Sei sulla pagina 1di 651

Methods for

General and
Molecular
Microbiology
3rd Edition

C. A. Reddy, Editor in Chief


T. J. Beveridge, J. A. Breznak, G. A. Marzluf, T. M. Schmidt,
and L. R. Snyder, Editors

ASM
PRESS

Washington, D.C.
Cover images:
From Beniac, D. R., G. J. Czamota, B. L. Rutherford, E.'F Ottensmeyer, and G. Harauz. 1997.
J. Microsc. 188:24-35; see chapter 4 (Beveridge et al.)
Rumen community, courtesy Frank Dazzo
G. L. Barron, University of Guelph; see chapter 42 (Thorn et al.)
M. McFall-Ngai, UW-Madison; see chapter 16 (Ciche et al.)

Copyright 0 2007 ASM Press


American Society for Microbiology
1752 N Street, N.W.
Washington, DC 20036-2904

Library of Congress Cataloginpin-Publication Data

Methods for general and molecular microbiology / C. A. Reddy, editor in chief ;


T.J. Beveridge ... [et al.], editors.-3rd ed.
D. : cm.
Includes indexes.
Rev. ed of: Methods for general and molecular bacteriology / Philipp Gerhardt, editor-in-chief ;
R.G.E. Murrav. Willis A. Wood. Noel R. Kriee.
-. leditorsl. c1994.
~

ISBN 978-1-55581-223-2
1. Bacteriology-Laboratory manuals. 2. Microbiology-Laboratory manuals. I. Reddy, C. A.
11. Methods for general and molecular bacteriology.
[DNLM: 1. Microbiological Techniques. 2. Molecular Biology-methods. QW 25 M59 20071

QR65.M26 2007
616.9'2010786~22
2007018796

All Righhts Reserved


Printed in the United States of America

1 0 9 8 7 6 5 4 3 2 1
Address editorial correspondence to: ASM Press, 1752 N St., N.W., Washington, DC 20036-2904,
U.S.A.
Send orders to: ASM Press, P.O. Box 605, Hemdon, VA 20172, U.S.A.
-
Phone: 800-546-2416; 703-661 1593
Fax: 703-661- 1501
Email: Books@asmusa.org
Online: estore.asm.org
PREFACE

Methods in General and Molecular Microbiology (MGMM) is MGMB (Bacteriology) to MGMM (Microbiology).
a substantially revised, updated, and expanded version of MGMM is intended as a laboratory manual of methods to
its successful predecessor, Methods in General and Molecular complement traditional microbiology textbooks and sys-
Bacteriology (MGMB), published by ASM Press in 1994, tematic treatises of general microbiology. However, aside
with my colleague Philipp Gerhardt serving as the Editor- from a chapter on bacteriophages, this manual does not
in-Chief. The objective of MGMM is to provide a compre- cover viruses, algae, or protozoa.
hensive yet moderately priced book that will serve as a first MGMM is organized into sections corresponding to key
source for traditional methods of microbiology as well as subject areas of general bacterial and archaeal biology (mi-
modern molecular biological methods commonly used with croscopy, growth, metabolism, and molecular genetics), in-
microbial cells. Both previous editions of this manual, cluding a new section, Community and Genomic Analysis.
MGMB and Methodsfor General Bacteriology (MGB, 1991), This is followed by the new section on mycology and ap-
were popular not only in North America, but also in many pendices on laboratory safety and culture preservation.
other developed and developing countries worldwide. It is Each section is divided into chapters, and each chapter has
hoped that the audience for this edition will be even wider a table of contents to help the reader see the organization
and will include not only card-carrying microbiologists, of the chapter and easily locate a specific topic of interest.
but also scientists in allied disciplines working with mi- A decimal numbering system is used throughout the man-
crobes as experimental models or as tools for various ual to facilitate quick identification, cross referencing, and
biotechnological applications. It is hoped that this manual indexing. A comprehensive list of references is provided at
will be on the bookshelf of every serious practitioner of mi- the end of each chapter.
crobiology in academic, industrial, governmental, and clin- The editors refrained from imposing a single, rigid for-
ical laboratories and that it will serve as a rich resource of mat on the authors other than requesting them to use
methods for the seasoned professional as well as for under- MGMB as a guide in preparing their manuscripts. The ed-
graduate and graduate students and postdoctorals. itors also did not delineate in detail the topic assigned to an
The primary stimulus for launching this new edition of individual author(s). In spite of this, the chapters turned
the manual (MGMM) came from the fact that over a out to be remarkably consistent in their format and scope.
decade had elapsed since publication of the previous edi- For many of the chapter topics covered in this manual, the
tion (MGMB) and during this time not only new methods state of the art has so developed that entire books dedicated
but also new areas of microbiology (such as community and to the topic covered by a single chapter are currently avail-
genomic analysis) had emerged. Considering the fact that able. Therefore, in many instances, the chapter authors
MGMB and its predecessors had been used widely around have primarily described reliable methods that are widely
the globe, it was also felt that a new section on general applicable to basic studies on the topic covered. Each chap-
methods in mycology would make this edition even more ter presents sufficient background principles to understand
useful to its worldwide readership. Owing to this increased the how and why of a given method, followed by a step-by-
scope and the need to accommodate additional method- step description of the procedure. Common problems, pre-
ologies, this edition contains 47 total chapters (as com- cautions, and pitfalls of the methods are presented as a p
pared to 31 chapters in MGMB). The section on systemat- propriate. In many cases, commercial sources for
ics from the previous edition has been dropped: much of equipment and materials are given. These suggestions,
the still-relevant material in that section has been incor- however, are not meant either to endorse or to exclude a
porated into other sections, whereas outdated material particular product. Many of the commercial sources for
from that original section was omitted altogether. products are also readily ascertained from catalogues pub-
Inasmuch as this edition covers methods for microbes rep- lished by various commercial firms, from annual buyers
resenting all the three domains of life, i.e., Bacteria, guides of various journals, and from extensive information
Archaea, and Eukarya, the title has been broadened from available on the Internet.

xvii
xviii rn PREFACE

As mentioned above, two sections appear in this edition Transmission Electron Micrographs, and Atomic Force
for the first time. Rapid advances in genomics and genome- Microscopy in the section Morphology and Ultrastructure;
based approaches have warranted the creation of the new Energetics, Stoichiometry, and Kinetics of Microbial
section, Community and Genomic Analysis. The ability to Growth and General Methods To Investigate Microbial
determine complete genome sequences and metagenomes Symbioses in the Growth section; Bacterial Respiration,
from microbial communities has revolutionized this field and Carbohydrate Fermentations, Metabolism of Aromatic
has extended our ability to obtain valuable information on Compounds, and two chapters on plant polymer-degrading
microbes that are yet to be cultured. Our rapidly expanding enzymes (Cellulases, Hemicellulases, and Pectinases and
knowledge in this area has been put together in seven excel- Lignin and Lignin-Modifying Enzymes) in the section on
lent chapters authored by leading researchers in this area. Metabolism; and Measuring Spontaneous Mutation Rates,
The new section on Mycology includes a comprehensive Genetics of Archaea, and Genetic Manipulations Using
chapter on general methods and three excellent chapters on Phages in the section on Molecular Genetics.
filamentous fungi, focusing on their physiology, metabolism, It goes without saying that a project of this size could
and genetic methods and on the principles and practice of not have been successfully completed without the excel-
DNA microarray technology. Again, each chapter is writ- lent cooperation and enormous effort on the part of the
ten by authorities in their respective areas of specialization. section editors and especially on the part of the authors,
In each of the sections retained from MGMB, existing many of whom have revised their chapters more than once;
chapters have been revised (quite extensively in many cases) the external referees, who provided rigorous and construc-
and a number of new chapters have been added. New au- tive critiques; and the dedicated publications staff of the
thors, revising existing chapters from the previous edition, ASM Press. Any corrections and constructive suggestions
have provided a fresh perspective, but in most cases the orig- for improvement from the users of this new edition are
inal authors of the analogous former chapter are credited. welcome.
Notable new chapters in MGMM, added to sections from
the previous edition, include Laser Scanning Microscopy, C. A. REDDY
Computational Image Analysis and Reconstruction from Editor-in-Chief
CONTENTS

Reviewers / xi 8. Antigen-Antibody Reactions / 138


Contributors / xiii LUCY AND JOSEPH s. LAM
M.MUTHARIA
Preface / xvii
Acknowledgments / xix SECTION II
GROWTH / 169
SECTION I
MORPHOLOGY Introduction / 171
AND ULTRASTRUCTURE / 1 JOHN A. BREZNAK,
EDITOR

Introduction to Morphology 9. Growth Measurement / 172


and Ultrastructure / 3 L. KOCH
ARTHUR
T. 1. BEVERIDGE,
EDITOR
10. Nutrition and Media / 200
1. Light Microscopy / 5 DAVIDEMERSONAND JANE TANG
AND CARL E ROBINOW
R. G. E. MURRAY
11. Enrichment and Isolation / 21 5
Sampling and Staining for light ANDREASTESKE, CYPIONKA, JOHN G. HOLT,
HERIBERT
2.
AND NOELR. KRIEG
Microscopy / 19
TERRYJ. BEVERIDGE,
JOHN R. LAWRENCE,
AND ROBERTG. E. MURRAY 12. Culture Techniques / 270
SYED
A. HASHSHAM
3. Laser Scanning Microscopy / 34
J. R. LAWRENCE
AND T. R. NEU
13. Energetics, Stoichiometry, and Kinetics
of Microbial Growth / 286
SYED
A. HASHSHAM
AND SAMW. BAUSHKE
4. Electron Microscopy / 54
TERRYJ. BEVERIDGE,DIANNE
MOYLES,
14. Physicochemical Factors
AND BOBHARRIS
in Growth / 309
JOHN A. BREZNAK N.
AND RALPH COSTILOW
5. Computational Image Analysis
and Reconstruction from Transmission 15. Phenotypic Characterization
Electron Micrographs / 82 and the Principles of Comparative
GEORGE
HARAUZ Systematics / 330
BRIANI.
TINDALL, JOHANNES SIKORSKI,
6. Atomic Force Microscopy / 96 ROBERTA. SMIBERT, AND NOELR. KRIEG
YVESF. DUFRBNE
16. General Methods To Investigate
7. Cell Fractionation / 108 Microbial Symbioses / 394
SUSAN SPROTT
F. KOVALAND G. DENNIS A. ClCHE AND SHANA K. GOFFREDI
TODD

vii
viii w CONTENTS

SECTION Ill 28. Measuring Spontaneous Mutation


METABOLISM / 421 Rates / 676
PATRICIA
L. FOSTER
Introduction to Metabolism / 423
C. A. REDDY,
EDITOR 29. Transposon Mutagenesis / 684
SILVIA ROSSBACH
AND FRANS
J. DE BRUIJN
17. Physical Analysis and Purification
Methods / 424 30. Plasmids / 709
SCOT B. MULROONEY,WILLIS
A. WOOD, MARCELOE. TOLMASKY,LUISA. ACTIS,
AND J. R. PATEREK TIMOTHY
J. WELCH, AND JORGE H. CROSA

18. Chemical Analysis / 462 31. Gene Transfer in Gram-Negative


LACYDANIELS, RICHARD S. HANSON, Bacteria / 735
AND JANE A. PHILLIPS JOSEPH E. PETERS

19. Enzymatic Activity / 504 32. Genetic Exchange in Gram-Positive


ROBERT
P. HAUSINGER
AND ALLEN
T. PHILLIPS
Bacteria / 756
J. KRISTICH,CHRISTINE
CHRISTOPHER E. SALOMON,
20. Permeability and Transport / 527 AND GARYM. DUNNY
E. MARQUIS
ROBERT

21. Bacterial Respiration / 539 33. Genetics of Archaea / 800


KEVINR. SOWERS,
PAULH. BLUM,
ROBERTP. GUNSALUS,
GARYCECCHINI,
AND SHILADITYA
DASSARMA
AND IMKESCHRODER

22. Carbohydrate Fermentations / 558 34. Genetic Manipulations Using


R. MEGANATHAN,YAMINIRANGANATHAN, Phages / 825
AND c. A. REDDY
GRAHAME HATFULL,
DEBORAHJACOBS-SERA,
MICHELLE
H. LARSEN,
AND WILLIAM
R. JACOBS, JR.
23. Metabolism of Aromatic
Compounds / 586 SECTION V
JEROME J. KUKOR,BORISWAWRIK, COMMUNITY AND GENOMIC
AND GERBEN J. ZYLSTRA ANALYSIS / 839

24. CelIulases, Hemicellulases, Introduction to Community


and Pectinases / 596 and Genomic Analysis / 841
E. HIMMEL,
MICHAEL JOHN 0. BAKER, THOMAS
M. SCHMIDT,
EDITOR
WILLIAM
S. ADNEY,
AND STEPHENR. DECKER
35. Characterization of Bacterial Genome
25. Lignin and Lignin-Modifying Sequences by Similarity
Enzymes / 611 Searching / 842
CARLOS
G. DOSORETZ
AND C. A. REDDY WILLIAM
R. PEARSON
SECTION IV
MOLECULAR GENETICS / 621 36. Reconstructing and Interpreting
Evolutionary Relationships / 856
Introduction to Molecular CHRISTOPHE J. DOUADY
AND CAMILLA L. NESB0
Genetics / 623
C. A. REDDY,
EDITOR 37. Microbial Nucleotide Fingerprints
L. R. SNYDER,
CO-EDITOR in Nature / 869
DAVIDM.KARL
26. Similarity Analysis of DNAs / 624
JOHN L. JOHNSON AND WILLIAM B. WHITMAN 38. Construction of BAC and Fosmid
Libraries from Naturally Occurring
27. Nucleic Acid Analysis / 653 Microbial Populations / 879
WILLIAM HENDRICKSON
AND DONWALTHERS EDWARD
E DELONG
Contents w ix

Single Cell Identification by Fluorescence Physiology, Metabolism, and Molecular


I n Situ Hybridization / 886 Aspects of Filamentous Fungi / 952
BERNHARD
M. FUCHS,JAKOB
PERNTHALER, GEORGE
A. MARZLUF
ANDRUDOLF
AMANN
Microbiological and Genetic Methods
Measurement of rRNA Abundance for Filamentous Fungi / 965
by Hybridization with ROWLAND H. DAVIS
Oligodeoxynucleotide Probes / 897 ANDA. JOHNCLUTTERBUCK
DANIEL
H. BUCKLEY
AND THOMAS
M. SCHMIDT
Principles and Practice of DNA
Analysis of Microbial Communities Microarray Technology / 978
with Denaturing Gradient Gel KRISHNAMURTHYNATARAJAN,
Electrophoresis and Terminal MATTHEW J. MARTON,
Restriction Fragment Length AND ALANG. HINNEBUSCH
Polymorphism / 909
TERENCE
L. MARSHAND CINDYH. NAKATSU APPENDICES

SECTION VI 46. Laboratory Safety / 997


MYCOLOGY / 925 W. EMMETT
BARKLEY
AND CLAUDIA
A. MICKELSON

Introduction to Mycology / 927 47. Culture Preservation / 1019


GEORGE
A. MARZLUF,
EDITOR ROBERTL. GHERNA
AND C. A. REDDY

42. Methods for Studying Terrestrial Fungal Author lndex / 1035


Ecology and Diversity / 929
R. G. THORN,J. SCOTT,AND M. A. LACHANCE Subject lndex / 1039
MORPHOLOGY AND
ULTRASTRUCTURE

Introduction to Morphology and 5. Computational Image Analysis and


UItrastructure 3 Reconstruction from Transmission
T. J. BEVERIDGE, Editor Electron Micrographs 82
GEORGE HARAUZ
1. Light Microscopy 5
R. G. E. MURRAY AND CARL F. ROBINOW 6. Atomic Force Microscopy 96
YVES F. DUFRENE
2. Sampling and Staining for Light
Microscopy 19 7. Cell Fractionation 108
TERRY J. BEVERIDGE, JOHN R. LAWRENCE, SUSAN E KOVAL AND G. DENNIS SPROTT
AND ROBERT G. E. MURRAY
8. Antigen-Antibody Reactions 138
3. Laser Scanning Microscopy 34 LUCY M. MUTHARIA AND JOSEPH S . LAM
J. R. LAWRENCE AND T. R. NEU

4. Electron Microscopy 54
TERRY J. BEVERIDGE, DIANNE MOYLES,
AND BOB HARRIS
Introduction to Morphology and Ultrastructure
T. J. BEVERIDGE

Microbiology and microscopy have enjoyed a strong syner- macromolecules within cellular space so that their location
gistic union since the earliest times of scientific investiga- can be seen (e.g., within nucleoids, division septa, secretory
tion. It is impossible for us to understand, today, how the apparatuses, endospores, etc.). Confocal laser scanning
crude lenses of Antonie van Leeuwenhoek or Robert microscopes (CLSMs) remain relatively expensive instru-
Hooke could have achieved the resolution they did, or how ments (compared to more simple light microscopes), but
the almost happenstance discovery of a fundamental differ- they are now readily available at the university, college, and
ential stain by Christian Gram would eventually partition (even) departmental levels. Because of their expense and
bacteria into two fundamental categories, gram-positive technical sophistication, most CSLMs are operated by a
and gram-negative bacteria. Because the individual cells of dedicated technical staff member who will aid students and
prokaryotes are so minute, they cannot be seen without researchers alike. Much of our current knowledge of the
some sort of microscopy. Accordingly, there necessarily has growth and spatial distribution of cells within biofilms has
been a fine history between microscopy and microbiology. come from the use of CSLMs.
Even the best light microscopes that are available today It is safe to say that almost all of our knowledge of the
can discriminate only shape and form unless distinctive fine structures in prokaryotes has been derived by using
staining procedures are used for differentiation of fine electron microscopes. Scanning electron microscopes
structures and added contrast. Consequently, other kinds of (SEMs) have clarified the topography of biofilms, colonies,
microscopy, often using wavelengths and energies different and (sometimes) individual cells. So-called environmental
from those of light, have been resorted to. Yet, light mi- SEMs can give images of specimens while they are partially
croscopy continues to be a hallmark of microbiology, and hydrated under high humidity. Transmission electron mi-
the first two chapters of this section cover the general croscopes (TEMs), though, have been our most important
methods employed. Chapter 1 outlines the basic compo- tool in microbiology for determining the macromolecular
nents of a simple compound light microscope and their op- structures of prokaryotes. Cell surfaces, juxtapositions of
eration. Chapter 2 broaches the various manipulations to enveloping layers, distributions of cytoplasmic constit-
microorganisms before they can be viewed; these include uents, intramembranous inclusions, and cytoplasmic parti-
the simple isolation of cells, chemical fixation, and both cles (e.g., polyphosphate, sulfur, and polyhydroxyalkoanate
general and specific staining protocols for cells and con- granules) have all been seen at high resolution, sometimes
stituent structures. confirming less resolving pre-TEM light microscopic evi-
It cannot be emphasized too much how important are dence.
these tried and true optical techniques to microbiology. It Electron microscopy requires difficult and (often) ex-
saddens those of us who specialize in microscopic imaging tensive preprocessing before specimens can be viewed.
to sometimes see how neglected and infrequently used is Frequently these procedures can induce artifacts that are
light microscopy in modern microbiological research labo- difficult to recognize for the uninitiated researcher. For this
ratories. Some laboratories do not even possess a workable reason, chapter 4 outlines tried and true protocols, using
light microscope! Researchers in these laboratories seem more traditional avenues for TEM, which most microbiol-
confident in their ability to maneuver relatively small mol- ogy laboratories should be capable of. Here, for even the
ecules of DNA as they manipulate the genetic message in difficult procedure of thin sectioning, cells can be grown,
cells and have little desire to actually visualize the cellular chemically fixed, stained, and embedded in plastic without
results. In these laboratories, possible altered phenotypes, specialized equipment. These samples can then be taken to
microbial contaminants, and lethal or growth effects are in-house TEM facilities or shipped over larger distances.
rarely monitored by light microscopy during such refined Because SEMs and TEMs are expensive to buy, operate,
molecular biological manipulations. Even more, by mi- and maintain, these microscopes are usually clustered
croscopys absence, a clear tangible distance is established within a university facility containing ancillary equipment
between the experimenter and that being experimented for the manipulation of your samples. Specimens can be
on, which lends a certain remoteness to the experimental thin sectioned, TEM grids can be prepared, and an opera-
process. We need to rediscover our traditional microscopic tor can be supplied for viewing specimens. Remember, the
roots in microbiology, and chapters 1 and 2 are a required technical staff members within these electron microscopy
knowledge base for students and researchers alike. facilities are expert microscopists but they may not be ex-
Chapter 3 covers confocal laser scanning microscopy, perienced in microbiology. For this reason, it can be ad-
which is fast becoming an essential part of microbiologys vantageous to network with an experienced microbiology
imaging armament. Good light microscopes combined with structuralist. Over the last decade, a renaissance in micro-
excellent coherent light sources (lasers) provide accurate biological ultrastructure has gradually occurred with the
resolution plus defined focal sections through relatively advent of cryogenic techniques. Here, cells are vitrified to
thick specimens. Specialized (but often user-friendly) com- preserve their native structures by ultrarapid freezing. One
puter software allows the easy manipulation of serial focal such cryogenic method, freeze substitution, is described in
sections so that cells located in x, y, and 2 axial planes of chapter 4 since it has brought remarkable clarity to a num-
the sample can be visualized and stacked to form three- ber of microbial structures, especially the enveloping layers
dimensional images. Fluorescent probes, excited at specific of bacteria. With some effort and dedication, freeze substi-
wavelengths, provide exquisite regional discrimination of tution can be done in a typical microbiology laboratory.

3
4 MORPHOLOGY AND ULTRASTRUCTURE

However, more dedicated cryogenic techniques, such as broken or permeabilized so that their cellular constituents
those required for the production of frozen foils or frozen can be fractionated to purity. It also gives valuable advice
hydrated thin sections, need more dedicated technical skill on how to determine so-called biochemical signatures of
and equipment (including cryoTEMs with electron energy structural components (e.g., the lipopolysaccharide of the
filters) and are best left to the experts. gram-negative outer membrane), which is essential for
As previously mentioned, user-friendly computer soft- tracking the various fractionations and (eventually) deter-
ware is an essential part of CSLMs and is included with mining purity. Many of the described methods are used to
them. However, computer manipulation of electron micro- obtain the components of cells that are eventually imaged
graphs is more difficult and uses sophisticated Fourier trans- by the TEM techniques described in chapter 4.
formation- or correlation averaging-based analysis to reduce One of the more difficult aspects of any form of mi-
the signal-to-noise ratio within the raw images. Chapter 5 croscopy is to be able to unequivocally identify specific
is a short but comprehensive explanation of image en- structures or macromolecules within a cell. Here, a specific
hancement in electron microscopy. It also includes the ex- probe to label such a constituent is an invaluable tool. One
citing new techniques of electron spectroscopic imaging (an of the best probes is an antibody specific to the structure of
electron-filtering method) and tomography (three-dimen- interest. Chapter 8 describes the techniques used to obtain
sional imaging by tilting the specimen) as applied to such such probes and how to ensure their specificity. These
small particles as ribosomal subunits so that rRNA can be probes can be polyvalent antibodies obtained from an ani-
seen as it folds its way around putative ribosomal proteins. mal after immunization or monoclonal antibodies devel-
Scanning probe microscopy, (SPM) which is represented oped through myeloma-spleen hybridomas. Highest speci-
by atomic force microscopy (AFM) in chapter 6, is an en- ficity is via monoclonal antibodies with discrimination of a
tirely different type of microscopy in that no lenses are used. single epitope, but highest labeling capacity is through poly-
Here, a sharp tip on a bendable cantilever is dragged over a clonal antibodies capable of labeling many epitopes at a sin-
sample in a raster pattern with the tip forced up and down gle time. Both factors have to be weighed against one an-
depending on the contours found on the specimen. The tip other when determining the best probe for your sample.
actually is in contact with the specimen and, depending on Once the immunological probe has been obtained, it must
the physical attributes of the samples surface, can detect be labeled with a visual marker that can be detected by mi-
the topography of macromolecules and even atoms. Unlike croscopy. For light microscopy, this label is usually a fluo-
other forms of scanning probe microscopy, such as scanning rescent marker (e.g., fluorescein; see chapters 2 and 3 ) , but
tunneling microscopy, AFM can be done under water, for electron microscopy an electron-dense marker (e.g., col-
which makes it a valuable microscopy for biological sub- loidal gold; see chapter 4) is needed. For AFM, the probe
stances. Chapter 6 explains the basic setup of the atomic must be large enough to be visualized against the typical to-
force microscope and how AFM is making inroads in mi- pography of the cell (e.g., a bacteriophage that is specific for
crobiology. Another valuable trait of the microscope is that the surface).
the tip, on a cantilever of a known spring constant, can Together, the chapters in this section are an integration
probe the physical nature of the sample to determine such of the methods generally used in microbiology to visualize
important traits as elasticity, viscosity, and adhesion. At one microorganisms and their constituent parts, along with the
time atomic force microscopes were highly specialized and techniques required for component isolation and visual de-
infrequently encountered in a research setting, but now tection. Although these chapters make a correlated union,
they are an item that all research-intensive universities they will also be helpful when applied to some of the tech-
have. niques described in other sections and chapters of this book.
It is one thing to discern the structures of microorgan- Indeed, the images that have been chosen to portray the
isms by looking at intact cells, but it is an entirely different various stains, cells, and structures in this section have also
and more demanding task to decipher the contours of their been chosen so as to help illustrate the microorganisms
individual components. For this, the components usually mentioned in other chapters of the book. One of the great
have to be isolated and purified. Once these steps are ac- pleasures of combining microbiology and microscopy is to
complished, these cellular constituents can not only be vi- enjoy the simplicity and beauty of microbial shape and
sualized by microscopy (usually by using either TEM or form, but each image also possesses a wealth of scientific in-
AFM), they can also be used for biochemical analyses to de- formation. This marriage of science and visual enjoyment
termine their chemical makeups and possible activities. has its roots in the 20th century and continues today with
Chapter 7 describes the manner in which microbes can be more technical sophistication and interpretative skills.
.

Light Microscopy
R. G . E. MURRAY AND CARL F. ROBINOW

1.1. BASIC MICROSCOPY ....................................... 6


1.1.1. Instrumentation ....................................... 6
.
2. Operating Steps ........................................ 7
.
3 . Operating Rules ....................................... 7
.4. Comfortable Microscopy ................................. 8
.5. Immersion Oils ........................................ 8
.6. Care and Cleaning ...................................... 8
.7. Troubleshooting ....................................... 9
1.2. HIGH-RESOLUTION MICROSCOPY ........................... 9
1.2.1. Resolution Requirements ................................. 9
.2. Objectives and Oculars .................................. 9
.3. Condensers ........................................... 11
.4.Mirrors .............................................. 11
.5. Filters ............................................... 11
.6. Light Sources ......................................... 11
.7. Optimum (Koehler) Illumination ........................... 12
1.3. DARK-FIELD MICROSCOPY ................................. 12
1.4. PHASE-CONTRAST MICROSCOPY ........................... 13
1.4.1. System Assembly ....................................... 14
.2. Specimen Preparation ................................... 14
.3. Phase Condenser for Low-Power Dark Field ................... 14
1.5. INTERFERENCE MICROSCOPY .............................. 14
1.6. FLUORESCENCE MICROSCOPY .............................. 15
1.6.1. Fluorescence Systems ................................... 15
.2. Confocal Scanning Microscopy ............................. 15
1.7. CELL MEASUREMENT ..................................... 15
1.7.1. Stage Micrometer ...................................... 16
.2. Eyepiece Graticule Micrometer ............................. 16
.3. Eyepiece Screw Micrometer ............................... 16
1.8. PHOTOMICROGRAPHY .................................... 16
1.9. REFERENCES ............................................. 17

The light microscope is essential in any laboratory or class- resolution of detail and the best of photomicrography. and
room that is concerned with microbes. cell cultures. and (iv) an instrument equipped for fluorescence microscopy.
the interactions of cellular life in nature or in experiments. Many modern microscopes are equipped to combine two or
Its primary functions are to assist in description and identi- more of these functions and are very satisfactory if the tran-
fication. the monitoring of growth or processes. the control sitions are easy to accomplish. An additional instrument. a
of biological experiments. and the observation of micro- stereoscopic dissecting microscope. is valuable for under-
scopic phenomena . Thoughtful and well-directed mi- taking the isolation of bacteria from nature on agar plates
croscopy provides basic information and an appreciation of when colonies early in growth are small and close together.
the living world . Furthermore. judicious use can save the Excellent microscopes are available. Those from first-
biologist from many a grievous error and will help in the rank manufacturers are all of comparable qualities and offer
anticipation of events or the recognition of novelty. All a range of accessories and support structures; the choice is
this can be accomplished at several levels of resolution and likely to be based on convenience factors and personal pref-
in a number of modes of preparation and presentation . erences. Other choices may involve technical require-
Light microscopy is made easy. interesting. and useful ments. and it is always advisable to make direct compar-
for bacteriological purposes if at least four different kinds of isons of the possible instruments and the alternative
instruments are readily available. permanently set up for accessories for a given material under given conditions of
work. and maintained in good order. These are (i) a rugged. work. The really sophisticated instruments are expensive.
well.equipped. but uncomplicated microscope for encoun- With care. good work can be done with first-class optics fit-
ters of the first kind. (ii) a phase-contrast microscope. (iii) ted to a general-purpose or even a student microscope
an optimally equipped and adjusted microscope for high stand. However. it is hard to equal the modern instruments

5
6 w MORPHOLOGY AND ULTRASTRUCTURE

from the best manufacturers. What matters is an awareness


that a good microscope (whatever its age), a discerning
choice of components, and a knowledge of how to use them
enhance the results and the pleasure derived from the work,
save time, and improve accuracy.
The descriptions that follow are based on an old-fash-
ioned basic microscope with separate light source, mirror,
and adjustable and centerable components. The optical
principles are no different for the enclosed modern instru- spedmen slide
ments with substage illumination, which seem simpler to
use but can suffer from similar problems. The use of poorly
adjusted and poorly illuminated microscopes is so wide-
spread that most scientists are no longer aware of what mi-
croscopes can attain and are astounded when given a
demonstration of the best resolution obtainable with even a
basic microscope when it is used in accord with the princi-
ples outlined below (see sections 1.1 and 1.2). condenser
tenses *- -
s t a g e y
1 .l. BASIC MICROSCOPY
tamp adjustment
The application of microscopy to the basic requirements of
bacteriology is simple, and the questions answered with its
help are as follows. Are there objects of bacterial size and
staining properties? Are they consistent in size and shape?
Are the bacteria gram positive or gram negative? Is there
more than one kind of organism present? . . . And so on
to more sophisticated questions. These primary questions
can usually be answered without attaining the highest reso- FIGURE 1 Basic light microscope and its parts. The assis-
lution, so a simplified procedure suffices as long as the user tance of R. Van Twest, Department of Microbiology, University
remembers that higher resolution demands improvement by of Guelph, is appeciated.
adjustments at almost every step.
The beginner would be well advised to have at hand a
fully illustrated manual for microscopy, of which there are
many available (1, 3, 6, 7, 9, 10, 14, 16, 22).
quire. It should have an iris diaphragm (aperture di-
1 .I . I . Instrumentation aphragm) and a movable filter carrier incorporated below
The basic microscope (Fig. 1) should be equipped with the the lower lens. Some condensers have a movable top ele-
following. ment (essential for work with high-power objectives) that
tilts out of the way for purposes of low-power examination.
1. Oculars (eyepieces) of a magnification of at least x 10. A condenser that has three lenses, is aplanatic, and when
Prolonged observation is more comfortable with the use of
under oil, has a numerical aperture (NA) of 1.3 to 1.4 is
a binocular system. The oculars fit into the microscope preferable when high-resolution as well as general use is ex-
tube, which may be fixed or adjustable in length (to 180
pected (see section 1.2.1).
mm, or whatever tube length the manufacturer recom-
6. The light source in modern microscopes is built into
mends). Observations on bacterial structure at high resolu-
the base to form a unit with a fixed mirror placed to illumi-
tion are assisted by higher-magnification (X12 or X15)
nate the condenser through a plane glass in a mount incor-
compensating oculars (see section 1.2.2). Another circum-
porating an iris diaphragm (field diaphragm). Filters can be
stance in which high-power (15x) oculars assist vision is
placed onto the opening when needed. The light itself is
the scanning of growth on agar plates with a 1OX objective.
usually mounted in a centerable collar so that the bulb can
2. Low-power, dry objectives (e.g., lox and 40X). be positioned to focus centrally in the axis of the condenser.
These are essential for looking at growing cultures and for
Some specialized microscopes add the optical train for epi-
identifying rewarding areas for study at high magnification,
illumination which directs the light to a beam splitter
according to the nature of the preparation. Ideally, the ob- halfway up the tube. This beam splitter sends the beam
jectives (including the oil-immersion objective) should all
down to the objective, which now plays the roles of both
be parfocal, i.e., close to being in focus when exchanged by
condenser and objective. Epi-illumination is most useful for
the Kosepiece carrier.
fluorescence microscopy (23). There are also inverted mi-
3. A n achromatic oil-immersion objective (90X to croscopes with the objective below the stage and a long-
1OOX). This is essential to bacteriologists for observing the
focus condenser above it; this setup is useful for examining
finest details in specimens.
tissue culture bottles and the like.
4. A stage with a mechanical slide-holding device or
with simple spring clips. If a mechanical stage is provided, it Old-fashioned but eminently useful microscopes employ
should be easily removable so that petri plates may be ex- an external light source and, in a gimbal under the micro-
amined at low power. scope, an adjustable two-sided mirror (flat on one side for
5. A substage condenser of the improved Abbe type. normal work and concave on the other for low-magnification
This is essential for use with oil-immersion objectives, be- examination without the condenser). The simplest form of
cause it provides the quality and quantity of light they re- lamp, a frosted bulb behind a bulls-eye lens, is adequate
1. Light Microscopy 7

for much work. However, the use of a lamp with a collec- 4. Replace the eyepiece to examine the specimen, and
tor lens, capable of projecting a sharp image of the light make any final adjustments for the best illumination and
source and provided with a filter carrier as well as an iris di- focus.
aphragm (field diaphragm), results in greatly improved im- 5. Turn the nosepiece to any other objective desired.
ages of details in stained preparations, especially when Many microscope makers provide parfocal objectives (i.e.,
Koehler illumination (see section 1.2.7) is used. A frosted- the focal position for one is nearly identical to that for all
glass diffusing screen held in the filter carrier of the con- the others). Clearance is sufficient for the dry lenses. If the
denser can be of value for low-power microscopy, especially lenses are from different manufacturers or otherwise mis-
when a three-element condenser is used. matched, it is wise to raise the tube a little before bringing
into place and focusing the oil-immersion objective, which
1 .I .2. Operating Steps has a small focal distance (often 0.2 mm). The clearance
will be even smaller when the specimen is mounted under a
Two basic kinds of bacteriological preparations are used for
coverslip (which should be no. 1 thickness).
simple microscopy: a specimen under a coverslip, usually a
wet mount (see chapter 2, section 2.2.1.1), or a stained
6. If going direct to oil immersion, apply a drop of oil
over the specimen on the slide before placing it on the
smear on the slide surface (see chapter 2, sections 2.2.1 and
stage. If the specimen has been scanned for a rewarding area
2.2.4). The former preparation is suitable for examination
with a lower power, it is convenient to apply the oil drop to
with all objectives. Start with a dry objective, either low
the slide in place with the nosepiece turned halfway to the
power ( l o x ) or high dry (40X), to see whether the spec-
correct objective before swinging the oil-immersion objec-
imen is rewarding and to select an area for study or to de-
tive into position.
tect motility. Then use an oil immersion objective (90X to
1OOX) for more definitive observation. A stained smear is
7. Using and focusing the oil immersion objective takes
a little practice and care even if the objective has a spring-
usually not suitable for study with dry objectives (although
loaded front end to protect both the lens and slide from a
they may be used to locate the bacteria when the smear is
ham-fisted operator. While looking from the side, lower the
very thin) because the smear must be covered with immer-
objective slowly onto the oil drop and down just a little
sion oil to provide a clear and translucent image of the bac-
more so that the lens almost touches the slide. Using the
teria. The steps described below assume that a progression
coarse focusing wheel and watching in the eyepiece, look
in levels of magnification will be used, although one can go
for the specimen while raising the objective gently. When
directly to using an oil immersion objective.
the specimen is found, use the fine focus wheel. Adjust the
Supposing that a slide preparation is to be examined by
illumination as described above, if needed. Focus with care;
use of the simple illumination source described above, the
contact between the objective and the slide can crack the
steps in operating the basic microscope are as follows.
slide and damage the lens mounting. Adjust for critical illu-
1. With the eyepiece and lox objective in place, the mination as described in step 3 above.
condenser lens fully up and almost level with the stage, and 8. O n a binocular microscope, adjust the eyepiece tube
the aperture diaphragm of the condenser fully open, tilt the that has a knurled ring for adjusting tube length so that
mirror to provide illumination up the optical axis. Adjust both eyes are in exact focus.
the objective by using the coarse control wheel to focus the 9. If the image is hazy, moves, or is only dim and not
light field, and then center it with the mirror. Use the flat sharp, there is something wrong. It may be necessary to
mirror for most microscopy; the concave mirror is used only clean the front lens of the objective (with lens paper only).
for low-power microscopy when a condenser is not being A common cause of hazy images is bubbles entrapped in the
used. immersion oil (take out the eyepiece and look down the tube;
2. Place a suitable slide on the stage, and focus an the bubbles are then easily identified). To remove the bub-
appropriate objective to get a sharp (not necessarily well- bles, raise the objective, turn the nosepiece, wipe off the oil
illuminated) image of the specimen. with lens tissue (the bubbles usually come up onto the objec-
3. Improve the illumination by adjusting the positions tive), and then turn back the objective and refocus. A poor
of the condenser and the aperture diaphragm to get the image may also be caused by having accidentally closed the
best image and appropriate contrast. The simplest way to diaphragm, moved the lamp, or moved the mirror. See sec-
determine the optimum condenser position is to remove tion 1.1.7 for other troubleshooting suggestions.
the eyepiece and look down the tube at the visible illumi-
nation of the back of the objective lens. While looking, ad- 1 .I .3. Operating Rules
just the position of the condenser to give the fullest and Remember the following simple rules of microscopy and
most homogeneous illumination of the objective. Any lack practice them.
of concentration can be recognized and adjusted by tilting
the mirror. Reduce glare (light scattering from lens Rule 1 . The focused image requires the best possible quality
mounts) by closing the aperture diaphragm of the con- and quantity of light.
denser (if available) just enough to impinge on the bright Rule 2. If there is too much light, it is best to reduce it with a
disk. This will approximate critical illumination, i.e., the neutral-density filter or with a voltage controller. The light
light source is centered and focused in the plane of the can also be moved farther away, but then the condenser
specimen. This whole procedure applies to all objective must be refocused. With oil-immersion (large-aperture)
lenses as long as they are first focused on the specimen. objectives, avoid reducing the illuminating aperture (by
Alternatively, or to check that the critical illumination is lowering the condenser or closing the aperture diaphragm
properly adjusted, place a wire loop against the lamp or more than 10% of the width of the opening), although this
against the frosted glass of other illuminators (the effective stratagem is helpful for small-aperture, dry objectives and
light source) while the objective is in focus on the speci- for use in looking at living, unstained cells.
men. Then adjust the condenser to get the sharpest image Rule 3 . If there is too little light for the objective used,
of the wire loop on the specimen. either there is something in the way, the alignment has
8 m MORPHOLOGY A N D ULTRASTRUCTURE

shifted, or a more appropriate illumination system (lamp chambered bottle in which the stopper is formed into a ves-
and condenser) is needed. sel for oil and the base is made to hold the solvent for clean-
Rule 4. The specimen must be part of a (nearly) homoge- ing. Squeeze bottles tend to generate bubbles unless used
neous optical system and therefore must be mounted in carefully.
oil, water, or another optically appropriate medium. No Oiling the condenser to the slide, which is necessary for
details can be discerned in dried, stained films of bacte- highest resolution with the oil-immersion objective (see
ria viewed with dry objectives. section 1.2.1), can be a messy procedure. It demands a high-
Rule 5. Keep all lenses clean and free from dust, finger- viscosity oil and some simple precautions both in mounting
prints, noseprints, and the grime from oily eyelashes and in demounting of a preparation, as follows.
(with lens paper only). 1. Place a drop of high-viscosity oil on the top lens of the
condenser, and then lower the lens below the stage level.
1 .I .4. Comfortable Microscopy Avoid bubbles in the oil drop.
Long hours at a microscope are needlessly tiring if the mi- 2. Place a small drop of oil on the underside of the slide
croscopist cannot sit comfortably and upright while looking below the area you wish to examine.
into the instrument. Inclined eyepiece tubes are no help if 3. Set the slide down on the microscope stage with the
the table or chair height is inappropriate and causes stoop- drop central in the hole, and hold the slide there while ad-
ing and straining. Binocular viewing also contributes to justing the mechanical stage or clips to keep the slide in
restful study. place.
Chairs (including stools in teaching laboratories) should 4. Raise the condenser so that drop meets drop and until
be adjustable. For the very tall, a block of wood can be cut the top condenser lens is fully covered with oil.
and used on a table of fixed height to raise the microscope 5. Place a drop of oil over the specimen, and focus with
to just the right position. The whole object of these adjust- the objective. Then adjust the condenser to give Koehler il-
ments is to allow the microscopist to look down the tube lumination (see section 1.2.7).
with minimal deflection of the neck or back. A comfortable 6. When demounting the preparation, lower the con-
position minimizes strain and the misery that can result. denser before picking up the slide. Lateral movement or
wide scanning may lead to oils being deposited or creeping
1.1.5. Immersion Oils under the stage. Remember this possibility, and clean the
stage underside when cleaning the top lens (with lens tis-
Immersion oils are designed for use with oil-immersion
sue!) after use.
lenses and provide an environment of the correct refractive
index for the front lens of the objective. They also generate
7. Only condensers with integral or screw-on top ele-
ments are suitable for oil immersion. Movable swing-out
a homogeneous system, approximating the refractive index
top elements on some universal condensers (i.e., those
of glass, and they clarify the dried cells of bacteriological
that can be slid aside with a lever for low-power work) must
slide preparations by permeating and embedding the stained
be oiled with great care or not at all because subsequent
cells.
cleaning is difficult.
The commercially formulated oils recommended by mi-
croscope manufacturers have been tested to be sure that
8. Cleaning procedures for condensers after use are the
same as these for objectives. Remove the condenser from its
they do not have acidic or solvent effects on lenses or their
substage mount to avoid (or clean up) oil that has flowed
mountings; substitutes are not recommended and may be a
around the condenser housing. Remove excess oil with lens
false economy. Certain properties can be chosen for special
tissue, and then clean the lenses with fresh lens tissue mois-
purposes (5), such as very-high-viscosity oil to fill wide gaps
tened with xylol or the appropriate solvent recommended
or to stay in place on horizontal or inverted microscopes
and low-fluorescence oil for fluorescence microscopy. For
by the microscope maker.
general purposes, use immersion oil of a fairly high viscosity 1 .I .6. Care and Cleaning
(e.g., type B, high-viscosity oil of 1,250 cP from Cargille
Microscopes are remarkably tough and durable. With rea-
Laboratories Inc., Cedar Grove, N.J.), which stays more or
sonable care and protection from the elements (particularly
less where it is placed and creeps less than low-viscosity oils.
the hostile acid-laden air of some laboratories), a micro-
The only problem with this level of viscosity is that bubbles
scope will last a lifetime or more. Take care of the micro-
are easily entrained, but this problem is avoidable.
scope as follows.
Oil immersion objectives should always be cleaned after
use or, at least, at the end of a days work. Use lens paper 1. Protect the microscope from dust and grit. Put a dust
moistened with xylol or another appropriate solvent as cover over the microscope when it is not in use; the best
recommended by the lens manufacturer. Ethanol and cover is a rigid, transparent, and all-enclosing glass or plas-
methanol are recommended by Wild-Leitz for its lenses, but tic bell. Do not allow dust to accumulate anywhere; it
xylol is also safe to use. The modern nonoxidizing oils are drifts into the lenses and mechanisms.
less of a problem than the traditional cedanvood oil (which 2. The moving parts, especially the rackwork and gears,
hardens, oxidizes, and leaves annoying residues), but they should be cleaned and treated with new grease at long in-
still slowly alter and may also creep where not wanted. Take tervals (the grease for model-train gears from a hobby shop
caution: modern immersion oils are safe, but before 1972 works well). Do not use thin oil on gears or bearing surfaces;
nondrying oils were often formulated with polychlorinated the tube or condenser may then sink from its own weight.
biphenyl compounds, which are now believed to be car- 3 . Clean the stage regularly, and mop up any spills.
cinogenic and toxic, so look out for old bottles! 4. Keep the lenses clean, and especially, clean up after a
Never use more oil than necessary; usually 1 drop will session of oil immersion work. Never use a finger instead of
do. A good oil bottle will help to reduce mess; e.g., use a an appropriate lens tissue (see below).
glass bottle with a broad base and an applicator (glass rod or 5. Do not attempt to repair an objective lens or the fine
wire loop) attached to the cap. Among the best is a double- adjustments. These repairs are best left to professionals.
1 . Light Microscopy 9

Good microscopes deserve a professional cleaning and ad- book by James (10). For additional advice, consult other
justment every 20 years or so! Microscope dealers usually books (see References), an experienced microscopist, or an
have or can recommend a repair facility. instrument technician to identify the problem. If the prob-
6. Keep the tube closed at all times with an eyepiece, and lem is optical, work along the light path in systematic order
keep all objective mounts filled or plugged, to minimize dust from the lamp and then through the condenser, specimen,
in the tube. This should not prevent you from removing an objective, and ocular. Adjust at each step, and determine
eyepiece to check the quality of illumination of the objec- the effect of adjustment on the problem; this is where Table
tive. 1 is useful.
The old soldiers among microscopes can be cleaned,
repaired if necessary, and put back to good use. The brass 1.2. HIGH-RESOLUTION MICROSCOPY
bodies on ancient ones (How elegant they are! How fine
The research microscope comes into its own for the resolu-
their lenses may be!) can be rubbed down with a lightly
tion of fine detail at high magnification. Its use is not called
oiled cloth or one lightly moistened with tarnish remover
for in determining the outcome of a Gram staining test, but
and then a dry cloth. Modern black finishes should just be
it will help to settle questions about flagella and matters re-
kept clean.
quiring the perception of fine detail; also, high resolution
Clean lenses make for a dramatic improvement in image
allows for the sharpest photomicrographs. For such purposes
quality over that achieved with dirty lenses. Optical sur-
it is essential to pay close attention to the quality of the op-
faces need special care in cleaning, and some general rules
tical components. Achieving high resolution as well as
apply- freedom from chromatic and spherical aberration requires
1. Keep a good supply of lens tissue (lens paper), a soft attention to basic principles (1, 13, 18).
brush (artists watercolor brush, 0.6 cm wide), and a nebu-
lizer bulb with a short narrow rubber tube attached near the 1.2.1. Resolution Requirements
microscopy area. Keep them scrupulously clean and store Resolution (R) is the shortest distance between points of
them in a dust-free box. detail which will still appear as a distinct gap in the images,
2. Nonadherent dust can be easily removed by using the visual or photographic. R = A/2NA, where A is the wave-
brush, puffing with the rubber bulb, or wiping with lens tis- length of the light used and N A is a measure of the light-
sue after breathing on the optical surface. gathering power of the objective. The purpose of a good re-
3. Finger marks, other adherent grease, and dirt must be search microscope is to help make R as small as possible.
polished off with lens tissue doubled over a finger and barely The smallest values of R require not only an objective of
moistened with xylol or the recommended solvent (see sec- high NA but also a condenser of equivalent quality (see sec-
tion 1.1.5). Do not flood lenses with solvents, and never use tion 1.2.3) and the use of short-wavelength light. Un-
(without specific recommendation) alcohol, ether, or ace- fortunately, light of the most effective wavelength (namely,
tone for fear of penetrating the cement between lenses. If UV light of about 365 nm) is not perceived by the eye and
the lens has a raised mount, clean the edges with moistened does not pass through glass. Indirect methods of microscopy,
lens tissue wrapped around an applicator stick. requiring a microscope with lenses (lamp, condenser, and
4. If xylol is not effective, try using tissue moistened with objective) made of quartz (permeable in various degrees to
distilled water. UV light), take advantage of the short wavelength of UV
5. The usual way of cleaning oil from objective and con- light, as is the case for fluorescence microscopy. The best re-
denser lenses is to wipe most of it away with lens tissue and sults with visible light are obtained in the green-yellow re-
finish with tissue barely moistened with xylol. Oil and gion of the spectrum because it is the wavelength to which
grease can also be efficiently removed with the surface of a the eyes are most sensitive (close to the mercury green spec-
freshly broken piece of polystyrene foam (common packing tral line) and for which microscope objectives are designed
material) by pressing it against the objective front lens and to transmit with a minimum of aberrations. So much for the
rotating it (H. Pabst quoted by James [lo]). The foam has X part of the expression for resolution.
lipophilic properties. Xylol dissolves the foam, so do not use The N A of oil immersion objectives is usually 1.30 or
both. 1.32, and the best ones may have an N A of 1.40. The N A
6. Never take an objective apart to clean the compo- represents the sine of one-half of the angle described by the
nents, because the interlens distances are critical. Dust can cone of light admitted by the objective lens, multiplied by
be puffed from the back lens or lifted with a superclean the refractive index of the medium through which light
brush. passes on its way to the lens. The maximum value of the
7. Dust on oculars is a constant problem and produces sine function cannot exceed 1.0, but the refractive index of
dark spots which move around as the lens is rotated. The the medium through which light passes on the way from the
eyepieces are quite simple in construction (top and bottom condenser to the objective can be raised to that of optical
lenses with a fixed diaphragm between), and both lenses glass (i.e., 1.5150) by the use of immersion oil. I t is this fact
unscrew. If cleaning of the external top and bottom surfaces that allows lenses of NAs of greater than 1.0 to be filled
does not remove a spot, there may be particles on the inner with light. Provided that certain other conditions are met,
surfaces. Be careful of bloomed lens surfaces (those with the resolving power of the costly objectives with NAs of
antireflective coating which appears iridescent blue), and 1.30 to 1.40 engraved on their glittering housings can be
use a brush or tissue with care because some blooms are soft; utilized to the fullest.
try puffing first.
8. Keep fingers off optical surfaces! 1.2.2. Objectives and Oculars
Large-aperture achromatic or apochromatic objectives are
1 .I .7.Troubleshooting spherically correct for one or three colors, respectively, in
The list of technical problems, causes, and remedies shown the center of the field of view (where most of the work is
in Table 1 is taken (with permission) from the excellent done at high magnification). The two kinds of objectives
10 M O R P H O L O G Y AND ULTRASTRUCTURE

TABLE 1 Common problems in light microscopy and their causes and remedies"
Problem Possible cause Remedy( ies)

Coarse adjustment is too stiff Mechanism adjustment was faulty With many stands, adjust simply by mov-
ing the two coarse control knobs in op-
posite directions
Dirt in rackwork Clean and apply new grease
Tube or stage sinks spontaneously under Incorrect adjustment of rackwork As with first remedy
its own weight (image drifts out of Lubrication with too-thin oil As with second remedy
focus) Faulty adjustment of focus control As with first remedy
Focus adjustment stops before image is Fine adjustment at the end of its travel Bring a lox objective into position with
focused the revolving nosepiece, set the fine
focus control at the middle of its
range, and then focus with the coarse
adjustment
Drift of focus with the slightest movement Objective insufficiently screwed into the Seat the objective fully into the nosepiece
of the fine adjustment (especially with revolving nosepiece
oil immersion objectives) Surface of the coverslip stuck to the Use less-viscous immersion oil; clip
objective by the layer of oil specimen firmly
Veiled, spotty image Dirt or grease (i) on the eyepiece (spots Clean where necessary
move when the eyepiece is rotated in
the tube), (ii) on the objective, (iii) on
the coverslip (spots move when the
specimen is shifted), or (iv) on any sur-
face of the illumination apparatus
Sharply focused spots or specks in the Dirt (i) on the light source or on the dif- Clean where necessary; when the contam-
image which change and disappear fusing screen in front of it with critical inated surface cannot be reached,
when the condenser is moved up and illumination, (ii) on the cover plate change the focusing of the condenser
down of a built-in lamp, or (iii) on a filter slightly or tolerate the problem for the
near the cover plate with Koehler sake of resolution
illumination
Hazy image that cannot be brought Wrong immersion medium (oil instead of Use correct immersion medium
sharply into focus air, air instead of oil, air bubble in oil)
Transparent contamination on objective Clean where necessary
front lens
Coverslips too thick or too thick a layer Use a correct coverslip or an appropriate
of mounting medium objective
Irregularly distributed remnants of immer- Clean the coverslip with dry cloth or
sion oil on the coverslip when high- paper tissue; beware of solvents, as
power dry objective is used some may weaken or dissolve mounting
medium
Slide upside down on the stage (only with Invert the slide; make sure a label is not
high-power objectives) stuck to the wrong side of the slide
Object field partially illuminated Filter holder partially in the light path Move the filter holder
Objective not clicked into position Reposition the objective
Condenser (or swing-out lens) not in the Realign the condenser
optical axis
Object drifts diagonally during focusing A lens is not centered in the optical axis Check centering of the lenses at all points
in the optical path
Object field unevenly illuminated Mirror not correctly in position Reposition the mirror
Condenser not centered (with critical Realign the condenser
illumination)
Irregularity in light source and/or diffusing Move the condenser slightly up and
screen (with critical illumination) down; use ground glass in front of the
light source
Drift of cloud across the field; after this, Air bubble in the immersion oil; oil in Wipe off the oil from the specimen, and
image out of focus (oil immersion) image space with a dry objective set up anew; clean the slide and objec-
tive carefully
~

(Continued on next page)


1. Light Microscopy 11

TABLE 1 (Continued)
Problem Possible cause Remedy( ies)
Sharply delineated bright spots in the Transverse reflections in the interior of Try another eyepiece; use correct Koehler
image the microscope (often sickle or ring illumination
shaped)
Longitudinal reflections in the tube, Use lenses with antireflective coatings;
causing round light spots change the combination objective-
eyepiece
Unsharp bright spots in the image Contamination at a lens surface, on upper When localization and removal of the
or underside of the object, or air in contamination are not possible, reduce
immersion oil of the condenser (differ- the effect by opening the condenser
entiate as explained before) diaphragm
- somewhat more
- .

Reprinted from reference 10 with permission.

work almost equally well, price notwithstanding, especially work, and is durable. However, it is not the best for opti-
when narrow-band-pass (interference) filters are used. mum illumination, because the front and back of the glass
The objective best suited to the task in hand should be that covers the reflecting metal produce separate and over-
complemented by an ocular of commensurate quality. What lapping reflections of the light source or of the opening of
is needed for apochromatic oil-immersion objectives are the field diaphragm when the Koehler procedure is fol-
compensating eyepieces. They are designed to correct the lowed, thus adding undesirable scattered light to the final
lateral chromatic error of magnification inherent in objec- image. Instead, use a first-surface mirror, which lacks pro-
tive lenses of high corrections and high apertures; experi- tective coverings and gives but a single reflection.
ence amply proves the soundness of the additional advice of
Shillaber (18) that the compensating ocular should be of 1.2.5. Filters
high power, preferably 15X or 20 X . The ability of apochro- The light used for microscopy is modified in intensity or
matic objectives to take a high-power eyepiece should be wavelength by the use of filters. Neutral-density filters (alu-
fully utilized; otherwise, the fine detail that they are capa- minized optical flats) allow the brightness to be adjusted for
ble of bringing out in a photomicrograph is likely to be comfortable vision. Color filters restrict the wavelength
lost. used. Two kinds of color filters are available: expensive
For those who wear glasses with major corrections, it is interference filters of narrow band-pass consist of two half-
convenient and helpful to buy high-eyepoint oculars which silvered glass plates facing each other across a precisely
are made so that glasses may be worn while observing. measured gap; Wratten filters consist of colored gelatin
mounted between two 5-cm2 flats of optically polished
1.2.3. Condensers glass. Wratten filters deteriorate with time because air
Another important station in the path of the light from creeps in from the sides, but they come in a wide range of
lamp to object to eye is the substage condenser. There is no colors and densities and are valuable accessories. A red fil-
need for oiling the condenser in routine use that does not ter (Wratten no. 29) or an orange filter (Wratten no. 22) is
require the highest resolution; remember that objective helpful in making dense structures transparent or enhanc-
NAs of 1.30 to 1.40 will be fully utilized only if the con- ing structures stained with blue dyes. A green filter
denser NA is at least as high. Such condensers are usually (Wratten no. 11 or no. 58B) is the most generally useful be-
also achromatic or aplanatic, and their properties should cause it gives the kind of light to which the human retina is
match the optical properties of the objectives described maximally sensitive and because many commonly used his-
above. tological stains are red. Their contrast is greatly enhanced
Remember the role of the refractive index in the expres- when the light is green, the complementary color.
sion for NA: the full resolving power of an objective of high
N A is utilized only when the medium through which light 1.2.6. light Sources
passes between the condenser and the lens system of the ob- Optimum performance of a highly resolving objective de-
jective is homogeneous and glasslike all the way. Therefore, pends on optimum illumination. The field of view must be
the maximum resolution of an oil-immersion lens requires evenly filled with light of a brightness comfortable for view-
oil between the condenser and the object slide as well as be- ing or sufficient for photography. This requires a more com-
tween the slide and the objective lens. The condenser iris plex lamp than that effective for ordinary observation. The
diaphragm (aperture diaphragm) restricts the beam and light from the usual coiled filaments is unevenly distributed
thus affects the N A attained (see section 1.2.7). over their surface and therefore over their image unless dif-
fused by ground glass. The best structureless, uniformly
1.2.4. Mirrors bright sources of light are small mercury or zirconium arcs
A mirror is used to reflect the beam of light along the axis and the less expensive conventional projection lamps with
of the condenser. The adjustable mirror issued with old- tungsten bead or ribbon filaments that can be imaged to fill
style microscopes has a plane side for work with a condenser the condenser. The Koehler system of illumination is de-
and a concave side for work at low powers without a con- signed to provide the appropriate quality of light and re-
denser. The plane mirror is made by depositing metal on the quires that the source be focused in the plane of the object
underside of an optically flat disk, does well in everyday being viewed. Therefore, the microscope lamp should be
12 MORPHOLOGY AND ULTRASTRUCTURE

equipped with a lens system to project a sharp image of the Note that if the margins of the field diaphragm give an un-
light source into the entrance plane of the substage con- even color fringe when white light is used, the condenser
denser. In front of this lens, there should be an iris di- needs centration. To center it, turn the two adjustment
aphragm, the so-called field diaphragm (see below), which knobs to give movement toward the red side, centering the
can restrict the area of illumination without interfering aperture with the mirror, and stop when the color fringes
with the quality of the light. There should also be a carrier are symmetrical.
for filters. 8. Open the field diaphragm until it just impinges on the
margin of the field of view. This prevents illumination of
1.2.7. Optimum (Koehler) Illumination the specimen outside the field of view, which scatters light
The Koehler system of illumination is useful for high-reso- into the image plane. The field diaphragm does not affect
lution microscopy because it provides appropriate illumina- resolution in the area illuminated and can be left in view.
tion of the field and makes good use of high-quality optics. 9. Removing the ocular (which should be 12X or 15X),
It is achieved by focusing an image of the filament or light look down the tube and restrict the condenser aperture di-
source at the level of the condenser diaphragm (i.e., the aphragm to abolish glare or reflections. This restriction
lower focal plane of the condenser), when the condenser is should not be more than 1/10 the diameter of the fully illu-
in the correct position relative to the specimen. Under minated back lens; excess restriction lowers resolution.
these conditions, an objective lens in focus on the specimen 10. Replace the eyepiece, and insert filters appropriate to
will be fully illuminated whatever the size of the source. observation or photography (section 1.2.5).
Effectively, this applies to every part of a source, so that The same principles apply to adjustment of the illumi-
there is even illumination across the field of view even
nation provided by modern integral substage systems or to
when a coil filament lamp is used. The principle can be ap-
incident-light systems (3). However, the centration of the
plied usefully to both high-power dry and oil-immersion ob-
light for these systems is attained by adjusting the position
jectives.
of the bulb in its mount with centering screws. The filament
With an eyepiece, objective, and condenser of matching
image is focused by positioning the lamp forward or back-
optical qualities and with a first-surface mirror receiving fil-
ward in its sleeve, because the diaphragm and the lenses are
tered green light from a bright and homogeneous source, fixed in place. The point of reference for alignment is usu-
the research microscope is ready to be set up in the best way
ally an integral field diaphragm.
for work and needs only a rewarding specimen preparation
The same principles apply also to dry lenses, but oil im-
to show its worth.
mersion of the condenser is then unnecessary as long as the
The steps allowing achievement of Koehler (nearly opti-
condenser is of adequate quality.
mum) illumination for an oil-immersion objective are as
follows.
1. With a specimen slide in place, switch on the lamp 1.3. DARK-FIELD MICROSCOPY
and adjust the condenser to give the smallest bright spot on Dark-field microscopy provides a useful means of looking at
the specimen. This approximates the focal position of the wet mounts of unstained specimens and detecting very
condenser. small structures by reflected and diffracted light, revealed
2. Hold a small white card against the underside of the like motes of dust in a beam of sunlight or like planets and
condenser, and observing in the mirror, focus the image of moons in a night sky. It is performed by illuminating the
the lamp filament on the card and move the lamp so that specimen with a hollow cone of light such that only the
the image covers the opening of the condenser. Alter- light diffracted by objects in the field of view is transmitted
natively, focus the image on the closed condenser (aper- up the microscope tube to the eye or camera; the beams
ture) diaphragm. If the physical arrangement is awkward, forming the cone of light focused on the specimen are at too
use a small mirror for observation or approximate the focal low an angle to be captured by the objective. The result is a
position on a card placed over the mirror. field of bright objects (spirochetes, bacteria, particles, or-
3. Insert appropriate filters in the light path, and fully ganelles, etc.) against a dark background. It is an appropri-
open all diaphragms. ate technique for all powers of objectives, but it has some
4. Oil the condenser with 1 drop of high-viscosity oil on requirements that impose the need for specialized con-
the top lens, and raise it to meet a similar drop on the un- densers for use with oil-immersion objectives.
derside of the slide (see section 1.1.5). Low-power ( l o x ) and high-dry ( 4 5 x ) objectives can be
5. If necessary, scan the preparation with a low-power used in a dark-field mode by using an ordinary condenser
objective to identify a rewarding area, using reasonable cen- equipped with a central patch stop in its filter holder. This
tration and illumination (section 1.1.1), and mark it by the patch stop allows light paths only in the periphery of the
coordinates provided on many mechanical stages. condenser and so attains a hollow cone of light effective at
6. Apply oil to the top of the slide (see section 1.1.5 for a particular condenser position. This means that the N A of
precautions), change to the oil-immersion objective, and the condenser must be considerably greater than that of the
focus on the specimen. A fuzzy or moving image may indi- objective being used in order to attain an appropriate illu-
cate bubbles in the oil; check by looking down the tube mination angle and the condenser diaphragm must be com-
after removing the eyepiece. pletely open. The cone of light accepted by an objective
7. Adjust the field (iris) diaphragm of the lamp to a min- limits the usefulness of patch stops, so they do not work for
imum opening, and bring it into view with the mirror. Focus objectives with an N A greater than 0.60 to 0.65. A simple
the image margin with the substage condenser in the plane version of low-power dark-field microscopy with 10X and
of the specimen (i.e., so that the objective and condenser 4 5 x objectives can also be attained by using a phase-
are both in focus). Readjust the field diaphragm so that its contrast condenser (see section 1.4); it is useful but does not
image just impinges on the field of view and centers exactly provide the highest-quality image. Few problems are en-
with the mirror. The optical alignment is then correct. countered when dry objectives are used with a condenser
1. Light Microscopy 13

particularly designed for dark-field microscopy, and an ex- 2. Replace the condenser with the dark-field condenser,
cellent image can be expected. and apply 1 drop of high-viscosity oil to the top lens.
Oil-immersion objectives (which collect light at an 3. Make a wet mount of the specimen on a clean, thin
angle proportional to the N A ) require specialized con- slide. If the slide is very clean, it is helpful to make a small
densers of high NA, which must be oiled to the slide so that wax pencil mark under the coverslip for preliminary focus.
an effective cone of light can be produced. These reflecting 4. Raise the condenser to meet the specimen slide, and
condensers consist of either a paraboloid mirror with a cen- observe the ring of light. Focus this to give the smallest spot.
tral stop or, more effectively, a central spherical reflecting An asymmetrical ring of light above or below focus indi-
surface which reflects much of the light entering the con- cates that the condenser is not properly illuminated.
denser to a cardioid (curved) peripheral mirror on the 5. Focus the oileimmersion objective on the specimen.
perimeter, reflecting a very low-angled beam in the form of The objective should be equipped in the body with a funnel
a cone (refer to texts for optical diagrams). stop or with an iris diaphragm controlled by a knurled ring.
However, when these condensers are used, most oil- In the latter case, start with the diaphragm half closed and
immersion objectives do not provide an adequately dark make final adjustments later for maximum darkness of the
ground without the presence of devices to restrict scatter field.
from the margins and mountings of their lenses. Excellence 6. Move the specimen slowly to a useful field, and make
requires an objective with an adjustable diaphragm in the final adjustments in the condenser focus for maximum
back focal plane or restriction in the back focal plane by use brightness of the reflections from the specimen.
of a funnel stop (a metal tube with a hole in the lower end)
that fits inside the objective to block transmitted rays scat- 1.4. PHASE-CONTRAST MICROSCOPY
tered from the periphery of the objective.
Phase-contrast microscopy is a system for gaining contrast
Intensive light is necessary to show very thin or very
in a translucent specimen without the help of stains (4, 10,
small objects. Sunlight from a heliostat has been used to
17, 22) and has the advantage of using high-resolution op-
show bacterial flagella in action.
tical components. It requires a specialized condenser and
It is sometimes difficult to get a good dark background
specialized objectives with appropriate annular modifica-
when there is poor centration of the condenser relative to
tions to the lens systems.
the objective. This requires some trial and error to gain
Stained specimens or biological material with opaque
symmetrical illumination. It is helpful to find the best set-
areas form images in a microscope because the various com-
ting for the condenser by using tongue or cheek scrapings
ponents transmit different amounts of light, so that ampli-
mounted in water under a coverslip; these large epithelial
tude and wavelength are modified by absorption and scat-
cells are easy to find. All the precautions about oiling the
tering in the specimen. Most cells and their components do
condenser and slide (section 1.1.5) must be followed. But
not cause enough amplitude modification to give useful
given a well-aligned microscope, all that matters then is the
contrast in the image. However, their materials are translu-
intensity, geometry, and quality of the cone of light with an
cent and have different refractive indices from neighboring
appropriate objective.
structures and from the mounting medium, and these differ-
The focus of the condenser is crucial in dark-field mi-
ences cause different degrees of phase retardation in the
croscopy. The apex of the cone of light must be at the level
light beam passing through a structure compared with
of the specimen, which is also the plane of focus of the ob-
phases of beams that have passed through other material or
jective. Above this focal point is a dark conical space, in
only through the mounting medium. The purpose of the
which the front lens of the objective is placed, symmetrical
phase microscope is to take advantage of changes in phase,
to the dark conical space formed below the specimen above
converting these into amplitude differences to form an
the condenser. When the dark-field condenser is brought up
image with enhanced contrast.
to the slide, with immersion oil making a complete contact,
A n instrument that is permanently set up for phase-
a bright ring of light is projected onto the specimen. As the
contrast microscopy is preferable to one that provides alter-
condenser is raised farther, the light comes to a bright in-
nating service with ordinary optics on the same stand.
tense spot, which must be close to the best focus. Because
Phase-contrast objectives are not suitable for the best
the large-aperture condensers for oil immersion microscopy
bright-field work, despite their high quality. It is better to
have a very short focal length, it is important to use a thin
have objectives dedicated to each purpose. Phase-contrast
slide (-0.8 mm thick) so that the condenser can focus
microscopy demands much more light than is adequate for
through it on the specimen. Standard slides (-1.2 mm)
the examination of stained specimens with ordinary optics.
may be too thick, thus making high-resolution dark-field
microscopy impossible.
A green filter should be used to reduce unavoidable chro-
matic aberration.
For many years, dark-field examination of exudates and
One of the advantages of phase-contrast microscopy
media was the accepted method for demonstrating the pres-
over the less-available dark-field microscopy is that the for-
ence of spirochetes. It is impressive to be able to see
mer uses the full aperture of the objective lens. Therefore,
Leptospira spp. in low-power dark-ground images by using a the best of phase-contrast objectives should be used. This
lox objective and 15 x oculars, but it is even more so when in turn implies the use of a substage condenser with a suit-
they appear in all their glory under oil immersion at an ef-
ably large aperture and of 12X to 15X compensating eye-
fective NA of 1.3. It is a pity that these beautiful images are
pieces, which fully utilize the fine resolution that the ob-
seldom seen nowadays because of the convenience of phase-
jectives are capable of giving. Medium-power (40X or
contrast microscopy.
60X ) oil-immersion phase-contrast objectives are available
A procedure for oil-immersion dark-field microscopy is and provide crisp, rewarding images for work with large
as follows.
cells. For the very best results, the condenser and slide must
1. See that the microscope and illumination system are be connected by oil when high-power (9OX or 1OOX)
fully aligned, as for bright-field microscopy. objectives with NAs higher than 1.0 are used. However,
14 MORPHOLOGY AND ULTRASTRUCTURE

an oiled condenser is not required for all applications. 1.4.2. Specimen Preparation
A dry condenser is adequate for observing the shapes The specimens for phase-contrast microscopy should be as
and forms of bacteria and their spores and for checking thin as possible and must be mounted in a fluid or gel and
motility. under a coverslip to give a homogeneous background to the
Phase-contrast microscopy works because a phase annu- images. For general work, a clean standard slide and cover-
lus is inserted at the lower focal plane of the condenser (to slip are satisfactory.
generate a cone of light) and because a phase plate is incor- When intracellular detail of living organisms is wanted,
porated in the back focal plane of the objective. An image attention must be paid to the refractive index of the
of the annulus will thus be formed on the phase plate. Some medium in which the cells are mounted. This is most easily
light beams are diffracted by the specimen (e.g., a living provided by dissolving gelatin or bovine serum albumin (15
cell) to go through all of the objective field, and some go di- to 30%) in the medium. According to the concentration
rectly through the specimen and the phase plate ring; both used, this equalizes or brings closer together the refractive
take part in forming an image. The phase ring in the objec- indices of the contents and medium, since phase retardation
tive is a flat, ring-form groove in an optical flat plate; its size is proportional to the refractive index multiplied by the
fits the cone of the direct light beams generated by the con- light path. Useful preparations can be made by mounting a
denser annulus. The groove is formed so that the phase ring water suspension on a slide that has a thin layer of agar
is less-thick glass, causing at least a 0.25-nm-wavelength dried on it and then applying a coverslip. The disturbing
phase retardation compared with the phase of a direct bright halos that usually surround dark-contrast images are
(nondiffracted) beam. The phase difference of the dif- then reduced or abolished, and the cells are more gray than
fracted and nondiffracted beams, some of which are also re- black. As an additional benefit, the cells then show internal
tarded by passing through a structure in the specimen, structures that are not easily discerned in life, e.g., nucleoids
causes either destructive interference (dark contrast) or and granules other than the obvious lipid droplets, or de-
constructive interference (bright contrast) when the beams veloping endospores. When the mounting fluid and an
form the image. Therefore, it is essential to center the an- adjacent structure are identical in refractive index, the
nulus plate in the condenser with respect to the analyzer structure will disappear; this technique of immersion refrac-
plate in the back focal plane of the objective. This is tometry can be used to measure the densities and masses of
achieved both by imaging with a focusing eyepiece (or tel- structures (15).
escope) and by centering the visible rings by using the two
adjusting knobs on the sides of the condenser. The bright 1.4.3. Phase Condenser for low-Power Dark Field
image of the annulus should be completely enclosed by the The condenser phase plate is, essentially, a form of patch
gray annulus of the phase plate when centration and con- stop, and so, given suitable geometry, it should produce
denser positions are correct; complete filling of the annulus dark-field images with ordinary low-power objectives. The
is not essential. annulus for the oil immersion phase commonly gives a rea-
sonable semblance of dark-field optics (section 1.3) with an
1.4.1. System Assembly ordinary lox objective and sometimes with a 4 5 x objec-
1. Set up the microscope as for Koehler illumination tive, after a little fiddling with centration and the position
(section 1.2.7) without the annulus in place (use the bright- of the condenser.
field position for the usual rotatable carrier in the condenser
of a phase microscope), and adjust the phase objective to
focus on a specimen. 1.5. INTERFERENCE MICROSCOPY
2. Revolve the condenser carrier to bring the appropri- Interference microscopes have definite applications in bac-
ate annulus into position below the condenser for the oh- teriology for discerning the structures of cells. Such micro-
jective in use. scopes are expensive, which makes them less available, and
3. Insert a focusing telescope instead of the eyepiece and their operation and the interpretation of the images ob-
focus on the gray ring image of the phase plate, which has tained are best learned by practice with experts. Like
distinct inner and outer edges. phase-contrast microscopes, they attain contrast in images
4.Look in the telescope for all or part of a very bright of translucent specimens by detecting phase changes in-
ring of light, the image of the condenser annulus, which has duced in light that traverses cell components with different
to be brought into register with the objective phase ring, masses and refractive indices. The two systems differ in
seen as faint rings for each side of the groove in the phase that the interference microscope develops separate object
plate. Achieve centering either by manipulating the con- and reference beams, rather than forming an image from
denser centering knobs or by nudging the annulus with a the direct and diffracted elements of a single beam from a
knurled ring on the carrier (systems vary according to man- condenser annulus as in the phase-contrast microscope
ufacturer). (10, 17, 20, 22).
5. Replace the telescope with the eyepiece, adjust the Interference microscopes provide superior images of the
fine focus, and regulate the light intensity (by using a trans- internal morphological details of cells without the halos that
former) for comfortable viewing. The image should not shift surround cells when viewed by phase-contrast microscopy.
asymmetrically on focusing; if it does, recheck step 4. A great advantage of the former is that phase changes may
6. If the image is unsatisfactory, check that all surfaces be measured to provide quantitative cytological data.
are clean, that the specimen is not too thick, that there Furthermore, the lack of an annulus avoids deterioration of
are no bubbles in the oil if you are using oil immersion, the image by optical effects from cell structures. The lenses
and that the correct annulus is in place. The image qual- are of high quality, are used to their effective NAs, and will
ity deteriorates if any part of the direct beam falls outside operate in either transmitted- or epi-illumination modes.
of the annulus in the objective phase plate or is not fully The quality of light and the adjustment of the microscope
concentric. Accurate focusing of the condenser is essen- to give critical or Koehler illumination at the outset are
tial. important.
1. Light Microscopy 15

The Nomarski-type interference microscope uses polar- substage condenser; and (iii) incident illumination (epi-
ized light (white or monochromatic) from a filter at the illumination) through a specialized objective, in which case
source to fill the condenser through a Wollaston (birefrin- the objective acts as both condenser and objective. The sec-
gent) prism. The prism generates two polarized beams at ond and third systems have the advantage of an effective
right angles, each filling the lenses. The resulting cone of dark-field system, because the optical path is such that a di-
light traversing the specimen and illuminating the objec- rect beam does not go to the eye and there is minimal in-
tive is a complex of these two beams; in effect, they are very terference with the detection of the weaker fluorescent
close together, in parallel, and uniform. They act as object light from the specimen. In the third system, which is the
and reference beams according to what they traverse in the best, the objective acts as the condenser and suffers no cen-
specimen. The beams are recombined above the objective tration problem and the weak fluorescence suffers minimum
by another Wollaston prism, allowing interference (con- attenuation from specimen thickness. The exciter beam is
structive or destructive) to take place. The plane-polarized reflected down to the objective from a side or rear port in
beam is recaptured by an analyzer polarizing filter, which the microscope tube by a beam-splitting mirror or prism
is usually in a fixed orientation (this means that the polar- that reflects the exciting wavelength but transmits visible
izing filter at the source has to be rotated to give the appro- light back from the objective to the eye.
priate orientation). If white light is used, with appropriate The manner of using the microscope and its filters may
manipulation parts of the image will contain interference be unique to the instrument, so consult the manual pro-
colors which are related to the amount and sign of the phase vided by the microscope manufacturer.
change. This information and photometric measurements
will allow generation of mass data. 1.6.2. Confocal Scanning Microscopy
In some interference microscope systems (including the The elaborate equipment (2, 19) required for confocal scan-
Nomarski type), the image has a pseudo-three-dimensional ning microscopy, utilizing an intense beam of light from a
appearance, which is a consequence of an angle of shear be- laser, is designed to scan the sample by illuminating and im-
tween the beams. aging one very small area at a time in a single focal plane of
the specimen. It is confocal because the scanning and the
image are both attained through the objective. An epi-
1.6. FLUORESCENCE MICROSCOPY illumination system focuses a small spot of light at a plane
Fluorescence microscopy employs all the principles of optics in the specimen. This illuminated spot is imaged through a
described for the research microscope (section 1.2). The conjugate aperture, which accepts only the direct beams
differences in practice and design relate to generation and (but not the diffracted beams) for forming the image. The
transmission of wavelengths of light suitable to the excita- specimen is scanned through the objective by a moving
tion of fluorochrome stains and of natural fluorescence beam producing a series of spots of light or aperture images.
in the specimen. The secondary emitted wavelengths are A raster scan allows the synthesis of a complete image in a
detected as an image of a fluorescing object. Because the detector system, and the image is displayed on a monitor.
excitation process usually requires short wavelengths in the Only structure that is in focus will form an image. This im-
near-UV or blue range, the lamp (a high-pressure mercury aging system will operate either with direct imaging by vis-
vapor arc lamp) and any lens between the lamp and object ible light or with fluorescence imaging by UV light.
must be made of material (quartz) appropriate for the pas- There are two major forms of the scanning equipment.
sage of that range of wavelengths. A first-surface mirror is One form involves tandem multiple-aperture arrays, and
essential (to avoid an interfering glass layer). The immer- the other involves a regulated movement of a very fine laser
sion oil for the objective and condenser must be nonfluo- beam. The principle is straightforward, but the equipment is
rescent (e.g., a special synthetic formulation or sandalwood very specialized and requires its own instruction manuals
oil). Quartz slides and coverslips must be used. Good advice and expert users.
on details comes from specific (11, 23) and general texts. The advantages of this method of microscopy are that
Most important are the light source and the arrange- it can be used at high magnification with the best of epi-
ment of filters in the light path, which vary in mechanical illumination objectives (including oil-immersion) to study
arrangement among manufacturers (who provide appropri- large objects that scatter a lot of light in other modes of mi-
ate sets of filters for specific fluorochromes). The light must croscopy. It can focus on the structure of a surface and, par-
include wavelengths appropriate to the fluorochrome being ticularly, can allow examination of structure within large
used, and high-pressure mercury vapor arc lamps have the cells. As far as bacteria are concerned, a major use is the study
most inclusive spectral range. Before entering the lens train, of the interactions of bacteria with or within eukaryotic cells.
the beam must be filtered to minimize heat transfers (i.e., Information is increased by using confocal scanning mi-
remove the longer wavelengths) and then passed through croscopy as a form of fluorescence microscopy; photomulti-
another filter to isolate the frequency appropriate to acti- plier imaging reduces the problems with dim signals, and
vation of the fluorochrome. When the beam has passed the confocal system evades fluorescence interference from
through the objective, the light is still UV, so a barrier filter features above and below the plane of focus. Because images
to stop the passage of eye-damaging frequencies is essential. can be generated at known depths in the specimen, a com-
This filter in the tube must allow the passage of the longer puter correlation of a stacked series of images generates
wavelengths arising from the excited fluorochrome. The three-dimensional information about the specimen.
choice of the latter may be governed by requirements of
multiple colors when mixed probes are used.
1.7. CELL MEASUREMENT
1.6.1. Fluorescence Systems It is important to determine the magnification imposed on
The illumination systems for fluorescence microscopy take bacterial cells either in projections for drawings or on pho-
several forms: (i) transmitted light through a substage con- tomicrographs. Equally important, a range of dimensions
denser; (ii) dark-field illumination through a specialized may be needed as part of experiments or for the description
16 m MORPHOLOGY AND ULTRASTRUCTURE

of cells for purposes of classification. Although a number of technical requirements are important, in all cases, so that
measuring techniques may be applied, they all must be the most faithful record may be obtained. There are excel-
based on a measurement standard provided by a stage mi- lent books available that amplify and explain the require-
crometer; the use of the stage micrometer is clearly de- ments (3,4,6,8, 15, 18,21). Chapters 3 through 5 provide
scribed in texts (12, 18). information on digital imaging and image processing by
computers.
1.7.1. Stage Micrometer Four types of photomicrography apparatuses are in gen-
A stage micrometer is a slide on which a number of lines eral use, as follows.
have been ruled by a grating engine or deposited photo-
graphically to show a precise scale. Usually, there are 10
1. A roll-film camera back and shutter (usually for 35-
mm film), together with an integral device that includes the
parallel lines at 0.1-mm (100 pm) spacing and 10 parallel
ocular and a beam-splitting prism with a side viewing arm
lines at 0.01-mm (10 pm) spacing. Image the micrometer
to allow focusing, form the working unit. The whole device
slide with the same care given to a specimen. Project the
rests on the microscope tube.
image onto the ground glass of a photomicrographic camera
2. Integral cameras have been developed by a number
for direct measurement, or record the image on film under
of microscope makers and come complete with an ex-
the same conditions used for photomicrographs. A number
posure meter and timing controls. These cameras are usu-
of measurements of the appropriate intervals are then used
ally adequate and produce effective routine photomicro-
to generate either the magnification on the film, the length
graphs.
required for a bar scale to apply to micrographs, or a set of
3. Digital and video cameras are available and used ef-
measurements of the cells under study.
fectively for recording images. Both allow for storage and
1.7.2. Eyepiece Craticule Micrometer can provide images for projection, computer archiving, or
A n eyepiece graticule micrometer is an optically flat glass image processing.
disk, about 12 mm in diameter, with an arbitrary but accu- 4. A n old-fashioned light-tight bellows is sometimes
rately proportioned scale engraved on it. The scale is usually used. It is located on a stand on which a microscope can
in numbered units, each with 10 divisions. The graticule also be placed. A light-excluding sleeve is fitted at the bot-
disk is placed in the plane of the intermediate image inside tom of the bellows and meshes with its mate, which slips
a Huygenian eyepiece, which has a diaphragm at the plane onto the top of the microscope tube. A plate carrier is fitted
of the intermediate image where the graticule disk can be at the top of the bellows and can be made for either cut film
rested (access is obtained by unscrewing the top element of or instant (Polaroid) negative film.
the eyepiece). In more complex eyepieces, the diaphragm is In all cases the success of photomicrography and the re-
below the lower field lens, and so there are fewer problems sulting photographic print depends on the following.
with parallax. 1. A first-class specimen preparation, appropriately
Each graticule must be calibrated for each eyepiece- mounted and exactly focused.
objective combination to be used. With a stage micrometer 2. Good optics, properly aligned and optimally
and an appropriate set of ruled lines in focus, rotate the eye- (Koehler) illuminated (section 1.2.7) to give the best possi-
piece so that the graticule scale is aligned to the rulings and ble image, and a suitable choice of color filters (section
note a number of coincidence points. Then it is easy to de- 1.2.5).
rive a statistical measurement of the graticule unit in mi- 3. A camera-microscope assembly that is free from vi-
crometers to two decimal places. Final direct measurements bration.
against the real specimen should not be considered more 4. Appropriate photographic materials and a processing
precise than one decimal place. technique appropriate to giving optimum grain size with ad-
There are graticules ruled in squares for counting of cells equate contrast and gray scale.
(see chapter 2.1.4) or in circles for rough sizing of cells. 5. A n appropriate final magnification, attained by print-
When counting, relate the squares to the specimen area and ing with an enlarger.
count the cells lying within the squares and touching two of
the sides. In this application, it is advantageous to have a The camera-microscope assembly can present some
special eyepiece allowing focus on the grid rulings by rota- problems, because fixing photographic devices on the mi-
tion of the top lens. croscope tube tends to allow the transmission of vibrations.
Partly, this is because pressing the shutter release removes
1.7.3. Eyepiece Screw Micrometer the beam splitter before activating the shutter, and each ac-
The eyepiece screw micrometer is a specialized eyepiece tion can generate persisting vibrations. When a shutter in-
(sometimes called a filar micrometer) for direct measure- tegral to the camera is used, it should be of the iris di-
ments after calibration against the rulings of a stage mi- aphragm type, and a focal-plane shutter must be avoided.
crometer. The units are on a wheel (with or without a Always use a cable release to actuate the shutter. Shutters
vernier reading scale) to be read against a fixed mark, and for the timing of exposures are best put in the light path be-
the wheel drives a hairline across the field, allowing a tween lamp and microscope. Vibration is no problem with
measurement in calibrated units by difference. As with an the bellows cameras as long as the light-tight collar around
eyepiece graticule micrometer, the cursor line and the ob- the eyepiece allows the camera and the microscope to be
ject must be in exact focus to minimize distortion by par- independent and not to touch each other. Vibrations must
allax. be kept to a minimum; a sturdy, heavy table that is not at-
tached to a wall is a help.
Film for black-and-white photomicrography should be a
1.8. PHOTOMICROGRAPHY panchromatic film of ASA 60 to 100 for general purposes
All types of light microscopy can make exact records on (to keep the exposures at a manageable level). For phase-
film, whatever the wavelength of light being used. The contrast photomicrography, a faster film (ASA 300 to 400)
1. Light Microscopy 1 17

is needed because the light levels are much lower. A n ex- An example of the booklets produced by manufacturers in-
posure meter allows repetition of values established by test- volved in aspects of microscopy. These booklets ure obuinable
ing. If an old bellows-type photomicrographic camera is from such firms and their agents in updated versions and are
available, acquire it because it is the best apparatus for the generally excellent.
highest-quality work, even though it is not the most con- 8. Engle, C. E. (ed.). 1968. Photography for the Scientist.
venient to use. Academic Press, Inc., New York, NY.
When exposing negatives and printing micrographs, General aspects of scientific photography, including photomi-
think about the final magnification and the detail to be crography.
shown. Use film processing that enhances contrast with rea- 9. Hartley, W. G. 1993. The Light Microscope-Its Use and
Development. Senecio Publishing Co., Oxford, United
sonably fine grain, as specified by the manufacturer. Grain Kingdom.
in a developed negative dictates that you should not enlarge 10. James, J. 1976. Light Microscopic Techniques in Biology and
the film more than 2 to 2.5 times in printing. Good mi- Medicine. Martinus Nijhoff Medical Division, Amsterdam,
croscopy of most ordinary bacteria allows a total magnifica- The Netherlands.
tion of up to x1,500, which is needed for fine detail and A fine book on theory and practice, with emphasis on the latter.
can be attained on the film (see section 1.6.1 for the deter- It provides good advice on special and advanced techniques, in-
mination of magnification). The larger images taken on a cluding phase-contrast, interference, dark-field, polarization,
bigger area (4 by 5 in. [-lo by 13 cm]) using cut film in an and fluorescence microscopy.
old-fashioned camera, for which enlargements of 1.25 to 1.5 11. McKinney, R. M., and W. B. Cherry. I985 Immun-
times are usually sufficient, are better than higher enlarge- ofluorescence microscopy, p. 891-897. In E. H. Lennette,
ments of much smaller images on 35-mm film. If the micro- A. Balows, W. J. Hausler, and H. J. Shadomy (ed.), Manual
graphs are not as sharp and as good as they appeared in the of Clinical Microbiology, 4th ed. American Society for
properly adjusted microscope, try again. Focusing is critical, Microbiology, Washington, DC.
whatever the camera used. The trick with bellows cameras 12. Mollring, F. K. 1981. Microscopy from the Very Beginning.
is to use a plain glass insert and to focus on the image ob- Carl Zeiss, Oberkochen, Germany.
tained with a focusing magnifier adjusted to focus on a mark Another excellent booklet produced by a manufacturer.
on the inside of the glass. The traditional ground glass and 13. Pluta, M. 1988 to 1993. Advanced Light Microscopy, Vol.
black hood can still be used effectively. 1-3. Polish Scientific Publisher/Elsevier, Amsterdam, The
Prints should be enlarged to a format that allows easy Netherlands.
visibility of the important detail, with the object occupying 14. Quesnel, L. B. 1971. Microscopy and micrometry, p. 1-
103. In J. R. Norris and D. W. Ribbons (ed.), Methods in
most of the frame. Microbiology, vol. 5A. Academic Press Ltd., London,
United Kingdom.
1.9. REFERENCES 15. Quesnel, L. B. 1972. Photomicrography and macropho-
tography, p. 276-358. In J. R. Norris and D. W. Ribbons
Books on light microscopy and photomicrography, ranging (ed.). Methods in Microbiolom. vol. 7B. Academic Press
from encyclopedias to paperbacks, are readily available. Ltd.; 'London, United Kingdrm.
However, books can give only basic principles. Satisfying Quesnel's two articles are useful resources for optical details
microscopy is best learned through direct instruction and and practical advice on microscopy and photomicrography for
practical experience, not a small part of which is the art of microbiology in particular.
preparing worthwhile specimens. No single book is directed 16. Richardson, J. H. 1991. Handbook for the Light Microscope.
toward microscopy for bacteriologists, but the principles are Noyes Publications, Park Ridge, NJ.
the same for all users. The list presented below is represen- 17. Ross, K. F. A. 1967. Phase Contrast and Interference
tative of the books available. Microscopy for Cell Biologists. Edward Arnold, London,
United Kingdom.
1. Barer, R. 1968. Lecture Notes on the Use of the Microscope. An excellent explanation of the theory and practice of phase-
Blackwell Scientific Publications, Oxford, United contrast and interference microscopy. Also practical discussion
Kingdom. of photographic techniques applied to microscopy.
The advice of a master microscopist. 18. Shillaber, C. P. 1944. Photomicrography in Theory and
2. Boyde, A. 1990. Confocal optical microscopy, p. 185-204. Practice. John Wiley &a Sons, Inc., New York, NY.
In P. J. Duke and A. G. Michette (ed.), Modern Nothing is likely to replace this classic text, which deals exhaus-
Microscopies. Plenum Press, New York, NY. tively but readably with the properties of objective lenses, ocu-
A description of equipment and applications. lars, and condensers. It sets out the prmtice of good illumina-
3 . Bradbury, S., and B. Bracegirdle. 1998. Introduction to tion, weighs the advantages of different mounting media, and
Light Microscopy. Springer-Verlag,New York, NY. deals with both theoretical and practical bench microscopy;
4. Bradbury, S., and P. Everett. 1996. Contrast Techniques in however, it antedates phase microscopy.
Light Microscopy. Bios Scientific Publishers, Oxford, 19. Shuman, H., J. M. Murray, and C. Di Lullo. 1989.
United Kingdom. Confocal microscopy: an overview. BioTechniques 7: 154-
Short handbook with simple explanations of lens systems. 613.
5. Cargille, J. I. 1975. Immersion Oil and the Microscope. This review includes an example of three-dimensional recon-
Technical reprint 10-1051. R. P. Cargille Laboratories, struction.
Cedar Grove, NJ. 20. Slayter, E. M. 1970. Optical Methods in Biology. John
Another excellent booklet produced by a manufacturer. Wiley & Sons, Inc., New York, NY.
6. Culling, C. F. A. 1974. Modern Microscopy-Elementary A source book for the theoretical bases of most forms of mi-
Theory and Practice. Butterworth & Co., London, United croscopy and for analytical processes including diffraction,
Kingdom. spectroscopy, and related optical techniques. It is concerned
Another short paperback book. with principles and not practice.
7. Delly, J. G. 1998. Photography through the Microscope. 21. Smith, R. F. 1990. Microscopy and Photomicrography-u
Eastman Kodak Co., Rochester, NY. Working Manual. CRC Press, Inc., Boca Raton, FL.
18 MORPHOLOGY AND ULTRASTRUCTURE

A professionally illustrated procedural manual with minimal 23. Wang, Y.-L., and D. L. Taylor (ed.). 1989. Fluorescence
theory. Useful for a beginner with no experience. Microscopy of Living Cells in Culture, part A. Fluorescent
22. Spencer, M. 1982. Fundamentals of Light Microscopy. Analogs, Labelling Cells, and Basic Microscopy. Academic
Cambridge University Press, Cambridge, United Kingdom. Press, Inc., New York, NY.
A useful general surwey. A volume with helpful technical advice.
i
.
.

Sampling and Staining for Light Microscopy


TERRY J . BEVERIDGE. JOHN R . LAWRENCE. AND ROBERT G . E. MURRAY

2.1. SAMPLING ............................................... 20


2.1.1. Air ................................................ 20
.2. Water .............................................. 20
.3. Soil ............................................... 21
.4. Direct Total-Cell Counting ............................... 21
.5. Direct Viable-Cell Counting .............................. 21
. .............................
6. Detection of Specific Bacteria 22
2.2. PREPARATION ........................................... 22
2.2.1. Living-Cell Suspensions ................................. 22
2.2.1.1. Wet Mounts ................................... 22
2.2.2. Negative Staining ..................................... 23
.3. Fixation of Smears and Suspensions ........................ 23
2.2.3.1. Heat Fixation .................................. 23
.2. Chemical Fixation............................... 23
2.2.4. Simple Staining ....................................... 23
.5. Permanent Mounts .................................... 24
2.3. CHARACTERIZATION ..................................... 24
2.3.1. Size ............................................... 24
.2. Shape .............................................. 24
.3. Mode of Division ...................................... 25
.4. Motility ............................................ 25
.5. Gram Staining ........................................ 25
2.3.5.1. Hucker Staining Method ......................... 26
.2. Burke Staining Method .......................... 26
.3. LIVE BacLight@and ViaGram@Methods ............. 26
2.3.6. Acid-Fast Staining ..................................... 27
2.3.6.1. Ziehl-Neelsen Staining Method ..................... 27
.2. Truant Staining Method .......................... 27
2.3.7. Endospores .......................................... 28
2.3.7.1. Popping Test .................................. 28
.2. Negative Staining .............................. 28
.3. Schaeffer-Fulton Staining Method................... 28
.4. Dorner Staining Method .......................... 28
2.3.8. Cysts .............................................. 28
.9. Capsules and Slime Layers ............................... 28
2.3.9.1. Duguid Negative Staining Method .................. 29
2.3.10. Flagella ............................................. 29
2.3.10.1. Leifson Staining Method ......................... 29
2.3.11. Cytoplasmic Inclusions ................................. 29
2.3.1 1.1. Staining of Poly-P-Hydroxyalkoanate................ 29
.2. Staining of Polyphosphate (Metachromatic or Volutin
Granules).................................... 29
.3. Periodate-Schiff Staining of Glycogen-Like
................................
Polysaccharides 29
2.3.12. Nucleoids ........................................... 30
2.4. SPECIAL TECHNIQUES .................................... 30
2.4.1. Spirochetes .......................................... 30
.2. Mycoplasmas ......................................... 30
.3. Rickettsiae .......................................... 31
2.4.3.1. Giemsa Staining Method ......................... 31
. 2. Gimenez Staining Method ........................ 31
2.4.4. Legionella ........................................... 31
2.5. SOURCES OF STAINING REAGENTS .......................... 32
.6. REFERENCES ............................................. 32
2.6.1. General References .................................... 32
.2. Specific References .................................... 33

19
20 1 MORPHOLOGY AND ULTRASTRUCTURE

Specific morphological details are required to characterize a centrations of bacteria. Better sites for productive
microorganism; these are usually determined by means of growth may be provided by interfaces on suspended
light microscopy, but some require more sophisticated tech- solids, surfaces of rocks, sands, sediments, and the sur-
niques such as laser scanning microscopy (chapter 3) or faces of water plants; these sites adsorb and provide nu-
electron microscopy (chapter 4). Some of the light mi- trients, making adhesion to surfaces advantageous and
croscopy methods employed are time honored and trace sometimes leading to the formation of biofilms.
their genesis to the early days of bacteriological science. Certain environments may support too many bacteria
This chapter is based on and is an updated version of a per unit of mass and must be diluted for examination or
chapter in the earlier edition of this book, Methods for cultivation. Sewage sludge contains an astonishing array
General and Molecular Bacteriology (12), to which R. G. E. of microorganisms intermixed with particulate materials.
Murray, R. A. Doetsch, and C. E Robinow were major con- Certain soils and marine muds contain only a few bacte-
tributors; we recommend that you read the former chapter. ria mixed with much opaque colloidal matter, some of
Here, we have concentrated on the general means of char- which may be hard to distinguish from bacterial forms.
acterizing bacteria by light microscopy because it includes Most bacteria constituting the natural flora of an envi-
the cytological approach to making the best of preparations ronment do not reveal particularly distinctive morpho-
for study and photomicrography. The methods are meticu- logical features, such as the star-shaped Prostheco-
lously described in section 2.2.7, Cytological Prepara- microbium species, sheaths of Sphuerotilus or Leptothrix
tions, of the previous edition of this book. spp., and trichomes of Curyophunon spp.
The first approach to the study of natural populations is
morphological classification, an assessment of relative 2.1 . I . Air
numbers, and formation of an idea of the complexity of the
bacterial community in advance of any attempt at cultiva- The field of aerobiology encompasses both indoor and out-
tion. More specialized methods estimate the proportions of door components and focuses on a wide range of microor-
growing and nongrowing cells and the productivity of the ganisms, including pathogenic and nonpathogenic vari-
populations (8, 13). Despite the problems in choice of eties. Although the numbers in a unit of volume may be
method and accuracy of results (3), light microscopy pro- quite small, the simplest approach to air sampling is the use
vides most of the basic biological information about an of open petri dishes containing a suitable nutrient agar near
ecosystem. a suspected contamination source and is applicable when
The establishment and maintenance of pure cultures the concentration of organisms is relatively high. When
(and even the struggle to maintain impure cultures of diffi- large volumes of air have to be sampled and the organisms
cult bacteria) require control by microscopy. Frequently, have to be characterized, bubble a defined volume (use a
contamination plagues the cultures maintained by those flowmeter in the system) through a flask of fluid medium,
less practiced in bacteriology, so that regular microscopy and then subject it to plate counting (7). Impaction and
and cultural control must be encouraged. The identifica- impingement samplers are also commercially available for
tion or description of bacteria inevitably requires the ap- sampling airborne organisms (26).
plication of determinative staining methods, including Filter sampling methods for bacteria (2) are derived from
those suitable for recognition of shape, special structures, those used to sample particulates of industrial or public
behavior, or life cycles, as well as those for the measure- health importance; commercial filtering units are readily
ment of cells. Digital image analysis techniques may also be available. The pore size used for bacteria is usually 0.22 to
applied to extract information regarding cell morphotypes, 0.45 pm. Bacterial spores and vegetative cells are both
numbers, and biovolumes (9). The information gleaned counted (section 2.1.4) either by performing direct micro-
from these techniques may be useful, but it is important to scopic counts or by depositing a plate-sized (90-mm-diameter)
understand the methodological limitations because any filter on a nutrient surface for CFU counting after incuba-
laboratory manipulation may introduce some alteration of tion. However, many vegetative bacterial cells die rapidly
form and structure, albeit small in most cases. Despite on an air filter because of excessive desiccation, and viable-
shortcomings, microscopic observations of bacteria are cell counting can be inaccurate. It is also possible to do
necessary, but usually not sufficient, factors for identifying direct counting of bacteria on membrane filters by using
them. Nevertheless, errors in identification are often trace- epifluorescence microscopy (section 2.1.4).
able to mistakes in judging the shape, Gram reaction, and
motility of a new isolate. 2.1.2. Water
Since bacteria have optical properties similar to those of
water and give minimal contrast with the use of ordinary
2.1. SAMPLING transmission light microscopy, examine specimens by using
phase-contrast or dark-field microscopy. Epifluorescence
It is difficult to observe living bacteria directly in natural microscopy used with fluorescent nucleic acid stains (acri-
habitats, and information so obtained may be meager.
dine orange, DAPI [4, 6-diamidino-2-phenylindole], etc.)
There are several reasons for this.
is also an efficient method for detection of bacteria (see
Not all environments contain large populations of bac- chapter 3). Direct microscopic examination of water sam-
teria per unit of mass. Organisms in marine and lake wa- ples generally reveals few bacteria (5, 11). Use filtration
ters, for example, usually must be concentrated by filtra- techniques to concentrate cells for estimation of the total
tion or centrifugation to obtain sufficient numbers for number per unit of volume, as well as for direct visualization
study. Waters are often low in nutrients, and minimal of different morphological types. Centrifuging water sam-
steady state and division often lead to extremely small ples from most sources leads to a remarkably large gelati-
cells (ultramicrocells). The presence of flocs or aggre- nous pellet, which includes polymers of diverse origins, en-
gates (ten to several hundred micometers in diameter) in trapped particulates, and a diversity of microorganisms. Wet
most aquatic environments leads to localized high con- mounts of the pellets examined by phase-contrast mi-
2. Sampling and Staining for Light Microscopy 21

croscopy or preparations stained with simple basic dyes or 2.1.4. Direct Total-Cell Counting
fluorescent stains reveal the biota for preliminary morpho- Direct counts can be made on bacterium-retaining filters ei-
logical classification (section 2.2.1). ther by the use of epifluorescence microscopy after appro-
As with air sampling (see above), membrane filters pro- priate staining of the cells on a portion of the filter mem-
vide a rapid and effective means of sampling from fluids for brane or by the use of phase microscopy on a clarified
direct microscopic counting of bacteria and viable-cell portion of the membrane (2). The counting is done for large
counting. Natural waters and other fluids (e.g., foods, oils, numbers by using an appropriate eyepiece graticule ruled in
and solvent emulsions) can all be sampled, although each squares (counting the cells within a square, counting a sta-
type of sample requires appropriate filters and methods. tistically sufficient number of squares, and relating the
General overviews of membrane filtration can be found in counts to the total area counted, the filtration area, and the
most microbiology textbooks, and a comprehensive techni- volume filtered). For smaller numbers, use a statistically ap-
cal review is provided by Brock (2). A recommended filtra- propriate number of total fields of the objective in use
tion procedure for direct counting ( 3 3 ) is as follows. (which has to be calibrated as to the area observed for sub-
1. Prestain polycarbonate filters (25-mm diameter; pore sequent relation of the counts to the filtration area and vol-
diameter, 0.2 pm) for 5 min in a 0.2% (wtlvol) solution of ume filtered). Use filters of low porosity (0.2 pm), because
Irgalan black (color index, acid black no. 107; Union many bacteria in waters are small. Filters used for fluores-
Carbide Corp., New York, NY) in 2% (vol/vol) acetic acid. cence microscopy should have minimum reflectance,
(Prestained filters are usually commercially available and should not fluoresce, and should be stained black (section
are preferred.) 2.1.2). The size of the filter, e.g., 25 or 90 mm in diameter,
2. Rinse the filter in cell-free distilled water and place it must be appropriate for the volumes and cell concentrations
wet on a cell-free glass filter apparatus (Millipore Corp., involved and for the available filtering apparatus (e.g., the
Bedford, MA). Stained filters, dried after rinsing, can be syringe attachment or vacuum system).
stored for future use. The filters must be made transparent for counting by
3. Fix the cells in 0.1% (vol/vol) aqueous glutaralde- phase or ordinary microscopy. Dry the filters after staining
hyde. them with a basic dye (0.01 % [wt/vol] thionine). Clear a
4. Place the sample over the filter, and then add acridine cut-out portion of the filter with a drop of immersion oil or
orange (40% dye content) to a concentration of 0.1% xylene on the slide and on the membrane before covering
(wtlvol) in 0.02 M Tris, pH 7.2 (at 20C), to make a final with a coverslip. When doing filtrations in the field, dry the
concentration of 0.02% (wtlvol). filter immediately after filtration; later, wet a cut-out por-
5. After staining for 3 min, draw the water sample-acri- tion for appropriate staining and then dry it for microscopy
dine orange through the filter membrane by suction. (as described above).
6. Remove the membrane from the filter apparatus and A simple method used for total counts is the acridine or-
place it over a drop of immersion oil (type 1, F, or A; ange direct count by fluorescence microscopy (2, 4, 6), al-
Cargille Laboratories, Inc., Cedar Grove, NJ) on a glass mi- though newer stains such as the LIVE/DEAD Gram stain
croscope slide. Place another drop of oil on top of the mem- provided by Molecular Probes, Inc. (Eugene, OR), are also
brane, followed by a glass coverslip. The steps must be per- quite straightforward to use. For acridine orange counting,
formed rapidly to prevent the filter membrane from add the stain to the sample by bringing the acridine orange
becoming dry. Examine the preparation by epifluorescence concentration to 0.01 to 0.1% (wtlvol) before filtration or by
microscopy with a 100- or 200-W halogen lamp, a BG-12 holding a small volume of acridine orange on the filter for 1
excitation filter, an LP-510 barrier filter, and an FT-510 min after filtering the sample. Other fluorochromes, such as
beam splitter. Bacteria on this filter will fluoresce green, and the fluorescein isothiocyanate, DAPI, and SYTO@series of
individual morphological types can be observed. Although nucleic acid stains, can also be used (more details are given
acridine orange is easy to use (although a teratogen), it suf- in chapter 3). Diluent or rinsing waters should be of an ap-
fers from a tendency for nonspecific binding to organic mat- propriate quality and free of cells; e.g., samples from the sea
ter, and depending on the sample source, other fluorescent require filter-sterilized artificial seawater medium. Formal-
stains should be considered. dehyde (2%, wt/vol) may be used to prevent growth in sam-
ples. Published applications of the methods should be con-
Another, time-honored technique for scanning the biota sulted, and consideration should be given to the sampling
that attach to surfaces, popularized by A. T. Henrici decades and counting strategies (2, 8, 15,33). Also see chapter 1.
ago, involves suspending slides or coverslips for hours or
days in the water being sampled and retrieving them for
staining and microscopy. Many organisms from fresh and 2.1.5. Direct Viable-Cell Counting
salt water are able to attach to surfaces. Viable-cell counting from a natural sample (without added
formaldehyde) requires filtration of a set of appropriate di-
2.1.3. Soil lutions and placing of the filters on a suitable nutrient agar
One simple means of obtaining a selection (but by no for counting of the colonies formed after incubation.
means a complete one) of soil microorganisms for micro- Viable-cell counts can also be estimated by using the fre-
scopic examination consists of placing the soil sample in quency of dividing cells (2, 39) or by counting cells on a
the bottom of a watch glass or small dish and adding water membrane filter that are incorporating a given substrate (2,
below the sample by using a Pasteur pipette until a water sur- 6, 49), and the proporton of live to dead cells can be esti-
face is formed above the sample. Surface tension will have mated by correlating to total counts. The accuracy of these
ensured the positioning of many soil cells at the air-water methods is not great.
interface. A coverslip can be floated on the surface and can The nalidixic acid method is representative of the ap-
then be examined by microscopy. In situ incubation of glass proaches to viable-cell counts made by estimating synthetic
slides will also provide an impression of the bacterial popu- activity in bacteria (8). Here, the drug inhibits DNA syn-
lations present. thesis and division and the cells grow longer and more
22 MORPHOLOGY A N D ULTRASTRUCTURE

stainable. The procedure compares a fixed (2% [wt/vol] should be given to factors such as nonspecific binding, auto-
formaldehyde) aliquot of the sample with an unfixed fluorescence, and excitation-emission spectra and factors
aliquot to which is added 0.025% (wtlvol) yeast extract as a that may influence the efficiency of a fluor (see chapter 3
growth substrate plus 0.002% (wtlvol) nalidixic acid after and reference 37).
an incubation period of 6 h. Filter the samples, and stain
with 0.01% (wtlvol) acridine orange or fluorescein isothio- 2.1.6. Detection of Specific Bacteria
cyanate for epifluorescence microscopy. Viable cells elon- Recognition of specific bacteria in an environment by mi-
gate when exposed to yeast extract and nalidixic acid, in- croscopy requires the application of either immunofluores-
ducing nonseptate filaments which are enumerated as cence techniques (see chapter 3), direct serological proce-
viable cells. Gram-positive cells are relatively insensitive to dures (see chapter 8), or molecular methods by using
nalidixic acid; in some environmental samples, the species-specific RNA or DNA probes. The increasing
nalidixic acid method may underestimate their numbers power of molecular probes makes them the most popular
and one of the methods described below may be more ap- and specific of all staining regimens today since probes for
propriate. The technique can be modified or expanded as multiple receptors can be run on the same sample at the
necessary, for example, with the use of in situ hybridization same time (e.g., Color Plate 1 and reference 48).
to allow assessment of the activity of specific species or
groups of organisms (35).
Although acridine orange has been a traditional stain for 2.2. PREPARATION
nucleic acids, more stains are now available. DAPI is popu- Microscopy is effective and important in the primary char-
lar; it stains blue and penetrates into living cells. Ethidium acterization of organisms either present in the sample or
bromide stains red but cannot enter cells to stain nucleic growing in liquid media. Low-power microscopy can also be
acids unless the semipermeability of the plasma membrane applied to colonies growing on agar plates, and valuable in-
has been disrupted. A combination of DAPI and ethidium formation about the arrangement and viability of cells
bromide can differentiate live (DAPI-stained, blue) from within a colony can be obtained. Proper study of single
dead (ethidium bromide-stained, red) cells. A wide range of colonies requires proper expertise and tools (often readily
nucleic acid stains (the SYTO@series) is available from var- manufactured out of simple laboratory materials by an en-
ious suppliers (e.g., Molecular Probes, Inc.). terprising student) which are beyond the scope of this chap-
Certain companies have attempted to maximize the ease ter. Excellent instructions were provided in the correspon-
of differentiating between live and dead bacteria. The most ding chapter of the previous edition of this book, and we
popular is Molecular Probes LIVE/DEAD BacLight@viabil- refer you to it (12).
ity kit where two fluorescent reagents are mixed together to
stain the cells (see section 2.3.5.3). There is no data avail- 2.2.1. Living-Cell Suspensions
able about how these two stains interact with archaea, but There are several approaches to the examination of living
presumably, similar responses should be seen. There are not bacterial cells, either in a natural specimen or in culture.
as many commercial kits available for determining yeast vi- The initial preparation is usually a smear or film made by
x.
abilit Molecular Probes, Inc., also markets a LIVE/DEAD
Yeast kit which can be used for yeasts and a number of fil-
spreading a droplet with a wire loop over a few square cen-
timeters of a microscope slide and then drying and fixing
amentous fungi. Here, a novel two-color fluorescent meta- the slide (section 2.2.3) before simple staining (section
bolic probe (FUN@l ) is used in combination with calco- 2.2.4). Cultures grown in liquid media can be used with the
fluor white M2R, which is a general stain for many fungi. proviso that dilution with sterile medium (e.g., by adding a
FUN 1 initially stains the cells a diffuse green, but this color loopful to a drop of medium on a slide) is generally required
is gradually metabolized into orange-red that is partitioned for reducing the population for microscopic study to a use-
into the intravacuolar regions. Only living cells stain this ful concentration. To easily detect bacterial cells in liquid
way since the conversion from green to orange-red requires suspension, there should be more than lo5 cells ml-.
an intact cytoplasmic membrane and metabolic capacity. Cultures grown on solid media can be suspended on a mi-
Another similar reagent (FUN@2) is also available, and if croscope slide with a sterile loop in a drop of the liquid
this reagent is used, the initial diffuse green becomes yel- medium or a diluent (not tap water) to a faint turbidity.
low-orange. Cells that are not metabolically active remain Collect cells that are in very low concentration either by
green with both FUN 1 and FUN 2. These reagents have centrifugation or accumulation on a membrane filter (e.g.,
proven reliable for Saccharomyces, Candidu, Neurosporu, and a 0.2-pm-pore-size membrane filter in a syringe adapter),
Aspergillus spp. Additional information on these fluorescent which is gently pressed onto a slide to transfer cells for sim-
stains for both bacteria and yeasts can be found on the com- ple staining (50). Remember that the age and conditions of
panys website at www.probes.com (product information a culture may affect the size and shape of cells, their surface
sheets MP 07007 and MP 07009). components (flagella, pili, and capsules), their inclusions
Other fluorescent stains can also be used as vital dyes. (sulfur, volutin and poly-P-hydroxyalkoanate granules, and
For example, 0.02 to 0.05% (wtlvol) fluorescein diacetate is endospores), and the selection of mutants. The culture con-
nonfluorescent when taken up by cells but is hydrolyzed by ditions must be clearly defined in describing bacterial form
cellular esterases (38). The released fluorescein is easily de- and structure. Examination of living cells under conditions
tected by either transmitted- or epifluorescence microscopy, appropriate for growth is important to defining growth habit
and the method may be applied to samples on filters as de- (e.g., mycelial or differentiated forms) and behavior (e.g.,
scribed above. Additional stains for metabolic activity motility), as described for the following methods.
(CTC) and specific enzyme activities (ELF-97) are dis-
cussed in chapter 3. A series of protocols for physiological 2.2.1 .I. Wet Mounts
characterization of bacterial cells is provided in Lisle et al. In preparing wet mounts of specimens, place a loopful or 1
(37). In the application of fluorescent stains, consideration drop (ca. 0.05 to 0.1 ml) of the sample on a clean and de-
2. Sampling and Staining for Light Microscopy rn 23

greased (by prior heating for 20 min at 400C) microscope Consequently, the dark film dries so that the bacterium ap-
slide and cover it with a glass coverslip. The latter should be pears to be somewhat larger than in life, even if no capsule
18 to 22 mm2 and of no. 1 thickness (-0.15 mm thick) to is present.
allow an oil-immersion objective to focus through it. To During storage, nigrosin solutions can acquire contami-
prevent convection currents, drifting, and drying, seal the nating organisms. A little formaldehyde (0.5%) helps pre-
edges of the coverslip to the slide by applying Vaspar (a vent growth and does not harm the solution. Flame sterilize
mixture of equal parts of petroleum and paraffin wax), loops, because the presence of dead bacteria leads to confu-
birthday-candle wax (using the wick as a brush after melt- sion. India ink also forms a sort of negative stain when
ing the wax, not lighting it, in a pilot flame), or nail polish. added to a wet mount and can display large capsules effec-
The motility of strictly aerobic bacteria can be observed tively if the coverslip is pressed down tightly.
only for a brief time in these preparations, because the bac-
teria cease moving once the oxygen is depleted. The inclu- 2.2.3. Fixation of Smears and Suspensions
sion of small air bubbles prolongs activity. Heat from the A smear of bacteria (section 2.2.1) merely dried on a mi-
light source may interfere with motility if observations are croscope slide can wash off during staining unless the cells
to be made for a long period. A green filter is easy on the are made to stick to the glass. Good preservation of struc-
observers eyes, and it utilizes the best corrections of the ture usually requires a treatment to inactivate enzymes and
lenses. to cross-link macromolecules. This fixation can be physical
Phase-contrast microscopy is recommended for examin- or chemical and usually results in the killing of the cells.
ing wet mounts, and the thinnest possible film should be (Remember that spores and some vegetative cells, e.g.,
used for best results. Placing a piece of blotting paper over those of Mycobacterium spp., are remarkably resistant to
the coverslip before sealing assists in drawing off sufficient heat and chemical killing. Also, aerosol droplets can be
fluid to give a satisfactory thin film. Good views of cells can formed and can spread to the surroundings, and fingers are
be obtained if a thin film of 0.75to 1.5% (wtlvol) agar (for easily contaminated.)
best results, use Noble agar or dialyzed agar) or 15 to 30%
(wtlvol) gelatin is dried on the surface of the slide to absorb 2.2.3.1. Heat Fixation
the drop of culture as it spreads under the applied cover- Heat fixation is the most common procedure used for stabi-
slip. The cells are immobilized and immersed in a higher- lizing bacterial smears. Allow the smear to dry completely.
refraction-index fluid and are close to the coverslip. Pass the underside of the slide several times over the flame
of a Bunsen burner to induce adherence. The smear is now
2.2.2. Negative
- Staining- ready for simple staining (section 2.2.4), Gram staining
Negative staining provides the simplest and often the quick- (section 2.3.5), or other procedures. When considering
est means of gaining information about cell shape, cell break- morphological interpretations, remember that the drying
age, and refractile inclusions in cells such as sulfur and poly- and heating cause some shrinkage and distortion.
p-hydroxyalkoanate granules and about endospores ( 16).
2.2.3.2. Chemical Fixation
Procedure Chemical fixation provides more accurate preservation of
1. Place a droplet of 7% (wtlvol) nigrosin on a coverslip shape and structure, although it is more time-consuming
(no. 1 thickness). than heat fixation. Useful procedures are as follows.
2. Mix a small sample of bacteria into the droplet.
3. Place another coverslip, rotated at 45, over the drop- Fixation with an Aldehyde
let, and slide the two coverslips apart to form a thin film on Add formalin to a suspension to give a 5% (vol/vol) solu-
each. Alternatively, spread the droplet over most of the tion (i.e., -1.7% [wt/vol] formaldehyde), and hold for a few
coverslip with a loop until the fluid starts to dry and there minutes before making and drying a smear. This is a simple
are thicker and thinner parts of the film visible. and adequate preparation for Gram staining. Alternatively,
4. Let the film dry completely. use 3% (vol/vol) glutaraldehyde in the same fashion.
5. Place the coverslip on a slide, film side down, and fix it Osmium Tetroxide Fixation of Wet Films
in place with several spots of birthday-candle wax along two Prior to drying a smear, expose it to the fumes from aqueous
edges. 1%(wtlvol) Os04for 2 min in a closed vessel. (Caution: 0s-
6. Observe thin areas of the film under oil immersion. mium vapors are harmful, and this step must be done in a
Only experience teaches how much of a culture to mix into fume cabinet!) Cell arrangements will be better preserved if
how large a droplet and the appearance of an effectively this step is included.
spread film. There is considerable advantage to microscopy on chem-
These air-mounted preparations reveal bacteria or ically fixed and lightly stained microorganisms on coverslips
fragments of them unstained and standing out brightly that are mounted in water and not allowed to dry at any time in
against a sepia background (Color Plate 2). They can be the processing.
used to monitor the progress and extent of cell disruption
and disintegration (see chapter 7). Negatively stained 2.2.4. Simple Staining
preparations should not be used for making cell length and Morphological studies of bacteria are generally done on
width measurements (see below) because the capsule or heat-fixed smears that are stained with basic dyes. When a
slime layer outside the cell wall may exclude the nigrosin single dye is used, the process is referred to as simple stain-
and because they display dried, unfixed, and partially col- ing. Simple staining of smears on slides is accomplished
lapsed cells. Furthermore, the negatively charged particles conveniently on a rack made of glass rods linked by short
of colloidal nigrosin do not react with the bacterial surface lengths of rubber tubing or of linked brass rods supporting
because, at physiological pH, it also is negatively charged. slides over a sink or suitable vessel for disposal of stains and
24 MORPHOLOGY AND ULTRASTRUCTURE

wash water. Coverslip preparations are best handled in amenable to improvement to satisfy conditions for particu-
Columbia staining dishes. lar bacteria and the manner in which they have been
grown. Cytologically rewarding preparations require great
Procedure for Slides care in fixation, staining, handling, and microscopy. The
1. Make a smear (section 2.2.1) of either living or chem- following are sound routine methods, but some of the pro-
ically fixed cells (section 2.2.3.2). cedures, especially those for cytoplasmic inclusions, are
2. Let the smear dry completely. most effective if applied to chemically fixed preparations
3. Heat fix (section 2.2.3) to gain adherence to the glass. with drying avoided at any stage.
4. Flood the heat- or chemically fixed smear briefly with
a solution of a basic dye such as crystal violet (for 10 s) or 2.3.1. Size
methylene blue (for 30 s), and then gently rinse with tap or Cell lengths and widths cannot be measured with great pre-
distilled water and blot dry with absorbent paper. cision because of certain unavoidable technical difficulties.
The boundaries of living bacteria do not appear sharp when
Preparation of Staining Solutions examined by phase-contrast or bright-field microscopy, and
Crystal violet (Hucker formula) in phase microscopy they and internal details are obscured
by halos. The halo problem can be reduced by mounting the
Crystal violet ........................... 2.0 g organisms in a medium of a refractive index close to that of
Ethanol, 95% (vol/vol) . . . . . . . . . . . . . . . . . . . 20 mi their cellular substance, which is done simply by using solu-
Ammonium oxalate, 1% (wtlvol), aqueous . . . . 80 ml tions of gelatin from 15 to 30% (wtlvol) (17), the best re-
sults being determined by experiment for each kind of or-
Dissolve the crystal violet in the ethanol. Then add the ganism and the detail to be observed. A less precise method
ammonium oxalate solution, and allow it to stand for 48 h to reduce the halos involves drying, ahead of time, a thin
before use. film of 15 to 30% (wtlvol) gelatin on slides and using these
to make wet mounts. The fluid swells the gelatin, and the
Methylene blue (Loeffler formula) cells become embedded in it; this process immobilizes them
under the coverslip and allows better phase relations be-
Methylene blue chloride . . . . . . . . . . . . . . . . . . 1.6 g tween the organism and its environment. Similar prepara-
Ethanol, 95% (vol/vol) . . . . . . . . . . . . . . . . . . . 100 ml tions made on a thin layer of agar can also be useful (see sec-
Potassium hydroxide, 0.01% (wtlvol), tion 2.2.1.1).
aqueous ........................... . l o 0 ml Cells in fixed and stained preparations all suffer some
degree of distortion, depending on the technique used;
Prepare a saturated solution of methylene blue by adding hence, one obtains only an approximation of the true di-
the dye to the ethanol. Then add 30 ml of the supernatant mensions. Dried, stained films, even if prepared from for-
solution to the potassium hydroxide. malin-fixed material, do not reveal the cell wall. Tannic
If the cells stain lightly or not at all with one of the dyes, acid-crystal violet staining (44) does reveal the cell wall
it may be necessary to stain them with carbol fuchsin (sec- and makes bacteria appear wider than they look when alive
tion 2.3.6.1); they may even be acid fast (section 2.3.6). A or dried, but this staining does not work well with gram-
few seconds of exposure to this dye is usually enough for negative bacteria.
most bacteria. Microscopic measurements are made with either a cali-
brated ocular micrometer or a filar micrometer eyepiece.
2.2.5. Permanent Mounts Calibration is attained by using a stage micrometer, which
For extended preservation, the stained and dried film of provides a set of engraved lines at defined intervals (chap-
bacteria on a coverslip should be cleared with xylene and ter 1). After calibration, the stage micrometer is no longer
then inverted on a drop of Canada balsam on a slide. Wipe required, but new calibrations must be made for each objec-
excess balsam away with a tissue moistened with xylene. tive used. For repeated measurements and counting, consid-
The balsam hardens in a day. Neutral mounting media con- eration should be given to digital image analysis; an over-
sisting of a plastic (polystyrene) dissolved in a solvent view and examples are provided in reference 9.
(toluene or xylene) and containing a plasticizer (tricresyl
phosphate) are marketed under such names as Permount 2.3.2. Shape
(Fisher Scientific, Pittsburgh, PA). (Caution: xylene, For determinative and descriptive purposes, individual bac-
toluene, and many polystyrenes can be carcinogenic.) Some of terial shapes are designated as straight, curved, spiral, coc-
these media may be painted onto a stained film on a glass cobacillary, branching, pleomorphic, square ended, round
microscope slide without the need for a coverslip. Permount ended, tapered, clubbed, or fusiform. Perhaps square, star
is neutral and does not become acid or discolor with age, shaped, stalked, and lobular should be added to these classi-
nor does it tend to trap bubbles under the coverslip. Stained cal terms. Arrangements of individual bacteria are described
films of bacteria on coverslips, mounted in water and sealed as single, pairs, short chains (fewer than five bacteria), long
with petroleum, clear nail polish, or candle wax, can be chains (five or more bacteria), packets, tetrads, octets,
kept if evaporation is prevented. They remain useful for clumps, filaments, and branching or mycelial forms.
only a few days kept in a moist chamber, but they provide In assigning the descriptive terms, it is assumed that nat-
excellent material for photomicrography. ural groupings are not disturbed. For example, excessive
shaking may break long chains or cause clumping of single
cells. To minimize the breakup of long chains or other frag
2.3. C H A RACTERlZATl ON ile associations, add formaldehyde to the culture to a final
The methods described below have been found satisfactory concentration of 1 to 2% (vol/vol). Centrifuge after 15 min,
for revealing determinative characteristics for a large num- and resuspend the culture in water for making films or n e g
ber of different bacteria. These techniques, however, are ative stains. This is highly recommended for streptococci.
2. Sampling and Staining for Light Microscopy 25

The influence of culture age and medium composition also atively dry agar surfaces on which the bacteria move as
must be taken into account. When there are doubts, there groups or microcolonies (32) or, in some instances (e.g.,
is no substitute for examining early growth in the undis- Proteus species), as associations of filamentous swarmer
turbed state (section 2.2.1). cells. A continuously shifting pattern of organisms develops
on the agar surface, ranging from short, tongue-like exten-
2.3.3. Mode of Division sions and isolated comet-shaped groups cruising away from
The mode of division used by a given bacterium is ordinare the mass to interlacing bands interspersed with empty areas.
ily not seen at first glance, and continuous observation of Swarming is being seen among an increasing number of
the living organism in a suitable medium is required (41). bacteria as the conditions for induction are becoming
Simple staining procedures may not adequately reveal de- known.
tails whether division is binary or ternary or takes place
through budding or fragmentation. It may be necessary to 2.3.5. Gram Staining
use cell wall staining (44) and even electron microscopy Gram staining is the most important differential technique
(chapter 4) to determine this behavior. applied to bacteria (42). In theory, it should be possible to
divide bacteria into two groups, gram positive and gram
2.3.4. Motility negative; in practice, there are instances when a given bac-
Translational movement of bacteria by flagellar propulsion terium or archeon is gram variable (22-24). Numerous
(swimming) may be observed in wet mounts (section modifications of Gram staining have been published since
2.2.1.1) of specimens by use of, in most cases, the low-power the method was first developed by Christian Gram in 1884.
or high-dry objectives. Bacteria vary in their translational The cells are stained with crystal violet and then treated
velocity, and slow organisms must be differentiated from with an iodine solution as a mordant (1). The purple-
those showing only Brownian motion. To ascertain the stained cells are then washed with ethanol. Gram-positive
presence of flagella in doubtful cases, as well as to determine cells retain the stain (Color Plate 3), whereas gram-negative
flagellar distribution (polar, peritrichous, or lateral), stain. cells do not. To make the contrast between the two results
ing procedures (section 2.3.10) and electron microscopy obvious, the preparation is counterstained with a contrast-
(chapter 4) may be required. ing red dye (e.g., carbol fuchsin or safranin) so that gram-
When a standard condenser and an oil-immersion ob- negative cells become red and are easily seen.
jective are used, the light must be reduced by the aperture The Gram reaction is determined by the interaction of
diaphragm and the condenser must be lowered to improve crystal violet and iodine and by the integrity and the struc-
contrast. Much better resolution and contrast are attained ture of the cell wall, most particularly the molecular archi-
with phase microscopy and (more dramatically) with dark- tecture of the peptidoglycan (murein) sacculus of bacteria
field microscopy. If the specialized condensers needed for (25, 27, 46). Cell walls of gram-positive cells do not allow
the latter technique are not available, an adequate dark the extraction of the crystal violet-iodine complex from the
field is possible by using the stratagem of employing a phase cytoplasm by a solvent. The crystal violet molecule is small
microscope condenser, with the oil-immersion phase plate enough to penetrate the interstices of the wall, but the dye-
in place, for illuminating the specimen observed with stan- iodine complex is too large to exit (25). If the walls are bro-
dard (not phase) 1OX or 45X objectives (chapter 1) and ken or their structure is compromised by autolysis, exposure
high-power eyepieces (12X to 16X). The condenser is not to lysozyme, or exposure to a wall-targeted antibiotic (e.g.,
immersed, but its position for attaining the best dark field is penicillin), the complex is extracted during the decoloriza-
discovered by trial. This trick makes it possible to see even tion step and the gram-positive bacteria stain as gram neg-
spirochetes under low power! ative (Color Plate 3). Gram-positive bacteria that have died
Observation of living cells sometimes detects another naturally stain as gram negative since autolysins have hy-
motion effectively described as twitching (32), which is a drolyzed their cell walls.
sudden movement or change of position that is believed to Archaea can also be stained by using the Gram reaction,
be associated with the extension and retraction of fimbriae but students beware! Their cell walls possess entirely differ-
(pili) on the cells. ent polymers and can be much simpler or more complex
Some prokaryotic protists exhibit a peculiar type of than those of bacteria; the staining response is not always
translational movement known as gliding when in contact unequivocal ( 1, 22-24).
with a solid surface. Gliding is a comparatively slow, stately, The description of novel bacteria requires recording of
intermittent progression parallel to the longitudinal axis of the Gram reaction, but this description should also include
the organism. It is characterized by frequent directional the structural profile of the cell wall observed in sections by
changes and the absence of external locomotory organelles electron microscopy (chapter 4).
(32). Gliding is often not obvious on the primary medium The smear of bacteria on the slide can be made directly
of isolation but is usually facilitated on solid media con- for routine Gram staining purposes from a liquid or solid
taining very small amounts of the nutrients appropriate to culture, as for simple staining (section 2.2.4), but is best
the group of bacteria involved. It is best observed on an agar made from a formalin-fixed, washed sample. The few extra
plate with a high-dry objective or with a low-power objec- minutes spent suspending the bacteria in 5% (vol/vol) for-
tive and high-power eyepiece. Single bacteria away from malin and concentrating and washing them by centrifuga-
the margin of the colony and tracks or trails are often con- tion pay off in good preservation of size and shape, good
sidered indicative of gliding motility. Young, active cultures staining, and the absence of messy precipitates from liquid
of filamentous gliders form wavy, curly, or Iacelike patterns. culture media. In either case, a thin smear should be used
Translocation by gliding movement can occur at velocities for air drying and heat fixation (thick clumps are hard to de-
of 10 to 15 Fm/s. colorize and can give a false positive).
Gliding should be differentiated from swarming, which is Gram-negative bacteria may seem to be gram positive if
an expression of flagellar motility on agar under special con- the film is too thick and the decolorization is not com-
ditions. Swarming is the active spreading of growth on rel- pleted. Gram-positive organisms, on the other hand, may
26 MORPHOLOGY AND ULTRASTRUCTURE

seem to be gram negative if the film is overdecolorized; this Stain Solution A


happens particularly if the culture is in the late stationary
phase of growth. Some Bacillus species are gram positive for Crystal violet (certified 90% dye content) . . . . 1.0 g
only a few divisions after spore germination. It is advisable Distilled water ......................... . l o 0 ml
to prepare light films (faint turbidity) of young, actively
growing cultures for best results, since older cultures tend to Bicarbonate Solution B
give variable reactions. A wise precautionary measure is to
use known gram-positive and gram-negative organisms as Sodium bicarbonate ..................... 1.0 g
controls. Distilled water ......................... . l o 0 ml
It is important to standardize the Gram staining proce-
dure, and two methods that give equivalent results are those Iodine Mordant
of Hucker and of Burke (20, 21), as follows.
Iodine ................................ 1.O g
2.3.5.1. Hucker Staining Method Potassium iodide ........................ 2.0 g
See references 6 and 20. Distilled water .......................... 100 ml

Solution A Prepare this mordant as for Huckers method.

Crystal violet (certified 90% dry content) . . . . . 2.0 g Decolorizing Solvent


Ethanol, 95% (vol/vol) . . . . . . . . . . . . . . . . . . . 20 ml Caution: this solvent is highly flammable.

Solution B Ethylether .......................... 1 volume


Acetone ............................ 3 volumes
Ammonium oxalate ...................... 0.8 g
Distilled water .......................... 80 ml Counterstain

Mix A and B to obtain the crystal violet staining reagent. Safranin 0 (85% dry content) . . . . . . . . . . . . . 2.0 g
Store it for 24 h, and pass it through filter paper before use. Distilled water ......................... . l o 0 ml

Mordant Procedure
1. Flood the smear with solution A, add 2 or 3 drops of
Iodine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.O g bicarbonate solution B, and let stand for 2 min.
Potassium iodide ........................ 2.0 g 2. Rinse off the stain with the iodine mordant, and then
Distilled water ......................... 300 ml cover the smear with fresh iodine mordant for 2 min.
3. Wash off the mordant with a gentle and indirect stream
Grind the iodine and potassium iodide in a mortar, and of tap water for 2 s; blot around the stained area with ab-
add water slowly with continuous grinding until the iodine sorbent paper, but do not allow the smear to dry.
is dissolved. Store the mordant in amber bottles. 4. Add the decolorizing solvent dropwise to the slanted
Decolorizing Solvent slide until no color appears in the drippings (less than 10 s),
Ethanol, 95% (vol/vol) and allow the smear to air dry.
5. Flood the smear for 5 to 10 s with the counterstain.
Counterstain 6. Wash the smear in a gentle and indirect stream of tap
water until no color appears in the effluent; blot the smear
Safranin 0 (2.5% [wt/vol] in 95 % [vol/vol] dry with absorbent paper.
ethanol) ............................ 10 ml
Distilled water ......................... 100 ml Several microbiological supply houses provide reliable
kits which contain all of the reagents necessary for Gram
staining; although these are more expensive than obtaining
Procedure
and making each reagent yourself, the convenience of a kit
1. Place the slide on a staining rack, and flood the smear may be worthwhile (see section 2.5).
with the crystal violet staining reagent for 1 min.
2. Wash the smear in a gentle and indirect stream of tap 2.3.5.3. LIVE BacLight@and Viacram@Methods
water for 2 s. The LIVE BacLight@Gram stain kit is supplied by Molec-
3. Flood the smear with the iodine mordant for 1 min. ular Probes, Inc. (Eugene, OR) and provides a one-step flu-
4. Wash the smear in a gentle and indirect stream of tap orescence analysis of the Gram state of living bacteria. The
water for 2 s; blot the film dry with absorbent paper. kit contains a mixture of green fluorescent SYT09 and red
5. Flood the smear with 95% ethanol for 30 s with agita- fluorescent hexidium iodide (a nucleic acid stain). SYT09
tion; blot the film dry with absorbent paper. stains both live gram-positive and gram-negative bacteria.
6. Flood the smear with the safranin counterstain for Hexidium iodide preferentially stains live gram-positive
10 s. cells, and this stain displaces the SYT09 dye. Accordingly,
7. Wash the smear with a gentle and indirect stream of gram-negative bacteria fluoresce green, whereas gram posi-
tap water until no color appears in the effluent, and then tives fluoresce red. The Gram response can be distin-
blot the film dry with absorbent paper. guished between the two types in mixed cultures. Dead
cells will give variable results, and it is best to use this
2.3.5.2. Burke Staining Method method on actively growing exponential cultures. SYT09
See reference 4. has an excitation-emission maximum of 490 nm/500 nm,
2. Sampling and Staining for Light Microscopy H 27

and hexidium iodide has an excitation-emission maximum Decolorizing Solvent


of 480 nm/625 nm. Since this is a relatively new Gram
staining method, it has not been tested on a large variety Ethanol, 95% (vol/vol) .................. 97 ml
of bacteria and some inconsistencies with traditional Gram HC1 (concentrated) ..................... 3 ml
staining responses may still be found. So far, there are
no data available about how the LIVE BacLight kits stain Counterstain
archaea.
ViaGram@Red+ Gram stain is another kit put out by Methylene blue chloride ................. 0.3 g
Molecular Probes, Inc., and it has the advantage of also dif- Distilled water ......................... 100 ml
ferentiating between live and dead cells based on their
plasma membrane integrity. The kit contains three
reagents, two nucleic acid stains for viability determination Procedure
and a fluorescently labeled wheat germ agglutinin (WGA) 1. Place a slide with an air-dried and heat-fixed smear on
for the determination of the Gram reaction. DAPI is a flu- a slide carrier over a trough. Cut a piece of absorbent paper
orescent blue stain used for cells with intact membranes to fit the slide, and saturate the paper with the carbol
(i.e., live cells), whereas SYTOXB Green can stain nucleic fuchsin stain. Carefully heat the underside of the slide by
acids only if the plasma membrane has been disrupted (i.e., passing a flame under the rack or by placing the slide on a
dead cells). The WGA is chemically attached to Texas hot plate until steam rises (without boiling!). Keep the prepa-
Red@-Xdye, and this entire complex selectively binds to ration moist with stain and steaming for 5 min, repeating
most gram-positve bacteria. Once bacteria are stained, here the heating as needed. (Caution: overheating causes spat-
are the Dossible results. tering of the stain and may crack the slide.) Wash the film
1. Gram negative, live: blue interior (DAPI) with a gentle and indirect stream of tap water until no color
2. Gram negative, dead: green interior (SYTOX Green) appears in the effluent.
3. Gram positive, live: blue interior (DAPI) and red sur- 2. Holding the slide with forceps, wash the slide with the
face (WGA-Texas Red) decolorizing solvent. Immediately wash with tap water, as
4. Gram positive, dead: green interior (SYTOX Green) described above. Repeat the decolorizing and washing until
and red surface (WGA-Texas Red) the stained smear appears faintly pink.
3. Flood the smear with the methylene blue counterstain
As with the LIVE BacLight@staining method, this is a for 20 to 30 s, and wash it with tap water as described above.
relatively new method and a full range of bacteria have not 4. Examine under oil immersion. Acid-fast bacteria ap-
yet been studied to ensure accurate correlation with more pear red, and non-acid-fast bacteria (and other organisms)
traditional Gram staining regimens. From our own experi- appear blue.
ence, WGA (i.e., WGA-Texas Red) does not always bind
to surfaces of gram-positive bacteria and clearly additional 2.3.6.2. Truant Staining Method
surface components such as capsules, S-layers, and sheaths See reference 5 1.
could affect this new stain. It is also possible to determine the acid fastness of an or-
For more information on both of these new fluorescent
ganism by using fluorescence techniques. A good example is
stains, consult the product information pages (MI 07008 the Truant technique. An advantage of this procedure is
and MP 07022 at www.probes.com). that slides of clinical specimens suspected of containing
mycobacteria, for example, may be screened by using a 60X
2.3.6. Acid-Fast Staining
rather than a lOOX objective; hence, an entire slide may be
A second important tinctorial property of certain bacteria is screened in a short time.
that of not being readily decolorized with acid-alcohol after
staining with hot solutions of carbol fuchsin. Acid-fast bac- FIuorescent Staining Reagent
teria include the actinomycetes, mycobacteria, and some
relatives which, it is believed, react in this way because of Auramine 0,C141000 . . . . . . . . . . . . . . . . . . 1.50 g
limited permeability of the waxy components of the cell Rhodamine B, C1749 . . . . . . . . . . . . . . . . . . . 0.75 g
wall. Dormant endospores are also acid fast, but the cause is Glycerol ............................. 75 ml
unknown. Phenol (heat-melted crystals) . . . . . . . . . . . . . 10 ml
Two methods are recommended for acid-fast staining. Distilled water ......................... 50 ml
2.3.6.1. Ziehl-Neelsen Staining Method Mix the two dyes with 25 ml of the water and the phe-
See reference 6. nol. Add the remaining water and the glycerol, and mix
again. Filter the resulting fluorescent staining reagent
Carbol Fuchsin Stain through glass wool.
Basic fuchsin .......................... 0.3 g
Ethanol, 95% (vol/vol) .................. 10 ml DecolorizingSolvent
Phenol, heat-melted crystals . . . . . . . . . . . . . . 5 ml
Distilled water ......................... 95 ml Ethanol, 70% (vol/vol) . . . . . . . . . . . . . . . . . . 99.5 ml
HC1 (concentrated) ..................... 0.5 ml
Dissolve the basic fuchsin in the ethanol, and then dis-
solve the phenol in the water. (Caution: phenol will harm Counterstain
skin and the fumes can be irritating.) Mix, and let the mix-
ture stand for several days. Pass the mixture through filter Potassium permanganate . . . . . . . . . . . . . . . . . 0.5 g
paper before use. Distilled water ......................... 99.5 g
28 MORPHOLOGY A N D ULTRASTRUCTURE

Procedure Air dry the specimen on a glass slide, heat fix, cover with
absorbent paper saturated with the dye, and place over a
1. Flood a lightly heat-fixed smear with the fluorescent boilingwater bath for 5 min. Wash the slide in tap water,
staining reagent for 15 min. and counterstain the film with safranin for 30 s as in Gram
2. Wash the slide with a gentle and indirect stream of staining (section 2.3.5). Then wash the slide and blot dry.
distilled water until n o color appears in the effluent. Endospores appear bright green, and vegetative cells appear
3. Flood the smear with the decolorizing solvent for 2 to brownish red.
3 min, and then wash the slide with distilled water as de-
scribed above. 2.3.7.4. Dorner Staining Method
4. Flood the smear with the permanganate counterstain See reference 28.
for 2 to 4 min.
5. Wash the slide with distilled water as described above, 1. Heat fix the sample on a glass slide, and cover it with
blot with absorbent paper, and dry. a square of absorbent paper cut to fit the slide.
2. Saturate the paper with carbol fuchsin, and steam for
Examine with a fluorescence microscope equipped with
5 to 10 min, as described in section 2.3.6.
a BG-12 exciter filter and an OG-1 barrier filter. Acid-fast
3. Remove the paper and decolorize the film with acid-
bacteria appear as brightly fluorescent yellow-orange cells
alcohol (3 ml of concentrated HC1 in 97 ml of 95%
on a dark background; non-acid-fast cells are dark.
ethanol) for 1 min; rinse with tap water, and blot dry. Use
2.3.7. Endospores forceps to handle the stain-covered slides.
Mature, dormant endospores of bacteria, when viewed un-
4. Place a drop of 7% nigrosin (section 2.2.3) on the
slide, cover with another slide, and draw the two apart to
stained, are sharp edged, even sized, and strongly refractile,
form a thin film of the nigrosin over the stained smear.
shining brightly in a plane slightly above true focus. It is the
5. Examine under oil immersion. Vegetative cells appear
core (protoplast) of the spore that is refractile; the surround-
colorless, the endospores appear red, and the background
ing cortex, coat, and exosporium appear dark and often diffi-
appears black to sepia.
cult to discern. Electron microscopy is needed for determining
the details of these peripheral structures. Do not assume that 2.3.8. Cysts
any highly refractile body within a bacterium is an endospore,
Bacterial cysts (e.g., as in Azotobucter spp.) stain weakly
particularly if the inclusion body is irregularly sized and if in-
with simple stains and generally appear as spherical bodies
formation concerning heat resistance is lacking; large poly-@-
surrounded by thick but poorly staining walls. The follow-
hydroxyalkoanate granules often appear in the cytoplasm of
ing method (52) is useful for demonstrating Azotobucter
Bacillus spp. before sporulation and can be confusing. cysts, including forms developed prior to the appearance of
2.3.7.1. Popping Test a mature cyst.
A direct test is provided by a visible phenomenon (16) occur- Reagent
ring in dormant endospores after immersion in an acid oxi-
dizer (e.g., 0.1% wt/vol KMnO, in 0.3 N HNOJ. The spore Glacial acetic acid ...................... 8.5 ml
cortex ruptures, and a portion of the spore protoplast includ- Sodium sulfate (anhydrous) . . . . . . . . . . . . . . . 3.25 g
ing all of the nucleoplasm, now readily stainable with basic Neutral red ........................... 200 mg
dyes, is herniated through the aperture. The dramatic sudden- Light green S.F. yellowish . . . . . . . . . . . . . . . . 200 mg
ness of the event (after about 5 min in the solution) makes the Ethanol, 95% (wtlvol) . . . . . . . . . . . . . . . . . . . 50 ml
term popping test appropriate. The test can be conveniently Distilled water ......................... 100 ml
performed by mounting a dried film of spores on a coverslip in
the reagent for 10 to 20 min; the consequent popping is visi- Add the chemicals and dyes to the water with continu-
ble with the oil immersion objective without staining, al- ous stirring for 15 min. Pass the mixture through a filter
though staining makes the popping more easily visible. (pore diameter, 0.5 pm).
2.3.7.2. Negative Staining Procedure
There are several methods for staining endospores in bacte- Immerse the specimen in the reagent, and examine the wet
ria. The simplest method is the use of a negative stain (sec- preparation. Vegetative azotobacters are yellowish green;
tion 2.2.2), which yields lifelike preparations (16). In this early encystment stages appear with a darker green cyto-
method, mix a small loopful of 7% (wtlvol) aqueous ni- plasm somewhat receded from the outer cell wall, from
grosin on a coverslip with an appropriate amount of culture, which it is divided by a brownish red layer. In mature cysts,
spread it into a thin film, and air dry. Invert the coverslip, the central body appears dark green and is separated by the
place it film side down on a microscope slide (so the film re- unstained intine from the outer, brownish red exine periph-
mains in air when the coverslip is examined under oil), and eral layers of the cyst.
maintain it in place with several spots of candle wax.
Endospores appear as highly refractile spherical or ellip- 2.3.9. Capsules and Slime layers
soidal bodies both within the bacteria and free. Capsules and slime layers are produced by bacteria capable
Endospores strongly resist staining by simple dyes until of forming them under specific culture conditions and are
they germinate. Dormant endospores, once stained, are best demonstrated in wet preparations because the highly
quite resistant to decolorization and hence are acid fast hydrated polymers constituting them are distorted and
(section 2.3.6). A useful positive-staining method for bac- shrunk by drying and fixation.
terial endospores is that of Schaeffer-Fulton, as follows. The capsules of specific pathogens (e.g., pneumococci,
Klebsiellu pneumoniue, and meningococci) can be displayed
2.3.7.3. Schaeffer-Fulton Staining Method effectively by using antisera specific for the capsule type,
In the Schaeffer-Fulton technique (47), 0.5% (wtlvol) and this method can provide a presumptive identification.
aqueous malachite green is used instead of carbol fuchsin. Suspend the clinical exudate or the organism in a drop of
2. Sampling and Staining for Light Microscopy 29

capsule-specific antiserum containing 0.01% (wtlvol) ing dye solution may be kept under refrigeration for 1 to 2
methylene blue under a coverslip. The cell is stained and months, but it should be used as soon as possible.
the surrounding capsule becomes outlined by a dark blue
line in a short time. For determinative purposes, this is Procedure
known as the Neufeld Quellung (or capsule-swelling) test. 1. Prepare an air-dried film on a slide. Using a wax glass-
Fluor-conjugated lectins may also be used to see capsules marking pencil, draw a rectangle around the film.
and more extensive exopolymer networks around cells and 2. Flood dye solution onto the slide within the confines
cell aggregates (chapter 3). of the wax lines. Leave for 7 to 15 min, the best time to be
If antisera are not available, Duguids method (see determined experimentally.
below) is the simplest and best staining method. 3. As soon as a golden film develops on the dye surface
and a precipitate appears throughout the film, remove the
2.3.9.1. Duguid Negative Staining Method stain with gently flowing tap water. Air dry.
See reference 29. 4. Examine under oil immersion. Bacterial bodies and
flagella stain red.
1. Place a large loopful of India ink on a clean slide, and
mix in a loopful of culture; then place a glass coverslip over 2.3.1 1. Cytoplasmic Inclusions
this in such a way that only part of the mixture is covered. Many bacteria grown under certain conditions produce de-
2. Press firmly down on the coverslip, using several posits within the cytoplasm which are termed inclusions.
thicknesses of absorbent paper, until the ink is sepia be- Among these are deposits of fat, poly-P-hydroxyalkoanate
neath the cover glass. (often referred to as poly-P-hydroxybutyrate), polyphos-
3. Examine with high-dry and oil immersion lens sys- phate, starch-like polysaccharides, sulfur, and various crys-
tems. Capsules appear as clear zones around the refractile
tals. Some of these are polymerized waste products, and oth-
organism and against the brownish black background full of ers are food reserves. Several staining procedures can be
dancing particles of India ink. used to reveal some of these inclusions, which are also evi-
dent by both phase-contrast and interference microscopy
2.3.1 0. Flagella because of their refractility.
Although Koch devised a technique for staining flagella
over a century ago, no easy or constantly reliable method is 2.3.1 1 . I . Staining of Poly-p-Hydroxyalkoanate
yet available. Since the width of individual bacterial flagella
lies below the limits of resolution for transmission light mi- 1. Prepare a heat-fixed film of the specimen on a slide
croscopy (flagella are 14 to 28 nm in diameter [53]), it is and immerse in a filtered solution of 0.3% (wtlvol) Sudan
necessary to tar and feather the flagella to make them vis- black B made up in ethylene glycol. Stain for 5 to 15 min.
ible. NanoOrange@ (Molecular Probes) is a new fluorescent Drain and air dry the slide.
protein stain that has also proven to be an effective method 2. Immerse and withdraw the slide several times in xy-
for screening bacteria for flagella (30). lene, and blot dry with absorbent paper.
Best results are obtained when using superclean slides; 3. Counterstain for 5 to 10 s with 0.5% (wt/vol) aqueous
this reduces drying artifacts and precipitation of stains. safranin.
Because flagella are sheared off easily by drying and me- 4. Rinse the slide with tap water, and blot dry.
chanical forces, there should be a minimum of agitation 5. Examine under oil immersion. Poly-P-hydroxy-
while making the smear. More information about the form alkoanate inclusions appear as blue-black droplets, and cy-
and disposition of flagella is obtained by electron mi- toplasm appears pink.
croscopy (chapter 4).
Some methods for staining flagella are labeled as simple 2.3.1 1.2. Staining of Polyphosphate (Metachromatic
(31), but few are simple in practice. Leifsons method has or Volutin Granules)
enjoyed good success for many years. 1. Prepare a heat-fixed film of the specimen on a glass
slide and stain for 10 to 30 s in a solution of Loeffler meth-
2.3.10.1. Leifson Staining Method ylene blue (section 2.2.4) or 1% (wtlvol) toluidine blue.
See reference 36. 2. Rinse the slide with tap water, blot dry, and examine
under oil immersion. With methylene blue, polyphosphate
Solution A granules appear as deep blue to violet spheres and the re-
maining cytoplasm appears light blue. Toluidine blue stains
Sodium chloride ........................ 1.5 g metachromatically, and the granules appear red in a blue cy-
Distilled water .......................... 100 ml toplasm.
Solution B 2.3.1 1.3. Periodate-Schiff Staining of Glycogen-Like
Pol ysaccharides
Tannic acid ............................ 3 .O g
Distilled water ......................... . l o 0 ml See reference 34.
1. Prepare a heat-fixed smear of the specimen on a glass
Solution C slide.
2. Flood the slide for 5 min with periodate solution (20
Pararosaniline acetate .................... 0.9 g ml of 4% [wt/vol] aqueous periodic acid, 10 ml of 0.2 M
Pararosaniline hydrochloride . . . . . . . . . . . . . . . 0.3 g aqueous sodium acetate, 70 ml of 95% ethanol [protect the
Ethanol, 95% (vol/vol) . . . . . . . . . . . . . . . . . . .lo0 ml solution from light!]). Wash the slide with 70% ethanol.
3. Flood the slide for 5 min with a reducing solution con-
Mix equal volumes of solutions A and B; then add 2 vol- taining 300 ml of ethanol, 5 ml of 2 M HC1, 10 g of potas.
umes of this mixture to 1 volume of solution C. The result- sium iodide, 5 ml of sodium thiosulfate pentahydrate, and
30 MORPHOLOGY A N D ULTRASTRUCTURE

200 ml of distilled water. (Add the ethanol and then the served by dark-field microscopy. The following positive
HC1 to the solution of potassium iodide and sodium thio- stain generally gives good results.
sulfate in distilled water. Stir, allow the sulfur precipitate to
settle, and then decant the supernatant for use.) Fontana Silver Staining
4. Wash the slide with 70% ethanol. Solution A (fixative)
5. Stain for 15 to 45 min (the time to be determined ex-
perimentally) with the following Schiff reagent. (Dissolve 2
Acetic acid ............................ 1 ml
g of basic fuchsin in 400 ml of boiling distilled water, cool
Formalin .............................. 2 ml
to 50C, and filter through paper. Add 10 ml of 2 M HC1
Distilled water ......................... . l o 0 ml
and 4 g of potassium metabisulfite to the filtrate. Stopper
tightly, and allow to stand for 12 h in a cool, dark place.
Add about 10 ml of 2 M HC1 until the reagent, when dried Solution B
Ethanol, absolute
on a glass slide, does not show a pink tint.) Store the
reagent in the dark. Solution C (mordant)
6. Wash the smear several times in a solution consisting
of 2 g of potassium metabisulfite and 5 ml of concentrated Phenol ............................... 1g
HC1 in 500 ml of distilled water. Tannic acid ............................ 5g
7. Wash the slide with tap water, and counterstain the Distilled water ......................... . l o 0 ml
smear with a 0.002% (wtlvol) aqueous malachite green for
2 to 5 s. Solution D
8. Wash the slide in tap water, blot dry, and examine Add a solution of 10% (wtlvol) aqueous ammonium hy-
under oil immersion. Polysaccharides appear red, and cyto- droxide dropwise to a 0.5% (wtlvol) aqueous solution of sil-
plasm appears green. ver nitrate until the precipitate that initially forms just re-
dissolves.
2.3.1 2. Nucleoids
The display of bacterial nucleoids requires special care in Procedure
fixation and in mounting of the preparation for high-reso- 1. Using ultraclean slides, air dry a film of the specimen
lution microscopy. They can be seen in living cells (section on a slide.
2.2.1) by phase-contrast microscopy as long as the refractive 2. Fix the film in solution A for 1 to 2 min, and rinse it
index of the surrounding medium is raised to a sufficient in solution B for 3 min.
level (15 to 30% gelatin in a fluid mount; the exact con- 3. Cover the film with solution C, and heat until steam
centration requires experimentation). Interference mi- rises for 30 s.
croscopy (chapter l ) may be used if it is available. 4. Wash the film with distilled water, and air dry.
Fixing and staining of bacterial nucleoids to show reli- 5. Cover the film with solution D, and heat until steam
able shape and form are difficult at the best of times (39), rises and the film appears brown. Wash the film, and air dry.
and the reader is referred to the excellent review article 6. Examine under oil immersion. Spirochetes appear
by Robinow and Kellenberger (43). The cytological brownish black.
methods given in the previous edition (12) are most reli-
able. Nucleoids can also be revealed by fluorescence 2.4.2. Mycoplasmas
methods (9, 18, 37), and more information can be found Mycoplasmas are the smallest free-living forms observable
in chapter 3. by light microscopy and are hard to recognize individually
because of extreme polymorphism. They are parasitic in an-
imal and plant tissues, and their growth and study require
2.4. SPECIAL TECHNIQUES specialized techniques. The inexperienced would be well
A great number of techniques based on light microscopy are advised to seek expert help when faced with the need to
special to particular areas of bacteriology. Medical microbi- recognize, cultivate, and identify mycoplasmas.
ology, in particular, has had to develop many methods to as- Mycoplasmas lack cell walls, and their cells are bounded
sist decisions about procedure, to attain an early presump- only by a plasma membrane. The individual cells tend to be
tive diagnosis, to detect bacteria that are difficult to stain or irregular, but a number of species and genera each have def-
that occur in small numbers, and to confirm the identity of inite overall shapes (star shaped, round, goblet shaped, fila-
a specific organism. These uses and attendant precautions mentous, and helical) which can be recognized by using
are detailed in chapters of the Manual of Clinical Micro- phase-contrast or dark-field microscopy (chapter l ) ,usually
biology, in which the staining methods are separately sum- assisted by electron microscopy of both negatively stained
marized (6), as well as in other references in section 2.6.1. and sectioned preparations (chapter 4). Some species show
Here, we give examples of some special methods not rou- a gliding motility on the surfaces of the tissues they infect.
tinely used. However, mycoplasmas growing in complex medium, tissue
culture, or plant or animal tissues can be hard to distinguish
2.4.1. Spirochetes among the large array of confusing artifacts and tissue com-
Spirochetes do not stain well with ordinary stains, but free- ponents.
living spirochetes show up well negatively stained in ni- They grow slowly on appropriate nutrient agar plates,
grosin films (section 2.2.2). A number of spirochetes have a and colonies are microscopic, often 5 100 Fm in diameter.
width near the limits of light microscope resolution; conse- Plates must be examined with a stereoscopic dissecting mi-
quently, these bacteria are best observed in life by direct croscope (magnification, X20 to X60) by using oblique il-
dark-field or phase-contrast microscopy. Spirochetes lumination to recognize the characteristic fried-egg
stained with Giemsa, a Romanowsky-type stain used for colonies, which have a central plug penetrating the agar
hematology (lo), fluoresce bright golden yellow when ob- and a superficial spreading periphery. Colony margins may
2. Sampling and Staining for Light Microscopy 31

also be examined in situ at high magnification by placing a Dissolve the dye in the ethanol, and then add the other
droplet of methylene blue onto the colony and covering it ingredients. Allow the stock solution to stand at 37C for
with a coverslip for examination by using transmitted light 48 h. For use, dilute the stock solution 1:2.5 with phosphate
with high-power objectives. buffer (pH 7.5) prepared by mixing 3.5 ml of 0.2 M
The standard stained smears made by using heat-fixed NaH2P04, 15.5 ml of 0.2 M Na2HP04, and 19 ml of dis-
films and Gram staining or staining with basic dyes are use- tilled water. Then filter the solution, which will keep for 3
less and uninformative. Stained preparations of mycoplas- to 4 days but should be filtered before each use.
mas require careful chemical fixation, and staining with
Giemsa (section 2.4.3.1) or Wrights (6) stain is preferred. Procedure
The osmotic requirements for integrity of shape and form 1. Heat fix the smear, and cover it with the staining so-
must be maintained until the chemical fixation is complete. lution for l to 2 min.
2. Wash the smear with tap water, and counterstain for 5
2.4.3. Rickettsiae to 10 s with 0.8% (wtlvol) aqueous malachite green.
Most rickettsiae are obligate intracellular parasites that are 3. Wash the smear again with tap water, and make a sec-
rod shaped, occur in pairs, and show marked pleomorphism. ond application of the malachite green. Rinse the slide
Infected tissue samples may be examined directly by fluo- thoroughly in tap water, and blot it dry.
rescence microscopy with specific antisera labeled with flu- 4. Examine under oil immersion. Rickettsiae appear red-
orescein isothiocyanate. With the Gram stain, rickettsiae dish against a green background.
appear gram negative.
Rickettsiae may be seen in films of infected tissue by di- 2.4.4. Legionella
rect microscopic examination after staining by the Giemsa The members of the family Legionellaceae cause problems of
or Gimenez procedure (see below). Most rickettsiae appear recognition and detection because several species in the
as pleomorphic coccobacillary organisms varying in length genera Legionella, Fluoribacter, and Tatlockia cause life-
from 0.25 to 2.0 pm. Pairs and short chains are most fre- threatening disease, yet they and other species are common
quently observed. Coxiella burnetii is the smallest (0.25 by in freshwaters (including tap water) and inhabit some
1.0 pm), and it appears as a bipolarly staining rod in the cy- species of amoebae. Because they grow only on special
toplasm of infected cells. The rickettsiae that cause typhus media, a degree of suspicion of their presence is needed.
and spotted fever are generally larger (0.3 to 0.6 by 1.2 pm). The appearance of simple to pleomorphic bacilli in tissues
Spotted-fever rickettsiae are found in the nuclei and cyto- and exudates or cultures from likely sources revealed by the
plasm of infected cells and often appear to be surrounded by nonspecific Gimenez (section 2.4.3.2) or Gram stain may
a halo, sometimes mistaken for a capsule. suggest the use of the direct fluorescent-antibody test as a
means of presumptive determination. This test involves the
2.4.3.1. Ciemsa Staining Method use of several group-specific polyvalent pools of fluorescent
antibody. Once a group is identified, a species-specific fluo-
Stock Solution
rescent antibody can be used. False-positive reactions are
rare and are usually laboratory induced (e.g., because of in-
Giemsa powder ......................... 0.5 g correct reagent or contaminations) when they occur. The
Glycerol ............................... 33 ml
commercial antibody is usually conjugated with fluorescein
Absolute methanol (acetone free) . . . . . . . . . . . 33 ml
isothiocyanate. The procedure (45), which is an example of
similar direct-fluorescent antibody tests for other organisms,
Dissolve the Giemsa powder in the glycerol at 55 to
is as follows.
60C for 1.5 to 2 h. Add the methanol, mix thoroughly,
allow to stand and decant. Store the sediment-free stock so- Preparation
lution at room temperature. For use, dilute l part of the Make smears, impressions of the cut surfaces of tissues, or
stock solution with 40 to 50 parts of distilled water or 0.5 M spreads of tissue scrapings on alcohol-cleaned slides.
phosphate buffer at pH 6.8. Distilled water and aqueous reagents must be filter steril-
Procedure ized, and the use of vessels and moist chambers (petri dish
with moist filter paper) must be regulated to avoid any bac-
1. Air dry the specimen on a glass slide, fix in absolute terial contamination of the slides from either other speci-
methanol for 5 min, and again air dry. mens or the environment, which may harbor members of
2. Cover the slide with freshly diluted Giemsa stain the Legionellaceae.
for 1 h.
3. Rinse the slide in 95% ethanol to remove any excess Procedure
dye, and air dry. For a smear, air dry and gently heat fix on a slide. For a
4. Examine under oil immersion for the presence of ba+ spread of tissue cells and impressions, fix in filter-sterilized
sophilic intracytoplasmic bacteria which will be small, pleo- 10% neutral formalin (add C a C 0 3 to the bottle, and let it
morphic, and purple. settle). Add enough of the appropriate conjugated antibody
to spread over the entire surface of the smear. Use a moist
2.4.3.2. Gimenez Staining Method chamber to prevent drying during the 20- to 30-min reac-
tion at room temperature. Immerse the slide, first in buffered
Stock Solution saline and then in distilled water. Drain, mount the smear in
a drop of buffered glycerol (9 volumes of glycerol, 1 volume
Basic fuchsin ........................... 10 g of 0.5 M NaHC03-Na2C03buffer [pH 9.0]), add a cover-
Ethanol, 95% (vol/vol) ................... 100 ml slip, and then seal with clear nail polish. Examine with a flu-
Phenol, 4% (wt/vol) aqueous . . . . . . . . . . . . . . 250 ml orescence microscope, preferably by epi-illumination (chap-
Distilled water ......................... 650 ml ter l ) , with appropriate exciting and barrier filters for the
32 w MORPHOLOGY A N D ULTRASTRUCTURE

fluorochrome in use. Useful practical hints and a more ex- 7. Herbert, R. A. 1990. Methods for enumerating microor-
tensive discussion of components are given by McKinney ganisms and determining biomass in natural environ-
and Cherry (1 1). ments. Methods Microbiol. 22:l-39.
A useful review and assessment of approaches to estimating bio-
muss in environments.
2.5. SOURCES OF STAINING REAGENTS 8. Karl, D. M. 1986. Determination of in situ biomass, via-
Most of the laboratory requirements are available from the bility, metabolism, and growth, p. 85-176. In J. S.
supply companies, but some stains are not widely stocked. E. Poindexter and E. R. Leadbetter (ed.), Bacteria in Nature,
Merck, Darmstadt, Germany, is a major source of biological vol. 2. Plenum Press, New York, NY.
Descriptions and assessments of the methods applied to deter#
stains, and this multinational company has acquired many mination of biomass and viable counts in the field.
of the suppliers of former times: BDH Chemicals, Harleco, 9. Lawrence, J. R., D. R. Korber, and G. M. Wolfaardt.
Hopkin &Williams, J. T. and E. Gum Co., and Raymond A. 2001. Digital image analyses of microorganisms. In G.
Lamb Co. A wide range of stains and other reagents are Bitton (ed.), Encyclopedia of Environmental Microbiology,
available through E. Merck subsidiaries. General staining John Wiley and Sons, New York, NY.
reagents (e.g., Gram stain), glass slides, and coverslips, etc., A useful review with clear examples of the applicationof imge-
are provided by a number of scientific suppliers. Merck sub- processing and analysis techniques.
sidiaries and general suppliers are as follows. 10. Lillie, R. D. 1977. H. A. Conns Biological Stains, 9th ed.
BD, Franklin Lakes, NJ (supplier of BBL reagents) The Williams & Wilkins Co., Baltimore, MD.
An extensive compendium of stains and dyes with discussion
BDH Chemicals, Toronto, Ontario, Canada of their chemical structures and applications. Also see refer-
BDH Chemicals, Poole, Dorset, United Kingdom ence 4 .
Difco Laboratories, Detroit, MI 11. McKinney, R. M., and W. B. Cherry. 1985. Immun-
ofluorescence microscopy, p. 891-897. In E. H. Lennette,
EM Science, Fort Washington, PA (in Canada, Cedarlane A. Balows, W. J. Hausler, Jr., and H. J. Shadomy (ed.),
Labs Ltd., Hornby, Ontario) Manual of Clinical Microbiology, 4th ed. American Society
Fisher Scientific Ltd., Pittsburgh, PA (in Canada, Nepean, for Microbiology, Washington, DC.
Ontario) A useful source of advice on the practice of immunofluores-
Merck Japan Ltd., Tokyo, Japan cence microscopy.
Sigma-Aldrich Canada, Oakville, Ontario 12. Murray, R. G. E., R. N. Doetsch, and C. F. Robinow.
1994. Determinative and cytological light microscopy, p.
Many of the newer fluorescent stains can be obtained 21-41. In P. Gerhardt, R. G. E. Murray, W. A. Wood, and
through Molecular Probes, Inc., Eugene, OR (www.probes N. R. Kreig (ed.), Methods for General and Molecular
.corn). Bacteriology, ASM Press, Washington, DC.
A more expanded source for observation of live culures grown
on agar, for chemical fixations, and for nucleoid visualization.
2.6. REFERENCES We highly recommend that you read this chapter!
13. Newell, S. Y., R. D. Fallon, and P. S. Tabor. 1986. Direct
2.6.1. General References microscopy of natural assemblages, p. 1-48. In J. S.
Poindexter and E. R. Leadbetter (ed.), Bacteria in Nature,
1. Beveridge, T. J. 2001. Use of Gram stain in microbiology. vol. 2. Plenum Press, New York, NY.
Biotech. Histochem. 76:111-118.
A survey with useful references to studies in the field.
2. Brock, T. D. 1983. Membrane Filtration: a Users Guide and
Manual. Science Tech Inc., Madison, WI. 14. Norris, 3. R., and D. W. Ribbons (ed.). 1971. Methods in
Microbiology, vol. 5A. Academic Press, Inc., New York,
A complete guide and instruction in the use and interpretation
of membrane filter techniques.
NY.
3. Brock, T. D. 1987. The study of microorganisms in situ:
This is an old but useful volume; the material presented in the
first part is particularly useful as a source of information sup-
progress and problems, p. 1-17. In M. Fletcher, T. R. G. plementary to that presented here. The relevant chapters are as
Gray, and J. G. Jones (ed.), Ecology of Microbial
Communities. Cambridge University Press, Cambridge, follows.
United Kingdom. I. Microscopy and micrometry, by L. B. Quesnel,
An assessment of the applications of counting techniques in mi- p. 1-103.
crobial ecology. 11. Staining bacteria, by J. R. Norris and H. Swain, p.
4. Clark, G. 1973. Staining Procedures Used by the Biological 105-134.
Stain Commission, 3rd ed. The Williams & Wilkins Co., 111. Techniques involving optical brightening agents,
Baltimore, MD. by A. M. Paton and S. M. Jones, p. 135-144.
A resource for methods used in association with H. A. Conns IV. Motility, by T. Iino and M. Enomoto, p. 145-163.
Biological Stains ( 10). 15. Pickup, R. W. 1991. Development of molecular methods
5. Hall, G. H., J. G. Jones, R. W. Pickup, and B. M. for the detection of specific bacteria in the environment.
Simon. 1990. Methods to study the bacterial ecology of J. Gen. Microbiol. 137:1009-1019.
freshwater environments. Methods Microbiol. 23: 181-210. A discussion of the strategiesfor sampling and detecting bacte-
A modern compilation of methods for bacteriologicalassessment ria in the environment with special reference to genetically mod-
of freshwaters. ified bacteria.
6. Hendrickson, D. A., and M. M. Krenz. 1991. Reagents 16. Robinow, C. F. 1960. Morphology of bacterial spores,
and stains, p. 1289-1314. In A. Balows, W. J. Hausler, Jr., their development and germination, p. 207-248. In I. C.
K. L. Herrmann, H. D. Isenberg, and H. J. Shadomy (ed.), Gunsalus and R. Y. Stanier (ed.), The Bacteria, vol. 1.
Manual of Clinical Microbiology, 5th ed. American Society Academic Press, Inc., New York, NY.
for Microbiology, Washington, DC. A classic account of bacterial spores and their study using light
A part of this chapter gives details of staining methods useful in microscopy. The same volume contains other illustrated articles
clinical microbiology. on bacterial structure.
2. Sampling and Staining for Light Microscopy 33

17. Robinow, C. F. 1975. The preparation of yeasts for light 34. Hotchkiss, R. D. 1948. A microchemical reaction result-
microscopy. Methods Cell Biol. 11:1-22. ing in the staining of polysaccharide structures in fixed tis-
Practical advice on using gelatin-agar slide cultures. sue preparations. Arch. Biochem. 16:131-141.
18. Rosebrook, J. A. 1991. Labeled-antibody techniques: flu- 35. Jowr, F., and P. LeBaron. 1997. Ecological implications of
orescent, radioisotopic, and immunochemical, p. 79-86. In an improved direct viable count method for aquatic bacte-
A. Balows, W. J. Hausler, Jr., K. L. Herrmann, H. D. ria. Appl. Environ. Microbiol. 63:3643-3647.
Isenberg, and H. J. Shadomy (ed.), Manual of Clinical 36. Leifson, E. 1951. Staining, shape, and arrangement of
Microbiology, 5th ed. American Society for Microbiology, bacterial flagella. J. Bacteriol. 62:377-389.
Washington, DC. 37. Lisle, J. T., P. S. Stewart, and G. A. McFeters. 1999.
The applications of immunomicroscopy in clinical microbi- Fluorescent probes applied to physiological characteriza-
ology tion of bacterial biofilms. p. 166-178. In R. J. Doyle (ed.),
Biofilms: Methods in Enzymology. Academic Press, New
2.6.2. Specific References York, NY.
19. Barer, R., R. F. A. Ross, and S. Thczk. 1953. Re- 38. Lundgren, B. 1981. Fluorescein diacetate as a stain of
fractometry of living cells. Nature (London) 171:720-724. metabolically active bacteria in soil. Oikos 36: 17-22.
20. Bartholomew, J. W. 1962. Variables influencing results, 39. Murray, R. G. E., and J. F. Whitfield. 1956. The effects
and the precise definition of steps in Gram staining as a of the ionic environment on the chromatin structures of
means of standardizing the results obtained. Stain Technol. bacteria. Can. J. Microbiol. 2:245-260.
37~139-155. 40. Newell, S. Y., and R. R. Christian. 1981. Frequency of
21. Bartholomew, J. W., and T. Mittwer. 1952. The Gram dividing cells as an estimator of bacterial productivity.
stain. Bacteriol. Rew. 16:l-29. Appl. Environ. Microbiol. 42:23-31.
22. Beveridge, T. J. 1997. The response of S-layered bacteria 41. Noller, E. C., and N. N. Durham. 1968. Sealed aerobic
to the Gram stain. FEMS Microbiol. Rev. 20:2-10. slide culture for photomicrography. Appl. Microbiol.
23. Beveridge, T. J. 1990. Mechanism of gram variability in 16:439-440.
select bacteria. J. Bacteriol. 172:1609-1620. 42. Popescu, A., and R. J. Doyle. 1996. The Gram stain after
24. Beveridge, T. J., and S. Schultze-Lam. 1997. The re- more than a century. Biotech. Histochem. 71:1415-1451.
sponse of selected members of the Archaea to the Gram 43. Robinow, C., and E. Kellenberger. 1994. The bacterial
stain. Microbiology 142:2887-2895. nucleoid revisited. Microbiol. Rev. 58:211-232.
25. Beveridge, T. J., and J. A. Davies. 1983. Cellular re- 44. Robinow, C. E., and R. G. E. Murray. 1953. The differ-
sponses of Bacillus subtilis and Escherichia coli to the Gram entiation of cell wall, cytoplasmic membrane and cyto-
stain. 1. Bacteriol. 156:846-858. plasm of Gram-positivebacteria by selective staining. Exp.
26. Buttner, M. P., K. Willeke, and S. A. Grinshpun. CeU Res. 4:390-407.
1996. Sampling and analysis of airborne microorgan- 45. Rogers, F. G., and A. W. Pasculle. 1991. Legionella, p.
isms, p. 629-640. In C. J. Hurst, G. R. Knudsen, M. 442453. In A. Balows, W. J. Hausler, Jr., K. L. Herrmann,
McInerney, L. D. Stetzenbach, and M. V. Walter (ed.), H. D. Isenberg, and H. J. Shadomy (ed.), Manual of Clinical
Manual of Environmental Microbiology. ASM Press, Microbiology, 5th ed. American Society for Microbiology,
Washington, DC. Washington, DC.
27. Davies, J. A., G. K. Anderson, T. J. Beveridge, and 46. Salton, M. R. J. 1963. The relationship between the na-
H. C. Clark. 1983. Chemical mechanism of the Gram ture of the cell wall and the Gram stain. J. Gen. Microbiol.
stain and synthesis of a new electron-opaque marker for 30:223-235.
electron microscopy which replaces the iodine mordant of 47. Schaeffer, A. B., and M. Fulton. 1933. A simplified
the stain. J. Bacteriol. 156:837-845. method of staining endospores. Science 77:194.
References 22 through 25 and 27 provide an upto-date expla- 48. Seto, S., G. Layh-Schmitt, T. Kenri, and M. Miyata.
nation of how the Gram stain works and why some prokaryotes 2001. Visualization of the attachment organelle and cytoad-
stain in an unexpected way. Also see references 42 and 46. herence proteins of Mycophma pneumoniae by immunoflu-
28. Dorner, W. 1926. Un prockd6 simple pour la coloration orescence microscopy.J. Bacteriol. 183:1621-1630.
des spores. h i t 6:8-12. 49. Tabor, P. S., and R. A. Neihof. 1982. Improved method
29. Duguid, J. P. 1951. The demonstration of bacterial cap- for determination of respiring individual microorganisms
sules and slime. J. Pathol. Bacteriol. 63:673. in natural waters. Appl. Environ. Microbiol. 43:1249-1255.
30. Grossart, H..P., G. E Steward, J. Martinez, and E Azam. 50. Traxler, R. W., and J. L. Arceneaux. 1962. Method for
2000. A simple, rapid method for demonstrating bacterial staining cells from small inocula. J. Bacteriol. 84:380.
flagella. Appl. Environ. Microbiol. 66:3632-3636. 51. Truant, J. P., W. A. Brett, and W. Thomas. 1962.
31. Heimbrook, M. E., W. L. L. Wang, and G. Campbell. Fluorescence microscopy of tubercle bacilli stained with
1989. Staining bacterial flagella easily. J. Clin. Microbiol. auramine and rhodamine. Henry Ford Hosp. Med. Bull.
27~2612-2615. 10:287-296.
32. Henrichsen, J. 1972. Bacterial surface translocation: a sur- 52. Vela, G. R., and 0. Wyss. 1964. Improved stain for visual-
vey and a classification. Bacteriol. Rev. 36:478-503. ization of Azotobacrer encystment. J. Bacteriol. 87:476-477.
33. Hobble, J. E., R. J. Daley, and S. Jasper. 1977. Use of 53. Wilson, D. R., and T. J. Beveridge. 1993. Bacterial fla-
Nuclepore filters for counting bacteria by fluorescence mi- gellar filaments and their component flagellins. Can. J.
croscopy. Appl. Environ. Microbiol. 33:1225-1228. Microbiol. 39:451-472.
Laser Scanning Microscopy
J. R. LAWRENCE AND T. R. NEU

3.1. INTRODUCTION.. ....................................... 34


.2. INSTRUMENTATION ..................................... 35
3.2.1. Lasers .............................................. 35
.2. Scanning and Detection Systems........................... 35
.3. Microscopes and Objective Lenses.......................... 36
3.3. PERFORMANCE ASSESSMENT AND GENERAL OPERATION ..... 37
.4. SPECIAL CONSIDERATIONS FOR 2P IMAGING ................ 39
.5. SAMPLE PREPARATION OPTIONS ........................... 40
3.5.1. Correlative Microscopy ................................. 40
3.6. FLUORESCENCE AND FLUORESCENT PROBES ................ 40
.7. LSMIMAGING ........................................... 41
3.7.1. Scanning in the xy Direction.............................. 41
.2. Scanning in the a Direction.............................. 43
3.8. 3-DIMAGING ........................................... 45
.9. MULTIPLE-PARAMETER IMAGING .......................... 45
3.9.1. Lectin Staining ....................................... 46
.2. Imaging of the Metals Nickel and Cadmium................... 46
3.10. DYNAMIC IMAGING ...................................... 46
.11. HANDLING 3-D DATA SETS ................................ 48
3.1 1.1. Quantification ....................................... 49
.2. Deconvolution ....................................... 50
3.12. LSM SAMPLING FREQUENCY .............................. 50
.13. LSM IMAGE ARCHIVING AND PRINTING .................... 50
.14. REFERENCES ............................................ 51

3.1. INTRODUCTION plane. This stray light results in degradation of the image
Light microscopy provides a principal method for the ob- quality and interferes with three-dimensional (3-D) repre-
servation and study of bacterial cells, aggregates, biofilms, sentation of the object. Both types of LSM, confocal or 1P
and communities. Conventional light microscopy tech- and 2P or multiphoton systems, act to eliminate light not
niques, including phase contrast, dark field, differential in- originating from the focal plane, resulting in a high-clarity
terference contrast, and fluorescence, all provide effective image of the specimen that may be considered an optical
means to observe bacterial cells in pure cultures and water thin section. The 1P and 2P optical microscopes may be
samples, on surfaces, and in sediments, with or without used to view virtually any specimen or preparation suitable
staining. In addition to these methods, there have been for conventional light and epifluorescence microscopy.
substantial innovations in light microscopy through the in- However, 1P and 2P microscopy is most frequently used in
tegration of conventional microscope optics, a scanned conjunction with fluors or fluor-conjugated probes in the
laser beam, confocal optics, and digital image formation. fluorescence mode to achieve optimal results. The option
Confocal laser scanning microscopy (CLSM) with one- of using reflection imaging to reveal minerals or colloidal
photon (1P) excitation became a practical technique with gold labels also exists but only for 1P excitation. The digi-
the introduction of the first commercially available models tal nature of the LSM images makes them amenable to
in the early 1980s. Laser scanning microscopes with two- image processing and analysis, allowing the user to obtain
photon (2P) excitation have been commercially available quantitative information or to make 3-D reconstructions of
since 1997. Since that time, laser scanning microscopy the material under study.
(LSM) technologies have been applied in many fields. The ultimate goal in the application of LSM is to study
Valkenburg et al. (55) published the initial study using and integrate physiological,biochemical, and molecular as-
CLSM to examine the nucleoid of Escherichia coli, and pects of living microbial cells. This goal requires the devel-
Lawrence et al. ( 3 3 ) published an extensive study of bacte- opment and application of noninvasive techniques to
rial biofilms. probe both cellular and extracellular processes of bacterial
In a conventional microscope, all the light passing cultures and naturally occurring assemblages of bacteria, in-
through a specimen or emanating from it is imaged directly cluding aggregates and biofilms.
and simultaneously, forming an image. However, all light The purpose of this chapter is to provide a basis for un-
from the illuminated region returns to the eye or the de- derstanding LSM and the approaches that may be used to
tector, including light from above and below the focal apply high-resolution digital microscopy to the study of

34
3 . Laser Scanning Microscopy 35

bacteria. Additional reviews of image analysis, digital imag- 3.2.1. Lasers


ing, and LSM for microbiology applications are also avail- Both 1P and 2P systems are equipped with a laser light
able (10, 27, 30, 32). source(s) that generates a range of excitation wavelengths.
Commercially available lasers for confocal 1P microscopy
include argon ion, helium-neon, argon-krypton, helium-
3.2. INST R UMENTATl0N cadmium, and UV varieties. The argon laser (25-, 50-, or
Modern 1P and 2P systems are complicated, sensitive, and 100-mW models with 5,000- to 10,000-h life spans) provides
expensive with multiple components, including lasers, de- two main excitation lines at a 488-nm wavelength (the blue
tectors, optical systems (glass or mechanical), step motors, line) and a 514-nm wavelength (the green line) as well as
and computer hardware and software. Many fundamental minor lines with wavelengths ranging from 274 to 528 nm.
aspects of CLSM are covered in various books (15,46), and To overcome limitations of the argon laser (i.e., limited ex-
readers are referred to these. The major manufacturers pro- citation, little separation of excitation lines, and limited flu-
vide a variety of configurations, including real-time in- orochrome selection), most systems are equipped with addi-
struments, which provide video rate image collection but tional helium-neon lasers (543- or 633-nm-wavelength line)
not high-resolution optical sectioning, and point scanning or mixed-gas argon-krypton lasers (488-nm [blue]-, 568-nm
systems, which maximize lateral and axial resolution at the [yellowl-, and 647-nm [red]-wavelength lines). The shift to
expense of image collection speed. The latter group may be alternate laser sources such as the Ar-Kr laser provides for
subdivided into the 1P CLSM which uses classical physical excitation of up to three fluorochromes with little spectral
barriers to remove out-of-focus light (pinholes or apertures) emission overlap. The addition of a UV laser (e.g., 351- to
and those multiphoton or 2P systems where the optical sec- 364-nm wavelength) further expands the excitation poten-
tioning is based on the physics of light absorption in the tial and the range of fluors. However, there are additional
focal plane. All of these systems may use either a standard safety and operational considerations, for example, the need
or an inverted light microscope as the fundamental plat- for quartz optics if excitation is performed using a 150-nm
form; some LSM systems are designed to be easily ex- wavelength. A development to watch is the appearance of
changed between microscopes. Depending on the manufac- diode lasers that offer specific excitation lines. For example,
turer and the intended applications, there are options a blue diode laser provides near-UV emission, allowing for
including multiple laser attachments, multiple- or single- excitation of DAPI (4,6-diamidino-Z-phenylindole) with-
pinhole arrangements, and a selection of photomultiplier out a UV laser. 2P LSM systems employ a high-peak-power,
tubes (PMTs). PMTs may have various levels of light sensi- infrared laser with an extremely short pulse in the femto-
tivity and response to specific emission wavelengths. to picosecond range and a repetition rate of about 80 MHz.
Additional equipment considerations include the host com- This laser produces the high photon density required to
puter and software (both of which are usually predefined by achieve the 2P effect of exciting the fluor only in the focal
the company) as well as an archiving system (e.g., compact plane of the microscope lens. Current 1P and 2P systems use
disks [CDs]) and vibration isolation tables. The room itself a fiber-optic system to connect the laser to the scan head
should also be well isolated from vibration, be air condi- of the microscope, greatly simplifying connection and
tioned, and have variable, controlled lighting levels. alignment.
In general, experience in light microscopy and fluores- 3.2.2. Scanning and Detection Systems
cence microscopy is a valuable starting point for LSM.
The core of the LSM system is the scan head, an integrated
Training is provided by the equipment manufacturers, and a
unit with galvanometric or acoustically controlled mirrors
variety of courses are offered worldwide for confocal 1P and
to scan the excitation wavelengths of the laser over the
2P microscopy and the specific applications. The major specimen. The laser light source is used to illuminate the
focus of most courses is cell biology, although the informa-
specimen in a point-by-point fashion (point scanning)
tion is of use and techniques are broadly transferable across
through a conventional objective lens, thereby exciting the
disciplines.
fluorescent probes that have been applied to the specimen
Although some equipment alignments may be done by
or creating a reflection image. The specimen may be
the user, in general these systems require technical adjust-
scanned in a raster pattern at resolutions equal to 512 by
ments by factory-trained engineers on at least an annual
512, 512 by 768, or 1,024 by 1,024 pixels or at even higher
basis. In most of the current LSM systems, there are really
resolution (new systems can do 4,096 pixels). Scan time
no user-repairable parts. It is, however, important to do the
may vary from a fraction of a second to several seconds.
following.
Note that the more rapid the scan, the lower the resolution
1. Establish user protocols for training and start-up and of the image, whereas the longer the scan, the greater the
shutdown of the instrument. photobleaching (destruction of a fluor by light, resulting in
2. Keep track of instrument use. a loss of signal). When a specimen is scanned, one or more
3. Keep a log of all the error messages displayed and their confocal pinholes allow only those fluorescence signals that
circumstances. arise from a focused xy plane to be detected by the PMT.
4. Check the alignment of the system periodically, espe- The pinholes are positioned confocally in front of the inci-
cially during extended use, and always at the beginning of dent laser light and in front of the photomultiplier detector,
each session. preventing fluorescent signals originating from above,
5. Log the results of alignment checks. below, or beside the point of focus from reaching the pho-
6. Periodically use reference samples and reference im- todetector. In contrast to 1P systems, 2P microscopy
ages relevant to the research area so as to assess instrument achieves the selection of light from a focused xy plane be-
performance. cause excitation occurs only in the true focal plane.
7. Ensure annual assessment and alignment to the man- The selection of the fluorescence wavelengths to be de-
ufacturers specifications. tected may be accomplished by using a beam splitter and
36 rn MORPHOLOGY A N D ULTRASTRUCTURE

optical glass filter sets. However, it is also possible to select This procedure can also increase resolution; e.g., a 20x,
excitation-emission wavelengths without the need for opti- 0.75-NA lens will increase in resolution up to approxi-
cal lenses through the application of a filter-free, prism mately four times during a zoom. In LSM the recorded dig-
spectrophotometer-based system. This approach reduces the ital image covers only the central part of the visual field of
loss of signal and frees the user to optimize the detection of the objective lens. Errors in the objective lens increase with
emission peaks, which can be very useful when more than the distance from the center; thus, it is better to use a high-
one fluorescent stain or probe is used. Regardless of the NA 60X lens and zoom than it is to use a similar lOOX lens
excitation-emission system, a PMT detects the emitted without zooming. Some of the issues associated with objec-
light at each point and converts this into a digital grayscale tive lens quality and corrections are illustrated in Color
image. Although custom PMTs are available for far-red im- Plate 4. Color Plate 4A shows an image of 6-pm-diameter
aging and potential exists for application of PMTs with var- Focal Check beads (Molecular Probes, Eugene, OR) stained
ious spectral ranges from 185 to 930 nm, the range of PMTs with green, red, and blue fluors and viewed with a 60X, 1.4-
in commercial LSM systems has been limited. One critical N A lens. The image shows a white peripheral ring around
limit is that the LSM PMTs generally have a response only the beads, indicating good alignment and correction of the
to changes in image brightness of one order of magnitude. lens and LSM system. The loss of resolution when the
1P and 2P systems may also incorporate either a fiber-optic image is formed by scanning in the xz direction is shown in
light guide below the condenser and connected to one of Color Plate 4B. Here effects of light wavelength and point
the PMTs or a separate photodiode for imaging of noncon- spread function error create distortion and misalignment of
focal transmitted laser images. images from the three channels. Panel C shows the effect of
using a 63 X, 0.9-NA lens on the appearance of the same
3.2.3. Microscopes and Objective lenses beads in the xy plane, and the further-reduced quality of the
Additional details regarding objectives and microscope ba- xq image is shown in panel D. When the same beads are
sics for light microscopy are found in chapter 1. Selection of viewed by using the outer edge of the 63X lens, the effect
the base microscope for LSM usually represents a compro- of the loss of correction with the distance from the central
mise. Inverted microscopes offer advantages such as easy ac- region of the scanned area is easily seen (Color Plate 4E). In
cess to the sample for application of extraneous items such panel F, the reader can see the loss of confocality (presence
as microelectrodes, whereas the upright microscope offers of multiple planes and bead colors) and chromatic correc-
the full range of objective lenses including water-immersible tion in a 20X, 0.4-NA lens.
ones and gives the possibility of viewing the sample without The thickness of an optical section obtained by confocal
a glass interface such as a coverslip. Some of the current 1P or 1P LSM is dependent on the NA of the objective lens
and 2P systems offer the convenience of being relatively and the diameter of the pinhole(s). In general, high-NA
easily switched between inverted- and upright-base micro- objectives such as those with NAs of 1.0 to 1.4 allow the use
scopes (however, in practice loss of alignment may be a of lower pinhole or aperture settings, thus generating sub-
problem, especially with UV lasers). micron optical sections. It is complicated to determine the
The primary imaging tool is the objective lens, and its exact thickness of an optical section. Neu et al. (44) used
selection is based on the type of sample, sample preparation, beads of known dimensions to show that a 40X, 0.55-NA
required resolution, and base magnification. For 1P and 2P water-immersible lens created an optical thin section that
microscopes, the major limitation for all objective lenses is was 5 pm thick when the system was optimized for the pin-
that the axial or pdimension resolution is poor relative to hole and PMT, etc. In contrast, based on the calculations of
the lateral resolution (Color Plate 4A and B). Older objec- Xiao and Kin0 (61), the theoretical thickness of an optical
tive lenses are not corrected for imaging in the far red, and section taken with this lens was 4 pm.
when wavelengths are extended into the UV range there Lenses with an N A lower than 0.5 will not provide con-
may also be losses of transmission and serious image aberra- focal images (Color Plate 4F). However, there are lenses
tion. New lenses provided by most manufacturers and avail- capable of extralong working distances (ELWDs) and water-
able for LSM are corrected from -350 to 1,000 nm, over- immersible lenses with NAs of 0.5to 0.9 that are particu-
coming these considerations. In general, one should use larly useful for imaging environmental samples. These
high-numerical-aperture (NA) oil or water immersion lenses offer additional advantages, particularly for living
lenses, i.e., those with NAs of 1.2 to 1.4. Another critical specimens, e.g., no coverslip, long working distance, high
factor in considering objective lenses is the working dis- NA, and superior brightness. There are a variety of high-
tance, i.e., that range of distances where the image formed N A water-immersible objectives supplied by the major
by the lens is clearly focused. Working distances may be as manufacturers (i.e., Leica, Nikon, Olympus, and Zeiss).
small as <I0 pm for some high-NA plan apochromat For fixed specimens, factors such as the refraction prop-
lenses, and some water immersion lenses (e.g., a 63X, 1.2- erties of mounting and immersion media must be consid-
N A lens) have been designed for LSM applications allow- ered since they will diminish signal intensity and resolution.
ing imaging through up to 220 pm of biological material. Therefore, both media should resemble each other in terms
Application of these water immersion lenses, which are of their optical properties and be matched to the objective
very expensive, assumes the use of fixed and stained speci- lens. See also chapter 1.
mens, optically appropriate mounting media, and high- Although penetration of biological specimens is greatly
quality coverslips (Corning Glass Works and NalgeNunc enhanced by 1P and 2P microscopy, it is still a function of
are suppliers of suitable coverslips and coverslip chambers) the transparency of the specimen, self-shading, diffraction
and proper adjustment of the objective lens as well as cor- by objects, and quenching of both the excitation and emis-
rection for the coverslip thickness and temperature. sion light by the specimen and the presence of the fluors.
Generally, as the N A increases, the working distance of the Solid surfaces may yield information from only the surface
objective lens decreases. The user should also consider that layer, whereas some biological materials such as bacterial
in LSM it is possible to zoom in while scanning, thereby in- biofilms may be imaged to a depth of about a millimeter. In
creasing magnification without changing the objective lens. 1P LSM applications, it is possible to effectively section
3. Laser Scanning Microscopy 37

TABLE 1 Advantages and disadvantages of CLSM


Advantages Disadvantages
Examination of fully hydrated samples up to several hundred micrometers Fluorescence of background
thick (dependentupon the objective lens selected)
Noninvasive optical sectioning with virtually no out-of-focus blur Bleaching in out-of-focusarea
xy, xz, and xt sectioning Cell damage in out-of-focus area
Fluorescence mode and reflection mode available Potential for chromatic aberration if UV laser is used
Simultaneous application of multiple probes and Depth of laser penetration
multichannel (4) imaging of digitally enhanced signals
Quantitative static and dynamic analyses of 3-D data sets, 3-D Low xz resolution
tomography, multicolor stereoscopic imaging, and computer animation

through several hundred micrometers with 63 X, 0.9-NA 1. The system should be allowed to warm up for -30
water-immersible lenses and up to 1 mm (in exceptional min prior to imaging. This warm-up is particularly critical
cases) when using ELWD lenses, for example, 20X ELWD with Ar-Kr systems to ensure a stable far-red (647-nm-
or 4 0 X ELWD or 40X, 0.55-NA water-immersible lenses. wavelength) excitation line.
Infrared illumination is less subject to scattering by biologi- 2. Internal mirrors and optical elements of the unit
cal specimens than visible light, which accounts for the should be periodically (annually) aligned by the manufac-
greater penetration observed with 2P LSM systems. Sytsma turer.
et al. (52) were the first to apply 2P excitation in a study of 3. Alignment must be confirmed prior to each use.
oral biofilms where they reported that a dense, mixed- Alignment is particularly critical for work with bacteria,
species biofilm of 100-p m thickness could be imaged after where a misalignment of 0.5p m cannot be ignored. Users
staining with acridine orange. However, the fluorescence should make up slides consisting of fluorescent beads of var-
decreased rapidly as a function of depth, and to counter ious intensities and Focal Check beads (Molecular Probes)
this effect the excitation intensity had to be adjusted (52). for routine evaluation of image brightness and alignment.
A subsequent study indicated that in the same sample, Focal Check beads allow assessment of images obtained from
there was a penetration of 70 ym for 2P excitation versus the same location but with different excitation-emission
15 y m for 1P excitation in oral biofilms stained with rho- combinations (multiparameter imaging or colocalization
damine B (57). A comparison of the advantages and disad- studies), ensuring that they are in perfect register. Color
vantages of 1P and 2P LSM systems is presented in Tables Plate 4 is an image of Focal Check beads showing the ap-
1 and 2. Figure 1 shows a head-to-head comparison of 1P pearance in the green, red, and far-red channels of a laser
and 2P LSM imaging of a thick biofilm specimen stained scanning microscope and the combined image from the
with SYBR green (Molecular Probes), illustrating the in- three channels.
creased resolution of the 2P system. 4. A variety of fluorescent beads ranging in diameter
from 100 nm to 1 mm should be used to assess both image
quality and resolution and to check the calibration of the
3.3. PERFORMANCE ASSESSMENT instrument.
A N D GENERAL OPERATION 5. Adjustment of the computer monitor should be car-
There are a number of points to consider when setting up ried out periodically under normal working and lighting
and operating an LSM system. conditions. A monitor test image (both color and black and

TABLE 2 Advantages and disadvantages of 2P LSM


Advantages Disadvantages
Excitation in the focal plane only with extremely small Whole spectrum of infrared light is available
excitation volume (femtoliter) only with 3 different mirror sets (700-1,200 nm)
Uncaging in extremely localized spots Tuning of infared laser is necessary
No out-of-focus bleaching Tuning determines the types of fluorochromes which can be excited
No out-of-focus cell damage
No background fluorescence
High depth penetration (0.5-2 mm)
Inherent depth resolution
Less scattering Resolution is slightly lower than that with CLSM
No pinholes
No expensive UV laser
No UV photo damage
No UV optics necessary
Fewer filter Droblems
38 1 MORPHOLOGY AND ULTRASTRUCTURE

FIGURE 1 1P and 2P LSM images of a fluidized bed reactor biofilm which degrades EDTA and is growing on pumice. The spec-
imen was stained with SYBR green, and serial sections were taken by using both LSM systems on the same sample sequentially.
Images show enhanced resolution of the 2P system relative to that of the 1P system with a 39-~m-thickbiofilm specimen.

white) should be displayed, and the brightness and contrast when viewed with epifluorescence or preliminarily scanned
controls should be adjusted until all intensities are visible with LSM.
and well defined. The monitor image should also be cen- 11. Switch to the 1P or 2P mode, and scan the sample by
tered and sized correctly. using a low zoom factor, with the PMT gain set at a low to
6. Periodically check the step motor and calibration in midrange level and, for 1P, with the pinhole aperture about
the 2 dimension to ensure that the stage motion corre- half open. Then, while scanning in real time, adjust the
sponds to that indicated by the software. This setting may gain of the PMT so that no part of the image is saturated
also be assessed by using a range of sizes of fluorescent beads (white or other color depending on the LUT used).
and coverslips of known thickness. 12. Optimize the intensity of the incident laser light such
7. Ensure that the correct optical filters or settings are in that it provides a strong fluorescent signal while minimizing
place for the fluorochromes to be used. In the event that the signal loss due to photobleaching. Laser intensity is usually
microscope is equipped with a filter-free, prism spectropho- controlled by using neutral-density filters or acoustic settings.
tometer-based system, the software allows the selection of 13. The size of the pinhole should be adjusted to achieve
an infinite range of excitation-emission lines, optimization the smallest aperture possible. This may involve a sequence
of the signal, and signal separation. of adjustments with progressive reduction in aperture size
8. The PMTs should be adjusted when there is no in- (note that larger apertures produce thicker optical sec-
coming signal so that their black level is at the point where tions). With some instruments, setting the Airy disk for
the boundary between scans is invisible against the dark each objective lens to 1 sets the optimal pinhole.
background. Furthermore, the gain and black level of the 14. By adjusting or balancing the PMT gain, laser inten-
PMTs should be adjusted to utilize the entire gray scale. sity, and pinhole size, the user will also establish the optimal
Note that optimum brightness in an image usually occurs conditions for a specific combination of fluorochrome( s)
when only a few pixels of the image approach saturation (a and/or specimen. Various fluorochromes also have different
gray level of 255 on a scale of 0 to 255 where 0 is black and quantum yields, and yield may change with the nature and
255 is white). Most of the commercial LSM systems include intensity of the excitation light source. It is important to
a digital indicator, so-called lookup tables (LUTs), where keep this in mind when fluorescent stains are added in com-
under- and oversaturated pixels appear in different colors, bination and the specimen is imaged by using more than
facilitating adjustments. one excitation wavelength. Figure 2 illustrates the excita-
9. Ensure that the objective lens used for a particular tion-emission spectra of various fluors and their potential
analysis has sufficient magnification, resolving potential, for interaction during imaging. Figure 2A illustrates the
and working distance. One also has to consider the nature ideal case, and Fig. 2B shows the potential for signal over-
of the sample, the presence or absence of a coverslip, and lap and bleed through with the use of more than one fluor
the type of mounting media, etc. in a sample.
10. Place a sample on the microscope specimen stage
and select a field for imaging using either standard epifluo- The above list assumes proper preparation of both the
rescence or phase-contrast microscopy. The ideal field for samples and the system, each of which is required to achieve
initial setup should provide a fairly even signal response high-quality, information-packed images.
3. Laser Scanning Microscopy 39

B
I I I
I I I
I I I
I I I
I I I
I I I
I I I

..
1
:-.,EX I I
I
I I

400 500 600 700


Wavelength (nm)
FIGURE 2 Graphs showing the nature of excitation-emissionspectra for an ideal series of green, red, and far-red-emittingfluors
(A) and problematic fluors where excitation (Ex) and emission (Em) peaks overlap, resulting in potential cross talk or bleed
through of the signal into adjacent imaging windows (B).

tion of suitable fluorochromes for microbiological samples.


3.4. SPECIAL CONSIDERATIONS In principle, it is possible to use the same fluorochromes
FOR 2P IMAGING that have been developed for 1P excitation. However, in
The microscopic application of 2P excitation is still not comparison to those for 1P excitation, the fluors for 2P ex-
fully explored and evaluations of basic protocols are neces- citation have a broader excitation cross-section. That
sary. These evaluations include, for example, the examina- means that the fluorochromes can be excited with a wider
40 MORPHOLOGY AND ULTRASTRUCTURE

range of wavelengths. In addition, for 2P excitation usually length on the horizontal axis) (e.g., reference 21 and
only one excitation wavelength is available at a time, and www.probes.com). Trpical excitation-emission spectra are
consequently, for multiple staining the fluorochromes must shown in Fig. 2. This spectral characteristic of fluors is fun-
have the same excitation cross-section while having differ- damental to understanding their behavior and selecting
ent emission peaks. A further point to consider is the com- specific fluors and fluor combinations for application in flu-
bination of 1P and 2P excitation. Observed differences in orescence and LSM studies. The signal emitted by a fluor
how they excite the same fluors indicate that they must be often provides information about its environment, includ-
performed sequentially rather than simultaneously. ing its binding site. Fluors are used independently or as re-
porters conjugated to probes for specific macromolecules.
These probes are considered in two classes, targeted (i.e.,
3.5. SAMPLE PREPARATION OPTIONS recognizing macromolecules) and responsive (i.e., changing
In practice, one of the appealing aspects of fluorescence mi- the fluorescent signal as related to the environment of the
croscopy is the lack of a requirement for fixation, dehydra- fluor). Bright and Taylor (8) indicated that the use of fluo-
tion, and the air drying of samples required by some other rescence has the following major advantages.
techniques. As noted previously, one of the goals of this ap-
proach is to examine living bacterial cells, aggregates, and 1. Sensitivity, with detection of lo- to M con-
biofilms. In most instances, immobilization of the cells is re- centrations of a fluor possible.
quired, and this may be carried out by using agar slide tech- 2. Specificity: with proper controls, probes, and condi-
niques or any one of the techniques outlined in the previ- tions, the identities and localization of a variety of
ous chapters on light microscopy (chapters 1 and 2) or in macromolecules can be determined.
Caldwell et al. (12) or Lawrence et al. (32). These methods 3. Spectroscopy and spectroscopic measurements may be
include light microscopy-friendly techniques such as used to obtain information about the environment of the
continuous-flow slide culture and a variety of coupon-based fluor, e.g., by using pH-sensitive fluors.
approaches. However, it is also possible to examine cells 4. Spatial resolution: the use of fluorescence allows
and biofilms that have been fixed and embedded (e.g., analysis at the limits of light microscopy, e.g., the localiza-
Epon, Periplast, and TissueTek) or frozen and sectioned tion of specific carbohydrates (43, 44) or metals by using
prior to being mounted on a slide and stained with a fluor Newport Green (60).
or fluor-conjugated probe (see reference 34). Hydrophilic 5. Temporal resolution: the user can assess time-depend-
resins such as Nanoplast which stabilize structures and min- ent processes related to metabolism, potentials (e.g., redox
imize artifacts may be employed with both transmission and electrical), and movement. For example, it is possible
electron microscopy and LSM (19, 47). Simple techniques to determine diffusion rates (18, 31), visualize enzyme ac-
may be effectively used, e.g., the use of embedding and tivities (56), and detect the rate of plasmid transfer (22).
paraffin sections in combination with rRNA-targeted For a fluor to be useful, it must meet certain criteria.
probes to study methanogens in anaerobic sludge granules
(49) and cryosectioning to examine biofilms (62). A solu- 1. Have appropriate excitation and emission wave-
tion of 0.1% (wtlvol) agarose (39) or 20% (wtlvol) DNA- lengths.
sequencing-grade acrylamide ( 14) for embedding of biofilms 2. Produce a detectable yield of visible light, i.e., a high-
and aggregates for confocal microscopy can also be used. quantum yield.
Advantages of gel embedding are that the sample may be ei- 3. Resist photobleaching.
ther fixed and dehydrated or fully hydrated when examined 4. Be minimally toxic.
and the gels are relatively nonfluorescent. A disadvantage is 5. Be able to penetrate to the appropriate location.
that the preparations may be relatively fragile. 6. Have a highly specific response to the target mole-
cule(s).
3.5.1. Correlative Microscopy
In reality, there are no ideal fluors or fluorescently con-
Correlative microscopy is a name used when two or more jugated probe combinations and few if any ideal specimens.
microscopic techniques are employed to examine the same Therefore, a number of considerations must be taken into
or parallel specimens. A number of methods should be con- account when setting out to apply fluorescence imaging.
sidered when designing an analytical approach to particular Additional assessments must be considered when combina-
specimens. Some authors have combined CLSM with con- tions of fluors and fluor-conjugated probes are used. These
ventional epifluorescence and transmission electron mi- are critical assessments for 1P confocal microscopy, al-
croscopy (35) or CLSM and scanning electron microscopy though there is extensive literature that provides basic in-
(29, 48), and others have combined CLSM with photon- formation on expected excitation-emission spectra. O n the
counting microscopy (45). other hand, for 2P or multiphoton laser microscopy the
present literature is limited and practical evaluation is re-
3.6. FLUORESCENCE A N D quired to assess the excitation-efnission spectra for any fluor
FLUORESCENT PROBES prior to its use.
When applying a fluor or a fluor conjugate to an un-
Fluorescence is a near-perfect tool for the in vivo or in vitro, known sample, the user should assess the following aspects.
nondestructive, noninvasive study of biological samples in-
cluding microorganisms. Fluors have unique and character- 1. Is there autofluorescence or background fluorescence
istic spectra for excitation and emission, which is a conse- originating from the unstained sample? Selection of the ap-
quence of their electronic configurations (Fig. 2). For many propriate filters may go a long way to limiting autofluores-
fluors, there are published absorption-excitation and emis- cence interference, as may the utilization of fluorescent re-
sion spectra which show the relative intensity of the fluo- porter molecules which have maximal separation in terms
rescence, in relation to the wavelength of light being of wavelength from autofluorescence wavelengths.
applied (relative intensity is the vertical axis versus wave- Quenching solutions such as 0.1% (wtlvol) NaBH, (sodium
3. Laser Scanning Microscopy 41

borohydride) used to reduce autofluorescence due to fixa- tetrazolium chloride; 1 to 20 mM) have demonstrated toxi-
tives such as glutaraldehyde may have applications in other city for microbial samples (13,54). In contrast, other fluors
preparations. However, autofluorescence can also be a (such as fluorescein and CellTracker) appear to have no
source of information; Lawrence et al. (28, 29) used the flu- negative effect when used on living bacteria (7, 11).
orescence of chlorophyll to identify and localize algae in 8. Although the above-described steps should result in
microbial mats and river biofilms. Likewise, autofluores- an optimized protocol for the application of a single fluor or
cence and LSM facilitated the examination of the distribu- fluor conjugate, most applications with 1P or 2P excitation
tion of photosynthetic bacteria in complex mat communi- have colocalization or multiparameter imaging as the de-
ties (58). Further, many bacteria may exhibit natural sired goal. Therefore, the implications of multiple fluors and
autofluorescence; for example, in methanogens autofluores- fluor conjugates must also be assessed.
cence is due to high levels of coenzyme F420. 9. Fluor interactions may occur, and observations should
2. What is the target molecule of interest (protein, car- assess the potential of fluor A to quench the fluorescence
bohydrate, or nucleic acid), and what is the probe (i.e., an- of fluor B, resulting in attenuation of the signal. For ex-
tibody, lectin, rRNA, or DNA)? ample, the SYT09 dye may quench dyes such as Cy3.
3. Based on the presence or absence of autofluorescence Overlap of excitation-emission spectra creates problems
and whether multiple probes are being applied, select the with the separation of emission signals and unacceptable
appropriate fluor based on excitation-emission spectra. See bleed through of the signal from fluor A into the observa-
Fig. 2 , which illustrates an ideal fluor combination where tion channel for fluor B or fluor C (Fig. 2B). These prob-
the excitation and emission spectra of the three fluors are lems can be avoided through sequential addition of stains or
well separated (Fig. 2A) and nonideal fluor combinations limitation of imaging to only one fluor at a time through se-
plus the nature of overlapping excitation-emission spectra, quential rather than simultaneous excitation-emission.
showing how tailing or bleed through may lead to conflict- 10. When the probe is a fluor conjugate, there exists the
ing signals during multiple staining (Fig. 2B). potential for one probe to interfere with the other or act as
4. Perform a practical assessment of fluor excitation- a target for other probes. The effects of incubation time,
emission spectra on the 1P or 2P system to be used, cover- concentration, fluor labeling, the order of fluor addition,
ing all available excitation-emission combinations of the and probe interactions must all be studied.
instrument. This step allows the assessment of such things
as bleed through of the fluor signal into other emission
A listing of fluors and fluor conjugates is provided in
Table 3 illustrating the range of fluors available and their
wavelengths and the presence of other secondary excitation
applications to the study of bacteria, aggregates, and
and emission peaks that may occur when 1P or 2P excita-
biofilms.
tion is applied.
5. Assess the quantum yield or signal strength and the
fade response curve of the fluor, i.e., is the fluor stable in
this application or should a fade retardant be considered? 3.7. LSM IMAGING
Differential fading in dual- or multiple-fluor applications After the LSM system has been aligned and the correct
may create problems for quantitative imaging. For example, fluor conjugate for the selected target in the specimen has
the results obtained using the LIVE/DEAD staining system been prepared for examination, there are a number of op-
(Molecular Probes) can be dramatically influenced by dif- tions available for imaging. The user may collect single im-
ferential fading of the two components. Fading can be con- ages in the xy, xz plane or a series of xy or xg images creat-
trolled through limiting observation and scan time and ing an image stack for 3-D reconstruction and/or digital
choosing the correct combination of zoom, pinhole, and image analyses. It is also possible to collect images by using
PMT settings and laser intensity. Antifade reagents are rec- combinations of one, two, three, or more excitation and
ommended for many fluor applications such as fluorescence emission wavelengths by using 1P (including UV) or 2P sys-
in situ hybridization (FISH). Florijn et al. (20) deal with tems. These images may be collected sequentially or simul-
these reagents, which include p-pheneylene-diamine, n- taneously depending on the LSM system in use and the suit-
propylgallate, 2-mercapto-ethylamine, and 1,4-diazabocy- ability of the fluors and fluor-conjugated probes selected.
clo-2,2,2-octane (DABCO). Note that fade retardants are One may also collect images by using fluorescence, reflec-
highly toxic, must be used with caution, and may be ordered tion (i.e., colloidal gold), and nonconfocal transmission
premixed from Molecular Probes or Citifluor (London, modes.
United Kingdom).
6. If a targeted conjugated probe is being used, then its 3.7.1. Scanning in the xy Direction
specificity for target molecules in the sample must be as- 1. After checking all alignments and optimizing the in-
sessed through the use of controls, including blanks; blind- strument as described in sections 3.3 and 3.6, the user may
ing of the probes and/or the reactive site; blocking of non- either switch between epifluorescence and LSM modes or
specific binding sites; or inhibition of nonspecific binding. scan continuously while examining the specimen and se-
Elaborate protocols have been established for FISH (1) and lecting locations for image collection. (Note that time is al-
for lectin use (44). ways of the essence in fluorescence microscopy; fluors decay
7. In addition, an important but understudied aspect is at room temperature, in water, in light, and under observa-
the influence of the fluor on the specificity of the conju- tion.) Continuous scanning can be done with experience if
gated probe, i.e., a fluorescein isothiocyanate-conjugated the fluorescence signals are stable and resist bleaching. The
probe may exhibit specificities at variance with those for actual taking or collection of images can be done in a ran-
the same probe conjugated to tetramethyl rhodamine iso- dom pattern or along a transect or consist of large-scale
thiocyanate (TRITC) or Cy5 (44). Assess the fluor for tox- montages composed of a series of adjacent microscope
icity. Is it toxic in the range required to monitor changes in fields. The latter approaches should be considered when
fluorescence and to observe a response in the test cells? collecting images for further analyses and statistical analysis
Some fluorochromes such as CTC (5-cyano-2,3-ditolyl of data (25,28,49). Once the appropriate location has been
42 m MORPHOLOGY AND ULTRASTRUCTURE

TABLE 3 Fluorescent comoounds and their aoolication i n microbial studies


Fluor Application

Compounds for cell labeling and enumeration Fluors which become fluorescent after binding to nucleic acids allow for
DAPI visualization and enumeration of individual cells (24)
Acridine orange
Hoechst 33258
Hoechst 33342
SYTO stains
SYTO green
SYTO blue
SYTO orange
SYTO red
SYTO far-red
SYBR green
PicoGreen
TOTO-l/TO-PRO-l
YOYO-l/YO-PRO-l
POPO-3
SYBR green 1/11
ChemChrome V6
Sytox stains Used for fixed or membrane-damaged cells
Propidium iodide Used for fixed or membrane-damaged cells
diOC6
Bis-oxonl
Probe labels" Suitable for conjugation to protein linkers on dextrans, lectins,
Alexa series (488 and 546) nucleotides, oligonucleotides, and peptides and for creation of
fluor-conjugated reporter molecules
Oregon Green (488) (difluorofluorescein)
Fluorescein isothiocyanate (494)
BODIPY FL (503)
Carboxytetramethylrhodamine(550)
Tetramethyl-rhodamine
Texas Red (596)
Cyanine dyes (Cy2, Cy3 [550], Cy5 [6491)
Compounds for identification
FISH compounds Detection of fluor-labeled oligonucleotide probe after hybridization
BacLight fluorescent Gram stain Two-component fluorescent stain allowing in situ Gram reaction
(pure cultures)
Green fluorescent protein Tracking of green fluorescent protein-labeled cells
Immunostaining compounds Fluorescent labels for antibody recognition of antigen
CellTracker Labeled cells may be monitored for several generations based on partitioning
of label to daughter cells (7)
Compounds for analysis of activity, viability,
and metabolism
RH-795 Analysis of cell membrane integrity and membrane potential
Rhodamine 123 Analysis of cell membrane integrity and membrane potential
Ethidium bromide Analysis of membrane integrity and detection of spore germination (16)
diBAC,( 3) Analysis of membrane integrity and detection of spore germination
LIVEDEAD BacLightTM(propidium Analysis of integrity of individual cell membranes and cell viability;
iodide-SYT09) see reference 14 for protocol
FISH compounds Analysis of cellular activity and ribosome content
FISH-MARbcompounds Analysis of ribosome content and response to substrate addition
In situ PCR compounds Detection of specific mRNA activity
Sulfofluorescein diacetate Detection of esterase activity

(Continued on next page)


3 . Laser Scanning Microscopy 43

TABLE 3 Continued
Fiuor Application
ELF-97 Detection of alkaline phosphatase activity; see reference 14 for protocol
Carboxyfluorescein diacetate Detection of esterase activity
Triphenyl tetrazolium chloride Detection of respiratory activity (dehydrogenase)
CTC Detection of respiratory activity (dehydrogenase);
see reference 14 for protocol
efp gene green fluorescent protein gene Analysis of gene expression of individual cells
Environmental reporters
BCECF-AM pH responsive
SNAFL-1 pH responsive
5- and 6-carboxyfluorescein pH responsive
Newport Green Sensitive to nickel concentrations;
see reference 60 for details
efp gene (green fluorescent protein gene) Analysis of gene expression of individual cells may provide information
on in situ conditions
Numbers in parentheses are excitation maximums in nanometers.
bFISH-MAR, FISH microautoradiography.

found, stop scanning, select the appropriate zoom, and op- age, start with the longest excitation wavelength and work
timize pinhole, brightness, and contrast values. It may also towards the shortest.
be necessary to select whether to collect the image as a di-
rect acquisition or through a mathematical filter. A number 3.7.2. Scanning in the xz Direction
of mathematical filter options are available, including line During an xy scan, the laser beam is scanned over a defined
averaging, frame averaging, and Kalman filtration. Kalman area; however, the beam can also scan along a single line ex-
filtration (a running-average filter that mathematically av- tending into the specimen (xz scan). When a line scan is re-
erages sequentially collected images, thereby reducing noise peated at many q levels, the resulting image (2 scan) is a
level in the image but maintaining edge features) is ex- sagittal section through the sample. q scan images lack the
tremely effective in reducing random noise and producing a resolution of xy images due to the nature of the corrections
clean image. Figure 3 illustrates the effect of mathematical in standard objectives and the point spread function of the
filtration. Once these decisions have been made, then it is lens. Therefore, xq images are exaggerated in the 7 dimen-
possible to start the collection of images. sion and a single point will appear to have an elongated
2. For quantitative imaging purposes, it is necessary to z axis (see beads in Color Plate 4B and D). This effect may
maintain the microscope settings (i.e., gain, laser intensity, be seen in Fig. 4B and C showing an xy-xzcombination at
and aperture size) throughout the collection of images from the same location in a microbial biofilm. It is also possible
an experimental series in order to create a valid compara- to calculate xz images from xy serial sections by using vari-
tive data set. Changes in any of the microscope settings will ous software packages.
alter the section thickness, the brightness, and the apparent Guidelines for xq scans are as follows.
size of the objects being imaged.
3. All LSM images are grayscale; color is applied by using 1. Focus through the material to establish the top and
color LUTs which assign colors according to the correspon- bottom or beginning and end of the region to be scanned.
ding grayscale value. Color is also assigned to indicate the Then take a single xy scan and use it as the basis to select
excitation-emission combination that gave rise to the par- the position and orientation of the xq line to be scanned.
ticular image. This assignment is arbitrary, but typically im- 2. For xq imaging, it may be necessary to increase bright-
ages are combined in the red-green-blue mode and stored in ness and contrast values and to adjust scanning time.
a tagged-image file format (TIFF) or a file format specific to Depending upon the instrument being used, this adjustment
the manufacturer (most often a modified TIFF). Most in- may not be possible or it can be done manually or automat-
struments will provide a display of each channel and, si- ically through the software. Typically, xz scans also benefit
multaneously, the combination of all channels. If all the from the application of line averaging, or Kalman filtration.
probes had the same excitation wavelength but emitted at 3. It may be helpful to do a quick scan (to minimize pho-
different wavelengths or there was minimal bleed through tobleaching) through the site first in order to assess whether
between channels, then simultaneous collection can be ad- the selected section thickness and orientation are of value.
vised. In general, one is using a variety of fluors with differ- Always remember that, depending on the objective lens,
ent excitation-emission spectra, e.g., SYT09, TRITC, Cy5, you may exceed the working distance if you select a large
and autofluorescence (if present). In this case, the images depth range for your sagittal section.
for each channel should be collected sequentially. These 4. It is possible to create an xz:series through a specimen;
images may then be overlaid in the operating software of however, as noted previously, this can be done through cal-
the LSM system, or the overlaying may be done by using a culation by using the xy series data set. Fig. 4 shows a typi-
variety of software packages (e.g., those of NIH IMAGE, cal xy and xz image pair taken through a biofilm stained
Image J, or Imaris). To reduce bleaching and sample dam- with SYTO9 (Molecular Probes).
44 a MORPHOLOGY A N D ULTRASTRUCTURE

FIGURE 3 1P LSM images showing a direct single-scan image (A) and the influence of image averaging on the quality and sig-
nal-to-noise ratio of an LSM image (B).

FIGURE 4 1P LSM micrographs showing the nature of the 3-D stereo pair (A) and a single xy (B) and a single xq (C) scan
through a microbial biofilm stained with the nucleic acid stain SYTO9 (Molecular Probes).
3 . Laser Scanning Microscopy n 45

3.8. 3-D IMAGING tection of emission signals has a number of advantages, in


The major advantage of LSM is the capacity to collect a se- particular by showing the identities and locations of bacte-
ries of images that allow the user to obtain 3-D spatial in- rial cells and of bacterial macromolecules. A variety of
formation. This capability follows from the optical section- probes such as fluor-conjugated antibodies, lectins, and nu-
ing capacity and the creation of a s series in perfect register. cleic acid stains, including rRNA probes, can be used to
This series of xy images is referred to as a z series since it is label bacteria and associated macromolecules. Although it
taken along the z axis or commonly as an image stack. is common to localize two to three signals, specific strategies
Serial optical sections can be used for a variety of 3-D re- allow collection of information on as many as seven differ-
construction techniques (see below and Table 4). When an ent labels in a specimen ( 2 ) . Several factors become critical
ideal z series is collected for display or analytical purposes, in collecting multichannel images.
the sections should meet the following specific criteria. 1. The nature of the emission and excitation optical fil-
1. They must be collected nondestructively where pho- ters or other mechanisms for selecting specific wavelengths
tobleaching is not a significant factor; however, in practice of light. For example, a long-pass filter will allow all wave-
during collection of a series photobleaching can be com- lengths greater than a set value to pass through and this may
pensated for by increasing brightness and contrast. create problems for excitation and detection of other fluors.
Photobleaching can also be controlled through the applica- In contrast, a band-pass filter allows only specific wave-
tion of fade-retardant solutions. The user should also con- lengths to pass, which can be desirable; however, it can re-
sider the total slice capacity of the sample (this is a fixed sult in insufficient light for detection of the fluor. For many
number of sections that are taken from a location in a spec- applications, significant advantages may be found in LSM
imen before the effects of photobleaching become signi- systems that do not use optical glass filters.
ficant). This value may be best considered empirically 2. Misalignments in the optical pathway can cause the
through trial, error, and experience. images from different channels to be offset. The presence of
2. Sections must be adjacent, without oversampling or un- these misalignments in the xy or xs plane may be checked
dersampling error. This requirement involves selection of the through imaging of Focal Check (6- or 15-pm-diameter)
correct sectioning interval. Theoretically, with a high-NA ob- beads in the dual- or three-channel mode and overlaying of
jective, sections can be collected at intervals as small as 40 the resulting images. Any misalignment is evident in the
nm. In practice, optical sections are 0.5 to 1.5 pm in thick- overlay of the channels and should be corrected by follow-
ness, and this factor should be reflected in the optical section- ing the alignment procedures outlined previously, arranging
ing interval, particularly in attempting to image bacteria. If for a service call, or investing in high-quality objectives.
the section interval is incorrect, oversampling or undersam- Color Plate 4 illustrates the alignment and misalignments in
pling errors occur. These errors are most important in analysis three-channel images by using 6-pm-diameter Focal Check
of a series to extract depth information or in 3-D reconstruc- beads.
tion. One quick test to assess sampling frequency is to project 3. Axial chromatic aberration in the objective lens re-
the series as a 3-D image; if there are visible discontinuities or sulting in offset images. If the cause of misalignment is in
steps in the image, then the sampling interval is incorrect. the objective lens, realignment of the LSM system cannot
3. Sections are collected within the working distance of correct the problem. For most lenses, maximum distortion
the objective. As discussed above, a specific objective lens occurs at the edge of the lens; 1.4-NA plan apochromat
has a working distance that is a function of its magnifica- lenses provide the largest fully corrected area (cf. Color
tion and NA, and this must be considered when obtaining Plate 4A and E). Software solutions are also available that
a z series. (Again, empirical observation also serves to es- allow toggling of the digital images into alignment and
tablish the working limits for specific samples.) cropping during the preparation of multichannel overlays.
4. Sections must be homogeneous; minimize nonuniform 4. Wavelength of light. The longer the wavelength of
bleaching by having the laser aligned. Also consider chang- light, the thicker the optical section; thus, sections collected
ing the fluor or the staining approach by using a more sta- in the far red are thicker than those collected in the red and
ble fluor or switch from a positive to a negative stain (11). green (17). Fully corrected 350- to 1,000-nm-wavelength
Heterogeneity may also occur as a result of vignetting, lenses minimize this problem.
which can be corrected by proper alignment or use of the 5. Field curvature and lateral aberration, which result in ob-
proper objective and zoom combination. jects at the edges of images appearing in slightly different loca-
5. Sections must be aligned and in register by ensuring tions. See points 2 and 3 regarding high-NA objective lenses
the critical alignments as discussed above. Color Plate 4 and the use of Focal Check fluorescent beads (Color Plate 4).
shows the nature of misalignments that may be detected 6. Loss of signal and resolution with increasing wave-
using Focal Check beads in multichannel mode. In this length of emission; thus, images originating in green and far
case, Color Plate 4A shows correct alignment whereas red are of considerably different quality.
Color Plate 4C and D illustrate inherent problems with the 7. Superior results may be obtained by sequential addi-
objective lenses. tion and viewing of probes with the lasar scanning micro-
6 . Sections must be calibrated so that image gray levels scope or by simultaneous application of equimolar mixtures
accurately represent concentrations or the dimensions of an of probes labeled with different dyes.
object (see further discussion below). 8. It is advisable to include a color wheel in multipara-
7. Sections must be isotopic such that the xy pixels cor- meter images to facilitate interpretation of the combina-
respond to the z dimension. To collect an xy series, the pro- tions of probes in the specimen.
cedure is essentially as described for the xy image collection. Amann et al. ( 2 ) present a method for multiple staining
using rRNA-targeted fluorescent probes. Lawrence et al.
(28) discuss multiple-parameter imaging and demonstrate
3.9. MULTIPLE-PARAMETER IMAGING the utility of nucleic acid staining to detect bacterial cells,
The simultaneous or sequential acquisition of images by lectin staining to assess bacterial exopolymers or glycocon-
using a range of excitation wavelengths and selective de- jugates, and autofluorescence signals to detect phototrophic
46 MORPHOLOGY AND ULTRASTRUCTURE

organisms. Lectins have tremendous potential as single that the approach can be used to qualitatively demonstrate
probes, in combination with other fluors and probes, or in the presence of heavy metals such as Ni in defined systems.
combination with one another. Some of the pitfalls of lectin
staining have been presented by Neu et al. (44). Next, we
1. Biofilms on 1-by-3-cm coupons were exposed to 100
p1 of a 1 mM Ni solution for 1 h and subsequently washed
present a brief protocol for the application of lectins to at-
two times with 100 pl of sterile distilled water.
tached or immobilized cells or biofilms.
2. Biofilms were then incubated with 100 pl of a 20 FM
SYT017 solution in the dark for 15 min. They were then
3.9.1. Lectin Staining washed three times with sterile distilled water.
Labeled lectins have been applied in many pure culture 3. Samples were then incubated in the dark for 15 min
studies to probe for cell surface structures. More recently, with 100 pl of a 2.4 FM solution of Newport Green.
they have been used to image and characterize the exopoly- 4. Samples were observed by using the 488- and 543enm-
meric substances of microbial aggregates and biofilms (28, wavelength excitation lines of the laser scanning micro-
43, 59). Neu et al. (44) provide a fairly detailed discussion scope for Newport Green and SYT017, respectively. Either
of their applications. A general protocol and considerations a 40X, 1.3-NA or a lOOX, 1.3-NA plan neofluor oil im-
are outlined below. mersion lens was used.
1. Fluorescent lectins conjugated with different fluors The method is suitable for qualitative detection and lo-
such as fluorescein, fluorescein isothiocyanate, Oregon calization of nickel and perhaps cadmium in association
Green, TRITC, Texas Red, and Alexa are available (e.g., with bacterial cells, colonies, and biofilms.
Sigma, St. Louis, MO, and Molecular Probes). Custom la-
beling (e.g., with Cy5 or Alexa) may be done by using com-
mercial kits according to the instructions (Research 3.10. DYNAMIC IMAGING
Organics, Cleveland, OH, or Molecular Probes). As noted previously, one of the major advantages of fluo-
2. Lectins are employed alone or in combination for dou- rescence microscopy, including conventional epifluores-
ble and triple staining. In brief, they are dissolved at 100 pg cence and 1P and 2P LSM, is the capacity for temporal res-
ml- in filter-sterilized (0.2-pm-pore-size filter) water. olution, where time-dependent processes may be monitored
Slides of 1 cm2 carrying a biofilm are directly covered with by fluorescence. These processes would include diffusion
100 pl of lectin solution. and transport of molecules, changes in intra- and extracel-
3. Next, samples are incubated in a humid chamber at lular pH, cellular metabolism, kinetics of binding, and
22 t 2C for various times to assess the time required for measurement of redox potential.
complete staining. After determination of optimal staining A number of approaches have been applied to study dif-
time, the effect of lectin concentration ( 5 pg cm- to 100 fusion in bacterial biofilms and serve to illustrate the appli-
Fg cm-) needs to be determined. cation of kinetic analyses using LSM systems. There are a
4. After staining, all the slides are carefully rinsed with number of fluor-conjugated probes that may be used to as-
filter-sterilized water four times to remove unbound lectins. sess permeability and diffusion coefficients of bacterial cells
For each rinse, the wash water must be carefully added to and polymers; these include ficols, size-fractionated dex-
the biofilm or bound cells and drawn off with filter paper. trans, and a range of fluorescent beads (10 nm to 15 um in
5. The sample is then transferred to the laser scanning mi- diameter). Lawrence et al. (31) used 1P LSM to monitor
croscope stage. To avoid disturbance of the biofilm, no cover- the migration of fluor-conjugated dextrans to determine
slip is used. If an upright microscope is employed, a 63 x ,0.9- effective diffusion coefficients for biofilm systems. Micro-
N A water-immersible lens is ideal for direct observation injection and 1P LSM were used by De Beer et al. (18) to de-
through the water droplet covering the biofilm. The lens must termine diffusion coefficients for biofilm materials. The more
be carefully immersed into the droplet and slowly brought standard fluorescence recovery after photobleaching (FRAP)
near the biofilm surface. For inverted microscopes, the sub- approach may also be applied to bacteria, aggregates, and
stratum with the attached biofilm must be mounted upside biofilms. Birmingham et al. (4) used FRAP to monitor diffu-
down into a coverslip chamber (NalgeNunc International) by sion and binding of dextrans in oral biofilms. Laca et al. (26)
using spacers. This provides a water space to protect the used CLSM to monitor diffusion of protein in alginate beads.
biofilm or attached cells, and the specimen must then be ex- The details of the procedures and necessary equations are
amined with relatively-long-working-distance lenses. The outlined in these and other papers (3,6, 9).
same procedure of staining as described above can be applied Fluorescent probes such as fluorescein, 5 - and 6-
to this inverted setup. Michael and Smith (40) also outline carboxyfluorescein, and dually labeled fluorescein-
procedures for lectin application and the use of positive and rhodamine dextrans, etc., exhibit pH-sensitive fluorescence
negative controls. See also reference 57a. Color Plate 5 uses a and consequently have potential for in situ measurements.
three-color stereo pair to present the result of staining with a Caldwell et al. (10) presented a series of pseudocolor images
combination of three fluor-conjugated lectins. This can be of these gradients by using 1P LSM and the probe 5- and 6-
best viewed by using stereo glasses. Color Plate 5 (panels B carboxyfluorescein. In a more recent study of in situ pH,
and C) presents high-magnification images of bacterial mi- Vroom et al. (57) applied 2P LSM to detect pH gradients in
crocolonies triple-labeled with three lectins and showing de- mixed-species biofilms. In their application, they were able
tail of the hydrated exopolymer structure. to demonstrate real-time imaging of pH gradients. Hunter
and Beveridge (23a) also demonstrated pH gradients in
biofilm regions and microcolonies; their study addressed
3.9.2. Imaging of the Metals Nickel and Cadmium concerns that pH-sensitive probes may be influenced by the
Wuertz et al. (60) presented a protocol for localization of the presence of proteins and other biofilm constituents, indi-
metals nickel and cadmium in biofilms. Conceptually the cating that these effects were not occurring in their system.
method is based on the observation that Newport Green at a A common approach to the study of pH is the use of ra-
concentration of 1 p M (22C, pH 7) increases its fluores- tiometric probes such as 5- and 6-carboxyfluorescein and
cence 16 times in the presence of 25 pM Ni. They concluded SNAFL-1 which exhibit no pH sensitivity when excited
3. Laser Scanning Microscopy 47

10 pm
FIGURE 5 Series of 1P LSM images showing the monitoring of pH by using a dually labeled (pH-sensitive fluorescein and pH-
insensitive rhodamine) 10,000-molecular-weight dextran in a microbial biofilm. (A) pH-sensitive imaging of fluorescein. (B) pH-
insensitive fluorescence of the rhodamine. (C) Grayscale representation of the standard curve for pH versus the ratio of images A
and B. (D) Contour map showing the distribution of pH levels within the microbial biofilm.
48 MORPHOLOGY A N D ULTRASTRUCTURE

by 435-nm-wavelength light but are sensitive when excited by probe and has the added advantage that straightforward cal-
490-nm-wavelength light. A ratio is taken of the pH-sensitive ibrations of the fluor response in buffer are sufficient to cre-
and pH-insensitive fluorescence signals, thereby eliminating ate a valid standard curve. A number of publications have
effects such as variable concentration. When imaging shown that this approach can be applied for determination
biofilms, one needs to consider the lack of discrete boundaries of ion concentrations in bacterial cells and pH gradients in
within the biofilm and the presence of multiple environmen- biofilms (51,53,57). The technique requires a modified 1P
tal variables (e.g., Eh and ion concentration), as well as the laser scanning microscopic equipped with an optical chop-
potential for fluor sorption to macromolecules and fluor up- per to create nanosecond laser pulses and time-gated detec-
take into microbial cells (see, e.g., reference 23a). With bac- tion to collect the fluorescence emission following the short
terial cells, the major concern is effective loading of the fluor excitation pulse. The reference by Sanders et al. (51) pro-
into the cells and its behavior in the presence of intracellular vides a starting point for understanding the nature and ap-
protein and other macromolecules. The calculation of pH plication of fluorescence lifetime imaging of pH by using
from the ratio images is based on ratiometric imaging of pH- SNAFL-1 and 1P LSM.
equilibrated cells, biofilm, or other specimens which have
been stained in the same fashion as those under investigation.
The cells used for equilibration may first be fixed in 70% 3.11. HANDLING 3-D DATA SETS
(vol/vol) ethanol or rendered permeable by using valinomycin There are many options for the presentation of 3-D data
and nigericin, which act to equilibrate potassium and proton sets; these are summarized in Table 4. The selection of the
gradients across the cell membrane. These cells or biofilm ma- option is dictated by personal choice, detection of specific
terials are then placed in appropriate buffers (pH 5.0 to 8.0) information, and cost. The easiest but least informative
with the fluor to obtain pH-equilibrated specimens that are presentation is the gallery in which all the images of a series
analyzed by using LSM and digital image analysis to establish are present sequentially (Fig. 6). Figure 7 shows the same
a standard calibration. Figure 5 illustrates the procedure, image stack (or series) presented as a simple black-and-
showing images taken in the green (pH responsive) and red white stereo pair. In general, the use of color to code for
(pH independent) wavelengths, the ratiometric image (with a depth information, create red-green anaglyph stereo images,
standard grayscale calibration curve), and the resulting con- and form three-color or multicolor stereo pairs (Color
tour map of pH in the biofilm material. Plates 5 and 6) is an effective method, although publication
The ratiometric method suffers from the drawback that costs may be inhibitory. Another option for the display of 3-
quantitative pH imaging requires time-consuming, cumber- D data sets is simulated fluorescence, whereby the mate-
some calibration procedures as described above. However, rial is viewed as though it were illuminated from an oblique
many ratio probes such as 5- and 6-carboxyfluorescein and angle and the surface layer were fluorescent. The applica-
SNAFL-1 may be used in conjunction with a technique re- tion of 3-D rendering through ray tracing or surface con-
ferred to as fluorescence lifetime imaging. This method was tour-based programs may also provide a useful presentation
introduced -10 years ago as an alternative method for of 3-D data sets, allowing the viewer to examine the data set
achieving contrast in fluorescence images. It is based on de- from various perspectives. For oral presentation, the evolu-
tecting difference? in the rates of decay of fluorescence of a tion of computer animation through programs such as
molecule. This type of imaging also produces images that QuickTime allows the LSM user to present 2: series as live
are independent of the concentration of the fluorescent showings of images collected in movie format. This format

TABLE 4 Methods for the presentation of 3-D data sets


Method Advantage Disadvantage
Gallery Simple presentation may allow specific No true 3-D representation; limited data content
marking of objects and depth
xyz projection View of single section in xy and sections in Maximum-intensity projection in xy is not possible
xx and yz along user-defined line
Red-green anaglyph Comparatively simple to prepare; usually Expense of color; requires special red-green glasses;
stereo projection part of LSM software up to 20% of people cannot see the stereo effect
Simulated fluorescence Provides a type of 3-D view of a Z series Presents only surface data
Stereo pair Can be either black and white or color, can Requires special viewer; not oral presentation friendly;
contain information from multiple probe high expense; up to 20% of people cannot create
signals, and can be very high quality the stereo effect
Polarized double Effective display of data on computer screen Requires special glasses and projector
projection
3-D rendering Detailed reconstruction of the data set; Tends to be limited to external features; 3-D space-filling
allows for rotation and fly through construction; extreme demand for computing time
Video Animations can be transferred to video; Requires additional equipment; not useful
easy to use for publication
Movie Audience can see Z series as collected; movie Requires laptop computer and special projection facility;
contains more visible information than the not a publishable format at this time
static presentation; easily appreciated
Digital presentation Allows use of single images or complex Not suitable for publication in most journals
animation; easy for presentation
3 . Laser Scanning Microscopy w 49

FIGURE 6 Gallery presentation of a Z series taken through a microbial biofilm stained with SYT09.

also facilitates the projection of full-color rendering and an- options available to LSM users with respect to quantifica-
imation of data sets, which provide a more information-rich tion and deconvolution. See also references 30 and 32 and
presentation than static formats. image-processing texts such as that by Russ (50).
Although there are many commercial and freeware sys-
3.1 1.l.Quantification tems for 3-D image analysis and processing, no single soft-
Processing and analysis of digital images are major topics in ware package offers all the required features. Some users
their own right, and thus we can only touch upon various may prefer commercial systems which are offered by the
50 MORPHOLOGY AND ULTRASTRUCTURE

FIGURE 7 3-D stereo pair presentation of the Z series shown in Fig. 6.

LSM companies. In addition there are separate commercial .corn). See, for example, Manz et al. (37). Another UNIX-
3-D imaging and image analysis systems, e.g., Imaris and based package for this purpose available over the Internet is
VoxelShop (Bitplane, Switzerland), MicroVoxel (Indec XCOSM, which can be applied to LSM images or other
Systems, Sunnyvale, CA), VoxelView (Vital Images, types of fluorescent images (http://3dmicroscopy.wustl
Fairfield, IA), VoxBlast (VayTek, Inc., Fairfield, IA), and .edu/-xcosm). Regardless of the software package used, it is
ImageSpace (Molecular Dynamics, Sunnyvale, CA). O n essential to have internal standards consisting of fluorescent
the other hand, freeware programs are available that pro- beads of known dimensions in each z series to control and
vide image processing and analysis for single 2-D sections. assess the deconvolution process.
The most widely distributed free software was NIH IMAGE,
now replaced by Image J, available for MS Windows, Mac
OS, Mac 0 s X, Linux, and the Sharp Zaurus PDA 3.12. L S M SAMPLING FREQUENCY
Nevertheless, other tools have
(http://rsb.info.nih.gov/ii/). In LSM there are both 2-D- and 3-D-related considerations
been developed and are available for download from the regarding sampling. In general, the Nyquist criterion indi-
Internet, e.g., CELLSTAT (41), which is a program for cates that the imaging system must sample with a spatial
UNIX workstations (http://www.cbm.biocentrum.dtu.dk), resolution of half that of the smallest object to be resolved.
and ImageTool (36), which is a PC-based freeware image For 3-D imaging, it is necessary to sample at twice the spa-
analysis package (http://ddsdx.uthscsa.edu/dig/itdesc.html) tial resolution of the optical system. In practical terms, this
particularly useful for object-based analyses. COMSTAT means x and y sampling at about 70-nm intervals and in the
(23) is an additional option written as a script in MATLAB 2 axis, which has far poorer resolution, an interval of 150
(http://www.im.dtu.dk/comstat).Some consideration nm is recommended. This sampling frequency creates a data
should be given to neural network systems for analysis of set sufficiently continuous for correct image restoration
fluorescence images (5). (deconvolution) and representation of the sample area in
subsequent processing and stereo presentation of the data.
3.1 1.2. Deconvolution For some analytical purposes (e.g., cell counting and area
As noted above, the major limitation of all light micro- measurements), the approach of sampling at discrete depths
scopic systems and particularly LSM is poor axial resolution. and locations for representative samples from a number of
The principle solution to this limitation is image processing replicate locations provides a viable alternative (38).
of the z series by computing of intensive restoration proce- Most microscopy studies rely upon relatively small areas
dures or deconvolution. Deconvolution refers to the sharp- of analysis; thus, typical data sets obtained may be prone to
ening, or deblurring, of an image obtained using an optical errors based on rare events. In practice, determinations of
microscope by removal of out-of-focus information, thereby representative samplings for microscopic study are not eas-
mathematically producing an optical thin section. The ily made. However, Korber et al. (25) suggested analysis
most common algorithm used for deconvolution is the near- areas exceeding >1 X lo5 pm2 and Maller et al. (42) ex-
est-neighbor deconvolution algorithm; however, others amined contiguous areas of >2.8 X lo5 pm2 for statistically
such as blind deconvolution, iterative maximum likelihood representative results. Alternatively, the user may randomly
estimation, and iterative constrained Tikhonov-Miller are select five or more replicate microscope fields (63X water-
included in certain software packages. The HUYGENS sys- immersible lens) per treatment replicate, allowing applica-
tem (http://www.svi.nl) was designed to run on Silicon tion of analysis of variance to determine significant effects
Graphics computers under the IRIX operating system, but is at P of <0.05 (28).
now available for a range of computer platforms. It is one of
the most advanced packages and includes many image-
processing features. Other specialized software packages are 3.13. L S M IMAGE ARCHIVING
available for the removal of out-of-focus haze such as AND PRINTING
AUTO-DEBLUR (http://www.aqi.com), MICROTOME Application of LSM techniques results in the creation of
(http://www.vaytek.com), and EPR (http://www.scanalytics vast image and data sets; our facilities can produce several
3. Laser Scanning Microscopy w 51

gigabytes per day. Thus, an important consideration is how Technical paper providing more complex mathematical treat-
to archive all this information. First, it is critical to have the ment of the data obtained through imaging FRAP.
most random-access memory possible and the largest hard 7. Boleti, H., D. M. Ojcius, and A. Dautry-Varsat. 2000.
drive(s) available for the operating computer. Second, op- Fluorescent labelling of intracellular bacteria in living host
tions for longer-term information storage include CDs and cells. J. Microbiol. Methods 40:265-274.
DVDs; for temporary storage and transport, a variety of Technical information on the use of CellTracker with bacteria.
mini-hard drives and USB memory sticks are available. For 8. Bright, G. R., and D. L. Taylor. 1986. Imaging at low
relatively secure (significant concerns about the stability of light level in fluorescence microscopy, p. 257-288. In D. L.
CD media have been raised), portable, and relatively uni- Taylor, A. S. Waggoner, E Lanni, and R. Birge (ed.),
Applications of Fluorescence in the Biomedical Sciences. Liss
versal storage media, CDs and DVDs remain the best, most
Inc., New York, NY.
cost-effective recommendation. Other options may emerge, An older book that still prowides an excellent owerview of and
and the user must continually address this question. entry point into the very large cell biology literature which con-
Archiving represents another major hurdle that should tains a vast array of vital information on fluors and techniques
be considered early in the process of developing an LSM- with potential for application in prokuryotic studies.
based research program. Images are stored in a variety of for- 9. Bryers, J. D., and F. Drummond. 1998. Local macromol-
mats such as TIFF, GIFF, U W , PICT, EPS, and JPEG and ecule diffusion coefficients in structurally non-uniform
manufacturers formats. Each of these has advantages and bacterial biofilms using fluorescence recovery after photo-
disadvantages. Some are dead ends which cannot be trans- bleaching (FRAP). Biotechnol. Bioeng. 60:462-473.
lated. Others offer various degrees of image fidelity and abil- Useful technical note on the FRAP method applied to micro-
ities to compress images, such as JPEG. In general, TIFF is organisms.
the most universally used and can be opened by most soft- 10. Caldwell, D. E., D. R. Korber, and J. R. Lawrence. 1992.
ware. Nevertheless, some of the advanced 3-D software Confocal laser microscopy and digital image analysis in
packages use different image formats to analyze 3-D data microbial ecology. Adw. Microb. Ecol. 12:l-67.
sets, e.g., image cytometry standard, which in contrast to The first comprehensiwe owerwiew of applications and potential
TIFF is designed for multidimensional image data. of 1P LSM and digital imaging focused on the area of microbial
Images may be printed for publication by using a vari- ecology; still prowides a good starting point.
ety of means including video printers, dye sublimation 11. Caldwell, D. E., D. R. Korber, and J. R. Lawrence. 1992.
printers, slide printers, color laser printers, and digital Imaging of bacterial cells by fluorescence exclusion using
printers. Increasingly, publishers can deal with the original scanning confocal laser microscopy. J. Microbiol. Methods
digital file. The further development of the Internet and 15:249-26 1.
the way publications will be done in the future may allow Technical note describing details of the fluorescent negatiwe
more publication of animated 3-D multichannel LSM staining technique for bacteria and biofilms .
data. 12. Caldwell, D. E., G. M. Wolfaardt, D. R. Korber, S.
Karthikeyan, J. R. Lawrence, and D. K. Brannan. 2001.
Cultivation of microbial consortia and communities,
3.14. REFERENCES p. 92-100. In C. J. Hum, R. L. Crawford, G. R. Knudsen, M. J.
1. Amann, R., R. Snaidr, M. Wagner, W. Ludwig, and McInerney, and L. D. Statzenbach (ed.), Manual of Enwi-
K. H. Schleifer. 1996. In situ visualization of high genetic runmental Microbiology, 2nd ed. ASM Press, Washington, Dc.
diversity in a natural microbial community. J. Bacterial. Owerwiew; features many devices for cultivation of bacteria and
178:3496-3500. biofilms that are comptible with microscopic studies.
A useful exumple of application of multiple fluors. 13. Choi, 1.-W., B. F. Sherr, and E. B. Sherr. 1999. Dead or
2. Amann, R. I., W. Ludwig, and K. H. Schleifer. 1995. alive? A large fraction of ETS-inactive marine bacterio-
Phylogenetic identification and in situ detection of indi- plankton cells, as assessed by reduction of CTC, can be-
vidual microbial cells without cultivation. Microbiol. Rew. come ETS-active with incubation and substrate addition.
59~143-169. Aquat. Microb. Ecol. 18:105-115.
Excellent review of both FISH and CLSM approaches with A good example of validation of a fluorescence technique for
good color stereo images of microbial specimens. natural bacterial populations and important considerations in
3. Axelrod, A., D. E. Koppel, J. Schlessinger, E. Elsen, and the process.
W. W. Webb. 1976. Mobility measurement by analysis of 14. Christensen, B. B., C. Sternberg, J. B. Andersen, L.
fluorescence photobleaching recovery kinetics. Biophys. J. Eberl, S. M@ller, M. Givskov, and S. Molin. 1999.
16: 1055-1069. Molecular tools for study of biofilm physiology. Methods
A more technical paper which provides the basis for using EnTymol. 3 10:2042.
FRAP to calculate the mobility of molecules. Excellent owerwiew of fluorescence-basedtechniques including
4. Birmingham, J. J., N. P. Hughes, and R. Treloar. 1995. those compatible with LSM.
Diffusion and binding measurements within oral biofilms 15. Conn, P. M. 1999. Methods in Enzymology, vol. 307.
using fluorescencephotobleaching recovery methods. Phil. Confocal Microscopy. Academic Press, New York, NY.
Trans. R. SOC.Lond. B 350:325-343. A comprehensiwe compendium of papers on various areas of
A practical application of FRAP in microbial samples. confocal microscopy with technical details and applications.
5. Blackburn, N., A. Hagstrom, J. Wikner, R. Cuadros- 16. Coote, P. J., C. M.-P. Billon, S. Pennel, P. J. McClure,
Hansson, and P. K. Bjomsen. 1998. Rapid determination D. P. Ferdinando, and M. B. Cole. 1995. The use of con-
of bacterial abundance, biovolume, morphology, and focal scanning laser microscopy (CSLM) to study the ger-
growth by neural network-based image analysis. Appl. mination of individual spores of Bacillus cereus. J.
Environ. Microbiol. 643246-3255. Microbiol. Methods 21:193-208.
An article for those with a greater interest in more sophisticated A good example of technical application of I P LSM and fho-
handling of images and data extruction. rescent labeling.
6. Blonk, J. C. G., A. Don, H. van Aalst, and J. J. 17. Cullander, C. 1994. Imaging in the far-red with electronic
Birmingham. 1993. Fluorescence photobleaching recovery light microscopy: requirements and limitations.J. Microsc.
in the confocal scanning laser microscope. J. Microsc. 176:281-286.
169:363-374.
52 MORPHOLOGY AND ULTRASTRUCTURE

18. De Beer, D., P. Stoodley, and Z. Lewandowski. 1997. 29. Lawrence, J. R., G. D. W. Swerhone, and Y. T. J. Kwong.
Measurements of local diffusion coefficients in biofilms by 1998. Natural attenuation of aqueous metal contamination
microinjections and confocal microscopy. Biotechnol. by an algal mat. Can. J. Microbiol. 44:825-832.
Bioeng. 53:151-158. The paper prooides a useful example of combining reflectance,
Excellent paper for those interested in calculation of diffusion fluorescence, and autofluorescence techniques.
from time course images of microinjected flwwconjugated 30. Lawrence, J. R., G. M. Wolfaardt, and T. R. Neu. 1998.
probes. The study of biofilms using confocal laser scanning mi-
19. Decho, A. W., and T. Kawaguchi. 1999. Confocal imag- croscopy, p. 431-465. In M. H. E Wilkinson and E Schut
ing of in situ natural microbial communities and their ex- (ed.), Modern Microbiological Methods Series, vol. 0 . Digital
tracellular polymeric secretions using Nanoplast resin. Analysis of Microbes. Imaging, Morphometry , Fluorometry
BioTechniques 27: 1246-1252. and Motility Techniques and Applications. John Wiley and
Useful starting point for combined use of hydrophilic resin em- Sons, Sussex, United Kingdom.
bedding and LSM . A comprehensive overview of applications of 1P LSM to mi-
20. Florijn, R. J., J. Slats, H. J. Tanke, and A. K. Raap. 1995 crobial biofilms .
Analysis of antifading reagents for fluorescence micro- 31. Lawrence, J. R., G. M. Wolfaardt, and D. R. Korber.
scopy. Cytometry 19:177-182. 1994. Monitoring diffusion in biofilm matrices using scan-
21. Haugland, R. P. 1998. Handbook of Fluorescent Probes and ning confocal laser microscopy. Appl. Enwiron. Microbiol.
Research Chemicals. Molecular Probes Inc., Eugene, OR. 60:1166-1 173.
Comprehensive guide to the fluorescent products provided by The first application of 1 P LSM to determine diffusion and pen-
Molecular Probes; includes much useful background informa- etrution of sizefractionated probes in complex biofilms.
tion, excitation-emission spectra, and literature. 32. Lawrence, J. R., D. R. Korber, G. M. Wolfaardt, D. E.
22. Hausner, M., and S. Wuertz. 1999. High rates of conju- Caldwell and T. R. Neu. 2001. Analytical imaging and
gation in bacterial biofilms as determined by quantitative microscopy techniques, p. 39-61. In C. J. Hurst, R. L.
in situ analysis. Appl. Environ. Microbiol. 65:3710-3713. Crawford, G. R. Knudsen, M. J. McInemey, and L. D.
A novel application of 1 P LSM for in situ analyses of single-cell Stetzenbach (ed.), Manual of Environmental Microbiology,
molecular events. 2nd ed. ASM Press, Washington, DC.
23. Heydorn, A., A. T. Nielsen, M. Hentzer, C. Sternberg, Comprehensive overview of digital microscopy as applied to a
M. Givskov, B. K. Ersboll, and S. Molin. 2000. variety of environmental samples.
Quantification of biofilm structures by the novel computer 33. Lawrence, J. R., D. R. Korber, B. D. Hoyle, J. W.
program COMSTAT. Microbiology 146:2395-2407. Costerton, and D. E. Caldwell. 1991. Optical sectioning
Paper that provides a good entry point into the use of statistical of microbial biofilms. J. Bacteriol. 173:6558-6567.
considerations in imaging of microbial biofilms. The first comprehensive study of biofilm architecture applying
23a.Hunter, R. C., and T. J. Beveridge. 2005. Application of 1 P LSM and digital image analyses.
a pH-sensitive fluoroprobe (C-SNARF-4) for pH microen- 34. Lisle, J. T., P. S. Stewart, and G. A. McFeters. 1999.
vironment analysis in Pseudomonas ueruginosa biofilms. Fluorescent probes applied to physiological characteriza-
Appl. Environ. Microbiol. 71:2501-2510. tion of bacterial biofilms. Methods Enzymol. 310:166-178.
A good example of ratiomenic applications of fluorescent pH A comprehensive owerwiew with practical protocols for applica-
reporters and useful controls for these studies. tion of a number of fluorescent reporters to microorganisms.
24. Kepner, R. L., and J.R. Pratt. 1994. Use of fluoro- 35. Liss, S. N., I. G. Droppo, D. T. Flannigan, and G. G.
chromes for direct enumeration of total bacteria in envi- Leppard. 1996. Floc architecture in wastewater and natu-
ronmental samples: past and present. Microb. Rew. 58: ral riverine systems. Environ. Sci. Technol. 30:680-686.
603-615. A useful example of the correlative microscopy approach.
Excellent owerwiew of the literature to 1994 on fluorochromes, 36. Liu, J., F. B. Dazzo, 0. Glagoleva, B. Yu, and A. K. Jain.
including applications and problems. 2001. CMEIAS: a computer-aided system for the image
25. Korber, D. R., J. R. Lawrence, M. J. Hendry, and D. E. analysis of bacterial morphotypes in microbial communi-
Caldwell. 1993. Analysis of spatial variability within motf ties. Microb. Ecol. 41:173-194.
and mot- Pseudomonas fluorescens biofilms using represen- Excellent publication prowiding information on ImageTool and
tative elements. Biofouling 7:339-358. examples of the application of digital imaging and software rou-
Paper describes application of geostatisticalmethods to biofilm tines in microbial ecology.
research and establishment of sampling requirements; also pro- 37. Manz, W., G. Arp, G. Schumann-Kindel, U. Szewzky,
vides images of montages of large biofilm areas. and J. Reitner. 2000. Widefield deconvolution epifluores-
26. Laca, A., L. A. Garcia, F. Argiieso, and M. Diaz. 1999. cence microscopy combined with fluorescence in situ hy-
Protein diffusion in alginate beads monitored by confocal bridization reveals the spatial arrangement of bacteria in
microscopy. The application of wavelets for data recon- sponge tissue. J. Microbiol. Methods 40:125-134.
struction and analysis. J. I d . Microbiol. Biotechnol. 23: Excellent coverage of deconwolution approaches, with exumples
155-1 65. and color illustrations using microbial specimens.
27. Lawrence, J. R., and T. R. Neu. 1999. Confocal laser 38. Manz, W., K. Wendt-Potthoff, T. R. Neu, U. Szewzyk,
scanning microscopy for analysis of microbial biofilms. and J. R. Lawrence. 1999. Phylogenetic composition, spa-
Methods Enzymol. 310:131-144. tial structure, and dynamics of lotic bacterial biofilms in-
A brief overwiew of how to do 1P LSM with microbial biofilms vestigated by fluorescent in situ hybridization and confocal
and flocs; also exumines specimen handling and upnght versus laser scanning microscopy. Microb. Ecol. 37:225-237.
inverted microscopes. 39. Massol-Deya, A., J. Whallon, R. F. Hickey, and J.
28. Lawrence, J. R., T. R. Neu, and G. D. W. Swerhone. Tiedje. 1995. Channel structures in aerobic biofilms of
1998. Application of multiple parameter imaging for the fixed film reactors treating contaminated groundwater.
quantification of algal, bacterial and exopolymer compo- Appl. Environ. Microbiol. 61:767-777.
nents of microbial biofilms. J. Microbiol. Methods 32: 40. Michael, T., and C. M. Smith. 1995. Lectins probe mo-
253-261. lecular films in biofouling: characterization of early films
Useful techniques paper describing how to use autoflumes- on non-living and living surfaces. Mar. Ecol. Progr. Ser.
cence, nucleic acid staining, and lectin staining with digital im- 119:229-236.
aging to quantify major biofilm components. One of the first applications of lectins in complex habitats.
3. Laser Scanning Microscopy 53

41. MGller, S., C. S. Kristensen, L. K. Poulsen, J. M. 53. Szmacinski, H., and J. R. Lakowicz. 1993. Optical
Carstensen, and S. Molin. 1995. Bacterial growth on sur- measurements of pH using fluorescent lifetimes and
faces: automated image analysis for quantification of phase-modulation fluorometry. Anal. Chem. 65:1668-
growth rate-related parameters. Appl. Environ. Microbiol. 1674.
61:741-748. Excellent starting point for understanding application of fluo-
42. Mqiller, S., D. R. Korber, G. M. Wolfaardt, S. Molin, and rescence life-time imaging.
D. E. Caldwell. 1997. The impact of nutrient composition 54. Ulrich, S., B. Karrasch, H. G. Hoppe, K. Jeskulke, and
on a degradative biofilm community. Appl. Environ. Mi- M. Mehrens. 1996. Toxic effects on bacterial metabolism
crobiol. 63:2432-2438. of the redox dye 5-cyano-2,3-ditolyltetrazolium chloride.
43. Neu, T. R. 2000. In situ cell and glycoconjugate distribu- Appl. Environ. Microbiol. 62:4587-4593.
tion of river snow as studied by confocal laser scanning mi- Good critical evaluation of the application of fluorescent probes
croscopy. Aquat. Microb. Ecol. 21:85-95. to assess bacterial metabolism.
Useful application publication illustrating information derived 55. Valkenburg, J. A,, C. L. Woldringh, G. J. Brakenhoff,
from 1P LSM based study. H. T. van der Voort, and N. Nanninga. 1985. Confocal
44. Neu, T. R., G. D. W. Swerhone, and J. R. Lawrence. scanning light microscopy of the Escherichia coli nucleoid:
2001. Assessment of lectin-binding analysis for in situ de- comparison with phase-contrast and electron microscope
tection of glycoconjugatesin biofilm systems. Microbiology images. 1. Bacteriol. 161:478483.
147~299-313. First application of confocal 1P LSM to study bacterial s ~ r u c -
Excellat example of an assessment of fluors and probes for use ture .
with 1 P U M , with additional infurmation on lectin applications. 56. Van Ommen Kloeke, F., and G. G. Geesey. 1999.
45. Palmer, R.,Jr., B. Applegate, R. Burlage, G. Sayler, and Localization and identification of populations of phos-
D. White. 1998. Heterogeneity of gene expression and phatase-active bacterial cells associated with activated
activity in bacterial biofilms, p. 609-612. In A. Rhoda, sludge flocs. Microb. Ecol. 38:201-214.
M. Pazzagli, L. J. Kricka, and P. E. Stanley (ed.), Example of the application of fluorescence imaging of alkaline
Bioluminescence and Chemical Luminescence: Perspectives for phosphatase activity in flocs.
the 21st Century. Proceeding of the 10th International 57. Vroom, J. M., K. J. de Grauw, H. C. Gerritsen,
Symposium on Bioluminescence and Chemiluminescence . D. J. Bradshaw, P. D. Marsh, G. K. Watson, J. J.
John Wiley and Sons, Chichester, United Kingdom. Birmingham, and C. Allison. 1999. Depth penetration
Useful example of correlative microscopy. and detection of pH gradients in biofilms by two-photon
46. Pawley, J. B. (ed.). 2006. Handbook of Biological Confocal excitation microscopy. Appl. Environ. Microbiol. 65:
Microscopy. Plenum Press, New York, NY. 3502-3511.
An excellent technical summary of all aspects of confocal mi- First substantial publication describing the use of 2P LSM to
croscopy. image microbial biofilms.
47. Perret, D., G. G. Leppard, M. Muller, N. Belzile, R. 57a.Wigglesworth-Cooksey, B., and K. E. Cooksey. 2005.
DeVitre, and R. Buffle. 1991. Electron microscopy of Use of fluorophore-conjugated lectins to study cell-cell in-
aquatic colloids: non-perturbing preparation of specimens teractions in model marine biofilms. Appl. Environ. Micro-
in the field. Water Res. 25:1333-1343. biol. 71428435.
Describes the use of Nanopht embedding techniques for deli- A good
. .
example of more extensive applications of lectins in ma-
cate microbial structures. rine habitats.
48. Podda, F., P. Zuddas, A. Minacci, M. Pepi, and F. Baldi. 58. Wiggli, M., A. Smallcombe, and R. Bachofen. 1999.
2000. Heavy metal coprecipitation with hydrozincite Reflectance spectroscopy and laser confocal microscopy
[Zn,(CO,),(OH),] from mine waters caused by photosyn- as tools in an ecophysiological study of microbial mats
thetic microorganisms. Appl. Environ. Microbiol. 665092- in an alpine bog pond. J. Microbiol. Meth. 34:173-
5098. 182.
49. Rocheleau, S., C. W. Greer, J. R. Lawrence, C. Cantin, Useful example of LSM imaging using reflectance, autofluores-
L. Laramee, and S. Guiot. 1999. Differentiation of cence , and staining to examine environmental samples.
Methanosaeta concilii and Methanosarcina barkeri in anaero- 59. Wolfaardt, G. M., J. R. Lawrence, R. D. Robarts, and
bic mesophilic granular sludge by fluorescent in situ hy- D. E. Caldwell. 1998. In situ characterization of biofilm
bridization and confocal scanning laser microscopy. Appl. exopolymers involved in the accumulation of chlorinated
Environ. Microbiol. 65:2222-2229. organics. Microb. Ecol. 35:213-223.
Excellent example of combined application of embedding, Application of fluor-conjugated lectins in complex biofilm sys-
rRNA probes, 1P LSM , and digital image analyses. tem with quuntification and analyses.
50. RUSS,J. C. (ed.). 2002. The Image Processing Handbook. 60. Wuertz, S., E. Muller, R. Spaeth, P. Pfleiderer, and
CRC Press, Boca Raton, FL. H.-C. Flemming. 2000. Detection of heavy metals in bac-
Excellent comprehensive and understandable publication on all terial biofilms and microbial flocs with the fluorescent
aspects of image processing. complexing agent Newport Green. J. Ind. Microbiol.
51. Sanders, R., A. Draaijer, H. C. Gerritsen, P. M. Houpt, Biotechnol. 24:116-123.
and Y. K. Levine. 1995. Quantitative pH imaging in cells A good practical example of the application of LSM and cali-
using confocal fluorescence lifetime imaging microscopy. bration of fluorescence imaging.
Anal. Biochem. 227:302-308. 61. Xiao, G. Q., and G. S. Kino. 1987. A real time confocal
Publication provides a basis for starting fluorescence lifetime scanning optical microscope. Proc. SOC. Photo Opt.
imaging using 1P LSM systems. Instrum. Eng. 809:107-113.
52. Sytsma, J., J. M. Vroom, C. J. de Grauw, and H. C. Presents a useful, simple equation for estimation of optical sec-
Gerritsen. 1998. Time-gated fluorescence lifetime imaging tion thickness.
and microvolume spectroscopy using two-photon excita- 62. Yu, F. P., G. M. Callis, P. S. Stewart, T. Griebe, and
tion. J. Microsc. 191:39-51. G. A. McFeters. 1994. Cryosectioning of biofilms for mi-
Provides current application of 2P LSM in conjunction with croscopic examination. Biofouling 8:85-91.
fluorescence lifetime imaging. An early example of this now-standard method.
.

Electron Microscopy
TERRY J . BEVERIDGE. DIANNE MOYLES. AND BOB HARRIS

4.1. INSTRUMENTATION ...................................... 56


4.1.1. Microscopy Procedures .................................. 56
. ........... 57
2. Transmission Electron Microscope and Resolving Power
..
3. Scanning Electron Microscope............................. 58
........................... 58
4. Specialized Electron Microscopes
4.1.4.1. STEMS ........................................ 58
. 2. Specialized Configurations for Spectroscopy and Other
..............................
Types of Microscopy 59
4.2. TECHNIQUES FOR TEM .................................... 61
4.2.1. Preparation ........................................... 61
4.2.1.1. TEM Grids..................................... 61
. .......................
2. Support Films for TEM Grids 61
4.2.2. Negative Staining ...................................... 62
....................................
4.2.2.1. Stain Choice 62
. ...............................
2. Staining Procedures 62
. .......................................
3. Washing 63
. ........................................
4. Pitfalls 63
. ....................................
5. Expectations 63
4.2.3. Thin Sectioning ....................................... 64
....... 64
4.2.3.1. Fixation with Glutaraldehyde and Osmium Tetroxide
. .........
2. Dehydration and Embedding with Plastic Medium 65
. ................. 65
3. Other Fixatives and Embedding Media
. .................................
4. Block Trimming 65
. .................................
5. Thin Sectioning 65
. ........................................
6. Knives 65
. ..........................
7. Staining of Thin Sections 65
4.2.4. Cryofixation and Freeze-Substitution........................ 66
.5. Metal Shadowing ....................................... 67
4.2.5.1. Shadow Casting of Whole Mounts .................... 67
. .................. 67
2. Freeze Fracturing and Freeze-Etching
. 3. Rotary Shadowing of DNA and RNA .................. 69
4.3. IMAGE ACQUISITION ..................................... 69
4.3.1. Film ................................................ 69
.2. Printing ............................................. 69
.3. CCD Cameras
4.4. INTERPRETATION
......................................... 69
........................................ 69
.
.
5. SELECTION OF THE BEST MICROSCOPY TECHNIQUE
6. MACROMOLECULE IDENTIFICATION TECHNIQUES
.......... 70
............ 70
4.6.1.PCF ................................................ 70
.2. Colloidal Gold-Antibody Probes............................ 70
4.6.2.1. Probe Production................................ 70
. ...........................
2. Direct Antigen Labeling 72
. ..........................
3. Indirect Antigen Labeling 73
. .......................
4. Thin-Section Antigen Labeling 73
4.7. MATERIALS .............................................. 74
4.7.1. Plastic Support Films.................................... 74
.2. Carbon Films ......................................... 75
.3. Negative Staining...................................... 75
.
4.8. COMMERCIAL SOURCES
...................
4. Fixing. Embedding. Sectioning. and Staining
...................................
75
76
.9. REFERENCES ............................................. 76
4.9.1. General References ..................................... 76
. .............................
2. Specific References by Subject 77
............. 77
4.9.2.1. Grids. Support Films. and Negative Staining
. ............................
2. Sectioned Preparations 77
. 3. Low-Temperature Embedding Resins .................. 77
54
4. Electron Microscopy w 55

.4. Visualizing Macromolecules by Using Metal Shadowing .... 77


.................
.5. Freeze Fracturing and Freeze-Etching 77
.6. Cryotechniques ................................. 78
.7. PCF ......................................... 79
.8. Immunogold Labeling ............................ 79
.9. Compositional Analysis ........................... 79
.lo. Image Processing ................................ 80
.ll.SEM ........................................ 80
.12. STM and AFM ................................. 80
4.9.3. Specific References on Interpretation ........................ 80

Microbiologists are dedicated to the study of very small tion of a conceptual bridge between cellular functions, the
cells. Today, as molecular biology is becoming an increas- chemistry of the parts, and the behavior of the macromole-
ingly important research tool for microbiology, microscopy cules that make up structure (Fig. 1 and 2). All students of
(especially high-resolution microscopy) is becoming an biochemistry, molecular biology, biotechnology, industrial
even more important implement for the identification of processes, and environmental studies really do require a mi-
subtle cellullar changes and cloned products. Unfor- croscopic view at one time or another. Accordingly, we also
tunately, this emphasis on molecular biology has, somehow, aim our chapter at these casual users as well as at neophytes
deterred many younger workers from recognizing the im- at EM. Simple, routine types of EM are described together
portance of microscopy in their molecular studies. Imagine! with hints based on experience of how best to go about
It is not unusual to enter a modern microbiological labora- preparing samples and maintaining equipment. The chapter
tory today and discover that no light microscope can be provides a guide to the kind of information that can be ob-
found, or to enter a department of microbiology and find tained from basic techniques and suggests combinations of
that n o electron microscopy (EM) facility exists. Yet, vi- techniques for more complete information.
ruses, bacteria, archaea, and most eucaryotic microorgan- The first level of ultrastructural information is provided
isms cannot be seen as individual cells (or particles) with- by transmission electron microscopy (TEM) and scanning
out the aid of, at least, a light microscope. Few dedicated
microbiological EM facilities remain, and microbiologists
are becoming more and more dependent on generic college,
university, or outside facilities that possess little experience
in microbes at the levels of both specimen processing and
the interpretation of results. It is analogous to taking a sore
tooth to an auto mechanic to be repaired instead of to a
dentist. Our own experience suggests that this neglect of
both light microscopy and EM in present-day studies is
having grave consequences for such disparate but important
aspects as culture contamination and proper identification
of molecular alterations in cells. For these reasons, mi-
croscopy in one form or another is inescapable. Researchers
who do not routinely examine their cultures under a micro.
scope run the very real risk of drawing conclusions based on
results from contaminated cultures. Chapters 1 and 2 have
explained the tried and true uses and advantages of light
microscopy, and this chapter deals with EM. It is our hope
that this chapter will explain some of the mysteries of EM
as well as make the various techniques more user friendly to
researchers who have lost the skills and recognized the im-
portance of its use.
Of all the research techniques available to microbiolo-
gists, EM is the only one that can accurately give shape and
form to cells and their component parts and identify posi-
tions of ultrastructural components (such as ribosomes, bi-
layers, and plasmids) and the viruses that afflict cells. There
has been a renaissance of interest in EM since new prepara-
tory procedures and equipment have made possible extreme
resolution with well-preserved material. The chromosomes FIGURE 1 Thin section of an unidentified gram-negative
and ribosomes are found to be dispersed throughout most of bacterium fixed via the glutaraldehyde-osmiumtetroxide pro-
the cytoplasm, and enveloping layers are more complicated tocol (section 4.2.2.1) showing the gram-negative envelope,
than was first suspected. The accurate identification and de- condensed nucleoid, and clustered ribosomes produced by this
termination of macromolecular distributions have provided method. An S-layer and capsule or pili (fimbriae) are also pres-
a better understanding of the integrative steps required in ent. With the thin sectioning technique, it is difficult to differ-
cellular metabolic processes and of their alteration during entiate between a capsule and pili, and a negative stain would
changes in growth or during division. EM assists the forma- help in identification. This is a TEM image.
56 MORPHOLOGY AND ULTRASTRUCTURE

know what sort of information can be expected from these


major techniques and to be aware of their limitations.
There are a number of EM books available and recom-
mended (1-1 l ) , but nothing can replace the experience of
the regular operators of a microbiological EM unit. The in-
struction from and the friendship of these highly qualified
personnel should be nurtured. No chapter or textbook on
EM can make you an expert microscopist; hands-on experi-
ence with guidance from someone who knows is essential.
Many new and complex derivatives of EM are available,
all with impressive and complicated names, e.g., scanning
transmission electron microscopy, energy-dispersive X-ray
spectroscopy (EDS), electron energy loss spectroscopy
(EELS), electron spectroscopic imaging (ESI), cryoTEM,
and cryoSEM; often these techniques can be intermixed
with one another (e.g., cryoTEM-EELS-ESI). Some micros-
copies cannot even be considered to involve typical lens-
style microscopes, and they depend on the physical probing
of specimens with sharp, pointed tips, thereby developing a
tunneling current (scanning tunneling microscopy [STM])
FIGURE 2 TEM image of a thin section of a freeze- or detecting atomic weak bonding forces (atomic force
substituted E. coli K-12 cell showing a well-preserved cyto- microscopy [AFM] and its derivatives). STM and AFM are
plasm and cell envelope. Compare the cytoplasm and cell en- described in more detail in chapter 6. To add to the confu-
velope of this cell with those shown in Fig. 1. (Reprinted from sion, new specimen-processing methods abound, e.g., cry-
reference 8 with permission of the publisher.) ofixation by freeze-plunging or freeze-slamming; freeze-
substitution; the use of thin, frozen films; and cryosectioning.
Preparative techniques allowing the localization of compo-
nent macromolecules have also become crucial. All are
electron microscopy (SEM). These are tried and true tech- highly specialized, requiring dedicated equipment and expe-
niques that have, over the last 40 years, deciphered the rienced personnel, but can be made available through col-
shapes, sizes, constituent arrangements, and cellular inter- laborative ventures.
actions of a great many bacteria (78, 81-83). Of the two
microscopies, SEM is better for determining the surface ap+
pearance or topography of cells but usually does not have 4.1. INSTRUMENTATION
enough resolving power to elucidate more than general fea-
tures (Fig. 3 ) . SEM is a good technique for deciphering cell- 4.1 .l.Microscopy Procedures
cell associations, growth habits, and the topography of cell Any electron microscope (Fig. 4) is an extremely compli-
aggregates such as biofilms. TEM offers greater resolving cated and expensive instrument. It is looked after by spe-
power for a variety of preparations and is capable, with suit- cialists and (usually) maintained, because of the expense of
able material, of detecting the positions of cellular parts and labor, by service contracts with the manufacturer.
macromolecules (Fig. 1 and 2 ) . Many procedures are suit- Most instruments have multiple users to justify the capital
able for TEM of bacteria, including negative staining, thin expense and complex operating problems. EM facilities often
sectioning, and the use of shadowed replicas; most of this provide thin sectioning, shadow casting, freeze-etching, and
chapter concentrates on these procedures. It is important to perhaps other kinds of assistance. Workers in charge of

FIGURE 3 SEM image of a gold-sputtered Chlorobium sp. FIGURE 4 Transmission electron microscope being oper-
covered with special surface appendages called spinae (arrow). ated by one of the authors (R. Harris) in the EDS mode to de-
Bar = 500 nm. (Reprinted from reference 8 with permission of termine the elemental composition of a natural bacterial com-
the publisher.) munity.
4. Electron Microscopy 57

these units establish rules and standards of operation, which mum. Cell contrast is poor because carbon, hydrogen, oxy-
protect the integrity of the equipment and the work that gen, and nitrogen (which make up most cellular mass) scat-
comes out of it, and are strict in enforcing them. Experience ter electrons poorly, and in comparison to resolution, cell
says that casual users of an electron microscope fit the de- contrast decreases with an increase in accelerating voltage
scription given by a museum guard of the museums young (high kilovoltages are used to penetrate thick specimens).
patrons: Lady, they will do anything. To this one must add, Consequently, the specimen must be as thin as possible and
And not tell you what it was! To assist in troubleshooting selectively stained or contrasted by using heavy-metal salts
and maintenance, each user must be considerate of the in- that increase the scattering power. Instruments in current
strument (some parts are delicate, and it does matter which use achieve enormous direct magnifications, but unless
knobs you twist) and of the next user (a golden rule) and there is a need to resolve macromolecules, most applica-
must be scrupulously honest about anything done or not tions are well served by magnifications of X5,OOO to
done. x 15,000 (allowing subsequent photographic or computer
Some procedural points to remember are as follows. enlargements of x5 to x 10). Higher magnifications require
more skill and a very well tuned instrument.
1. Get checked out on every instrument and procedure The value of TEM in microbiology is its outstanding
to know what to do and your limitations.
ability to elucidate details of cell structure, provided that
2. If equipment does not work, behaves strangely, or re-
the preparative methods and the choice of EM technique
quires unusual force, stop the equipment in a safe position,
are suitable to the structure involved, e.g., the use of heavy-
think, and seek help.
metal salts to surround the bacterium and penetrate into
3 . Do not be tempted to undertake repairs. Own up and surface irregularities supplies contrast by differentially im-
confess the problem. peding electrons, producing the effect of negative staining
4. Keep records, even if that is not the habit of the unit, (section 4.2.2), analogous to the use of nigrosin in light mi-
and see that operating times are recorded and that every
croscopy (chapter l). However, unless there are breaks in
malfunction is reported.
the cell wall or membrane to allow penetration of the metal
5. Make notes of the steps in procedures, have them at salts, no intracytoplasmic features can be seen. Macromo-
hand when at work, and use a checklist, which should in-
lecular arrangements and cellular components can be stud-
clude the following: positions of switches and settings at
ied if cells are broken and fractionated (chapter 7) before
start-up; start-up procedures; readiness requirements; align-
negative staining (Fig. 5).
ment procedures for everyday operation for condenser ad-
Microbes are too thick for adequate resolution of inter-
justment, centering of the objective aperture, and astigma-
nal structures, but thin sectioning (into slices 50 to 100 nm
tism compensation; the setting for wide-field scanning;
thick) of chemically fixed and stained cells embedded in
operating procedures and camera operation; warning signs
plastic allows their cytoplasmic arrangements to be dis-
and responses to them; standby and safety actions; the turn-
cerned (Fig. 1 and 2). In this technique, heavy-metal stains
off procedure; and positions of switches and settings after
of the sections are needed to positively stain the cells (as op-
turn-off.
posed to negative staining) and provide differential contrast
Good preparations, procedures, patience, and opera- for imaging. With bacteria, a thin section will show the
tional skills will be rewarded by good micrographs and will arrangements of DNA, ribosomes, and cytoplasmic granules
be appreciated by both the supervisor and the microscope. as well as the juxtapositioning of cell envelope layers (Fig.
4.1.2. Transmission Electron Microscope
and Resolving Power
The value of a transmission electron microscope (Fig. 4) lies
in its capacity to resolve objects that cannot be resolved by
any form of light microscopy. The wavelength of electrons,
which is shortened as the accelerating voltage i s increased,
permits the resolution of objects as small as 2 A (0.2 nm or
0.0002 pm), whereas the limit of light microscopy is close
to 0.2 km. With biological specimens, resolutions of -1.0
nm are achieved under only ideal conditions of operation,
since cells are not infinitely thin and their substance con-
tributes to noise. The practical limit of real resolution,
e.g., with thin biological membranes such as bacterial S-lay-
ers, is about 1.0 to 1.5 nm. Even then it takes real skill and
a well-tuned instrument to do better than 2.0 to 2.5 nm.
Thick specimens and support films increase electron scat-
tering, and the resolution deteriorates accordingly.
It is useful to recall some of the limitations of TEM.
Because specimens must be examined in a high vacuum to
permit electron flow, cells need to be dried (unless they are
held frozen [i.e., vitrified] in a special cryospecimen holder
at extremely low temperatures). In addition, typical accel-
erating voltages (60 to 120 kV) subject the specimen to so
much energy that chemical bonds (including covalent link- FIGURE 5 TEM image of a negatively stained preparation
ages) can be broken, molecular conformations can be al- of isolated membrane vesicles, pili, and pyocin particles from
tered, and substantial organic mass can be lost. Specimen Pseudomonas aeruginosa. Negative stains are particularly good
exposure to the electron beam should be kept to a mini- for visualizing small particles for identification.
58 MORPHOLOGY AND ULTRASTRUCTURE

1 and 2 ) . As mentioned previously, cellular elements (C, H, not as expensive as transmission electron microscopes and
0,and N, etc.) do not scatter electrons proficiently. Most of are relatively easy to maintain and operate, and because
the electrons of the electron beam pass cleanly through a their sample preparatory procedures are relatively straight-
specimen, and these unscattered electrons do not carry forward, they are more frequently encountered than trans-
structural information. However, some electrons do have mission electron microscopes by microbiologists. They are
their trajectories altered as they pass through the specimen more likely to be used for a first look by students of micro-
in the transmission electron microscope by elastic or inelas- bial structure. Environmental scanning electron micro-
tic scattering; this scattering by structures in the specimen scopes are becoming more readily available so that even hy-
provides information about the specimen. The elastic scat- drated specimens can be viewed, which gives a tremendous
tering is responsible for the high fidelity of TEM images. advantage over dried, coated specimens and provides a won-
Electron beams are usually generated by heating a derful glimpse of microorganisms existing under quasinatural
pointed tungsten filament to a high temperature in the conditions (Fig. 6). Cryostages also provide a means of look-
electron gun at the top of the of the transmission electron ing at frozen, hydrated specimens. Yet, resolving power and
microscope column and focused by powerful symmetrical restriction to topography make scanning electron micro-
electromagnetic lenses that are stacked on top of one an- scopes only partially useful for an encompassing EM analysis
other in the column. More sophisticated electron guns can of microbial cells, and TEM must be, at some point, always
be outfitted with a lanthanum hexaboride (LaB6) crystal to considered. However, field emission and low-voltage systems
give a more brilliant coherent electron beam. Essentially, are rapidly increasing the resolution gap between SEM and
the column of a transmission electron microscope consists TEM. SEM remains a useful tool for looking at microorgan-
of condenser lenses (which focus the electron beam on a isms in situ (e.g., to examine surfaces in natural environ-
small area on the specimen) and magnifying lenses (func- ments for adhering microorganisms) (Fig. 6).
tioning as objective, diffraction, intermediate, and projec-
tor lenses) immediately below. Electrons cannot be seen, 4.1.4. Specialized Electron Microscopes
and reliance is placed on a phosphorescent screen, which is This section is intended to identify and give a general ex-
excited by the incident electrons, to emit visible light. The planation of highly sophisticated EM equipment and tech-
emitted light intensity is low, and a darkened room is re- niques but not procedures or methods for their use. The
quired to clearly see the images. Electron-sensitive photo- equipment and techniques have been chosen because they
graphic film or a charge-coupled device (CCD) camera is are important to research in microbiology. Special equip-
necessary to record the images. ment and expert operators are required, so suitable problems
must be taken to them. However, it is necessary to have a
4.1.3. Scanning Electron Microscope broad familiarity with them so as to know what they may be
In SEM, the electron beam rapidly scans the surface of the able to contribute to a research project.
specimen to induce radiation of low-energy secondary elec-
trons or higher-energy backscattered primary electrons. 4.1.4.1. STEMs
These electrons form an image on a cathode ray tube by Scanning transmission electron microscopes (STEMs) come
synchronizing the movement of the electron beam in the mi- bine some of the attributes of SEM and TEM; as in SEM, a
croscope (and the information being collected) with the narrow electron beam is scanned back and forth over the
information displayed (during a raster) on the tube; the specimen; as in TEM, a transmitted signal can be recorded.
process is similar to the formation of a television picture. The great advantage of a STEM is that a highly concen-
The specimen must first be chemically fixed, dried (best by
criticalepoint drying or freeze-drying to avoid distortion),
and given a thin metal coating to make it conductive (sput-
tered in vacuo with gold, platinum, or a 60:40platinum-
palladium mixture; the last of these gives a less granular
metal layer). The resolution achieved by SEM has been
markedly improved since its introduction (at best, -30 nm
in commercially available models) but is still lower than
that routinely available by TEM. Newer low-voltage and
field emission scanning electron microscopes are beginning
to approach TEM resolution, and environmental scanning
electron microscopes can view specimens under high rela-
tive humidities. A scanning electron microscope can dis-
play only the images of surfaces, but it does so in a three-
dimensional mode that avoids the tedious preparation of
the replicas of intact, fragmented, or frozen and fractured
bacteria used in TEM for similar purposes. In bacteriology,
SEM is most useful for discerning surface appendages and
other surface structures (Fig. 3) and for defining shape and
topographic relationships such as those in colonies or on FIGURE 6 ESEM image of a wet and unaltered sample of a
surfaces of infected tissues (70-73). biofilm growing in a sulfur spring located near Ancaster,
Preparation of specimens for SEM involves fixation on a Ontario, Canada. Double arrows show the chains of cells
conductive surface such as graphite that can be affixed to (cyanobacteria) growing in the biofilm which are partially ob-
the specimen holder. Heavy-metal coating done under vac- scured by exopolymeric substances. The single arrow points to
uum prior to examination is usually required to avoid spec- a break in the exopolymeric substances. This image was kindly
imen charging and to give a strong surface signal for imag- supplied by S. Douglas of the NASA-Jet Propulsion
ing. Because scanning electron microscopes are (usually) Laboratory, Pasadena, CA.
4. Electron Microscopy 59

trated and narrow electron beam can be directed onto and detector is separated from the high vacuum of the electron
through a thicker than normal specimen. Because STEMSuse microscope column by a small borehole covered with a thin
a scanning mode, secondary and backscatter electron de- foil (usually of beryllium, germanium, or aluminum) which
tectors can be placed above the specimen so that, at a flick limits access to X rays with energies above those of the foil.
of a switch, a STEM becomes a scanning electron micro- For this reason, low-atomic-number elements (e.g., C, H,
scope. Secondary electrons produce topographical images of N, and 0)are not easily resolved, although recent advances
the specimen, whereas the backscattered electrons provide with plastic foils and windowlessdetectors are making the
density information. If an energy-dispersive X-ray spec- detection of such elements feasible. The borehole limits the
trometer (see below for more details) is attached to a number of X rays from regions other than the sample and
STEM, a compositional analysis of the elements in the the foil prevents the lowest-energy X rays from flooding the
specimen can be obtained. By selecting the energy line of a detector. EDS has been used to great advantage for the
single element (e.g., Ca or Mg) and using a computer to identification of fine-grained minerals formed on bacterial
combine the data with raster coordinates obtained in the surfaces by biogeochemical means in the new field of geo-
STEM mode, a point map of the distribution of that ele- microbiology (Fig. 7) (61).
ment can be obtained (63,64,68,69). Magnetosomes (Fe), WDS is another technique for obtaining elemental
polyphosphate (P) and sulfur (S) granules, and bacterium- analysis; it is restricted to scanning electron microscopes
associated minerals such as clays (Si) can be readily identi- and relies on physical crystals (e.g., those of lithium fluoride
fied. The detection of toxic heavy metals complexed to bac- or lead stearate) to separate and expand the X-ray emission
teria from the environment (61) has provided some spectrum focused on the detector. Analysis of the spectrum
exciting examples of structural analysis by using STEMs. provides compositional analysis. Compared with EDS,
WDS is rarely used on microbiological samples since it re-
4.1.4.2. Specialized Configurations for Spectroscopy quires long counting times and can be used only if the ma-
and Other Types of Microscopy terial is very stable under the electron beam.
There are three basic ways to obtain elemental analyses by EELS requires a detector underneath the specimen (usu-
EM: EDS (Fig. 4), wavelength-dispersive X-ray spec- ally below the viewing screen) which is aligned to the opti-
troscopy (WDS), and EELS. The application of EDS has al- cal axis of the electron beam and is, therefore, restricted to
ready been mentioned (section 4.1.4.1). This can be done a transmission electron microscope or a STEM. Only the
by SEM but is better done by scanning transmission elec- inelastically scattered electrons are used for EELS. These
tron microscopy because higher-resolution (elemental) electrons have lost energy after atomic interaction with the
point mapping can be achieved for single elements. A trans- specimen which results in lower energies and longer wave-
mission electron microscope with an EDS detector will lengths; both of these traits provide elemental signatures of
serve if no elemental imaging is required and if a full- the atom(s) with which interaction occurred. Although
spectrum analysis is satisfactory. As a specimen is irradiated these inelastic electrons lessen the quality of a TEM image
by the electron beam, its constituent atoms emit signature by producing chromatic aberration, they are very useful for
X rays, and the X-ray emission spectrum is read by a detec- compositional analysis. Since the EELS detector operates in
tor placed above and to the side of the specimen. The X-ray the vacuum of the electron microscope column without

FIGURE 7 Unstained thin section of a Pseudomonas aeruginosa cell which has accumulated a lan-
thanum mineral phase on its surface. The lower inset uses SAED to show that it is a crystalline min-
eral phase due to the periodicity of the reflections in the diffractogram. The inset to the right is an
EDS spectrum showing a high La concentration. The P and 0 could be due to the mineral, the cell,
or the plastic. The high Cu is from the copper TEM grid. (From S. Langley and T. J. Beveridge, Can.
J. Microbiol. 45:616-622, 1999.)
60 MORPHOLOGY AND ULTRASTRUCTURE

metal foil filters (unlike the EDS detector), the technique seen. No matter what the resolving power of the micro-
can resolve a full spectrum of elements, including those of scope, there must be enough signal for our eyes to readily
low atomic number. Compared with EDS, short counting decipher an object and, hopefully, to see its macromolecu-
times (10 s versus 100 s) are required but high doses of en- lar structure. In the early 1970s, optical diffractometers
ergy are required; highly beam-sensitive bacteriological using lasers (as a coherent light source) and images of peri-
samples should not be analyzed unless low-temperature odic structures (as diffraction grids) were used to painstak-
(cryo-) stages are used to stabilize the specimen. However, ingly reconstruct images through mechanical means. The
so far, EELS is the best technique for analyses of elements of easy use of computers and the digitization of images has
low atomic number. caused a major improvement in image processing. Selected
One of the most exciting advances in TEM uses a mod- negatives are digitized by using either a high-resolution tel-
ified version of EELS and is called ESI (62, 66). Two con- evision camera, a CCD camera, or a narrow-beam flat-bed
figurations are possible; one is a below-column spectrometer scanner, and the information is then fed into a computer.
resembling that described above but with additional lenses Over the last few years, great advances have been made
and a filter attached. This configuration suffers from the dis- with CCD cameras with improved digitization. Two basic
advantage of a small sampling size, and the large number of computer programs are used: Fourier analysis, which deals
lenses makes image acquisition difficult. Its advantage is with highly periodic objects, and correlation averaging,
that this ESI spectrometer can be added to most modern which is used for more irregularly shaped objects (such as ri-
transmission electron microscopes. The other configuration bosomes) (Color Plate 7) or periodic objects with lattice de-
requires a more dedicated instrument and relies on an in- fects. The structures of such diverse paracrystalline objects
column prism which allows for a larger sampling size and as viruses, bacteriophages, flagella, pili, Slayers, recombi-
fewer lenses. Each ESI configuration separates the energy nant protein crystals, and parasporal bodies (Fig. 11 in
lines in the inelastic signal and allows the operator to chapter 5 shows a reconstruction of an S-layer) and such ir-
choose a single elemental signal as the sole information sig- regular particles as flagellar basal bodies and eucaryotic, ar-
nal to use in the formation of a TEM image. As a conse- chaeal, and bacterial ribosomes have been reconstructed by
quence, an elemental image at TEM resolution (- 1.0 nm) using computer-based techniques. Chapter 3 provides a
is formed which has a much higher resolution than a point more in-depth treatment of computer-based reconstruction
map formed by scanning transmission electron microscopy- techniques.
EDS. This technique is complicated, requiring great exper- Cryo-transmission electron microscopes are relatively
tise, and has only rarely been used in microbiology. rare in microbiology, but they are rapidly revealing many se-
Remarkably, the configuration of rRNA has been visualized crets about microbial structure. These exotic microscopes
in both small and large subunits of ribosomes by following are difficult to maintain, and the preparation of specimens
the phosphorus line of the rRNA (Color Plate 7). By using is more of an art than a recipe. The specimen holder and
the same phosphorus line, the bilayer profiles of mem- specimen chamber are maintained at liquid-nitrogen or
branes, such as plasma membranes and outer membranes, liquid-helium temperature so that snap-frozen specimens
can also be imaged because of the phosphorus in phospho- remain in a vitrified state (see section 4.2.4). Thin, frozen
lipids and lipopolysaccharides. Because of the need for ex- foils of small particles (e.g., membranes or ribosomes) (37d)
tremely fine compositional analyses and because microbes or cryosections of microorganisms (37a-d) can be exam-
are so small, ESI holds tremendous promise for TEM analy- ined. Here, no heavy-metal stains can be used and we must
sis in microbiology. rely on the actual mass and its differential organization
A comprehensive list of articles describing these meth- within the section or foil for visualization against the over-
ods of elemental analysis and their use in microbiology can all mass of the frozen water in which the cells or particles
be found in references 60 through 69. are embedded. It is remarkable how well the cryoimages of
Diffraction is another method for structural analysis but bacteria compare with those from freeze-substitution or,
is applicable only to crystals and highly ordered macromo- even, conventional embedding. Subtle differences in shapes
lecular arrays. X-ray diffraction, which uses a beam of X and dimensions of cell envelopes, ribosomes, and nucleoids
rays, has been influential in determining molecular struc- are seen (37a, b, and c) but general shape and form is main-
ture at the atomic level and in understanding the molecular tained. One of the most drastic differences is that a definite
conformations of proteins and nucleic acids. Like X rays, periplasmic space is seen in gram-positive bacteria (37b and
electrons diffract, and a transmission electron microscope c). New, very sophisticated cryo-transmission electron
can be used as an electron diffractometer by focusing inci- microscopes are now becoming available that are capable
dent electrons on extremely small areas. If the magnifying of doing tomography (producing a tilt series that can be
lenses are turned off in TEM for the viewing of a periodic used to produce a computer-generated three-dimensional
object, an electron diffraction pattern will be obtained. image).
Since the electron beam is concentrated on a small region Scanning probe microscopy (SPM), which includes
of the specimen, this technique is called selected-area elec- STM and AFM, is unlike any previously described mi-
tron diffraction (SAED). Detection of high-level periodic- croscopy. Here, no optical lenses are used, but instead, an
ity in most microbiological specimens is quite sensitive to atomically sharp tip (usually fabricated from silicon nitride
electron bombardment, and SAED is restricted to biostruc- [SijN3]) attached to a cantilever of known rigidity is
tures with a high proportion of covalent bonding, to inor- dragged over the surface of the specimen. The up and down
ganic mineralized material such as magnetosomes (78), and movement of the tip provides topographical (z-axis) detail,
to external mineral deposits (61). The combination of EDS and (as in SEM) as it is rastered over x,y coordinates, a
or EELS with SAED is a most powerful technique for the three-dimensional image of the surface is generated (Fig. 5
analysis of fine-grained minerals associated with microor- in chapter 6). STM and AFM can provide atomic detail of
ganisms (61). hardy, robust inorganic samples (e.g., minerals and metal
A major challenge with imaging by EM is to obtain a foils), but biological samples are orders of magnitude more
good signal-to-noise ratio so that structure can be clearly difficult. Yet, since AFM can be done under water, it holds
4. Electron Microscopy 61

great promise for microbiology; chapter 6 provides an in- may be stored indefinitely by being wrapped in folds of lint-
depth treatment of these new, exciting SPMs. free tissue.
4. Fill a Coplin jar two-thirds full with the working solu-
tion of 0.2% Formvar@.
4.2. TECHNIQUES FOR TEM 5. Place two or three clean slides in the Coplin jar con-
This section describes a limited number of TEM methods taining the Formvar@solution, immersed to three-quarters
that, from our experience, are considered useful to the of their length. Allow them to stand for 3 to 5 min, with-
worker who has need of basic structural information about draw one slide at a time with a slow, even motion, and hold
microorganisms. Although we refer directly to procaryotes the slide just above the solution for 10 s to allow excess to
in our descriptions, most of the general principles are also drain off in the solvent atmosphere. Now, completely with-
suitable for eucaryotic microbes. Emphasis is placed on the draw the slide and place it standing on lint-free tissue for
use of negatively stained preparations and thin-sectioned about 30 s allowing it to dry completely. With an alcohol-
materials for examination by TEM. No attempt is made to cleaned razor blade, score the Formvar@film around the en-
describe the operation and maintenance of the microscope tire perimeter of the slide. Be careful that small glass chips
(4, 6, 10) or other associated instruments. Materials that do not land on the Formvar@film!
are required are mentioned (section 4.7), and their sources 6. Place a 6-in. (ca. 15*cm)-high,flat-bottomed, glass
are given (section 4.8). It is assumed that instruments are crystallizing dish on black paper. Fill the dish almost to the
functional and that experienced operators are available for top with deionized water. Take a lint-free tissue, and drag it
assistance and instruction. The aim here is to supply the across the surface of the water to remove dust particles.
neophyte with helpful information on techniques and in- 7. While holding the coated slide at a 45 angle over the
terpretation. General reading is provided in references 1 dish, slowly immerse the slide into the water (breathing on
through 11, and more specialized sources of information are dry Formvar@coatings helps to free them from the glass).
given in references 12 through 100. The Formvar@film will slowly release from the glass surface
where you have made your cuts and float off onto the water
4.2.1. Preparation surface. Inspect each film visually to judge uniformity and
4.2.1 . I . TEM Grids thickness; they should appear gray to silver-gray, represent-
ing a thickness of -60 nm according to interference color
See references 4 through 7 and 12.
charts. The theoretically ideal thickness of 10 to 20 nm is
Specimens in the form of thin sections, intact organisms,
rarely achieved. If uniformity or thickness of the films is un-
and cell components must be supported in the electron
satisfactory, repeat the above-decribed steps with appropri-
beam in accurate relation to the objective pole pieces of the
ate modifications, as follows. If the thickness of the film is
microscope. This support has to be relatively transparent in
not uniform, then the slide was taken out of the solution too
the electron beam and is provided by an ultrathin film (sec-
quickly or did not have enough time to drain in the solvent
tion 4.2.1.2). To accomplish this, thin metal disks with a
atmosphere. If the film is too thick, dilute the Formvar@so-
regular pattern of holes (grids) are usually covered with thin
lution with solvent. If film is too thin, increase the per-
plastic or carbon films. Standard grids, of a diameter (2.3 or
centage of Formvar@to 0.3% and repeat the process.
3.0 mm) appropriate to the microscope used, are commer-
cially available (see section 4.8, items k, 0, p, r, u, x, 2, and
8. Once satisfactory films are obtained on the water sur-
face, use narrow-pointed forceps (Dumont no. 5) to care-
cc). Grids are made of several materials and are available in
fully align the grids (dull side down) onto the films. To
several mesh sizes, but the most commonly used grids are of
maintain a degree of consistency, avoid using film areas
copper and have a mesh size of 200 (mesh size indicates the
with obvious irregularities. Usually a film will accommodate
lines per inch). The grids have a dull (matte) side and a
25 to 50 individual grids.
shiny (bright) side. Before support films are applied, the
grids must be clean and therefore must be treated with ace-
9. To retrieve the film-covered grids, cut a circular 9.0-
cm-diameter filter paper (Whatman no. 1) into strips larger
tone and/or other solvents; however, supply houses can fur-
than the size of the film containing the grids and bend up
nish precleaned grids.
one end of the filter paper strip (0.5cm). While gripping
4.2.1.2. Support Films for TEM Grids the bent portion of the filter paper strip, hold the paper over
the Formvar@-coveredgrids and lower it gently onto them
Most thin sections as well as suspensions of bacterial cells
until the paper has uniformly absorbed water. Just before the
for metal shadowing or for negative staining must be sup-
filter paper is completely wet, lift it quickly from the surface
ported on thin films covering the holes in the grid. The film
of the water (some filter papers are so hairy that the fibers
materials commonly used are nitrocellulose or collodion
damage the film; avoid these). Do not let the paper strip get
(Parlodion@), polyvinyl formol (Formvar@), and carbon.
completely wet or the film will separate from the paper and
Films of the first two substances, which are plastics, are fre-
sink to the bottom of the dish.
quently stabilized by evaporative de osition of an addi-
10. Place the filter paper supporting the grids onto an-
tional thin layer of carbon. Formvaris the most satisfac-
other piece of filter paper (grid side up) in a petri dish, and
tory plastic-film material and is used as follows.
partially cover the dish with its lid while the contents dry.
1. Have available the materials listed in section 4.7.1. 11. After drying, lift the grids by their edges with narrow-
2. Prepare a working solution of 0.2% (wtlvol) Formvar@ pointed forceps. The plastic film will break cleanly around
in ethylene dichloride (chloroform can also be used). Mix the perimeter of the grid and will be retained over the grid.
thoroughly. Films that lift away from the filter paper as the grid is lifted
3. Degrease microscope slides (standard slides for light or that do not break cleanly are too thick; some areas of the
microscopy) by washing with soap (or detergent) and water, film may show this property while others are satisfactory.
and rinse thoroughly in distilled or deionized water. Blot dry Films that are too thin will tear in the electron beam. Films
with lint-free tissue (lens tissue), which minimizes static that have large holes in them may also fail. Holes also result
charge buildup; never wipe completely dry. Cleaned slides from too high an ambient humidity during drying.
62 w MORPHOLOGY AND ULTRASTRUCTURE

Once in the microscope, because of ionization and con- photungstate salt. Uranium salts are also widely used be-
sequent changes in electrostatic charge, plastic films expand cause at pHs below 4.5 they dissociate into small uranyl
and contract to cause specimen drift as the electron beam is (UO?) ions, which penetrate into surface irregularities
intensified and decreased. Any movement will degrade the and improve the definition of fine detail. A t higher pHs,
photographic image in proportion to the magnification the uranyl ions polymerize into larger ions and can even
used. Often, adjustments of beam intensity and a brief wait precipitate from the solution. Uranyl acetate, which is gen-
will establish a level of stability. Formvat-@films prepared as erally available in the EM laboratory because it is also used
described above are quite satisfactory for routine purposes for staining thin sections, is useful at concentrations of 1 to
using magnifications of up to X20,OOO. For fine detail and 2% (wtlvol), but its pH is low (-2.5 to 4.0). When uranyl
higher magnifications, however, the stability of plastic films acetate is used as a negative stain, both positive and nega-
is improved by the deposition of a thin (2.0-to 3.0-nm) tive staining may be seen on different areas of the same grid.
layer of carbon. Thin films of pure carbon avoid drift and In addition, uranyl acetate acts as a fixative for proteins and
improve resolution even more because of a more even and nucleic acids, can contract or round up mycoplasmas, dis-
finer substructure, but they are fragile. rupts some protein structures, and should be used only with
Formvar@-coveredgrids are usually placed within a car- well-washed preparations. Uranyl oxalate can be more use-
bon evaporator and are carbon coated by the heating of car- ful than uranyl acetate because the oxalate anion is more
bon electrodes in a 1.33- to 0.13-nPa vacuum. Evaporators tightly complexed to the uranyl cation and resists precipita-
are essential equipment of all EM laboratories; make sure tion at more neutral pH. Uranyl formate, sodium silico-
that you have an operation manual and have been checked tungstate, and various other salts are also sometimes used.
out on their use. Grids without Formvar@can also be car- Of all negative stains, uranium (uranyl) salts have the high-
bon coated. Evaporate carbon onto a freshly cleaved mica est atomic number, 92, and provide the greatest specimen
surface under the same conditions; float the carbon film contrast. (Caution:salts of uranium are slightly radioactive
from the mica and place grids on this film in the same way and toxic. Care in both use and disposal is advised.)
that Formvard grids are prepared. Grids with only a carbon Some workers recommend the addition of very small
coating are extremely fragile and must be handled with care. amounts of a wetting agent (such as peptone, glycerol,
They are especially useful for high resolution of subcellular serum albumin, starch, bacitracin, or sucrose) to some stains
components and viruses. to improve spreading (12). Not all investigators find this
N o matter how good the specimen preparation, accurate useful or necessary, especially if high magnifications are to
visualization of the specimen will clearly depend on the be used and uncluttered backgrounds are desired.
quality of the support film on the grid. For most routine in-
vestigations, Formvar@- and carbon-coated 200-mesh-size 4.2.2.2. Staining Procedures
copper grids are recommended. The appropriate thickness There are many variations for negative staining (5, 10, 12).
of the coat can be gauged only by experience and by the A standard procedure is a two-step method of first applying
silver-to-gray interference pattern of the raft of grids float- the specimen to the grid and then staining with 2%
ing on water (as described above). (wtlvol) aqueous ammonium molybdate, as follows.
4.2.2. Negative Staining 1. Press a small piece of Parafilm@M (American National
Can Co. [see section 4.8 for addresses of suppliers]) onto the
4.2.2.1. Stain Choice laboratory bench surface, and then remove the tissue cover-
Negative staining is amongst the highest-resolving tech- ing to provide a clean surface. Since the Parafilm@surface is
niques that are easily available. It can be thought of as thin hydrophobic, a small drop of the sample suspension can eas-
embedding since the sample is immersed in a thin film of a ily be applied with a Pasteur pipette. Also, next to the drop
dilute heavy-metal salt, which is then dried down to a glassy of suspension, place a small drop of 2% (wtlvol) ammonium
consistency surrounding the object. Negative staining is es- molybdate onto the Parafilm@(for step 3).
pecially good for visualizing subcellular particles such as ri- 2. Place a coated grid, with the film side down, on the
bosomes, cell wall or membrane fragments, viruses, and (in drop of sample suspension. The surface tension of the
its most refined form) protein macromolecules. Most mate- meniscus spreads the suspended solids and allows them to
rial does not have to be chemically fixed before a negative adsorb to the grid. Large debris falls to the bottom of the
stain is applied, but some fragile specimens may require pre- drop and will not obscure the grid. Usually 10 to 60 s is
fixation (see section 4.2.2.2, step 5). When objects are neg- enough time for particle adsorption. Small concentrations
atively stained, they appear white against the dark back- of detergents, organic solvents, or other surface-active agents
ground of the stain. Since the heavy-metal solution can reduce adsorption and affect stainability. Trial and error
penetrates into and around the contours of subunit arrange- will determine the optimum time for adsorption, which is
ments, fine infrastructure can often be seen (see pyocins also affected by the nature and charge of the film and the
and flagella in Fig. 5). sizes and nature of the particles. To improve both adherence
Several different heavy-metal salts, usually in the range and distribution of the particles, the hydrophilicity of the
of 0.5 to 4.0% (wtlvol), have been used as negative stains. support film can be improved by the use of coated grids that
In our experience, ammonium molybdate is most useful be- have been stored in a refrigerator or exposed to UV irradia-
cause it spreads well, does not react with proteins or most tion, to glow discharge in a high-vacuum evaporator, or to
other components in a suspension, and can be readily ad- discharge from a Tesla coil. The exposures minimize elec-
justed in tonicity as required. However, solutions of sodium trostatic charges.
or potassium phosphotungstate are perhaps the most widely 3. With narrow-pointed forceps, remove the grid. By
used and are generally satisfactory at a neutral pH. Contrary touching the edge of the grid to a piece of filter paper
to popular belief, phosphotungstic acid is almost never used, (Whatman no. l), remove the small drop still adhering to
because of its very low pH; it is usually brought to neutral the grid. Quickly touch the film side of the grid to the small
pH with sodium or potassium hydroxide to become a phos- drop of 2% ammonium molybdate on the Parafilm@.
4. Electron Microscopy 63

4. After 10 to 60 s, withdraw the grid from the drop of tive apertures in the microscope. Specimen drift is avoided
stain in the fashion described above. Staining times may re- by patience, experimentation with beam intensity, and the
quire some experimentation, especially with different sorts use of carbon-coated plastic or pure carbon support films.
of specimens. Beam damage to fine structure, as well as puddling of stain
5. Allow the sample to dry and examine it in the trans- about or on the specimen, can be minimized by learning to
mission electron microscope. For very delicate samples achieve focus as quickly as possible with the lowest possible
(such as bacterial S-layers), macromolecular arrangements beam intensity and making the shortest possible photo-
can be perturbed by high surface tension during drying. This graphic or CCD exposure that produces a good, accurate
effect can be generally overcome by prefixation with 0.1 to image. A short exposure at a relatively low magnification
1.0% (vol/vol) glutaraldehyde in water or a suitable buffer. usually produces the best results; by trial and error, each mi-
This prefixation is easily done by incorporating the fixative croscopist will determine the magnifications most easily fo-
into step 2 (see above) and washing. (Caution: glutaralde- cused and producing the most consistent results.
hyde is volatile and toxic; prefixation should be done in a Solutions of stains and wash solutions (buffers, salts, and
suitably vented fume cabinet or hood.) even deionized, distilled water) may become contaminated
with bacteria over time, and a quick examination of ni-
There are several alternative procedures for negative
grosin films by light microscopy will soon confirm the con-
staining of bacterial suspensions. One involves mixing
dition of the solutions (chapter 2.2.3). Reduce the problem
equal volumes (or other ratios) of stain and suspension,
by keeping working stains in the refrigerator in a rack of
placing the grid onto a drop of the mixture, and then with-
small vials, which are filled as needed (with sterility pre:
drawing excess fluid with filter paper, either immediately or
cautions) from stock bottles. Contaminated solutions may
after an appropriate interval of time (e.g., 10 to 60 s). It is
be filtered but are best made up anew. A convenient
usually best to do this immediately after mixing, since in-
method for storage and filtered delivery is the use of a plas-
teractions of stain and particles can, with time, produce
tic syringe with an attached 0.45- or 0.20-p,m-pore-size
confusing results. Sometimes, to supply a more uniform par-
Swinnex filter. Solutions of uranyl acetate require protec-
ticle distribution, it is advantageous to pick up a thin film of
tion from light. This is done by covering the container with
the mixture in a platinum-iridium loop just bigger than the
aluminum foil.
grid (e.g., -3.2 mm) and to break this flat film over a grid
The distribution of negative stain and specimens on
resting on blotting paper. The grid is then allowed to dry
the support film may be uneven or unsatisfactory; it is
and is examined. Occasionally, stain-suspension mixtures
therefore efficient to prepare two or more grids of the same
are sprayed onto grids, but this technique requires a special
material. A grid should be scanned thoroughly at low mag-
apparatus. This procedure is most useful for quantification
nification. A satisfactory grid prepared by the two-step
of viruses or other small particles (12) and should be done
method will show a gradient of specimen and stain dis-
in a special chamber for safety.
tributed from one side to the other (remember, fluid was
withdrawn by touching one edge of the grid to the filter
4.2.2.3. Washing paper). If the particles are in low concentration in the
A useful accessory procedure after application of a suspen- original suspension or if drop withdrawal techniques are
sion but before negative staining (in the two-step method) unsatisfactory, specimens and stain may be found on only
is to wash the grid to eliminate excessive debris, salt, and one small area of the grid.
protein that can be precipitated by some stains and that can Ammonium molybdate produces less contrast than does
obscure detail. Washing can be done by touching the coated phosphotungstate or uranyl acetate, and its use may require
grid surface rapidly and repeatedly in succession to the sur- some practice. However, photographic negatives (with ap-
faces of several fresh drops of distilled water or other fluid propriate development) will have more contrast than antic-
on ParafilmB. ipated and present a great range of printable tones. Digitized
In addition to water, various solutions may be tried as images from CCD cameras can be easily manipulated by
washes, but these can be risky. The presence of mineral salts computer for added contrast. Even molybdate tends to
and organic compounds, pH, and other conditions are heavily surround most whole bacteria with an almost im-
sometimes essential for the maintenance of structure, but penetrable mass of stain; shorter staining times and less con-
they can also produce an obscuring film as the specimen centrated (0.1 to 0.5%) solutions are often required for
dries. In fact, during drying, tremendous concentration gra- good results with these relatively large objects.
dients can be produced, and these can denature proteins.
Salts have a tendency to form large precipitates on the grid. 4.2.2.5. Expectations
Of all washing fluids other than water, 1% (wtlvol) aqueous Negative staining is the simplest and fastest EM method
ammonium acetate seems best since it is nonreactive with that can be used, but to obtain very good results it can take
bacteria, mycoplasmas, membranes, bacteriophages, and many different attempts with several combinations of stain-
most intracellular components and because any excess is ing and specimen concentrations. This can be frustrating,
volatile in the vacuum of the transmission electron micro- but do not give up; the results are well worth the effort!
scope. What sort of structural information on bacteria can be
obtained by negative-staining techniques? The size and
4.2.2.4. Pitfalls shape of the cell can be ascertained. The presence of flagella
For examination of negatively stained specimens, it is par- (perhaps already suggested by motility studies) can be af-
ticularly important to balance contrast and resolution by firmed, and the distribution and fine structure of the flagella
using a n appropriate accelerating voltage (usually 60 to 80 can be determined (Fig. 5 and 8). Other surface appendages
kV). Contamination of the microscope column, apertures, such as pili (fimbriae) (Fig. 5) or the presence of S-layers or
and specimen by evaporated stain or other substances is capsular materials can be revealed. Even the differences be-
minimized by the use of a liquid nitrogen-cooled anticonta- tween walled and wall-less procaryotes (mycoplasmas or
mination device (or cold trap) and self-cleaning gold objec- bacterial L forms) become apparent.
64 MORPHOLOGY AND ULTRASTRUCTURE

icals and should be treated with care! Surgical gloves and a


well-vented fume cabinet (hood) with the glass door pulled
down to protect the face are recommended.

4.2.3.1. Fixation with Glutaraldehyde


and Osmium Tetroxide
1. To the cell suspension ot liquid culture, add an equal
volume of 5% (vol/vol) glutaraldehyde in aqueous 100 mM
HEPES buffer (pH 6.8) containing 2 mM MgClz (final glu-
taraldehyde concentration, 2.5%). Establish the pH of the
growing culture immediately before fixation, and alter the
FIGURE 8 TEM image of a negatively stained flagellum iso- pH of the fixative solution accordingly. (Sodium cacodylate
lated from Vibrio cholerae. Notice how the stain has penetrated at a 0.2 M concentration is another effective buffer often
between the flagellin subunits of the filament and the protein used for the fixative, but it contains arsenate, which vapor-
subunits of the hook (here artificially straightened). The fla- izes and is toxic. Be careful!) See sections 4.7 and 4.8 for
gellar rings of the basal body are also well differentiated. (From sources and chemical formulations. Alternatively, cen-
E G. Ferris, T. J. Beveridge, M. L. Marseau-Day, and A. D. trifuge the cells and suspend the pellet in 2.5% glutaralde-
Larson, Can. 1.Microbiol. 30:322-333, 1984.) hyde in HEPES buffer. A wash in fresh medium or in an
appropriate buffer may precede this step. For some procary-
otes, especially some mycoplasmas, it may be preferable to
make up the glutaraldehyde (perhaps at a different concen-
Sometimes negative stains penetrate through the surface tration) in fresh culture medium to balance osmolality or
layers of bacteria so that membrane invaginations (e.g., pH. (For easier manipulation and to reduce the number of
mesosomes, which are considered to be artifacts) and the in- centrifugations, it might be convenient to embed a concen-
ternal membranes of photosynthetic, methylotrophic, and trated slurry of cells in molten 2% [wt/vol] Noble agar or
nitrifying bacteria can be seen. Indeed, bacteria can be lysed another highly refined gel and allow it to solidify. Cut into
and their component parts can be isolated (see chapter 7) so cubes of 1 mm3, and process the cubes in small bottles
that they can be negatively stained (Fig. 5), and this is one through the reagents suggested below.)
of the most powerful methods of ultrastructural determina- 2. Allow the cells to fix for 1 to 4 h at room temperature.
tion. The subunit arrangements of periodic structures, such The time is flexible; any time beyond 1 h is usually adequate,
as S-layers (chapter 5, Fig. l l ) , flagella, pili, spinae, and and bacteria held in glutaraldehyde for several days can even
porins (after delipidation of outer membranes), are also re- be shipped across the country if appropriately packaged to
vealed. Even minute structural differentiations within some meet stringent transport regulations for dangerous chemi-
appendages can be seen, e.g., the filament, hook, and basal- cals. (The glutaraldehyde-fixed pellet may be enrobed in
body rings of a flagellum (Fig. 8). With experience, gram- agar at this stage for easier handling; see steps 6 through 8.)
positive and gram-negative bacteria can be readily differen- 3. Wash the pellet twice in HEPES buffer, centrifuging
tiated by their negatively stained envelope profiles. between washes.
Examples of the applications of negative staining may be 4. Suspend the washed pellet in 1% (wtlvot) buffered os-
found in references 77 through 85. mium tetroxide (0~0~) for 1 to 4 h at toom temperature.
Caution: if the odor of osmium tetroxide is detectable, eyes
4.2.3. Thin Sectioning may be subject to dangerous vapors. Persons wearing con-
See references 4 through 7, 10, and 13. tact lenses are particularly vulnerable. For safety, a commer-
For conventional thin sectioning, bacteria must be cially available eyewash station (Nalge Co.) should be
chemically fixed, washed, dehydrated, and embedded in a available, properly labeled, and strategically located.
liquid plastic that then solidifies with appropriate properties 5. Centrifuge and wash the pellet (now usually black due
for the cutting of very thin sections. The sections are to a compound called osmium black) one to three times by
stained with solutions of heavy-metal salts to give sufficient suspension in the buffer, recentrifuging each time.
contrast for examination by TEM (Fig. 1). The chemicals 6. Make up and melt 2% (wtlvol) Noble@agar in deion-
used in this technique (fixatives, stains, dehydration sol- ized water, and cool to 50C. Add a volume of agar equal to
vents, and plastics) can be very damaging to the specimen, that of the pellet, and mix quickly with a warm Pasteur
and structural artifacts can be introduced. A n experienced pipette.
microscopist should help with the assessment of the initial 7. With the pipette, quickly dispense the suspension
preparations. Although many different combinations can onto an alcohol-cleaned glass slide, and allow it to solidify.
be used, it is generally adequate to use sequential fixation in The bacteria should be in high concentration and evenly
glutaraldehyde and osmium tetroxide, followed by dehydra- distributed. With a clean razor blade, dice into 1-mm cubes.
tion and embedding in Epon 812@,LR White@,or Spurrs 8. Suspend the cubes for 15 min at room temperature
medium@ (section 4.7.4). There are other choices of em- first in deionized water to equilibrate them and then in 2%
bedding plastics, but these three work reasonably well for aqueous uranyl acetate for 1 to 2 h at room temperature.
routine purposes and are a good starting point. Once expe- Wash with two to five changes of deionized water. The rea-
rience is gained with a sample and the way it reacts to fixa- son for replacing the buffer with water is that UO:+ will
tion and embedding, modifications can be tried. Caution: polymerize from the solution at pHs above 4.5; it is best to
chemical fixatives, heavy-metal stains, and plastic resins are have an unbuffered system. Make sure that a phosphate-
chosen because of their high reactivity with and penetra- buffered system has not been used (unless the cells are well
tion into biological specimens. Remember, microbiologists washed of the buffer), because uranyl and phosphate will
are made out of biological stuff too. These are highly toxic chem- rapidly complex together and precipitate from the solution.
Next Page

4. Electron Microscopy 65

4.2.3.2. Dehydration and Embedding previously (4, 6, 10). Recent models of ultramicrotomes
with Plastic Medium often incorporate trimming devices, and separate specimen
(This is a simple method for Spurr@'sresin or LR White@ trimmers are also available. The preparatory procedure is as
resin which is commonly used. For Epon 812@,follow the follows.
manufacturer's instructions.) 1. With pliers or a Beem capsule press (E. E Fullam),
1. Wash the uranyl acetate-treated agar cubes once in press firmly over the entire capsule to loosen the enclosed
30% (vol/vol) ethanol for 10 min. (Use 2- to 3-ml volumes block. Now press from the pyramidal end, and proceed up-
in 7-ml screw-cap glass vials.) ward to force out the cured block. Alternatively, release
2. Replace with 50, 70, 80, and 95% (vol/vol) ethanol, blocks by cutting the capsule lengthwise on two sides with
successively, for 10 minutes each. a razor blade. Mount the block in a holder, pyramidal end
3. Replace with 100% (vol/vol) ethanol for 45 min, up, and place the holder in a microtome chuck.
changing once at the 20-min point. 2. Under a stereo-binocular microscope, use an alcohol-
4. Replace with 1.0 ml of fresh 100% ethanol. Add 1.0 ml cleaned razor blade to carefully trim the top of the pyramid
of complete plastic medium, and mix thoroughly but gently. (which will be the front or cutting face when mounted in
Allow to stand, with occasional mixing, for 30 min or until the ultramicrotome) until the cell mass has been reached.
cubes sink to the bottom of the vial. (The medium is most Cut down and away from this surface on all four sides at an
easily dispensed by using disposable plastic syringes.) angle of about 30" (the angle of the pyramid formed by the
5. Replace the mixture with 2.0 ml of plastic medium, block in the capsule is about 60),and leave a small, trun-
and allow to stand for 1 to 4 h until the cubes settle to the cated pyramid (having a face about 1.0 mm2) that encom-
bottom. One change of the 100% plastic medium is recom- passes the specimen to be sectioned. With care and prac-
mended during this final infiltration step. tice, this may be adequate trimming-especially if the face
6. Insert typed labels into appropriately-sized Beem@ is cut in the shape of a trapezoid. However, grooves in-
capsules or flat embedding molds, and add to each a drop of evitably left by the razor blade are rather large and run at
complete plastic medium. right angles to the face; as a result, water may run onto the
7. With a sharpened wooden applicator stick, place one face during sectioning and interfere with good results.
cube (from step 5) into each Beem@capsule or mold. Fill Better to further smooth the face in the microtome with a
the capsule with plastic medium. (Use 0.55 ml, so that the glass knife.
final block fits precisely into the microtome chuck.) 4.2.3.5. Thin Sectioning
Embeddings can also be done in gelatin capsules (available
at a local drug store). Because procedures and equipment differ in each laboratory,
8. With caps off (to allow volatilization of any residual no attempt is made here to describe the sectioning process.
solvent), place the capsules in a 60 to 80C oven for poly- Descriptions of pitfalls are thoroughly described in TEM
merization for the specified time (Epon 812@, 48 h; LR texts as well as in operating manuals available from the in-
Whitem, 22 h; and Spurr@, 16 h; some plastics [e.g., LR strument makers (4, 6, 10).
White? cure in the absence of oxygen and must be 4.2.3.6. Knives
capped). Immediately upon removal, the blocks in the cap-
sules may feel slightly spongy, but they will harden to opti- Both glass and diamond knives can be used for cutting thin
mal cutting quality after 2 to 4 h at room temperature. sections. Glass knives, which are less expensive, are usually
prepared by the use of a knife-making apparatus, which is
4.2.3.3. Other Fixatives and Embedding - Media available in all TEM laboratories. The making of glass
Many different fixatives have been devised for special pur- knives is described in texts (4, 6, 10) and in manuals ac-
poses. These include permanganates, which have applica- companying the instruments. For sectioning, the choice of
tions for thick-walled microorganisms such as yeasts and knife depends on financial limitations, operator preference
fungi, and other aldehydes (e.g., acrolein, formaldehyde, and experience, and the embedding polymer. In some in-
and crotonaldehyde). Principles of their use (including ef- stances, glass knives are preferable and produce superior re.
fects of pH, buffers, tonicity, concentration, temperature, sults. A diamond knife produces more consistent results as
and duration of fixation) are discussed in numerous books the operator becomes familiar with its edge and other qual-
(4-7, 10, 13). Similarly, numerous embedding media are ities, especially when a standard embedding medium is con-
used (4-7, 10, 13), and their selection depends on their in- sistently used. The angle of the edge of the diamond as the
filtration into cells, polymerization and cutting character, diamond sits in its holder must be specified when purchas-
miscibility with water, and availability. Other embeddin ing a diamond knife; an angle of 42 to 45" is satisfactory for
media in common use include Durcupan@, Araldite 8, most conventional embedding plastics.
Maraglas@,Vestopal@,various methacrylates for special pur-
poses, and numerous epoxy resins. The reader is referred to
4.2.3.7. Staining - of Thin Sections
the literature and to local experts for advice on the use and Even though most embedding procedures have a uranyl
properties of these media. The Lowicryls@ are particularly acetate-staining step, additional staining of the thin section
useful for low-temperature embeddings and the preservation is usually required for added contrast. The use of uranyl ac-
of antigenic sites for colloidal gold-antibody labeling (sec- etate followed by lead citrate is the most common staining
tion 4.6.2) (14-17). Caution:Lowicryls can be highly aller- procedure. For uranyl acetate staining, use the following
genic, and surgical gloves must be worn. procedure.
1. Place a drop of 1to 2% (wtlvol) uranyl acetate on a sheet
4.2.3.4. Block Trimming of clean dental wax or Parafilm@,and float a grid bearing a thin
Before sectioning, remove the hardened blocks from Beem@ section, section surface down, on the surface of the stain.
or other capsules and trim them to the proper size and 2. Cover with a petri dish lid to maintain humidity and
shape. Various methods of trimming have been described protect from dust, and stain for 5 to 7 min. Longer staining
Computational Image Analysis and Reconstruction
from Transmission Electron Micrographs
GEORGE HARAUZ

5.1. MACROMOLECULAR STRUCTURE DETERMINATION BY TEM ... 82


.2. THE NECESSITY FOR COMPUTATIONAL IMAGE ANALYSIS ..... 83
.3. COMPUTER AND SOFTWARE REQUIREMENTS ................ 84
.4. WHAT IS SINGLE-PARTICLE ANALYSIS? ..................... 84
.5. SINGLE-PARTICLEANALYSIS OF A MULTISUBUNIT ENZYME
COMPLEX: A PHOSPHOENOLPYRUVATE SYNTHASE FROM
THE ARCHAEON STAPHYLOTHERMUS m r N u s ............. 87
.6. WHAT IS ELECTRON CRYSTALLOGRAPHY? .................. 88
.7. ELECTRON CRYSTALLOGRAPHIC ANALYSIS OF A NATURALLY
OCCURRING 2-D PROTEIN ARRAY: A N S-LAYER FROM THE
EUBACTERIUM AEROMONAS SALMONICIDA ................. 91
.8. CONCLUDING REMARKS .................................. 92
.9. SOFTWARE SOURCES ..................................... 93
.lo. GLOSSARY .............................................. 93
.11. REFERENCES ............................................ 93
5.1 1.1. General References ................................... 93
.2. Specific References ................................... 94

Personal computers have become an essential fixture of itive stains, and an earlier book by Hayat and Miller (7) is
every office and laboratory. Almost everyone has had some also recommended in this regard. Of course, a TEM facility
familiarity with using a computer to send messages or ana- should also have references with alternative protocols, e.g.,
lyze and display laboratory data, including image data, at those by Nasser Hajibagheri (10) and Hoppert and
one point or another. It has become commonplace to ren- Holzenburg (8), the latter being aimed specifically at mi-
der and submit figures for publication electronically. In this crobiologists. Researchers who are interested in TEM of bi-
chapter, some specialized applications are described in ological macromolecular specimens per se should consult
which computers are essential for manipulating and en- Harris (5) and Sommerville and Scheer (12). Finally, the
hancing images obtained by using a transmission electron computational techniques for analyzing and reconstructing
microscope, particularly to understand the tertiary and qua- macromolecular structures from TEM images, the subject of
ternary structures of biological macromolecules and their this chapter, are reviewed in monographs by Frank (1, 2),
complexes. In order to be able to employ these approaches Hader (4),and Misell (9). Perhaps the best source for an
fruitfully, a certain facility and intuition for mathematics introduction to computational image analysis is the opus
are required. More commonly, one would enlist the assis- magnum by Russ (11). I t is richly illustrated with examples
tance of a specialist with the appropriate skills and compu- of electron micrographs of specimens from the biological
tational equipment. Thus, it is not possible, in a single and materials sciences and is accompanied by a general pur-
chapter, to present a how to do it scheme of even selected pose software package called the image-processing tool kit.
aspects of specimen preparation, transmission electron mi-
croscopy (TEM) imaging, or computational analysis of the
images. Instead, the conceptual principles of some tech- 5.1. MACROMOLECULAR STRUCTURE
niques that are commonly encountered in the literature are DETERMINATION BY TEM
outlined in a simplified way in order to help readers with a One of the fundamental tenets of molecular biology and
biological background understand them. A glossary at the biochemistry is that the structure of a macromolecule, such
end (section 5.10) redefines terms in boldface. as a protein, determines its function. The two most com-
This chapter must be read with some knowledge of the monly known methods of determining the structures of sol-
basic principles of TEM of biological material. Chapter 4 is uble proteins are X-ray crystallography (diffractometry)
a great place to start, and there are numerous excellent and nuclear magnetic resonance (NMR) (3). However,
monographs that are recommended for reference on this neither method is universally applicable. The former ap-
subject. The most recent edition of the book by Hayat ( 6 ) proach requires three-dimensional (3-D) crystals of suitable
gives a good encyclopedic overview of conventional tech- size (-0.5 mm or larger) and high regularity, and this con-
niques for preparation of biological specimens for TEM, dition often represents a bottleneck in structural analyses.
viz., thin sectioning. Of relevance here are its two chapters NMR is generally applicable to soluble proteins of <20
summarizing the variety and properties of negative and pos- kDa, which must be highly concentrated and enriched with

82
5. Computational Image Analysis and Reconstruction 83

isotopes such as I3C and I5N. This goal is achieved by ex- changing rapidly, and when any computer purchased is soon
pressing the proteins in recombinant form in bacteria grown out of date, the only truism is the principle of garbage in,
in minimal media with, say, [3C]glucose as the sole carbon garbage out. Get good data.
source. In contrast, TEM represents a direct approach to vi-
sualizing macromolecules and their modes of assembly and
thus serves as an alternative or complement to X-ray crys- 5.2. THE NECESSITY FOR COMPUTATIONAL
tallography or NMR (3). The resolutions of structures thus IMAGE ANALYSIS
obtained are usually in an intermediate range (1 to 5 nm), Once good micrographs are available, then computerized
but h i g h resolution (0.3 to 1 nm) has been achieved with image analysis can be used to ameliorate some inherent lim-
certain proteins arranged in highly ordered 2-D arrays (i.e., itations of TEM of biological macromolecules. The first lim-
2-D crystals) (21,22). In general, though, TEM is an excel- itation is that the images are fairly noisy in a visual sense,
lent way to investigate the quaternary or domain arrange- thus limiting the resolution of structural detail that can be
ment of fairly large macromolecules greater than about 10 interpreted. The sources of the noise include the uneven
nm in size. (Sometimes this is called blobology.) background of the carbon support or ice film, the irregular
One significant advantage of TEM of biological distribution of negative stain, and radiation-induced struc-
macromolecules is that only small amounts of material are tural alterations of the structure under investigation.
required, about 1/1,000 of that needed for other biophysical Although experimental approaches such as low-dose imag-
approaches. The sample must be as biochemically homoge- ing are designed to minimize noise, it can never be entirely
neous as possible and can be prepared for imaging in one of eliminated. Thus, one of the major themes of computational
many different ways. Generally, the specimen support is a processing and analysis of macromolecular images is that of
standard TEM grid that is coated with a layer of fenestrated increasing the signal-to-noise ratio by averaging similar im-
(or holey) plastic, which itself supports an ultrathin carbon ages of different individual complexes. This process of form-
film (2 to 5 nm thick) to which the purified macromole- ing characteristic projections with an enhanced signal-to-
cules are allowed to adsorb. Contrast is achieved by nega- noise ratio and better resolution of structural detail will be
tive staining, usually with 2% (wtlvol) uranyl acetate, al- illustrated below. Secondly, the TEM image represents a 2-D
though one should always experiment with other stains as projection of a 3-D object. This concept will also be exempli-
well (see chapter 4). If the macromolecule or complex is fied later. In order to get the 3-D structure, one requires a
large enough (>lo nm in diameter), it can be imaged in large number of different views (i.e., from different orienta-
frozen-hydrated form. Then the specimen support is merely tions) of the object. Computers are then essential for deter-
the fenestrated plastic film, whose holes contain a thin layer mination of the relative angular orientations of different
of sample. The grid is rapidly frozen by plunging into liquid projections and for reconstructing an image of the 3-D struc-
pr?pane o ethane maintained at liquid-nitrogen tempera- ture from the 2-D data that are available.
tures so tt?at vitreous (noncrystalline) ice is formed to trap For purposes of illustration in this chapter, two comple-
the macromolecules in their native state. Stains are not mentary approaches to analyzing macromolecular images
used, and contrast is achieved instrumentally by varying the have been chosen. The first is called single-particle analy-
degree of focus (5,9,23). sis, where the purified protein or complex is imaged as iso-
In any structural biology project, there are limitations to lated single particles in random orientations on a carbon
TEM which must be dealt with for high-resolution work. film or in vitreous ice by low-electron-dose TEM. Two good
There are potential specimen preparation artifacts, especially recent reviews of single-particle analysis for asymmetric
due to dehydration- and/or radiation-induced structural al- complexes are those by Czarnota et al. (14) and
terations. There are contrast limitations necessitating the use Radermacher (25). The single-particle approach is the
of heavy atom salts or metal shadowing. There are resolution method of choice for probing the structures of highly sym-
limitations due to stain and image noise. Moreover, TEM metric structures such as icosahedral viruses (3 1). Usually,
images are only 2-D and the structure being investigated is resolution achievements can approach 1 nm in 3-D for
3-D. These points will all be illustrated below. The driving asymmetric molecules and even better for symmetric ones
force behind many recent advances in TEM technology has such as viral capsids. It is thought that there is no inherent
been to solve problems such as these. The technique of limitation in the technique to achieving even atomic reso-
cryoTEM allows one to image freeze-dried or frozen-hydrated lution (33).
specimens trapped in their native state. One routinely per- The second approach that will be briefly described is 2-
forms low-electron-dose TEM, meaning that the specimen is D electron crystallography, which can be applied when the
irradiated only during actual image formation, not during fo- specimen is arranged in the form of a 2-D crystal (e.g., a
cusing which is done on an adjacent region. There are vari- monolayer of protein). Again, the best-quality TEM images
ous flavors of TEM involving the use of combinations of are obtained, and crystallographic computational ap-
elastically and inelastically scattered electrons to enhance proaches can be used to form a projection map of the unit
contrast without the need for stains and/or high-resolution cell at generally higher resolution than is achievable by
elemental mapping by either dark-field TEM, scanning TEM, single-particle analysis. Combining projection maps at dif-
or electron spectroscopic imaging (chapter 4). One can note ferent orientations yields the 3-D structure; reference 32
here that scanning probe microscopies (including atomic provides a nice schematic illustration of this process. The
force microscopy) are also viable and fruitful imaging tech- computational procedures for electron crystallography were
niques (chapter 6). All of these considerations are important originally developed with specimens such as bacterial sur-
to TEM of biological macromolecules in general and dictate face arrays (e.g., S-layers). Instructive reviews with micro-
requirements for specialized instruments that might not nec- biological samples include references 9, 16, 29, and 32.
essarily be found in every center or university. For present Electron crystallography is increasingly being applied to de-
purposes, suffice it to say that one should strive to obtain the termine the native 3-D structures of numerous membrane
best possible TEM image of the biological macromolecule proteins in situ (20, 35); secondary structure elements can
that one can. In these days when computer technology is generally be defined. Indeed, atomic resolution can also be
84 w MORPHOLOGY AND ULTRASTRUCTURE

obtained, as first achieved for an archaeal bacteri- incorporating them into 3-D reconstructions from TEM
orhodopsin (21) and shortly thereafter for other proteins data is Insight 2000 (h). One should always be careful not
such as bacterial porins (22). to use any computer program as a black box without un-
derstanding some of the underlying principles behind its
development.
5.3. COMPUTER A N D SOFTWARE
REQUIREMENTS
As indicated above, personal computers are an essential 5.4. WHAT IS SINGLE-PARTICLE ANALYSIS?
component of any laboratory, and few researchers work It is best first to describe what is meant by single-particle
without them. In the computers memory, an image is sim- analysis. In Fig. 1, a hypothetical macromolecular complex is
ply a large set of numbers indicating degrees of darkness and shown. This structure consists of 12 spheres (representing the
lightness. Any point in a digital image is called a pixel, protein subunits) arranged in what is called D6 symmetry (19,
meaning picture element, and has a certain size (with re- 26). One way of representing this 3-D structure is by shaded
spect to the object) associated with it. For high-resolution surface representations as shown in Fig. la. Here, we get a
work, one requires fine sampling, i.e., pixels spanning a visual impression of the macromolecular complex as a real
small distance. Modern transmission electron microscopes 3-D object from the depth cues given by the simulated light
can capture images digitally directly, or special photo- source. This sort of representation is common in computer
graphic film can be scanned at high resolution. In popular graphics, especially in the entertainment industry. However,
computing, one hears of images being scanned at a certain the representation in Fig. l b is that of 2-D projections, which
dpi, meaning dots per inch. Scanners with resolutions of is what we get in a TEM image. When the macromolecular
600 dpi or better are commonplace and inexpensive. It is complex is imaged from a particular direction, its mass at each
more appropriate here to think of scanning TEM film with point along that direction is summed (figuratively speaking)
pixel sizes of about 5 km, at which point the size of the sil- and recorded on the micrograph. The degree of darkness on
ver grains of the emulsion becomes the limiting factor. the image at any point is proportional to the amount of
With respect to an image of a protein complex, pixel sizes macromolecular mass above that point. A good analogy is that
should be no larger than about 0.5 nm at the object level. of a chest X ray-the details of both front and back ribs, and
(To convert pixel sizes from the object level to the micro- all the organs in between, are projected onto a 2-D film and ap-
graph level, one simply needs to use the magnification fac- pear superimposed.
tor.) Thus, the new 4,000 dpi scanners would have a pixel As shown by the various images in Fig. 1 (remember that
size of 6.35 km, more appropriate for our purposes. One im- these are all of the same 3-D object), if the macromolecular
portant point to note is that TEM image data are not in complex lies in a certain way on its support in the transmis-
color, but instead are represented by shades of gray, also sion electron microscope, then the electron beam will im-
called gray levels, i.e., numbers ranging from 0 (dark) to pinge on it from a certain direction. In negatively stained
4,095 (light), say. Image data require substantial memory for preparations, this complex will tend to lie on the carbon
programs to be able to manipulate them quickly (of the support film on its flat side. Thus, we would usually see the
order of many hundreds of megabytes), significant hard top-down views of the structure, and other projection views
drive space for storage (of the order of gigabytes), and a such as side ones would occur less frequently. In frozen-
good-quality display that can render many colors. The more hydrated preparations where the sample is effectively being
powerful the computer, the better. imaged in a solution, a far greater variety of orientations is
Although there is a wide range of general software for
manipulating image data, the specialized software that is di-
rectly applicable to TEM images is more limited (see sec-
tion 5.9; italic letters in parentheses refer to items listed
there). Some examples are Northern Eclipse 6.0 (a) and
Scion Image ( b ) , which run on personal computers with a
recent version of the Windows operating system. The Scion
program is adapted from the NIH Image software package
that runs on Macintoshes (c), and both packages can be
downloaded for free. The suites of computer programs for
high-resolution analysis of macromolecular structure are
quite complex, however, and have been developed on
UNIX workstations. Two common packages are IMAGIC-
5 (d) (34) and SPIDER (e) (18) and comprise both single-
particle analysis and 2-D electron crystallography. All of the
work presented here was performed by using IMAGIC-5. A
new package called EMAN (f) (24) is based partly on
IMAGIC-5 and focuses on single-particle analysis, but it is
noteworthy because it is downloadable for free and provides
extensive guidance for novice users. The SEMPER system is
partly of historical importance because many techniques for FIGURE 1 Simulated macromolecular complex, consisting
analysis of TEM images of bacterial S-layers were developed of 12 spheres arranged in D6 symmetry, i.e., with one sixfold ro-
by using it, but it remains current and easy to use and is of tational axis of symmetry (the top-down view) and two twofold
very general applicability, and a modern version is now rotational axes of symmetry (the side views). In panel a, the 3-
available that can run on almost any computer platform (8) D structure is depicted via shaded surface representations.
(27). Finally, structural molecular biologists are ultimately Here, the brighter regions act as a depth cue. In panel b, 2-D
interested in atomic models of their proteins. A powerful projections of the 3-D structure are shown, in which brighter
suite of programs for manipulating molecular models and regions represent a greater amount of molecular mass.
5. Computational Image Analysis and Reconstruction 85

seen. It is far easier to prepare macromolecules for TEM by macromolecular complex has the same 3-D structure, each
negative staining, and this method should be used first before image in Fig. 2 would be identical if they were noise free.
proceeding to cryoTEM of vitreous ice-embedded material. The computer programs that have been written for single-
The effects of noise on the image are depicted in Fig. 2. particle analysis do a number of complicated tasks. First of all,
Here, we have added randomly generated numbers with a they select automatically (or allow one to select interac-
Gaussian distribution to the gray level of each pixel of a set tively) the images of different macromolecular complexes
of images of the macromolecular complex lying in the top- from a transmission electron micrograph that contains views
down orientation. The simulated molecules are obscured by of many of them. Then the programs align each macromolec-
the noise, as is the case in TEM, and it is difficult to assess by ular image with respect to a reference to facilitate comparison
visual inspection which projection view is represented. For and sort out into smaller subgroups those that represent the
this reason, computational single-particle analysis was devel- same projection (the characteristic projections mentioned
oped. Consider that each of the images in Fig. 2 represents a above). The sorting (sometimes called clustering) is done via
2-D top-down projection of a different individual macromo- a multivariate statistical analysis. Finally, we average together
lecular complex. However, since the preparation was bio- the images of macromolecular complexes viewed in the same
chemically pure to begin with, and since each individual orientation. (This process is simple arithmetic.) Since the
macromolecular structural information is constant, it rein-
forces itself, whereas the noise, which is random, tends to
cancel itself out. The effect of averaging is shown in Fig. 3.
With increasing numbers of images being averaged, the final
image of the macromolecular complex becomes clearer. In
other words, the signal-to-noise ratio is increased.
In single-particle analysis, minimally hundreds and
preferably thousands of images of a macromolecular complex
are required in order to be able to get large enough subgroups
representing the same 2-D projection for averaging. When a
number of averages of different 2-D projections are obtained,

FIGURE 3 When noisy images like those in Fig. 2 are aver-


aged together, the signal-to-noise ratio increases roughly ac-
cording to the square root of the number of images being aver-
aged. The constant structural information in each image is
reinforced, whereas the random noise cancels itself out. The
number of images being averaged in each panel is as follows:
FIGURE 2 In transmission electron micrographs of biologi- panel 1, 1; panel 2, 2; panel 3, 9; panel 4, 25; panel 5, 100;
cal macromolecules, noise obscures structural detail severely. panel 6,225; panel 7,400; panel 8,625; and panel 9,900. Each
Here, the top-down 2-D projection from Fig. l b has been du- image must represent the same 2-D orientation and the
plicated a number of times to represent different individual macromolecule must be in the same place in each image in
macromolecular complexes lying in the same orientation on a order for the average to make sense. Thus, in single-particle
support film. Random noise has been added to each point analysis, a large population of images of different macromolec-
(pixel) in each image. Thus, the numbered images portrayed ular complexes is aligned in order to facilitate comparison and
here are all different because the noise is different in each, even sorted into homogeneous subgroups for averaging. Details of
though the complexs structural information is the same. these processes are found in references 2, 14, 25, and 33.
86 MORPHOLOGY AND ULTRASTRUCTURE

they can be used to reconstruct a 3-D image of the struc- D structure yet. The methods for determining the relative
ture. To return to the chest X-ray analogy, in order to see angles amongst different projections are mathematically
how the organs and bones in the chest are really arranged quite complex; some approaches for asymmetric objects are
with respect to one another, one needs an X ray from described in references 14, 25, and 33, and the strategy
front to back, side to side, and all the oblique views in used for icosahedral viruses is reviewed in reference 31.
between. In fact, this angular sampling is what medical Once the angles relating different projections are known,
X-ray computer-assisted tomography (CAT scanning) then a 3-D reconstruction from projections can be per-
achieves. formed. The computational principles of reconstruction
In single-particle analysis of TEM images, we do not from projections are depicted in Fig. 4 for a simple exam-
know a priori how the different views are related to one an- ple and are used in many fields such as medicine as well as
other-remember, we do not know the macromolecule's 3- in structural biology.

Z 1 I

v v v
I I I

v v v
I I I

v v v
I I I

v v v
I I

FIGURE 4 Principles of reconstruction from projections. (a) In this example, a 2-D image is projected into a 1-D image by sum-
ming all the density values along a line at a certain angle. (b) The process of reconstruction by back projection involves smearing back
the densities of each 1-D projection along the direction in which the projection was collected into a blank 2-D image and adding to-
gether all the back projections possible. Here, the reconstructions from 1, 2,3, ..., 9 different 1-D projections (i.e., collected at differ-
ent angles of 0, 20,40, ..., 160") are shown. In panel b, image 1, the density from each point in a 1-D projection was back projected
at an angle of 0" into the 2-D image that will form the reconstruction. The angle 0" is the one at which projection 1 was collected.
In panel b, image 2, the 1-D density data are back projected into the 2-D image at the angle 20" associated with this projection and
added to the data back projected from projection 1. On it goes, with images 3 and 4, etc., showing the cumulative results of back pro-
jecting density data from differetlt angles and adding them to the previously back-projected information. A reasonable representation
of the original object, a collection of three disks, is already obtained from nine projections. Reconstruction quality improves with in-
creasing numbers of input projections that span all possible views. The reconstruction approach shown here is called tomography,
meaning that 2-D slices of a 3-D object are reconstructed. Tomography requires a specialized geometry in which the 3-D object is tilted
about an axis by a certain angle, e.g., using a tilt stage in a transmission electron microscope. In single-particleanalysis, such a geom-
etry generally does not apply and 3-D volume images are reconstructed directly (i.e., not slice by slice) from 2-D projection images.
5. Computational Image Analysis and Reconstruction 87

5.5. SINGLE-PARTICLE ANALYSIS OF dimer.) Thus, it was of interest to find out how this en-
zyme was arranged, and its size (about 20 nm in diameter)
A MULTISUBUNIT ENZYME COMPLEX: made it suitable for TEM investigation. In Fig. 5, a trans-
A PHOSPHOENOLPYRUVATE SYNTHASE mission electron micrograph of a negatively stained prepa-
FROM THE ARCHAEON S7WHYLOTHERMUS ration of the enzyme lying on a carbon support is shown,
MARINUS along with a set of individual smaller images of different
The deep-sea archaeon Staphylothermusmarinus possesses a complexes extracted from the larger micrographs. The lat-
phosphoenolpyruvate synthase that is unusual in that it is ter images were the input to the programs used for single-
a large protein complex of molecular mass 2.25 MDa, particle analysis.
comprising 24 identical subunits (13, 23). (The corre- Two transmission electron micrographs of a frozen-
sponding enzyme from mesophilic eubacteria is a simpler hydrated preparation (i.e., embedded in vitreous ice) are

FIGURE 5 Homogeneous preparation of the 2.25-MDa phosphoenolpyruvate synthase from S. marinus, adsorbed to a thin car-
bon support, negatively stained with 2% (wtlvol) uranyl acetate to provide contrast, and imaged by TEM. (a) Field of view show-
ing a number of different individual complexes. The scale bar represents 20 nm. (b) A set of smaller images of different individual
complexes, extracted from larger micrographs like those in panel a. The scale bar represents 20 nm. These smaller images will be
processed by computer programs that perform single-particle analysis to align, sort, average, and reconstruct them. In these images,
the lighter regions represent proteinaceous material whereas the darker areas represent negative stain that embeds it. Some indi-
vidual complexes are circled and denoted by A, B, C, and D.
88 w MORPHOLOGY AND ULTRASTRUCTURE

shown in Fig. 6. Here, there is neither stain nor an underly-


ing carbon support, and the individual complexes can be
seen partly because of a phase-contrast effect. The images in
Fig. 6 are of the same region of the specimen grid but differ
in that they were recorded at different levels of defocus, i.e.,
at levels away from the true focus of the instrument. The
concept of focus is somewhat less intuitive in TEM than in
light microscopy; biological electron microscopists gener-
ally defocus (i.e., underfocus) to get a better-looking image.
In cryoTEM, one achieves higher contrast in terms of abil-
ity to distinguish the macromolecules within the ice by de-
focusing, but at the cost of resolution of detail. In Fig. 6, one
image is slightly blurrier than the other, but the macromo-
lecular complexes stand out a bit better. As in Fig. 5, single
macromolecular images were selected from the larger micro-
graphs and subjected to single-particle analysis. Moreover,
the micrographs of the frozen-hydrated preparations at dif-
ferent defocus levels were combined via an approach called
contrast transfer function correction to maximize the
amount of structural information obtained (23). This
process is very difficult to explain without using mathemat-
ics. Suffice it to say that structural detail of different sizes is
not recorded with the same efficiency at different defocus
levels. We can predict theoretically how the blurring occurs
at each level. Given a pair of (or even more) images
recorded at different defocus levels, we can extract the best
information from each micrograph and put complementary
data together to yield the least blurry image possible.
Partial results of single-particle analyses of both frozen-
hydrated and negatively stained preparations are shown in
Fig. 7. The image averages in Fig. 7a to c show projection
views (amongst many others obtained) of the frozen-
hydrated macromolecular complex along axes that portray
twofold, threefold, and fourfold rotational symmetry. The
image average in Fig. 7d shows the projection view of the
negatively stained macromolecular complex along its three-
fold rotational symmetry axis, its most stable orientation in
this kind of preparation. For all preparations available in
this study (23), projection views at different orientations
such as those in Fig. 7 were then combined to give 3-D re-
constructions. In Fig. 8, surface representations of various 3-
D reconstructions of the complex are depicted, showing the
underlying octahedral arrangement of this 24-meric com-
plex and the overall consistency (yet subtle differences) ob-
tained from different TEM preparations. The resolution of
structural detail was better with the frozen-hydrated-
preparation data, as is usually the case. Finally, an atomic
model of the enzyme was generated and 24 subunits were fit FIGURE 6 Homogeneous preparation of the 2.25-MDa
into the 3-D reconstruction obtained by single-particle phosphoenolpyruvate synthase from S. marinus, encased in a
analysis, as shown in Color Plate 8. This exercise provided thin layer of vitreous ice and thereby trapped in a native, hy-
some useful insights into how this enzyme complex drated state. The sample was kept at liquid-nitrogen tempera-
achieved structural stability under extreme conditions of ture in the transmission electron microscope during imaging.
temperature (23). Presently, single-particle analysis in con- Panels a and b represent the same region of the specimen, but
junction with cryoTEM is an established tool for probing the images were recorded at different defocus values. The
the structures of numerous macromolecular complexes, and image in panel a is closer to the true focus position of the mi-
its greatest notable recent success has been the elucidation croscope than the one in panel b, and is somewhat crisper but
of the architecture of prokaryotic ribosomal subunits (re- shows less contrast. This effect is due to the contrast transfer
viewed in references 17 and 33). function, which changes with defocus ( 5 , 9, 23). Single-parti-
cle analyses of such frozen-hydrated material also incorporate
contrast transfer function correction (e.g., reference 23). In
5.6. W H A T IS ELECTRON these cryoTEM images, the darker regions represent the pro-
CRYSTALLOGRAPHY? teins. These images, like those in Fig. 5, were captured in digi-
In electron crystallography, the macromolecule of interest is tal form directly from the cryo-transmission electron micro-
already arranged in the form of a 2-D array or crystal, one scope at a pixel size of 0.344 nm at the object level (23). Each
protein thick. In the simple example shown in Fig. 9a, each panel in this figure is 600 by 600 pixels in size and thus spans a
repeating motif in the 2-D crystal is a top-down view of the region on the specimen that is 206.4 nm in extent.
5. Computational image Analysis and Reconstruction rn 89

FIGURE 7 In cryoTEM images of frozen-hydrated preparations, biological macromolecules can exhibit a greater variety of ori-
entations than those adsorbed to a support film. Single-particle analyses of contrast transfer function-corrected cryo-transmission
electron micrographs (as those in Fig. 6) of the S. marinus phosphoenolpyruvate synthase yield projection averages viewed along
twofold (a), threefold (b), and fourfold (c) rotational symmetry axes, consistent with an underlying octahedral shape. Single-
particle analyses of TEM images of negatively stained preparations (as those in Fig. 5) yield primarily the threefold rotationally sym-
metric view (d) because this is the stablest way in which this complex can lie o n the support film. In all of these images, contrast
is such that lighter regions represent proteinaceous material. The scale bar represents 5 nm for all panels. Panels a through c are re-
produced from reference 23 by permission of Academic Press.

FIGURE 8 Projection averages at many different orientations, including those in Fig. 7, were used to reconstruct the 3-D struc-
ture of the archaeal phosphoenolpyruvate synthase from negatively stained preparations (1,028 input images) (a) and frozen-
hydrated preparations (b through d) at high defocus (2,467 input images) (b) and low defocus (5,419 input images) (c) and after
contrast transfer function correction (3,776 input images) (d). Here, the reconstructed 3-D objects are represented by shaded sur-
faces and are viewed from different orientations. This particular macromolecular complex has an octahedral shape and thus has
fourfold, threefold, and twofold axes of rotational symmetry. There are always subtle differences among reconstructions from dif-
ferent preparations, and one must be cautious not to overinterpret details that might be spurious.
90 w MORPHOLOGY AND ULTRASTRUCTURE

simulated structure first seen in Fig. 1. Again, with added in Fig. 9a and 9b into a n alternative representation in
noise in Fig. 9b (as in a TEM image), individual motifs are which bright central spots indicate that we have repeating
obscured. In electron crystallography, a mathematical tool structural information (Fig. 9c and 9d, respectively). By se-
called the Fourier transform allows us t o convert the images lectively filtering these data as in Fig. 9d, i.e., zeroing out all

FIGURE 9 Basic principles of 2-D electron crystallography in general and of Fourier filtering in particular. (a) Simulated macromo-
lecular complex, as shown in Fig. 1, arranged in a 2-Dplanar array (one complex thick). (b) Random noise has been added to each
point in the image. (c) A Fourier transform of the image in panel a reveals a set of bright spots representative of the underlying regu-
larity of the structure. Crystallographers are able to extract a great deal of information from the intensities and patterns of these spots.
(d) A Fourier transform of the noisy image in panel b also reveals a set of bright spots, but now there is a diffuse background due to
the noise, and the spots farther away from the origin are obscured. (e) The image in panel d can be filtered by computationally se-
lecting only those values around the bright peaks and discarding all other values which represent primarily image noise. The bright
peaks are arranged in a pattern that can be predicted by the way the repeating motifs are arranged with respect to one another, i.e.,
distances and angles between them. (f) By inverse Fourier transformation of the image in panel e, an improved image of the crystalline
array is obtained for comparison with the original noisy image in panel b and the noise-free image in panel a.
5 . Computational image Analysis and Reconstruction 91

pixels in the Fourier transform not associated with repeated


information (Fig. 9e), and then doing an inverse Fourier
transform, we obtain a less noisy image of the planar array
(Fig. 9f). The jargon used to describe this process is Fourier
filtering, or sometimes image reconstruction, which should
not be confused with 3-D reconstruction from projections
that has already been mentioned. In practice, some fine
tuning is performed by using computational alignment al-
gorithms similar to those used in single-particle analysis, a
scheme known as correlation averaging. The reader is re-
ferred to reference 16 for an overview of these approaches.
In order to get a 3-D structure of the repeating motif of
the crystal, one would tilt the specimen physically in the
transmission electron microscope and obtain images at dif-
ferent angles of tilt, i.e., the different orientations (Fig. 10).
Each image of the tilted crystal would be processed as those
in Fig. 9, to provide a good 2-D projection, and the 3-D
structure would be reconstructed from the set of 2-D pro-
jections as shown in Fig. 4. The angles of tilt are already
known for each view. A good conceptual illustration of
this tomographic reconstruction process is found in refer-
ence 32.

5.7. ELECTRON CRYSTALLOGRAPHIC


ANALYSIS OF A NATURALLY O C C U R R I N G
2-D PROTEIN ARRAY: A N S-LAYER F R O M THE
EUBACTERIUM AFROMONAS SALMONKIDA
Slayers, the proteinaceous surface arrays found on some
gram-positive and gram-negative eubacteria and many ar-
chaea (28), are naturally occurring 2-D crystals that are
amenable to image analysis by electron crystallography. We
use here as an example the Slayer from Aeromonas salmoni-
cida (Fig. l l a ) (30). The Slayer is a virulence factor for this
gram-negative fish pathogen, and as in all Slayers, the pro-
teinaceous subunits self-assemble into oblique (p2), square
(p4), or hexagonal (p3 or p6) motifs, depending on the par-
ticular protein being produced. The prefix "p" refers to the
patterns of symmetry evident in the array (19, 26).
In this example, it is difficult to see in the untreated elec-
tron micrograph (Fig. 1l a ) what the individual repeating
motifs look like, but after filtering the Fourier transform

FIGURE 10 Specimens in the transmission electron micro-


scope can be tilted about a single axis (or sometimes about two
orthogonal axes) at defined angles and then imaged. In mod-
em transmission electron microscopes, this entire process is
computer controlled and automated. Here, a partial tilt series
around a vertical axis of a segment of a simulated 2-D crystal is
shown at tilt angles of 0" (a), 15" (b), 30" (c), 45" (d), and 60"
(e). In practice, most transmission electron microscope speci-
men stages are limited to tilt angles of t60",where 0" repre-
sents the untilted specimen, but one would collect data at finer
tilt increments such as every 3 or 5". The limitation is the cu-
mulative electron dose that the specimen can withstand before
being distorted or destroyed. The object being tilted can be re-
constructed tornographically as shown in Fig. 4. The Venetian
blind effect of specimen grid bars makes it difficult to go any
higher. This effect is already visible here via the confusing su-
perimposition of repeating motifs at 60". Since the object is not
imaged fully from all possible orientations, the resolution of the
final 3-D reconstruction is limited (the so-called missing-cone
effect).
92 MORPHOLOGY AND ULTRASTRUCTURE

FIGURE 11 Example of 2-D electron crystallography. (a) Slayer from the eubacterium
Aeromonas salmonicida with p4 symmetry (with unit cell dimensions a = b = 12 nm), negatively
stained with 2% (wtlvol) uranyl acetate and imaged by TEM. The various possible planar symme-
try groups are defined in references such as 16, 19, and 26. The distance between the center of each
repeating motif and its neighbor along the lattice line (i.e., not diagonally) is thus 12 nm. (b)
Fourier transform of the image in panel a. The arrangement of peaks is not as good as that in the
simulated image in Fig. 9, reflecting the underlying imperfections of the 2-D array. (c) After filter-
ing of the image in panel b and inverse Fourier transformation, an improved image of the bacterial
surface array is obtained. The regional imperfections of the crystal, in terms of degree of staining
and distortion, are evident. (d) Magnified view of a region of the filtered crystal. In panel a, the pro-
teinaceous material is dark and the negative stain is light, as it would appear on the original elec-
tron micrograph recorded on film. In panels b through d, the contrast has been inverted.

shown in Fig. 1lb, an image with enhanced signal-to-noise Finally, electron crystallography is, at present, the most
ratio as shown in Fig. 1l c and d is obtained. Such analyses productive method for determining the structures of mem-
have become almost routine for investigating bacterial S- brane proteins, and a tremendous amount of research activ-
layers (e.g., references 16 and 28). Because Slayers are ity is presently under way in this field (20, 35). Many of
highly sensitive to damage by the electron beam, long expo- these examples, including various membrane channels (22),
sures necessary for techniques such as selected-area electron are of direct interest to structural microbiologists. In addi-
diffraction (chapter 4) are usually avoided, making image tion to membrane proteins, soluble proteins (such as en-
reconstruction the preferred method of obtaining high- zymes) can be induced to form planar arrays, and electron
resolution, noise-reduced images. Most work has been done crystallography can be used to solve their structures (15).
on negatively stained preparations like that in Fig. 1la, but
some cryoTEM of frozen-hydrated specimens has been re-
ported (see reference 30). More recently, some image recon- 5.8. CONCLUDING REMARKS
struction has been achieved on Slayers probed by scanning TEM, particularly cryoTEM, has become a powerful tool for
tunneling or atomic force microscopies (chapter 6). analyzing the tertiary and quaternary conformations of bio-
5. Computational Image Analysis and Reconstruction 93

logical macromolecules and their complexes. Computa- somewhat misplaced from where it should be, ideally.
tional image analysis and reconstruction are integral tools Correlation averaging is a tool used to reposition each unit
in this process. This chapter has illustrated two approaches cell computationally to give a better averaged image.
to manipulating and processing macromolecular TEM im- Fourier filtering The Fourier transform is a useful mathe-
ages that one would commonly find in the structural and matical tool that can be used to show the degree of repeating
molecular microbiological literature. information in any signals, including images. By selecting
only this repeated information and inverse transforming,
the level of noise can be reduced significantly because it is
5.9. SOFTWARE SOURCES random. One occasionally reads about the fast Fourier
a. Northern Eclipse 6.0. EMPIX Imaging, 3075 Ridgeway transform, which is simply an algorithm for rapidly comput-
Dr., Unit #13, Mississauga, Ontario, L5L 5M6, Canada. ing a Fourier transform for images of certain sizes (i.e., num-
http://www.empix.com. bers of pixels).
b. Scion Image. Scion Corporation, 82 Wormans Mill Image reconstruction A n imprecise term often used to
Court, Suite H, Frederick, MD 21701. http://www denote Fourier filtering.
.scioncorp.com. Single-particle analysis A n electron micrograph of a pro-
c. NIH Image. Research Services Branch, National tein sample will portray many protein macromolecules (or
Institute of Mental Health, National Institute of complexes). Each macromolecule possesses the same 3-D
Neurological Disorders and Stroke, c/o NIMH Public structure, but each looks different in the electron micro-
Inquiries, 6001 Executive Blvd., Room 8184, MSC 9663, graph because it lies in a certain orientation and its image is
Bethesda, MD 20892-9663. http://rsb.info.nih.gov/ noisy. Single-particle analysis combines all of the images of
nih-image. different individual macromolecules together to reduce
d. IMAGIC-5. Michael Schatz, Image Science GmbH, noise and obtain the 3-D structure.
Heilbronner StraBe 10, D-10711 Berlin, Germany.
http://www.ImageScience.de. 5.1 1. REFERENCES
e. SPIDER. J. Frank, Wadsworth Center for Laboratories
and Research, New York State Department of Health, The reference lists here are not exhaustiwe and are provided as
Empire State Plaza, Albany, NY 12201-0509. http://www reasonable places to start gathering more information. The
.wadsworth.org/spider-doc/spider/docs/spider.html. choice of general references reflects as much what I had in hand
f. EMAN. S. Ludtke, Vema and Marrs McLean Depart- (or rather, on shelf) as any other criterion. Specific references
ment of Biochemistry, Baylor College of Medicine, could number in the thousands.
Houston, TX 77030. http://blake.bcm.tmc.edu/eman.
g. Image Objects (formerly SEMPER). Synoptics Ltd., 5.1 1. I . General References
Beacon House, Nuffield Rd., Cambridge, CB4 lTF, United 1. Frank, J. (ed.). 1992. Electron Tomography: Three-
Kingdom. http://www.synoptics.co.uk. Dimensional Imaging with the Transmission Electron Micro-
h. Insight 2000. Accelrys Inc. (formerly Molecular scope. Plenum Press, New York, NY.
Simulations Incorporated), 9685 Scarnton Rd., San Diego, 2. Frank, J. 1996. Three-Dimensional Electron Microscopy of
CA 9212 1-3752. http://www.accelrys.com. MacromolecularAssemblies. Academic Press, New York, NY.
Both references 1 and 2 are good, albeit specialized, owerwiews
of computerized 3 - 0 electron microscopy, the first one edited
5.1 0. GLOSSARY and the second one written by a pioneer in the field. Reference
1 describes 3-D reconstruction techniques that are applicable to
2-D electron crystallography Electron beams in standard any structures that can be imaged by TEM, even those as large
transmission electron microscopes cannot penetrate thick as organelles and whole cells. Reference 2 is devoted to single-
samples like whole bacteria or 3-D crystals of proteins. particle analysis like that described here.
Occasionally, 2-D crystals of proteins that are one molecule 3. Glasel, J. A,, and M. P. Deutscher (ed.). 1995.
thick can be produced, which are suitable for viewing by Introduction to Biophysical Methods for Protein and Nucleic
TEM because now the electrons can get through to form an Acid Research. Academic Press, San Diego, CA.
image. These images can be manipulated computationally An overview in one volume of the myriad techniques for prob-
by using the same conceptual tools that X-ray crystallogra- ing the structures of biological macromolecules.
phers use for 3-D crystals. 4. Hader, D..P. (ed.). 2001. Image Analysis: Methods and
3-D reconstruction from projections A TEM image is Applications, 2nd ed. CRC Press, Boca Raton, FL.
like an X-ray image in that 3-D information is projected into A compendium of papers describing warious applications of
a 2-D image. By combining computationally the data from imge analysis in the biological sciences, including electron mi-
many 2-D images in which the object is viewed from differ- croscopy.
ent orientations, the 3-D internal density distribution can 5. Harris, J. R. 1997. Negative Staining and Cryoelectron
be calculated, i.e., reconstructed. Microscopy. Bios Scientific Publishers, Oxford, United
Contrast transfer function correction A transmission Kingdom.
A lot of excellent examples of high-quality TEM images of bio-
electron micrograph is blurry in a way defined by the con- logical macromolecules are presented here. A good place to see
trast transfer function, which changes with the level of de- what to aim for in ones own work, especially for cryoTEM.
focus at which the image was recorded. The information 6. Hayat, M. A. 2000. Principles and Techniques of Electron
from two or more micrographs, recorded at different defocus Microscopy. Biological Application, 4th ed. Cambridge
levels, can be combined to give a minimally blurry result. A University Press, Port Chester, United Kingdom.
great deal of physics and mathematics are involved. General, standard reference that should be in every TEM lab.
Correlation averaging In 2-D electron crystallography, 7. Hayat, M. A., and S. E. Miller. 1990. Negative Staining.
the first step is Fourier filtering, which essentially averages McGraw-Hill, Toronto, Canada.
the information from all repeating units. However, protein More detailed description of a common preparation technique,
crystals are never perfect. Each repeating unit (unit cell) is of interest to the experienced prmtitioner.
94 w MORPHOLOGY A N D ULTRASTRUCTURE

8. Hoppert, M., and A. Holzenburg. 1998. Electron this groups work on ribosomes. Reference 17 is dedicated to re-
Microscopy in Microbiology. Bios Scientific Publishers, viewing contemporary progress in structural ribosomology.
Oxford, United Kingdom. 18. Frank, J., M. Radermacher, P. Penczek, J. Zhu, Y. Li, M.
A good resource written specifically for microbiologists. Ladjadj, and A. Leith. 1996. SPIDER and WEB: process-
9. Misell, D. L. 1978. Practical Methods in Electron ing and visualization of images in 3D electron microscopy
Microscopy, vol. 7. Image Analysis, Enhancement, and Inter- and related fields. J. Struct. Biol. 116:190-199.
pretation. North-Holland, Amsterdam, The Netherlands. References 18, 24, 27, and 34 describe software packages
An excellent classic in the field and a good review of techniques available for analysis of TEM images of macromolecules.
such as helical reconstruction. 19. Hammond, C. 1992. Introduction to Crystallography.
10. Nasser Hajibagheri, M. A. (ed.). 1999. Electron Micro- Oxford University Press and Royal Microscopical Society,
scopy Methods and Protocols. Humana Press, Totowa, NJ. Oxford, United Kingdom.
Another general reference that every TEM lab should have This is handbook 19 of the series produced by the Royal
available; includes techniques for macromolecules as well. Microscopical Society, other volumes of which are noted in
11. RUSS,J.C. 1998. The Image Processing Handbook, 3rd ed. chapter 4. References 19 and 26 provide the reader with expla-
CRC Press, Boca Raton, FL. nations of symmetry groups found in macromolecular crystals,
Although image processing is encountered by everyone daily, including planar ones.
most books on the subject are written by and for physicists, 20. Hasler, L., J. B. Heymann, A. Engel, J. Kistler, and T.
engineers, and computer scientists and combine a great deal Walz. 1998. 2-D crystallization of membrane proteins: ra-
of mathematics with trivial examples such as models faces. tionales and examples. J. Struct. Biol. 121:162-171.
This book helps nonspecialists get an appreciation of the References 20 and 35 review some of the exciting advances in
tricks of the trade, presents a variety of real microscopic ex- the field of structure determination of membrane proteins by
amples, and is accompanied by software to help put principles cryoTEM.
into practice. 21. Henderson, R., J. M. Baldwin, T. A. Ceska, F. Zemlin,
12. Sommerville, J., and U. Scheer (ed.). 1987. Electron E. Beckmann, and K. H. Downing. 1990. Model for the
Microscopy in Molecular Biology: a Practical Approach. IRL structure of bacteriorhodopsin based on high-resolution
Press, Oxford, United Kingdom. electron cryo-microscopy. J. Mol. Biol. 2 13:899-929.
A standard reference book on preparing biological macromole- One of the classic milestones of biological macromolecular
cules, including DNA. TEM.
22. Jap, B. K., P. J. Walian, and K. Gehring. 1991. Structural
5.1 1.2. Specific References architecture of an outer membrane channel as determined
13. Cicicopol, C., J. Peters, A. Lupas, Z. Cejka, S. Miiller, by electron crystallography. Nature 350: 167-170.
R. Golbik, G. Pfeifer, H. Lilie, A. Engel, and W. A good example of the power of electron crystallography in elu-
Baumeister. 1999. Novel molecular architecture of the cidating membrane protein structure, of direct interest to struc-
multimeric archaeal PEP-synthase homologue (MAPS) turd and mokcular microbiologists.
from Staphylothermusmarinus. J. Mol. Biol. 290:347-361. 23. Li, W., F. P. Ottensmeyer, and G. Harauz. 2000.
References 13 and 23 are intended to provide background for Quaternary organization of the Staphylothermus marinus
the specific example used here to illustrate single-particle analy- phosphoenolpyruvate synthase: angular reconstitution
sis of a large macromolecularcomplex. from cryoelectron micrographs with molecular modelling.
14. Czarnota, G. J., D. R. Beniac, N. A. Farrow, G. Harauz, J. Struct. Biol. 132:226-240.
and F. P. Ottensmeyer. 2001. Three-dimensional electron 24. Ludtke, S. J., P. R. Baldwin, and W. Chiu. 1999. EMAN:
microscopy of biological macromolecules: quatemion- semiautomated software for high-resolution single-particle
assisted angular reconstitution of single particles, p. 275- reconstructions. J. Smct. Biol. 128:82-97.
294. In D. Hader (ed.), Image Analysis: Methods and 25. Radermacher, M. 2001. Three-dimensional reconstruc-
Applications, 2nd ed. CRC Press, Boca Raton, FL. tion of single particles in electron microscopy, p. 295-327.
References 14 and 25 describe two slightly different approaches In D. Hader (ed.), Image Analysis: Methods and Appli-
to angular reconstitution, i.e., the computational determination cations, 2nd ed. CRC Press, Boca Raton, FL.
of relative angular orientations amongst different projections of 26. Rhodes, G. 1993. Crystallography Made Crystal Clear: a
asymmetric macromolecules. Guide for Users of Macromolecular Models. Academic Press,
15. Ellis, M. J., and H. Hebert. 2001. Structure analysis of San Diego, CA.
soluble proteins using electron crystallography. Micron 27. Saxton, W. 0. 1996. SEMPER: distortion compensation,
32:541-550. selective averaging, 3-D reconstruction, and transfer func-
16. Engelhardt, H. 1988. Correlation averaging and 3-D re- tion correction in a highly programmable system. J. Struct.
construction of 2-D crystalline membranes and macromol- Biol. 116:230-236.
ecules. Methods Microbiol. 20:357413. 28. Sleytr, U. B., and T. J. Beveridge. 1999. Bacterial Slayers.
This review does for Fourier filtering and correlation averaging Trends Microbiol. 7:253-260.
what references 14, 2 5 , 3 1 , and 33 do for single-particle analy- An overall review of bacterial surface arrays, including TEM.
sis-it brings together the important concepts for a biologically 29. Stewart, M. 1990. Electron microscopy of biological
oriented reader without obscuring them by excessive use of macromolecules: frozen hydrated methods and computer
equations. image processing, p. 9-39. In P. J. Duke and A. G.
17. Frank, J., P. Penczek, R. K. Agrawal, R. A. Grassucci, Michette (ed.), Modern Microscopies-Techniques and
and A. B. Heagle. 2000. Three-dimensional cryoelectron Applications, Plenum Press, New York, NY.
microscopy of ribosomes. Methods Enzymol. 3 17:276-291. Quick big picture review.
The research groups of Frank and van Heel were pivotal both 30. Stewart, M., T. J.Beveridge, and T. J. Trust. 1986. Two
in developing techniques of single-particle analysis and in eluci- patterns in the Aeromonas salmonicida A-layer may reflect
dating the structures of ribosomal subunits to high resolution in a structural transformation that alters permeability.
work spanning well over the past two decades. Reference 33 J. Bacteriol. 166:120-127.
ranks also with references 2, 14, 25, and 31 in terms of re- This reference is intended to provide background to the specific
viewing single-particle analysis and cryoTEM and summarizes example used here to illustrate 2-D electron crystallography.
5. Computational Image Analysis and Reconstruction 95

31. Thuman-Commike, P., and W. Chiu. 2000. Recon- 33. van Heel, M., B. Gowen, R. Matadeen, E. V. Orlova, R.
struction principles of icosahedral virus structure determi- Finn, T. Pape, D. Cohen, H. Stark, R. Schmidt, M.
nation using electron cryomicroscopy. Micron 31:687- Schatz, and A. Patwardhan. 2000. Single-particle elec-
711. tron cryomicroscopy: towards atomic resolution. Q. Rev.
Of interest to wirologists; a good recent review of single-particle Biophys. 33:307-369.
analysis of highly symmetric objects. 34. van Heel, M., G. Harauz, E. Orlova, R. Schmidt, and M.
32. Unwin, N., and R. Henderson. 1984. The structure Schatz. 1996. The next generation of the IMAGIC image
of proteins in biological membranes. Sci. Am. 250: processing system. J. Stwct. Biol. 116:17-24.
78-94. 35. Walz, T., and N. Grigorieff. 1998. Electron crystallo-
A popular review intended for a general audience and of walue graphy of two-dimensional crystals of membrane proteins.
for a beginner because of the clarity of the illumations. J. Struct. Biol. 121:142-161.
Atomic Force Microscopy
YVES F. DUFRENE

6.1. SAMPLE PREPARATION .................................... 97


6.1.1. Substrates ............................................ 97
.2. Macromolecules ........................................ 97
.3. Cell Surface Layers ..................................... 97
.4. Whole Cells .......................................... 99
6.2. IMAGING ................................................ 99
6.2.1. Imaging Modes ........................................ 99
.2. Scanners, Cantilevers, and Probes ........................... 101
.3. Imaging Parameters ..................................... 101
.4.Imaging Environment .................................... 102
6.3. FORCE MEASUREMENTS ................................... 102
6.3.1. Principle of Force-Distance Curves .......................... 102
.2. Probing Molecular Interactions ............................. 103
.3. Measuring Cell Surface Elasticity ........................... 104
.4. Stretching Single Molecules ............................... 104
6.4. REFERENCES ............................................. 105

Light microscopy and electron microscopy have long been dent beam as in other classical microscopies, but by sensing
recognized as key tools in microbiological science (see the force between a very sharp probe and the sample sur-
chapters 1 and 4, respectively). Light microscopy is useful face. Thus, an AFM image is generated by recording the
for counting and identifying microbial cells as well as for force changes as the probe (or sample) is scanned in the x
determining their general morphological details. Because and y directions. The sample is mounted on a piezoelectric
the resolution is limited to 200 to 500 nm, i.e., the wave- scanner which ensures 3-D positioning with high resolu-
length of the light source, information at the (supra)molec- tion (Fig. 1).The force is monitored by attaching the probe
ular level is not accessible. High-resolution images of mi- to a soft cantilever, which acts as a spring, and measuring
crobial samples can be obtained by electron microscopy, the bending, or deflection, of the cantilever. The larger the
which uses high-energy electrons instead of light as the in- cantilever deflection, the higher the force that will be ex-
cident beam. Elegant techniques have been developed for perienced by the probe. Most instruments today use an op-
transmission electron microscopy (TEM) such as the use of tical method to measure the cantilever deflection with
freeze fracturing and surface replication to visualize, for ex- high resolution; a laser beam is focused on the free end of
ample, cell surface layers and the use of negative staining the cantilever, and the position of the reflected beam is de-
for studying purified structures such as flagella and fimbriae tected by a position-sensitive detector (photodiode).
(chapter 7). These approaches are limited by the require- This chapter focuses on the use of AFM in microbiology.
ment of vacuum conditions during the analysis, i.e., native, Rather than providing an exhaustive review of the litera-
hydrated samples cannot be directly investigated unless so- ture in this area, the chapter emphasizes methods and gives
phisticated cryoTEM methods are employed. Furthermore, recommendations for reproducible, reliable experiments.
electron microscopy does not easily allow quantitative Selected data are also presented to highlight the various ap-
height measurements. plications offered by the technique for microbiology (for re-
The advent of atomic force microscopy (AFM) (7), one views of papers published in this area, see references 17
technique in a family of new microscopies called scanning through 20 and 22). Section 6.1 concentrates on sample
probe microscopies, has recently opened a wide range of preparation procedures: selection of appropriate substrates
novel applications for microbiologists. The technique pro- and immobilization protocols available for isolated
vides three-dimensional (3-D) images of the surface ultra- macromolecules, cell surface layers, and whole cells.
structure with unprecedented resolution, in real time, Section 6.2 deals with the various aspects of AFM imaging:
under physiological conditions, and with minimal sample different imaging modes together with common problems
preparation. In addition, force measurements make it pos- and artifacts, imaging parameters, and imaging environ-
sible to probe physical properties such as molecular inter- ments. Section 6.3 focuses on AFM force measurements:
actions and mechanical properties. Hence, the combina- the principle of force-distance curves and their application
tion of AFM imaging and force measurements provides to probe molecular interactions and mechanical properties.
new avenues to study biological phenomena such as molec- It is hoped that this contribution will be useful to microbi-
ular recognition, protein folding, and cell adhesion (20a). ologists to evaluate the advantages and limitations of AFM
The general principle of AFM is surprisingly simple in their specific fields and to define appropriate procedures
(Fig. 1). AFM imaging is performed, not by using an inci- that will lead them to successful experiments.

96
6. Atomic Force Microscopy rn 97

Laser literature describing sample preparation protocols and im-


Photodiode I aging conditions for biomolecules is abundant and not spe+
cific to microbiology (14, 27, 33, 48, 53, 66). Therefore,
only the general features of these procedures will be men-
tioned.
Different methods are available to attach macromole-
cules to a substrate. For AFM in air, a simple immobilization
procedure consists of depositing a drop of an aqueous solu-
tion containing the macromolecule of interest onto the sub-
strate and letting the drop evaporate (38).Alternatively,
the molecules can be sprayed from aqueous solution or in
the presence of glycerol onto the substrate (46). Physical
adsorption in the presence of appropriate electrolytes is an-
other approach, which offers the advantage of allowing the
FIGURE 1 General design of an atomic force microscope. sample to be directly imaged under liquid without the need
for air drying. Addition of certain divalent cations may
sometimes promote the attachment.
When firm, irreversible attachment is desired, macromol-
6.1. SAMPLE PREPARATION ecules can be covalently bound to the substrate by using
chemically derivatized alkanethiol or silane monolayers. In
6.1 .I. Substrates the first case, gold substrates are functionalized with alka-
Sample preparation is a key factor that determines the suc- nethiols (for details, see section 6.3.2) terminated by car-
cess of an AFM experiment. One point of particular impor- boxyl functions which are then reacted with amino groups of
tance is that the sample must be well attached to an appro- proteins by using 1-ethyl-3~(3-dimethylaminopropyl) car-
priate solid substrate. For many applications, the substrate bodiimide (EDC) and N-hydroxysuccinimide (NHS) (59).
should typically be flat and easy to handle, prepare, and Alternatively, NHS-terminated monolayers can be directly
clean. Mica is certainly the most widely used substrate for formed by immersing the gold substrate in a solution of
biological applications. This layered mineral has strong 2-D dithio-bis-succinimidylundecanoate (73). The other ap-
bonds but is only weakly bound in the third dimension. As proach is based on the attachment of silane molecules such as
a result, it is easily cleaved, for instance by using an adhe- 3-aminopropyltriethoxysilane(APTES)onto glass or mica to
sive tape, to produce clean surfaces that are atomically flat create a positive surface charge which can then bind anionic
over large areas. Glass, in the form of glass coverslips, is an- molecules (43).
other suitable substrate for AFM studies. The fact that it is
transparent may be an important advantage for experiments 6.1.3. Cell Surface layers
in which AFM and light microscopy are combined. Because of their well-organized structure and high rigidity,
Furthermore, its surface is fairly smooth and can be chemi- microbial cell surface layers made of 2-D protein crystals
cally modified for specific applications (see below). Glass (i.e., S-layers) (Fig. 2) are ideal model systems for high-
coverslips, however, are always coated with organic con- resolution imaging and for monitoring of structural changes
taminants and particles that should be removed before use. in membrane proteins or other biological processes. AFM
This removal can be achieved by washing in concentrated yields structural information directly under physiological
acidic solution (HC1-HNO,, 3: 1) followed by ultrasonica- conditions, which makes it a complementary tool to X-ray
tion in several Milli-Q (Millipore) water solutions. and electron crystallography (57).
Both mica and glass have a hydrophilic surface. For some During the past decade, various immobilization strate-
applications, best results may be obtained by using hy- gies have been established for AFM of cell surface layers.
drophobic substrates, such as highly oriented pyrolytic The procedure is often based on physical adsorption from
graphite, a layered material from which flat, clean surfaces aqueous solution in the presence of appropriate electrolytes.
can easily be obtained by cleavage. Substrates may also be The optimal solution composition differs according to the
prepared by coating solids with a thin layer of metal such as type of specimen. Purple membranes from the archeon
gold. Gold-coated surfaces prepared by thermal evaporation Halobacterium contain bacteriorhodopsin, a light-driven
are relatively flat and can be easily functionalized with self- proton pump, in the form of highly ordered 2-D lattices.
assembled monolayers of organic alkanethiols (for details, Appropriate buffer conditions for adsorption are as follows
see section 6.3.2). The functionalized surfaces can then be (52, 54): the pH may vary from 4 to 10; 150 mM KC1 is
used as such or as substrates to covalently attach biomole- added with 10 mM morpholineethanesulfonic acid (MES),
cules (section 6.1.2). Finally, isoporous polymer membranes HEPES, or Tris for adjustment of the pH. The purple mem-
can be used to immobilize large objects such as whole cells branes are diluted in the buffer to a concentration of about
(section 6.1.4). 50 pg/ml, and a drop of this solution is deposited onto
In the following sections, various immobilization proce- freshly cleaved mica. After adsorption for 10 to 30 min,
dures are described. The protocols differ according to the samples are gently washed with buffer to remove mem-
type of sample investigated, i.e., macromolecules, cell sur- branes that are not firmly attached. By using this procedure,
face layers, or whole cells. individual bacteriorhodopsin molecules in the membrane
were imaged at subnanometer resolution (Fig. 2). Further-
6.1.2. Macromolecules more, force-induced conformational changes were directly
Since its invention, AFM has proved useful in imaging the visualized (50, 57).
structure and the conformation of isolated biomolecules, in- To adsorb in a defined orientation specimens that carry
cluding polysaccharides, proteins, and nucleic acids. The surface charges in an uneven distribution, such as purple
98 MORPHOLOGY A N D ULTRASTRUCTURE

sorption buffer made of 10 mM Tris-HC1 (pH 7.5), 150 mM


KC1, and 25 mM MgClZ onto freshly cleaved mica (65).A
volume of 3 pl of protein crystal solution (0.1 mg/ml) is
then injected into the drop. After 2 h of contact time, the
sample is rinsed with a recording buffer made of 10 mM
Tris-HC1, pH 7.5, and 150 mM KC1. AFM images revealed
2-D crystals with p42( 1)2 and p4 symmetry (65). Imaging
both crystal types before and after cleavage of the N termini
allowed the cytoplasmic surface to be identified. Flexibility
mapping and volume calculations identified the longest
loop at the extracellular surface; this loop exhibited a re-
versible force-induced conformational change.
A n elegant method has been developed to investigate
S-layers from Bacillus species in conditions relevant to their
native state (60,75). The proteins are recrystallized on a lipid
monolayer in a Langmuir-Blodgett trough, and the compos-
ite lipid-S-layer structure is then deposited onto a flat sub-
strate. Under these conditions, s-layers attach to the lipid
film with their inner face, which corresponds to the orienta-
tion found in the living organism. The protocol is as follows.
A 1-mg/ml chloroform-methanol (9: 1, vol/vol) solution of
the lipid 1,2-dipalmitoyl-sn-3 -phosphatidylethanolamine
(DPPE) is spread onto a subphase of 1 mM borate buffer (pH
9.0; 10 mM CaClZ) in a Langmuir-Blodgett trough. After
FIGURE 2 High-resolution AFM image, obtained with the evaporation of the solvent, the lipid molecules are com-
specimen in aqueous solution, of purple membrane adsorbed to pressed to a surface pressure of 30 mN/m. The clear super-
freshly cleaved mica (57). A fully reversible, force-induced natant of the S-layer protein solution is carefully injected be-
conformational change was observed: at the top of the image, neath the lipid monolayer into the subphase. After 18 to 20
the force applied to the atomic force microscope stylus was 100 h of recrystallization at 20"C, 1-by-1-cm silicon wafers are
pN. During scanning of the surface line by line, the force was carefully placed onto the liquid surface and removed after 15
increased until it reached 150 pN at the bottom of the image. min by being lifted horizontally from the surface with a pair
White outlines indicate bacteriorhodopsin trimers. (Reprinted of tweezers. Samples are rinsed twice with Milli-Q water and
from reference 57 with permission of the publisher.) mounted into the AFM liquid cell while avoiding dewetting.
AFM imaging in buffer solution may be facilitated by co-
valently immobilizing the sample on the substrate. This
type of procedure has been developed for the hexagonally
membranes, the surface properties of the substrate can be packed intermediate (HPI) layer of Deinococcus radioduruns
modified by coating with a polycation such as poly-L-lysine (35). First, silanization of gtass substrates is performed with
(53). The procedure involves dipping a mica or glass sub- the NHz-terminated reagent, silane APTES. Glass cover-
strate into a 1- to 5-mg/ml solution of poly-L-lysine for a few slips are covered with 30 ml of a 2% (wt/vol) solution of
minutes, rinsing with water, and drying. Before adsotbtion APTES in 95% (vol/vol) aqueous acetone and shaken for 3
of the specimen, the quality of the coating should be min. After removal of the solution, the substrates are cov-
checked, i.e., one should ensure that the substrate surface is ered with 30 ml of acetone and shaken for 5 min and the
not covered with polycation aggregates. acetone is removed. The last step is repeated 10 times, after
OmpF porin is a channel-forming protein of the outer which coverslips are put into an oven at 100C for 1 h. The
membrane of Eschen'chiu coli that forms 2-D crystals. The sample is then immobilized onto the derivatized glass sur-
sample preparation (63) consists of preincubating freshly face by means of the photoactivatable heterobifunc-
cleaved mica with 20 p1 of buffer made of 100 mM NaC1, tional cross+linkerN-5~azido-2-nitrobenzoyloxysuccinimide
20 mM HEPES, and 2 mM MgC12 (pH 7) for 5 min. After (ANB-NOS). Glass substrates are covered with 30 ml of 0.1
removal of the buffer, 1 to 3 pl of sample solution is added. M Na2C03, pH 9.0, containing 2 mg of ANB-NOS dis-
This solution contains porin trimers dissolved at 1 mg/ml in solved in 1 ml of dioxane and are shaken for 4 h. The ex-
octylpolyoxyethylene, mixed with dimyristoyl phos- cess reagent is removed by washing two to three times with
phatidylcholine at a lipid-to-protein ratio (wt/wt) of 0.2. four different solutions: NaZC03 and 200 p l of butylamine,
After 10 min of contact time, the sample is washed with Na2C03,water, and acetone. Then, a volume of 1 to 2 pl
buffer to remove membranes that are not firmly adsorbed. of the protein solution (1 to 10 mg/ml) is placed between
The presence of Mg2+ is essential to compensate for the two derivatized coverslips which are squeezed between two
negative surface charges of both the sample and the sub- glass disks so as to bring the hydrophilic sample into close
strate at pH -7. This protocol allowed high-resolution im- contact with the hydrophobic cross-linker. The azide is ac-
aging of porin OmpF 2-D crystals, with a lateral resolution tivated with UV irradiation at 366 nm at a distance of 10
of 1 nm and a vertical resolution of 0.1 nm (64). The con- cm for 3 min, and the sample is thoroughly rinsed with
tours determined by AFM agreed well with 3-D structural water or buffer to remove excess protein. The above-
information from X-ray crystallography. In addition, volt- described immobilization strategy made it possible to image
age- and pH-dependent conformational changes of the ex- the HPI layer surface in buffer solution with a lateral reso-
tracellular loops of the protein were demonstrated (49). lution of 1 nm, the images being consistent within a few
For E. coli aquaporin Z, a membrane channel protein, angstroms with data obtained by electron microscopy (36).
the adsorption protocol involves first putting a drop of ad- It was also shown that the inner surface of the HPI layer had
6. Atomic Force Microscopy 99

a protruding core with a central pore exhibiting two con- A


formations, one with and the other without a central plug.
In addition, individual pores were observed to switch from
one state to the other, indicating that conformational microbial cell
changes can be detected at subnanometer resolution (51). 3% agar gel
6.1.4. Whole Cells
There are several difficulties associated with the imaging of sample holder
whole microbial cells. First, as opposed to animal cells, mi-
crobial cells have a well-defined shape and have no ten-
dency to spread over substrates. As a result, the contact area B
between cells and the substrate is very small, often leading
to cell detachment by the scanning probe. A second prob-
lem is the vertical motion of the sample (or probe), which ,microbial cell
is typically limited to a few micrometers. This makes it dif-
ficult to image the surfaces of large objects, such as micro.
bial cells. For those reasons, immobilization by means of
simple adsorption procedures is often inappropriate for in-
vestigating whole microbial cells.
For AFM studies related to bioadhesion issues, one im- FIGURE 3 Schematic illustration of two immobilization
mobilization method consists in exploiting the natural abil- methods for AFM investigation of native, single microbial
ity of microorganisms to firmly attach to solid substrates by cells. (A) Agar gel; (B) porous membrane.
means of extracellular polymeric substances. The sample
preparation is dependent on the type of organisms and sub-
strates investigated. It typically involves incubating the sub-
strate with a cell suspension for a given period of time, re- tissue and then placed onto the sample holder. The use of
sulting in the formation of a biofilm of adhering cells that such a soft, deformable immobilization matrix enabled in
can then be imaged by AFM (6). situ investigation of the growth process of Saccharomyces
For imaging of single cells, pretreatments such as air dry- cerevisiae cells and direct visualization of micrometer-sized
ing or chemical fixation can be used to promote cell attach- bud scars (25).
ment. The air-drying method typically involves placing a The second method consists of trapping spherical cells
5- to 50-yl droplet of a concentrated cell suspension on a 1-by- in a filter membrane with a pore size comparable to the di-
1-cm2substrate, mica or glass, and allowing drying in air (4, mensions of the cell (21, 31, 37, 70). Typically, a concen-
40). Best results are obtained when using submonolayer cell trated cell suspension (10 ml; lo6 cells per ml) is gently
coverages, which can be achieved by rinsing the substrate sucked through an isopore polycarbonate membrane
(three times in Milli-Q water) prior to air drying. Sometimes, (Millipore) with a pore size slightly smaller than the cell
covalent bonding of bacteria to silanized glass can be used size. After cutting of the filter (1 by 1 cm), the lower part is
since it does not appreciably affect the cell viability (10). carefully dried on a sheet of tissue and the specimen is then
Glass slides are soaked for 10 min in a 10% (wtlvol) silane attached to the sample holder by using a small piece of ad-
solution, made of either 3-aminopropyltrimethoxysilaneor hesive tape. This procedure offers two important advan-
3-aminopropyldimethoxysilane in methanol, and then tages, i.e., it is fairly simple and straightforward, and it does
rinsed with copious amounts of methanol followed by Milli- not involve a macromolecular system such as agar, thus pre-
Q water to remove excess silane. Then, a bacterial suspen- venting the risk of cell surface contamination. An illustra-
sion (10' cells/ml) containing 100 mM EDC and 40 mM tion of this immobilization procedure is given in Fig. 4,
NHS is put in contact with silanized slides and allowed to which shows an AFM height image, recorded under water,
shake (125 rpm) overnight. After rinsing with water, sam- of two dividing daughter cells of Lactococcus lactis. Cells are
ples are either air dried by gently blowing air across the slide trapped into a pore of the membrane, allowing repeated im-
(for imaging in air) or kept hydrated (for imaging under aging without cell detachment or cell damage.
water).
The above-described treatments may cause significant
rearrangement or denaturation of the surface molecules, 6.2. I M A G I N G
yielding a surface which is no longer representative of the
native, hydrated state. To circumvent this problem, immo- 6.2.1. Imaging Modes
bilization procedures that do not involve drying or chemi- A number of AFM imaging modes are available. The most
cal fixation have been developed, namely, immobilization widely used is the contact mode, in which sample topogra-
in agar gels (Fig. 3A) and in porous membranes (Fig. 3B). phy can be measured in different ways. In the constant-
In the first method ( 2 5 ) , 5 p.1 of a highly concentrated cell height mode, one simply records the cantilever deflection
suspension is deposited onto a clean coverglass. About 200 while the sample is scanned horizontally, i.e., at constant
p.1 of 3% (wtlvol) molten agar (50C) is dropped onto the height. Minimizing large deflections, thus maintaining the
cell layer. Another piece of coverglass is quickly put on top applied force at small values, is often necessary to prevent
of the agar before hardening to create a flat surface. After 30 sample damage. This is achieved in the constant-deflection
min, the solidified sample is turned upside down and the mode, in which the sample height is adjusted to keep the
lower coverglass is pulled from the agar surface. Most cells deflection of the cantilever constant by using a feedback
are localized on that side of the agar disk, and weakly cap- loop (Fig. 1). The feedback output is used to display a true
tured cells can be easily removed by washing with water. height image. In many cases, small cantilever deflections do
The cell-free side of the disk is carefully dried on a sheet of occur because the feedback loop is not perfect and the
100 MORPHOLOGY AND ULTRASTRUCTURE

recorded as a function of time following addition of protease


revealed the progressive formation of large depressions,
about 500 nm in diameter and 50 nm in depth, reflecting
the erosion of the mannoprotein outer layer. In other work,
AFM was combined with thin-section TEM to investigate
the changes in the cell walls of Staphylococcus uureus cells as
they grow and divide (70). Nanoscale perforations were
seen around the septa1 annulus at the onset of division and
found to merge with time to form a single, larger perfora-
tion. These holes were suggested to reflect so-called muro-
somes, i.e., cell wall structures possessing high autolytic ac-
tivity and which digest peptidoglycan. After cell separation,
concentric rings were observed on the surface of the new
cell wall and suggested to reflect newly formed peptidogly-
can. Besides that of fungi and bacteria, it is worth noting
that AFM may also be used to reveal the surface architec-
ture of diatoms and viruses (15).
Accordingly, these data show distinct advantages of AFM
over classical electron microscopy: (i) the ability to measure
critical dimensions on the nanoscale such as the thickness of
hydrated cell wall layers or the size of supramolecular struc-
tures and (ii) the possibility of probing surface ultrastructure
in a hydrated state and in real time. Great advances are also
FIGURE 4 AFM height image (2range = 750 nm) recorded being made with cryoelectron microscopy, in which hy-
under water showingLactococcus h t k cells trapped by a pore of drated specimens can be examined in a vitrified state. This
a polycarbonate membrane. By trapping the cells mechanically is briefly described in chapter 4 in section 4.1.4.2.
into a porous membrane, single cells can be imaged under phys- Several other AFM imaging modes have been devel-
iological conditions without any pretreatment. oped. Although their use in microbiology has been limited
so far, they are briefly considered here because of their great
potential for imaging surface topography at minimal applied
forces and for mapping physical properties. Dynamic imag-
resulting error signal can be used to generate a so-called de- ing modes consist of oscillating the probe or sample at a
flection image. given frequency. In tapping mode AFM (TMAFM), or in-
For microbial specimens, both height and deflection im- termittent contact mode, the probe is excited externally
ages are often useful because they yield complementary in- and the amplitude and phase of the cantilever are moni-
formation. The height image provides quantitative height tored near the resonance frequency of the cantilever.
measurements, thus allowing an accurate measure of surface TMAFM has been used to record high-resolution images of
roughness, the height of surface features, or the thickness of purple membranes without detectable deformation (47), to
biological layers. The deflection signal is more sensitive to image Shewanella putrefaciens bacteria adhering to iron-
fine surface details than the height signal because the fre- coated and uncoated silica surfaces (26), to examine bio-
quency response is much higher. This characteristic makes films formed by Pseudomonas sp. on hematite and goethite
it valuable to reveal the surface ultrastructures of curved or minerals (24), and to observe changes in bacterial cell mor-
rough samples such as microbial cells, for which conven- phology resulting from adhesion-modifying chemicals (10).
tional height images are often of poor resolution. Although Force modulation microscopy is another dynamic mode
lateral dimensions are accurately measured in the deflection which measures the amplitude and phase shift of the can-
imaging mode, this is not the case for vertical variations. tilever while the sample or the probe is vibrated, thereby al-
Hence, one should keep in mind that deflection images do lowing estimation of spatial viscoelasticity variations.
not provide quantitative height measurements. The com- Further information on the different AFM imaging modes
plementarity of both height and deflection imaging modes can be found in the literature (14,44,48).
is illustrated in Fig. 5 for the surfaces of native, dormant Time resolution is a crucial limitation of current AFM
spores of the filamentous fungus Aspergillus oryzue (71). In imaging modes. For instance, recording a high-resolution
the height image (Fig. 5A), one can see that the spore sur- image of a 2-D protein crystal typically takes about 30 s, but
face is rough and heterogeneous. The vertical cross section for cells this procedure may take minutes, meaning that
shows a -35-nm step height, reflecting the thickness of the many dynamic processes occurring at short time scales are
outer cell wall layer, which is in good agreement with elec- not accessible. Interestingly, novel scanning probe instru-
tron microscopy data. The high-resolution deflection image ments are currently being developed with increased imaging
recorded for the outer layer surface (Fig. 5B) shows well- rates (millisecond resolution), suggesting that within a few
ordered structures called rodlets, that are 10 nm in diameter years microscopists should have new possibilities for prob-
and assembled in parallel to form fascicles interlaced with ing molecular and cellular dynamics (32). Another issue is
different orientations. the alteration of the specimen by the tip during scanning.
Because AFM can be operated in aqueous solution, it Here also, significant progress is being made to improve ex-
can be used to visualize in real time how cell surfaces inter- isting imaging modes. In particular, the recent active reso-
act with external agents or change during cell growth. For nance control in TMAFM makes it possible to reduce tip-
instance, real-time imaging was used to monitor the diges- specimen interactions, thus yielding images of biological
tion of S. cerewisiae cell walls ( 3 ) . Images of the cell surface specimens with improved resolution (32).
6. Atomic Force Microscopy H 101

-
50 nm

0 0.5 1 1.5
Distance (pm)
FIGURE 5 AFM height (A) and deflection (B) images recorded under water for the surfaces of
dormant spores of the fungus Aspergillus oryxae. The height image (A) together with the vertical cross
section (taken along the dashed line) reveal the presence of a heterogeneous outer layer -35 nm
thick. The deflection image (B) shows that the outer laver is made of rodlet structures 10 ? 1 nm in
diameter.

6.2.2. Scanners, Cantilevers, and Probes tive force measurements, spring constants must be deter-
By applying appropriate voltages, piezoelectric materials ex- mined experimentally (13, 67).
pand in some directions and contract in others in a defined The size and shape of the AFM probe play an important
way. This i s the basic idea behind the piezoelectric scanner role in determining the resolution and the contrast of the
(Fig. I), which is used to position the sample in the x, y, and images. At first glance, it is surprising to see that
g directions with high accuracy and precision. Scan ranges (sub)nanometer resolution is routinely acquired on recon-
are typically up to 100 p m laterally and a few micrometers stituted surface layers by using commercial probes with 10-
vertically. One problem associated with the piezoscanner is to 50-nm radii of curvature. It is thought that nanoscale
that the expansion (i.e., the response) is not exactly linear protrusions or asperities extending irregularly from the
with the applied voltage and shows hysteresis when the ef- probe are responsible for the high resolution.
fects of increasing and decreasing applied voltages are com- A common source of artifacts in imaging is the broaden-
pared. For large displacements, this nonlinearity can be im- ing of the surface features due to the finite size of the AFM
portant and be a source of inaccuracies. probe. This effect is important when imaging height varia-
Today, most AFM cantilevers and probes are made of sil- tions in the range of the probe radius of curvature, i.e., 10-
icon (Si) or silicon nitride (Si3N4) by using microfabrica- to 50-nm. In this case, the sample interacts with the sides of
tion techniques. Because the cantilever deflection is usually the probe and the resulting image will be a combination of
monitored by means of a laser beam, the backside of the the real sample topography and of the probe geometry. The
cantilever is often coated with a thin layer of gold to main consequence is that surface depressions appear smaller
enhance its reflectivity. The probe shape is either conical while protrusions appear broader. By using image-processing
(Si) or pyramidal (Si3N4), with apex radii of curvature of methods, the probeesample interaction can be modeled and
about 10 nm (Si) and 20 to 50 nm (Si3N4).For topographic an image resembling more closely the actual surface may be
imaging, cantilevers are often used as received. However, restored (76). Another common problem is the shadowing
organic contaminants on the probe may substantially influ- or multiplication of small structures produced by multiple
ence the image quality. The use of a mixture of concen- probe effects. These are due to the presence of multiple as-
trated sulfuric acid and hydrogen peroxide (piranha solu- perities on the probe apex which usually originate from
tion) is an easy and effective way to remove this organic contamination. If this problem is encountered during the
contamination (41). For biological specimens, best results course of an experiment, it is recommended that the probe
are generally obtained with cantilevers exhibiting small be changed before proceeding further.
spring constants, i.e., in the range of 0.01 to 0.10 N/m. Note
that actual spring constants may substantially differ from 6.2.3. Imaging Parameters
values quoted by the manufacturer. Therefore, when accu- In the constant-deflection mode, the sample height is ad-
rate knowledge of the force is required, such as in quantita- justed to keep the deflection of the cantilever constant by
102 w MORPHOLOGY A N D ULTRASTRUCTURE

using a feedback loop. To achieve accurate height adjust- the aqueous solution when filling the space between the
ments, the feedback loop can be varied in different ways by sample and the probe holder.
using various gain parameters (for more details on feedback High-resolution imaging requires vibrational and ther-
gain parameters, see reference 48). Most AFM systems use mal stability. External vibration isolation, especially in the
at least two gain controls, i.e., proportional and integral 10. to 100-Hz frequency range, can be accomplished by
gains. It is often recommended to use a proportional gain mounting the microscope stage on a mechanically isolated
higher than the integral gain. The gains should be high platform, such as an optical table with air bearings or heavy
enough so that the probe can track accurately every surface stone plinths hanging from elastic cords. To minimize ther-
feature; however, a too-high gain will result in oscillations mal drift, it is important to wait at least 10 min so that the
and instabilities. imaging force can be maintained constant and at low val-
The operator should optimize the rate at which the ues. If a longer time is required to achieve thermal stability,
image is acquired. A n AFM image is obtained by raster one should keep in mind the possible risk of surface alter-
scanning the probe over the sample, i.e., the sample or ation due to contamination or cell lysis.
probe is moved along a line in the so-called fast scanning di- Another important factor for achieving (sub)molecular
rection and then moves up to another scan line in the slow resolution is to use an adequate liquid composition because
scanning direction. The scan rate is defined as the rate in it will directly affect the forces between the probe and the
the fast scanning direction and thus depends on the scan- sample. For macromolecules such as polysaccharides (38)
ning frequency and on the image size. The scan rate should and DNA (28), excellent results have been obtained by im-
not be too high when nanometer resolution is desired be- aging under alcohols. For ordered microbial layers, aqueous
cause at high scan rates the probe may not respond faith- buffers are generally used, the buffer composition having a
fully to nanoscale features. On the other hand, low scan profound influence on the image resolution. For instance,
rates may cause sample damage when imaging soft, fragile for purple membrane and OmpF porin surfaces (55), it was
samples. Therefore, scan rates are often in the range of 0.5 possible to improve the spatial resolution by changing the
to 2 p,m/s. pH and electrolyte concentration, best results being ob-
The force acting between the probe and the sample is tained with 150 mM KC1 or NaCl (in 10 mM Tris-HC1, pH
another key parameter to control in order to obtain reliable, 7.6). Accordingly, a general recommendation for micro-
high-resolution images. Strong imaging forces may dramat- scopists investigating new microbial samples is to investi-
ically reduce the image resolution and cause molecular gate the effects of pH and ionic strength on the image qual-
damage or displacement. When imaging in air, a layer of ity in order to define an optimal imaging environment.
water condensation and other contamination often covers
both the probe and the sample, forming a meniscus pulling
the two together. The resulting strong attractive force, usu- 6.3. FORCE MEASUREMENTS
ally 10 to 100 nN, makes high-resolution imaging difficult
and sometime causes sample damage. The capillary force 6.3.1. Principle of Force-Distance Curves
can be eliminated by performing the imaging in aqueous so- Measuring the force acting between the AFM probe and the
lution. By selecting appropriate buffer conditions, it is gen- sample is important in defining the imaging force and thus
erally possible to maintain an applied force in the range of optimizing the image resolution. As we shall see, AFM force
0.1 to 0.5 nN. measurements also provide microbiologists and biophysi-
In addition to the sample surface topography, lateral cists with the opportunity to probe the sample physical
forces (friction) between the probe and the sample may sig- properties. Remarkably, the force sensitivity of the instru-
nificantly contribute to the contrast of an AFM image. To ment, in the piconewton range, allows the forces associated
assess the contribution of lateral forces to the apparent with single biomolecules to be measured.
topographic contrast, it is advisable to compare images ob- Force measurements are performed by monitoring, at a
tained by forward and backward scanning. Identical forward given (x,y ) location, the cantilever vertical deformation, or
and backward images indicate that frictional effects are deflection, as a function of the vertical displacement of the
likely to be minimal. It may also be useful to change the piezoelectric scanner (z). This process yields a raw voltage-
scanning angle, say by 90, to evaluate whether the ob- displacement curve which can be converted into a force-
served contrast is affected by the scanning direction. distance curve as follows. By using the slopes of the curves
in the region where the probe and the sample are in con-
6.2.4. Imaging Environment tact, the voltage can be converted into a cantilever deflec-
For most microbiological applications, water is the most rel- tion value. In order to minimize the possible effects of re-
evant factor in the imaging environment. Imaging under pulsive surface forces and/or sample deformation, it is
water requires the use of a liquid cell, which is provided with recommended to consider the slope of the retraction curve.
most commercial microscopes. Depending on the cell de- The cantilever deflection is then converted into a force (F)
sign, the sample can be attached to the sample holder either by using Hookes law: F = -k x d, where k is the cantilever
mechanically or by using a small piece of double-sided ad- spring constant and d is the cantilever deflection. The curve
hesive tape or glue. When using the second option, keep in can be corrected by plotting F as a function of (z - d). The
mind the possible risk of sample contamination. While zero separation distance is then determined as the position
mounting the wet sample into the cell, it is important to of the vertical linear parts of the curve in the contact re-
avoid passing the sample through the air-water interface gion. Interestingly, spatially resolved mapping can be per-
and so also avoid contact with air bubbles, which may reor- formed by recording multiple force-distance curve arrays in
ganize or denature the structures of interest. Some liquid the (x,y) plane (for more details on the principle of force-
cells use an O-ring to seal the sample and probe holder. distance curves, see references 9 and 11).
However, best results can be obtained by leaving the system Different parts may be distinguished in a force-distance
open. In this case, it is strongly recommended to protect the curve (Fig. 6 ) . At long probe-sample separation distances,
scanner head with a piece of parafilm to avoid contact with the force experienced by the probe is null. As the probe ap-
6. Atomic Force Microscopy H 103

lipopolysaccharide molecules on the cell surface and by the


t 3 production of the capsular polysaccharide, i.e., colanic acid
(61).
Q,
2
0
I * Reproducible, quantitative force measurements require
detailed control of the probe surface chemistry. The surface
LL
0- chemistries of most commercial probes are fairly complex,
and the probe surface is often contaminated with gold and
other materials. Probes of well-defined chemical composi-
tions can be designed by functionalizing the surface with or-
ganic monolayers terminated by specific functional groups
(e.g., OH and CH3). The most common method is based on
4
the formation of self-assembled monolayers of alkanethiols
I on gold surfaces. The procedure involves coating, by ther-
0 mal evaporation, microfabricated cantilevers with a thin ad-
Separation distance -F hesive layer (Cr or z), followed by a 15- to 100-nm-thick
FIGURE 6 AFM force measurements: different parts of a Au layer, immersing the coated cantilevers in dilute (0.1 to
force-distance curve. Labels indicate the approach (1,2,3)and 1 mM) ethanol solutions of the selected alkanethiol, rinsing
retraction (3,4,4') parts. with ethanol, and drying by using a gentle nitrogen flow.
Although the protocol is fairly simple, it is important to val-
idate the quality of the surface modification. This can be
proaches the surface, the cantilever may bend upwards due done by treating model substrates (glass, mica) in the same
to repulsive forces until the probe jumps into contact when way as the probes and characterizing them by means of sur-
the gradient of attractive forces exceeds the spring constant face analysis techniques (e.g., contact angle measurements
plus the gradient of repulsive forces. Upon retraction of the or X-ray photoelectron spectroscopy) ( 16). Another impor-
probe from the surface, the curve often shows a hysteresis tant point is to use the functionalized probes immediately
referred to as the adhesion pull-off force, which can be used after they are prepared in order to minimize surface con-
to estimate the surface energies of solids or the binding tamination.
forces between complementary biomolecules. In the pres- Chemically functionalized probes can be used to map
ence of long, flexible molecules, an attractive force, referred physicochemical properties, such as surface hydrophobicity
to as an elongation force, may develop nonlinearly due to and surface charge. By using OH- and CH3-terminated
macromolecular stretching. The next sections show how probes, patterns of nanoscale rodlets were visualized under
the different portions of force-distance curves can be used physiological conditions on the surfaces of spores of
in microbiology for probing molecular interactions, measur- Phanerochaete chrysosporium (16). Multiple (1,024) force-
ing sample elasticity, and stretching single molecules. distance curves recorded over 500-by-500-nm areas at the
spore surface, either in deionized water or in 0.1 M NaCl so-
6.3.2. Probing Molecular Interactions lutions, always showed no adhesion for both OH- and CH3-
Cellular events such as microbial adhesion, microbial ag- terminated probes. Control experiments indicated that the
gregation, and molecular recognition play a pivotal role in lack of adhesion is due to the hydrophilic nature of the
the natural environment, in medicine, and in biotech- spore surface. Following a similar approach, COOH probes
nological processes. Understanding the molecular basis of allow the mapping of surface charges and local isoelectric
these phenomena requires knowledge of the physical prop- points at cell surfaces (2).
erties of cell surfaces and of their molecular interactions. A n alternative approach consists of immobilizing micro-
Until recently, these properties were not directly accessible bial cells directly on the AFM probe. In this case, the im-
to study. aging capability of the atomic force microscope cannot be
The approach portion of force-distance curves can be used, meaning that cell surface morphology cannot be con-
used to characterize van der Waals and electrostatic forces, trolled while measuring forces. A typical procedure is to ad-
solvation, and hydration and steric and bridging forces asso- sorb poly( ethyleneimine) from a 1% (wtlvol) aqueous solu-
ciated with polymer-covered surfaces (9). By measuring elec- tion onto the probe (58). After rinsing of the probe with
trostatic forces, at different pH values, between standard sil- deionized water, a cell pellet is transferred onto the
icon nitride probes and the surfaces of purple membranes, poly( ethy1eneimine)-coated probe by using a micromanipu-
the surface charge density of the latter was estimated to be lator and the coated probe is treated with a drop of 2.5%
-0.05 C/m2 (8). In the same way, the surface potential of (vol/vol) glutaraldehyde at 4C for 1 to 2 h and finally
2-D bacteriorhodopsin crystals could be determined and the rinsed in water. This method has been used to measure the
isoelectric point was found to be about 4 (29). In the pres- forces between E. coli-coated probes and solids of different
ence of 20 mM KC1, long-range repulsion forces were mea- surface hydrophobicities (58). Both attractive forces and
sured at the surfaces of purple membranes and OmpF porins cell adhesion behavior were promoted by substrate surface
by using silicon nitride probes, reflecting the electrostatic hydrophobicity, pointing to the role of hydrophobic inter-
double-layer repulsion (55). These forces could be adjusted actions. In the above-described method, denaturation of
by changing the pH or the electrolyte concentration to op- cell surface molecules is likely to occur. This problem can be
timize the image resolution. No repulsion force was found for avoided by using probes coated with native living cells (42).
the HPI layers, indicating that the surface charge of the lat- To this end, glass beads are incubated in a solution of
ter was either too small to be.detected or positive. Force- APTES (5 to 10% in acetone) for 6 h and the cells are then
distance curves can detect subtle differences in cell surface linked to the amino-functionalized beads by spinning a cell-
composition. For instance, interaction forces between sili- bead mixture at 7,000 x g for 2 min. A single bacterium-
con nitride probes and confluent monolayers of isogenic coated bead is then attached to a cantilever with epoxy. By
strains of E. coli mutants were affected by the lengths of core using this approach, the forces between living Shewanella
104 MORPHOLOGY AND ULTRASTRUCTURE

oneidensis bacteria and goethite were measured quantita- bridging one or more grooves can be obtained. The force
tively (42). measurement consists of placing the probe at the midpoint
Finally, AFM probes can also be functionalized with of the unsuspended wall component region and increasing
biomolecules in order to measure intermolecular forces be- or decreasing the force as a linear function of time.
tween individual ligands and receptors (30, 39). Such Comparison of the cantilever deflections for the specimen
nanobioprobes were used to measure the specific inter- and the hard substrate allows an accurate determination of
actions between cell surface lectins and mannose residues the specimen elasticity. Applying this approach to the pro-
involved in the flocculation (aggregation) of the yeast teinaceous sheath of the archeon Methanospirillurn hungutei
Saccharomyes curlsbergensis (69). To measure these interac- GP1 yielded an elastic modulus of 2 X 10 to 4 X lo1
tions at the single-molecule level, gold-coated AFM probes N/m2, indicating that the sheath could withstand an inter-
were functionalized with thiol-terminated hexasaccharide nal pressure of 400 atm, well beyond that needed for a eu-
molecules, a strategy which provides enhanced accessibility bacterial envelope to withstand turgor pressure (77). For
of the carbohydrate moieties to the lectin binding sites. The murein sacculi of gram-negative bacteria in the hydrated
force curves recorded for S. curkbergensis in flocculating state, elastic moduli of 2.5 X lo7N/m2 were measured (78).
conditions and carbohydrate-coated probes showed adhe- Interestingly, lateral variations of elasticity on single cells
sion forces of 121 t 53 pN, reflecting the specific interac- can be detected by using spatially resolved force maps. To
tion between individual cell surface lectins and glucose this end, arrays of force-distance curves are recorded in par-
residues. In the presence of mannose, the adhesion was allel with topographic images. Each curve is then converted
strongly reduced, indicating that mannose had blocked the into a force-indentation curve to extract local elastic modu-
lectin receptor sites, thus confirming the specificity of the lus (or Youngs modulus) values. Applied to budding yeast
measured adhesion force. It is believed that such nanobio- cells, this approach yielded Youngs modulus values of 6.1
probes have important promise in clinical microbiology and and 0.6 MPa for the bud scar and surrounding cell surface,
pathogenesis to investigate the interactions between micro- respectively (68). This result showed that the bud scar is 10
bial pathogens and host cells and localize cell surface recep- times stiffer than the surrounding cell wall, a finding which
tors, which may eventually help in developing new thera- is consistent with the accumulation of chitin in this area.
peutic approaches.
6.3.4. Stretching Single Molecules
6.3.3. Measuring Cell Surface Elasticity How stiff is a single polysaccharide molecule? What are the
The mechanical properties of microbial cell walls and sur- forces required to unfold a protein? What are the interac-
face appendages play an important role in controlling tions responsible for the anchorage of proteins in cell mem-
events such as cell growth, cell division, and cell adhesion. branes? How strong are the forces driving the formation of
By pressing, in a controlled way, the AFM probe onto cell supramolecular assemblages?These questions, of great rele-
wall components or whole cells, it is possible to measure vance in cellular and molecular microbiology, can now be
quantitatively sample elasticity. addressed by means of AFM force spectroscopy.
So far, both the AFM probe and the sample were as- When the AFM probe is pulled away from a sample that
sumed to be nondeformable. In practice, most microbiolog- is made of long, flexible macromolecules, the retraction
ical samples are fairly soft and can be significantly deformed curves often show attractive elongation forces, developing
by the probe, resulting in a contact region which is no nonlinearly, which reflect the stretching of the macromole-
longer a vertical line such as that represented in Fig. 6 but cules (Fig. 6). In such experiments, referred to as single-
a curve. By subtracting the approach curves obtained for a molecule force spectroscopy experiments, it is common to
soft sample and for a rigid reference material (substrate), it present the force curve as the positive pulling force versus
is possible to generate a so-called force+indentationcurve, the extension (e.g., see Fig. 8). Two models from statistical
the shape of which provides direct information on the sam- mechanics are often used to describe the elasticity of long,
ples elastic properties (for methodological details, see refer- flexible molecules, i.e., the worm-like chain (WLC) and
ence 74). the freely jointed chain (FJC) models (34). The WLC
Multiple force-indentation curves recorded in buffer so- model describes the polymer as an irregular, curved fila-
lution for a gram-negative magnetic bacterium, Mugneto- ment, which is linear on the scale of the persistence length,
spirillurn gryphiswuldense, were used to determine the effec- a parameter which represents the stiffness of the molecule.
tive compressibility of the cell wall, i.e., -42 mN/m (5). By In the FJC model, the polymer is considered as a series of
using approach curves, significant differences in the surface rigid, orientationally independent statistical (Kuhn) seg-
softnesses of the fibrillated Streptococcussulivarius HB strain ments, connected through flexible joints. An extended FJC
and the nonfibrillated S. sulivarius HBCl2 strain were (FJC+) model has been developed in which Kuhn seg-
demonstrated (72). In agreement with AFM and electron ments can stretch and align under force.
microscopy images, a long-range repulsion was measured on In recent years, force spectroscopy experiments have
the fibrillated strain and attributed to the compression of provided new insight into the nanomechanical properties of
the soft cell surface fibrils. single DNA, protein, and polysaccharide molecules (for re-
The AFM depression technique is an original ap- views, see references 12, 23, and 34). For dextran, a poly-
proach, complementary to the indentation method, for saccharide found in bacterial and yeast species, the defor-
probing the elastic properties of isolated bacterial cell wall mation at low forces was dominated by entropic forces and
components (77, 78). The main features of the procedure was described by an extended FJC model (45,62). At strong
are as follows. A drop of a solution containing the cell wall forces, the dextran filaments underwent a distinct confor-
components (archeal sheaths and peptidoglycan sacculi, mational change corroborated by molecular dynamic calcu-
etc.) is placed onto a hard substrate and allowed to dry. The lations. The polymer stiffened, and the segment elasticity
substrate is made of GaAs or Si3N4 and contains grooves was dominated by the bending of bond angles. Although
that are narrow compared to the width or the length of the the biological relevance of this behavior remains unclear, it
material to be investigated so that single wall components may significantly improve the polymer ductibility.
6. Atomic Force Microscopy 105

6
Q1
2
0
LL

0 20 40 60 80
Extension lnml
FIGURE 7 Combining AFM imaging and force spectroscopy to unzip proteins from the HPI layer
of Deinococcus rudioduruns (56). (A) Control AFM topograph of the inner surface of the HPI layer.
(B) Force-extension curve recorded for this inner surface region showing a saw-tooth pattern with
six force peaks of about 300 pN. (C) The same inner surface area imaged after recording of the force
curve; a molecular defect the size of a hexameric HPI protein complex has clearly been created.
(Reprinted from reference 56 with permission of the publisher.)

Another elegant study involving a microbial specimen is ditions in which electron transfer from the bacterium to
the unzipping of bacterial proteins from the HPI layer of the mineral is expected. Specific signatures in the force-
Deinococcus radiodurans by using combined AFM imaging extension curves were well-fitted with a WLC model and
and force spectroscopy (56). Force-extension curves suggested to reflect the unfolding of a 150-kDa putative
recorded for the inner surface of the HPI layer showed saw- iron reductase.
tooth patterns with six force peaks of about 300 pN (Fig. 7). Long, flexible macromolecules were stretched at the sur-
This behavior, well-fitted with a WLC model, was attrib- face of living fungal spores (71). As can be seen in Fig. 8,
uted to the sequential pulling out of the protomers of the force-extension curves recorded for germinating spores of
hexameric HPI protein complex. After recording of the Aspergillus oryzae showed single or multiple attractive forces
force curve, a molecular defect the size of a hexameric com- of 400- 2 100-pN magnitude, along with characteristic
plex was clearly visualized by means of high-resolution im- elongation forces and rupture lengths ranging from 20 to
aging. Hence, AFM allows for correlation of force measure- 500 nm. Elongation forces were well fitted with an FJC+
ments with resulting structural changes. model by using fitting parameters that were consistent with
Stretching macromolecules directly on living cells is the stretching of individual dextran molecules (45, 62).
very challenging due to the complex and dynamic nature of The measured forces, attributed to the stretching of long
the surfaces. The forces between living Shewanella oneiden- cell surface polysaccharides, may be of great biological rele-
sis bacteria and goethite, both commonly found in Earth vance. Incubating the spores of A. oryzae in liquid medium
near-surface environments, were measured quantitatively for a few hours leads to their germination and, in turn, to
(42). Energy values derived from these measurements their aggregation. Hence, one may expect that the elastic
showed that the affinity between S . oneidensis and goethite properties of long surface macromolecules may play a signif-
rapidly increases by two to five times under anaerobic con- icant role in modulating spore aggregation and other bioin-
terfacial processes. Macromolecular stretching was also per-
formed on the surface of Psewlomonas putida in solvents
spanning a range of polarity and ionic strengths to learn
about the macromolecule elasticity and adhesion. Shorter
lengths were observed at higher ionic strength, indicating
M- that the polymer chains were less extended in the presence
of salt (1).

6.4. REFERENCES
Abu-Lail, N. I., and T. A. Camesano. 2002. Elasticity of
Pseudomom put& KT2442 surface polymers probed with
0 250 500 750
single-moleculeforce microscopy. Langmuir 18:407 1-4081.
Ahimou, F., F. A. Denis, A. Touhami, and Y. F. DufrEne.
Extension (nm) 2002. Probing microbial cell surface charges by atomic
force microscopy. Langmuir 18:9937-9941.
FIGURE 8 Stretching cell surface macromolecules. Typical Ahimou, F., A. Touhami, and Y. F. DufrEne. 2003. Real-
force-extension curve recorded under water between a silicon time imaging of the surface topography of living yeast cells
nitride probe and the surface of a germinatingAspergillus oryzue by atomic force microscopy. Yeast 20:25-30.
spore (thin line, starting at 0 mm extension). Elongation forces Amro, N. A., L. I? Kotra, K. Wadu-Mesthrige, A.
were well described by an extended freely jointed chain model Bulychev, S. Mobashery, and G.-Y. Liu. 2000. High-
(thick line, starting at 200 mm extension) with parameters resolution atomic force microscopy studies of the Esche-
consistent with values reported for the elastic deformation of richia coli outer membrane: structural basis for permeability.
single dextran and amylose polysaccharides (71). Langmuir 16:2789-2796.
106 MORPHOLOGY AND ULTRASTRUCTURE

5. Arnoldi, M., C. M. Kacher, E. Bauerlein, M. 25. Gad, M., and A. Ikai. 1995. Method for immobilizing
Radmacher, and M. Fritz. 1998. Elastic properties of the microbial cells on gel surface for dynamic AFM studies.
cell wall of Magnetospirillurn gryphiswaldense investigated by Biophys. J. 69:2226-2233.
atomic force microscopy. Appl. Phys. A 66:S613-S617. 26. Grantham, M. C., and P. M. Dove. 1996. Investigation of
6. Beech, I. B., C. W. S. Cheung, D. B. Johnson, and J. R. bacterial-mineral interactions using fluid tapping mode
Smith. 1996. Comparative studies of bacterial biofilms on atomic force microscopy. Geochim. Cosmochim. Acta
steel surfaces using atomic force microscopy and environ- 60:2473-2480.
mental scanning electron microscopy. Biofouling 10: 27. Hansma, H. G., and J. H. Hoh. 1994. Biomolecular im-
65-77. aging with the atomic force microscope. Ann. Rev.
7. Binnig, G., C. F. Quate, and C. Gerber. 1986. Atomic Biophys. Biomol. Strmt. 23:115-139.
force microscope. Phys. Rev. Lett. 56:930-933. 28. Hansma, H. G., J. Vesenka, C. Siegerist, G. Kelderman,
8. Butt, H.-J. 1992. Measuring local surface charge densities H. Morrett, R. L. Sinsheimer, V. Elings, C. Bustamante,
in electrolyte solutions with a scanning force microscope. and P. K. Hansma. 1992. Reproducible imaging and dis-
Biophys. J. 63578-582, section of plasmid DNA under liquid with the atomic
9. Butt, H.+J., M. Jaschke, and W. Ducker. 1995. force microscope. Science 256:1180-1184.
Measuring surface forces in aqueous electrolyte solution 29. Hartley, P., M. Matsumoto, and P. Mulvaney. 1998.
with the atomic force microscope. Bioelectrochem. Bioenerg. Determination of the surface potential of two-dimensional
38:191-201. crystals of bacteriorhodopsin by AFM. Langmuir 14:5203-
10. Camesano, T. A., M. J. Natan, and B. E. Logan. 2000. 5209.
Observation of changes in bacterial cell morphology using 30. Hinterdorfer, P., W. Baumgartner, H. J. Gruber, K.
tapping mode atomic force microscopy. Langmuir Schilcher, and H. Schindler. 1996. Detection and local-
16:4563-4572. ization of individual antibody-antigen recognition events
11. Cappella, B., and G. Dietler. 1999. Force-distance curves by atomic force microscopy. Proc. Natl. A c d . Sci. USA
by atomic force microscopy. Surf. Sci. Rep. 34:l-104. 93:347 7-348 1.
12. Clausen-Schaumann, H., M. Seitz, R. Krautbauer, and 31. Holstein, T. W., M. Benoit, G. V. Herder, G. Wanner,
H. E. Gaub. 2000. Force spectroscopy with single bio- C. N. David, and H. E. Gaub. 1994. Fibrous mini-
molecules. Cum. Opin. Chem. Biol. 4:524-530. collagens in Hydra nematocysts. Science 265:402-404.
13. Cleveland, J. P., S. Manne, D. Bocek, and P. K. Hansma. 32. Horber, J. K. H., and M. J. Miles. 2003. Scanning probe
1993. A nondestructive method for determining the spring evolution in biology. Science 302:1002-1005.
constant of cantilevers for scanning force microscopy. Rev. 33. Ikai, A. 1996. STM and AFM of bio/organic molecules
Sci. Instrum. 64403405. and structures. Surf. Sci. Rep. 26:261-332.
14. Colton, R. J., A. Engel, J. E. Frommer, H. E. Gaub, A. A. 34. Janshoff, A., M. Neitzert, Y. Oberdorfer, and H. Fuchs.
Gewirth, R. Guckenberger, J. Rabe, W. M. Heckel, and 2000. Force spectroscopy of molecular systems-single
B. Parkinson (ed.). 1998. Procedures in Scanning Probe molecule spectroscopy of polymers and biomolecules.
Microscopies. John Wiley & Sons Ltd., Chichester, Angew. Chem. Int. Ed. 39:3213-3237.
England. 35. Karrasch, S., and A. Engel. 1998. AFM imaging of HPI
15. Crawford, S. A., M. J. Higgins, P. Mulvaney, and R. layers in buffer solution, p. 433-439. In R. J. Colton, A.
Wetherbee. 2001. Nanostructure of the diatom frustule as Engel, J. E. Frommer, H. E. Gaub, A. A. Gewirth, R.
revealed by atomic force and scanning electron mi- Guckenberger, J. Rabe, W. M. Heckel, and B. Parkinson
croscopy. J. Phycol. 37543-554. (ed.), Procedures in Scanning Probe Microscopies. John
16. DufrCne, Y. F. 2000. Direct characterizationof the physic- Wiley & Sons Ltd., Chichester, England.
ochemical properties of fungal spores using functionalized 36. Karrasch, S., R. Hegerl, J. Hoh, W. Baumeister, and A.
AFM probes. Biophys. J. 78:3286-3291. Engel. 1994. Atomic force microscopy produces faithful
17. DufrCne, Y. F. 2001. Application of atomic force mi- high#resolution images of protein surfaces in an aqueous
croscopy to microbial surfaces: from reconstituted cell sur- environment. Proc. Natl. A c d . Sci. USA 91:836-838.
face layers to living cells. Micron 32: 153-165. 37. Kasas, S., and A. Ikai. 1995. A method for anchoring
18. DufrCne, Y. F. 2002. Atomic force microscopy, a powerful round shaped cells for atomic force microscope imaging.
tool in microbiology. J. Bacteriol. 184:5205-5213. Biophys. J. 68:1678-1680.
19. DufrCne, Y. F. 2003. Recent progress in the application of 38. Kirby, A. R., A. P. Gunning, and V. J. Morris. 1996.
atomic force microscopy imaging and force spectroscopy to Imaging polysaccharides by atomic force microscopy.
microbiology. Cum. Opin. Microbiol. 6:317-323. Biopolymers 38:355-366.
20. DufrCne, Y. F. 2004. Using nanotechniques to explore mi- 39. Lee, G. U., L. A. Chrisey, and R. J. Colton. 1994. Direct
crobial surfaces. Nat. Rev. Microbiol. 2:451-460. measurement of the forces between complementary
ZOa.DufrCne, Y. F. 2003. Atomic force microscopy provides a strands of DNA. Science 266:771-773.
new means for looking at microbial cells. ASM News 40. Lister, T. E., and P. J. Pinhero. 2001. In vivo atomic force
69:438-442. microscopy of surface proteins on Deinococcw rudiodurans.
21. Dufrsne, Y. F., C. J. P. Boonaert, P. A. Gerin, M. Langmuir 17:2624-2628.
Asther, and P. G. Rouxhet. 1999. Direct probing of the 41. Lo, Y. S., N. D. Huefner, W. S. Chan, P. Dryden, B.
surface ultrastructure and molecular interactions of dor- Hagenhoff, and T. P. Beebe. 1999. Organic and inorganic
mant and germinating spores of Phanerochaete chrysospo- contamination on commercial AFM cantilevers. Langmuir
rium. J. Bacteriol. 1815350-5354. 15:6522-6526.
22. Firtel, M., and T. J. Beveridge. 1995. Scafining probe mi- 42. Lower, S. K., M. F. Hochella, and T. J. Beveridge. 2001.
croscopy in microbiology. Micron 26:347-362. Bacterial recognition of mineral surfaces: nanoscale inter-
23. Fisher, T. E., P. E. Marszalek, and J. M. Fernandez. 2000. actions between Shewanella and ol-Fe00H. Science
Stretching single molecules into novel conformations 292: 1360-1363.
using the atomic force microscope. Nat. Sauct. Biol. 43. Lyubchenko, Y. L., A. A. Gall, L. S. Shlyakhtenko,
7:719-724. R. E. Harrington, P. I. Oden, B. L. Jacobs, and S. M.
24. Forsythe, J. H., P. A. Maurice, andL. E. Hersman. 1998. Lindsay. 1992. Atomic force microscopy imaging of dou-
Attachment of a Pseudomom sp. to Fe(111)-(hydr)oxide ble stranded DNA and RNA. J. Biomol. Sauct. Dyn.
surfaces. Geomicrobiology 15:293-308. 9589-606.
6. Atomic Force Microscopy w 107

44. Magonov, S. N., and M.-H. Whangbo (ed.). 1996. hesion monitored by atomic force microscopy. Proc. Nutl.
Surface Analysis with STM and AFM. VCH, New York, NY. Acad. Sci. USA 95~11059-11064.
45. Marszalek, P. E., A. F. Oberhauser, Y.-P. Pang, and J. M. 62. Rief, M., F. Oesterhelt, B. Heymann, and H. E. Gaub.
Fernandez. 1998. Polysaccharide elasticity governed by 1997. Single molecule force spectroscopy on polysaccha-
chair-boat transitions of the glucopyranose ring. Nature rides by atomic force microscopy. Science 275:1295-1297.
396:661-664. 63. Schabert, F. A. 1998. Imaging of reconstituted OmpF
46. McIntire, T. M., and D. A. Brant. 1997. Imaging of indi- porin in solution using AFM, p. 429-432. In R. J. Colton,
vidual biopolymers and supramolecular assemblies using A. Engel, J. E. Frommer, H. E. Gaub, A. A. Gewirth, R.
noncontact atomic force microscopy. Biopolymers 42: 133- Guckenberger, J. Rabe, W. M. Heckel, and B. Parkinson
146. (ed.), Procedures in Scanning Probe Microscopies. John
47. Moller, C., M. Allen, V. Elings, A. Engel, and D. J. Wiley &a Sons Ltd., Chichester, England.
Muller. 1999. Tapping mode atomic force microscopy pro- 64. Schabert, F. A., C. Henn, and A. Engel. 1995. Native
duces faithful high-resolution images of protein surfaces. Escherichia coli OmpF porin surfaces probed by atomic force
Biophys. J. 77:1150-1158. microscopy. Science 268:92-94.
48. Morris, V. J., A. R. Kirby, and A. P. Gunning (ed.). 65. Scheuring, S., P. Ringler, M. Borgnia, H. Stahlberg,
1999. Atomic Force Microscopy for Biologists. Imperial D. J. Muller, P. Agre, and A. Engel. 1999. High resolu-
College Press, London, England. tion AFM topographs of the Escherichia coli water channel
49. Miiller, D. J., and A. Engel. 1999. Voltage and pH- aquaporin 2. EMBO J. 18:49814987.
induced channel closure of porin OmpF visualized by 66. Shao, Z., J. Mou, D. M. Czajkowsky, J. Yang, and 1.-Y.
atomic force microscopy. J. Mol. Biol. 285:1347-1351. Yuan. 1996. Biological atomic force microscopy: what is
50. Muller, D. J., G. Biildt, and A. Engel. 1995. Force- achieved and what is needed. Adw. Phys. 45:l-86.
induced conformational change of bacteriorhodopsin. 67. Tortonese, M., and M. Kirk. 1997. Characterization of
J. Mol. Biol. 249:239-243. application specific probes for SPMs. SPIE 3009:53-60.
51. Miiller, D. J., W. Baumeister, and A. Engel. 1996. 68. Touhami, A., B. Nysten, and Y. E Dufrene. 2003.
Conformational change of the hexagonally packed inter- Nanoscale mapping of the elasticity of microbial cells by
mediate layer of Deinococcus radioduruns monitored by atomic force microscopy. hngmuir 19:4539-4543.
atomic force microscopy. J. Bacteriol. 178:3025-3030. 69. Touhami, A., B. Hoffmann, A. Vasella, F. A. Denis, and
52. Muller, D. J., M. Amrein, and A. Engel. 1997. Y. F. Dufrene. 2003. Aggregation of yeast cells: direct
Adsorption of biological molecules to a solid support for measurement of discrete lectin-carbohydrate interactions.
scanning probe microscopy. J. Struct. Biol. 119:172-188. Microbiol. SGM 149:2873-2878.
53. Muller, D. J., A. Engel, and M. Amrein. 1997. 70. Touhami, A., M. Jericho, and T. J. Beveridge. 2004.
Preparation techniques for the observation of native bio- Atomic force microscopy of cell growth and division in
logical systems with the atomic force microscope. Biosens. Staphylococcus aureus. J. Bacteriol. 186:3286-3295.
Bioelecrron. 12:867-877. 71. van der Aa, B. C., R. M. Michel, M. Asther, M. T.
54. Muller, D. J., G. Buldt, and A. Engel. 1998. Preparation Zamora, P. G. Rowhet, and Y. F. Dufrene. 2001.
and observation of purple membranes by AFM, p. 425428. Stretching cell surface macromolecules by atomic force
In R. J. Colton, A. Engel, J. E. Frommer, H. E. Gaub, A. A. microscopy. hngmuir 17:3116-3119.
Gewirth, R. Guckenberger,J. Rabe, W. M. Heckel, and B. 72. van der Mei, H. C., H. J. Busscher, R. Bos, J. de Vries,
Parkinson (ed.), Procedures in Scanning Probe Microscopies. C. J. P. Boonaert, and Y. F. Dufrene. 2000. Direct prob-
John Wiley & Sons Ltd., Chichester, England. ing by atomic force microscopy of the cell surface softness
55. Miiller, D. J., D. Fotiadis, S. Scheuring, S. A. Muller, of a fibrillated and non-fibrillated oral streptococcal strain.
and A. Engel. 1999. Electrostatically balanced sub- Biophys. J. 78:2668-2674.
nanometer imaging of biological specimens by atomic 73. Wagner, P., M. Hegner, P. Kernen, F. Zaugg, and G.
force microscope. Biophys. J. 76:llOl-1111. Semenza. 1996. Covalent immobilization of native
56. Muller, D. J., W. Baumeister, and A. Engel. 1999. biomolecules onto Au( 111) via N-hydroxysuccinimide
Controlled unzipping of a bacterial surface layer with ester functionalized self-assembled monolayers for scan-
atomic force microscopy. Proc. Natl. Acad. Sci. USA 96: ning probe microscopy. Biophys. J. 70:2052-2066.
13170-13174. 74. Weisenhorn, A. L., M. Khorsandi, S. Kasas, V. Gotzos,
57. Muller, D. J., J. B. Heymann, F. Oesterhelt, C. Moller, and H.-J. Butt. 1993. Deformation and height anomaly of
H. Gaub, G. Buldt, and A. Engel. 2000. Atomic force mi- soft surfaces studied with an AFM. Nanotechnology 4:106-
croscopy of native purple membrane. Biochim. Biophys. 113.
Acta 1460:27-38. 75. Wetzer, B., D. Pum, and U. B. Sleytr. 1997. Slayer sta-
58. Ong, Y.-L., A. Razatos, G. Georgiou, and M. M. bilized solid supported lipid bilayers. J. Struct. Biol.
Sharma. 1999. Adhesion forces between E. coli bacteria 119:123-128.
and biomaterial surfaces. Langmuir 15:2719-2725. 76. Wilson, D. L., K. S. Kump, S. J. Eppell, and R. E.
59. Patel, N., M. C. Davies, M. Hartshorne, R. J. Heaton, Marchant. 1995. Morphological restoration of atomic
C. J. Roberts, S. J. B. Tendler, and P. M. Williams. 1997. force microscopy images. Langmuir 11:265-272.
Immobilization of protein molecules onto homogeneous 77. Xu, W., P. J. Mulhern, B. L. Blackford, M. H. Jericho,
and mixed carboxylate-terminated self-assembled mono- M. Firtel, and T. J. Beveridge. 1996. Modeling and mea-
layers. Langmuir 13:6485-6490. suring the elastic properties of an archaeal surface, the
60. Pum, D., M. Weinhandl, C. Hodl, and U. B. Sleytr. sheath of Methanospirillurn hungatei, and the implication
1993. Large-scale recrystallization of the Slayer of Bacillus for methane production. J. Bacteriol. 178:3106-3112.
coaguhns E38-66 at the air/water interface and on lipid 78. Yao, X., M. Jericho, D. Pink, and T. Beveridge. 1999.
films. J. Bacteriol. 175:2762-2766. Thickness and elasticity of gram-negative murein sacculi
61. Razatos, A., Y.-L. Ong, M. M. Sharma, and G. measured by atomic force microscopy. J. Bacteriol. 181:
Georgiou. 1998. Molecular determinants of bacterial ad- 6865-6875.
7
.

Cell Fractionation
SUSAN F. KOVAL AND G . DENNIS SPROTT

7.1. CELL BREAKAGE ......................................... 109


7.1.1. Mechanical Breakage .................................... 109
7.1.1.1. Pressure Shearing ................................ 109
. ........................... 110
.2 Ultrasonic Disintegration
. ............................ 111
3. Ballistic Disintegration
. ................................... 111
4. Solid Shearing
. ............................. 111
5. Freezing and Thawing
7.1.2. ChemicalLysis ........................................ 111
7.1.2.1. Detergent Uses and Actions ......................... 111
. ................................. 112
2. Ionic Detergents
. .............................. 112
3. Nonionic Detergents
. ...................................... 112
4. Bile Salts
. .......................... 112
5. Problems with Detergents
. ................................... 113
6. Dithiothreitol
7.1.3. Osmotic Lysis ......................................... 113
. 4. Enzymatic Lysis ........................................ 113
7.2. GENERATION OF PROTOPLASTS AND SPHEROPLASTS ......... 113
7.2.1. Protoplasts from Gram-Positive Bacteria ...................... 114
. 2. Spheroplasts from Gram-Negative Bacteria .................... 114
. 3. Osmotically Sensitive Forms from Archaea .................... 114
7.3. CENTRIFUGATION ........................................ 114
7.3.1. Differential Centrifugation ................................ 115
. 2. Rate-Zonal Centrifugation ................................ 116
. 3. Isopycnic Separations .................................... 116
. 4. Preparation and Fractionation of Density Gradients .............. 116
. 5. Rotor. Tube. and Bottle Selection........................... 117
. 6. Gradient Media ........................................ 118
7.3.6.1. Sucrose and Ficoll................................ 118
.
.2 Cesium Chloride and Potassium Bromide ............... 118
. ........................................
3. Percoll 118
. 4. Metrizamide.................................... 119
7.4. ISOLATION OF CELL COMPONENTS ......................... 119
7.4.1, Appendages ........................................... 119
7.4.1.1. Flagella........................................ 119
. .................................. 120
.2 Fimbriae (Pili)
. 3. Spinae........................................ 120
7.4.2. Walls and Envelopes .................................... 121
. 3. Wall Macromolecules .................................... 122
7.4.3.1. Murein and Pseudomurein .......................... 122
.
.2 Teichoic Acids.................................. 123
. 3.LPS .......................................... 123
. ....................................... 127
4. S-Layers
. ...............................
5. Exopolysaccharides 128
7.4.4. Plasma Membranes ..................................... 129
.
.5 Membranes of Gram-Negative Bacteria ....................... 129
. 6 Specialized Membranes ................................... 130
7.4.6.1. Photosynthetic Membranes ......................... 130
. 2. Purple and Red Archaeal Membranes .................. 130
7.4.7. Periplasm ............................................ 130
7.4.7.1. Chloroform Release of Periplasm ..................... 130
. ........................ 131
2. Osmotic Release of Periplasm
. ........................ 131
3. Calcium Release of Periplasm
7.4.8. Nucleoid ............................................. 131
. 9. Ribosomes ........................................... 131

108
7. Cell Fractionation 109

.lo. Endospores .......................................... 132


.........................................
.l 1. Gas Vesicles 132
.12. Inclusion Bodies ...................................... 132
.13. Magnetosomes ........................................ 133
7.5. REFERENCES ............................................. 133

This chapter presents techniques for the fractionation of eel. This choice will depend on how easily the microorganism is
lular components, including organelles and appendages, be- broken and on how the method of breakage affects the
ginning with external surfaces and ending with internal structural intactness and functional activity of the isolated
components. The structural and physiological diversity of component(s). The factors pertinent to cell fractionation,
prokaryotes makes it impossible to present a single universal often dictating the approach needed, are the differences in
method for cell fractionation or to summarize all of the ap- wall structures and the bonding forces within walls (and as-
proaches developed to fit each bacterium. Extra effort was sociated surface materials) responsible for maintaining cell
made to provide methods for the isolation of components shape.
from both bacterial and archaeal phylogenetic lineages. In Fundamental differences exist in the cell envelope struc-
an era of molecular biology, genomics, and proteomics, many tures of bacteria and archaea, which influence the choice of
researchers may not have experience in breakage and frac. cell breakage method. The Gram staining reaction subdi-
tionation procedures (excluding those for nucleic acids), so vides bacteria into gram-negative, gram-positive, and gram-
it is hoped that such individuals will find a revisit to some of variable species on the basis of wall structure (chapter
the germane, and also sometimes older, literature rewarding. 2.3.5.), and this staining may be useful as a first step in as-
Large variations in genes exist even within the sub- signing a method for breakage. In general, the gram-positive
groupings of bacteria and often lead to changes in structure; cocci have thick peptidoglycan layers; this characteristic
these structural variations can affect cell fractionation. and their shear-resistant shape make them the most re-
Indeed, a rational fractionation scheme can be developed silient to mechanical breakage. The Gram reaction has lim-
only on the basis of solid knowledge of a particular organ- ited value for archaea because their cell walls can be so fun-
ism. Since the information needed is often lacking for damentally different in structure and chemistry from those
newly isolated or poorly studied bacteria, some preliminary of bacteria as to defy conventional interpretation. Archaea,
exploration may be essential. It is important, for example, which include the methanogenic, extremely halophilic, and
to determine at what stage in the growth cycle and in which sulfur-dependent groupings, have at least five different en-
growth medium the component of interest is produced op- velope structures (71). Archaea that have only a proteina-
timally. Such factors can influence not only the yield, but ceous Slayer as the sole wall component are relatively frag-
also the chemical structures of cell components. ile (although the S-layer of some hyperthermophiles may be
It is recommended that the isolation of any cellular covalently bonded). Archaea with lysozyme-insensitive
component be monitored by electron microscopy (chapter pseudomurein may be more difficult to break. Thennoplasma
4) at all stages of the procedure. Electron microscopy often and Methanoplasma species resemble the mycoplasmas with
provides the only adequate means of monitoring the frac- respect to the complete absence of a wall structure.
tionation and the state of intactness of the cell component. The extent of breakage can be assessed by comparing the
This is especially important during the preliminary explo- cell counts for the suspension before and after treatment.
ration of a bacterium. For routine assessments, it is adequate to view wet mounts
Glassware must be carefully cleaned and sometimes ster- by phase-contrast microscopy or by negative staining with
ilized to avoid contamination from stray microbial cells and nigrosin (chapter 2.2.2.). Microscopy, providing direct ob-
enzymes, especially if the fraction is to be stored. Also, it is servation of the numbers and intactness of cells (chapter
preferable to store samples at -20C or below, provided 2.1.4. and 2.1.5.), has an advantage over techniques which
that the component resists freeze-thaw damage. Samples monitor the loss of cell viability, since viability may be lost
can be stored for short periods of time at 4"C, with the without a concomitant release of internal contents.
growth of microbial contaminants inhibited temporarily Alternatively, it is possible to make measurements to reflect
with poisons such as 0.05% (wtlvol) sodium azide. Con- the release of cytoplasmic contents into the supernatant fol-
tainment facilities may be needed to comply with safety reg- lowing centrifugation (e.g., 8,000 X g for 15 min). Markers
ulations if pathogenic bacteria are used, as well as to protect for cytoplasm include specific cytoplasmic enzyme activi-
against oxidative inactivation of specific components of ties, ions such as K+,and UV-absorbing cofactors or nucleic
anaerobic prokaryotes. acids.
The fractionation of cytoplasmic or organelle-associated
proteins is not discussed, since gel electrophoresis and the 7.1 .l. Mechanical Breakage
chromatography of soluble proteins are described in chapter Mechanical methods for cell breakage are the most univer-
21. It should be appreciated, however, that many of the sally successful. Each method, however, must be carefully
techniques described in this chapter, from cell breakage to assessed in light of the species studied and the cell fraction
centrifugation theory, will have similar applications. to be isolated.
7.1 .I . I . Pressure Shearing
7.1. CELL BREAKAGE Pressure shearing using the French pressure cell is probably
Because of differences in the architectures of prokaryotic the most widely used and useful method of cell breakage,
cell surfaces, there is perhaps no greater area of variability, particularly for gram-negative bacteria (including the
or importance, than the initial choice of breakage method. cyanobacteria) and gram-positive bacteria (particularly the
110 MORPHOLOGY AND ULTRASTRUCTURE

gram-positive bacilli). Gram-positive cocci are not broken may be used, but Tris buffer or chelating agents should be
readily by this device, and ballistic disintegration or the en- avoided because they cause outer membrane damage. Add a
zymatic digestion of the cell wall is more effective for break- small amount of pancreatic RNase and DNase I (-0.1
ing them. The French pressure cell is effective for many ar- mg/ml; omit Mg2+ to encourage ribosome dissociation), and
chaea, although Methunobrevibacter and Thermoplasma spp. break the suspension with a French pressure cell operated at
are particularly resistant. A n apparatus capable of achieving a cell pressure of 16,000 lb/in2 (1 lb/in2 = 6.895 kPa). After
higher pressures, such as the EmulsiFlexTM high-pressure breakage, add MgCl2 to give a final concentration of 1 mM,
homogenizers sold by Avestin (www.avestin.com), can and remove unbroken cells by centrifugation at 5,000 X g
overcome these difficulties. The EmulsiFlexTMseries, simi- for 5 min. The purpose of the M_g2+is to activate DNase.
lar in principle to the French pressure cell, have capacities RNase acts in the absence of Mg2 . Omission of the DNase
ranging from small scale (0.5 to 3.5 ml) to continuous flow allows for the isolation of short strands of DNA by this
(5 to <1,200 liter&) depending on the model chosen. The breakage method; viscosity is generally not a problem be-
pump operating these units is driven with compressed air or, cause of DNA shearing.
in the case of the larger units, with electrical power.
The French pressure cell consists of a steel cylinder, pis- 7.1.1.2. Ultrasonic Disintegration
-
ton, and pressure relief valve fitted with a replaceable nylon A variety of ultrasonic probe devices are available to break
ball. Additional equipment includes a loading stand and a both prokaryotic and eukaryotic cells. Programmable ultra-
10-ton (89,000 N ) motor-driven hydraulic press which can sonic liquid processors are now available (e.g., SonicatorTM
deliver a constant force over a range of piston speeds. The from Misonix Inc., Farmingdale, NY, and Sonic Dismem-
Aminco pressure cell, accessories, and a suitable hydraulic branator 500TMfrom Fisher Scientific). Operators will find
pump can be obtained from SLM Instruments, Inc. the timer-programming feature convenient, although older,
(Urbana, IL), or Canberra Canada Ltd. (Kanata, Ontario). nonprogrammable sonicators (e.g., MSE and Braun types)
Alternatively, the pump, which is the most expensive part function just as efficiently in most laboratories. The rapid
of the apparatus, can be purchased through a local supplier vibration of the probe tip produces high-intensity sound
of hydraulic equipment, although commercial units may re- waves, which generate microscopic gas bubbles (especially
quire some modification by a machine shop to fit the cell on small nucleation particles such as bacteria). Creation of
unit safely and satisfactorily. Aminco cells are available these transient cavities (cavitation) is thought to create
with maximum capacities of 3.7, 35, or 40 ml of sample. high shear gradients by microstreaming (59). This breakage
Both the 35- and 40-ml-capacity cells can be fitted with a method is tempting to use, since the probe devices are rela-
one-way return valve for continuous operation with larger tively inexpensive and effective. However, there are inher-
volumes of sample or for repeated processing. The larger ent disadvantages.
cells are recommended for most applications, since they are The major disadvantage is that breakage is not instanta-
more easily controlled than the smaller ones. neous; a cell suspension must be treated for 30 s to several
To use a French pressure cell, load the cell suspension minutes before a reasonable proportion of the cells are bro-
(up to 30% wet cell paste by volume) into the pressure cell, ken. During this time, subcellular particles released from
taking care to avoid a gas pocket. Close the relief valve, and broken cells are subjected to the same high shear forces as
place the entire assembly into the hydraulic press. Lower the unbroken cells. Membrane vesicles can be degraded
the hydraulic press piston onto the cell piston, and allow it into small lipoprotein fragments, which can no longer be
to reach the maximum force desired. Then slowly open the sedimented by ultracentrifugation, and extensive redistri-
relief valve to the point where the piston moves downward bution of membrane proteins among various membranes
while maintaining pressure. The cells are broken by the can occur (e.g., between the inner and outer membranes of
high shear force generated as the suspension passes through gram-negative bacteria).
the small orifice of the relief valve. In addition, it is difficult to control the temperature of
This method has several advantages. Heating can be the sample during breakage. Foaming can cause protein de-
minimized by precooling the press cylinder and piston; heat naturation, and cavitation promotes oxidation of oxygen-
is generated only when the cells pass through the relief sensitive enzymes and unsaturated lipids. These problems
valve. The effluent temperature will rise by 5 to 10C, but can be minimized by subjecting the sample to several short
the effluent can be rapidly cooled if it is collected in a metal bursts (15 to 30 s) rather than one continuous treatment,
tube or beaker in an ice bath. Anaerobiosis may be main- with cooling periods between the bursts. Covering the sam-
tained by flushing the chamber with argon or N2 while ple with argon during treatment may minimize oxidation. It
loading the cell suspension with a syringe preflushed with is difficult to obtain reproducible breakage, since the effec-
N2 and collecting the effluent in a serum vial continuously tiveness of the probe depends upon sample viscosity and the
flushed with N2. Breakage is virtually instantaneous, and size, shape, and composition of the sample vessel (e.g., a
the broken suspension is not subjected to additional shear glass beaker is far more effective than a plastic one).
forces that could damage subcellular particles. If a satisfac- Caution: wear ear protection, unless the probe is enclosed in
tory hydraulic press is used, very reproducible conditions a sound-absorbing cabinet. Avoid holding the vessel in the
can be obtained. The extent of breakage is little influenced fingers, because ultrasonic transmission is effective through
by the density of the cell suspension, the growth phase at glass.
which the cells are harvested, or the breakage medium in Although ultrasonic treatment is not recommended for
which the cells are suspended. Multiple passes are done as primary cell breakage, it may be useful for small-scale prepa-
needed. ration of walls or envelopes, for distinguishing soluble from
particulate proteins, or for the initial identification of S-
Breakage Procedure for Estherichia coli Cells layers (by electron microscopy). In these procedures, very
Harvest cultures, wash them once, and resuspend them at short bursts of ultrasonic energy are required so that subcel-
0.01 to 0.1 times the original culture volume in 0.01 M lular organelles are not damaged. For the processing of dif-
HEPES, pH 7.4, at 0C. Other buffers such as phosphate ficult cells, glass beads (10 to 50 pm in size) (see section
7 . Cell Fractionation w 111

7.1.1.3) can be added to the liquid to facilitate breakage. tar and pestle; they also may be broken by forcing a frozen
Staphylococcus species should be pretreated with lyso- suspension or paste through a small hole or slit. Hughes et
staphin. Ultrasonic treatment is useful for lysing sphero- al. (59) have provided a complete description of these
plasts (as in the procedure of Osborn and Munson [lo91 for methods.
the separation of plasma and outer membranes of gram-
negative bacteria), for dispersing clumps of subcellular or- 7.1 .I .5. Freezing and Thawing
ganelles, and for suspending centrifuge pellets. Freezing and thawing have long been used to release high-
M, DNA from a variety of cells (121). The efficiency of the
7.1 .I . 3 . Ballistic Disintegration process depends on the organism and the operator. A
The term ballistic disintegration describes a variety of meth- washed cell pellet is frozen in liquid Nz and ruptured by
ods in which prokaryotes are broken by the shear forces de- grinding the frozen cells with a pestle in a precooled mortar.
veloped when a suspension of cells together with small glass This procedure of lysis has been used for the isolation of
or plastic beads is shaken or agitated violently. Many com- DNA from Methanothemus fewidus (152) and several other
mercial devices have been used, and they are detailed in re- methanogens (62) and involves further treatment with
views by Salton (120) and by Hughes et al. (59). sodium dodecyl sulfate (SDS).
The Mickle shaker has been replaced by the MSK Tissue
DisintegratorTM (B. Braun, Melsungen, Germany) that is 7.1.2. Chemical Lysis
available from many laboratory supply houses in the United
States. The Braun disintegrator has a 65-ml sample con- 7.1.2.1. Detergent Uses and Actions
tainer that is shaken horizontally at 2,000 to 4,000 oscilla- Detergents have multiple uses in cell fractionation. (i)
tions/min. Liquid C02 delivered to the sample container When nonmembranous organelles such as ribosomes and
provides cooling. Most samples can be disintegrated in 3 to nucleoids are sought, detergents provide a gentle means of
5 min at a temperature of <4"C. The Braun disintegrator is lysing the cells once the integrity of the peptidoglycan
often the method of choice for the isolation of cell walls (gram-positive bacteria) or the outer membrane plus pepti-
and particulate enzymes from gram-positive bacteria. The doglycan (gram-negative bacteria) has been damaged. (ii)
following procedure (156) can be used with most microor- Detergents are used to selectively solubilize the plasma
ganisms. membranes of gram-negative bacteria while leaving the
Mix a cell suspension (30 ml, containing 20 to 50 mg outer membranes intact (section 7.4.2). (iii) Detergents
[dry weight]/ml) in a suitable buffer with 20 ml of glass also can be used to remove membrane contamination from
beads (Ballotini #12; 100 to 200 pm) in the sample con- ribosomes, polysomes, cell walls of gram-positive bacteria,
tainer (an air space of at least 20% of the container volume or other cell fractions after cell breakage. (iv) Extraction
is essential for breakage), and pass liquid COz through the and recovery of proteins expressed in E. coli can be simpli-
apparatus for 0.5 min to cool the container. Shake the con- fied by the use of kits containing chemical lysis agents that
tainer at 3,000 strokes/min for 3.5 to 5.0 min depending on gently disrupt the cell wall. These kits provide a simple,
the species. The beads may be removed by low-speed cen- rapid alternative to mechanical methods for releasing ex-
trifugation or by passage through coarse-grade sintered glass. pressed target proteins in preparation for purification or
Because glass beads may liberate alkali when shaken vio- other applications. Harsh conditions associated with pres-
lently, with potential damage to alkali-sensitive compo- sure disruption and sonication are avoided. One example is
nents, they must be acid washed, usually with nitric x i d , BugBusterTM. This protein extraction reagent from
before use. Suitable glass beads are supplied by Sigma Novagen (www.novagen.com) is a proprietary formulation
Chemical (St. Louis, MO). (Some workers prefer to use of a mixture of nonionic detergents advertised not to dena-
plastic beads [118], to minimize enzyme denaturation.) ture soluble proteins. A companion nuclease can be pur-
When this procedure is used for the isolation of cell walls, chased that reduces the viscosity of the extract. The low-
the sample should be treated promptly as described below to viscosity clarified extract is fully compatible with affinity
inactivate autolytic enzymes (156). chromatography resins for fusion proteins. Si ma also sup-
Bacterial endospores can be broken with the Braun dis- plies a comparable reagent called CelLytic B&.
integrator by shaking dry spores with dry, acid-washed glass Detergents are amphipathic molecules with both hy-
beads (5 g of dry spores plus 15 g of beads) as described above drophilic and hydrophobic regions and are sparingly soluble
and then suspending the sample in buffer (137). Beads in water. At very low concentrations, detergents form true
with a 470-pm diameter are appropriate; acid-washed beads solutions in water. As the concentration is increased, addi-
with diameters of 425 to 600 Fm are available from Sigma. tional molecules of detergent aggregate to form micelles.
The Bead-BeaterTM is sold by Biospec Products Here, the hydrophilic regions are exposed to water and the
(Bartlesville, OK) as an apparatus for homogenizing and dis- hydrophobic regions are shielded from water on the inside
rupting microorganisms. To use this apparatus, mix glass of the micelle. The concentration at which micelles begin
beads with up to 20 g (dry weight) of cells (500-pm diameter to form, as the amount of added detergent is increased, is
for yeast, fungi, and algae; 100 to 150 pm in diameter for bac- the critical micelle concentration (CMC). The CMC and
teria) in a 30- or 60-ml chamber. A Teflon rotor within the the size and shape of the detergent micelle are characteris-
chamber is driven by a conventional blender motor and pro- tic of each detergent. Membranes undergo various degrees
vides breakage by bombardment with the beads. An external of disintegration at different detergent concentrations. At
jacket may be filled with ice water to minimize heating dur- low concentrations, lysis or rupture is seen first; at interme-
ing the 2 to 3 min normally needed to achieve breakage. For diate detergent-to-membrane ratios (0.1 to 1 mg/mg of
small samples (- 1 ml), a mini Bead-Beater is available. membrane lipid), membrane proteins may be selectively
extracted; and at higher ratios (2 mg/mg of lipid), the mem-
7.1 .I .4. Solid Shearing brane may be solubilized by the formation of soluble mi-
Cells may be broken by grinding a cell paste or lyophilized celles of lipid-detergent, protein-detergent, and lipid-
cells with abrasives such as alumina by using an agate mor- protein-detergent (54). Early reviews by Helenius et al. (54)
112 4 MORPHOLOGY AND ULTRASTRUCTURE

and Zulauf et al. (160) summarize the properties of many of properties, but they have several advantages. They are avail-
the detergents used in cell fractionation. able as pure compounds of defined structures, and they gener-
Detergents may be grouped into three classes, which dif- ally have much higher CMCs, which makes them more useful
fer in micelle properties, protein binding, and response to in chromatography and facilitates removal by dialysis or gel
other solutes. These classes are ionic detergents (cationic, filtration. They are available in different acyl chain lengths,
anionic, and zwitterionic), nonionic detergents, and bile which in turn results in ranges in properties such as CMC, the
salts. A variety pack of detergents may be purchased from ease of dialysis, and micelle M,. High cost is a major disad.
Calbiochem, La Jolla, CA, to explore suitability for specific vantage.
applications .
7.1.2.4. Bile Salts
7.1.2.2. Ionic Detergents Bile salts are sterol derivatives, such as sodium cholate, de-
The most commonly used ionic detergents are SDS (also oxycholate, and taurocholate. Because of the poor packing
known as sodium lauryl sulfate), sodium N-lauroylsarcosine ability of the bulky sterol nucleus, these detergents form
(Sarkosyl), alkyl benzene sulfonates (common household small micelles (often of just a few molecules), and unlike
detergents), and quaternary amine salts such as cetyltri- those for other detergents, the micelle M, is a function of
methylammonium bromide. Ionic detergents tend to form concentration. As these detergents are salts of very insolu-
small micelles (molecular mass [MA, -10 kDa) and exhibit ble weak acids with pK,s in the range of 6.5 to 7.5, they
rather high CMCs (the CMC of SDS is about 0.2% [wt/vol] must be used at an alkaline pH. To avoid solubility prob-
at 20C in dilute buffers). The CMC and the micelle solu- lems, stock solutions are often prepared by dissolving the
bility of ionic detergents are strongly influenced by the free acid in excess NaOH. Ionic composition, pH, and total
ionic strength of the solution and the nature of the counte- detergent concentration affect their usefulness and must all
rions present. For example, a 10% (wtlvol) solution of SDS be maintained constant when these detergents are used.
is soluble down to about 17"C, whereas a similar solution of For the release of genomic DNA from rod-shaped
Tris dodecyl sulfate is soluble at 0C. Potassium dodecyl sul- halobacteria, sodium taurocholate may be preferred to
fate is soluble only at elevated temperatures, and K+ must sodium deoxycholate, since it dissolves more readily in the
be excluded from all buffers when this detergent is used. presence of high salt concentrations. The lysis of cells by
The guanidinium salt of SDS is also insoluble. bile salts is effective for only select members of the archaea
Detergents such as SDS, which have a hydrophilic group and bacteria (70).
that is strongly ionized, are not affected by pH and are not
precipitated by 5% (wtlvol) trichloroacetic acid. Ionic de- 7.1.2.5. Problems with Detergents -
tergents bind strongly to proteins, and for SDS, this strong In excess, detergents act by forming mixed micelles with
binding usually results in unfolding and irreversible denatu- lipids or by binding to proteins, forming soluble protein-
ration of proteins. This is the basis for their use in polyacry- detergent complexes. Since most detergents are used at con-
lamide gel electrophoresis (chapter 15.5.1). Although ionic centrations well above the CMC, the ratio of detergent to
detergents can pass through dialysis membranes, they bind protein or lipid is much more important than the actual de-
strongly to proteins and cannot be removed completely by tergent concentration. As a general rule, one must use at
dialysis. least 2 to 4 mg of detergent per mg of sample protein to en-
sure a satisfactory excess of detergent. For example, if 2%
7.1.2.3. Nonionic Detergents - (vol/vol) Triton X-100 is used for membrane solubilization,
Commercial nonionic detergents include the polyoxyethyl- the maximum protein content of the sample should be less
ene detergents such as Triton X-100, Nonidet P-40, Brij 58, than 5 to 10 mg/ml.
and Tween 80 (54). These detergents are not pure but a Triton X-100, one of the most useful nonionic deter-
mixture of related compounds. In general, these detergents gents, presents problems in protein assays because it has an
have high-molecular-mass (50-kDa or greater) micelles and aromatic moiety that prevents the measurement of Also and
low CMCs (0.1% or less), which limits their usefulness in because it (and other nonionic or cationic detergents)
procedures such as gel filtration and electrophoresis and forms a cloudy precipitate in chemical protein assays.
makes them difficult to remove by dialysis. Although their Proteins can be assayed in the presence of these detergents
properties are not strongly affected by pH or ionic strength, by the modification of the procedure of Lowry et al., de-
they may be precipitated by 5% (wtlvol) trichloroacetic scribed by Peterson (113), in which an excess of SDS is
acid. The advantage of these detergents is that they bind added to the sample. This addition leads to the formation of
primarily to hydrophobic sites on the surfaces of proteins stable mixed micelles of the detergents that do not interfere
and thus do not cause extensive denaturation or loss of bio- with the assay.
logical activity. Since nonionic detergents are uncharged, Triton X-100 and other similar nonionic detergents are
they do not interfere with separations that are based on the soluble in ethanol-water mixtures, and proteins dissolved in
charge character of the cellular component. As noted in these detergents may be freed from the detergents by
section 7.4.2, the outer membrane resistance of many gram- ethanol precipitation as follows. Place the sample on ice,
negative bacteria to nonionic (124) or weakly ionic (36) and add 2 volumes of ice-cold absolute ethanol with stir-
detergents allows the application of these detergents to the ring. Allow the sample to stand overnight in a freezer, and
isolation of cell surface components. collect the protein precipitate by centrifugation. Efficient
There are a number of nonionic detergents consisting of precipitation requires a protein concentration of at least 0.2
acyl derivatives of sugars that have been synthesized for scien- mg/ml. Dilute samples may be concentrated by using an ul-
tific rather than commercial applications. Examples include trafiltration apparatus (Amicon Corp., Beverly, MA) with a
octyl-P-D-glucopyranoside, octyl-P-D-thioglycopyranoside, filter with an appropriate pore size, although this procedure
and dodecyl-P-D-maltoside. These are available from supply is limited by the possibility that detergent micelles will also
houses such as Calbiochem and Sigma. These detergents are be concentrated. Triton detergents can be removed by an
similar to Triton X- 100 in their membrane protein-solubilizing SM-2 resin (Bio-Rad Laboratories, Richmond, CA) (58).
7. Cell Fractionation m 113

SDS can be removed from samples by acetone precipita- 2. Resuspend thawed cells in 10-ml ABE buffer (50mM
tion as follows. Stir 6 volumes of anhydrous acetone into ammonium bicarbonate-50 mM disodium EDTA, pH 8.0).
the sample at room temperature, and recover the precipitate Chelators of divalent cations (e.g., EDTA) facilitate lysis in
by centrifugation. Wash the precipitate several times with cases where the cell wall has cationic cross bridging.
acetone-water (6:1, vol/vol). Since the precipitate is often 3. Add pronase to give 40 pg/ml, and incubate for 1 h at
waxy and difficult to work with, it may be dispersed in water 37C. If DNA is to be isolated, nucleases in the pronase
with a Potter-Elvehjem homogenizer and then lyophilized. (Roche, Indianapolis, IN) must be inactivated by autodi-
A n alternative method for SDS removal, used often in gestion in 50 mM Tris-HC1 (pH 8.0) for 1 h at 37C. Note
DNA purification, is precipitation of the potassium salt of that proteinase K cannot be substituted for pronase with
SDS made by the dropwise addition of potassium acetate to some strains.
a final concentration of 0.3 to 0.5 M. 4. Add dithiothreitol (200 pg/ml) for 15 min at 23C.
5. Finally, add SDS (1 mg/ml), and heat the mixture at
7.1.2.6. Dithiothreitol 55C for 15 min to promote lysis.
Dithiothreitol will induce the lysis of the sheathed
methanogens in the absence of an osmotic protectant, pro- Note that not all steps are necessary for all strains; the
viding that the pH is moderately alkaline (133). The pro- protease digestion step is likely to be unnecessary for most
cedure can be scaled up to allow the isolation of cellular or- applications, excluding nucleic acid isolations, and can
ganelles or enzymes from Methanospirillurn hungatei or often be omitted.
Methanosaeta concilii but is generally ineffective with most
7.1.4. Enzymatic Lysis
other methanogens (100, 133).
Osmotic lysis may be achieved in the many cases where os-
7.1.3. Osmotic Lysis motically sensitive cell forms can be generated by enzymatic
Osmotic lysis in hypotonic solution may be achieved for activity, as described next in section 7.2.
some bacteria without the need of first weakening the cell
wall. Extreme halophiles such as Halobacterium, as well as
many moderate halophiles, require salts to maintain struc- 7.2. GENERATION OF PROTOPLASTS
tural integrity and lyse in deionized water. The lytic suscep- A N D SPHEROPLASTS
tibility of a new culture should therefore be tested by sus- The strength of most bacterial walls resides in the murein
pension in low-ionic-strength solutions. Because of its (peptidoglycan) component, which may be damaged or de-
simplicity, osmotic lysis is recommended as a first option for stroyed by specific enzymes (e.g., lysozyme), leading to os-
lysis, provided that the component to be isolated is stable motically sensitive cells. The surrounding medium is gener-
under these conditions. Gas vesicles have been isolated ally hypotonic (or can be made so by dilution) so that lysis
from Halobacterium salinarum by using salt dilution (130). results, the extent of which should be monitored by phase-
Environmental cations can affect susceptibility to lysis in contrast microscopy (chapter 1). Lysis may be prevented by
water (86,95). For example, 18 marine and 2 terrestrial gram- adding 0.3 to 0.8 M sucrose and 0.5 to 10 mM Mg2+ to the
negative isolates were found to give a range of lytic responses reaction mixture to generate round and osmotically sensi-
in distilled water; some lysed after exposure to Mg2+-contain- tive, intact cells. These sensitive forms are protoplasts (from
ing solutions and others were sensitized by washing in 0.5 M gram-positive cells), free of wall material but with the
NaCl (e.g., Pseudomom ueruginosa). Others resisted lysis in plasma membrane remaining, or spheroplasts (from gram-
distilled water even after washing with NaCl (e.g., E. coli). negative types), with both outer and plasma membranes re-
Many methanogenic bacteria that have S-layers as the sole maining. Protoplasts and spheroplasts may not be perfectly
wall component (e.g., Methanococcus woltae and Methano- round, and it is important to check their morphology with
caldococcus jannaschii) and moderate halophiles (e.g., that of untreated cells. In addition to sucrose and Mg2+,
Methanosarcina mazei) lyse in water. other osmotic stabilizers may be used (e.g., polyethylene gly-
A general procedure for osmotic lysis in low-ionic- col, sodium succinate, and certain salts from 0.2 to 0.8 M).
strength solutions, from Laddaga and MacLeod (86), is as The enzymes that degrade murein can be divided into
follows. three classes: carbohydrases, acetylmuramyl-L-alanine ami-
1. Harvest exponential-growth-phase cells at 8,000 X g dases, and peptidases (66). Those most commonly used in
for 10 to 20 min, and wash them in an equal volume of 0.5 forming protoplasts and spheroplasts are the muramidase
M NaCl up to three times (the outer membranes of gram- subgroup of the carbohydrases, notably lysozyme. Structural
negative bacteria can sometimes be removed with further variations in the murein can lead to differences in sensitiv-
washes in 0.5M sucrose). ity to murein-lytic enzymes. For example, the extent of 0
2. Resuspend the cell pellet in deionized water. acetylation of murein directly affects its susceptibility to egg
white lysozyme (31). Since the extent of 0 acetylation may
Extraordinary resistance to lysis may require individual- increase during the stationary phase (65), it is generally
ized procedures. The sulfur-dependent, wall-less ther- recommended that exponential cultureslbe used.
mophile Thewnoplasma acidophilum grows optimally at pH 2 Osmotically sensitive forms may also be generated by in-
and can be lysed by raising the pH to 6 or 7. The organism teractions with antibiotics. Penicillins (p-lactam antibi-
otherwise resists mechanical breakage, nonionic detergents, otics) cause specific inhibition of murein biosynthesis and
pronase, and trypsin and is osmotically stable (87). lead to unbalanced growth and lysis of P-lactamase-negative
A procedure combining chemical and osmotic tech- bacteria in a hypotonic environment. In the presence of hy-
niques is effective with a wide variety of methanogens, as pertonic sucrose, osmotically sensitive spheres are formed
follows (100). from many rod-shaped gram-negative (spheroplasts) and
1. Centrifuge a 10-ml sample of cell culture (0.3 to 0.5 mg gram-positive (protoplasts) bacteria, such as Bacillus subtilis
[dry weight]/ml), freeze overnight at -20C as cell paste, and Bacillus meguterium. For the technique to work effec-
and thaw. tively, rapidly growing mid-exponential-phase cells are
114 MORPHOLOGY AND ULTRASTRUCTURE

used. The method, with pros and cons for its use, has been Some modifications to this method are described else-
described in detail by Kaback (68). Staphylococcus aureus where (109), as is an efficient procedure for the production
forms protoplasts during growth in media containing sub- of viable spheroplasts from E. coli (97). This procedure is
lethal doses of D-cycloserine in 1 M sucrose (147). based on Ca2+ pretreatment, glycerol stabilization, EDTA
incubation, and heat shock and utilizes much less lysozyme
7.2.1 Protoplasts from Gram-Positive Bacteria than standard procedures. Factors influencing the efficiency
A procedure for generating protoplasts from gram-positive of spheroplasting include growth medium (cells grown in a
bacteria that are sensitive to lysozyme is as follows. rich medium are more suitable than cells from defined
media), harvesting temperature (0C versus room tempera-
1. Wash the bacteria in a buffer of pH 6 to 8, and resus- ture), factors which might destroy the permeability barrier
pend (1 to 10 mg [dry weightllml) in buffer containing to sucrose, and the concentration of lysozyme.
hypertonic sucrose (usually 0.2 to 1.0 M) and lysozyme
(Sigma) at 0.1 to 1.0 mg/ml. Treatment for 30 min at 20C 7.2.3. Osmotically Sensitive Forms from Archaea
should result in complete protoplast formation. The enzyme The formation of osmotically sensitive forms in archaea is
may be used over a broad pH range, with optimum pHs of 6 less well understood, but has been achieved in selected cases
to 7, and over a wide temperature range from 4 to 60C (30, (Table 1). Spheroplasts of Halobacterium spp. may be formed
119). Lysozyme can be inhibited by the presence of salts in by resuspending the cells in 0.1 M 2-(N-morpholino)
excess of 0.2 M (132). ethanesulfonic acid (MES) buffer (pH 7.0) containing 0.5 M
Many gram-positive bacteria are not sensitive to hen egg sucrose, 0.25 M NaCl, and 0.01 M MgClz (63). Also,
white lysozyme but may be sensitive to muramidases of Methanospirillumspp. and Methanosaeta concilii form sphero-
broader substrate ranges (Table 1). These include lyso- plasts when exposed to dithiothreitol at alkaline pH (see
staphin from Staphylococcus staphylolyticus, mutanolysin section 7.1.2.6) (13).
from Smeptomyces globisporus (Sigma), and a fungal murami- Strains of Methanosarcina barkeri, which have cell walls
dase from Chuhropsis sp. (Miles Laboratories, Inc., Elkhart, composed largely of protein, form metabolically active pro-
IN). The latter enzyme, unlike egg white lysozyme, is active toplasts when incubated at 37C with pronase (2 mg/ml)
against murein acetylated at the C-6 position of muramic and 0.5 M sucrose (67). During growth, some bacteria enter
acid (52). Caution should be used when working with these a lytic phase, often as a result of nutrient depletion. In these
enzymes since, unlike egg white lysozyme, they are often cases, it may be possible to prevent lysis by incorporating su+
contaminated with proteases, lipases, or nucleases. crose directly into the growth medium. This concept may
Methods can sometimes be found to cause the murein of be used to form protoplasts during growth limitation of
insensitive strains to become sensitive to lysozyme. For exam- Methanobacterium bryantii in a medium deficient in nickel
ple, Mycobacterium s m e p t i s becomes sensitive to lysozyme and ammonium but containing 20 mM MgC12 to prevent
after prolonged exposure to DL-methionine ( 157). Protoplasts lysis (61), and to form protoplasts from Methanosarcina bark-
of Bacillus megaterium can be formed following incubation for eri cultures stabilized by 0.3 M sucrose or glucose (27).
2 h at 30C in nutrient broth containing 100 U of penicillin Finally, a pseudomurein endopeptidase has been discovered
per ml. The treated cells are washed once in 0.85% (wtlvol) in Methanobacterium wolfei and found to be effective in
-
saline and resuspended for 1 h in 0.3 M sucrose containing
500 pg of lysozyme per ml (96). Partially lysed and weakened
preparing protoplasts of Methanothermobucter themoau-
totrophicus stabilized with 0.8M sucrose (74). The useful-
cells of cyanobacteria (Oscillatoriaspp.) can be made by treat- ness of this enzyme is limited by its O2sensitivity, but it was
ment with ampicillin before lysozyme (148). the first enzyme known to hydrolyze pseudomurein. It is
Clinical isolates of Staphylococcus spp. can be made more possible to purify this enzyme aerobically and recover ac-
sensitive to lysostaphin by growth in P medium lacking glu- tivity by incubating under reducing conditions with
cose (44). Na2S-9H20or dithiothreitol (104).

7.2.2. Spheroplasts from Gram-Negative Bacteria


Virtually all gram-negative bacteria have a murein structure 7.3. CENTRIFUGATION
that is sensitive to lysozyme. However, since their outer The techniques of differential centrifugation and density
membranes are impermeable to lysozyme, their integrity gradient centrifugation are widely used in the purification
must be compromised before lysozyme will penetrate to the of components from cell lysates. Detailed discussions of
murein. This can be done in various ways (Table 1). The both the theoretical and practical aspects of preparative
basic lysozyme-EDTA procedure for forming spheroplasts centrifugation are provided in comprehensive reviews (37,
and causing the lysis of a wide variety of gram-negative bac- 115,117,140).
teria (68) is as follows. The sedimentation rate of particles subjected to a cen-
trifugal force is proportional to the force applied, and also to
1. Harvest bacteria in mid to late exponential growth
the properties of the suspension fluid, as shown below for a
phase, and wash twice with 10 mM Tris-HC1 (pH 8.0) at
spherical particle:
4C. Resuspend the cells (1 g [wet weight]/80 ml), and stir
at room temperature in 30 mM Tris-HC1 (pH 8.0)contain-
ing 20% (wt/wt) sucrose.
18n
2. Add 10 mM potassium EDTA (pH 7.0) and 0.5 mg of
lysozyme per ml (final concentrations), and incubate for 30 where w is the sedimentation rate, d is the particle diameter,
min at room temperature. Add lysozyme slowly under the ppis the particle density, pl is the liquid density, g is the cen-
surface of the liquid to decrease aggregation of the sphero- trifugal force, and n is the viscosity of the liquid. At a con-
plasts. stant centrifugal force and viscosity, the sedimentation rate
3. If lysis is required, dilute the sucrose either directly by is proportional to the size of the particle and to the differ-
dialysis or by centrifugation and resuspension of the pellet. ence between the densities of the particle and the liquid.
7. Cell Fractionation rn 115

TABLE 1 Some commonly used methods to lyse bacteria and archaea to form osmotically sensitive cells in hypotonic solutions
Method Species Reference
Lysozyme Bacillus megaterium 132
Bacillus subtilis 132
Micrococcus lysodeikticus 132
Lysozyme-EDTA-Tris (pH 8 ) Salmonella enterica serovar Typhimurium 68
Escherichia coli 68
Enterobacteria 68
Pseudomonas aeruginosa 68
Micrococcus denitrificans 68
Asotobacter vinelandii 68
Bacillus subtilis 68
Bacillus megaterium 68
Cbstridium thermouceticum 68
Aquaspirillum serpens 23
411 gram-positive and gram-negative bacteria 146
Thiobacillus strain A2 92
Wolinella succinogenes 78
Yersinia pestis 105
NaC1-sucrose-lysozyme 5 Alteromonas spp. 86
7 Vibrio spp. 86
2 Cytophuga spp. 86
4 Pseudomoms spp. 86
3 Alcaligenes spp. 86
EDTA-Tris (pH 8.6) Pseudomonas aeruginosa 40
Alcaligenes faecalis 43
Escherichia coli 43
Mutanolysin 2 1M NaCl Streptococcus mutans 129
Streptococcus sanguis 129
Streptococcus salivarius 129
Lysoamidase Staphylococcus aureus 114
Penicillin G Various gram-positive and gram-negative bacteria 68
Lysozyme-penicillin" Fusobucterium varium 21
Enterococcus faecium 21
Serine protease Methanobacteriumformicicum 20
Pronase Methunosarcina barkeri 67
Dithiothreitol (pH >8) Methanospirillurn spp. 13
Methunosueta concilii 13
Pseudomurein endopeptidase Methunothermobucter thermoautoaophicus 74
Nalidixic acid-glycine incubation,followed by freeze-thaw Escherichia coli K-12 159
Aeromonus hydrophila 159
Staphylococcus epidermidis 159
'Lysozyme susceptibility follows growth in medium containing penicillin G.

Simple differential centrifugation (section 7.3.1) is a tech- the separation of particles with very different sedimentation
nique employed to differentially pellet cell fractions. velocities and thus for the isolation of partially purified frac-
Density gradient centrifugation can be based on either the tions. For example, centrifugation for 5 to 10 min at 3,000
size difference among particles (rate-zonal centrifugation to 5,000 X g will pellet intact bacteria, leaving most cell
[section 7.3.2]), or the density difference among particles fragments (walls, membranes, and appendages) in the su-
(isopycnic separation [section 7.3.31). pernatant. Cell wall fragments and large membrane struc-
tures can be pelleted by centrifugation at 20,000 to 50,000
7.3.1. Differential Centrifugation x g for 20 min, whereas centrifugation at 200,000 x g for 1
In differential centrifugation (pelleting), samples are cen- h (in an ultracentrifuge) is required to pellet small mem-
trifuged for a given time at a given speed, resulting in a su- brane vesicles or ribosomes. For pelleting in a n ultracen-
pernatant and a pellet fraction. This technique is useful for trifuge, thick-walled tubes can be used without tube caps in
116 rn MORPHOLOGY AND ULTRASTRUCTURE

fixed-angle rotors. For ultracentrifugation of radioactive or solute density gradient (sucrose gradients are commonly
biohazardous material, polycarbonate bottles with a three. used for membranes and organelles with a density of less
piece cap assembly are available for general-purpose fixed- than 1.3 g/ml; cesium chloride is used for denser structures
angle rotors and are used frequently for differential cen- such as viruses) until an equilibrium state is reached, at
trifugation where band recovery is not a problem. which time each particle has migrated to a point in the gra-
Low-speed centrifugation may be performed anaerobi- dient where the particle has the same density as the sur-
cally by using standard anaerobic methodologies and glass rounding solution (Table 2). This point is the isopycnic po-
centrifuge tubes modified to accept serum bottle closures sition of the particle. Since the equilibrium density is an
(134). equilibrium position, it may be approached by sedimentation
from less dense regions of the gradient, from more dense re-
7.3.2. Rate-Zonal Centrifugation - gions (by flotation), or from particles dispersed throughout
Zonal centrifugation is effective for separating subcellular the gradient (Fig. 1). Since the sedimentation velocity of a
structures having similar buoyant densities but differing in particle becomes progressively lower as the particle ap-
shape or particle size, e.g., ribosomal subunits, classes of proaches the region in the gradient where it has the same
polysomes, and various forms of DNA molecules. Cen- density as the solution, very long centrifugation times are re-
trifugation is carried out either in swinging-bucket rotors or quired in order to approach equilibrium. This is particularly
specially designed zonal rotors, and a shallow linear gradient true for small membrane vesicles such as chromatophores
(usually sucrose) is employed to prevent convection in the and plasma membrane fragments from cells broken with a
tubes or the chamber of the zonal rotor during centrifuga- French press, since the sedimentation velocity of these vesi-
tion. The preparation of linear gradients is described in sec- cles is low even in the absence of the gradient. If centrifugal
tion 7.3.4. A density range is chosen such that during the forces in the order of 100,000 to 200,000 x g are used, at
separation the particle densities are always greater than the least 18 h will be required for a reasonable separation, and
density of the liquid. The sample is applied as a zone or nar- periods as long as 72 h are needed for critical separations.
row band at the top of the gradient, and the run is termi-
nated before the separating particles reach the bottom of 7.3.4. Preparation and Fractionation
the tube. Times and speeds must be determined empirically. of Density Gradients
For subcellular particles, a gradient of 15 to 40% (wtlwt) su- Density gradients may be subdivided into two main cate-
crose is commonly used, and centrifugation at 100,000 X g gories according to the method of preparation: step (dis-
for 1 to 4 h is sufficient for the separation of most subcellu- continuous) gradients and linear (continuous) gradients.
lar particles. Step gradients can be prepared by hand layering solutions
The use of conventional sucrose or glycerol gradients re- of different densities in the centrifuge tube. Sucrose gradi-
sults in an increase in the concentration of the gradient ma- ents are prepared often as a series of five to seven layers.
terial, and therefore of the liquid viscosity, as the particle When wettable tubes such as polycarbonate ones are used,
moves down the gradient. Consequently, the rate of move- each layer can be added by allowing it to run down the side
ment can decrease as the particle moves downward even of the tube from a pipette held at an angle against the side of
though the centrifugal force increases, and this decrease ul- the tube. When nonwettable tubes such as polyallomer or
timately results in less resolution between separating zones. polypropylene ones are used, the pipette tip must be kept in
When a cesium chloride gradient (section 7.3.6.2) is used, contact with the meniscus during the addition of the solu-
the viscosity decreases with increasing concentration, re- tion to avoid mixing. Alternatively, an easy way to layer so-
sulting in an accelerating gradient and better separations. lutions is to float on the bottom sucrose layer a thin disk of
cork with a diameter just less than the diameter of the tube.
7.3.3. lsopycnic Separations The solutions are added dropwise just above the center of
Isopycnic separation is often used to separate particles and the cork disk. The solutions will run over and under the cork
membrane fractions on the basis of buoyant density instead disk, without disturbing the heavier sucrose solutions under-
of sedimentation velocity. Membrane fragments derived neath. The sample is layered on top, and centrifugation is
from the same subcellular membrane may differ greatly in started immediately. Before and during centrifugation, the
size and, hence, in sedimentation velocity but should have steps in these preformed gradients are smoothed by diffusion.
the same buoyant densities. The sample is centrifuged in a Bottom- and middle-loaded samples, first having been mixed

TABLE 2 Buoyant densities of fractions isolated from prokaryotic cells


Fraction Buoyant density (g/ml) Medium Source Reference
Plasma membrane 1.14-1.16 Sucrose Gram-negative enterics 109
1.17 Sucrose Methanospirillurn hungutei 135
Outer membrane 1.22 Sucrose Gram-negative enterics 109
Protein sheath 1.35 Sucrose Methanospirillurn hungutei 135
Fimbriae (pili) 1.117-1.137 Sucrose Escherichia coli 72
1.3 CsCl Pseudomonus ueruginosa 110
Spinae 1.37 CSCl Marine pseudomonad 33
Flagella (intact) 1.30 CsCl Escherichiu coli 28
Endospores 1.27 Percoll Cbsnidium perfnngens 144
1.21 Percoll Bacillus subtilis 144
7. Cell Fractionation 117

ticles such as cells, cell organelles, and viruses. Shorter cen-


trifugation times and weaker g forces can be used, which as-
sists in the preservation of delicate structures. Smaller par-
ticles such as macromolecules and associated complexes
require much longer centrifugation times to band, during
which a gradient can be self-generated.
Various methods can be used for fractionating sucrose
gradients. The gradients may be pumped out through a
stainless steel cannula (e.g., a blunted syringe tip) inserted
into the bottom of the tube by using a peristaltic pump.
Alternatively, the bottom of the tube, or the side just below
the band of interest, may be pierced with a needle and sam-
ples may be collected by gravity flow. Although simple to
perform, these methods in which samples are collected
starting from the bottom of the gradient can lead to cross
contamination of upper bands with those appearing lower
in the tube. In cases where the component of interest is the
upper band, the gradient should be fractionated by upward
displacement. The centrifuge tube is fitted with a tight-
fitting rubber bung, pierced by a stainless steel cannula suf-
ficiently long to extend to the bottom of the tube. A sucrose
solution about 5% greater in density than the bottom of the
gradient is pumped through the cannula, thus displacing the
gradient through a short exit tube leading to a fraction col-
lector. Alternatively, if bands are clearly visible, samples
may be taken carefully from the top of the gradient by
pipette.
FIGURE 1 Isolation of cytoplasmic membranes by discon-
tinuous sucrose gradient centrifugation. A membrane fraction 7.3.5. Rotor, Tube, and Bottle Selection
from Methanospirillurn hungutei spheroplast lysate in 33% su- There are basically four types of rotors used for centrifugal
crose was loaded on 40 to 50% sucrose steps and centrifuged separations: (i) swinging bucket (swing out), (ii) fixed
at 4C for 48 h (110,000 X g, RmaJ in a Beckman SW27 angle, (iii) vertical tube, and (iv) ional. It is important to
swinging-bucket rotor. A thin section of the upper band re- choose the correct rotor in order to achieve optimum sepa-
vealed closed vesicles bounded by the double-track membrane, ration results. This topic is discussed in references 37 and
typical of cytoplasmic membranes. See reference 135 for fur- 117. The Beckman Coulter Internet site (http://www
ther details. .beckmancoulter.com) is also an excellent source of infor-
mation about the practical aspects of ultracentrifugation.
Swinging-bucket rotors are recommended for most
with gradient medium to make the sample solution denser preparative rate-zonal separation methods because of their
than the remainder of the gradient, can float upward to find relatively long path length. Vertical-tube rotors are also use-
their own buoyant density. ful for rate-zonal separations, especially for large particles
Linear gradients of sucrose can be formed by allowing a such as cells and subcellular organelles. The main advan-
step gradient to age in the centrifuge tube overnight at 4 tage of vertical-tube rotors is the short time needed to
to 8C. A continuous gradient will form by diffusion. A va- achieve separation compared with that for other rotor types.
riety of commercial gradient-forming devices are available, Fixed-angle rotors are not normally used for rate-zonal sep-
but these are unnecessary if the molecular mass of the gra- aration. Beckman Coulter has published an excellent cen-
dient medium is less than 1,000 Da. Diffusion of higher- trifuge product selection guide (BR-8101D) that includes a
molecular-mass molecules such as Ficoll 400 is too slow, table for discontinued rotors and their recommended re-
requiring a gradient maker for the preparation of linear gra- placements. This guide may be useful when one is following
dients. Linear gradients are called preformed gradients for a centrifugation procedure from the older literature. It is ex-
the obvious reason that they are formed prior to centrifuga- tremely dangerous to use older rotors that are not compati-
tion. Continuous-density gradients may also be formed by ble with the newer models of ultracentrifuges or to exceed
centrifuging a solution, which initially is of the same den- the maximum speed recommended for a particular rotor. Be
sity throughout the centrifuge tube, for sufficient time and aware that certain rotors may be derated (based on the total
at sufficient speed. During centrifugation, a gradient is number of run hours) but still useable.
formed as the density decreases at the top of the tube and For isopycnic separations, sample particles will cease sed-
increases toward the bottom. This type of gradient is known imenting when they reach their buoyant density and wall
as a self-forming gradient but cannot be formed with all effects are much less important than they are for rate-zonal
types of gradient media. It is very useful for cesium chloride separation. Swingingbucket, fixed-angle, and vertical-tube
gradients. rotors may all be used for isopycnic separations; however,
Unless one is following a specific protocol, a choice will the density range and slope of an equilibrium gradient will
have to be made on the gradient type for a particular sepa- depend upon the type of rotor used for isopycnic separation.
ration. In the simplest terms, preformed gradients are used Fixed-angle and vertical-tube rotors have some advantages
when the particles to be purified are large enough to reach over swinging-bucket rotors for these separations. Because
their isopycnic positions in less time than it takes a gradient the effective path length is shorter for these rotors than for
to self-form. This is very important in the case of large par- swinging-bucket rotors, the time period of centrifugation is
118 MORPHOLOGY AND ULTRASTRUCTURE

considerably shorter for the equilibrium banding of parti- 7.3.6. Gradient Media
cles. The increased number of tubes in fixed-angle and
vertical-tube rotors means that the sample capacity per 7.3.6.1. Sucrose and Ficoll
rotor is much greater. Sucrose concentrations in gradients are reported in the lit-
A wide variety of materials have been used for the man- erature in several ways: density, percent sucrose by weight
ufacture of centrifuge tubes and bottles, each with its own (wtlwt), percent sucrose per unit of volume (wt/vol, or
distinct combination of properties to meet a variety of ap- grams/lOO ml), or molar concentration of sucrose. In dupli-
plications. It is important to choose a material that is com- cating experiments reported in the literature, note how the
patible with the sample and the gradient medium. Available gradient solutions were mixed: wt/wt or wtlvol. The most
options include transparent, translucent, and opaque tubes; useful unit is percent sucrose by weight. Standard chemistry
tubes that can be sliced, punctured, or sterilized and reused; handbooks (e.g., Handbook of Biochemistry and Molecular
and tubes resistant to a variety of chemical compounds. Biology, [8]) contain tables defining both densities and vis-
This selection can be overwhelming to the novice user. cosities of various concentrations of sucrose (percent) in
Thankfully, selection considerations are discussed in detail water at different temperatures. For each density of sucrose,
in the Beckman Coulter centrifuge product selection guide the percent wt/wt and wt/vol is given so conversions can be
(BR-8101D). The tube size may vary somewhat from the ac- made. Sucrose gradients can be run at different tempera-
tual filling capacity. Centrifuge tubes should be filled (to tures (e.g., 4, 10, or 20C), but remember that the density
within -3 mm of the tube rim) to prevent collapse of the of the sucrose will vary according to temperature. This vari-
tubes during centrifugation. A n exception is thick-walled ation may affect band position.
tubes that can be used uncapped and filled to half the max- When preparing sucrose solutions, one may assume that
imum volume. The maximum filling volume of thick- water or dilute buffers have a density of 1 g/ml. Therefore,
walled tubes will depend on the tube angle of the rotor to to prepare a 54% (wt/wt) sucrose solution, for example, dis-
be used. It is important to dry the outside of the tube before solve 54 g of sucrose in 46 ml of water. This will give 100 g
inserting it into the rotor or bucket. Any residual moisture of solution, and since the density of a 54% (wtlwt) sucrose
on the outside of the tube can act as a lubricant under cen- solution is 1.2451 g/ml, the final volume will be 80.3 ml.
trifugal force and contribute to the collapse of a tube, par- Preparing solutions in this manner avoids the necessity of
ticularly with open-top tubes. having to make viscous solutions up to fixed volumes.
To use a tube closure or not: that is the question. With- Sucrose allows for a density range of up to 1.35 g/ml. It is
out a doubt, the most convenient tube closure is none at all. important to check the maximum density of the gradient
This applies to all tubes run in swingingbucket rotors. material for each rotor. Since sucrose solutions are relatively
Thick-walled polyallomer tubes can be run in all fixed- viscous, linear gradients are generally employed.
angle rotors without caps, provided that they are partially Ficoll 400 (Amersham Pharmacia Biotech, Inc.,
filled. Polycarbonate tubes are used like thick-walled Piscataway, NJ) is a hydrophilic polymer of sucrose with a
polyallomer tubes, with or without caps, in fixed-angle ro- molecular weight of 400,000. Solutions of up to 50%
tors. Ultraclear, polypropylene, and polyethylene tubes can (wt/vol) Ficoll can be made, which allows a density range of
also be used without caps in fixed-angle rotors. up to 1.2 g/ml. Its advantages over sucrose for density gradi-
Tubes need to be capped for one of four reasons: (i) to ent centrifugation lie in its lower osmotic pressure, which
prevent the collapse of thin-walled tubes during centrifuga- can be important in preserving the morphologies and activ-
tion, (ii) to contain radioactive or biohazardous samples, ities of subcellular fractions.
(iii) to maintain anaerobic conditions for oxygen-labile
components, and (iv) to maintain sterility. Thin-walled 7.3.6.2. Cesium Chloride and Potassium Bromide
polyallomer tubes must be capped when used in fixed-angle Cesium chloride solutions have low viscosity so that pre-
rotors. For swinging-bucket rotors, these thin-walled tubes formed gradients of this solute are difficult to prepare and
should be filled to within 2 to 3 mm of the top, as tube caps self-forming gradients are used. The sample is mixed with
are not used in swinging-bucket rotors. The standard tube enough CsCl to provide a density equal to the average den-
cap assembly (with an 0 ring sealing system) is definitely sity of the subcellular particles. This homogeneous suspen-
one of the more tedious aspects of ultracentrifugation. sion is placed in the centrifuge, and the gradient is formed
Fortunately, Beckman Coulter has introduced alternatives. during centrifugation as a result of the sedimentation of the
Polycarbonate bottles with three-piece cap assemblies are CsCl in the centrifugal field. CsCl allows a density range of
available for fixed-angle rotors. These are frequently used for up to 1.81 g/ml. KBr can also be used and is less expensive
differential Centrifugation where the bottle does not need to than CsCl(69).
be punctured for band recovery. Bottle assemblies can be
filled to half the maximum volume. OptiSealTMpolyallomer 7.3.6.3. Percoll
tubes have the simplest closure; simply insert a tube plug and Percoll is a density gradient medium containing colloidal
press, and an 0 rin seals securely against the tube's inner silica particles that are rendered nontoxic by a coating of
surface. Quick-Seal8 tubes (polyallomer or ultraclear) have polyvinyl-pyrrolidine (1 12). Percoll is recommended for
a 3-mm-long inlet through which the tube is filled. The top separation of cells because it has low osmolarity, low viscos-
of the tube is then heat sealed by using a handheld or table- ity, and large particles (30 nm in diameter) so that it does
top tube topper. Quick-Seal@tubes can be used in all swing- not permeate bacterial walls or membranes. It is used also to
ing-bucket and most fixed-angle rotors and must be used in separate viruses and subcellular organelles. I t can be pur-
vertical rotors. Tube size must be compatible with the rotor chased from Amersham Pharmacia Biotech as a sterile sus-
and the sample volume. Many Beckman rotors can accom- pension which may be diluted as desired and used in gradi-
modate smaller tubes by using adapters or spacers. The ents that are either preformed or self-formed during the
Beckman g-Max system uses a combination of short poly- centrifugation run to produce a density range of up to
allomer Quick-Seal@ tubes and floating spacers to permit 1.3 g/ml. Since Percoll cannot penetrate walls or mem-
faster run times at maximum g forces. branes, intact cells band at the buoyant densities of the in-
7 . Cell Fractionation m 119

tact cell. In contrast, gradient media with smaller particles sheared by forcing of the cells through a 22-gauge needle
(e.g., Metrizamide) permeate coatings above the membrane and subsequent homogenization of cells in a Potter-
and thus indicate the buoyant densities of the protoplasts, Elvehjem Teflon glass tissue grinder.
as exemplified by lysozyme-sensitive spores (91). Density 3. Sediment deflagellated cells by differential centrifuga-
marker beads of differing densities and colors can be pur- tion (6 to 10,000 X gfor 15 min).
chased (Amersham Pharmacia Biotech) for calibration. 4. Centrifuge the cell-free supernatant at 100,000 X g in
The use of marker beads is important for checking the shape an ultracentrifuge for 90 min to pellet the filaments.
of the gradient, as the rotor angle and the tube size affect 5. The sheared filaments can be further purified by iso-
the geometry (shape) of the gradient formed. pycnic density gradient centrifugation using KBr (69).
Resuspend the pellet of filaments from step 4 into 10 ml of
7.3.6.4. Metrizamide buffer overnight at 4C. Add 5 g of KBr (Sigma; ca. 99%),
Metrizamide, or 2(3-acetamido-5-N-methylacetamido- and dissolve the KBr with gentle stirring. Centrifuge at
2,4,6-triiodobenzamido)-2-deoxy-~-glucose (Fluka, Buchs, 210,000 x gfor 24 h a t 5C (Beckman SW41Ti or SW50.1
Switzerland), is a density gradient medium which has a mo- rotor).
lecular weight of 789, a density of 2.17 g/ml, and very low A single diffuse band of filaments usually occurs in the
viscosity. The last property is particularly useful in applica- bottom third of the centrifuge tube. Remove the band from
tions in which it is desirable to isolate membrane fractions the top with a Pasteur pipette. KBr can be removed by dial-
by flotation to assess the association of the membrane with ysis against distilled water or buffer, and the filaments can be
mutant proteins or products of genes cloned into multicopy collected by ultracentrifugation at 100,000 X g for 90 min.
vectors. Spheroplast lysates of E. coli are brought to a den-
sity of 1.29 g/ml with Metrizamide and placed into a cen- Procedure for Isolation of intact Flagella
trifuge tube. The sample is layered over with a 1.27-g/ml The DePamphilis and Adler (28) method can be used suc-
Metrizamide solution. Rapid flotation of membranes occurs cessfully with E. coli and Bacillus subtilis. Refer to the origi-
while the gradient is formed during centrifugation, leaving nal paper for details of the procedure, which may be of use
cytoplasmic proteins near the bottom of the tube. The short for work with other bacteria. In all steps, care must be taken
sedimentation distance and high force obtainable in a not to break off the basal bodies from the filaments.
Beckman TLlOO benchtop ultracentrifuge can be used to
advantage (141). 1. Grow 1 to 3 liters of bacteria, and harvest cells by cen-
trifugation.
2. Gently resuspend cells in 50 ml of 20% (wt/wt) su-
7.4. ISOLATION OF CELL COMPONENTS crose (final volume, -70 ml).
3. To form protoplasts or spheroplasts, add in the follow-
7.4.1. Appendages ing order 7 ml of 1 M Tris-HC1, (pH 7.8), 2 ml of 0.25%
(wtlvol) lysozyme in 0.1 M Tris-HC1 (pH 7.8) containing
7.4.1 .l. Flagella
.., 0.2 M NaCl, and 6 ml of 0.1 M EDTA in 0.1 M Tris-HC1
Intact flagella of most prokaryotes consist of three major (pH 7.8). Mix the suspension after the addition of each
components: filament, hook, and basal body (chapter 4, reagent, and add the EDTA within 30 s after the lysozyme.
Fig. 8). The first two components are external to the cell Incubate at 30C for 1.5 h with gentle shaking. A good
wall and are relatively easy to isolate from other cell com- yield of intact flagella depends on the formation of at least
ponents. Flagellar filaments can be removed mechanically 80% protoplasts or spheroplasts.
from cells by shear forces in a blender or Potter-Elvehjem 4. Add 7 ml of 20% (vol/vol) Triton X-100. This addi-
homogenizer which removes filaments without damaging tion should produce a clear, viscous lysate.
the cell walls of most organisms. Sheared flagella from some 5. To free the flagellum preparation of DNA, add 0.8 ml
bacteria do retain the hook; however, experience has shown of 1 M MgClz followed by 1.5 mg of DNase I. Incubate the
that this retention is rare. When flagella are released spon- mixture at 30C for 20 min. Do not add Mg2+ before the
taneously from autolyzed cells, the hook often remains at- Triton X-100, since this prevents solubilization of the outer
tached to the filament. Often, filaments can be sheared membrane of gram-negative bacteria.
from cells, the cells can be placed in fresh medium, and re- 6. Dilute the lysate immediately to 260 ml with cold 0.1
growth of the filament can be monitored. M Tris-HC1 (pH 7.8) containing 0.5 mM EDTA (Tris-
Note: check the culture supernatant by electron mi- EDTA buffer). Rapidly pour 87 ml of cold-saturated
croscopy (negative stains, chapter 4.2.2 and chapter 4, (NH&S04 (in Tris-EDTA buffer) into the diluted lysate to
Fig. 8) after harvesting for any filaments that have been re- give 25% of saturation. Stir the suspension slowly at 5C
moved during either growth or centrifugation. Often con- for 2 h.
centration and ultracentrifugation of the culture super- 7. Centrifuge the suspension at 12,000 X g for 25 min in
natant will produce a good yield. a fixed-angle rotor. Collect the viscous white material float-
ing at the meniscus and adhering to the side of the tube.
Procedure for Isolation of Flagellar Filaments Discard the fluid.
1. Grow 1 to 3 liters of bacteria. Harvest cells by cen- 8. Rinse the tube with Tris-EDTA buffer, and combine
trifugation, and resuspend cells in approximately 10% of the with recovered material to give a final volume of 40 ml.
culture volume in 0.5 M HEPES or Tris buffer, pH 7.5. The 9. Add 1 ml of 20% (vol/vol) Triton X-100 and dialyze
density of the cell suspension is important for the removal the suspension at 5C against 1 liter of Tris-EDTA buffer.
of filaments. The initially turbid suspension should clear completely
2. Homogenize cells in a rotating-blade blender (Sorvall upon dialysis.
Omni-Mixer; Waring) at maximum speed for about 60 s. 10. Dilute the dialyzed preparation to 50 ml with Tris-
The volume of the cell suspension must at least be enough EDTA buffer. To separate the intact flagella from other cel-
to cover the blades. For some bacteria, flagella can also be lular debris, layer in duplicate tubes 25 ml of the diluted
Next Page

120 MORPHOLOGY AND ULTRASTRUCTURE

preparation onto a gradient consisting of 2 ml of 20% found on flagellated cells, in which case isolation and pu-
(wt/wt) sucrose and 3 ml of 60% (wt/wt) sucrose (in Tris- rification will involve separation of these two kinds of ap-
EDTA). After centrifugation for 1 h a 40,000 X g in a pendages. They are very stable protein assemblies and are
swinging-bucket rotor (Beckman SW28), remove and dis- firmly attached, making them more resistant to shear forces
card the liquid above the sucrose layers. than flagella.
11. Add 5 ml of Tris-EDTA buffer to the sucrose layers.
Mix gently. Remove sucrose by dialysis overnight against Procedure for Removal of Fimbriae from Cells
Tris-EDTA buffer. Dilute the suspension to 25 ml with 1. Grow 1 liter of cells in appropriate medium and har-
buffer, and add 0.15 ml of 20% (vol/vol) Triton X-100. vest by centrifugation. Resuspend the cells in 50 ml of 10
Centrifuge at 4,000 x g for 10 min to remove unlysed cells mM Tris-HC1 (pH 7.2).
and large aggregates. Retain the supernatant. 2. Remove fimbriae by blending for 2 min at maximum
12. To further purify the intact flagella, use isopycnic speed in a rotatingblade blender (e.g., Sorvall Omni-
density gradient centrifugation in CsC1. Dilute the super- Mixer; Waring). See comments in section 7.4.1.1.
natant material from step 11 to 27 ml with Tris-EDTA 3. Remove cells by differential centrifugation (6,000 to
buffer. Add 12.1 g of CsCl (all at once), and dissolve rap- 10,000 x g for 15 min), and retain the supernatant fluid
idly. Centrifuge at 50,000 X g for 50 h (Beckman SW28 ("shear supernatant") which contains the fimbriae.
rotor). A band of flagella should be present near the center Procedures for Purification of Fimbriae
of the gradient. Collect this band, and remove the CsCl by
dialysis against Tris-EDTA buffer. The appearance and Procedure 1
number of bands in CsCl gradients and the purity and qual- Fimbriae can be separated from other contaminating
ity of the intact flagella recovered are markedly influenced materials in the shear supernatant by differential centrifu-
by the rate of addition of CsCl and the age of the cells at gation. A detailed description of the purification of gono-
harvesting. coccal fimbriae is given in reference 53, whereas a method
for the common fimbriae of nonflagellated E. coli is by pre-
Isolation of Intact Flagella from Archaea cipitation in 0.1 M MgC12 (final concentration) (19). The
Kalmokoff et al. (69) devised a procedure appropriate to fimbriae are collected by centrifugation, resuspension in
Methanococcus voltae, whose cell envelope consists of only buffer, and reprecipitation twice with 0.1 M MgC12.
the plasma membrane and a protein surface array. This pro-
cedure is based on the temperature-inducedphase separation Procedure 2
of the nonionic detergent Triton X-114. At temperatures The shear supernatant (containing 0.5 M NaC1) of
above 20'32, Triton X-114 separates into two phases: hy- sheared Pseudomom aeruginosa contains flagella and fim-
drophilic proteins separate into the upper aqueous phase, briae, which are both precipitated by the addition of poly-
and hydrophobic integral membrane proteins separate into ethylene glycol 6000 (110). After 18 h at 4"C, collect the
the lower detergent-rich phase. Since the envelopes of ar- precipitate by centrifugation and resuspend the pellet in
chaea contain only a few hydrophilic proteins, this tech- 10% (wt/wt) (NH4)2S04(pH 4). After 2 h at 4"C, the fim-
nique allows a selective enrichment of the wall protein and briae form a precipitate while the flagella remain in suspen-
flagella. For osmotically fragile methanogens, cells can be sion. Remove the flagella by repeating the (NH4)2S04pre-
lysed by dilution in distilled water. Crude flagellar prepara- cipitation step. Dissolve the final pellet in water, dialyze to
tions are purified as described in reference 69. The proce- remove the (NH4)2S04,and subject the suspension to CsCl
dure is not suitable for some archaea whose flagellar fila- density gradient centrifugation.
ments are sensitive to Triton (142). Procedure 3
Procedure The flagella and fimbriae of E. coli precipitate upon the
1. Prepare spheroplasts (see section 7.2) from 1 liter of addition of (NH4)2S04 to give 20% saturation (72).
medium, if possible, and lyse them in distilled water with Resuspend the pellet in buffer, and layer on top of a linear
gentle mixing. Add DNase (1.5 mg) with 1 mM MgCl2 to sucrose gradient (15 to 50% [wt/wt] sucrose). Remove con-
reduce viscosity. taminating flagella by incubating the fimbriae for 1 h at
2. Centrifuge the lysate at 22,000 X g for 30 min at 4"C, 37C in 0.4% (wtlvol) SDS. Separate the dissociated fla-
resuspend envelopes in 20 ml of cold 10 mM Tris-HC1 (pH gella from the intact fimbriae by gel filtration on Sepharose
7.5), and treat with 1% (vol/vol, final concentration) CL-4B by using a buffer containing 0.4% SDS and 0.1%
Triton X-114 for 30 min at 4C with occasional mixing. (wt/vol) EDTA. Only fimbriae elute in the void volume.
3. Incubate samples at 37C to induce phase separation. When appropriate, isolation is assisted by taking advan-
Centrifuge at 300 X g for 3 min. Do not centrifuge at higher tage of the buoyant density of fimbriae versus the densities
speeds, since the flagella will be pelleted in the lower deter- of other structures (Table 2).
gent phase.
4. Remove the upper aqueous phase (-17 ml), and con- 7.4.1.3. Spinae
centrate to 1 ml with an ultrafiltration apparatus (e.g., Spinae are nonprosthecate, rigid, helical protein structures
PM30 filter, model 8010; Amicon Corporation). Check the that extend outward from the cell surface of some bacteria
sample by electron microscopy for the presence of flagella; (chapter 4, Fig. 3, and reference 32); they may coexist with
bacterial flagella are -20 nm in diameter, and methanogen flagella and require appropriate culture conditions for ex-
-
flagella are 11 nm in diameter. pression (34). They are readily detached from the cell sur-
face by shear force, but the base may remain on the cell.
7.4.1.2. Fimbriae (Pili) Spinae are not efficiently detached from the cell by homog-
Fimbriae are proteinaceous, filamentous surface structures enization in a Potter-Elvehjem homogenizer. Their large
composed mostly of identical subunits (chapter 4, Fig. 1). size and characteristic density (Table 2) allow purification
They are narrower in diameter (< 10 nm; see chapter 4, Fig. by differential and density gradient centrifugation. Here
5) than flagella and have no basal structure. They are often is a procedure for the isolation of spinae from a marine
Antigen-Antibody Reactions
LUCY M . MUTHARIA AND JOSEPH S. LAM

8.1. HANDLING ANIMALS ..................................... 139


.................................. 139
8.1.1. Animal Care Guidelines
. 2. Animals............................................. 139
. ..........................................
3. Age and Size 140
........................................ 140
8.1.3.1. Rabbits
. ......................................... 140
2.Mice
. .......................................... 140
3.Rats
. ...................................... 140
4. Chickens
............................... 140
8.1.4. Injection Route and Dosage
. .................................. 140
5. Immunization Protocols
. ....................................... 141
6. Blood Collection
........................................ 141
8.1.6.1. Rabbits
. .........................................
2.Mice 141
. .......................................... 141
3.Rats
...................................... 141
8.1.7. Serum Preparation
8.2. ANTIGEN PREPARATION .................................. 142
........................................ 142
8.2.1. Particulate Ags
..................................
8.2.1.1. Bacterial Cells 142
. ....................... 142
2. Sheep Erythrocytes as Carriers
. ..........................
3. Bacterial Cells as Carriers 142
........................................... 143
8.2.2. Soluble Ags
.................................... 143
8.2.2.1. Purification
. ................................... 143
2. Electroelution
8.2.3. Haptens ............................................. 143
.
.4 Adjuvants............................................ 144
................................ 144
8.2.4.1. Freunds Adjuvant
. ......................................... 144
2.Alum
....................................... 144
.3.ISCOMs
. ..................................... 144
4. Liposomes
. ............................ 144
5. Ribi Adjuvant Systems
. ..............................
6. DNA Immunization 146
8.3. CONVENTIONAL ANTIBODY PREPARATION .................. 146
.............. 146
8.3.1. Absorption of Cross-Reactive and Nonspecific Abs
. ........................... 147
2. Ammonium Sulfate Precipitation
. 3. Ion Exchange Chromatography............................. 147
. ......................................... 148
4. Gel Filtration
. ................................. 148
5. Affinity Chromatography
................................... 148
8.3.5.1. Affi-Gel Blue
. ............................... 148
2. Protein A-Agarose
. ...................................... 148
3. Protein G
8.4. MAb PREPARATION ....................................... 149
..................................... 149
8.4.1. Production Protocol
. .........................................
2. Isotyping Abs 149
. ................................. 149
3. Ascites Fluid Production
. ........................................ 150
4. Ab Purification
...................................... 151
8.4.4.1. Protein A
. .............................. 151
2. DEAE Affi-Gel Blue
8.5. DETECTION OF REACTIONS ................................ 152
8.5.1. Agglutination......................................... 152
...................... 152
8.5.1.1. Qualitative Slide Agglutination
. .......................
2. Microtiter Plate Agglutination
. ............................. 152
3. Passive Agglutination 153
...................................... 153
8.5.2. Quellung Reaction
. .......................................... 153
3. Precipitation
.... 153
8.5.3.1. Radial Single and Ouchterlony Double Immunodiffusion
. ................ 154
2. Rocket Immunoelectrophoresisand CIE
138
8. Antigen-Antibody Reactions 139

.3. XIE ......................................... 155


8.5.4. ELISA .............................................. 156
8.5.4.1. Requirements .................................. 156
.2. System Classification............................. 157
.3. Procedure ..................................... 158
-4. Chemiluminescence ELISA ........................ 159
8.5.5. Immunoblotting of Ags .................................. 159
8.5.5.1. General Procedure ............................... 160
.2. Detection of Ags ................................ 160
.3. Dot Blotting ................................... 161
.4. Detection of Glycoproteins ........................ 161
8.5.6. Immunofluorescence Techniques ........................... 162
8.6. COMMERCIAL SOURCES AND THEIR URLS ...................163
.7. REFERENCES ............................................. 163
8.7.1. General References ..................................... 163
.2. Specific References ..................................... 164
8.7.2.1. Handling Animals ............................... 164
.2. Ag Preparation ................................. 164
.3. Adjuvants .................................... 164
.4. Antibody Preparation ............................ 165
.5. Agglutination .................................. 165
.6. Quellung Reaction
.7. Immunodiffusion
.............................. 166
............................... 166
.8. ELISA ....................................... 166
.9. Immunoblotting ................................ 167
.lo. Immunofluorescence ............................. 167

Reactions between antigens (Ags) and antibodies (Abs) are Use of Laboratq Animals, the Animal Welfare Acts, and
usefully exploited in many areas of life science research (1- other applicable federal regulations and policies. In most
4). Antibodies have many virtues as biological reagents, in- cases, the use of animals for immunization and the produc-
cluding specificity for an Ag, availability, and the usually tion of Abs can be classified in the acute-care and low-pain
visible secondary reactions due to the divalent nature of the categories. However, it is necessary for the researchers to be
Ab. The monoclonal Ab (MAb) technology developed by aware of the established guidelines. In brief, all animals
Kohler and Milstein (66) allows for the production of un- should be cared for, transported, and handled in accordance
limited quantities of Abs against virtually any molecule. A with the Animal Welfare Act or its equivalent. The proce-
MAb is known for its unique epitope specificity and repro- dures used should be designed and performed with due con-
ducibility in Ag-Ab reactions. Although MAbs have obvi- sideration of their relevance to human and animal health;
ous advantages over conventional polyclonal Abs, one the humane use of animals is imperative, and where neces-
should also consider the disadvantages of MAbs, i.e., high sary the minimum number of animals should be considered.
production costs and the requirement for highly trained Anesthesia is to be used to minimize distress and pain, and
personnel to produce and maintain the MAbs. In most animals that are suffering chronic pain or distress that can-
cases, the generation of polyclonal Abs requires nothing not be relieved should be painlessly euthanized. Appropriate
more than an immunogen, a rabbit, and a syringe. The re- living conditions should be provided, and investigators or
sulting polyclonal antisera will be adequate for most needs personnel handling the animals must be trained and quali-
in microbiology and at a fraction of the cost of MAb pro- fied. A copy of the Guide for the Care and Use of Laboratury
duction. As there is an immense volume of information Animals can be obtained from the Animal Care Committee
concerning all aspects of Ag-Ab reactions in the literature, of your institution or directly from the Animal Welfare
the objective of this chapter is to provide readers with sim- Division of the National Institutes of Health in the United
ple and useful protocols and an introduction to some of the States.
more novel techniques.
8.1.2. Animals
Rabbits, mice, and rats are the laboratory animals most
8.1. HANDLING ANIMALS commonly used for Ab production. The preference for any
The success of Ab production depends on many factors, in- particular species depends on the volume of serum required
cluding Ag quality, dose, and route of immunization and an- and can be influenced by the specificity of the generated
imal host and health. With few exceptions, animals are used serum. In general, rabbits are the animals of choice for poly-
to generate Abs; therefore, the knowledge of animal han- clonal Ab production due to their size and, more impor-
dling, use, and welfare is of primary concern for anyone who tantly, their gentle natures and the ease of handling. Also,
needs to produce Abs. anti-rabbit immunoglobulin Abs (secondary Abs) conju-
gated to either enzymes, biotin, fluorescence reagents
8.1 .l.Animal Care Guidelines (chapters 2 and 3), or electron-dense colloidal gold particles
Institutional animal facilities and programs should be oper- (chapter 4) are commercially available. For MAb produc-
ated in accordance with the governmental requirements tion, mice or rats are usually chosen because most of the
and recommendations as outlined in Guide for the Cure and myeloma cell lines available are of murine origin. Taking
140 w MORPHOLOGY AND ULTRASTRUCTURE

into consideration the points discussed in section 8.1.1, n o able to immunize more than one animal with the same Ag,
individual should handle animals without some form of and if they provide adequate titers of Ab you can pool the
training. The details on how to handle each type of animal sera collected. For rabbits, use a minimum of two animals;
should be learned from experienced and qualified person- for mice, use at least five; and for rats, use a minimum of
nel. For detailed descriptions of the proper handling of ani- three. Note that antisera should not be pooled when the ob-
mals, one can consult several review chapters on the subject jective of the study is to evaluate the responses of different
(6,8-10). animals to an Ag or to monitor the Ab response during the
course of immunization.
8.1.3. Age and Size
8.1.3.4. Chickens
8.1.3.1. Rabbits Hen egg yolk has been successfully used to generate Abs
Many breeds of rabbits, weighing from 2 to 7 kg, are readily ( 5 ) . Each egg may contain in excess of 100 mg of egg yolk
available from approved commercial sources. When large immunoglobulin Y (IgY), and the Ab is easily purified by
Ab quantities are required, larger animals should be consid- precipitation from the water-soluble fraction of the egg
ered. Lop-eared rabbits, which have a body weight of up to yolk. Because such large amounts of Abs can be obtained,
7 kg and large ears and veins, allow for easy bleeding. egg yolk Abs have been used for passive immunization stud-
However, the most frequently used type of rabbit is the New ies ( 5 , 7).
Zealand White, adults of which weigh 5 to 6 kg and have
the advantage of white skin and easily visible veins. 8.1.4. Injection Route and Dosage
Generally, 8- to 10-week-old animals, ca. 2 kg, are pur- Many factors determine the immunogenicity of an Ag. The
chased, and by the time they have been immunized and are dose and injection route significantly influence the out-
ready to be bled, they have been kept for another 4 to 6 come. The routes of injection commonly used on the three
weeks. It is worth noting that female rabbits are usually types of animals described above include intradermal (i.d.),
more tame and easier to handle than male animals. It is not subcutaneous (s.c.), intramuscular (i.m.), intraperitoneal
uncommon for rabbits to be kept for more than 6 months (i.p.), and intravenous (i.v.). Other routes of immunization
for continuous immunization and bleeding. However, unless such as ingestion, inhalation, and skin application are also
rabbits are housed in open pens, after 6 to 8 months they possible but are not as common. The i.d., s.c., and i.m. routes
may have outgrown the standard-sized cages used by most are normally chosen due to slow Ag release, which can en-
institutions. Long-term care can be expensive. As an alter- hance a humoral response. i.p. injection allows an Ag to
native, several smaller breeds of rabbits such as the Dutch come into contact with the lymphatics sooner than the pre-
Belted or the San Juan strains can be used and sera can be viously described methods and is easy in rodent-mice are
pooled to obtain a larger volume. The latter choice will often immunized by this method to prime their spleen cells
guarantee that the standard-sized cages will be adequate to for MAb production. However, this route is not recom-
meet long-term animal-holding requirements. mended for rabbits due to their large size. i.v. injection is
often used for administering a booster dose for rapid response
8.1.3.2. Mice to an Ag, for instance, when wishing to produce agglutinat-
Many laboratory strains of mice are available from commer- ing Abs to microbial surface Ags. When this route is chosen,
cial suppliers, and some institutions may even breed the the researcher should be cautious to exclude air bubbles or
more common strains such as the Swiss White, which is adjuvants from the injecting material. The practice of in-
often used for microbiological work. Because of their small jecting into the footpads of rabbits to get a slow Ag release
size, one cannot expect to obtain more than 0.2 to 0.3 ml of should not be used since the footpad is a sensitive site and
blood unless by cardiac puncture. BALB/c mice are the injection causes severe pain and discomfort. For all injec-
source of myeloma cell lines, and for obvious reasons, these tions, the materials should be suspended in isotonic saline to
mice are used for immunization to obtain primed spleno- avoid causing a burning sensation in the animal. Very low
cytes for plasmacytoma fusion experiments for MAb pro- doses of Ag may cause either nonresponsiveness or toler-
duction. Animals that are 5 to 8 weeks postweaning, weigh- ance, while very high doses can also cause tolerance. In gen-
ing -20 g, are usually purchased. After being housed for the eral, an Ag dose of 50 to 500 pg can be administered to elicit
duration of the immunization procedures, they are large a good response. Table 1 summarizes the maximum volumes
enough to yield sufficient amounts of splenocytes. that can be injected into animals via the various routes.
8.1.3.3. Rats 8.1.5. Immunization Protocols
Commonly available strains of rats are the Hooded Lister Circulating Abs to a specific Ag do not appear in significant
and the two albinos, Wistar and the Sprague-Dawley. Rats amounts until at least 7 days after immunization. Most of
are stronger and more agile than mice; they need to be han-
dled with caution. Do not h andl them without proper training.
e
Animal size depends on how long one is intending to keep
them. Animals that are 5 to 8 weeks postweaning can weigh TABLE 1 Volumes of A g A b mixtures for injection
between 75 and 150 g. They are slightly small, but after they
have been kept for the duration of the immunization proce- Maximum vol (ml) for route of injection:
dures and are ready to be bled, they should have grown to Animal i.d. S.C. i.m. i.p. i.v.a
sufficient size to give a good yield of blood.
A frequently asked question is how many animals are Rabbit 0.1 0.4 2.0 5.0 1.0
needed for the generation of sufficient Abs. If the amount Mouse 0.1 0.1 0.2 1.o 0.2
of Ag is not limiting, then more than one animal should be Rat 0.1 0.2 0.5 2.5 0.5
used because even genetically identical animals will re-
spond to the same Ag differently ( 3 ) . Therefore, it is advis- 'Freund's adjuvants should not be used with i.v. injections
8. Antigen-Antibody Reactions w 141

the Abs in an early (or primary) response belong to the IgM tip of a fresh scalpel blade will open the vein up. The
class, while Abs from a secondary response are mostly IgG. droplets of blood are collected in a suitable container, usu-
However, if the Ag is pure carbohydrate, the secondary- ally a screw-capped glass bottle. With sufficient practice, 10
response Abs will be mainly IgM. The amount of Ab formed to 20 ml of blood can easily be obtained. If the inflicted in-
after a second or booster injection of an Ag is usually much cision is small enough, the bleeding will stop rapidly when
greater than that formed after the first injection; thus, for the procedure is completed and gentle pressure is applied to
the production of high-titer Abs, the following schedules the cut by using cotton wool. The animal will heal within a
can be used (e.g., for rabbits). short time. Alternatively, the rabbit can be lightly anes-
thetized; a 22-gauge needle can be inserted into the vein,
1. Test bleed. To collect preimmune serum (this should be bevel up; and blood can be allowed to drip from the needle
done before the first injection) for use as a control to detect
into the collection container. If sterile blood is required, it
cross-reactive Abs can be withdrawn directly from the vein into a syringe and
2. Day 1. Inject 0.1 ml each (1:l emulsified mixture of
needle or into a Vacutainer (Becton Dickinson, Rutherford,
protein Ag with Freunds complete adjuvant or another
suitable adjuvant; see section 8.2.4) at four sites s.c., usually
NJ). The latter method will require proper instruction and
practice to avoid causing hematoma formation in the veins.
at the dorsal part of the body near the shoulders or the hips.
Larger volumes of blood can be obtained by bleeding
Freunds complete adjuvant should not be used more than
from the central ear artery or by using cardiac puncture on
once because repeat injections can cause granuloma forma-
anesthetized animals. The animal dies after cardiac puncture
tion and ulceration of the tissue at the injection site (44,
and is heavily sedated prior to starting the procedure.
53). The reaction to only one injection with this adjuvant
Cardiac puncture is not recommended for routine use but is
is usually mild.
useful if the animal is to be exsanguinated. The following
3. Day 4. Repeat the day 1 injection steps, except now steps can be followed for bleeding from the central ear artery.
Freunds incomplete adjuvant should be used (see section
8.2.4). After this step, the animals are allowed to rest for 14 1. Fifteen minutes prior to bleeding, administer S.C. or
days to allow the primary response to subside to a baseline i.m. a dose of anesthetic-either 0.125 ml of Innovar-
level; otherwise, a secondary response may not be achieved. Vetkg of body weight (11) or 0.4 ml of a mixture contain-
4. Day 18. Inject 1.0 ml (1:l mixture with Freunds in- ing 1 mg of oxymorphone and 0.5 mg of acepromazinekg
complete adjuvant) i.m. into the thigh muscles of one leg of (or as advised by a veterinary or animal technician). The
the animal. drugs will tranquilize the animals for up to 45 to 60 min.
5. Day 22. Bleed, and collect serum. 2. Dilate the central ear artery by warming the ear with
6. Day 25. Repeat the above-described step of i.m. injec- a lamp or gently rubbing it with ones hands.
tion on another leg of the animal. 3. Insert a 20- or 21-gauge needle into the central ear ar-
7. Day 29. Bleed, collect serum, and pool it with the pre- tery, bevel up, and allow blood to drip into the container.
vious sample. Again, for collection of sterile blood, aspirate the blood into
a sterile syringe. To prevent hematoma formation, apply
1.m. injections can be kept up once a month or once
pressure over the puncture site until bleeding has ceased; it
every 2 weeks, and serum can be collected 3 to 4 days later.
will take a few minutes since the puncture site is an artery.
To avoid batch-to-batch variability, the immune sera col-
lected at different times should be pooled, unless one is at- 8.1.6.2. Mice
tempting to monitor the immune response throughout a
time course. The general immunization schedule given Only a small volume of blood can be obtained from a mouse
above has been used successfully with a wide range of Ags, due to its small size. The easiest and most humane way is to
including soluble proteins, enzymes, whole-cell bacterial put the animal under light anesthesia, dip the tail into a
suspensions, lipopolysaccharide (LPS), and polysaccharide- beaker of warm water to dilate the tail veins, and nick a tail
protein conjugates. Variations can be made at some steps; vein (usually at about the 1 oclock position relative to the
for instance, an i.v. injection without adjuvant may be given dorsal part of the tail) with a scalpel. One can massage the
as a booster dose (on days 18 and 25) to elicit a more rapid tail lightly while withdrawing blood into a Pasteur pipette.
response before bleeding to collect antiserum. Adjuvants Approximately 0.2 ml can be obtained this way. Alter-
are not used for i.v. immunizations. natively, bleeding can be accomplished by withdrawing
from the venous plexus, located in the orbit behind the eye-
8.1.6. Blood Collection ball. The latter procedure is a more delicate operation and
Blood should be collected in a dry, sterile container without should not be attempted without instruction. One to three
adding coagulating or anticoagulating agents. milliliters of blood can be obtained from the heart or from
the heart cavity immediately after the animal is killed.
8.1.6.1. Rabbits
Restrain rabbits by wrapping each animal with a large towel 8.1.6.3. Rats
so that only the head and the ears are exposed. This is more Blood can usually be collected from the tail vein of a rat by
efficient than the use of commercially built rabbit-holding the procedure described above for mice. Again, once the an-
boxes. If the latter are used, the animals may kick or bounce imal is killed, larger amounts can be obtained from the heart.
violently and may sustain severe injuries such as a dislo-
cated spine. Alternately, one could anesthetize the animals 8.1.7. Serum Preparation
lightly to calm them down for all bleeding steps. Routinely, Freshly collected blood should be allowed to clot at room
blood is withdrawn from the marginal vein of the ear. The temperature for 30 min, followed by incubation at 37C for
vein closer to the head can be occluded by using a paper clip another 30 min. The cell-free fluid or serum, which con-
or by holding firmly with the fingers. This procedure pro- tains the Abs, is collected by centrifugation. Alternatively,
duces a better blood flow at the more proximal part of the incubate the clotted blood at 4C overnight to allow maxi-
vein for puncturing. A small cut to the dilated vein with the mum shrinkage of the clot before centrifugation and the
142 MORPHOLOGY AND ULTRASTRUCTURE

collection of serum. The low-temperature protocol yields 2. Mix 4.5 ml of bacterial supernatant with 6 ml of 2.5%
more serum per milliliter of blood and protects the Abs (vol/vol) SRBC (bacteria/SRBC ratio of 3:4 [vol/vol]; note:
from hydrolysis by the action of naturally occurring pro- commercially available SRBC are usually prepared as a
teases. Aliquot the serum into 0.5- to 2-ml volumes, and 2.5% solution in citrate buffer and are usually stable at 4C
store the aliquots frozen at -20C. For short-term storage at for at least 1 month). Isotonic solutions such as phosphate-
4C, add an antimicrobial agent to the serum. buffered saline (PBS) and Alsevers solution (see recipe
below) should be used for all manipulations of the SRBC
suspension. Incubate at 37C for 30 min with occasional
8.2. ANTIGEN PREPARATION shaking.
8.2.1. Particulate Ags 3. Sediment the SRBC by centrifugation at 200 X g for
10 min, and wash the cells twice with 10 ml of saline or iso-
8.2.1 . I . Bacterial Cells tonic buffer.
Due to their immunological foreignness and complex 4. Resuspend the pellet with 6 ml of PBS, and the coated
chemical compositions, bacteria are naturally immunogenic SRBC are ready for use in injections (Table 1) or in hemag-
and when injected into animals will induce an Ab response. glutination assays.
Therefore, the use of whole-cell suspensions to immunize The attachment of LPS onto SRBC has the advantage of
animals is common, provided that a protective or polyva- presenting the Ags on highly immunogenic particulate carri-
lent A b response against multiple components or antigenic ers. In addition, the amount of LPS presented to the animals
determinants of the microorganism is desired (5 1). Effective will be much smaller than a dose of pure LPS (LPS is endo-
immunizing doses of whole-cell suspensions (see section toxic); thus, this type of immunogen will be relatively non-
8.1.5) range from 5 x lo8 to 1 X 10 cellslml. The estima- toxic to the animals while effective in eliciting a response to
tion of cell counts per milliliter can be achieved by standard LPS. By treating RBC with 0.005% (wtlvol) tannic acid,
methods including serial dilutions, plate counts, or optical low-M, proteins can be adsorbed to the RBC surface.
density measurements at 600 nm (OD600) of standard cell
suspensions (e.g., an OD600 of 20.6 for Escherichia coli indi- Alsevers Solution (Citrate Saline Solution)
cates a cell concentration of approximately 109/ml of the Alsevers solution is an isotonic, anticoagulation blood pre-
suspension). Alternatively, the cell suspension can be com- servative that permits the storage of whole blood at refrig-
pared to McFarland standards (14), which use various con- eration temperatures for 10 weeks or more.
centrations of BaS04 suspended in water to give a rough es-
timate of bacterial concentration from lo8 to 10 cells/ml. Dextrose ........................... .20.50 g
Comparison of the test and reference suspensions is made Sodium citrate (dihydrate) .............. 8.00 g
easier if they are held in front of a white sheet of paper. Citric acid (monohydrate) . . . . . . . . . . . . . . 0.55 g
In some cases, vaccines with live, attenuated bacteria are Sodium chloride ...................... 4.20 g
necessary; however, infecting animal hosts with live micro- Distilled water ....................... to 1 liter
organisms may have some unexpected effects. Apart from
the fact that live bacteria may be more toxic and distressing Tanning of SRBC for Coating with Protein Ags
to animals, they can undergo antigenic and phase variations 1. Add 3 ml of 0.005% (wtlvol) tannic acid to a cen-
due to the pressure of host defense mechanisms. Bacterial trifuge tube containing 3 ml of 2.5% SRBC. Incubate at
cells can be killed either by heating at 100C for 30 min or 37C for 10 min.
by suspending the cells in formalin-saline overnight at a 2. Centrifuge the cells at 2,000 X g for 5 min, and wash
final formalin (formaldehyde) concentration of 0.3% once in 5 ml of PBS. Centrifuge as before, and resuspend
(vol/vol). Cells should be centrifuged and washed twice in the pellet in 3 ml of PBS.
isotonic saline to remove free formalin. Killing should be as- 3. To each centrifuge tube containing 3 ml of tanned
certained before injection. SRBC, add 3 ml of 0.3-mg/ml soluble protein, mix gently,
As a common practice, Ags should be divided into at and incubate at 37C for 15 min.
least 10 doses and stored frozen with or without adjuvants 4. Centrifuge, and wash twice as described above. Re-
to ensure the consistency of each dose. Repeated thawing suspend each pellet in 3 ml of PBS. These cells are now
and freezing of Ag should be avoided. ready for injection or for passive hemagglutination.
8.2.1.2. Sheep Erythrocytes as Carriers 8.2.1.3. Bacterial Cells as Carriers
Both carbohydrate and protein Ags can easily be adsorbed Smooth LPS containing 0 Ag sugars is generally strongly
onto sheep red blood cells (SRBC) to render them agglu- immunogenic, whereas semirough and rough LPS (contain-
tinable with antiserum specific for the coating Ags. Thus, ing only core oligosaccharides as terminal substituents) or
hemagglutination is easy and convenient for assessing Ag- capsular polysaccharides are often weakly immunogenic. To
Ab reactions. For the same reasons, SRBC can easily be elicit a response to epitopes of the core region, Ags such as
used as a carrier for purified carbohydrate Ags that by them- pure oligosaccharides, lipid A, and rough LPS can be at-
selves are normally not very immunogenic in rabbits (17). tached onto bacterial cells of a rough strain, for instance, E.
By treating red blood cells (RBC) with 0.005% (wtlvol) coli 15, according to the following method by Galanos et al.
tannic acid, low-M, proteins can be efficiently adsorbed and (18) with modifications by Bogard et al. (12).
the complexes can be used as immunogens or components
in passive agglutination reactions (section 8.5.1.3). 1. Prepare heat-killed cells of the rough strain at a con-
centration of 5 x lo9 cellslml in 1% (vol/vol) acetic acid.
Attachment of LPS Ags onto SRBC 2. Heat the cell suspension to 100C for 1 h, wash three
1. Boil an overnight culture of gram-negative bacteria times in distilled water, and lyophilize.
for 1 h; centrifuge at 2,000 x g for 10 min to sediment cell 3. Dissolve lipid A or rough LPS in 0.5% (vol/vol) tri-
debris. The supernatant contains a crude LPS preparation. ethylamide at a concentration of 1 mg/ml, and add the
8. Antigen-Antibody Reactions 143

lyophilized acid-treated bacteria to a final concentration of 2. Wash the gel slices in PBS or distilled water for a few
1 mg/ml. minutes. Finely chop the slices with a razor blade, or re-
4. Stir slowly for 30 min at room temperature. peatedly squeeze the gel pieces through the barrel of a stan-
5. Dehydrate the mixture in vacuo with a SpeedVac cen- dard 5-ml syringe with the help of a small amount of buffer.
trifuge (Savant Instruments Inc., Hicksville, NY). For the latter option, do a final squeezing step with a 21-
gauge needle attached.
Capsular polysaccharides made up of homopolymers or
containing sialic acids induce T-cell-independent immune
3. For immunization, the homogenate can be used di-
rectly or lyophilized and ground into a fine powder.
responses and produce low levels of humoral Abs. To improve
However, it is advisable to electroelute (see below) the
their immunogenicity, the polysaccharides can be covalently
proteins from these chopped-up gel slices before injec-
linked to carrier proteins to form glycoconjugates. These gly-
tion, since acrylamide is not easily degradable in an animal
coconjugates induce T-cell-dependent immune responses
host.
with the production of serum with high Ab titers (26).
8.2.2. Soluble Ags 8.2.2.2. Electroelution
Complete Ags usually have high molecular masses, al- Gel slices can be placed into a small dialysis tube contain-
though some naturally occurring immunogens can be small ing 1 ml of 0.2 M Tris-acetate (pH 7.4), 1.0% (wtlvol) SDS,
(e.g., insulin [6 kDa] and ribonuclease [14 kDa]). Molecules and 100 mM dithiothreitol per 0.1 g (wet weight) of poly-
with molecular masses of <3 to 5 kDa are not good im- acrylamide gel. The dialysis tubing is then placed into a
munogens. As a rule, polypeptides of >20 kDa should be horizontal electrophoresis chamber. Alternatively, gel slices
reasonably immunogenic and should induce a T-cell- can be placed into an ISCO sample cup (Canberra Packard,
dependent response with an increased level of specific IgG. Mississauga, Ontario, Canada) with a dialysis membrane at-
Smaller peptides can be conjugated to larger protein carri- tached to the bottoms of both chambers. The running
ers, adsorbed onto particulate carriers (such as SRBC as de- buffer contains 50 mM Tris-acetate (pH 7.4), 0.1% SDS,
scribed in section 8.2.1), or polymerized by treatment with and 0.5 mM sodium thioglycolate, and electroelution is run
a cross-linking bifunctional agent, such as 0.5% (vol/vol) for 3 h at 100 V. The gel slices can be removed and stained
glutaraldehyde (final concentration), for 1 h at room tem- with Coomassie blue to verify the removal of Ags. The pro-
perature. Here, the material is dialyzed against buffer to re- teins are then dialyzed against distilled water (to remove all
move the excess cross-linking agent. The disadvantage of detergents and other chemicals) and lyophilized, and the
glutaraldehyde treatment is the irreversible modification of product can now be incorporated into an emulsion with an
certain native epitopes. adjuvant for immunization (25).
The purity of soluble Ags from microorganisms depends
on the extraction method used. Soluble cell products such 8.2.3. Haptens
as toxins and enzymes are usually purified by a combination Ags with molecular masses of <10 kDa are usually weak im-
of column chromatography methods including ion ex- munogens, with the exceptions described previously. Thus,
change, gel filtration, chromatofocusing, and affinity tech- one needs to conjugate low-molecular-mass molecules with
niques. These methods are further enhanced by separation little or no immunogenicity onto carriers before immuniz-
using high-pressure liquid chromatography (HPLC) and fast ing animals in order to raise specific Abs. The carriers can
protein liquid chromatograpy systems. Chapter 7 specifi- be globular protein molecules, such as bovine serum albu-
cally deals with the fractionation of Ags from cells. The min (BSA), keyhole limpet hemacyanin, or particulate Ags
higher the purity, the easier it will be to raise specific Abs to as described above (section 8.2.1). For more detail on the
the Ag of interest. The following protocol outlines fre- choice of peptide sequences and the strategy for the cow
quently used rapid methods for purifying Ags from poly- pling of peptides to carriers, consult the manual by Harlow
acrylamide gels, provided the protein band of interest has and Lane (4).
been identified. A common procedure for conjugating haptens to protein
carriers uses glutaraldehyde, which cross-links molecules
8.2.2.1. Purification through their amino groups. The following procedure has
Protein Ags are usually separated by using the standard been used successfully to conjugate cadmium-binding pep-
sodium dodecyl sulfate (SDS)-polyacrylamide gel elec- tide (29) and can be used to conjugate other small peptides
trophoresis (PAGE) method of Laemmli (reference 23 and with 9 to 25 amino acids to BSA.
chapter 7) or, if native protein is important for eliciting an
Ab response to conformational epitopes, by omitting SDS. 1. Add 2 ml of 0.2% (vol/vol) glutaraldehyde to a 2-ml
The more commonly used methods for identifying the loca- solution containing 3 mg of your peptide plus 1 mg of BSA.
tion of the band of interest in the gel are either staining a To slow the rate of coupling, the pK of the buffer used in the
vertical strip of the gel to locate the Ag band or staining the reaction mixture should be above the pK of the peptides
entire gel lightly. The former has the advantage of avoiding amino groups (if known) so that NH2 and not NH3 will be
chemical fixation of the entire gel and denaturing the Ag of targeted.
interest. Normally, 0.05% (wtlvol) Coomassie brilliant blue 2. Incubate the mixture at room temperature for 2 h with
R-250 is used to stain the gel. The sensitivity is roughly 1 to gentle mixing. Dialyze for 24 h against several changes of 5
2 pg per band in a lane of standard width. If the more sen- mM Tris-HCI (pH 8.0).
sitive silver staining method has to be used to see the band, 3. Concentrate the contents of the dialysis bag either by
the quantity is insufficient for immunization. using a SpeedVac (Savant) or by sprinkling dry flakes of
polyethylene glycol (PEG; molecular mass, 6 kDa or
Direct Use of Gel Slices higher) onto the surface of the dialysis bag placed in a
1. Excise the band of interest with a razor or scalpel Pyrex dish. This can be accomplished in a matter of min-
blade. Before discarding the rest of the gel, stain it to check utes if sufficient PEG flakes are used to totally cover the
the accuracy of the excision. dialysis bag.
144 MORPHOLOGY AND ULTRASTRUCTURE

Alternatively, 1 to 2% glutaraldehyde can be used to it is not known whether ISCOMs can be used as effectively
cross-link the peptides amino groups such that it polymer- with other microbial products. Here is how to prepare them.
izes, increasing Ag size, but this may interfere with the epi-
1. Using 5-ml layers, prepare a discontinuous sucrose
topes of interest.
gradient ranging from 10 to 40% (wtlwt) (chapter 7) in TN
8.2.4. Adjuvants buffer (0.05 M Tris, 0.1 M NaC1, pH 7.2) containing 0.2%
Quil A (wtlvol).
With few exceptions, adjuvants are always used to enhance
the Ab response to an Ag. Several reviews can be consulted
2. Solubilize viral glycoproteins or other Ags with 2%
(vol/vol) Triton X-100 in TN buffer, and then place 200 pg
for more specific detail on adjuvants (37, 43, 45, 55, 56).
of the Ag in 200 pl of 8% (wtlwt) sucrose in TN buffer con-
Table 2 summarizes commonly used adjuvants and their
taining 1% Triton X-100 on top of the gradient.
characteristics.
3. Centrifuge at 150,000 X g for 4 h at 20C in an ultra-
8.2.4.1. Freunds Adjuvant centrifuge using an SW50 Beckman rotor.
Both complete and incomplete forms of Freuds adjuvant
4. Collect 250-pl fractions, and assess the quantity of
Ags either by protein assay or by the measurement of ra-
are readily available commercially, and here is how they are
dioactivity if the Ag is labeled. The fractions in which Ags
used.
are detected can be pooled. To remove excess Quil A, the
1. To aqueous Ags (usually suspended in isotonic saline), pooled fractions are centrifuged again in a 10 to 40% su-
add an equal volume of Freunds adjuvant (either the com- crose gradient as described above but without Quil A.
plete or the incomplete form). Mix ingredients vigorously Fractions are collected as before, pooled, dialyzed against
until a thick emulsion is formed. To help generate the emul- 0.5M ammonium acetate, pH 7.0, and lyophilized.
sion, take the mixture into a glass syringe fitted with an 18- 5. The amount of Quil A will be approximately 5%
gauge needle, forcing the material in and out of the syringe (wtlvol) in the ISCOM, i.e., <2 pg of QuilA/10 pg of pro-
until the syringe plunger is almost impossible to move. tein. The effective dose of Ag in the ISCOM will be in the
Alternatively, mix the Ag and adjuvant in a vortex vigor- range of 5 to 10 pg. Note that in a mouse weighing 20 g, 10
ously until a thick emulsion develops. The emulsion is to 50 pg of Quil A is toxic (48); however, nontoxic deriva-
deemed ready when a droplet of it does not disperse imme- tives exist (50).
diately when added to a saline solution.
2. Take up the volume needed for injection into a fresh 8.2.4.4. Liposomes
syringe, add a needle of an appropriate size, and the im- Liposomes are micelles that take the shape of concentric
munogen is ready for use. spheres consisting of phospholipid bilayers. Proteins and
other Ags can be trapped inside the liposomes. Further-
8.2.4.2. Alum more, depending on the nature of the interaction between
The following procedure can be used for alum (34). Ag molecules and liposomes, hydrophilic epitopes can be
well exposed by the liposomes. A commonly used mixture
1. Add 5 ml of 10% (wtlvol) aluminum potassium sulfate of lipids, composed of egg lecithin, cholesterol, and stearyl-
(dissolved in distilled water) to a 50eml conical centrifuge
amine in molar ratios of 7:2:1, forms positively charged li-
tube. Then add 22.8 ml of 0.25 N NaOH dropwise to the
posomes (30). If negatively charged liposomes are desired,
tube while vortexing.
phosphatidic acid or dicetylphosphate replaces stearyl-
2. Allow the mixture to settle for 10 min at room tem-
amine. Negatively charged liposomes are reported to be
perature.
superior to positively charged ones as adjuvants to elicit an
3. Sediment the Al(OH)3 by centrifugation at 1,000 X g
Ab response to diptheria toxin (42). While there are many
for 10 min. Wash with 50 ml of distilled water, and repeat
combinations of phospholipids that can be used, the follow-
the centrifugation. Discard the supematant.
ing general protocol can be used (35).
4. The pellet contains the Al(OH)3, or alum adjuvant,
which has a binding capacity of 50 to 200 pg of protein Ag. 1. Dissolve 75 mg of phosphatidylcholine and 11 mg of
5. Add an aliquot of Ag to alum, allow the two compo- cholesterol in chloroform by using a round-bottomed flask.
nents to mix at room temperature for 10 min, centrifuge at A thin film will develop after low+powervacuum rotary
10,000 X g for 10 min, and check the supernatant for un- evaporation at 37C for a short time or under a nitrogen at-
bound Ags. The sediment is ready for injection. mosphere.
2. BSA liposomes can be prepared by adding 10 mg of
8.2.4.3. ISCOMs BSA in 10 ml of PBS (0.15 M NaC1, 10 mM phosphate
Immunostimulatory complexes (ISCOMs) are adjuvant sys- buffer, pH 7.4) to the flask with the thin film of lipid and
tems composed of cholesterol, lipid, phospholipid, and incubating at 37C for 1 h with gentle rotation. Altem-
saponins (46, 47, 55). The early formulations of ISCOMs atively, the mixture can be subjected to bath sonication for
used Quil A, a mixture of saponins (46), which showed var- 30 min.
ious levels of toxicity in animals. A recent-generation 3. Wash the BSA liposomes three times with PBS
ISCOM containing a 7:3 ratio of Quil A and Quil C (100,000 X g, 30 min) to remove unattached BSA.
(ISCOPREPTM703)is nontoxic as an animal and human Resuspend the pellet in a small volume of saline or PBS.
vaccine (50).Preparation of ISCOM with Quil A can be The amount of bound BSA can be assessed by a standard
done according to the method of Morein et al. (46). Since protein assay. The Ag-incorporated liposomes are ready for
Quil A behaves like a detergent in water and forms micelles use.
at the critical micelle concentration of 0.03% (wtlvol), it
easily forms a complex with Ags in which the Ag is present 8.2.4.5. Ribi Adjuvant Systems
in an accessible and multimeric fashion. ISCOMs prepared Adjuvant systems are available from Ribi ImmunoChemical
from Quil A have successfully elicited humoral responses Research, Inc. (Hamilton, MT), and they contain a combi-
against viral products in animal protection studies. Thus far, nation of mycobacterial cell walls, trehalose dimycolate, and
8 . Antigen-Antibody Reactions a 145

TABLE 2 Adjuvants and their characteristics


Adjuvant(s) (Source) Characteristics Reference(s)

Freunds complete ad- Most commonly used adjuvant 40


juvant (FCA) Mix 1:l (vol/vol) with Ag until thick emulsion is formed
Serves as a depot for slow release of Ags
Emulsion of oil (Bayol F), detergent, and an extract from mycobacteria
Must never be used for i.v. injections
Disadvantages: (i) elicits granuloma at site of injection if used repeatedly and (ii) is
difficult to prepare as an emulsion

Freunds incomplete Contains the oil and detergent only without bacterial extract 40
adjuvant Mix 1:l (vol/vol) with Ag as described above
Do not use in i.v. injections
Normally used to replace Freunds complete adjuvant in subsequent injections

Alum Aluminum hydroxide salt (gel-like) which also facilitates slow release of Ags 16,48, 53
Attracts immunocompetent lymphocytes to the area of injection to induce an improved
Ab response
Elicits IgGl and IgE responses in mice
Is less toxic than Freunds complete adjuvant

Liposomes Can be made as lecithin-cholesterol-dicetylphosphate in a molar ratio of 7:2: 1 19, 35,42,52


Are useful to entrap Ags for slow release
Advantages: (i) function as a depot of Ags, (ii) help Ags to retain T-cell dependency of
native protein
Disadvantage: entrapped Ags, not exposed on the outer lipid layer, may not be detected
by immune system

LPS B-cell mitogen 41, 49, 51


Stimulates natural (nonspecific) Abs if administered without Ag but stimulates specific
Ab response when injected with an Ag
Usually, the lipid A region is the immunopotentiating component; however, mannan 0
side chain sugars from Klebsielh strain 03 and E. coli 09 were shown to have adjuvant
effects as well
Toxicity is the major disadvantage

Muramyl dipeptide Active component of mycobacterial extract used in Freunds complete adjuvant 38
Is prepared in many forms and derivatives
Stimulates specific Ab response to a wide variety of natural Ags such as bacteria, viruses,
and fungi
Is nontoxic

Bacterial toxins and Genetically detoxified derivatives of cholera toxin or heat-labile E. coli toxin 13, 16
proteins Potent adjuvants for induction of mucosal, humoral, and cell-mediated immunity

Quil A Composed of saponin, a mixture of water-soluble triterpene glycosides from the South 43,46,47,
American tree Quillaia saponaria 50,55
Advantage: is highly surface active and forms stable complexes known as ISCOMs with
viral envelopes
Has been reported to be superior to Freunds complete adjuvant in eliciting Ab response
to human serum albumin. Use of 5-10 pg of Ags in the ISCOM for each dose will
elicit better response than 100-200 p g of aggregated Ag without Quil A

Synthetic adjuvants, A synthetic MDP muramyl dipeptide was shown to stimulate IgG2 to human serum 38,43
formulation 1 (e.g., albumin in mice
SFA-1, Syntex) Toxicity in animals and low viscosity; the latter means that Ag-Ab mixture will be easy
to mix and to administer to animals

(Continued on next page)


146 MORPHOLOGY AND ULTRASTRUCTURE

TABLE 2 (Continued)
Adjuvant(s) (Source) Characteristics Reference(s)
Adjuvant system (Ribi Composed of monophosphoryl lipid A 31, 32, 39
Immunochem From 5-50 pg of monophosphoryl lipid A was shown to augment humoral responses to
Research, Inc., both polysaccharide and protein Ags
Hamilton, MT) Nontoxic and has low viscosity; thus, Ag-adjuvant mixtures are easy to prepare

SBAS4 (SmithKline System containing alum and 3-deacylated monophosphoryl lipid A 36, 39, 54
Beecham Adjuvant Known to elicite humoral and cellular immune responses
System 4)

Bacterial DNA Synthesized as oligodeoxynucleotides containing methylated CpG motifs derived from 20, 28, 33
bacterial DNA
Also called immunostimulatory CpG ODNs
Is a potent stimulator of cytokines leading to induction of mucosal, humoral, and cell-
mediated immune responses
Can be administered to potentiate immune responses to protein Ags

a nontoxic lipid A derivative (monophosphoryl lipid A). The directly in Ag-Ab reactions, absorbed to deplete it of non-
adjuvant effect appears to be based on the ability of mono- specific or cross-reactive Abs (section 8.3.1), or fraction-
phosphoryl lipid A to inactivate suppressor T cells while stim- ated to purify Abs free of other serum proteins (sections
ulating a polyclonal B-cell response (32,36,38,39). 8.3.2 to 8.3.4). The two principal types of proteins in serum
are globulins and albumin, which can be fractionated by
8.2.4.6. DNA Immunization various means. The most common purification methods in-
Nucleic acid (DNA or RNA) immunization and genetic volve ammonium sulfate precipitation, dialysis to get rid of
vaccines introduced in the early 1990s are some of the most the salts, and anion exchange chromatography for purifica-
important discoveries and novel strategies in vaccine devel- tion. This combination is tedious but inexpensive and can
opment (20, 21, 24, 27). The essential features of a DNA effectively isolate all IgG molecules from serum. Gel filtra-
vaccine are a bacterial plasmid vector engineered to carry a tion may be used instead of anion exchange chromatogra-
DNA insert encoding the protein immunogen(s) of inter- phy when IgM purification is desired. More recently, the use
est, a eukaryote gene promoter, and a poly(A) site to enable of affinity chromatography can dramatically speed up the
expression of the protein in mammalian cells. The vectors process, but more importantly, it can help to target the spe-
are usually maintained in and purified from E. coli. Bacterial cific desired Ab if purified Ag is incorporated onto an affin-
DNA is a potent adjuvant that stimulates immune cells and ity matrix. Affi-Gel blue (Bio-Rad) replaces the salt precip-
Ag-presenting cells and induces B-cell proliferation and the itation step because the Cibacron Blue@dyeof its matrix (2
secretion of Abs. The immunostimulatory properties are at- mg/ml) has a strong affinity for binding albumin (at 11
tributed to unmethylated CpG dinucleotide motifs found mg/ml); therefore, it can be used as a one-step method to
with high frequency in bacterial and viral DNA (28). yield a relatively clean IgG fraction. HPLC and fast protein
For immunization, the recombinant plasmid DNA is de- liquid chromatography methods involving a high-pressure
livered directly into muscles of the host animal and the pump and special columns provide better speed and resolu-
DNA is taken up by tissue and translated in vivo into pro- tion for the separation of Abs from other serum proteins;
tein Ags. DNA vaccines induce both humoral (Ab) and however, the equipment is expensive and may not be read-
mucosal immune responses and elicit strong, sustained T- ily available.
cell-dependent responses. DNA vaccines are typically ad-
ministered to the host by injecting tissues or by using a gene 8.3.1. Absorption of Cross-Reactive
gun that provides high-pressure contact delivery. Recent and Nonspecific Abs
studies describe systemic and mucosal routes with or with- In addition to the Ag-specific Abs, polyclonal immune sera
out adjuvants with the induction of mucosal and humoral contain a variety of Abs representative of immune re-
responses (15, 28). There are advantages of using nucleic sponses to Ags that an animal is exposed to from its envi-
acid vaccines; e.g., a variety of well-characterized plasmid ronment, gut, or foods. These so-called background, or non-
vectors are available, recombinant plasmids are easily pre- specific, Abs include those to Ags of bacteria, other
pared and stable, insert DNA (encoding the immunogen) is microorganisms, and macro-Ags such as pollen and dust
stable, and there is never a need to prepare the Ag for im- mites, etc. When tested against bacterial whole-cell lysates,
munization (15, 20, 24, 28). these serum Abs interact with common and conserved anti-
genic determinants of bacterial surface molecules including
LPS, capsular polysaccharide, flagella, and outer membrane
8.3. CONVENTIONAL ANTIBODY proteins. To increase the sensitivity of sera and enhance the
PREPARATION detection of specific Ags, cross-reacting or background Abs
The polyclonal serum is expressed from the blood clot by are removed from the polyclonal antisera by adsorption
centrifugation, and approximately 52% of its volume can be with whole bacteria to remove cross-reacting Abs to cell
collected as serum (section 8.1.7). The serum can be used surface-localized Ags or bacterial cell lysates are absorbed
8. Antigen-Antibody Reactions H 147

onto nitrocellulose disks to deplete the sera of all cross- saturation, although a small amount of albumin may be co-
reacting Abs (61, 62). precipitated.

Procedure Procedure
1. Lyse E. coli cells from a 100-ml overnight culture by 1. To prepare a saturated stock solution of (NH4)2S04,
incubating the cells in a lysis buffer (50 mM Tris [pH 81, 2 add 100 g of the crystals for every 100 ml of distilled water
mM EDTA, 2% [wt/vol] SDS, 4% [vol/vol] P-mercap- and leave the mixture stirring for 1 to 2 days on a magnetic
toethanol, 1% [wt/vol] lysozyme) for 1 h at 37C. stir plate at room temperature. There should always be
2. Centrifuge the cell lysate at 12,000 X g at 4C for 30 undissolved crystals on the bottom; otherwise, the solution
min. Discard the pellet, and collect the supernatant. is not saturated. Check and adjust to pH 7.0. Store at 4C
3. Immerse five pieces of nitrocellulose (NC) mem- since Ab precipitation is usually performed at this tempera-
branes, 35 cm2, in the cell lysate supernatant, and incubate ture.
for 1 h on a shaker at room temperature. 2. To 20 ml of serum (already cooled to 4"C), slowly add
4. Wash the treated N C membranes three times (10 min 10 ml of saturated (NH4)2S04dropwise from the stock so-
each) in Tris-buffered saline (TBS; 10 mM Tris-HC1, [pH lution while the solution is stirred. Continue the stirring for
81, 150 mM NaC1). at least 2 h at 4C.
5. Block the membranes for 1 h with 3 to 5% (wtlvol) 3. Sediment the pellet by centrifugation at 10,000 x g
skim milk in TBS. The purpose of this step is to fully satu- for 10 min.
rate any remaining binding sites on the N C membranes to 4. Resuspend the pellet with a small volume of buffer (5
avoid nonspecific binding of Abs to the membranes in sub- ml), e.g., PBS or TBS (50 mM Tris, 1.5 M NaC1) at pH 7.0,
sequent adsorption steps. and then add 2.5 ml of saturated (NH4&304 as in step 2.
6 . Wash the membranes again three times for 5 min each Sediment the pellet by centrifugation as before.
with TBS. Now the membranes are ready to be used for ad- 5. Resuspend the pellet with 10 ml of buffer, and trans-
sorption of background Abs from the antiserum. If the fer it into a dialysis bag. Dialyze overnight at 4C against
membranes are not being used right away, they can be air two to three changes of 2-liter volumes of buffer. The dia-
dried; wrapped in plastic film, sealed in a plastic bag by lyzed material can now be stored frozen or can be
using a bag sealer device, or sealed in a zip-lock bag; and lyophilized prior to further steps such as anion exchange
stored at -20C until use. Before use, remove the mem- chromatography.
branes from the freezer, allow them to thaw at room tem-
perature for 20 min, and soak for 1 to 3 min in PBS. 8.3.3. Ion Exchange Chromatography
7. To begin the adsorption process, immerse one mem- For the purification of IgG, ion exchange chromatography
brane in 10 to 20 ml of diluted antiserum (1/100 in saline or is a natural second step after ammonium sulfate precipita-
PBS) and incubate for 1.5 h at room temperature. tion to reduce albumin. The most commonly used matrix
Adsorption is done in petri dishes. Use a rotary shaker to for Ab purification is one to which an ionizable DEAE
provide good mixing during the incubation. group is attached. DEAE-Sephacel (cellulose) and DEAE-
8. After the incubation, remove the membrane and re- Sepharose from Pharmacia are convenient, preswollen, and
place it with a second membrane. Repeat this process until ready to use. They have high capacities, and a 10-ml col-
all five membranes have been used. umn can be used to bind 100 to 200 mg of protein. At pH
9. Now the antiserum is ready for use for Western im- 8.0, both matrices are strongly cationic for binding Abs.
munoblotting and should yield results with little to no Immunoglobulins have PIS in the range of 6 to 8; thus, they
background protein band reactivity. If not immediately will be anionic at pH 8.0 and will bind to DEAE at low salt
used, the freshly adsorbed antiserum should be stored frozen concentrations. Elution of Abs can easily be achieved either
in the presence of a small amount of a preservative such as by increasing the concentrations of competing ions (e.g.,
sodium azide. We often collect the used serum, add a pre- C1-) in the column buffer or by lowering the pH. The re-
servative, store it frozen, and reuse it for blots up to five covery of Abs should be almost 100%.
times or until the intensity of the reaction against the im-
munizing Ag is appreciably decreased. Procedure
1. Pack and equilibrate a DEAE-Sephacel or DEAE-
8.3.2. Ammonium Sulfate Precipitation Sepharose column (1.5 by 10 cm) with 10 mM Tris buffer,
Ammonium sulfate is the most widely used salt for the pre- pH 8.0, at room temperature. To avoid the formation of air
cipitation of proteins. (NH4)2S04 has the advantage of a bubbles which will diminish the efficiency of the column,
high level of solubility which is only minimally dependent the buffer and gel slurry should be allowed to equilibrate to
on temperature; its solubility varies only -3% between 0 room temperature before starting. The size of the gel bed
and 25C. In contrast, Na2S04, which is indicated in some will be determined by the amount of Abs to be bound. As
methods, is five times as soluble at 25 as at 0C. Proteins are described above, one can pack a 10-ml column for every
polyvalent ions, with surface charges that interact with 100 mg of protein to be bound. If chromatographic columns
water molecules through hydrogen bonding. As SO4'- ions are not available, a plastic disposable syringe will suffice as
also attract water molecules, they compete with proteins long as some fiberglass is used as a bottom screen to prevent
and, at high concentrations, will strip away the solvation leakage of the slurry.
layer. The proteins will then have an increased tendency to 2. Wash the column with at least 5 column volumes of
interact with one another and precipitate from the solution. the 10 mM Tris buffer. A gravity feed with a head pressure
The gamma globulin fraction, containing Abs, is obtained of 100 to 200 mm will be sufficient (65) if a peristaltic pump
after repeated precipitations with (NH4)2S04added to is not available.
make a 33.3% saturation solution (see the recipe below). 3. Apply the ammonium sulfate-precipitated and dia-
Unwanted proteins, including albumin, will remain in the lyzed Ab solution onto the column. If the dialyzed Abs have
solution. A higher yield c.an be achieved by a single 50% been lyophilized, reconstitute them in 10 mM Tris.
148 w MORPHOLOGY AND ULTRASTRUCTURE

4. Wash the column with 2 to 5 bed volumes of Tris protein carriers. More appropriately, the coupling of micro-
buffer. Collect these fractions to ensure that Abs are not bial polysaccharides can be achieved through the binding of
lost. their carboxyl groups to one of the following gels: AH-
5. Develop the column with a linear gradient of increas- Sepharose 4B (Pharmacia), Affi-Gel 102 (Bio-Rad), and
ing concentrations of NaCl made from 100 ml of 10 mM Aminoethyl Bio-Gel (Bio-Rad). Detailed descriptions of the
Tris buffer in the first chamber and 100 ml of 0.3 M NaCl principles and procedures of the coupling of ligands to afin-
in 10 mM Tris buffer in the second chamber of a gradient ity gels can be found in a document entitled AfJlnity Chrom-
maker. The usual total volume of the gradient is 10 to 20 atography: Principles and Methods, which can be obtained from
times the bed volume. The gradient is allowed to run over Pharmacia free of charge.
4 h, and 5-ml fractions are collected. Gradient makers can
be purchased from commercial sources, and as long as both 8.3.5.1. Affi-Gel Blue
chambers are of the same size, linear gradients can easily be In addition to the aforementioned activated gel matrices for
achieved. To mix the solutions, a magnetic bar is placed in preparing affinity columns, one product from Bio-Rad
each chamber and the gradient maker is placed on a mag- known as AEi-Gel blue is worth mentioning (59). Affi-Gel
netic plate. Mouse IgG2a is usually eluted earlier, and IgGl blue (50 to 100 mesh size) is a beaded, cross-linked agarose
is usually eluted later. Mouse IgG3 may not be stable at low with covalently attached Cibacron Blue F3GA dye. There
ionic strength; thus, the concentration of the Tris buffer is ca. 1.9 mg of dye/ml of gel, and the dye has a very high
used for the purification of IgG3 should be increased to 50 affinity for albumin. It can bind 2 1 1 mg of albumin/ml of
or 100 mM. Alternatively, the column can be eluted with a Affi-Gel blue. Thus, a single passage of serum through a
stepwise salt gradient. A series of 50-ml volumes in 10 mM minicolumn of this gel can yield a relatively pure im-
Tris (pH 8.0) are used. The first contains no salt, and then munoglobulin fraction with albumin virtually removed.
increasing salt concentrations of 50 mM NaC1, 100 mM This method is very rapid compared to the steps involved in
NaC1, 150 mM NaC1, 200 mM NaC1, 250 mM NaC1, and ammonium sulfate precipitation, dialysis, and concentra-
300 mM NaCl are added. Most of the Abs should be eluted tion to isolate immunoglobulins. Minicolumns with 5- to
by the time the 200 mM NaCl step is added. 10-ml volumes can be prepared in Econo-columns (Bio-
6. The fractions are monitored by using a spectropho- Rad) or in syringes. To reconstitute the column, albumin
tometer fitted with a UV detector or by assaying for pro- can be removed by using 3.0 M potassium thiocyanate or 8
teins. The purity of the eluted Ab can be assessed by stan- M urea (65), and then the column should be reequilibrated
dard agarose gel electrophoresis or SDS-PAGE. with running buffer of near-neutral pH.
8.3.4. Gel Filtration 8.3.5.2. Protein A-Agarose
Gel filtration chromatography separates proteins according Protein A is a cell wall component of Staphylococcus aureus
to their molecular sizes. Since IgM has a molecular mass of with a strong affinity for the Fc region of IgG. Protein A has
-900 kDa, it can be separated easily from other Abs with a molecular mass of 42 kDa and consists of four globular
average molecular masses of 150 kDa. Many commercial immunoglobulin-binding sites at the N-terminal end.
gels are suitable for this purpose, including Sepharose 6B, Protein A interacts with the immunoglobulins of 65 mam-
Sephacryl S-500, and Sephadex G-200 (all from malian species (69). Purified protein A can be covalently
Pharmacia), Ultrogel AcA 22 (LKB), and Bio-Gel P-300 attached to gel beads such as agarose and used as an affinity
(Bio-Rad). Sephacryl (a mixture of dextran and acry- gel for the purification of IgG. The binding of protein A to
lamide) and Ultrogel (a mixture of agarose and acrylamide) IgG is generally at least five times stronger than binding to
are more resistant to compression and easier to pack and IgM. However, protein A-agarose columns can be used to
can be run at higher pressures and flow rates than gels like purify IgM if buffers of high ionic strength and high pH are
Sephadex (65). The columns used are typically long, thin used (60, 70). Protein A-agarose can be obtained from
ones (15 to 30 mm in diameter). When packed and prop- Pharmacia or Bio-Rad; the latter product is known as the
erly degassed, the gel generally occupies approximately 70% MAPS (MAb purification system), which can be purchased
of the column and the space between the gel beads occupies as a kit with the column and all the necessary buffers. In
the remaining 30%. Either TBS or PBS can be used at pH general, IgGZa, IgGZb, and IgG3 can be easily purified by
7 to 8 at an NaCl concentration of 2 1 0 0 mM to prevent this method, and IgGl and IgM can be purified when high
adsorption effects. Sodium azide at 10 mM may be added in pH and high ionic strength are employed (63). Elution of
running buffers to prevent microbial growth. For best reso- the Abs is usually achieved by using acidic buffers such as
lution, the flow rate of buffers should be low, which means citrate or glycine-HC1 buffer at pH 5.0 or lower. The buffer
that it will take 1 to 3 days to perform a proper run. A n ob- conditions described below for HPLC purification of MAbs
vious disadvantage besides the time this takes to perform is can be used. Naturally, columns should be run at a much
the dilution of the Abs being eluted. However, concentra- lower flow rate, for example, 2 to 5 ml/h, than those in
tion of the fractions is easily achieved by lyophilization or HPLC systems (5 to 10 ml/min). In order to avoid clogging
concentration by using PEG (see section 8.2.3). of the rather expensive protein A-agarose columns, the
methods described for the pretreatment of ascitic fluid
8.3.5. Affinity Chromatography could be used. Samples should at least be passed through a
Ag covalently bound onto a chromatographic gel matrix pro- 0.45 to 0.22 pm-pore-size filter before being applied to the
vides specific binding to isolate the Ab of choice from serum. column. Acid-eluted Abs should be neutralized with 1 M
Some of the commonly known gels are CNBr-activated Tris, pH 8.0 (65), immediately after elution from the col-
Sepharose 4B and Sepharose 6B (Pharmacia), which are umn to avoid any loss of Ab activity.
ready for the direct coupling of Ags through the primary
amino groups of the Ags. For obvious reasons, direct coupling 8.3.5.3. Protein G
of high-molecular-mass polysaccharides onto CNBr-activated Protein G is a cell wall component of alpha-hemolytic
gels is not possible until the polysaccharidesare conjugated to group C and G streptococci (57,58). Native protein G has
8. Antigen-Antibody Reactions 149

a strong affinity for the Fc regions of all IgG Abs, including Incubate overnight at 37C and 5% COZ. This allows the
IgG classes that have low affinity for protein A; however, cells to recover from the PEG shock before being subjected
this protein also binds albumin (57). Recombinant protein to the selective medium containing HAT and 20% FCS. At
G in which the albumin site has been eliminated is now this time, prepare feeder cells from the spleen and thymus of
available for immunoglobulin purification and immunode- a nonimmunized mouse (e.g., a CD1 mouse); incubate the
tection procedures (58). cell suspension (at lo6 cells in medium containing 10%
FCS) overnight at 37C and 5% C02.
7. O n the next day, add to the fused cells the feeder
8.4. M A b PREPARATION cells and 50 ml of medium containing a 2X concentration
Since the first report by Kohler and Milstein (66) of cell fu- of HAT (HAT can be obtained as a 50X stock from most
sion between a hypoxanthine-aminopterin-thymidine suppliers of tissue culture materials) to yield a density of 5
(HAT)-sensitive variant of MOPC-21 myeloma cells and x lo4 to 5 x lo5 myeloma cellslml. This is the concentra-
spleen cells immunized with SRBC, the hybridoma tech- tion at which myeloma cells grow best.
nique has been well exploited by scientists in all life science 8. Feeder cells are added at a concentration of lo5
disciplines. The main advantages of MAbs over conven- cells/ml. Transfer aliquots of the cell suspensions onto 24- or
tional Abs include increased reproducibility and specificity, 96-well tissue culture plates.
improved sensitivity, and the ability to produce unlimited 9. Leave the fused cells undisturbed for 10 to 14 days in
amounts of immunoglobulins. These advantages usually an incubator set at 37C with 5% C02and 95% humidity.
outweigh the expense and the fact that the people involved Hybrid clones that can survive the HAT selection will ap-
in MAb production require a fair amount of training. How- pear as plaques (or colonies of clones) on the bottoms of the
ever, one should consider the real need for MAbs before em- wells. These plaques can be confirmed by viewing with an
barking on the process. For most intended purposes, there is inverted microscope.
nothing wrong with rabbit polyclonal Abs, which cost a 10. Remove culture Supernatants, assay for specific ac-
fraction of what it would cost for MAbs. For consideration tivity against the immunizing Ag (e.g., by enzyme-linked
of the legal ramifications of the use of animals for MAb pro- immunosorbent assay [ELISA]), and identify the wells pos-
duction, consult the review by Kuhlmann et al. (67). The itive for activity.
following protocol is for the production of murine MAbs by 11. Clone cells in positive wells by using the limiting
cell fusion by using a special mixture of PEG and dimethyl dilutions (71). The medium used should contain 20%
sulfoxide. (vol/vol) FCS and hypoxanthine-thymidine obtained as a
50X stock from suppliers. Reclone cells in the same manner.
8.4.1. Production Protocol 12. Culture supernatants from the twice-cloned cell lines
usually contain microgram quantities of MAbs and are
Most protocols used for this purpose are based on the pro-
ready for use for most Ag-Ab reactions. In order to produce
tocol of GalfrC and Milstein (64) with PEG as a fusogen in-
larger quantities (such as milligram-per-milliliter amounts)
stead of Sendai virus as used in earlier studies. The follow-
of MAbs, the method of ascites fluid production described
ing is a brief outline of the procedure described by Lam et al.
below can be followed. Several systems have also been de-
(68) for the production of MAbs against LPS; all reagents scribed for scaling up the production of MAbs from hy-
are from Sigma.
bridoma cells grown in serum-free media by using shaker
1. In a 50-ml sterile centrifuge tube with 10 ml of flasks, roller bottles, and hollow fiber and capillary systems
serum-free medium (either RPMI 1640 or Dulbeccos mod- (refer to the Invitrogen website at http://www.invitrogen
ified Eagle medium [DMEM]), mix lo8 spleen cells from an .com for the GIBCO BRL Guide to Hybndoma Technology).
immunized mouse (BALB/c) with lo7 myeloma cells in 10
ml of the same medium. This ratio of 1O:l of the two cell 8.4.2. lsotyping Abs
types may be varied depending on personal experience. Before a MAb can be further characterized or exploited, it
2. Centrifuge at 400 X g for 5 min. Resuspend the pel- is necessary to identify the Ab isotype, including its class
let with 20 ml of serum-free medium, split the cell suspen- and subclass and the presence of a K- or A-light chain.
sion into four equal aliquots of 5 ml each, and put the Commercially produced anti-mouse (and anti-rat) Ab
aliquots into 15-ml sterile centrifuge tubes. The purpose of reagents are readily available such that there is no longer a
working with four smaller cell pellets is to allow better ex- need to produce Abs to each of the immunoglobulin iso-
posure of cells to the fusogen in the step below. Repeat types for identification. Depending on the commercial
step 2. mouse Ab typing kit purchased, a simple double immuno-
3. To one of the four cell pellets, dispense 0.5 ml of the diffusion test, a more sensitive passive agglutination test, a
fusogen (40% [wt/vol] PEG [molecular weight of 1,050],4% dot-blotting method with precoated nitrocellulose strips, or
[vol/vol] dimethylsulfoxide, 56% [vol/vol] DMEM or RPMI a highly sensitive ELISA method can be used. Table 3 sum-
1640) in small dropwise quantities in exactly 2 min. (The marizes the various commercial kits.
other three tubes with the cell pellets should be kept on ice
until ready for use.) 8.4.3. Ascites Fluid Production
4. Add 20 ml of serum-free medium over 3 to 5 min. The term ascites refers to an accumulation of fluid in the
Centrifuge at 400 X g for 10 min. abdominal cavity. In medicine, ascites is usually associated
5. Tap the tube gently on the bench to dislodge the cell with circulatory congestion due to disorders of the heart,
pellet. Gently resuspend the cell pellet in 12.5 ml of lungs, kidney, or liver. For research purposes, ascites is in-
medium containing 20% (vol/vol) fetal calf serum (FCS). It duced in mice as a source of the production of highly con-
is all right to leave some clumps of cells. Repeat steps 3 centrated MAbs. Ascites fluids like serum contain 10 to 20
through 5 for each of the remaining cell pellets. mg of Ab/ml. This level of Abs is at least 100 to 1,000 times
6. Combine the four fused cell suspensions, each 12.5 more than that obtained in tissue culture supernatants of
ml, and transfer to a large (75-mm) tissue culture flask. hybridoma lines. Both Freunds incomplete adjuvant and
150 MORPHOLOGY AND ULTRASTRUCTURE

TABLE 3 Commercial sources of immunoglobulin isotype determination reagents


Commercial reagents or kits Techniaue used Remarks
Bio-Rad mouse typer isotyping kit ELISA Complete kit includes rabbit anti-mouse sera, goat
anti-rabbit Ab-peroxidase or horseradish conjugates,
substrate solutions, and anti-tc and anti-A reagents
Monoclonal typing kit, mouse or rat, Gel diffusion Simple to use, but one has to wait 24-48 h before
immunodiffusion (ICN Immunobiologicals) reading results
Monoclonal typing kit, mouse, Hemagglutination More sensitive than gel immunodiffusion and
hemagglutination (ICN Immunobiologicals) can detect 1-20 p.g of MAbs
Mouse MAb isotyping reagents (Sigma) Choice of uses 6 reagents are provided to identify all classes and subclasses,
but no anti-light chain reagents. The reagents can be
used in either ELISA or immunodiffusion methods
Sigma ImmunoType kit Dot blotting Precoated strips and reagents for color development are
provided

pristane, a defined mineral oil (see below; 3, 72,73), can in- 6. Separate the fluid containing the Abs from the pellet.
duce ascites production in mice. The latter has been partic- Note that the pellet contains the healthiest hybridoma cells
ularly useful for priming BALB/c mice for the injection of and they should be resuspended in hypoxanthine-thymidine-
myeloma cells to induce Ab production; ca. 5 to 10 ml of as- or HAT-containing DMEM to be propagated for freezing
cites fluid can be generated by this procedure. However, be- and storage. Cell lines suspected of containing contaminat-
fore choosing to use this route of Ab production, one should ing microbes can be cleaned by passage through an animal
realize that both Freund's adjuvant and pristane cause im- by using this protocol. The normal white blood cells from
munosuppression yet induce tumor formation in the peri- the ascites fluid will eventually die and should not pose any
toneal cavities of the injected animals. Without doubt, this problem to the purity of the hybridomas. The ascites fluid is
will cause stress to the animals; repeated injection of the an- now ready to be used for Ag-Ab reactions, with or without
imals over prolonged periods and more than one harvest of further purification.
ascites fluids should be avoided to minimize stress. The pro-
Many undesirable components such as lipids, adjuvants,
cedure is outlined below.
and fibrins are included in ascites fluid collected from the
animals. These impurities can clog up affinity columns and
1. For pristane priming, inject 0.1 ml of pristane the more expensive HPLC columns; thus, they must be re-
(2,6,10,14-tetramethylpentadecane;Aldrich Chemicals;
moved before proceeding with purification. The following
catalog no. T2-280-2) i.p. into a BALB/c mouse and, 1
methods can be used separately or in combination to
week later, inject an additional 0.2 ml of pristane i.p. (Most
achieve this goal.
protocols recommend one to two 0.5-ml doses for priming;
however, we find that increasing the second dose to 0.5 ml 1. Add CaClz to a final concentration of ca. 1 mM (per
or higher did not increase the volume of ascites fluid recov- Bio-Rad instructions for Affi-Prep protein A columns).
ered. By using two smaller doses of pristane, animals are sub- Allow the mixture to sit for 2 h at 4C. Use a wooden ap-
jected to less stress and the survival time is longer than plicator stick or other suitable probe to remove the fibrin-
when 0.5 ml is used.) lipid clot. Centrifuge at 10,000 x g for 10 min at 4"C, col-
2. Optimally, pristane priming should not be done more lect the clear supernatant, and filter it through a 0.22- or
than 60 days prior to the injection of the hybridoma cells. 0.45-pm-pore-size sterile filter. The supernatant is now
3. One week after the administration of the second dose, ready for injection into HPLC columns.
lo6 hybridoma cells (0.1 ml) can be injected i.p. into the 2. Ascites fluid can also be poured through a small
animal. Note that a solid tumor with little or no ascites flu- Sephadex G-25 column to adsorb clogging materials. The
ids production can sometimes occur, as a result of either the amount of Sephadex to use is 1 ml/ml of ascites fluid. The
pristane priming conditions or the low level of secretion Sephadex column should be preequilibrated with 50 mM
that can be characteristic of certain cell lines (65). (When Tris, adjusted to pH 8.6 with HC1. Again, filter the eluant
tumors are obtained, harvest the hybridoma cells from the with a sterile filter (as described above) before subjecting it
peritoneal cavity and suspend them in culture medium con- to further purification (see protein A Sepharose protocols
taining 20% [vol/vol] FCS; store cells frozen in liquid nitro- in the Pharmacia manual).
gen or inject 106cells i.p. into a mouse primed with Freund's 3. Glass wool can also be used to adsorb clogging agents.
incomplete adjuvant.) Dilute ascites fluid samples 1:l with 50 mM Tris, pH 8.6, and
4. Once the abdominal area of the animal begins to en- apply them to a small column made of either syringes or
large, it should be watched closely to avoid overextension of Pasteur pipettes. A small volume may be retained by the glass
the peritoneal cavity and skin. When the bulging of the ab- wool.
domen is deemed to be large enough or accounts for 20% of
the total body weight, the animal is euthanized and ascites 8.4.4.Ab Purification
fluid is drained asceptically with a 20-gauge needle and Since any of the methods described in section 8.4.3 can be
syringe. used to purify MAbs, this section will be devoted to de-
5. Remove the needle and dispense the fluid into sterile, scribing two rapid methods, HPLC and a less expensive
15-ml, conical centrifuge tubes with caps. Sediment the affinity protocol, that have been used successfully to purify
cells by centrifugation at 1,000 x g for 10 min. MAbs of both IgG and IgM isotypes (Fig.1).
8. Antigen-Antibody Reactions 151

0.60 I
L
Alternatively, a protein A Superose column (Pharmacia)
can be used. The MAb purification system buffer from Bio-
Rad could also be used; however, premade buffers are more
0.50 - expensive.
2. Dilute samples ( 1 5 ) with binding buffer, and filter
through a 0.45- or O.ZZ-p,m-pore-size filter if the ascites
fluid has not been pretreated as described previously.
0.40 - 3. Load 0.2 to 1.0 ml of the diluted sample, and run it
r through the column at 5 to 10 ml/min. The flow rate may
be reduced if the column cannot withstand the pressure of
a high flow rate. All proteins that do not bind should elute
within the first 5 min. To remove unbound proteins, run the
column for 5 to 10 min so that the A2a0 remains at the base-
line for at least 5 min.
4. Elute the immunoglobulin fraction with the first elu-
tion buffer (0.1 M citric acid buffer, pH 4, containing 0.15
M NaC1) for at least 5 min at the same flow rate as before.
Repeat with the second elution buffer (0.1 M citric acid
buffer, pH 3, containing 0.15 M NaC1) at 5 to 10 ml/min for
5 min.
5. Wash the column with PBS containing azide, and
store and seal the column in the same buffer. Note that the
eluted immunoglobulin should not be stored in the elution
buffer, as the acidity may be deleterious to the IgM if the Ab
is exposed to it for too long.
Time (min) 6. Dialyze the eluted IgM against PBS (2 4-liter vol-
umes) overnight at 4C.
FIGURE 1 HPLC separation of IgM MAb from ascites fluid. 7. Concentrate to the desired volume by placing the IgM
The first peak represents unbound material, and the second solution into dialysis bags and sprinkling PEG flakes (mo-
peak represents pure IgM Abs eluted after the addition of elu- lecular mass, 20 kDa) over the tubing. This process should
tion buffer 1. The arrow indicates the time at which the elution not take any more than 30 to 60 min.
buffer was added. 8. Assay for total protein content and analyze the sam-
ple by SDS-PAGE to ensure that only the heavy (-55 to 60
kDa)- and light (-28 kDa)-chain bands are seen (Fig. 2).

8.4.4.1. Protein A Figure 1 shows the purification of IgM MAb from ascites
fluid by HPLC using this method.
While protein A has been used effectively to purify IgG
Abs, it generally binds poorly to IgM and murine IgG1.
8.4.4.2. DEAE Affi-Gel Blue
When protein A-Sepharose affinity chromatography is used
to purify IgM, antibody recovery is ca. 14 to 29% (63, 70). The DEAE Affi-Gel blue method is a one-step procedure
In recent years, many companies (e.g., Pharmacia, Bio-Rad, for the isolation of IgG free from protease, nuclease, or al-
and Pierce) have reported that efficient binding between bumin (59). Recovery can be as high as 77 to 80%. (Note
IgM and protein can be accomplished by using buffers of that the DEAE Affi-Gel blue is not the same as the Bio-Rad
high pH and high ionic strength. Pharmacia uses 1.5 M Affi-Gel blue described in section 8.3.5.1.) IgG will be
glycine buffer, pH 8.9, containing 3 M NaC1, while other bound to the DEAE functional groups and therefore must
company protocols are proprietary (e.g., the Bio-Rad MAb be eluted.
purification system buffers). The following procedure can
be used for purifying either IgM or murine IgGl and a vari- Procedure
ation for purifying other isotypes is summarized in Table 4. 1. Preequilibrate a small column containing 5 to 10 ml
of DEAE Affi-Gel blue gel with 20 mM Tris-HC1, pH 7.2.
Procedure 2. Apply 1 to 5 ml of ascites fluid (pretreated as de-
1. Equilibrate a column (Bio-Rad Affi-Prep protein A scribed above). Wash the column with 2 bed volumes of the
preparative cartridge; binding capacity, -35 mg of im- same buffer.
munoglobulin) with binding buffer (see Table 4 for the 3. Wash the column with 1 bed volume of buffer but
recipe) for 12 min at 5 to 10 ml/min (15 column volumes). now containing 25 mM NaCl to elute transferrin. Elute the

TABLE 4 Conditions for HPLC purification of various immunoglobulin isotypes


Isotype(s) of MAb Binding buffer Elution buffer 1 Elution buffer 2
IgM and murine IgGl 1.5 M glycine-3 M NaCl, 0.1 M citric acid-O.15 M NaCl, 0.1 M citric acid-0.15 M NaC1,
pH 8.9 pH 4.0 pH 3.0
Murine IgG2a and IgG3 50 mM Tris-HC1, pH 8.6 0.1 M citric acid, pH 5.0 0.1 M citric acid, pH 4.0
Murine IgG2b 50 mM Tris-HC1, pH 8.6 0.1 M citric acid, pH 4.0 0.1 M citric acid, pH 3.0
Next Page

152 MORPHOLOGY AND ULTRASTRUCTURE

kDa 1 2 3 gella (H Ags), which contain repeated epitopes, and whole


? .
cells, which have large numbers of Ags, are ideal candidates

-
for agglutination reactions. Ags used in agglutination reac-
97.4 - tions can be particulate (e.g., whole cells and Ag-coated
particles; see section 8.2.1).
66.2 - w.u Diagnostic polyclonal antiserum should be extensively
42.7 - adsorbed with a bacterial strain deficient in the target Ag,
e.g., a nonflagellated, unencapsulated, or 0 Ag-deficient
strain (see section 8.3.1). Adsorption reduces nonspecific,
31.0- cross-reactive activity and increases the specificity of the
serum. The adsorbed polyclonal antiserum contains a mix-
ture of Abs of different affinities to different determinants,
21.5- and the combined activities allow detection of an organism
bearing these Ags. The major problems encountered in the
14.4 - use of polyclonal antisera include batch-to-batch variation
. " in Ab quality and residual nonspecific activity. In addition,
while adsorption increases the specificity of the serum,
there is considerable reduction of Ab titer. These problems
FIGURE 2 SDS-PAGE profiles of IgM MAb purification by are virtually eliminated by the use of highly specific MAbs
HPLC. Lanes: 1, molecular mass standards; 2, ascites fluid be- in serotyping with exposed specific epitopes.
fore purification (note the large number of bands that are nor- Despite the high affinities of MAbs, their application
mally found in animal serum proteins or body fluids); and 3 , can be limited due to the fixed affinities of these reagents.
purified IgM MAb eluted from the column (the second major This problem can be overcome by using mixtures of specific
peak in Fig. 1).Note the presence of only two major bands r e p MAbs. They are invaluable in the serotyping of non-
resenting the heavy and light chains at apparent molecular typeable and polyagglutinable bacterial isolates and in the
masses of 60 and 28 kDa, respectively. identification of organisms expressing a specific antigenic
determinant. The choice of polyclonal versus monoclonal
antisera depends on the problem under investigation.
Bacterial agglutination techniques are routinely used for
MAbs with 3 bed volumes of buffer containing 50 mM the detection of Abs to bacterial surface Ags, notably LPS,
NaC1. cell walls, and K and H Ass. Serological classification of
bacteria is based on selective agglutination of the bacterial
cells by Abs to these Ags (74, 76, 79, 80).
8.5. DETECTION OF REACTIONS
Ag-Ab reactions have wide application in all areas of bio- 8.5.1 .I. Qualitative Slide Agglutination
logical, medical, and biochemical research where Abs are Slide agglutination assays can be performed on a slide, in
used as probes for the presence, structure, and even function tubes, or on microtiter plates.
of a given Ag. In many instances, there is a limited amount
of Abs, Ags, or both and the economical use of these
1. Suspend a loopful of bacterial cells (or other particu-
late Ass) in 1.5 ml of normal saline (0.85% [wt/vol] NaC1).
reagents becomes paramount. Furthermore, in diagnostic
labs, screening systems must be easy to use routinely with a
2. Mix 25 ~1 or a loopful of the bacterial suspension with
an equal volume of the diluted test antiserum on a glass
large number of samples yet be rapid, sensitive, and specific.
slide.
The classical serological methods, including immuno-
precipitation, agglutination, immunodiffusion, and comple-
3. Use normal cell suspensions mixed with normal saline
or nonimmune serum as negative controls to detect the
ment fixation, have traditionally been used in diagnostic
nonspecific agglutination of the cells.
laboratories. In recent years, these techniques have been re-
placed by highly sensitive and rapid immunoassay tech-
4. Aggregation should be visible in about 1 min by eye
or by low-power phase-contrast microscopy (chapter 1).
niques such as ELISA and Western immunoblotting. The
reaction mechanisms and application of both classical and Note: (i) Do not use bacterial cells that autoagglutinate
modern techniques are discussed below. in normal saline or water in the absence of an Ab in these
reactions. Minor autoagglutination can be overcome by
8.5.1. Agglutination using 0.1% (wtlvol) skim milk to block adherence or by sus-
Agglutination reactions are among the easiest of immuno- pending the cells in a 1:lOO dilution of normal serum.
logical tests to perform and evaluate and have long been These reagents, when used, should also be used in the con.
used in bacterial identification and serological classification trols. (ii) LPS 0 Ag-deficient strains may agglutinate with
(74,75,76,79,80). Here, the Ag (generally a suspension of more than one serotype-specific serum. (iii) Heat serum at
cells or Ag-coated particles) is mixed with the test Ab, and 56C for 30 min to inactivate complement before testing
a positive reaction is observed as aggregation or clumping for agglutination. (iii) Dilute serum to avoid a prozone ef-
and is termed agglutination. The titer of an antiserum is the fect, i.e., failure to agglutinate at high Ab concentrations.
highest dilution of the serum that gives a visible aggregation
reaction. If the Ag is heterologous to the Ab or if there is 8.5.1.2. Microtiter Plate Agglutination
no Ag present, the suspensions remain unchanged. 1. Plate agglutination allows simultaneous testing of one
For agglutination to occur, the Ag must have more than or more antisera against one or more particulate Ags. Make
one epitope to allow the formation of large aggregates of serial doubling dilutions of the test serum or sera in normal
cross-linked Ag-Ab complexes. Bacterial Ags such as LPS saline or PBS, pH 7.4 (see section 8.5.4), containing 150
and its 0 Ag, carbohydrate-capsular Ags (K Ags), and fla- mM NaC1. To each serum dilution, add 0.005% (wtlvol)
Introduction to Growth 171 13. Energetics, Stoichiometry, and Kinetics
JOHN A. BREZNAK, Editor of Microbial Growth 286
SYED A. HASHSHAM AND SAM W. BAUSHKE
9. Growth Measurement 172
ARTHUR L. KOCH 14. Physicochemical Factors
in Growth 309
10. Nutrition and Media 200 JOHN A. BREZNAK AND RALPH N. COSTILOW
DAVID EMERSON AND JANE TANG
15. Phenotypic Characterization and
11. Enrichment and Isolation 215 the Principles of Comparative
ANDREAS TESKE, HERIBERT CYPIONKA, Systematics 330
JOHN G. HOLT, AND NOEL R. KRIEG BRIAN J. TINDALL, JOHANNES SIKORSKI,
ROBERT M. SMIBERT, AND NOEL R. KRIEG
12. Culture Techniques 270
SYED A. HASHSHAM 16. General Methods To Investigate
Microbial Symbioses 394
TODD A. CICHE AND SHANA K. GOFFREDI
Introduction to Growth
JOHN A. BREZNAK

The study of the growth of bacterial cultures . . . is the stoichiometry, and kinetics of microbial growth. This chap-
basic method of microbiology (1). This quote, excerpted ter reviews the chemical and thermodynamic underpin-
by Gerhardt to introduce this section in Methods for nings of microbial growth wherein materials (elements of
General and Molecular Bacteriology, is as true today as when the substrate[s] and their associated electrons) are con-
written by Jacques Monod nearly 60 years ago. Despite the verted into energy and cell material (biomass), emphasizing
impressive impact that molecular biological methods have the importance of mass and redox balance for obtaining an
made on understanding the phylogenic diversity and phys- accurate interpretation of the energetics of microbial
iological properties of microbes-especially those that have growth on various substrates, or on a given substrate under
not yet yielded to cultivation in vitro-it is the study of the various culture conditions. It ends with a discussion of the
growth of microbes, either in the laboratory or in their nat- kinetics and modeling of batch and continuous (e.g.,
ural habitat, that continues to provide some of the deepest chemostat) culture systems.
insights into the role of particular microbes in the real Chapter 14 by this writer and Costilow required rela-
world. Hence, most of the chapters in this section are con- tively minor revision from the previous edition, as the main
cerned with microbial growth: its achievement, measure- physicochemical factors that affect microbial growth (tem-
ment, and interpretation in the laboratory, as well as ther- perature, pH, water activity, etc.) remain the same.
modynamic relationships and physicochemical factors However, a new addition to this chapter is a discussion of
affecting growth. microaerophilesand methods for growth of oxygen-requiring,
Chapter 9 by Koch has been subject to minor updating but at the same time oxygen-sensitive, microbes under hy-
since the last edition but remains largely intact, reflecting poxic conditions.
the fact that the mathematics of microbial growth, as well The last two chapters are also important new inclusions
as the methods for its measurement, are fundamentally un- in this section. Chapter 15 by Tindall et al. provides an ex-
changed. It remains a cornerstone chapter of this section, tensive description of methods for phenotypic characteriza-
important for beginning microbiologists and seasoned in- tion of microbes. It is an update of a previous chapter by
vestigators as well, and it includes statistical and computer Smibert and Krieg that was included in a section entitled
methods for growth measurement. Systematics in the last edition. However, with reorgani-
In chapter 10, dealing with microbial nutrition and zation of the current edition, and considering that many of
media, Emerson and Tang of the American Type Culture the characterization methods depend on growth of the mi-
Collection (ATCC) update and expand the former chapter crobes in question, this section seemed a logical place for it.
by Cote and Gherna (also of the ATCC). Retained is an A n important addition to this chapter is a discussion of
overview of macro- and micronutrient requirements of mi- principles of comparative systematics, thereby providing an
crobes and a description of defined and undefined media intellectual context within which the results of phenotypic
for cultivation of metabolically diverse microorganisms, characterization tests can be interpreted effectively.
from autotrophs to heterotrophs, and including both Chapter 16 by Ciche and Goffredi is new to this edition
chemotrophs and phototrophs. Also retained are methods and describes general methods to study the growth and ac-
for sterilization of media and medium components, includ- tivities of microbes on, or within, other organisms, i.e., mi-
ing heat-labile components. In chapter 11, Teske and crobial symbioses. In light of the fact that microbial sym-
Cypionka update the former chapter on enrichment and bioses are extremely diverse and include microbial
isolation written by Holt and Krieg. Notable additions in- associations with animals, plants, and even other microbes,
clude methods for enrichment and isolation of iron- and this chapter focuses on general methods to establish that a
sulfide-oxidizing bacteria by using gradient culture systems, symbiosis is actually occurring (i.e., a Kochs postulates
as well as methods for extinction and high-throughput cul- approach to verification); methods to characterize envi-
turing, single-cell isolation, and rRNA-based molecular ronmental symbioses, wherein one or more of the micro-
methods to follow sought-after microbes from environmen- bial partners, or the host, have not yet been cultured indi-
tal samples through enrichment to pure culture. vidually in vitro; and model symbiotic systems, wherein the
In chapter 12 on culture techniques, Hashsham has re- partners not only can be individually cultured in vitro but
vised, updated, and merged previous chapters by Krieg and also are amenable to genetic manipulation. The emphasis
Gerhardt (solid, liquid/solid, and semisolid culture) and here is on some well-studied animal-microbe symbioses as
Gerhardt and Drew (liquid culture) into one comprehen- examples.
sive and unified treatment encompassing batch, continu-
ous, and specialized (e.g., fed-batch, immobilized-cell) cul- REFERENCES
tivation at the laboratory scale. Hashsham and Bauschke
follow this in chapter 13 with one of the most important 1. Monod, J. 1949. The growth of bacterial cultures. Annu.
new additions to this section: a discussion of the energetics, Rew. Microbiol. 3:371-394.

171
4
.

Growth Measurement
ARTHUR L. KOCH

9.1. PRINCIPLES .............................................. 173


9.1.1. Definitions of Growth ................................... 173
. 2. Balanced Growth ....................................... 173
. 3. Changes in Cell Composition at Different Growth Rates.......... 174
. 4. Unbalanced Growth .................................... 174
. 5. Pitfalls in Growth Measurement ............................ 174
. 6. Mycelial Growth ....................................... 174
. 7. Cell Differentiation..................................... 175
. 8. Cell Adsorption to Surfaces............................... 175
. 9. Growth in Natural Environments ........................... 175
9.2. DIRECT COUNTS ......................................... 175
9.2.1. Microscopic Enumeration ................................. 175
. 2. Electronic Enumeration .................................. 176
. 3. Flow Cytometry ....................................... 177
9.3. COLONY COUNTS ........................................ 177
9.3.1. Diluents ............................................. 178
. 2. Spread Plates.......................................... 179
. 3. Layered Plates ......................................... 180
. 4. Pour Plates ........................................... 181
9.4. MOST PROBABLE NUMBERS ................................ 181
9.5. BIOMASS MEASUREMENT .................................. 182
9.5.1. Wet Weight ........................................... 182
. 2. Dry Weight ........................................... 183
. 3. Water Content ........................................ 183
.4.Volume .............................................. 183
.5. Wet and Dry Densities ................................... 183
9.6. LIGHT SCATTERING ...................................... 184
9.6.1. Turbidimetry .......................................... 184
9.6.1.1. Procedure...................................... 186
.
.2 Calibration..................................... 186
. 3. Klett-Summerson Colorimeter....................... 188
9.6.2. Nephelometry ......................................... 188
9.7. STATISTICS. CALCULATIONS. AND CURVE FITTING ........... 189
9.7.1. Population Distributions ................................. 189
. ........................................ 190
2. Statistical Tests
. 3. Error Propagation ...................................... 190
. 4. Ratio Accuracy ........................................ 191
. 5. Coincidence Correction .................................. 191
. 6. Exponential-Growth Calculations ........................... 191
. ........................
7. Plotting and Fitting Exponential Data 192
. 8. Least-Squares Fitting.................................... 193
9.7.8.1. By Pocket Calculator ............................. 193
9.7.9. Nonexponential Growth ................................. 195
9.8. REFERENCES ............................................. 197
9.8.1. General References ..................................... 197
. 2. Specific References ..................................... 197

Growing bacterial cultures and knowing their growth rate cessfully does without. However. it is truly needed for critical
and density of growth are essential for microbial physiology. work in microbial physiology and hence for much of molecu-
But obtaining this knowledge is not simple and is painstak- lar biology. This circumstance means that much of the work
ing. Moreover. it is o1d.fashioned . Much modern biology and techniques presented here are old hat and the present
avoids this care. and about half of the molecular biology SUC- chapter is anachronisticand is archaeology. but it is essential.

172
9. Growth Measurement w 173

9.1. PRINCIPLES nificantly, sooner or later the bacteria will come to achieve
Principles of bacterial growth are also discussed in chapter a stable growth state characteristic of any particular con-
13 of this volume and in references 1 through 8. stant environment no matter what the condition of the cell
was initially. Once this balanced growth state has been
9.1 .I. Definitions of Growth achieved, if the conditions remain the same the culture will
To measure bacterial growth, precise definitions are needed. remain in balanced growth indefinitely (until a nutrient is
Probably the most basic definition of growth is based on the exhausted or the culture is altered by mutation and selec-
ability of individual cells to multiply, i.e., to initiate and tion). The criteria given above are too stringent to be fully
complete cell division. This definition implies monitoring met, but it is readily possible to study cultures that are sub-
the increase in the total number of discrete bacterial parti- stantially held in balanced growth by maintaining growing
cles. There are two basic ways to do this: by microscopic cultures at low density by dilution and by using only a sin-
enumeration of the particles or by electronic enumeration gle measure (such as biomass measured turbidimetrically) to
of the particles passing through an orifice. This definition monitor growth. If the doubling time remains constant over
would also falsely include the fragmentation of nongrowing an extended period, it is a good and useful assumption that
filamentous organisms as growth, although this kind of ap- growth is balanced.
parent growth cannot continue indefinitely. Consider any extensive property (such as biomass, pro-
A second definition of growth involves determining the tein, DNA, or RNA) of a culture in balanced growth and
increase in CFU. Since some cells may be dead or dying, call this property X.The rate of formation of X will be pro-
this definition of growth may be different from the one portional to the amount of biomass, m. Call the propor-
based on the detection of discrete particles. In the long run, tionality constant C1. Then,
the increase in the number of organisms capable of indefi- dX/dt = Clm
nite growth is the only important consideration. This is the
reason why colony counting and most-probable-number Because growth is balanced, X/m is constant; call this con-
(MPN) methods of measurement are so important. Viable- stant Cz. Then,
counting methods, which seem so natural to a microbiolo- dX/dt = (Cl/Cz)X
gist, are really quite special in that cultures are diluted so
that individual organisms cannot interact. For example, where (C&) is a proportionality constant (equal to the
these methods cannot in principle be applied to obligately specific growth rate, most usually designated by p). Through
sexually reproducing organisms requiring male-female in- the laws of calculus, this equation can be integrated and a
teraction or to colonial organisms such as myxobacteria boundary condition (X = Xo when t = 0) can be imposed,
that under certain conditions need to be part of a large mass yielding
of organisms that produces a sufficient amount of exoen- X =X 0P
zyme. Even when applied to procaryotes, there are special
restrictions and limitations; e.g., exogenous C02 must be That is, for every cellular substance or even an extracellular
available in sufficient concentration, although it need not product of the cells, after growth becomes balanced the in-
be supplied if many organisms are present (see chapter crease of X will be exponential in time. If the ratio of one
6.5.4). substance to every other substance is to remain constant,
A third definition of growth is based on an increase in the same proportionality constant must apply for every
biomass. Macromolecular synthesis and increased capability choice of X. This common value of p is called the specific
for synthesis of cell components are the obvious basis for growth rate or growth rate constant. Any substance that
the measurement of growth by the bacterial physiologist, you wish to choose and call X will increase exponentially,
the biochemist, and the molecular biologist. From their but also any combination of substances, or, indeed, the rate
point of view, cell division is an essential, but minor process of change of any substance will increase exponentially with
that seldom limits growth; what usually limits growth is the the same specific growth rate, p. Practically, this allows the
rate at which enzymatic systems utilize resources to form rate of oxygen uptake or the production of a metabolite to
biomass (cytoplasm). be used as an index of growth.
A fourth definition of growth is based upon the action of Intensive properties, such as p and the ratio of the con-
the organisms in chemically changing their environment as centrations of different cell constituents, must necessarily
a consequence of the increase in biomass. remain constant under conditions of balanced growth. In
addition, the distribution of cell sizes stays constant. A n av-
9.1.2. Balanced Growth erage cell will have a constant rate of carrying out every cel-
The four definitions of growth mentioned above become lular process, and newly arisen daughter cells will have a
synonymous under a single circumstance: balanced growth constant probability of being able to form a colony (i.e., the
(17). A n asynchronous culture can be said to be in balanced percentage of nonviable cells will remain constant).
growth when all extensive properties increase proportion- Therefore, under these special conditions, no matter what
ally with time. Extensive is a term from physical chem- measure of growth is used (whether it is particle counting,
istry and refers to the properties of the system that change colony formation, chemical determination of a cell sub-
when there are altered amounts of substances of various stance, consumption, or excretion of a substance), the same
kinds in the system. Thus, biomass, DNA, RNA, and cell specific growth rate will be obtained and that rate will be
number are extensive properties of a culture in a fixed vol- constant through time in a constant environment.
ume, but temperature or constituent ratios (such as DNA or Such favorable conditions in principle occur when bac-
RNA per cell) are intensive properties that do not change teria are cultivated under long-term continuous culture.
during balanced growth. The application of this physico- Balanced growth should arise during chemostat growth that
chemical principle to bacteriology lies in the thought that is limited by the rate of addition of a single nutrient and
if a culture is grown for a long enough duration when during turbidostat growth when the culture is mechanically
growth is sparse enough not to alter the environment sig- diluted to maintain a constant amount of biomass in the
174 GROWTH

growth vessel and growth is limited by the nature of nutri- habit of growth. This can occur even under mildly toxic
ents and not their amount. Balanced cultures can be formed conditions with bacteria that ordinarily divide regularly.
by repeated dilution under batch growth (6, 62), if carried The second pitfall is the differential viability of injured bac-
out in such a way that the culture never goes into the lag teria under different culture conditions. Repair processes
phase or into the stationary phase. may permit the recovery of viable cells under some but not
other conditions (11). A third major pitfall is the possible
9.1.3. Changes in Cell Composition development of resistant stages. Bacteria known to form re-
at Different Growth Rates sistant stages, such as spores, pose no problem since the con-
Organisms respond to environmental conditions, both trols and measurements to correct for such forms are well
physical and chemical, by altering their own composition. known; but when resistant stages are not suspected, error
These changes have been well documented for certain en- can arise. The fourth pitfall pertains to the way in which
teric bacteria (37, 46) but may occur with all procaryotes. the inoculum is exposed to the new environment. Different
In general, favorable growth conditions mean faster growth, results may be obtained if the concentration of an agent is
which requires a higher concentration of ribosomes and as- raised gradually, if it is raised discontinuously, or if a high
sociated proteins. In terms of gross composition, the most concentration is temporarily presented and then removed
obvious change is an increase in RNA content. Also, under or lowered. The cell concentration at the time of challenge
favorable growth conditions the cells can lay down reserve can be critical. Bacteria may have special ways, sometimes
materials such as glycogen and poly-P-hydroxybutyric acid. inducible and sometimes unknown, to protect themselves
These changes in composition lead to the possible pitfall of against toxic agents. The protection may be dependent on
the microbiologist falsely relating one measure of growth to the number of organisms cooperating in detoxification.
others when comparing growth in different environments. The phenomena related to the fourth pitfall can be illu-
minated with a single example from work on the interac-
9.1.4. Unbalanced Growth tion of rifampin and Escherichia coli (42). Rifampin is a large
Although balanced growth conditions lead to reproducible molecule that can only very slowly enter the cell. Doses of
cultures, much of physiology deals with the responses of or- rifampin can be detoxified by a sufficiently large number of
ganisms to changes in their environment that lead to pro- cells, and the process appears to be inducible. Therefore,
gressive changes in the organism during the ensuing un- higher doses of drug can be tolerated when the cells have
balanced growth. When a stationary-phase culture is previously been gradually exposed to the antibiotic.
inoculated into fresh medium, the properties of organisms Independent of this phenomenon, fast-growing bacteria are
change drastically through the course of the batch culture more resistant than slow-growing ones. This observation
cycle. Though only well documented for certain enteric can be explained largely by the shorter time available for
bacteria (1, 14), similar changes probably apply with all the drug to penetrate relative to the doubling time of the
procaryotes. The exact sequence of changes in composition growing bacteria. For example, under conditions where the
and morphology depends on the medium and on the age low dose by itself would not inhibit growth, a higher dose of
and condition of the inoculum. Culture cycle phenomena antibiotic that is subsequently diluted can block growth.
have relevance to growth measurements. These phenomena Thus, when growth is slowed by the brief high dose, the in-
are particularly important in ecological studies, where the hibition remains permanent because growth is slowed and
conditions under which organisms grow are critical and, to now sufficient time is available to allow penetration of
a large degree, uncontrollable. The changes in characteris- enough drug to maintain the blockage of growth.
tics of cells are also involved in response to the fluctuations
in natural conditions (37,40). 9.1.6. Mycelial Growth
A typical bacterial culture cycle progresses as follows. The evaluation of mycelial growth (the first pitfall) can be
When a stationary-phase culture is diluted into rich easier and also can be much more difficult than is the eval-
medium, macromolecular synthesis accelerates. The com- uation of well-behaved organisms that engage in binary
ponents of the protein-synthesizing system (i.e., ribosomal fission and then promptly separate. The problems and
proteins and RNA) are made first. Only after considerable methods for mycelial growth have been discussed by Calam
macromolecular growth does cell division take place. (16) and Koch (39) and are briefly dealt with here. Filtering
During this lag phase, the average size of cells increases filamentous cells, with or without drying, is easier than fil-
greatly. Later when the capacity of the medium to support tering smaller nonfilamentous cells. In addition, the in-
balanced exponential rapid growth is exceeded, cellular crease in the physical size of a mycelial colony of filamen-
processes slow down. Different processes slow differentially tous cells can be monitored. In the extreme case, a tube 1 m
in a way to produce small, RNA-deficient cells. Finally, the long containing a layer of nutrient agar is inoculated at one
cells largely die (20) and may eventually lyse. end, and the mycelial mass grows along the tube and may
Workers using the techniques from this book may em- even be continued successively into other tubes (59). Such
ploy intentional perturbations of growth. These perturba- colonial growth is essentially one-dimensional, whereas
tions can result from nutritional shifts or deficiencies, the growth of mycelial colonies on agar surfaces is two-
use of growth inhibitors (particularly antibiotics), or the ef- dimensional. In liquid shake cultures (chapter 12.2.2)
fects of radiation or other extreme physical conditions (e.g., mycelial growth is three-dimensional. Under all three con-
high or low temperature and osmotic pressure). Their ef- ditions the linear dimensions increase linearly (not expo-
fects on growth can be quite complex, and the bacteriolo- nentially) with time because of the nutrient limitation
gist must be both cautious and critical. created by the cells.
The rate of increase in size of the colony in one, two, or
9.1.5. Pitfalls in Growth Measurement three dimensions depends on the rate of elongation of the
There are four classes of pitfalls in bacterial growth mea- terminal hyphae that happen to be growing on the surface
surement. The first major pitfall is the tendency of most or- out from the edge of the colony, but the mobilization of re-
ganisms toward either clumping or a having a filamentous sources into the mycelial mass depends on the surface area
9. Growth Measurement 175

of the colony. Therefore, with shake cultures particularly, under the fluorescence microscope. Fifth, sophisticated
the results depend on the nature and size of inoculated frag- mass spectrometry of the gases emanating from natural soil
ments; with large fragments, growth becomes limited at an samples has been used to provide information about the
earlier stage by diffusion of nutrients into the mycelial mass. growth of organisms within the sample (51). Sixth, agents
Thus, the major pitfall with the mycelial habit of growth is that block cell division but not enlargement have been
that the growth quickly deviates from exponential and de- used. Cells that become bigger under such conditions can
pends on the geometry of growth and the nature of the in- be considered to have been alive.
oculum. In shake cultures, the apparent growth may depend
on the shape of the vessel and on the shaking speed, its
character (circular or reciprocal), and the distance moved, 9.2. DIRECT COUNTS
because all of the above can affect the tendency of the
mycelia to break into smaller pieces. 9.2.1. Microscopic Enumeration
Microscopic enumeration in a counting chamber is a com-
9.1.7. Cell Differentiation mon technique that is quick and cheap and uses equipment
The change of enteric bacteria from large RNA-rich forms readily available in the microbiological laboratory.
in the exponential phase to small RNA-poor forms in the Microscopic direct counts can also be made by membrane
stationary phase has many of the aspects of differentiation. filter sampling and staining (chapter 2).
Microbiology, however, has much clearer examples of cell The counting chamber technique is subject to errors, but
differentiation in the cases of transition of a rod to a coccus, these can be overcome to a large degree by using improve-
of a vegetative cell to an endospore, and in the formation of ments suggested by the work of Norris and Powell (53). The
exospores, cysts, buds, and prosthecae. The tendency to major difficulty in direct microscopic enumeration is the re-
form filaments in certain circumstances can also be consid- producibility of filling the counting chamber with fluid. In
ered a differentiation. These changes and their reversal pose the technique recommended below, the thickness of the
potential pitfalls for all approaches to growth measurement. fluid filling is measured by focusing on bacteria attached to
the top and bottom interfaces and measuring the distance
9.1.8. Cell Adsorption to Surfaces between them with the micrometer scale on the micro-
Many bacteria naturally adhere to certain surfaces or can scope. The horizontal dimensions between the scribe marks
adapt and mutate to achieve a high avidity for solid surfaces in commercial chambers are quite accurate and cause no
including glass. Many experiments with chemostat culture problem.
have failed to achieve their primary goal because the or- A second major difficulty is the adsorption of cells on
ganisms adhered to the vessel walls. Therefore, plastic or the surfaces of glassware, including pipettes. The procedure
Teflon should be used whenever there is long-term contact described below avoids adsorption during the dilution
of the organism with a culture vessel (37). Other ap- process but desirably encourages it in the counting chamber.
proaches to minimize the effects of growth on vessel walls In the dilution process, it can be decreased by carrying out
include the use of large culture volumes in large containers, dilutions in high-ionic-strength medium (e.g., physiological
the use of violent agitation, and the frequent subculture of saline or many minimal media without their carbon source)
the cells into fresh glassware. In addition, the use of deter- instead of water or in a solution of formaldehyde (any con-
gents, vegetable oils, silicone coatings, and a high-ionic- centration between 0.5 and 5% is satisfactory) that has
strength medium may to some degree alleviate this problem. been neutralized with K2HP04together with a trace of an-
This problem is particularly pertinent during dilution of ionic detergent such as sodium dodecyl sulfate (53). The
the culture for measurement by highly sensitive means, such formaldehyde stops growth and motility, and the K2HP04-
as microscopic and plate counts. It is therefore given further detergent combination prevents aggregation, but the deter-
consideration in the discussions of those methods in sec- gent may lyse certain organisms even though it is used only
tions 9.2.1, 9.3.1, and 9.3.2. in minute amounts. Alternatively, plastic containers and
Currently, a great deal of research is being devoted to the plastic pipette tips can be used for the dilution to decrease
study of biofilms. This is the flip side of what was said above the loss by adsorption on surfaces. In the procedure de-
since many organisms do bind to surfaces and many surfaces scribed below, 0.1 N HC1 is used as the final diluent to favor
in nature have communities with multiple species. On adsorption on glass surfaces of the chamber.
many of these surfaces, polysaccharides are present, secreted The Hawksley counting chamber (A70 Helber;
by the cells. These help bacteria adhere. Hawksley, Ltd., Lansing, England) is recommended in pref-
erence to the Petroff-Hausser counting chamber, and there
9.1.9. Growth in Natural Environments are other varieties from other suppliers that will serve; for
If the problem at hand is to measure growth under natural example, Preiser Scientific, Inc, or Arthur H. Thomas Co.,
conditions, tremendous difficulties must be overcome (12). Philadelphia, PA. The prime advantage of the former is that
In nature, growth almost always involves mixed cultures its optical path with an ordinary coverslip is short enough
with bacteria attached to each other or to solid particles. that the chamber can be used under an oil-immersion ob-
Six kinds of approaches have been used to deal with the jective at high power. Although most counts are done under
measurement problem in these cases. First, 14C02fixation a high-dry objective, it is sometimes necessary to use the
has been used (72) to monitor biomass increase. Second, oil-immersion objective, either because there are too many
tritiated thymidine autoradiography has been used to iden- cells or because they tend to clump. Both chambers are 20
tify cells engaged in DNA replication (13). Third, antibod- pm deep with scribe marks defining counting areas of 50 by
ies (60) or oligonucleotide probes (see chapter 39) tagged 50 pm.
with fluorescent labels have been used to scan soil samples One major advantage of microscopic examination is that
and identify particular organisms (60). Fourth, nucleic acids one can gain additional information about the size and mor-
within cells have been stained with DAPI (4,6-diamidino- phology of the objects counted. Oil-immersion and high-
2-phenylindole) and acridine orange and then observed power objectives make counting more tedious, but critical
176 GROWTH

distinctions can be made. The manufacturers of chambers total bacteriacounted x dilution factor x 4 x 10'
supply alternative procedures. Additional discussion has
been presented by Meynell and Meynell (6) and Postgate number of small squares x filling depth (in micrometers)
(58). For a discussion of statistical considerations, see sec- If the coverslip is precisely positioned, the volume of the
tion 9.7. filling on top of a small square is 50 X 50 X 20 pm3 = 5 X
Procedure lo4 pm3 = 5 x lo-@ml. The reciprocal of this, 2 x 107/ml,
is the usual factor in the formula quoted in the instructions
1. Clean the chamber and the coverslip with water con- supplied by the manufacturer; thus, the two formulas are the
taining a small amount of anionic detergent; rinse with same if the filling depth is exactly 20 p,m. For the procedure
water and then alcohol, blot, and let air dry. to be successful, only a few cells need be attached to each
2. Make a preliminary estimation of the concentration of surface for the thickness measurement. It is most conven-
cells. Proceed to the next step if the concentration is less ient, however, if most of the cells are on one surface for
than 3 x 10" cells per ml. Otherwise, make a primary dilu- counting. Then, the final act of counting a small square is
tion of the cell suspension in Norris-Powell diluent ( 5 1 ) the focusing through the suspension to count the cells not
prepared as follows. Add 5 ml of formalin (37% formalde- attached and the ones on the upper interface that has fewer
hyde) to 1 liter of water. Adjust to pH 7.2 to 7.4 (indicator cells. Then, tally the square, refocus on the original surface,
paper is sufficiently accurate) by adding solid K,HP04; the and move on to the next square.
amount of the phosphate needed will vary with the amount
of formic acid in the formalin. Add a few milligrams of
sodium dodecyl sulfate, and repeat until bubbles do not 9.2.2. Electronic Enumeration
break immediately when air is passed through the solution The Coulter Counter (Coulter Electronics, Hialeah, FL),
with a Pasteur pipette. its commercial competitors, and particularly the laboratory-
3. Carry out a final single dilution of the sample in a built versions have been important in the development of
ratio of at least 1:l with 0.1 N HC1. This will kill the cells, bacteriology over the last 40 years. Such instruments are
had the formaldehyde not sufficed, and gives their surface a used routinely in clinical hematology. They are also very
net positive charge so that they will not aggregate but will useful in the enumeration of nonfilamentous yeasts and pro-
instead adsorb onto glass. tozoa but not of mycelial or filamentous organisms.
4. Immediately fill the hemocytometer counting cham- Although use of these counters has led to important con-
ber with approximately 5 pl of the diluted sample, using a cepts in bacteriology, the technique is difficult to apply in a
Pipetteman, Eppendorf, Centaur, or other plastic-tipped valid way to estimate the volume of bacteria because of
pipette. Let the chamber stand for 1 to 2 min. their small size and usually elongated shape. Attempts to
5. Examine with a phase-contrast microscope under a improve and validate the technique for use in bacteriology
high-dry or oil immersion objective. Most of the cells will have been made at the research level by people with back-
have attached to the bottom interface; a few cells will have grounds in physics or engineering. Evidence of the difficul-
attached to the top interface; and only a few cells will re- ties involved is the fact that many of the people who helped
main suspended and exhibit Brownian motion. develop the technique for measuring distribution of cell
6. First, focus on the cells that have attached to the bot- volumes no longer use it. This article, therefore, only pre-
tom interface, and read the markings on the dial of the fo- sents the .principles, mentions the difficulties and the at-
cusing knob. O n many microscopes of high quality, this dial tempted solutions to these difficulties, and directs the
reads directly in micrometers. Next, focus on the cells on reader to published literature. Then, the reader will be able
the top interface. Note the distance between the top and to consider the applicability of the technique to a specific
bottom, and augment the difference by one bacterial diam- bacteriological problem.
eter. This total distance will quite accurately measure the The principle of electronic enumeration is as follows. A
filling, which is nominally 20 pm. When gaining familiar- fixed volume of a diluted cell suspension is forced to flow
ity with the technique, make depth measurements in sev- through a very small orifice connecting two fluid compart-
eral well-separated regions of the chamber to find the uni- ments. Electrodes in each compartment are used to measure
formity of the depth of filling. the electrical resistance of the system. Even though the
7. Count the cells lying within small squares. Optimally, medium conducts electricity readily, the orifice is so small
the number in each small square should be in the range of 5 that its electrical resistance is very high. Consequently, the
to 15. Score the cells that lie on the boundaries of a square electrical resistance of the rest of the electrical path is neg-
if they are on the upper or right side but not if they are on ligible by comparison. When a cell is carried through the
the lower or left side. A hand tally counter is convenient to orifice, the resistance further increases since the conductiv-
count the cells within a square. A second hand tally is con- ity of the cell is lower than that of the medium. This change
venient to keep track of the number of squares. At least 600 in resistance is sensed by a measuring circuit and converted
total organisms should be counted for accurate work (see into a voltage or current pulse. An electronics circuit simi-
below), but this need not be done with a single filling. Some lar to that used in counting radioactivity counts the pulses.
authors recommend multiple fillings of the chamber, thus A discriminator circuit eliminates very small pulses. A n
averaging the variability of the fillings; however, this is not upper discriminator eliminates very high pulses, which
necessary with the method described here, because the might be due to dirt or other irrelevant particles. In ad-
thickness of the filling is measured each time. It is best to vanced models, the pulses may be analyzed by size and the
count squares chosen in a systematic fashion, such as the pulse sizes may be stored in a multichannel analyzer. Later,
four corner squares and the major diagonal squares. This the data are recovered and plotted in a histogram, and the
prevents counting the same square twice and averages a numbers, mean size, and standard deviation are calculated.
possible geometric gradient of cells in the chamber. All the data may be collected, and the discrimination
8. Calculate the number of cells per mi of undiluted cul- against pulses that are too high or too low is carried out as
ture by use of the following formula: the data are analyzed. The instrument needs some method
9. Growth Measurement 177

of forcing an accurately known volume through the orifice pulse generated by a cell passing through an orifice.
during the counting period. This is usually done by displac- Attempts have been made to overcome this difficulty by
ing the fluid in contact with a mercury column past trigger- using a relatively long pore (100 pm) (43), but the result-
ing electrodes that conduct effectively through the mercury ing slower flow and longer path increase the chance of clog
but not through the diluent medium. ging. A second approach involves special hydrodynamic fo-
There are three major problem areas in electronic enu- cusing of solutions in such a way that the bacterial cells pass
meration. First, some bacterial cells are very small (less than very nearly down the center of the pore surrounded by fluid
0.4 pn3),and the resistance pulses produced as they pass containing no particles (63,65, 75).
through the orifice are comparable to the noise generated Additional information about this technique can be
by the turbulence that develops in the fluid flowing through found in references 2, 22, and 44 and in instruction manu-
the orifice. The discriminator dial on the instrument can be als for the instruments.
set to reject the turbulence noise, but then sample informa-
tion, particularly about newly divided cells, may be lost. 9.2.3. Flow Cytometry
Blanks can be run, and their values can be subtracted, but Flow cytometry has become an extremely powerful method
blanks are particularly variable for small cell sizes. In addi- for the studies of many aspects of the biology of eucaryotes,
tion, a pattern of turbulence can become established, re- but the methods are only now coming into their own in the
main for a while, and then be replaced with another pat- study of the biology of procaryotes. The delay was simply
tern. Finally, the overall error increases when the blank has because the latter are smaller and require more develop-
large statistical variation (section 9.7.3). There is little ment. The literature is extensive (50,55,71). The flow cy-
problem in the study of bacteria growing in rich medium in tometry instruments that are now common in hospitals and
which even the newly formed cells are larger than the research laboratories operate by forming a small-diameter
pulses produced by the turbulence or if interest is restricted stream of the sample suspension. They were originally de-
to the relatively few large bacteria such as Azotobacter agilis signed for applications in immunology but now are com-
and Lineola longa. mercially available from several manufacturers. The sample
The second major problem in electronic counting results suspension is encased in a stream of fluid that is added in
from the failure of cells to separate promptly from each such a way that the stream of the sample is made still nar-
other after cell division. This, and the tendency to form fil- rower in diameter. The flowing stream is examined with
aments and aggregates, can be minimized by careful choice laser light of various frequencies and angles, and the output
of the organism and the conditions. The choice of E. coli for of the measurement circuits is used to detect when a parti-
physiological and genetic studies was fortuitous because of cle passes through. The design permits the analysis of bio-
the relatively small extent to which it remains as pairs or mass by light-scattering methods and by staining of chemi-
chains or forms aggregates. Various physical techniques cal components such as DNA with fluorescent dyes. The
such as mild ultrasound treatment or vigorous blending in a electronic circuits allow cells to be counted very rapidly.
Vortex mixer can be used to try to separate pairs, disperse Growth can be monitored by measuring the increase in
aggregates, or break up filaments. There is a related problem counts in sample counted for a fixed duration corresponding
of coincidence (section 9.7.5), i.e., the passage of more than to a fixed volume. The problems for the application to pro-
one cell through the orifice in a short enough time that a caryotes are in sensitivity and background noise. General
single larger cell is registered by the electronics. This prob- applications in microbiology are described in references 23
lem can be dealt with (see below) by increasing the dilution and 61. Commercially available equipment has been used to
so that the probability of coincidence is altered. study a variety of problems in ecology (15). Other applica-
Coincidence is an especially vexing problem when cell size tions are described in references 8 and 52.
distributions are to be accurately measured. Another instrumental approach (66) depends on flow
The third major problem in electronic counting is clog- directly on a microscope slide on an inverted microscope. It
ging of the orifice. The resistance change is a smaller pro- was developed in Norway by Steen and has the property
portion of the total orifice resistance when the orifice di- that it is much cheaper and has been adapted to microbio-
ameter is larger. Consequently, for small rod-shaped logical applications to access the level of many cellular
bacteria such as E. coli, orifices with diameters in the range functions.
of 12 to 30 pm must be used. The exact choice is a trade-
off between an increased signal and the increased noise and
chance of becoming clogged. Clogging is best prevented by 9.3. COLONY COUNTS
ultrafiltration of all reagents. Alternatively, the diluent can Bacteriology really became an experimental science when
be prepared and allowed to settle for a long time (months) Robert Koch listened to Fannie Hesse and developed the
in a siphon bottle so that the particulate-free solution can agar plate. This allowed not only the cloning of pure strains
be withdrawn away from the bottom. Choosing solvents but also the enumeration of colonies arising from individual
that do not tend to generate particulate matter can also be viable cells or CFU. Various colony count methods have
of help. Kubitschek (44) recommended 0.1 N HC1 for this been used: (i) pour plates, in which an aliquot sample of di-
reason; moreover, it is entirely volatile and does not leach luted cells is pipetted into a n empty sterile petri dish,
materials out of the glass, which later may form precipitates. molten but cool (45C) agar medium is poured onto the
However, the problems in practice are severe and become sample, and the contents are mixed by swirling and then al-
worse in some of the modifications needed to size bacterial lowed to harden; (ii) spread plates, in which the sample is
cells more accurately. pipetted onto the surface of solidified agar medium in a
Electronic counting has influenced the study of bacterial petri dish and the cells are distributed with a wire, glass, or
growth more because of its presumed ability to measure the Teflon spreader; (iii) thin-layer plates, in which the sample
size distribution of bacterial cells than because of its ability is pipetted into a tube containing a small volume (2.5 to 3.5
to enumerate them. In fact, it is very difficult to measure ml) of molten but cool soft (0.6 to 0.75%) agar medium, the
cell size accurately because of the nature of the resistance mixture is poured onto hardened sterile agar medium in a
178 GROWTH

petri dish, and this overlay is allowed to harden; (iv) lay- Certain organisms are very sensitive to substances pres-
ered plates, which are like the thin-layer plates except that ent in agar. Meynell and Meynell(6) presented an excellent
an additional layer of agar medium is poured onto the newly discussion of these problems. Injured organisms may have
congealed soft-agar medium containing the cells so that all additional special requirements, and the entire 26th sympo-
colonies are subsurface; and (v) membrane filter methods, sium of the Society for General Microbiology (25) was de-
in which the diluted cells are filtered onto an appropriate voted to these problems. Genetically defective organisms
presterilized membrane filter, which is then placed on an pose their own individualistic problems that can be research
agar medium plate or onto blotter pads containing concen- problems on their own, e.g., the ability of various repair mu-
trated liquid medium. Sometimes it is necessary to carefully tants to form countable colonies (18).
prewash the membranes and the pads with water or medium The problem of quantifying the number of organisms in
and then resterilize them. cultures of strict anaerobes is dealt with in references 19,30,
The pour plate methods have variations in which the and 31 and in chapter 14.
cells are grown in roller tubes or microtubes (56) and are
examined with low-power microscopes when the colonies 9.3.1. Diluents
are small (57). There are many individual variations or Bacterial cells often must be diluted from their original
techniques, sometimes resulting from historical accidents dense concentration to a sparse concentration suitable for
and sometimes resulting from special bacteriological cir- observation in a microscope, measurement of cell numbers,
cumstances. Automation of colony counting has put addi- analysis for genetic or metabolic properties, or washing
tional special restrictions on techniques but allows colonies preparatory to study. Whatever the use, the diluted cells
on petri dishes to be counted rapidly with reduced operator must retain their original characteristics. The preservation
error. of viability and metabolic activity is particularly important
Two colony count methods are detailed below, and an- when measuring the CFU. Diluted cells are more likely to
other is described briefly. The first is the spread plate, in be harmed by an unfavorable environment than are cells in
which all colonies are surface colonies. It is chosen for pres- dense suspensions, in which the environment contains
entation because it is reliable and because surface colonies higher levels of materials leached from the cells. Con-
are required to produce the proper color responses with sequently, care must be taken to use a suitable diluting
many indicator agars. In many cases, different colors are solution.
given from subsurface colonies because the oxygenation is Distilled or tap water alone should usually not be used,
different; therefore, the acid production and reducing po- because they are osmotically hypotonic to all bacterial cells
tential are different from those on the surface. and unbuffered against pH change. Traces of heavy-metal
The second method is the layered plate. It is very useful contaminants and detergents in water supplies may also
because all colonies are subsurface and therefore much cause inactivation of cells in very dilute suspension.
smaller and compact. They can be intensely colored. Many Physiological saline (0.85% NaC1) also is usually inade-
more colonies may be present, and yet the coincidence by quate because it is isotonic only to mammalian cells and is
fusion of colonies is small; this means that several thousand unbuffered. Viability may be reduced by 50% or more when
colonies per plate can be used to give statistically more such diluents are used.
meaningful results. This approach is especially recom- A common general-purpose diluent is phosphate-
mended because the main difficulty with usual colony- buffered saline with a small amount of protein, usually gel-
counting methods is the lack of dynamic range. Rules have atin or peptone. The presence of the protein stabilizes frag-
been issued that between 30 and 300 colonies are required. ile bacteria by binding metals and detergents, and the
The lower limit is set by statistical accuracy, and the upper diluent is buffered at neutrality by the phosphate salts.
limit is set by coincidence limitations. This 10-fold range is Buffered saline with protein contains 8.5 g of NaC1,0.3 g of
inconvenient for many purposes, because in many cases one anhydrous KHZPO,, 0.6 g of anhydrous NalHP04, and 0.1
cannot predict the number within a factor of 10 when g of gelatin or peptone per liter of distilled or deionized
choosing the dilution factor. The extra care needed to pre- water. It is adjusted to pH 7.0 when necessary.
pare the overlay plates is justified because it allows one to For critical or unusual situations, pretest the diluent for
count in the larger range of 30 to 2,000. A second major ad- adverse effects and use a diluent adapted to the particular
vantage is flexibility for nutrient supplementation. Minimal conditions. For plate counts, do not transfer bacteria grow-
agar can be used to pour the basal layer in a large number of ing rapidly at an incubator temperature into cold diluent.
petri plates for indefinite storage. Stock supplies of the min- To minimize death or multiplication, do not suspend them
imal soft agar can also be kept on hand. Then 10- to 50-fold in diluent for more than 30 min at room temperature. The
excesses of needed special nutrients can be added to the growth medium devoid of the carbon source may be the best
aliquots used for the molten top agar. In some cases dyes and diluent for cultures as it may slow or arrest further growth.
chromogenic substrates are added as needed to the soft agar MgClz (0.2 g/liter, sterilized and cooled separately as a 4%
to allow screening of the colonies. Toxic substances can be aqueous stock solution) may be added to preserve mem-
incorporated to measure frequencies of resistant mutants. brane integrity, especially with gram-negative bacteria.
The third method is the pour plate. This method, al- With anaerobic bacteria, the diluent should contain a re-
though commonly employed, must be used with caution be- ducing agent and be freed from dissolved oxygen (see chap-
cause the elevated temperature or the sudden change in tem- ter 14.6.2 and 14.6.4).
perature may kill some bacteria. The method also lacks the A detergent (e.g., 0.1% Tween 80) is sometimes sug-
advantages of the foregoing two methods. Before routine use, gested as a dispersing agent. However, this is effective
test and compare pour plates with one of the other methods. mainly for mycobacteria and other cells with hydrophobic
Several articles and books have been devoted to at- surfaces and may be harmful to other cells. Polymerized or-
tempts to speed and automate growth measurement (2, 29). ganic salts of sulfonic acids of the alkyl-aryl type are effec-
This is a field that is in so much flux that further discussion tive dispersing agents and apparently are not harmful to
is not practical here. representative bacteria (48).
9. Growth Measurement 179

Selection and standardization of diluents are especially samples were used. As pipette and volumetric apparatuses
important in microbiological tests for safeguarding public have been improved, smaller volumes have been used,
consumption of food and water, which are detailed in stan- economizing on reagents and mixing time. Modern plastic-
dard methods manuals published by the American Public tipped semiautomatic pipettes allow very small samples of
Health Association. A particularly good review of the liter- bacterial culture to be used, but then the problem is proper
ature and methods for using diluents is contained in Standard sterilization. This can be done in an autoclave or in a mi-
Methods for the Examination of Dairy Products, 16th ed. (49). crowave oven. Commonly, presterilized tips are used. Good
Meynell and Meynell (6) have also reviewed the use of laboratory technique, however, depends on the continued
diluents, and an entire symposium has dwelt on the death sterility of the racks of pipette tips. When a microwave
and survival of vegetative microorganisms ( 2 5 ) . Further de- oven is used, the energy is absorbed by bacteria or other bi-
tails of diluents are provided in section 9.2.1 and chapter ological material much more readily than by the plastic (be-
20.1.1. cause the bacteria have dipoles and ions), so that the plas-
tic remains cool but the cells and even spores are heated to
9.3.2. Spread Plates lethal temperatures. For many purposes, 0.1-ml serological
pipettes may be used to deliver a full 0.1 -ml volume of cul-
Prepare a suitable agar medium in ordinary glass (old-fashe ture into 0.9 or 4.9 ml of diluent (usually contained in tubes
ioned but ecologically sound) or plastic (modern and con-
measuring 13 by 100 mm) or into 9.9 ml of diluent (usually
venient) 9-cm petri plates. Various procedures have been
contained in tubes measuring 16 by 150 mm). These dilu-
suggested to pour and dry the plates. The following proce-
tion factors are about optimal for ease of making an accu-
dure is recommended. rate dilution and ease of mixing adequately (which is the
1. Place the covered container of molten agar, shortly most critical factor). Moreover, with this rule it is easy to
after removal from the autoclave and after addition of check that the dilution was properly carried out and that no
mildly thermolabile substances (sugars, dyes, antibiotics, trivial error was introduced.
and chromogenic substances), into a dishpan or other large 7. Pipette 0.1 ml of the final dilution onto the agar sur-
vessel containing hot tap water (45 to 50C). The agar face of the petri plate. Form two or three free-falling drops
cools quickly (temporarily warming the water further) but off center on the surface, and then blow the remaining fluid
uniformly throughout the vessel and reaches a temperature onto the surface. If using a pipetting device, push to the sec-
plateau above its solidification point. For 1 liter of agar, 30 ond stop of the pipettor. The cells may have a tendency to
min is sufficient. With this procedure there is no gelling become immediately attached in situ, so do not delay in
around the edges of the container. spreading the drops.
2. Pour the agar into the petri plates. If the agar is suffi- 8. Sterilize a spreader (Fig. 1) by dipping it in 70% alco-
ciently cooled, little condensation will form on the under- hol (do not use the autoclave), shaking off the excess alco-
surface of the petri plate lid. Further, covering the plates hol, and igniting. A spreader can be made quickly from a
with a sheet of newspaper can minimize condensation. If glass Pasteur pipette by using a Bunsen burner with a wing
bubbles form on the agar surface, direct the flame of a tip.
Bunsen burner momentarily downward on them until they 9. Spread the plate. Try to achieve a uniform coverage as
burst (be careful not to melt plastic petri plates). With 9-cm close to the edges as possible. The major difficulty with this
petri plates, 15 to 20 ml of agar is satisfactory, corresponding method is learning the technique for uniform distribution of
to a thickness of 0.24 to 0.32 cm. If thicker, the contrast will the cells. Therefore, after the plates have been incubated,
be less between the colonies and their background. If thin- examine the distribution of colonies on all of the plates to
ner plates are used, colonies may be small and present in re- learn how uniform your spreading technique has become.
duced numbers, possibly because some nutrient is limiting Plates with a larger number of colonies are especially useful
or the plate becomes locally dry. in this regard, even if they contain too many colonies to
3. Dry the plates (to remove the water of condensation count. Also note whether the number of colonies is larger
on the plate lids and water of syneresis on the agar) at room in the vicinity of the original droplets. If so, then the tech-
temperature overnight or for 24 h, depending on the rela- nique must be improved. The agar surface should be dry
tive humidity. Frequently the humidity is lower in incuba- enough that the 0.1 ml delivered from the pipette is ab.
tion rooms or laminar flow hoods, so use them, although sorbed in 15 to 20 s by the agar. Some workers prefer to
some modern incubation sites have controlled humidity. work with turntables, which speed the work of spreading
4. Store the plates indefinitely at 4C in closed plastic the sample uniformly.
containers. Be sure to allow them to warm to room temper- 10. Incubate the inverted plates in a constant-tempera-
ature before use. ture room or chamber with good temperature regulation
5. Prepare a suitable dilution of the cell suspension based (chapter 14.3). Incubating in closed containers is advanta-
on all the information and hunches at your disposal. geous, but do not overfill the containers, as this will in-
Calculate to get 100 to 200 colonies, but use the results of crease the time required for the temperature of the plates to
plates containing between 30 and 300. If the organism become equilibrated to the temperature of the incubator.
forms only small colonies, up to 500 may be counted. Make This is especially important when using temperature-
the dilutions with a diluent solution that does not favor ad- sensitive mutants. Storing in closed containers also avoids
sorption to glass, if ordinary glassware and pipettes are to be the effects of any noxious gases that may be present in gen-
used. High ionic strength, pH between 4 and 5 (if not harm- eral-purpose constant-temperature rooms (e.g., acetic acid
ful), and the presence of small amounts of anionic deter- fumes from gel destaining). Opaque closed containers pro-
gents (if not toxic) are helpful in this regard (section 9.3.1). tect against inactivation of colored drugs and dyes by keep-
Alternatively, use presterilized plastic vessels and pipette ing out light. Finally, in closed containers the plates do not
tips with a Pipetteman-type device. dry out and so can be retained for slow- or late-developing
6. The actual dilutions can be carried out in many ways. colonies (the container can also contain filter paper or paper
Historically, large volumes of diluent (99 ml) and l-ml towels that are kept moist to maintain high humidity).
180 GROWTH

counts are not registered as a result of dirt and imperfections


in the petri dishes (which would not cause difficulty to a
human eye).

9.3.3. layered Plates


1. Prepare the petri plates with a base of 1.5% agar
medium approximately 0.4 cm thick. Pour the plates on a
level surface. Check the work area with a level carefully; if
the surface is not true, use any shim material (metal strips,
wood strips, or pieces of paper) to level a piece of plate glass,
and pour the three layers of agar while the plate rests on this
level surface.
Prepare small test tubes (13 by 100 mm) with 2.5 to 3 ml
of soft 0.7% agar medium. It is convenient to prepare stock
bottles with this strength of agar in the basal medium. Melt
the agar in these bottles. (A microwave oven is the quick
way, but experience is necessary to find the setting that
melts but does not boil the agar explosively.) In any case,
the agar must be thoroughly melted, or mixing cannot be
uniform. Add nutrients, dyes, and inhibitors at this point,
and then pipette aliquots into small test tubes (13 by 100
mm) previously placed in a water bath or heating block at
45C. This pipetting can be done while the agar medium is
still very hot, decreasing the chance of contamination. The
agar will remain liquid at 45C for several hours. It remains
FIGURE 1 Spreaders. The one at the top is bent from a liquid for a longer time at 50C, but this temperature is
paper clip and fitted into a length of Teflon tubing. The one at likely to cause some killing.
the bottom is made by fusing the end of a Pasteur pipette and 2. Dilute the sample as in the previous procedure (sec-
then bending it sharply in two places by using a Bunsen burner tion 9.2.1).
with a wing tip to define the spreading region. The handles of 3. Bring the base-medium petri plates from storage to at
both are bent to the convenience of the user. Both spreaders least 25C (30C is better).
have low heat capacity. The spreader at the top absorbs fewer 4. Pipette 0.1 ml of the diluted sample near the inside lip
bacteria because of its Teflon construction. of the tube. Do this on an identifiable side of the tube (e.g.,
the side with a trademark, or a side that has a frosted spot
or an identifying mark made with a marking pen).
Another point of concern has to do with COZ. Even organ-
5. Immediately pour the soft agar out of the tube onto
the base agar. Pour it over the side of the tube on which the
isms not usually isolated in a high-COZ atmosphere may
sample has been pipetted. This will wash all of the organ-
have a COzrequirement. This may be particularly evident
isms onto the base agar in such a way that they will be quan-
when single cells are spread at high dilution and incubated
titatively transferred and uniformly mixed with the rest of
in normal laboratory air. Some of these considerations may the soft agar. They will not have a chance to become ad-
be of small importance in any particular case, but all should
sorbed to the glass of the test tube or to cluster in spots on
be kept in mind.
the base agar.
11. Observe the plates before the colonies have fully ma- 6. Tilt and swirl the petri plate so that the melted agar
tured. It is often possible to see that too many colonies are de-
covers the surface.
veloping. To obtain reliable information with less coincidence
correction, it may be possible to make fresh dilutions of a sam-
7. Place the petri plate on the level work area while the
agar congeals.
ple that had been retained in the cold or to count colonies
under a dissecting microscope while they are still small.
8. Carry out the platings needed for the remainder of the
experiment.
12. Count the colonies. Depending on the circumstance, 9. Pour or pipette 2.5 to 3 ml of additional soft agar onto
various types of illumination are advantageous and may
the congealed agar surface, and distribute by rocking. Then,
not be obtainable with commercial colony counters.
let this third layer congeal on the level surface.
Experiment with various types of magnifying glasses that
10. Incubate the plates either upright or upside down,
can be worn or clipped onto one's own glasses. Also try var-
since contamination and degree of dryness are much less
ious lamps that have a magnifying lens as an integral part.
critical with this technique than with spread plates.
The colonies may be enumerated by marking the bottom of
the petri plate with a marking pen, or they may be counted
11. Examine and count the colonies. The subsurface
colonies are compact and lens shaped with well-defined
by hand with an electronic counter, hand tally, or televi- edges and are smaller than the usual surface colonies. This
sion-based scanning equipment. One technique that clearly
makes subsurface colonies a little more difficult to count,
marks the individual colonies is to stick the point of a col-
but the magnifying glass of a colony counter helps.
ored pencil into the colony. Not all brands of colored pen-
Magnifying glasses on a headband also help, and these
cils transfer color well; Eberhard Faber Mongol colored pen-
glasses have built-in prisms that reduce eyestrain. A dissect-
cils do this well, but some colors work better than others
ing microscope can also be used.
(e.g., French Green no. 898). Several colors can be used for
differential counting. If using an electronic scanning The extra trouble involved in the overlay technique is
counter, pay careful attention to be sure that false-positive worthwhile when many colonies are present. The colonies
9. Growth Measurement 181

should be uniformly distributed, and this can be checked by alter the reliability of the count or interfere with the object
visual inspection. A fraction of a plate can be counted, and of the experimental plan, the MPN method can be used to
then the count can be prorated. A low-power dissecting avoid these difficulties.
binocular microscope allows virtually all errors due to coin- Modern developments in laboratory techniques can be
cidence to be eliminated because the colonies can be visu- used to speed the execution of the MPN method. Machines
alized in three dimensions. It is laborious to count under are available to fill the wells of plastic trays that have as
such conditions, but it can save a very carefully performed many as 144 or more depressions. Scanning devices de-
experiment from being incomplete and inconclusive. signed for other purposes can be used to aid in counting the
number of wells with no growth. Similarly, automatic and
9.3.4. Pour Plates semiautomatic pipettes can be used to fill small test tubes.
1. Prepare a number of tubes with 20 to 30 ml of thor- Because these procedures make it possible to examine many
oughly molten agar. more cultures, the classical tables of fixed numbers of tubes
2. Place in a 45C water bath or heated aluminum block. and fixed dilution series are obsolete and should be aban-
Allow adequate time for temperature equilibration. doned. A different approach and method of calculation is
3. Mix the sample aliquot. Do this well but without caus- needed and is provided below.
ing bubbles to form. The MPN method used at a single level of dilution when
4. Pour the contents into a sterile petri dish, and allow the mean number of bacterial cells capable of indefinite
them to set. growth is 1.59 per tube is statistically the most accurate
5. Incubate, examine, count, and calculate. (24). This number would result in 20.8% of the tubes remain-
ing sterile (Fig. 2). Consequently, if the expected number
is known precisely and the assay is conducted for confirma-
9.4. MOST PROBABLE NUMBERS tion, all the available tubes should be seeded with a dilution
The concentration of viable cells can be roughly estimated that is expected to have the value 1.59. The accuracy falls
by the MPN method (also called the fraction-negative or
endpoint-quanta1 method), which involves the mathemat-
ical inference of the viable count from the fraction of mul-
tiple cultures that fail to show growth in a series of dilution Mean cell number per tube
tubes containing a suitable growth medium. B
This method consists of making a number of replicate di-
lutions in a growth medium and recording the fraction of
tubes showing bacterial growth. The tubes exhibiting no
growth presumably failed to receive even a single cell that
was capable of growth. Since the distribution of such cells
must follow a Poisson distribution (see below), the mean
number plated at this dilution can be calculated from the
formula Po = Crn, where m is the mean number and Po is
the ratio of the number of tubes with no growth to the total
number of tubes. The mean number, estimated by -In Po
(In designates the natural logarithm), is then simply multi-
plied by the dilution factor and by the volume inoculated
into the growth tube to yield the viable count of the origi-
nal sample. The MPN method is a very inefficient method
from the point of view of statistics, because each tube cor-
responds to a small fraction of the surface of a petri dish in
a plate count. Consequently, very many tubes or wells in a
titer plate must be used, or the worker must be prepared to
settle for a very approximate answer.
When is the MPN method of advantage? First, it can be
used if there is no way to culture the bacterium on solidified
medium. Second, it is preferred if the kinetics of growth are
highly variable. Suppose some cells grow immediately and
rapidly and end up making a large colony on solid agar that

lot
spreads over and obscures colonies of the organism of inter-
est that form later. The small-colony formers may be more
numerous but unmeasurable on plates because of the fewer
but highly motile or rapidly growing bacteria. Third, it is
preferred if other organisms not of interest are present in the I I I I I
sample and no selective method to eliminate them is avail- 0 10 20 30 40
able. The method has utility when the bacterium of inter- Percentage of sterile tubes
est produces some detectable product (e.g., a colored mate-
rial, specific virus, antibiotic, or other metabolite). Then, FIGURE 2 Errors that arise in using the MPN method. The
even though contaminating organisms may overgrow the CV for a single level of dilution is shown as a function of the
culture, the numbers of the bacterium in question can be es- percentage of sterile tubes (bottom abscissa) and the mean
timated by the fraction of the tubes that fail to produce the number of cells per tube (top abscissa). The respective optima
characteristic product. Fourth, if agar and other solidifying occur at 20.8% and 1.59 mean cells per tube for a varying num-
materials have some factors (such as heavy metals) that may ber of tubes (n) prepared at a single dilution level.
Next Page

182 rn GROWTH

off rapidly with deviations from this optimum, particularly same number of levels are always used, the program can be
outside the range of a n average of 1 to 2.5 cells per tube. customized to fix these parameters and to delete some of the
Thus, the dilutions 10-fold away from the optimum con- requests that are made by the supplied program.
tribute almost no information about the mean number of A statistician might properly point out that the program
cells. Consequently, if the viable count is known within a does not compute the error statistics. Although that feature
narrow factor, the optimum strategy is to use a narrow range could have been included in the program, it was omitted to
of dilution factors. If the uncertainty is greater, a wider make the program briefer, simpler, and more readily under-
range should be used. standable and adaptable. The program is short enough that
Usually, prior knowledge is not available and growth the user, it is hoped, will undertake the one-time-only job
tubes at several dilutions are needed. The simplest approach of keying it in. However, there are two even better reasons
is to separate the dilution levels 10-fold. For this approach, for not calculating the error directly from the statistical the-
little is lost by discarding the data from dilutions at which ory for the MPN. First, the most reliable data come from
the percentage of sterile tubes lies outside the range of 8 to the level in which 20% of the cultures show no growth. The
36% or outside the range of 5 to 50% if a larger range of error can be quickly estimated with adequate accuracy by
error is acceptable. The error from a dilution within the neglecting the rest of the data and using the level with the
range can then be read or interpolated from Fig. 2. This closest proportion to 20% and values read from Fig. 2.
method is simple but more wasteful than is necessary. Second, as mentioned in section 9.2, the errors in pipetting
Consequently, statistical methods have been developed to and sampling are not included in such an error computa-
combine data from different dilution levels when specified tion. Consequently, it is strongly recommended that several
numbers of tubes are run at each level. independent samples be taken and processed independently
Classically 10 cultures at each of three successive 10-fold by MPN determinations. Consider an example in an eco-
dilutions have been used. Of the 1,000 possible outcomes of logical context in which the problem is to estimate the
such tests, Halvorson and Ziegler (28) listed the 210 cases number of cells in a stream. It is far better to take samples
that could happen with P values of >0.01%. Finney (24) from different locations within the stream or sample at a se-
gave a good exposition of the statistical aspects and the ap- ries of different times than to take one sample and analyze
proaches to calculate the MPN of replicates at each dilu- it with many tubes. With more original samples and fewer
tion. He pointed out that there could be a definite advan- tubes in each individual MPN determination, the estima-
tage in computation when carrying out error calculations if tion of the mean and standard deviation (and standard
the dilution series extends over a range such that most di- error) of the independent MI values will give an estimate
lution levels show some tubes with growth, but there is a of the combined error due to variation of the biological
higher dilution in which every tube shows no growth. Even source, errors in the dilution procedure, and errors in the
then, the proper statistical calculations are cumbersome and MPN determination itself.
very wasteful of time and material.
With modern computer programs (see Appendix, The
MPN Method), it is easier, better, and more flexible to do 9.5. BIOMASS MEASUREMENT
away with tables and to calculate the MPN for an individu- A measure of the mass of bacterial cell constituents fre-
ally chosen and optimized arrangement of tubes and dilu- quently is used as the unit basis for measurement of a meta-
tions. Even for a standardized series of dilutions (for exam- bolic activity or the concentration of a morphological or
ple, the data shown in Table l ) , a computer program is of chemical constituent; i.e., biomass and cell numbers pro-
value since sometimes the possible outcomes obtained are vide the two basic parameters of bacterial growth.
not listed in the published tables. The methods for measuring biomass seem obvious and
Table 1 shows an example for the case of 10 tubes at each straightforward, but in fact they are complicated if accuracy
of three 10-fold dilutions. Thus, the numbers of tubes ( t l , t2, is sought. Furthermore, the results may be expressed in dif-
and t3)are each 10. The volumes of solution pipetted are ul = ferent ways, and, in some of these ways, the values may be
10, u2 = 1, and u3 = 0.1. The data for the example are gl = 8, more relative than absolute.
gz = 5, and g3 = 1. Although 30 tubes are used, the accuracy
is less than if all 30 tubes had been used at a single dilution (as 9.5.1. Wet Weight
given by the curve method n = 30 in Fig. 2) and would have A nominal wet weight of bacterial cells originally in liquid
been much poorer if a wider range had been chosen. suspension is obtained by weighing a sample in a tared pan
When designing an MPN measurement, one must after separating and washing the cells by filtration or cen-
choose first how many dilution tubes at each level and how trifugation. In either case, however, diluent is trapped in the
many dilution levels are to be used for the analysis and how interstitial (intercellular) space and adds to the total weight
far apart they will be spaced. To do this, one must consider of the sample. The amount of interstitial diluent may be
the expected titer and the confidence that one has in that substantial. A mass of close-packed, rigid spheres contains
estimate. The less sure one is of the titer of the sample, the in its interstice 27% of the volume of the cells. This is in-
wider is the range of dilution levels that one should test; dependent of size for uniform spheres, and a pellet of bacte-
conversely, if the titer can be quite accurately estimated or rial cells close packed by centrifugation may contain an in-
inferred, one should choose fewer levels. This flexibility, terstice of 5 to 30%, depending on cell shape and amount of
compared with existing tables, is really the point of using deformation.
the computer program, because it allows the investigator to A method for obtaining the actual wet weight of the
make optimum use of the MPN technique. cells themselves is to correct the nominal wet weight of a
The program, as written, prints the output only on the packed-cell pellet by subtracting the experimentally deter-
screen. It is expected that the user will wish to modify the mined weight of diluent in the interstitial space. This is de.
program to obtain a printed output instead. Printer direc- termined by use of a nonionic polymeric solute that perme-
tions have been omitted because format statements vary ates into the interstitial space but is too large to penetrate
widely among computer systems. If the same volumes or the into bacterial cell walls, such as dextrans of very high aver-
10
Nutrition and Media'
DAVID EMERSON AND JANE TANG

10.1. INTRODUCTION ......................................... 201


.
2. TERMINOLOGY .......................................... 201
.
3. BASIC NUTRITIONAL REQUIREMENTS ..................... 201
10.3.1. Macronutrients ...................................... 202
10.3.1.1. Carbon ..................................... 202
.
2. Nitrogen .................................... 202
.3.Sulfur ...................................... 202
.
4. Phosphorus .................................. 202
.
5. Potassium and Magnesium ....................... 202
.
6. Others ..................................... 202
10.3.2. Micronutrients ....................................... 202
10.3.2.1. Minerals .................................... 202
.
2.Vitamins .................................... 202
10.4. COMPLEX NUTRITIONAL COMPONENTS .................... 205
10.4.1. Peptones. Extracts. and Hydrolysates ....................... 205
10.4.1.1. Acid Hydrolysates ............................. 205
.
2. Enzymatic Digests ............................. 205
.
3. Extracts of Meat and Yeast ....................... 205
.
4. Mixed Hydrolysates ............................ 205
10.4.2. Other Organic Nutrients ............................... 205
10.4.2.1. Fatty Acids .................................. 205
.
2. Others ..................................... 205
10.4.3. Supplements ........................................ 205
10.4.3.1. Blood and Serum .............................. 206
.
2. Rumen Fluid ................................. 206
.
3. Soil Extract .................................. 206
.
4. Other Supplements ............................ 206
10.5. NONNUTRITIONAL MEDIA COMPONENTS .................. 206
10.5.1.Agar .............................................. 206
.
2. Reducing Agents ..................................... 206
.
3. Redox Indicators ..................................... 207
.
4. Buffers ............................................ 207
10.5.4.1. Inorganic Buffers .............................. 207
.
2. Organic Buffers ............................... 207
10.6. PREPARATION OF MEDIA ................................. 208
10.6.1. Sterilization ......................................... 208
10.6.1.1. Autoclaving .................................. 208
.
2. Filtration
.
....................................
3. Tyndallization ................................ 208
208
10.7. QUALITY CONTROL TESTING ............................. 208
.
8. MEDIA FOR SPECIFIC GROUPS ............................. 209
10.8.1. Media for Heterotrophic Microbes ......................... 209
.
2. Phototrophs ......................................... 209
10.8.2.1. Oxygenic Phototrophs .......................... 209
.
2. Anoxygenic Phototrophs ........................ 209
10.8.3. Biodegradation ....................................... 210
.
4. Methanogenesis ...................................... 210
.
5. Methane-Oxidizing Bacteria ............................. 210
.
6. Sulfur Metabolism .................................... 210
10.8.6.1. Sulfate Reduction ............................. 210
.
2. Sulfur Disproportionation ....................... 211
.
3. Sulfur Oxidation .............................. 211
10.8.7. Dissimilatory Metal Reduction ........................... 211
'This chapter is an updated and significantly revised version of an earlier chapter in Methods fur General and MokcuLrr
.
Bacteriology by Rosalie ].Cote and Robert L Ghema (see reference 9) .
200
10. Nutrition and Media H 201

...............................
10.8.7.1. Iron Reduction 2 11
.2. Manganese Reduction.......................... 2 11
..........................
.3. Arsenic and Selenium 21 1
10.8.8. Fe Oxidizers ....................................... 211
.9. Nitrifying Bacteria.................................. 212
.lo. Halophiles........................................ 2 12
.11. Oligotrophs ....................................... 212
10.9. SOURCES .............................................. 2 12
10.9.1. Sources of Manufacture .............................. 212
.2. Culture Collections ................................. 212
.3. Others. .......................................... 212
10.10, REFERENCES ........................................... 2 13

10.1. INTRODUCTION 10.2. TERMINOLOGY


Nutritional diversity is the hallmark of prokaryotes. As a re- A sometimes bewildering array of terms are encountered to
sult, the media requirements for prokaryotes reflect the di- describe the general properties of different media. The fol-
verse array of organic and inorganic energy sources that lowing is a glossary of media descriptions.
these organisms can utilize. To illustrate this point, at the
Defined or synthetic medium: a medium in which the chemi-
ATCC there are approximately 18,000 strains of prokary-
cal composition is known.
otes that require over 2,000 different kinds of media to
maintain; in comparison, there are approximately 36,000 Complex or undefined medium: a medium in which the
strains of fungi but these metabolically diverse eukaryotes chemical composition is not known or defined. Ex-
require only about 100 different kinds of media for growth. amples include yeast extract or peptone.
The preparation of the appropriate medium is where every Minimal medium: a medium that is designed to provide a
microbial investigation begins, whether it is the routine use given organism with only the essential nutritional re-
of Escherichia coli to develop a clone library or an attempt to quirements that it cannot synthesize by itself. Generally
isolate a novel anaerobic hyperthermophile from a deep-sea all the components are defined.
hydrothermal vent. Every medium shares in common the Mineral medium: a defined medium that contains only es-
necessity of providing an organisms energy requirements, sential minerals, trace elements, vitamins (if necessary),
sources of carbon, minerals, and any supplementary nutri- and a simple energy source, for example, glucose.
ents, as well as an appropriate atmosphere and a stable pH. Auxotroph medium: a medium, usually defined, that contains
The roots of general microbiology lie in the seemingly an essential growth factor (for example, an amino acid)
simple concept pioneered by Beijerinck and Winogradsky in which an organism requires. Auxotroph media are often
the late 1800s of using a selective medium to enrich from the used in the selection of genetically altered microbes.
environment a particular group of organisms. For example,
Selective medium: a medium designed either to select for
medium that lacks a source of combined nitrogen can be used
growth of cells possessing a specific trait (for example, an-
to select for organisms capable of fixing nitrogen, or medium
tibiotic resistance) or to inhibit a group of organisms.
that contains methanol as the sole energy source can be used
Examples include adding Brilliant Green to inhibit gram-
to select for methylotrophic bacteria. While selective enrich-
positive bacteria and select for gram-negative organisms.
ment may be seemingly simple, more than 100 years later
we are still conceiving of new combinations of organic and/or Enrichment medium: a medium designed to promote the
inorganic compounds to discover whole new metabolic growth of organisms with a specific physiological trait
classes of organisms. Recent intersections between geology from a mixed population (for example, sulfate reduc-
and microbiology, as well as environmental engineering and tion). Enrichment can also be achieved by adding a spe-
microbiology, have provided even more creative approaches cific supplement to an otherwise nonselective medium.
for developing selective media for novel organisms; see, for For example, adding cysteine to enrich for Francisella,
example, Amend and Shock (2). This chapter places some which specifically promotes the growth of this organism.
emphasis on the nutritional needs of some of the broad Differentialmedium: a medium designed to distinguish a par-
groups of prokaryotes that have been discovered through ticular microbe or group of microbes based on nutritional
using selective media as an enrichment tool. properties. For example, MacConkey medium differenti-
Other important aspects of nutrition that are considered ates between lactose fermenters and nonfermenters.
in this chapter are requirements for vitamins, trace miner- Bioassay medium: a medium designed to determine the con-
als, other growth factors, buffers to control pH, sterilization, centration of an essential growth compound by quanti-
and preparation of solid media. A section on quality control fying the growth of a microbe that requires that com-
of media is also included. In general, specific media formu- pound in the medium. A standard curve is developed by
lations are not included in this chapter. The best general growing the organism on a range of concentrations, from
sources for media formulations are the websites of the large very limiting to nonlimited, of the compound.
culture collections; for example, the ATCC, the DSMZ, or
the JCM, where complete listings of media formulations are
available free of charge. These can be searched either by or- 10.3. BASIC NUTRITIONAL REQUIREMENTS
ganism or by media name. The URLs for these sites are The essential nutritional requirements for prokaryotes can
listed under the sources section (section 10.9.2). In addi- be divided into two classes: macronutrients and micronutri-
tion, the Handbook of Microbiological Media (4) has a large ents. The macronutrients serve as the primary energy
number of media formulations. sources for growth as well as the chemical building blocks
202 GROWTH

for cellular structures. The micronutrients, vitamins, miner- enzymes, as well as being needed for carbohydrate metabo-
als, and other cofactors, can provide essential functionality lism and transport processes. Among the halophilic archaea
to enzymes and other parts of the cellular machinery. it plays a key role in osmoregulation (15). Potassium re-
quirements are normally met by the addition of potassium
10.3.1. Macronutrients salts (sulfate, phosphate, or chloride) to the medium.
More than 30 elements are considered essential for cell Magnesium stabilizes ribosomes, cell membranes, and
growth; however, there are six nonmetals (carbon, oxygen, nucleic acids. A number of enzymes also use magnesium as
hydrogen, nitrogen, sulfur, and phosphorus) and two metals an essential cofactor, for example, ATPase. Magnesium sul-
(potassium and magnesium) that constitute 98% of the dry fate or chloride salts are the usual form added to the growth
weight of prokaryotic organisms (15). These elements must be medium to satisfy this requirement. ,

present in relatively high concentrations in all growth media.


10.3.1.6. Others
10.3.1.1. Carbon Sodium is necessary for certain marine bacteria, pho-
The majority of known prokaryotes are heterotrophic; that totrophs, and some anaerobes. Although calcium is not a
is, they use carbon-containing organic compounds to satisfy requirement for growth, it stabilizes cell walls and plays a
their carbon and energy requirements. The different classes role in the heat stability of bacterial endospores (25).
of organic compounds are shown below (see Table 3, in sec-
tion 10.8.1). Some bacteria are very versatile in their abil- 10.3.2. Micronutrients
ity to utilize a wide range of compounds as sole sources of The micronutrients (minerals, vitamins, and other cofac-
carbon and energy, e.g., pseudomonads, whereas others are tors) are essential for optimal growth; however, the absolute
limited in their ability to decompose certain organic sub- quantities that are required are often very small (micro- or
stances, e .g., methylotrophs. nanomolar concentrations, or less). For this reason, they are
Autotrophy, or the ability to fix COZ as the principal normally added to growth media from concentrated stock
source of carbon for cellular materials, is also widespread solutions. Ideally, in characterizing a microbe its specific vi-
throughout the prokaryotic world. Autotrophs require a tamin requirements should be determined; however, this is
source of C02in the gas phase; however, this is often sup- tedious and often not done; it is simply easier to add a stock
plemented by inclusion of bicarbonate salts in the liquid solution of vitamins. One common problem in growing mi-
phase. C02-bicarbonateor bicarbonate-carbonate mixtures crobes is that growth may seem to diminish upon serial
can also act as a buffering system for media. transfers of the culture; often this is a result of some essen-
tial micronutrient being diluted out of the original culture
10.3.1.2. Nitrogen medium.
Nitrogen is the next most abundant constituent in the cell,
making up 8 to 15% of the dry weight (15, 25). It is the 10.3.2.1. Minerals
major noncarbon constituent of proteins, nucleic acids, There are a number of elements that, although required in
peptidoglycan, and coenzymes. Microorganisms can obtain tiny amounts (milligrams per liter), are critical to the over-
nitrogen either from inorganic sources such as salts of am+ all nutrition of microbes because of their role as cofactors in
monium (NH4+)or nitrate (N03-) or by taking up N-rich a wide variety of enzymatic reactions. These elements in-
organic compounds such as amino acids. Unique to all of clude iron, manganese, cobalt, copper, molybdenum, zinc,
life are the nitrogen-fixing or diazotrophic prokaryotes that nickel, chloride, and boron. Some organisms require sele-
contain nitrogenase and are capable of fixing atmospheric nium and tungsten. In the case of complex media, these el-
nitrogen gas (N2) as a source of nitrogen. ements are usually abundant enough in the organic supple-
ments so it is not necessary to add them to the media
10.3.1.3. Sulfur separately. A n important exception to this can be iron. Due
Sulfur is an absolute requirement for microorganisms be- to its prevalence in respiratory enzymes and cytochromes it
cause of its structural role in building amino acids (cysteine can potentially be growth limiting, especially in complex
and methionine), coenzymes, and a number of vitamins (bi- media in which the overall organic carbon content is low.
otin, thiamine, and lipoic acid) (5, 25). Microorganisms In most mineral salts media it is recommended to add
generally acquire sulfur from inorganic sulfate salts or from minerals from a stock solution. In Table 1 a recipe for
organic sulfurecontaining amino acids. In addition, sulfur ATCC's trace mineral supplement (based on Wolfe's min-
compounds are involved in the energy metabolism of quite eral solution) is provided along with the primary function of
a large number of microbes, either serving as electron ace each element (9,38). Many stock mineral solutions call for
ceptors for anaerobic respiration, or as oxidizable substrates the use of a chelating agent, such as nitrilotriacetic acid or
(see section 10.8.6.2.) EDTA, to prevent precipitation of the relatively high con-
centrations of metals in the solution. Stock solutions of
10.3.1.4. Phosphorus minerals should always be filter sterilized. While the heat of
Phosphorus, usually in the form of phosphate, is needed for autoclaving does not harm the metals themselves, it can
ATP synthesis and serves as a cellular constituent for nu- cause precipitation reactions. After the mineral solution is
cleotides, phospholipids, and coenzymes, as well as cell wall diluted into the final growth medium (usually 1:lOO or
components of gram-positive bacteria (teichoic acids). Or- 1:l,OOO), the solutions can be autoclaved without harm.
ganisms obtain phosphorus from inorganic phosphate ions or
organic sources, such as glycerophosphate or phosphate esters. 10.3.2.2. Vitamins
Growth factors are specific organic compounds that a cell
10.3.1.5. Potassium and Magnesium may not be able to synthesize and are usually required in
Potassium is required by all organisms, and it is the princi- very small (parts per million) amounts. There are three
pal inorganic cation in the cell. It is a cofactor for several main classes of growth factors: (i) amino acids for certain
10. Nutrition and Media 203

TABLE 1 Trace mineral supplements


Ingredients Concn (g per liter) Function
EDTA 0.5 Chelating agent
MgS04 . 7H2O 3.0 Cocatalyst, contributes to pigmentation of some organisms
MnS04 . H 2 0 0.5 For enzymes reacting with active oxygen species, and water-splitting enzymes
of photosystem 11
1.0 Electrolyte
0.1 For enzymes, electron carriers, and oxidoreductases
0.1 For making cobalamin (BIZ)
0.1 Cocatalyst; membrane stability
0.1 For different enzyme classes; dehydrogenases, polymerases, anhydrase, proteinases
0.01 For enzymes reacting with oxygen; oxygenases, oxidases, superoxide dismutase,
and certain nitrite reductases
AIK(S04)Z . 12Hz0 0.01 Function unknown, probably for enzymatic activities
H3B03 0.01 Uptake of Ca2+and Mg2+; increase retention of Ca2+in cells; synthesis
of certain quorum signal compounds
NaZMo04.2HzO 0.01 For oxidoreductases and nitrogenases
NazSeO3 0.001 Found in tRNA and selenocysteine of some oxidoreductases
NiClz 6H20 0.02 For Ni-tetrapyrrole of methanogens, in urease, hydrogenase, CO dehydrogenase
Na2W04. 2H20 0.01 Substitute for Mo in some oxidoreductases, but cannot be replaced by Mo

proteins, (ii) purines and pyrimidines for nucleic acids and role in one-carbon (C,) metabolism and transfer. Folic acid
coenzymes, and (iii) vitamins for coenzymes or prosthetic is only slightly soluble in water, but the ammonium salt is
groups of certain enzymes (9). very soluble. It is stable to autoclaving.
Vitamins play catalytic roles within the cells and usually
serve as components of coenzymes or as prosthetic groups of 10.3.2.2.3. Biotin
enzymes. There are certain bacteria that have complex vi- Biotin is responsible for biosynthetic reactions that re-
tamin requirements. The lactic acid bacteria, which include quire carbon dioxide fixation and release, as well as the syn-
Streptococcus,Leuconostoc, and Lactobacillus,are well known thesis of fatty acids. Free biotin is only slightly soluble in
for this, and they are used in vitamin assay procedures. For water, but its salts are freely water soluble. Biotin is stable to
growth in complex media, yeast extract (see section autoclaving and acids but can be oxidized to the sulfoxide
10.4.1.3) is often added as a supplement that is rich in vita- and sulfone. For bacteria that can convert the oxidation
mins and can fulfill the requirements for most prokaryotes. products to biotin, these compounds can promote their
In defined and mineral media, vitamins are normally added growth. Oxidation is a problem only in extremely diluted
from a stock solution. Stock solutions of vitamins should be solutions (<1 p,g/ml) and if n o reducing agents are present.
made in ultrapure water and filter sterilized. They are nor-
mally added to growth media after the medium has been au- 10.3.2.2.4. Nicotinic Acid and Its Derivatives
toclaved. Some vitamins are light sensitive (e.g., riboflavin Nicotinic acid serves as the precursor of NAD and
and vitamin B6), so stock solutions should be kept at 4C in NADP, which are involved in redox reactions. Nicotinic
the dark. It is recommended that vitamin stock solutions be acid and its amide are stable to autoclaving at neutral pH.
made fresh every 6 months. Some organisms (Hamophilus influenza) are unable to syn-
Below is a group of commonly used vitamins, with some thesize NAD from nicotinic acid, and thus, they require the
detail as to their functions and properties. This information addition of NAD (factor V) as the coenzyme. NAD is de-
is largely drawn from an earlier edition of this book (9). stroyed by autoclaving, acids, and alkalis, so it must be ster-
Table 2 summarizes this list of vitamins and their functions. ilized by filtration and added aseptically to the separately
autoclaved medium.
10.3.2.2.1. PABA
p-Aminobenzoic acid (PABA) serves as a precursor for 10.3.2.2.5. Pantothenic Acid and Related
tetrahydrofolic acid biosynthesis; thus, it plays a part in the Compounds
metabolism of thymine, methionine, serine, the purine Pantothenic acid is the precursor of coenzyme A and a
bases, and vitamin BIZ. The presence of these compounds component of the acyl carrier protein. It also plays a part in
may decrease or eliminate the requirement of a bacterium the metabolism of fatty acids. A few bacteria utilize the in-
for PABA in the medium. tact coenzyme as a growth factor, but most others require
pantothenic acid or pantotheine for growth. The require-
10.3.2.2.2. Folic Acid Group ment of the individual forms of these related compounds re-
Folic acid is primarily found in cells as tetrahydrofolate. flects the differences in the synthetic and transport abilities
It functions as a cofactor in the synthesis of thymine, purine of the bacteria. Free pantothenic acid and many of its salt
bases, serine, methionine, and pantothenic acid. It plays a forms are very hygroscopic; thus, it is marketed as the calcium
204 GROWTH

TABLE 2 Vitamins and their functions


Vitamin Functions
pAminobenzoic acid Precursor of tetrahydrofolic acid; involved in one-carbon transfer
Biotin Carbon dioxide fixation and release; fatty acid biosynthesis
Coenzyme M Involved in methane formation
Cyanocobalamin (BIZ) Molecular-rearrangement reactions; synthesis of deoxyribose
Folic acid One-carbon metabolism
Lipoic acid Transfer of acyl groups
Pantothenic acid Precursor of coenzyme A; metabolism of fatty acids
Pyridoxine (B6) Deamination/transamination
Nicotinic acid Precursor of NAD and NADP; involved in redox reactions
Riboflavin (BJ Precursor of FMN and FAD; involved in redox reactions
Vitamin K Precursor of menaquinone
Thiamine (B, ) Transketolase; decarboxylation

salt, which is only slightly hygroscopic, freely soluble in water, 10.3.2.2.9. Vitamin BIZ
and autoclavable. Like its more complex derivatives, pan- Cyanocobalamin (vitamin B12) functions in methyl
tothenic acid is readily hydrolyzed, so its growth-promoting (CH,) transfer reactions. The coenzyme form (cyanocobal-
activity is destroyed by acid or alkaline hydrolysis. amin coupled to adenine nucleoside) plays a part in a num-
ber of carbon rearrangement reactions and in the biosyn-
10.3.2.2.6. Riboflavin and Its Derivatives thesis of ribonucleotide reductase. Vitamin B12 itself is
Riboflavin 5-phosphate (flavin mononucleotide) and relatively stable to light and autoclaving. While vitamin
flavin adenine dinucleotide are the major coenzyme forms B12is synthesized by many bacteria, it is one of the vitamins
of riboflavin. They serve as cofactors in redox reactions. most commonly added as a supplement to minimal media.
Riboflavin is slightly soluble in cold water but is easily
soluble by warming. It is destroyed by visible light, espe- 10.3.2.2.10. Lipoic Acid
cially at neutral pH or above; thus, it is important to Lipoic acid is essential for propagating members of the
store the solutions in the dark. Riboflavin is stable to lactic acid bacteria. Its function is in the transfer of acyl
autoclaving. groups and in the irreversible oxidative decarboxylation of
2-0x0 acids. Acetate can alleviate the requirement of lipoic
10.3.2.2.7. Thiamine acid for many lipoate auxotrophs. Lipoic acid is stable to
A large number of bacteria require thiamine, its precur- acid and autoclaving but is labile to oxidizing agents.
sors, or its coenzyme form, thiamine pyrophosphate (cocar-
boxylase) to grow. Thiamine pyrophosphate is involved in 10.3.2.2.11. Vitamin K
the decarboxylation of 2-0x0 acids and in the transketolase Vitamin K and related compounds are the precursors of
reaction. At pH 5.0 or above, thiamine is cleaved into its menaquinones, which function as carriers for redox reac-
component moieties in aqueous solutions when autoclaved; tions. A few organisms (e.g., Porphyromonas levii, Prevotella
thus, the solutions need to be filter sterilized or autoclaved melaninogenicu, and Prevotella nigrescens) need vitamin K or
at pH below 5.0 if the intact form of this vitamin is required. related compounds to grow (13, 23). Vitamin K3 (mena-
dione; 2.methyl-l,4-naphthoquinone) is destroyed by light,
10.3.2.2.8. Vitamin B6 Group alkali, and reducing agents. Vitamin K1 (2-methyl-3-
The vitamin B6 group is comprised of pyridoxine, pyri- phytyl-1,4-naphthoquinone)is more heat stable and bio-
doxal, pyridoxamine, and their phosphorylated derivatives. logically reactive for some bacteria. Both forms are fat solu-
They are precursors of pyridoxalphosphate and serve as ble, and they can be added to media from an ethanol stock
coenzymes in amino acid transformation (deamination and solution. Vitamin K stock solutions should be prepared fresh
transamination) reactions. Pyridoxal is preferred by some at monthly intervals and stored at refrigeration temperature
bacteria, and adequate amounts of it are formed during au- in the dark.
toclaving if sufficient pyridoxine is included in the medium.
Some bacteria require pyridoxamine 5-phosphate for 10.3.2.2.12. Purines and Pyrimidines
growth; but for most auxotrophs the phosphorylated forms The relationships of nucleosides between folic acid and
are inactive probably due to the lack of the necessary trans- vitamin BIZwere mentioned above (sections 10.3.2.2.2 and
port systems for these bacteria. 10.3.2.2.9). Some bacteria do not have the ability to syn-
All three forms of the vitamin are stable to autoclav- thesize purines and pyrimidines, for example, Bacteroides
ing; however, under the temperatures and pressures of au- frugilis ATCC 29766 and Vibrio cholerae ATCC 14033. In
toclaving pyridoxal and pyridoxamine may react with this case, the nucleosides need to be added to the mineral-
many naturally occurring compounds and become inac- based media in milligram concentrations. The purine salts
tive. Thus, it is crucial that these vitamins be filter steril- are more soluble than the free bases. Guanine and xanthine
ized. All forms of this vitamin are sensitive to light, espe- can be dissolved in a minimal amount of hydrochloric acid;
cially at alkaline pH. the other compounds are soluble in hot water.
10. Nutrition and Media 205

10.4. COMPLEX NUTRITIONAL 10.4.1.3. Extracts of Meat and Yeast


COMPONENTS Extracts of meat and yeast contain large amounts of amino
Complex media contain many undefined ingredients, i.e., acids, vitamins, and trace elements. Plant extracts contain
crude extracts of animals and protein digests. These highly high concentrations of carbohydrates. They are often in-
nutritious substances promote bacterial growth by providing cluded in complex media as a source of nutrition. The com-
an excess of both micro- and macronutrients (9). Complex monly encountered meat extracts are beef extracts and
media are used routinely to propagate a variety of microor- brain heart infusion, which are made by boiling the sub-
ganisms and are especially useful when maximal growth strate for a prescribed time (20 min at l0OOC). They are free
rates and yields are the objective. The most common unde- from fermentable carbohydrates but contain glycolic acid,
fined components are listed below (4, 9). lactic acid, and creatinine, which can serve as carbon
sources. Yeast extract is produced by the autolytic reaction
10.4.1. Peptones, Extracts, and Hydrolysates of the proteolytic enzymes from the yeast cells and consists
Peptones are hydrolyzed proteins that result from enzymatic of a concentrated solution of hydrolyzed yeast protein,
hydrolysis, or acidic/alkali digestion resulting in individual which is high in water-soluble vitamins and carbohydrates
amino acids and small peptides. Casein is the most common as well. Commercially available powdered forms may not be
protein used for forming peptones, but more complex ani- equivalent in the growth-promoting potential to freshly
mal and plant hydrolysates can serve as raw materials for prepared autolysates of yeast. The latter have been shown to
these digests as well. The protein source and the method be more effective in media for cultivating mycoplasmas, and
and degree of digestion are important in determining the preparation of fresh yeast autolysate was shown to be essen-
suitability of a particular digest in a specific medium. tial for the culture of acetogenic spirochetes from the ter-
Although medium manufacturers rigidly evaluate the diges- mite gut (22).
tion products, lot-to-lot variations are expected, especially 10.4.1.4. Mixed Hydrolysates
with respect to those undefined factors that promote the
growth of certain very fastidious organisms. Peptones and Mixed hydrolysates are processed yeast or meat extracts to
hydrolysates are usually added to media at a concentration supplement the nutritional aspects of peptones by providing
of 0.5 to 1.0%. some amino acids, carbohydrates, nucleic acid fractions, or-
ganic acids, vitamins, and trace minerals lost during their
10.4.1 .1, Acid Hydrolysates manufacture. Mixed hydrolysates include polypeptone (ca-
sein and meat), tryptose (mixed enzymatic sources), and
Acid hydrolysates use heat and low pH to digest the pro- Biosate (yeast extract and casein digest). These are gener-
teins. As a result tryptophan and, to a lesser degree, serine ally added to media at final concentrations of 0.3 to 0.5%.
and threonine are destroyed in the process. Further treat-
ment during the manufacture of acid hydrolysates also lim- 10.4.2. Other Organic Nutrients
its the vitamin content of the end products. In addition,
neutralization of the final products results in a high salt 10.4.2.1. Fatty Acids
content. Among the common forms of acid-hydrolyzed ca- Certain groups of microorganisms can utilize fatty acids as
sein are Acidicase and Casamino Acids. These and other carbon and energy sources, and some groups are unable to
acid-hydrolyzed caseins consist predominately of free amino synthesize certain fatty acids required for their lipids; these
acids but are deficient in tryptophan and cystine. They are must be provided in the medium. For example, members of
used in assay media that are low in these growth factors, as Mycobacterium need oleic acid included in their propaga-
well as in media for toxin production. tion media (Middlebrook 7H10 agar, ATCC medium 173).
Several of the volatile fatty acids can be substituted for
10.4.1.2. Enzymatic Digests rumen fluid to cultivate the microorganisms present in the
Enzymatic digests best conserve the intact nutritional con- rumen. Volatile fatty acids can be prepared as a stock mix-
tent of the original protein source. Common enzymatic di- ture and added to the propagation media. It should be re-
gests of casein include Casitone, Trypticase peptone, and membered that fatty acids can be toxic even at low con-
tryptone. They consist mostly of free amino acids and small centrations; this is especially true for unsaturated fatty
peptides and are used frequently in growth media for het- acids. Thus, it may be necessary to include a detoxifying
erotrophs. Since the digests are low in carbohydrates, they compound in the medium. Bovine serum albumin is com-
can be used in fermentation test media for certain sugars. monly used for this purpose.
Examples of enzymatic digests of meat protein are pep-
tone, proteose peptone, Myosate, and Thiotone. They con- 10.4.2.2. Others
sist of a general mixture of amino acids and peptides suit- Some Streptococcus pneumonia strains and oral treponemes
able for growing most heterotrophs. Peptones with the require choline to grow. This organic compound serves as a
designation proteose are hydrolyzed in such a way to have precursor for certain cellular lipids. In S. pneumoniae, the
a high peptide content and are recommended for cultivat- choline residues make up the lipoteichoic cell membrane.
ing fastidious organisms. Thiotone is a digest high in sulfur Purines and pyrimidines are the building blocks of nucleic
amino acids and can be used in test media for detecting hy- acids and may be required in auxotrophic strains that are
drogen sulfide. unable to synthesize these compounds.
Enzymatic digests of plant protein include Phytone,
Soytone, soya peptone, and soy peptone. They are high in 10.4.3. Supplements
carbohydrate content and vitamins and can support rapid Many fastidious organisms grow in media that contain ad-
growth of most bacteria; however, acid production from the ditional undefined supplements. These substances include
carbohydrates may also cause a rapid decline in culture den- growth factors, blood components, and other compounds or
sity in the stationary phase, especially under fermentative extracts serving as sources for vitamins, amino acids, or un-
conditions. defined growth factors.
206 rn GROWTH

10.4.3.1. Blood and Serum aqueous extracts have proven useful in isolating planktonic
Many clinical isolates grow on media containing blood or Herpetosiphon in freshwater (32). Dry pellets of rabbit dung
blood components. Routine cultivation media for human or are boiled in water for 20 min, and the filtrate is adjusted to
animal pathogens contain 5 to 15%defibrinated blood. The pH 7.2 before adding to enrichment media for isolation of
most common source of blood is from sheep, although rab- appropriate organisms.
bit blood and horse blood are also used occasionally for cer-
tain fastidious microbes. A few organisms, such as members
of the genus HuemophiZus, may be inhibited by fresh sheep 10.5. NONNUTRITIONAL MEDIA
blood. Instead, chocolate medium (GC medium, ATCC COMPONENTS
medium 814)in which the blood is heated to 70 to 80C for
10.5.1. Agar
15 min is used. In addition, species of Huemophilus and
Neisseria require X factor (heme) and V factor (NAD). One of the major innovations in the growth of microbes
Serum can serve as an alternate to blood. Horse and was the use of agar as a solidifying agent for culture media,
bovine sera are widely used in cultivating fastidious or- an idea suggested by Fannie Eilshemius Hesse, the wife of an
ganisms, i.e., Jijxotrichia, Mycobacterium, Streptobacillus, and associate of Robert Koch (5). The ability to cultivate bac-
Treponema. Fresh rabbit serum is crucial for cultivating teria on solid media revolutionized almost every aspect of
Borrelia species. Lyophilized pooled rabbit serum is needed microbiology. With the exception of some marine bacteria,
for cultivating species of Leptospiru. Certain members of the agar is not digested or used as a nutrient source by most mi-
Mollicutes (Mycoplasma, Spiroplasma, Mesoplasma, and pleu- crobes.
ropneumonia-like organisms [mycoplasmas]) require serum Agar is the most commonly used gelling agent extracted
as a source for sterols to grow. from species of red algae. It is a sulfuric acid ester of the lin-
It is important to ensure that the blood and serum used ear gelactan (26). A valuable property of agar is that it
in formulating bacteriological media are free of pathogens melts at 80 to 95C but does not solidify until cooled down
and potential health risks, especially in light of the recent to 35 to 40C. Typically 15 g of agar per liter is used for solid
bovine spongiform encephalopathy outbreaks. Serum may culture media, and lower concentrations can be used to pro-
have to undergo heat treatment for inactivating certain duce soft or semisolid media. Most manufacturers sell differ-
proteins to prevent a possible inhibiting effect on microbes. ent grades of agar that vary in the amount of impurities,
Media containing blood and serum are stable and usable if such as trace elements and residual organic matter, that
stored properly under refrigeration. they contain. Noble agar is generally the highest grade and
most expensive. Agarose, typically used for gel electro-
10.4.3.2. Rumen Fluid phoresis, is an even more purified form of agar that can be
To cultivate some ruminal bacteria, complex media may used if it is suspected that impurities are a problem; how-
have to be supplemented with rumen fluid. Rumen fluid can ever, while agarose has a higher gelling strength than agar,
be obtained from universities and colleges with animal sci- it is also much more expensive.
ence or veterinary programs that have access to fistulated Other different types of solidifying agents include gela-
cows (this is a not a stressful experience for the cow). tin, gellan gum, Gelrite, and silica gels. Gelatin can be di-
Although the exact content of the rumen fluid is not gested by many organisms, so it is most useful when identi-
known and varies with the diet of the animal, it serves as a fying bacteria with proteolytic activity by observing
supplement of amino acids, peptides, vitamins, fatty acids, liquefaction of the gelatin medium. Silica gel is an inor-
carbohydrates, and other intermediate metabolites. Results ganic solidifying agent, useful in culturing autotrophs in the
may vary from batch to batch as well as with age. A mixture complete absence of organic substances. Silica gel can also
of volatile fatty acids can replace rumen fluid to fulfill the be used in testing the ability of heterotrophs to utilize cer-
nutritional requirements of some ruminal organisms tain organic compounds as single carbon sources, as well as
(Ruminobacter, Ruminococcus, and rumen treponemes) (9). in vitamin assays. Gelrite (Kelco Division of Merck & Co.,
Inc., San Diego, CA) or Phytagel (Sigma Chemical Co.)
10.4.3.3. Soil Extract has a higher melting temperature than agar, so it is used for
Many soil microbes grow better and sporulate more readily culturing thermophiles that grow at temperatures close to
in media containing soil extracts, although the composition lOOC,which will liquefy the agar. Unlike agar, Gelrite re-
is unknown and the reasons for the effects are not clear (7, quires a divalent cation (Mg2+ or Ca2+) to solidify and it
9).One standard soil extract uses African violet soil sup- solidifies very rapidly at about 90C, making it somewhat
plemented with sodium bicarbonate. This has been used as difficult to work with. One solution to this is to mix agar
a supplement to cultivate Azotobacter, Rhizobium, and some and Gelrite in a 1:3 ratio; this results in a solution that is
Bacillus species. easier to work with but that remains solid at temperatures of
up to 80 to 90C.Gellan gum has been shown to be effec-
10.4.3.4. Other Supplements tive for culturing novel soil microorganisms on solid
A number of undefined supplements are frequently added to medium (19).It requires a divalent cation, e.g., CaC12, for
mimic the natural habitat from where the organisms were solidification.
isolated. For example, tomato juice is included in media to
cultivate lactic acid bacteria (i.e., Lactobucillus, Leuconostoc, 10.5.2. Reducing Agents
Pediococcus, Bifidobacterium, and Propionibacterium). V-8 While reducing agents do not contribute directly to an or-
canned vegetable juice is supplemented in media for propa- ganisms nutrition, they are a necessity for the optimal
gating species of Actinomadura and Microspora, as well as growth of a number of strict anaerobes by lowering the
organisms isolated from plants (Pseudomom tolaasii) (9). redox potential of the medium to a point compatible with
Rabbit dung is another example. The pellets are often used the stability and activity of many cellular enzymes (see
as a bait in the enrichment of myxobacteria (33),and the chapter 14). Sodium sulfide is probably the most commonly
10. Nutrition and Media 207

used reducing agent. Liquid stock solutions are stable for 10.5.4.1. Inorganic Buffers
months when stored under Nz and can also be repeatedly
frozen and thawed. Sulfide reduces growth media rapidly. It 10.5.4.1.1. Phosphates
is important to remember that hydrogen sulfide gas is ex- Historically, phosphate is probably the most commonly
tremely toxic and is generated rapidly if sodium sulfide so. used buffering system, made by mixing the monobasic salts
lutions come in contact with acid. Likewise, stocks of of either KH2P04 or NaH2PO4 with their dibasic forms,
sodium sulfide should be filter sterilized and not autoclaved. K2HP04or Na2HP04, to provide buffering capacity in the
Another quite commonly used reducing agent is cysteine. range from pH 6 to 8. The exact pH depends upon the ratio
Both cysteine and sulfide can be used as sulfur sources by a of monobasic to dibasic salts. These buffers can cause pre-
number of prokaryotes; sulfide has an advantage in enrich- cipitation reactions with metals, principally Ca2+, Fe3+,
ment media of not being used as an energy source for fer- and Mg2+, especially above pH 7.0, resulting in these met-
mentative microbes. Another very effective reducing agent als no longer being available for microbial nutrition.
for methanogens is 2-mercaptoethanesulfonic acid or coen-
zyme M, which is, itself, a key intermediate in methane for- 10.5.4.1.2. Bicarbonate-C02
mation. A stock solution of 5% (wtlvol) coenzyme M can The bicarbonate-CO2 buffering system is the most wide-
be made in deionized water and filter sterilized. The stock spread in nature, and as a result it is commonly used for
solution is diluted 1 : l O O into the culture medium. methanogens, sulfate-reducing bacteria, and anoxygenic
Coenzyme M takes some time to fully reduce the culture photosynthetic bacteria, as well as a variety of other
medium; between 30 and 60 min is common; however, it lithotrophic prokaryotes. It is especially advantageous for au-
has proven a very effective reducing agent for a wide vari- totrophic organisms since it provides both buffering capacity
ety of methanogens (D. Boone, personal communication). and a source of C. Bicarbonate buffers are more cumbersome
Other reducing agents that are used in more specialized ap- to prepare than most other buffers; however, once proce-
plications include iron sulfide (FeS), titanium citrate, and dures are established, they quickly become routine and re-
hydrogen in the presence of palladium chloride (PdC12) quire little more time than using more conventional labora-
catalyst; these are discussed in more detail in chapter 14. tory buffers. Typically a calculated concentration of sodium
bicarbonate (usually between 10 and 50 mM) is added to the
10.5.3. Redox Indicators medium, either before or after autoclaving, and then the pH
Redox indicators are dyes that either change or lose color is adjusted by gassing the medium with C02 after autoclav+
depending on the redox potential of the medium. Important ing. Using the equations in chapter 14, it is possible to cal-
factors to consider with any redox indicator are its mid- culate the ratios of bicarbonate:COz that should be used, or
point potential and potential toxicity. The former consider- this can be done empirically by gassing a known volume of
ation will guide the selection of the most appropriate redox medium with a known flow rate of C02 for different times
indicator when a particular redox potential for a medium is until the final pH is achieved. Bicarbonate buffers must be
desired. These dyes are not measures of specific redox po- used in a contained culture vessel, otherwise the COz will
tentials. Instead, they indicate whether the redox potential escape to the atmosphere, causing the pH to rise as the bi-
of the medium is above or below their mid-point potential. carbonate dissociates to equilibrate the [COJ. This is espe-
The most commonly utilized redox indicator is resazurin. At cially important if the bicarbonate is added prior to auto-
neutral pH, this dye is blue under aerobic conditions. claving. In general, autoclaving is not recommended for
However, once the O2is gassed out of the medium and it is bicarbonate buffers, because at high concentrations enough
heated either by boiling or autoclaving, an irreversible re- C02 may be driven out of the solution to pressurize the
action leads to the formation of resorufin, which is pink. headspace and create an explosion hazard in glass bottles.
Upon addition of a reducing agent, such as sodium sulfide, A n advantage of this buffering system is that it can be used
capable of poising the redox around - 110 mV or less, the over the range of pH 6 to 8 with few problems, such as metal
resorufin (Eo= -51 mV) becomes clear. Reduced forms of precipitation reactions.
resazurin are sensitive to photo-oxidation and may produce 10.5.4.2. Organic Buffers
toxic intermediates; therefore, resazurin should not be used
in anaerobic phototrophic media that will be illuminated. 10.5.4.2.1. Tris
Redox indicators are discussed in more detail in chapter 14. Tris-hydrochloride is a commonly used biological buffer.
Its best buffering capacity is between pH 7.5 and 8.5, so if it
10.5.4. Buffers is to be used at pH 7.0 a concentration in the range of 50
Many metabolic reactions coupled to cell growth result in mM should be used. Tris is prepared by either adjusting the
the consumption or release of acidic or basic products, which pH of the Tris-hydrochloride with sodium hydroxide to the
can result in a substantial shift in the pH of the medium. For desired pH or mixing a ratio of Tris-HC1 with a basic form,
example, most fermentations result in the production of or- e.g., tris[hydroxymethyl]aminomethane,to achieve the de-
ganic acids which can easily reduce the pH of a culture sired pH. The Sigma catalog lists the ratios of these buffers
medium to below pH 5. The most common way to alleviate for different pHs. Tris buffers are temperature sensitive; for
this problem is to add a buffering agent to the medium. The every degree Celsius above 25"C, Tris buffers decrease by
important things to consider in choosing a buffer are (i) its approximately 0.025 pH unit; for every degree Celsius
buffering range, (ii) its potential toxicity, (iii) its capacity to below 25"C, there is an approximate increase in pH of 0.03
bind metals or otherwise interact with other media compo- unit. Tris buffers are also known to react with some metal
nents, and (iv) the potential to be metabolized by the or- cations, e.g., Ni2+ and Ca2+.
ganisms that are growing in it. In chapter 14, there is an ex-
cellent description of the chemistry and buffering capacity of 10.5.4.2.2. Good's Buffers
buffers, and so this chapter highlights only some of the The zwitterionic, organic buffers originally synthesized
buffers commonly used in media preparation. by Good et al. (14) have continuously grown in popularity
208 rn GROWTH

for use in microbiological media. There are a number of tions can be avoided by physically separating the reactants.
these available from commercial sources, and together they Here are a few general rules to follow when autoclaving.
can cover the pH range from about pH 5.5 to 11. Three of
Sterilize glucose separately from amino acids, peptones,
the more common examples are MES [2-(N-morpholino)
or compounds containing phosphate.
ethanesulfonic acid], with a pH range of 5.5 to 6.7; HEPES
[N-(2-hydroxyethy1)piperazine-N'-(4-butanesulfonic Autoclave phosphates separately from amino acids, pep-
acid)], with a pH range of 6.8 to 8.2; and TAPS {N- tones, or other mineral salt components.
[Tris(hydroxymethyl)methyl]-3-aminopropanesulfonic Alkaline media (pH, >7.6) should not be autoclaved.
acid}, with a useful pH range of 7.7 to 9.1. These acidic Prepare the medium at neutral pH and adjust it to the de-
buffers are simply added to the medium prior to autoclaving sired alkalinity after it has cooled down from autoclaving.
at an appropriate concentration, and the pH is adjusted Acidic media (pH, <6.0) also should not be autoclaved.
with sodium or potassium hydroxide. They are chemically Prepare the agar and the acidic ingredients separately in
inert and nontoxic. Their only drawback is that they are double strength (i.e., one liter's worth of ingredients dis-
relatively expensive, especially if large volumes of a medium solved in 500 ml of water, and 15 or 20 g of agar in 500
are required. ml of water), and then autoclave them. After cooling
down to about 50"C, combine the two solutions asepti-
10.5.4.2.3. Others cally before pouring to plates or distributing into tubes.
Organic acids, for example acetate and citrate, can be If precipitation problems are encountered with an agar-
used as buffering agents at low pH, 4.0 to 5.5, when there containing mineral salts solution, try autoclaving the
are no good inorganic or synthetic buffers available. These agar separately from mineral salt solutions. There may be
have the obvious drawback that they are common microbial calcium and magnesium salts present in certain brands of
food sources. Protein and amino acids also have buffering agar which precipitate with other mineral salts.
capacity; in fact most complex media do not have specific
buffers added, in part due to the buffering capacity of the The sterilizing effectiveness of the autoclave machines
proteinaceous components of these media. should be routinely monitored. This is usually performed by
using a commercially available product, Kilit (BBL), which
contains spores of Bacillus stearothermophilus.
10.6. PREPARATION OF MEDIA
10.6.1.2. Filtration
10.6.1. Sterilization For heat-labile and volatile liquids, filtration is the method
After the ingredients of a given medium are weighed out of choice for sterilization. A number of filtration systems are
and dissolved, either together or separately, they must be available commercially for different volumes of solution.
sterilized. While this step seems easy, most microbiologists The membrane filters are convenient to use, and their com-
have experienced the frustration of carefully mixing a positions need to be compatible with the chemical nature of
complex set of ingredients, adjusting the pH, dispensing it the solutions to be sterilized. Most media and components
into culture flasks, and autoclaving the medium, only to are compatible with cellulose acetate or nitrocellulose filters.
find upon opening the autoclave an hour later a precipi- Nylon or Teflon membranes should be used for solvents.
tated mess. To avoid this outcome, thought must be given Most bacteriological media are passed through filters
to the potential reactions of media ingredients at the heat with a pore size of 0.2 pm. Most microorganisms are trapped
and pressure in an autoclave, especially as a function of by this pore size; however, smaller organisms (i.e., Pseu-
pH. If precipitation problems are encountered, or sus- d o m o m diminuta, mycoplasmas, rickettsias, and viruses)
pected, it is best to either filter sterilize the medium or pass through these filters. If this is a concern, 0.l-Fm-pore-
heat sterilize the components separately and add them to- size filters can be used instead.
gether after autoclaving. In most cases the pH of the If carbohydrates are to be used in sole-carbon source test-
media is adjusted prior to sterilization; however, it is some- ing procedures, it is recommended to filter sterilize them,
times necessary to check and readjust the pH after heat rather than autoclave. As mentioned above, vitamin and
sterilization. It is also important to sterilize the medium as mineral stock solutions should be filter sterilized.
soon as it is prepared, to minimize the opportunity for
growth of microbial contaminants. Several methods used 10.6.1.3. Tyndallization
for sterilization are as follows. Tyndallization method alternates heating and cooling by
exposing the solutions to steam, or heating to 100C in a
10.6.1 . I . Autoclaving water bath, for 30 min each day for three successive days. In
Autoclaving utilizes high-pressure steam to kill microorgan- this case the vegetative cells of heat-resistant endospore-
isms. This is probably the most commonly used method of forming bacteria (e.g., Bacillus) are killed by heating at
sterilization, exposing the media to steam at 115 lbs/in2 and 100C. The endospores germinate when the medium cools
121"C for a minimum of 15 min. It is important that the in- down, and they in turn are killed by the subsequent heat
ternal temperature of the liquid in the autoclave reaches treatment. This method is useful for solutions which con-
121C for the set time period of sterilization. Thus, longer tain powdery substances and is most often used with media
running times are required for large loads and/or containers containing elemental sulfur, which turns into an unwork-
of larger volume. able mass when autoclaved.
Although autoclaving is a convenient and effective
method of sterilization, many formulations cannot tolerate
the high heat and pressure that contribute to undesirable 10.7. QUALITY CONTROL TESTING
chemical reactions (e.g., precipitation and browning) and For routine maintenance and characterization media, espe-
breaking down of components. In most instances these reac- cially in clinical and quality control laboratories, it is
10. Nutrition and Media 209

important to ensure that media perform properly. Media best, yielding the most rapid growth rates and highest cell
purchased from commercial companies usually have gone yields. Many standard microbiological media can be pur-
through quality control procedures; however, it is always chased premixed from commercial sources. This makes
important to record the lot number of any individual media preparation easy and fast and ensures a quality prod-
medium in case problems arise. For media that are produced uct. For more specific physiological or genetic studies more
in-house, a testing protocol should be established. The pro- selective or defined media are often used. It is also impor-
tocol should include standard operating procedures for test- tant to remember that genetic drift or alteration is a real
ing, the expected results, and criteria for acceptance. and often rapid phenomenon, so organisms selected for a
A n important aspect of quality control includes quality specific trait should always be maintained on a medium that
control organisms that will produce a positive or negative selects for that trait.
reaction on the media to be tested. These organisms should
be obtained from a reputable source. For example, the 10.8.2. Phototrophs
ATCC has many quality control organisms, and every effort
is made to minimize transfers and authenticate their identi- 10.8.2.1. Oxygenic Phototrophs
I -

ties and properties. Cyanobacteria are nutritionally among the real minimalists
Frozen stocks (in 10% [vol/vol] glycerol or 5% [vol/vol] of the microbial world, requiring little other than a few min-
dimethyl sulfoxide) of the quality control organisms should eral salts and light to grow. Most are capable of synthesizing
be made and stored. After three passages of the working all of their vitamins, although a few require vitamin BIZ.
stock culture, it is necessary to go to another frozen stock Despite these minimal media requirements, a number of
culture. Minimizing transfers is crucial in the performance cyanobacteria have proven quite challenging to grow, in part
of these quality control organisms. because they can be quite susceptible to toxic compounds
A good source of information on quality control of that contaminate media components. For example, a num-
media is the Difco Manual ( 3 ) , which provides the cultural ber of marine strains, especially Trichodesmium spp., are ex-
reactions to each of its media and lists the organisms used quisitely sensitive to Cu, and so in some cases extraordinary
for testing as well as positive and negative results. The care needs to be taken in preparing media and trace ele-
incubation conditions are also included. This manual can ments solutions (see Waterbury [37] for a much more de-
serve as a guide to establish a media quality control pro- tailed discussion of media preparation for cyanobacteria).
gram. Light requirements are generally met by cool or warm white
fluorescent lamps; lighting intensities should try to mimic
those of the isolates natural environment.
10.8. MEDIA FOR SPECIFIC GROUPS
In this section we consider media designed for some major 10.8.2.2. Anoxygenic Phototrophs
metabolic groups of prokaryotes. This discussion is repre. A large number of microbes are capable of carrying out pho-
sentative but by no means exhaustive; references and tosynthesis under anaerobic conditions using reduced S
sources with more detail are provided for different groups. compounds or H2in place of H 2 0 as a source of electrons
for the incorporation of C02 into cell biomass. Two major
10.8.1. Media for Heterotrophic Microbes groups of obligate anaerobic photosynthetic bacteria are the
In general when we think of a bacterial growth medium, we purple sulfur photosynthetic bacteria represented by the fam-
think of a medium containing an organic energy and carbon ily Chromtiaceae and the green sulfur bacteria of the family
source. If an organic compound can be made available to Chbrobiaceae. Interestingly, both of these groups of organ-
microbes, there is a good chance that there is a prokaryote isms are found primarily in aquatic habitats including
that can extract energy from it. Major classes of organic anoxic water columns, littoral sediments, or microbial mats
compounds that are used for growth are listed in Table 3. and have similar media requirements; however, phylogenet-
When a complex medium is used, several of these or- ically they are very distinct. The Chlorobiaceae form their
ganic sources may be available simultaneously, whereas de- own phylum within the domain Bacteria, while the
fined or minimal media are generally designed to provide Chromtiaceae are members of the gamma-proteobacteria.
only a single primary energy source. In general, for routine Both groups of organisms can be grown with a mineral salts
growth of a microbe in the laboratory a complex medium is medium that contains trace elements, vitamin BIZ, and

TABLE 3 Major classes of organic growth compounds


Comuound Examples Comments
Carbohydrates Glucose, maltose, ribose Excellent energy sources, readily soluble
Polysaccharides Cellulose, starch May be difficult to solubilize
Proteins/amino acids Peptones, albumin Excellent energy sources
Lipids Palmitic acid, stearic acid Rich energy sources, although limited range of organisms;
solubility may be a problem
Organic acids Acetate, butyrate pH of medium must be adjusted, buffers are recommended
Alcohols Ethanol, sorbitol Some are toxic at higher concentrations
Aromatic hydrocarbons Benzoate, toluene Low solubility, volatility, toxicity
Aliphatic hydrocarbons Hexadecane Low solubility, volatility
210 GROWTH

sodium sulfide as a source of sulfide. A n important physio- the headspace of the culture vessel. Glass media vessels
logical trait of most genera of the green sulfur bacteria is a should be vented regularly with a needle to prevent the
preference for growth under low light intensity. See the buildup of potentially explosive concentrations of gas. An
chapter by Overmann (28) in The Prokuryotes, for an up-to- anaerobic gassing station is essential for preparing
date and detailed discussion of the cultivation and lifestyle methanogen medium with prepurified 100% Nz and 80%
of the Chlorobiaceae, and the chapter by Pfennig and Truper H2/20% C02or 70% N2/30% C02.The gas must be com-
(29) for a discussion of cultivation of Chromatiuceae,espe- pletely oxygen free; if good-quality gas is not available,
cially vis&vis light requirements. traces of O2can be removed by passing the gas stream over
hot copper turnings or purchasing a commercially available
10.8.3. Biodegradation oxygen trap. See the paper by Sowers and No11 (34) for a
The capacity for microbes to degrade and in many cases min- more detailed discussion of preparing a laboratory for the
eralize xenobiotics and organic pollutants is one of their most growth of methanogens.
impressive traits. This is a large and ever-growing field and so
is dealt with only in a cursory way here. Classes of toxicant 10.8.5. Methane-Oxidizing Bacteria
compounds include aromatic hydrocarbons (e.g., benzene, to- The methane-oxidizing or methanotrophic bacteria are a
luene, and xylene), aliphatic hydrocarbons (e.g., gasoline), ubiquitous group of organisms capable of oxidizing methane
chlorinated aromatic hydrocarbons (e.g., polychlorinated in the presence of air for growth (18). They are commonly
biphenyls), and chlorinated aliphatics (e.g., tetrachloroeth- found in aquatic and terrestrial environments often associ-
ylene). Aside from their inherent toxicity, the property that ated with aerobic-anaerobic interfaces, where they can take
makes many of these compounds recalcitrant is their insolu- advantage of the methane that is supplied by methanogen-
bility and/or volatility. This has to be taken into account in esis in the anaerobic environment. These organisms are
preparing the media. In some cases the compound of interest unique in being able to incorporate the formaldehyde that
may need to be dissolved in a potent organic solvent, for ex- is formed as an intermediate of methane oxidation directly
ample, hexadecane, and added to the medium. In the case of into cell carbon; thus, true methanotrophs do not require
toluene, the compound is sparingly soluble and quite volatile; an exogenous supply of organic matter.
therefore, it should be maintained in vapor phase in a properly A much larger group of bacteria, the methylotrophs are
sealed culture vessel to prevent its depletion by volatilization. capable of growing on methanol by a similar mechanism.
Again, some of these compounds are themselves organic sol- Most methanotrophs are obligate, requiring either methane
vents, and so they can degrade or partition into rubber stop- or methanol for growth; on the other hand methylotrophs
pers, requiring the use of Teflon-coated caps. For enrichment that are unable to grow on methane are generally metaboli-
studies, often a mineral medium is used so that the only car- cally diverse and often are able to grow on a variety of organic
bon and energy source available is the organic compound in compounds as well. Methanotrophs can be grown on a min-
question. In the case of anaerobic organisms capable of eral salts medium with trace metals; some require vitamins,
dechlorination, the chlorinated compound can serve as the others do not. Methanol is the preferred electron donor, since
electron acceptor and the organism may use an organic com- the alcohol can easily be dissolved in the medium. Methane
pound, for example, pyruvate, as the electron donor; thus, the is necessary, especially in the enrichment stage, to select for
dechlorination is the result of anaerobic respiration (10). true methanotrophs and must be added as a gas by bubbling
a stoppered tube for a short time with a gas mixture. Consult
10.8.4. Methanogenesis a more specialized text (18) for advice on specific methane-
Methanogens, which constitute a large and well-studied containing gas mixtures to use for enrichment and cultiva-
group of Archueu, are unique in the microbial world for their tion of different methanotrophs. It is important to remember
ability to form methane. Despite being metabolically lim- that methane (natural gas) is very flammable.
ited only to methane production, methanogens are a physi-
ologically and morphologically varied lot, and there is a 10.8.6. Sulfur Metabolism
large literature devoted to them (34,36,40). Methanogens
are among the strictest of anaerobes, meaning that they re- 10.8.6.1. Sulfate Reduction
quire the addition of a reducing agent (see section 10.5.2) Sulfate reduction is the most common mechanism for car-
to the growth medium. It is best to assume that they cannot bon mineralization in anaerobic marine sediments, making
tolerate any exposure to oxygen, so a redox indicator should it, on a global scale, an important pathway for both sulfur
be included as well. Specially adapted culture vessels have and carbon cycling. The sulfate-reducing bacteria (SRB)
been designed for growing methanogens, the most notable are very diverse; however, known isolates are concentrated
being the Balch tube, an 18- by 150-mm glass tube with a in the delta-proteobacteria. There are several excellent re-
serum vial-type lip that can accommodate a thick butyl rub- views on the feeding and care of SRB (20, 30). In general,
ber stopper and be sealed with an aluminum crimp cap. This these organisms require a source of sulfate, an organic elec-
allows the tubes to be pressurized with the COZ-H2 gas mix- tron donor, and strictly anaerobic conditions, although ad-
tures that many of these organisms require for optimal dition of a reducing agent is usually not necessary. Sulfate is
growth. Although methanogens are comprised of over a generally provided by simply dissolving sodium sulfate into
hundred different species, metabolically they grow on a lim- the growth medium. Organic acids like acetate or lactate
ited range of substrates. The classic substrates are C02plus are commonly used as electron donors for SRB, but a wide
HZ. Another well-studied group are the aceticlastic variety of alcohols and fatty acids can also be used. Se-
methanogens that can utilize acetate. Other potential sub- lectivity for certain species can be achieved by using a spe-
strates for methanogens are methanol, ethanol, methylated cific electron donor. Some SRB, for example, Desulfovibrio
amines, and methylated sulfides. In every case, CH, is the desulfuricuns and Desulfovibrio vulgaris, are capable of litho-
primary product. It is important to remember when using trophic growth on SO4by coupling H2 oxidation or formate
the latter set of soluble substrates that CH4 will build up in oxidation to sulfate reduction.
10. Nutrition and Media 21 1

10.8.6.2. Sulfur Disproportionation some aromatic compounds. In addition some strains can ox-
This is a newly discovered metabolism that results in the idize H2 and formate as well and grow as chemolithoau-
disproportionation of either elemental sulfur or thiosulfate totrophs. Addition of extracellular quinones, principally as
to sulfide and sulfate. In the case of sulfur the reaction is not the humic acid analog, anthraquinone-2,6-disulfonate
energetically favorable by itself; however, if there is Fe(II1) (AQDS) has also been shown to stimulate Fe reduction by
present in the medium that can react with the sulfide that acting as an intermediary electron shuttle (8).
is produced and remove it from the medium as FeS, the low
concentration of the sulfide product drives the reaction to
10.8.7.2. Manganese Reduction
be thermodynamically favorable. Thus far, only one organ- Manganic oxides are also excellent electron acceptors for
ism, Desulfocapsa sulfexigens, has been described as a true anaerobic respiration, although relatively less common
chemolithoautotroph that grows by sulfur disproportiona- than iron. The most common Mn(IV) oxidation state does
tion; however, it is likely that others await discovery (12). not exist in a chelated form, so Mn minerals such as col-
Several sulfate reducers are known to also be able to grow loidal Mn(IV) or vernadite (Mn02)must be prepared syn-
by disproportionation of thiosulfate, although these organ- thetically and added as the insoluble oxides to the medium
isms require a source of organic carbon as well (30). (21). The carbon sources used may be similar to those used
for Fe reduction.
10.8.6.3. Sulfur Oxidation
The oxidation of different reduced forms of S is, in most
10.8.7.3. Arsenic and Selenium
cases, a thermodynamically favorable reaction; thus, there Arsenic and selenium are metals known for their toxicity
is a broad range of lithotrophic bacteria and archaea that and, while naturally occurring, are most often associated in
can derive energy by using reduced S compounds as elec- more concentrated forms with industrial processes such as
tron donors (20). The most commonly used form of sulfur mining and the production of semiconductors. The oxidized
for growth of these is thiosulfate, since the sodium salt is form of arsenic is arsenate, As(V), which can be reduced to
readily soluble at neutral pH and can be autoclaved. Other arsenite, As(II1). The oxidized forms of selenium are sele-
sulfur compounds are tetrathionate, which can be made sol- nate, Se(VI), and selenite, Se(IV), which can be reduced to
uble in acidic stock solutions, and elemental sulfur, which is elemental selenium, Se(0). A t present there are at least five
completely insoluble and must be sterilized by tyndalliza- species of bacteria that are known to respire anaerobically
tion (10.6.1.3). Hydrogen sulfide (H2S) gas may also be by coupling oxidation of organic acids to Se reduction (27).
used. Please remember that H2S is very poisonous and it is Other bacteria that can reduce Se(V1) or Se(1V) are
also critical to keep acids away from sodium sulfide solu- known, but they are not able to grow via metal reduction.
tions, since the acidification causes the sulfide to rapidly Sulfurospirillumbamesii is also able to respire on As(V), and
form volatile H2S gas. several other species have also recently been shown to be
capable of dissimilatory As reduction. The oxidized forms of
10.8.7. Dissimilatory Metal Reduction both As and Se are available as salts and require no special
handling. It is important to remember that the soluble
10.8.7.1. Iron Reduction forms are toxic.
Over the past decade a growing number of anaerobic bacte-
ria have been recognized that can utilize ferric (Fe) oxides as 10.8.8. Fe Oxidizers
an electron acceptor for carrying out anaerobic respiration Prokaryotes that can utilize Fe(I1) as an energy source are
(24). Two organisms that are well-known for carrying out another unusual group. Physiologically these organisms fall
this reaction are Geobucter metallireducens and Shewanella into two categories, acidophiles and neutrophiles. Fe(11) is
oneidensis; these organisms represent the extremes of meta- quite stable in the presence of air at a pH of 4 or less, and
bolic diversity that are associated with these processes as thus, acidophilic Fe oxidizers largely avoid competition
well. G. metallireducens has a limited metabolic repertoire, with abiotic oxidation. At neutral pH, chemical oxidation
consisting of electron acceptors or donors that it can use for of Fe(11) predominates unless the oxygen concentration is
growth. S. oneidensis is a facultative anaerobe and is meta- kept very low, which is the case in the oxic-anoxic bound-
bolically very diverse, capable of utilizing a wide variety of ary areas where these organisms grow. Interestingly, acido-
electron donors and acceptors. However, when respiring on philic Fe oxidizers, such as Acidothiobacillus ferrooxidans and
Fe(II1) the two organisms have quite similar requirements: Leptospirillum ferrooxidans, cannot tolerate the presence of
both require a source of amorphous Fe oxide, an electron organic compounds, including agar, in their medium. As a
donor, and buffering capacity. There are a variety of sources result these organisms are normally maintained in a liquid
of Fe(II1) that have been used (21). Chelated forms of mineral salts medium. Clever plating methods have been
Fe( 111), including ferric citrate, Fe-pyrophosphate, and Fe- employed that use acidophilic heterotrophs to scavenge or-
nitrilotriacetic acid are the easiest to work with and can ganic matter in the agar; these are overlayered with a thin
all be purchased as powders that can be dissolved in the cul- hard layer of agar that can then support the growth of the
ture medium. Especially during enrichment studies it is Fe oxidizers (17) .
probably best to work with more naturally occurring forms Neutrophilic Fe oxidizers can be grown in media de-
of Fe oxides, including synthetically produced Fe oxide, or signed to create opposing gradients of Fe(11) and 0 2 ; the mi-
naturally produced ferrihydrite (35); these are insoluble crobes grow as a band at the oxic-anoxic interface (1l). The
forms that require that the cells attach to the oxides during most commonly employed method is to make a synthetic
growth. FeS solution which is mixed with 1% agarose to make a
While acetate is the most oft-used electron donor for the thin, solid layer in the bottom of a tube. This is overlayed
growth of Fe-reducing bacteria, a wide range of organic with a semisolid mineral salts medium (containing 0.15%
compounds have been shown to be coupled to Fe reduction, agarose) with an air space on top, which naturally causes
including short- and long-chain fatty acids, alcohols, and Fe( 11) to diffuse up from the bottom and O2to diffuse down
212 GROWTH

from the air. The lithotrophic Fe oxidizers grow at the in- 10.9. SOURCES
terface, and the gel prevents the cells from sinking out of
the interface zone. FeC03 or FeC12 can be substituted for 10.9.1. Sources of Manufacture
FeS in these media. Becton Dickinson Microbiology Systems, 7 Loveton Circle,
Sparks, MD 21 152-0999. (Tel) 800-638-8663.
10.8.9. Nitrifying Bacteria www.BD.com
The nitrifiers are another group of phylogenetically diverse Bellco Glass, Inc. 340 Edrudo Rd., Vineland, NJ 08360.
bacteria that share a quite limited lithotrophic metabolism, (Tel) 800-257-7043.
the ability to gain energy from the oxidation of ammonia or www.bellcoglass.com (Good source for anaerobic culturing
nitrite (1, 6). The ammonia oxidizers, consisting of the glassware)
beta-proteobacteria Nitrosomonas and related genera, grow BioMerieux, Inc., 595 Anglum Rd., Hazelwood, MO
in mineral media with NH4C1 or (NH4)2S04as an energy 63042-2320. (Tel) 800-638-4835.
source. The nitrite oxidizers, consisting of the alpha-
proteobacteria Nitrobacter spp. and of the Nitrospira spp., www.biomerieux-vitek.com
which form a separate phylum, grow on NaNOz or KNOZ as Difco Laboratories: see Becton Dickinson Microbiology
an energy source. Many of these organisms have slow dou- Systems.
bling times (12 to 24 h). Ammonia oxidation results in Gibson Laboratories, Inc., 1040 Manchester St., Lexington,
acidification of the medium, so buffering is required, usually KY 40508. (Tel) 800-477-4763.
with HEPES or CaC03, and pH indicators can also be www.gibsonlabs.com
added as an indicator that growth is occurring. All known Oxoid Inc., Suite 100, 1926 Merivale Rd., Nepean, Ontario
nitrifiers can grow autotrophically; thus, the addition of car- K2G 1E8, Canada. (Tel) 613-226-1318.
bonate to medium can be a source of carbon. Some strains
have been shown to be capable of incorporating organic www.oxoid.co.uk
compounds, thus carrying out mixotrophic growth; how- Raven Biologics Laboratories, 8607 Park Dr., P.O. Box
ever, in some cases excess organic compounds can be toxic 27261, Omaha, NE 68127. (Tel) 800-728-5702.
to these organisms. www.ravenlabs.com
Remel Inc., 12076 Santa Fe Dr., P.O. Box 14428, Lenexa,
10.8.1 0. Halophiles KS 66215. (Tel) 800-255-6730.
Halophilic bacteria and archaea require the addition of salt www.remelinc.com
to the medium, from 10 to 30% by weight, depending on Sigma, P.O. Box 14508, St. Louis, MO 63178. (Tel) 800-
the organism. In most cases, NaCl is the primary form of 521-8956.
salt; however, many halophilic archaea have requirements
wwwsigma-aldrich.com
for Mg salts as well. These organisms are nutritionally di-
verse and in general do not have any other extraordinary
nutritional requirements beyond the addition of salt. The
high salt concentrations can present problems when work- 10.9.2. Culture Collections
ing with agar-solidified medium, as the salt causes the agar These sites have search capacities for finding media recipes.
to be softer than normal. NaCl also affects the pH of the American Type Culture Collection (ATCC)
medium due to the high ionic strength. http://www.atcc.org
Deutsche Sammlung von Mikroorganismen und
10.8.1 1. Oligotrophs Zellkulturen GmbH (DSMZ)
While the physiological concept of microbes capable of http://www.dsmz.de
growing under, or requiring, very low nutrient concentra-
Japan Collection of Microorganisms (JCM)
tions is not new, in the past few years good progress has
been made in using very low nutrient media to enrich and http://www.jcm.riken.go.jp
grow bacteria that had previously been thought to be un-
culturable. A nice example of this is the enrichment and
isolation of members of the Sarl 1 clade of marine bacte- 10.9.3. Others
ria. This clade had been identified through analysis of Cyanobacteria
16s rRNA clone libraries as ubiquitous in the open
ocean; however, these organisms resisted attempts at cul- http://www-cyanosite.bio.purdue.edu/
tivation. By using very dilute seawater amended with mi- Provides media recipes and information on culturing a wide
cromolar or smaller amounts of nitrogen and phosphorous range of cyanobacteria including prochlorophytes.
and organic substrates, Rappe et al. were successfully able Methanogens
to cultivate members of Sarll (31). A similar approach http://me thanogens.pdx.edu/
that uses a dilute gel-encapsulated medium has been used A culture collection of methunogens; provides media recipes and
to cultivate a number of novel bacteria from the Sargasso detailed information on cultivation.
Sea and from soil (39). Another recent study used pro-
gressively diluted nutrient media to aid in the isolation of Biodegradation
previously uncultured, but numerically prevalent, ultra- http://umbbd.ahc.umn.edu/
microbacteria from freshwater lakes (16). The secret to Does not provide media recipes, but does have a wealth of in-
the success of these types of studies is in designing a formation on the metabolic pathways of organisms that break
growth medium that carefully simulates both qualitatively down most toxic organic compounds, and includes links to
and quantitatively the natural habitat of the organisms the culture collections where specific media recipes can be
under study. found.
10. Nutrition and Media H 213

10.10. REFERENCES and isolation in pure culture of novel members of the


1. Abeliovich, A. 2002. The nitrite-oxidizing bacteria. In divisions Acidobacteria, Actinobacteria, Proteobacteria, and
M. Dworkin (ed.), The Prokaryotes: An Evolving Electronic Verrucomicrobia. Appl. Environ. Microbiol. 68:2391-2396.
Resource for the Microbiological Community. Springer- 20. Kelly, D. P., and A. P. Wood. 1998. Microbes of the sul-
Verlag, New York, NY. fur cycle, p. 31-57. In R. S. Burlage, R. A. Atlas, D. Stahl,
2. Amend, J. P., and E. L. Shock. 2001. Energetic of overall G. Geesey, and G. Saylor (ed.), Techniques in Microbial
metabolic reactions of thermophilic and hyperther- Ecology. Oxford University Press, New York, NY.
mophilic Archaea and Bacteria. FEMS Microbiol. Rew. 25: 21. Kostka, J. E., and K. H. Nealson. 1998. Isolation, culti-
175-243. vation and characterization of iron- and manganese-reduc-
3. Anonymous. 1998. Difco Manual, 11th ed. Becton ing bacteria, p. 58-78. In R. S. Burlage,.R. A. Atlas, D.
Dickinson Microbiology System, Sparks, MD. Stahl, G. Geesey, and G. Saylor (ed.), Techniques in
4. Atlas, R. M. 1996. Handbook ofMicrobiologicalMedia, 2nd Microbial Ecology. Oxford Press, New York, NY.
ed. CRC Press, Boca Raton, FL. 22. Leadbetter, J. R., T. M. Schmidt, J. R. Graber, and J. A.
5. Black, J. G. 1999. Growth and Culturing of Bacteria. John Breznak. 1999. Acetogenesis from Hz plus COZ by spiro-
Wiley & Sons, Inc., New York, NY. chetes from termite guts. Science 283:686489.
6. Bock, E., and M. Wagner. 2002. Oxidation of inorganic 23. Lev, M. 1959. The growth-promoting activity of compounds
nitrogen compounds as an energy source. In M. Dworkin of the vitamin K group and analogues for a rumen strain of
(ed.), The Prokuryotes: An Evolving Electronic Resource for Fusifmes nigrescens.1.Gen. Microbiol. 20:697-703.
the Microbiological Community, 3rd ed. Springer-Verlag, 24. Lovley, D. R. 2000. Fe(II1) and Mn(IV) reduction, p. 3-
New York, NY. 30. In D. R. Lovley (ed.), Environmental Microbe-Metal
7. Bridson, E. Y. 1978. Natural and synthetic culture media Interactions. ASM Press, Washington, DC.
for bacteria, p. 91-281. In M. Rechcigl (ed.), CRC 25. Madigan, M. T., J. M. Martinko, and J. Parker. 2003.
Handbook Series in Food and Nutrition, vol. 111. CRC Press, Brock Biology of Microorganisms. Prentice-Hall, Upper
Cleveland, OH. Saddle River, NJ.
8. Coates, J. D., D. J. Ellis, E. Roden, K. Gaw, E. L. Blunt. 26. Matsuhashi, T. 1998. Agar, p. 335-375. In S. Dumitriu
Harris, and D. R. Lovley. 1998. Recovery of humics- (ed.), Polysaccharides: Structural Diversity and Functional
reducing microorganisms from a diversity of sedimentary Versatility. Marcel Dekker, New York, NY.
environments. Appl. Enoiron. Microbiol. 64: 1504-1509. 27. Oremland, R. S., P. R. Dowdle, S. Hoeft, J. P. Sharp, J. K.
9. Cote, R. J., and R. L. Gherna. 1994. Nutrition and Schaefer, L. G. Miller, J. S. Blum, R. L. Smith, N. S.
media, p. 155-178. In P. Gerhardt, R. G. E. Murray, W. A. Bloom, and D. Wallschlaeger. 2000. Bacterial dissimilatory
Wood, and N. R. Krieg (ed.), Methods for General and Mo- reduction of arsenate and sulfate in meromictic Mono Lake,
lecular Bacteriology. ASM Press, Washington, DC. California. Geochim. Cosmochim. Acta 64:3073-3084.
10. DeWeerd, K. A., L. Mandelco, R. S. Tanner, C. R. 28. Overmann, J. 2002. The family Chlorobiaceae. In M.
Woese, and J. M. Suflita. 1990. Desulfomonile tiedjei gen. Dworkin (ed.), The Prokaryotes: An Evolving Electronic
nov. and spa nov., a novel anaerobic, dehalogenating, Resource for the Microbiological Community, 3rd ed.
sulfate-reducing bacterium. Arch. Microbiol. 154:23-30. Springer-Verlag,New York, NY.
11. Emerson, D., and C. Moyer. 1997. Isolation and charac- 29. Pfennig, N., and H. G. Truper. 2002. The family
terization of novel iron-oxidizingbacteria that grow at cir- Chromatiuceae. In M. Dworkin (ed.), The Prokuryotes: An
cumneutral pH. Appl. Enwiron. Microbiol. 63:4784-4792. Evolving Electronic Resource for the Microbiological
12. Finster, K., W. Liesack, and B. Thamdrup. 1998. Community, 3rd ed. Springer-Verlag,New York, NY.
Elemental sulfur and thiosulfate disproportionation by 30. Rabus, R., T. Hansen, and F. Widdel. 2001. Dissimilatory
Desulfocapsa sulfoexigens sp. nov., a new anaerobic bac- sulfate- and sulfur-reducing prokaryotes. In M. Dworkin
terium isolated from marine surface sediment. Appl. . (ed.), The Prokaryotes: An Evolving Electronic Resource for
Environ. Microbiol. 64:119-125. the Microbiological Community, 3rd ed. Springereverlag,
13. Gibbons, R. J., and J. B. MacDonald. 1960. Hemin and New York, NY.
vitamin K compounds as required factors for the culti- 31. Rappe, M. S., S. A. Connon, K. L. Vergin, and S. J.
vation of certain strains of Bacteroides melanogenicus. Giovannoni. 2002. Cultivation of the ubiquitous marine
J. Bacteriol. 80:16+170. bacterioplankton clade. Nature 4 18:630-633.
14. Good, N. E., D. Winget, W. Winter, T. N. Connolly, 32. Reichenbach, H. 2002. The genus Helpetosiphon. In M.
S. Izawa, and R. M. M. Singh. 1966. Hydrogen ion buffers Dworkin (ed.), The Prokaryotes: An Evolving Electronic
for biological research. Biochemistry 5467477. Resource for the Microbiological Community, 3rd ed.
15. Gottschal, J. C., W. Harder, and R. A. Prins. 2002. Springer-Verlag, New York, NY.
Principles of enrichment, isolation, cultivation, and preser- 33. Reichenbach, H., and M. Dworkin. 2002. The
vation of bacteria. In M. Dworkin (ed.), The Prokayotes: Myxobacteria. In M. Dworkin (ed.), The Prokaryotes: An
An Evolving Electronic Resource for the Microbiological Evolving Electronic Resource for the Microbiological
Community, 3rd ed. Springer-Verlag, New York, NY. Community, 3rd ed. Springer-Verlag,New York, NY.
16. Hahn, M. W., H. Lunsdorf, Q. Wu, M. Schauer, M. G. 34. Sowers, K. R., and K. M. Noll. 1995. Techniques for
Hofle, J. Boenigk, and P. Stadler. 2003. Isolation of novel anaerobic growth, p. 15-48. In H. J. Schreier and K. R.
ultramicrobacteria classified as actinobacteria from five Sowers (ed.), Archaea: A Laboratory Manual, Methanogens.
freshwater habitats in Europe and Asia. Appl. Environ. Cold Spring Harbor Laboratory Press, Plainview, NY.
Microbiol. 69:1442-1451. 35. Straub, K. L., M. Hanzlik, and B. E. Buchholz-Cleven.
17. Hallberg, K. B., and D. B. Johnson. 2001. Biodiversity of 1998. The use of biologically produced ferrihydrite for the
acidophilic prokaryotes. Adw. Appl. Microbiol. 49:37-84. isolation of novel iron-reducing bacteria. Syst. Appl.
18. Hanson, R. S. 1998. Ecology of methylotrophic bacteria, Microbiol. 21:442-449.
p. 137-162. In R. S. Burlage, R. Atlas, D. Stahl, G. 36. Tumbula, D. L., T. L. Bowen, and W. B. Whitman.
Geesey, and G. Saylor (ed.), Techniques in Microbial 1995. Growth of methanogens on solidified medium, p. 49-
Ecology. Oxford Press, New York, NY. 56. In H. J, Schreier and K. R. Sowers (ed.), Archaea: A
19. Janssen, P. H., P. S. Yates, B. E. Grinton, P. M. Taylor, Laboratory Manual, Methanogens. Cold Spring Harbor
and M. Sait. 2002. Improved culturability of soil bacteria Laboratory Press, Plainview, NY.
214 GROWTH

37. Waterbury, J. B. 2002. The cyanobacteria-isolation, 39. Zengler, K., G. Toledo, M. Rappe, J. Elkins, E. J.
purification, and identification. In M. Dworkin (ed.), The Mathur, and M. Keller. 2002. Cultivating the uncultured.
Prokaqotes: An Evolving Electronic Resource for the Microbi- Proc. Natl. Acad. Sci. USA 99:15681-15686.
ological Community, 3rd ed. Springer-Verlag, New York, NY. 40. Zinder, S. H. 1998. Methanogens, p. 113-136. In R.S.
38. Wolin, E. A., M. J. Wolin, and R. S. Wolfe. 1963. Burlage, R. Atlas, D. Stahl, G. Geesey, and G. Saylor (ed.),
Formation of methane by bacterial extracts. j . Biol. Chem. Techniques in Microbial Ecology. Oxford Press, New York,
238:2882-2886. NY.
11
Enrichment and Isolation
ANDREAS TESKE. HERIBERT CYPIONKA. JOHN G . HOLT. AND NOEL R . KRIEG

11.1. BIOPHYSICAL ENRICHMENT .............................. 219


11.1.1. Low-Temperature Incubation ............................ 219
11.1.1.1. Psychrophiles ............................... 219
. .........................
2. Listeria monocytogenes 219
. 3. Marine Luminescent Bacteria.................... 219
11.1.2. High-Temperature Incubation ........................... 219
11.1.2.1. Thermophiles in Milk .......................... 219
. 2. Thermus Species ............................. 219
. 3. Themplusma Species ......................... 220
11.1.3. High-Temperature Treatment ........................... 220
11.1.3.1. Thermoduric Organisms in Milk .................. 220
. ........................
2. Mesophilic Sporeformers 220
. 3. Thermophilic Sporeformers...................... 220
11.1.4. Drying ........................................... 220
.5. Motility ........................................... 220
11.151. Swarming Motility of Clostridium tetani ............ 220
. 2. Swimming Motility of Treponemes ................ 221
. 3. Swimming Motility of Spirillum volutans............ 221
. 4. Gliding Motility of Cytophagas. Flexibacters. and
Myxobacters ................................ 221
. ............... 221
5. Gliding Motility of Beggiutoa Species
11.1.6. Filterability ........................................ 222
11.1.6.1. Spirochetes ................................. 222
. ....................... 222
2. Thin. Flagellated Bacteria
11.1.7. Visible Illumination for Phototrophs ...................... 223
11.1.7.1. Unicellular Cyanobacteria ...................... 223
. 2. Purple Nonsulfur Bacteria...................... 223
. 3. Green Sulfur Bacteria......................... 223
11.1.8. Visible Illumination To Enhance Pigment Production .......... 224
.9. Sonic Oscillation Selection of Sporocytophuga myxococcoides..... 224
.l0. Use of Antiserum Agglutination or Magnetic Beads Coated
with Antibodies ..................................... 224
.
.11 Cell Density ....................................... 224
11.1.11.1. Flotation on Water for Lumpropedia hyalinu ........ 224
. 2. Swirling for Achromatium oxaliferum............. 224
. ................. 224
3. Density Gradient Centrifugation
. 4. Sampling the Neuston Layer for Nevskia ramosa ..... 225
11.1.12. Radiation Resistance ................................. 225
11.1.12.1. UV Irradiation for Cyanobacteria ................225
.
.2 X-Ray and Gamma-Radiation Resistance for
................................. 225
Deinococci
11.1.13. Magnetic Field for Magnetotactic Bacteria .................. 225
............................ 225
.14. Isoelectric Focusing of Cells
11.2. BIOCHEMICAL ENRICHMENT .............................. 226
11.2.1. Alkali Treatment for Mycobacteria ....................... 226
.2. Incubation at Alkaline pH for Vibrios and Sporosarcina ureae .... 226
.3. Acid Treatment for Legionella Species ..................... 227
.4. Incubation at Low pH ................................ 227
11.2.4.1. Lactobacilli................................. 227
. 2. Thiobucillus thwoxidans........................ 227
. ............................. 227
3. Frateuria Species
11.2.5. Inhibition by Toxic Metals .............................. 227
11.2.5.1. Tellurite Inhibition for Corynebacteria and Certain
.................................
Streptococci 227
. .. 227
2. Thallium Inhibition for Mycoplasmas and Enterococci
. ................ 227
3. Selenite Inhibition for Salmonellae

215
216 m GROWTH

11.2.6. Phenylethanol Inhibition for Gram-Positive Cocci............ 227


.7. Dye Inhibition for Gram-Negative Bacteria. Mycobacteria. and
Arthrobacters ...................................... 228
.8. Salt Inhibition ...................................... 228
11.2.8.1. Staphylococci ................................ 228
.2. Halobacteria ................................ 228
.3. Halomonas Species ............................ 228
11.2.9. Bile Inhibition for Enteric Bacteria ....................... 228
.10. Antibiotic Inhibition ................................. 228
.11. Dilute Media ....................................... 229
11.2.11.1. Caulobacters ............................... 229
. ............................. 229
2. Aquatic Spirilla
. .......................... 230
3. Sphaerotilus Species
.
4. Marine Oligotrophic Bacteria Growing on Natural
Seawater Medium ............................ 230
11.2.12. Special Substrates ................................... 230
11.2.12.1. Tryptophan for Pseudomonads .................. 230
.
2. Nz for &ospirillum and kotobacter Species ........ 231
.
3. Hz for Aquaspirillum autotrophicum ............. 231
..
4. Methanol for Hyphomicrobia ................... 231
5. Cellulose for Cellulolytic Cytophagas ............. 231
.
6. Agar for Agarolytic Cytophagas ................. 231
.
7. Lactate for Propionibacteria .................... 231
....... 232
.
8. Ammonium or Nitrite for Nitrifying Bacteria
.
9. Nitrate plus Organic Acids for Pseudomonads ...... 232
.10. Sulfate plus Organic Acids for Sulfate Reducers ..... 232
.11. Elemental Sulfur for Desulfiromonas acetoxidam .... 232
.12. Thiosulfate. Sulfite. or Elemental Sulfur for Bacteria
that Carry Out Disproportionation of Intermediate
Sulfur Compounds .......................... 233
.13. Fe and Mn Oxides for Iron- and Manganese.Reducing
Bacteria.................................. 233
.l4. Chlorinated Compounds for Dehalogenating
Bacteria.................................. 233
.................. 234
.15. Toluene for Toluene Oxidizers
.16. Bacterial Cells as Substrate for Myxococci ......... 234
.17. Unusual Carbon Sources for Nitrogen Fixers ....... 234
11.2.13. Chemotactic Attraction ............................... 234
.14. Gradient Culture .................................... 235
11.2.14.1. Gradient Culture for Neutrophilic Fe-Oxidizing
Bacteria.................................. 235
. ..... 236
2. Gradient Culture for Sulfide-OxidizingBacteria
11.2.15. Cyclic AMP and N-Acyl Homoserine Lactones as Growth
Inducers .......................................... 236
11.3. BIOLOGICAL ENRICHMENT ............................... 237
11.3.1. Bacterial Parasitism by Bdellovibrios ...................... 237
.2. Plant Symbiosis by Rhizobia ............................ 237
.3. Animal Parasitism ................................... 237
.4. Plant Parasitism..................................... 237
. .............................
5. Baiting for Actinoplanetes 237
.6. Uncultured Symbionts and Parasites...................... 238
11.4. ISOLATION ............................................. 238
11.4.1. Spatially Streaking or Spreading on Solid Medium ............ 238
...
11.4.1.1. Isolation of Microcolony-Forming Bacteria on Filters 238
11.4.2. Serially Diluting in Solidified Medium..................... 241
. .............
3. Serially Diluting to Extinction in Liquid Medium 241
11.4.3.1. MPN Dilutions .............................. 241
. ..........................
2. Extinction Culturing 242
.3. High-Throughput Culturing..................... 242
11.4.4. Washing Filamentous Bacteria ........................... 243
. .................................
.5 Isolating Single Cells 244
11.5. rRNA-BASED METHODS IN CONJUNCTION WITH
PHENOTYPIC CHARACTERIZATION ........................ 246
11.5.1. rRNA Sequencing as a Guide for Enrichment and Isolation ...... 246
.2. Uses of Whole-Cell FISH .............................. 247
.3. Molecular Monitoring of the Path from Environmental Samples
to Enrichments and Pure Cultures ........................ 248
1 1 . Enrichment and Isolation 217

11.6. MEDIA AND REAGENTS .................................. 249


11.6.1. Agrobacterium Biovar 1 Isolation Medium .................. 249
. 2. Agrobacterium Biovar 2 Isolation Medium .................. 249
. ................. 250
3. Alkaline Peptone Water for Vibrio cholerae
..4. Aqumpirillum gracile Isolation Medium.................... 250
............................ 250
5. Arthrobacter Selective Agar
.6. ASN-I11 Medium .................................... 250
.7. Azotobacter Medium .................................. 250
. . . 250
8. Basal Enrichment Medium for Members of the Rhodospirillaceae
.9. Basal Inorganic Media A and B.......................... 250
................... 251
.l 0. Basal Salts Medium for Themus Species
........................ 251
.11. BCYE Agar for Legionella Species
.12. BG-11 Medium ..................................... 251
.................................... 251
.l 3. Bile Esculin Agar
.14. BMS Agar (Potato Agar) .............................. 252
.l5. Bordet-Gengou Agar. Modified.......................... 252
................................. 252
.16. Brilliant Green Agar
....................... 252
.l 7. Brilliant Green Lactose Bile Broth
.18. Brucella Agar...................................... 252
.19. CAS Medium ...................................... 252
..................................
.2 0. Casein-Starch Agar 252
.2 1. CHSS Medium for Spirillum volutans..................... 252
........................... 253
.2 2. Cystine Tellurite Blood Agar
.2 3. Dehalococcoides ethenogenes Medium ................... 253
.2 4.EAgar ........................................... 253
.2 5.EBroth ........................................... 254
.2 6. Frateuria Enrichment Medium.......................... 254
.............................. 254
.2 7. Furazolidone (FTO) Agar
.2 8. Giesberger Base Medium.............................. 254
.2 9. Glucose-Tryptone Agar............................... 254
.30. GYC Agar ......................................... 254
.3 1. GYPT Medium ..................................... 254
.32. Halophile Medium................................... 254
.33. Hyphomicrobium Medium .............................. 254
.34. Iverson Medium..................................... 255
.35. Iron- and Manganese-Reducer Medium .................... 255
.36. Lactate Medium..................................... 256
.37. Lowenstein-Jensen Medium............................ 256
.38. MacConkey Agar .................................... 256
...................................
.3 9. Mannitol Salt Agar 256
.4 0. Marine Basal J3 Medium for Lithoautotrophic Marine
.......................................
Beggiatoa spp 256
.................. 257
.4 1. Marine Medium for Luminescent Bacteria
.
.42 Mineral Agar ....................................... 257
................................. 257
.43. Mitis-Salivarius Agar
.4 4. MN Medium ....................................... 257
.4 5. Modified Campy BAP ................................ 257
.4 6. Modified Rogosa Medium.............................. 257
......................... 258
.4 7. Modified Wolfe Mineral Medium
.48.MPSSAgar ........................................ 258
.
.49 N-Acetyl-L-Cysteine-SodiumHydroxide Reagent ............. 258
.5 0. Nevskia Medium.................................... 258
. .
5 1 Nfb Medium ....................................... 258
................................. 258
.5 2. Nitrification Medium
........................ 258
.5 3. Pfennig and Biebl Sulfur Medium
.54. Phenylethyl Alcohol Agar............................. 259
.55. Pringsheim Soil Medium............................... 259
.5 6. Pseudomonas Isolation Medium......................... 259
.
5 7 PTN Medium for Listeria Species ........................ 259
.58. RGCA-SC Medium .................................. 259
........................... 259
.5 9. Salmonella-Shigella (SS) Agar
..................................... 260
.60. Selenite F Broth
.............................. 260
.6 1. Serpens Isolation Medium
.62. Soybean-Casein Digest Broth........................... 260
.63. Sphaerotilus Medium................................. 260
.64. Sporocytophaga Medium ............................... 260
.65. ST6CX Medium for Cellulolytic Cytophagas ................ 260
218 GROWTH

.66. Standard Methods Agar ............................... 260


.67. Succinate-Salts Medium ............................... 260
.68. TCBS Agar ........................................ 260
.69. TGYM Medium ..................................... 261
.70. Thallous Acetate Agar ................................ 261
.7 1. Thayer-Martin Agar .................................. 26 1
.72. Thermoplcrrma Isolation Medium ........................ 261
.73. Thiobacillus thiooxidam Medium ........................ 261
.74. T N Medium for Listeria Species ......................... 262
.75. Tryptic Soy-Yeast Extract Agar with Urea .................. 262
.76. Tryptophan Medium .................................. 262
.77. Veldkamp Medium ................................... 262
.78. Violet Red Bile Agar ................................. 262
.79. Widdel and Pfennig Medium ............................ 262
.80.Wiley and Stokes Alkaline Medium ....................... 263
3 1 . Yeast Extract Mannitol Agar ............................ 263
...............................
3 2 . Yeast Extract Milk Agar 263
.83. YP Medium ........................................ 263
11.7. REFERENCES ............................................ 263
11.7.1. General References .................................. 263
.2. Specific References .................................. 263

Since the last edition of this book, the development of mo- Enrichments make it possible to assess the differential ef-
lecular biological approaches has revolutionized microbiol- fects of environmental factors imposed on mixed microbial
ogy. The possibility to analyze microbial communities with- populations and permit the selection of organisms capable
out the necessity of cultivating their members has of attacking or degrading particular substrates or of thriving
demonstrated an overwhelming microbial diversity. In under unusual conditions.
1987, 12 divisions were known in the bacterial domain. Successful isolation of a given organism into pure cul-
Within 10 years this has increased to 36, and more are com- ture requires a sufficiently high proportion of that organ-
ing (88). While about 6,000 species are described today, es- ism in the mixed population. Isolation is easiest when the
timates based on reassociation kinetics of DNA isolated organism is the numerically dominant member of the pop-
from natural environments come to 13,000 species already ulation. Enrichment methods are designed to increase the
in 30 g of forest soil (197) or even 1 billion prokaryotic relative numbers of a particular organism by favoring its
species worldwide based on a DNA-DNA homology of 70% growth, its survival, or its spatial separation from other
within a species (209). At the same time it became increas- members of the population. Biophysical enrichments
ingly clear that our success in cultivation of microbes from make use of such conditions as growth temperature, heat
natural environments is minute. Usually less than 1% of the treatment, sonic oscillation, or UV irradiation to kill or
prokaryotes present in a natural environment grow on lab- inhibit the rest of the population. Such methods may also
oratory media (13). Therefore, many people came to the take advantage of some physical property of the desired
conclusion that microbial ecology should abandon cultiva- organism, such as its size, motility, surface charge, or the
tion attempts and rely on molecular biology only. Most of presence of certain antigens, which can allow the organ-
the microbes in natural environments were regarded as un- ism to be preferentially separated from the rest of the com-
culturable. However, the huge numbers of prokaryotes in munity. Biochemical enrichments employ toxic agents to
natural environments and their diversity are a fact. Mother kill or inhibit the rest of the population without affecting
Nature has cultivated them where they are. Obviously, they the desired organism; alternatively, they may provide nu-
are not unculturable per se. It is only our techniques that are trient sources that can be used preferentially by a particu-
not efficient. With improved cultivation techniques and lar component of the mixed population. Biological en-
more sensitive analysis of cultures the success can be in- richments may make use of specific hosts for selective
creased, although cultivation of 100% of a natural commu- growth of a particular organism, or they may take advan-
nity is not feasible. In the meantime it has turned out that tage of some pathogenic property, such as invasiveness,
cultures of bacteria are still required to understand their which the rest of the population does not possess. In many
properties and interactions. And just the molecular tech- enrichment procedures several physical, chemical, and/or
niques in themselves turned out to be very helpful for culti- biological methods may be used in combination to achieve
vation experiments. They can help to study the composi- a maximum effect.
tion of enrichment cultures in order to track a member of Most enrichments are carried out in closed systems,
interest. In the future, studies on mixed cultures will be pos- such as batch cultures in a flask or tube, where the con-
sible, where some members can be analyzed without sepa- centrations of nutrients and metabolic products in the cul-
rating them from their natural-environment mates. A new ture vessel continually change during bacterial growth.
section (11.5) focuses on the use of enrichment and culti- Open systems have also been used for enrichment. For ex-
vation procedures with molecular methods, such as whole- ample, the use of a chemostat (chapters 12.2.5 and 13.2)
cell fluorescent in situ hybridization (FISH) or denaturing enables one to provide a constant environment for culti-
gradient gel electrophoresis (DGGE), to monitor the vation of bacterial cells by continuously supplying a
progress of an enrichment, to evaluate the presence of con- growth-limiting nutrient and continually removing meta-
taminants, and to identify new isolates. bolic products. Alteration of the dilution rate controls the
1 1 . Enrichment and Isolation 219

concentration of the growth-limiting nutrient, which dif- 1 1 .I .I .2. Listeria monocytogenes


ferentially affects the growth rate of the various organisms It may be difficult to isolate Listeria monocytogenes from ma-
in mixed culture, making it possible for one or another terial heavily contaminated with other bacteria (e.g., feces,
member of the mixed population to become predominant; silage, sewage, and clinical specimens from the cervix,
for an example, see section 11.2.11.2. The use of gradients vagina, meconium, nasopharynx, or tissues). Such materials
(11.2.14) is another approach that allows the organisms to may be enriched by the following procedure (174). Add a 1-
find optimum conditions instead of forcing them into a to 2-g sample of the suspected material to each of two flasks
fixed situation. containing 100 ml of sterile enrichment broth. Examples of
Bacteria are usually isolated from enrichment cultures by suitable enrichment broths are TN medium (section
spatially separating the organisms in or on a solid medium 11.6.74) and PTN medium (section 11.6.57). Incubate one
and subsequently allowing them to grow into colonies. For flask at 37C and the other at 4C. Plate 0.1-ml samples
organisms that cannot grow on solid media, dilution to ex- from the flask incubated at 37C onto Tryptose agar (Difco)
tinction by allowing separation of cells into individual tubes daily for 7 days. Incubate the plates at 37C for 24 h, and
of a liquid medium can be used. Because such ordinary iso- examine the plates under oblique lighting for the presence
lation methods do not absolutely ensure purity, more diffi- of distinctive blue-green colonies. Plate 0.1-ml samples
cult methods may be needed whereby an individual bacte- from the flask incubated at 4C onto Tryptose agar at 7-day
rial cell from a mixed population is spatially isolated under intervals for a period of up to 2 months. Incubate and ex-
a microscope before being cultured into a clone. amine the plates as described for the broth held at 37C.
This chapter is designed to demonstrate the multiplicity Although the incubation at 37C may give results more
and in many instances the considerable ingenuity of en- quickly, the cold enrichment usually gives a higher propor-
richment and isolation methods for bacteria by presenting tion of positive cultures. The reason why cold incubation
specific, selected examples. The wide-ranging examples for enriches for L. monocytogenes has been variously attributed
enrichment and isolation methods in this chapter in the to an ability to survive longer than many other bacteria at
previous edition were retained and expanded by new ap- 4C (215), an ability to multiply at 4C (179), and release
proaches, such as gradient culture, new electron acceptors of the organisms from an intracellular location (59).
(metal oxides and humics, halogenated organics, and dis-
proportionating sulfur species), oligotrophic cultivation in 11 .I .1.3. Marine Luminescent Bacteria
seawater, and high-throughput cultivation. Luminescent bacteria can be isolated from fresh herring
Although many different methods are described, this (39). Although the isolates are psychrophilic, pure cultures
chapter is not intended to be a comprehensive compendium can be obtained within a 2-week course. Place a fresh her-
for all bacterial taxa. The general references (1-1 1) given at ring in a bowl, covered with salt water (3%)by half. Put the
the end of this chapter and those given for enrichment and bowl into a refrigerator at 8 to 10C for 2 days (not longer,
isolation of mutant cells in chapter 28 should be consulted not warmer, otherwise a bad smell will develop!). Light-
for additional methods and organisms. emitting pinpoints can be detected, often on the sideline
Following the procedures described here will usually lead organ. The room has to be perfectly darkened, and the eye
to the decribed microorganisms. One should, however, keep has to adapt to the darkness for at least 5 min. To isolate
in mind that we-in contrast to Mother Nature-so far pure cultures, light-emitting pinpoints are transferred to
have cultivated only a small percentage of the prokaryotes agar medium (section 11.6.41) by means of sterile tooth-
on earth. Any variation of an enrichment procedure might picks. A freshly grown plate can be bright enough for read-
give rise to a different outcome. Thermodynamics can give ing a watch nearby.
a guideline for the detection of new organisms. In principle,
any situation with free energy could provide the energetical 11.1.2. High-Temperature Incubation
basis for microbial growth. In many cases, like dispropor-
tionation of elemental sulfur, the concentrations of sub- High-temperature incubation favors the selection of ther-
strates and products have to be controlled by further reac- mophilic bacteria because the growth of other organisms is
tions in order to allow growth. Whenever an exergonic inhibited at high temperature. The definition of ther-
reaction is possible, there is a good chance to find a microbe mophilic varies depending on the particular organisms
that can use it for growth. being studied. The term usually refers to organisms that are
unable to grow at temperatures below ca. 45 to 50C; how-
ever, it has also been applied to other organisms, for exam-
11.l.BIOPHYSICAL ENRICHMENT ple, Campylobacter species (e.g., Campylobacter jejuni, C.
coli, and C. lari) that can be cultured at 42C but not 25C.
11.I . I . low-Temperature Incubation (139) Another term, extreme thermophiles, is usually applied to
Low temperature retards the growth of many mesophilic organisms that can grow at temperatures above 70C.
bacteria but not psychrophilic and psychrotrophic bacteria.
Therefore, incubation of enrichment cultures at 0 to 5C 11 .I .2.1. Thermophiles in Milk
can favor the growth of the latter organisms. For milk thermophiles, plate serial dilutions of milk in stan-
dard methods agar (section 11.6.66) and incubate the cul-
11.1.1 . I . Psychrophiles tures at 55C (14).
Inoue (92) isolated psychrophilic bacteria from Antarctic
soil by spreading dilutions of the soil over plates of a glu- 11 .I .2.2. Thermus Species
cose-yeast extract-peptone agar and incubating them for 14 Themus quaticus strains have an optimum growth tem-
to 24 days, with the temperature maintained throughout at perature of 70C, a maximum of 79C, and a minimum of
below 5C. In this manner nine strains of obligately psy- 40C. T. ruber strains have an optimum temperature of
chrophilic bacteria with maximum growth temperatures of 60C, a maximum of 70C, and a minimum of 37C (214).
ca. 20C were obtained (93). For enrichment (33,214), inoculate 0.5- to 1.0-ml samples
220 GROWTH

of microbial mats, water from hot springs, thermally pol- Closcridium species, prereduced starch broth (PY broth [see
luted water, or hot tap water into 10-ml portions of +
chapter 15.4.311 1.0% soluble starch) is often satisfactory.
Thermus medium (basal salts medium [section 11.6.101 plus If small numbers of spores are present in the samples, the
0.1% pancreatic digest of casein and 0.14% yeast extract). proportion may be increased by adding the sample to a suit-
Incubate in covered water bath shakers for 1 to 3 days at 70 able sporulation medium and incubating the culture for var-
to 75C for T. aquaticus or 55 to 65C for T. ruber. For iso- ious periods (2 to 21 days) prior to the heat treatment. For
lation, streak turbid cultures onto the Thermus medium so- most Bacillus species, a suitable sporulation medium is nu-
lidified with 2 to 3% agar. Wrap the petri dishes in house- trient broth (nutrient agar [see chapter 15.4.281 without the
hold plastic wrap to avoid drying out during the incubation agar component) plus 0.5% yeast extract, 7 X lop4 M
period (32), and incubate them aerobically at the appro- CaC12, 1 X M MgC12, and 5 X lo- M MnC12. Also
priate optimum temperature. Colonies of T. aquaticus are see chapter 4.4.9. For most Clostridium species, no one
yellow to pale or colorless. Colonies of T. ruber are red medium is optimum for production of spores, but chopped-
(214). meat medium (see chapter 15.4.12) with and without glu-
cose often supports sporulation (86).
11.I .2.3. Thermoplasma Species The heat treatment employed may have to be modified
Thennoplasma species are best enriched and isolated from for some types of sporeformers, since the endospores of some
coal refuse piles (28, 58). Filter a liquid sample through a bacterial strains are not as heat resistant as others. Try a
membrane filter with a pore size of 0.45 p m and then lower temperature (e.g., 75 or 70C) if satisfactory results
through a second filter with a pore size of 0.22 pm. Incubate are not obtained at the usual temperature of 80C.
aerobically in Thermoplasma isolation medium (section
11.6.72) at 55C for 4 to 6 weeks or until turbidity devel- 11 .I .3.3. Thermophilic Sporeformers (14)
ops. The organism can be isolated either by performing Thermophilic sporeformers may be present, for example, in
dilution to extinction in liquid medium (section 11.4.3) or various sweetening agents used in ice cream. Initially pre-
by obtaining colonies on medium solidified with 10% pare a 20% (wt/vol or vol/vol) solution of the sweetening
(wtlvol) hydrolyzed starch of the type used for gel elec- agent (beet or cane sugar, lactose, cerelose, invert syrup,
trophoresis (175). Adjust the pH to ca. 3 immediately be- corn syrup, maple syrup, liquid sugar, or honey), and heat to
fore pouring the plates. Incubate the plates within a sealed 100C for 5 min to kill nonsporeformers. Dilute with sterile
chamber in a humid atmosphere consisting of ca. 60% air water to give a final concentration of 13.3%.
and 40% C02 (vol/vol). Thennoplasma colonies appear For thermophilic, aerobic flat-sour organisms, plate 2.ml
within 7 days (or more), are small and colorless to brown- portions of the heated, diluted sugar solution in glucose-
ish, and have a characteristic fried-egg appearance when tryptone agar (section 11.6.29). Incubate in a humid cham-
viewed under a dissecting microscope (175). ber at 55C for 48 h. Characteristic surface colonies are
round, are 2 to 5 mm in diameter, have a typical opaque
11.I .3. High-TemperatureTreatment central spot, and are surrounded by a yellow halo in the
Treatment of a sample at an appropriate high temperature medium. Subsurface colonies are compact and may be pin-
may select heat-resistant bacteria from a mixture of micro- point in size; these should be subcultured and streaked onto
organisms. The heat treatment may be relatively mild (as in glucose-tryptone agar.
pasteurization), which even certain vegetative cells can sur- For thermophilic, anaerobic hydrogen sulfide producers,
vive, or much more severe, which only sporeformers can add samples of the heated, diluted sugar solution to deep
survive. tubes of sulfite agar (1% tryptone or Trypticase, 0.1%
sodium sulfite, and 2% agar). The tubes of medium should
11 .I .3.1. Thermoduric Organisms in Milk (14) be melted and cooled to 55C prior to inoculation. Allow
Pasteurize a sample of raw milk by heating it to 62.8C and the tube contents to solidify,and incubate at 55C for 72 h.
holding it at this temperature for 30 min. Cool, add dilu- Sulfide producers will form blackened spherical areas in the
tions to standard methods agar (section 11.6.66), and incu- medium.
bate the plates at 32C. For instance, Microbacterium
lacticum can be isolated with the same heat treatment and 11.I .4. Drying
subsequent plating on yeast extract milk agar (section A number of bacteria that normally reside in the soil and
11.6.82) with incubation at 32C for 7 days. produce spores, cysts, or other dormant forms are resistant
to dehydration. A soil sample is air dried to kill many of the
11 .I .3.2. Mesophilic Sporeformers vegetative cells and is then plated onto a suitable agar
Heat water samples or soil suspensions at 80C for 10 min, medium. This method is useful for members of the genera
and then streak onto plating media. isolation media for Bacillus, Azotobacter, and Arthrobacter (48).
Bacillus species differ according to the special nutritional
needs of the species (e.g., use of uric acid as a substrate for 11.1.5. Motility
B. fastidiosus), and the reader is referred to references 48 and Motility has been used as the basis for several ingenious
212 for details. For Clostridium species, streak a roll tube of methods for enrichment and isolation. The principle is
prereduced chopped-meat agar (see chapter 15.4.12) or that bacteria that swim in liquid media or swarm or glide
streak a plate of freshly prepared agar medium and incubate across solid media may be able to outdistance other micro-
it in an anaerobe jar. organisms.
Another approach is to add some of the sample in which
spores are suspected to a suitable growth medium, heat at 11 .I .5.1. Swarming Motility of Clostridium tetani
80C for 10 min, and then incubate the broth for about 1 (181)
day to permit growth to occur before streaking onto solid Inoculate the specimen (soil, animal feces, or clinical mate-
media. For Bacillus species, nutrient broth containing 0.5% rial) onto a small area of a freshly prepared plate of blood
yeast extract is usually a suitable growth medium; for agar (see chapter 15.4.9). Incubate the plate at 37C in an
11. Enrichment and Isolation 221

anaerobic jar for 1 day, and examine the agar surface care- in the enrichment culture and thus can reach the distal end
fully for evidence of swarming (a thin layer of growth that of the capillary first. As soon as some spirilla reach the dis-
has spread outward from the inoculated area). It may be tal end, break the capillary behind them, expel the spirilla
helpful to scrape the surface of the medium with a needle to into a tube of semisolid CHSS medium (section 11.6.21),
verify the occurrence of swarming. Suspend some growth and incubate at 30C. Confirm the purity of the cultures by
from the edge of the swarming area in broth, and streak phase-contrast microscopy.
onto solid media containing 5% agar to obtain isolated
colonies. 1 1 . I .5.4. Gliding Motility of Cytophagas,
Flexibacters, and Myxobacters
11.1.5.2. Swimming Motility of Treponemes Spread dilutions of the specimen (soil, water, or animal
(80, 83, 167-1 69) dung which has been in contact with the soil) onto agar
The following method can be used for enrichment of cul- media that have a low nutrient concentration (e.g., 0.1%
tivable Treponema species, many of which occur as part of tryptone or Tiypticase, or l/lO-strength nutrient agar).
the normal flora of the oral cavity and intestinal tract of hu- Another procedure is to smear terrestrial plant leaf material,
mans and animals. Cut a well into the center of a thick algal fronds, or marine plants on nutrient-poor media.
layer of suitable agar medium (section 11.6.58) contained Cytophagas and flexibacters are able to migrate on the sur-
in either petri dishes or beakers. The well should be at least face of solid media; their colonies can be recognized as thin,
7 mm deep and 2 to 10 mm in diameter and should not be often nearly translucent colonies with fingerlike projec-
cut to the bottom of the dish. Inoculate the well with the tions, which develop far beyond the streak of deposition
specimen (sample from the oral cavity, intestinal contents, line. Subculture from these colonies. The incorporation of
or feces). Inoculate large wells (diameter, 10 mm) with up penicillin G (15 U/ml) and chloramphenicol (5 pg/ml)
to 0.2 ml of sample; inoculate small wells (diameter, 2 mm) into the agar media often helps to suppress the growth of
by stabbing ca. 2 mm obliquely into one side of the well. Do other bacteria (206). Many of the fruiting myxobacters are
not allow any of the sample to be deposited on the surface bacteriolytic and can be enriched on bacterium-rich sub-
of the agar. Immediately place the plate in an anaerobic jar, strates to the point at which they produce characteristic
and incubate at 37C for 4 to 7 days. In contrast to most fruiting bodies, which can then be transferred to agar
other bacteria, treponemes can migrate through agar media. media. Place natural materials such as dung from herbivo-
Look for treponemal growth occurring as a haze in the rous animals, decaying plant material, or bark onto mois-
medium at some distance from the well. Subculture a sam- tened paper contained in petri dishes or household plastic
ple from the outermost portions of this hazy region into a crisper boxes, and incubate for up to 3 weeks.
suitable prereduced semisolid medium (e.g., broth contain- Myxococci can be enriched on sterilized, urine-free
ing 0.15% agar). Since more than one kind of treponeme rabbit dung pellets (preferably from rabbits fed on non.
may be present, streak the subculture onto a roll tube of pre- antibiotic-containing feeds) placed on moistened soil in a
reduced medium to obtain isolated colonies. These appear petri dish (135). It is best to moisten the substrate with a so-
as hazy, whitish, dense areas in the medium. lution of cycloheximide (30 to 50 mg per liter of distilled
A similar approach has been successfully used for the iso- water) to inhibit fungi. Keep the preparations moist while
lation of anaerobic marine spirochetes (43). incubating. Check periodically for the presence of charac-
teristic fruiting bodies with the aid of a dissecting micro-
11.I .5.3. Swimming Motility of Spirillurn volutans scope. Transfer material from the fruiting body to an agar
(91, 118, 166) medium by using a sterile fine metal or glass needle (135).
Obtain mixed cultures of the giant microaerophilic bac- As with other gliding bacteria, pure cultures can be ob-
terium Spirillum volutans by preparing a hay infusion with tained by repeatedly removing cells from the leading edge of
stagnant pond water. After a surface scum develops, exam- the colony and inoculating them onto the center of a fresh
ine samples taken just beneath the scum. Look for very large plate of agar medium. Bacteriolytic species can be inocu-
spirilla (1.4 to 1.7 pm in diameter and up to 60 pm long) lated onto an agar medium containing a suspension of heat-
with bipolar flagellar fascicles that are clearly visible by killed bacteria such as Escherichia coli.
dark-field microscopy. Enrich the culture by inoculating
some of the hay infusion into Pringsheim soil medium (sec- 1 1 . I .5.5. Gliding Motility of Beggiatoa Species
tion 11.6.55) and incubating at room temperature. Even (45, 66, 147, 186)
with this enrichment, S. volutans will be vastly outnum- Beggiatoa species form colorless filamentous chains of cells,
bered by other bacteria. However, it can be isolated by using which exhibit gliding motility on surfaces. The organisms
a capillary tube (166), as follows. Soften the center of a occur in aerobic freshwater or marine environments rich in
short section of a sterile, cottoneplugged piece of 5 m m - HZS; the cells can oxidize the sulfide to elemental sulfur,
diameter glass tubing in a flame. Pinch the tubing with which is deposited intracellularly.
square-ended forceps until it is almost closed. Reheat the For enrichment, first prepare extracted hay by the fol-
flattened portion, and draw it out rapidly to form a long lowing procedure. Cut dried hay into small pieces, and ex-
capillary tube 15 to 30 cm long and 0.1 to 0.3 mm wide, tract by boiling the hay in a large volume of water. Change
oval in cross section. Seal the ends of the capillary in a the water three times during the extraction. The final wash
flame. Break the capillary near one end with sterile forceps, should have an amber color. Drain the hay, and place it on
and draw up 10 to 20 cm of sterile Pringsheim soil medium trays at 37C to dry. The enrichment medium consists of
(supernatant). Then, dip the capillary into the enrichment 0.8% (wtlvol) of the dried, extracted hay in tap water, dis-
culture, and draw up another 2 to 4 cm, making sure that n o tributed in 70-ml volumes into 125-ml cotton-stoppered
air space occurs between the sterile medium and the cul- Erlenmeyer flasks. Sterilize by autoclaving. Inoculate the
ture. Seal the tip of the capillary, leaving a small air space. flasks with 5-ml portions of mud containing decaying plant
Mount the capillary on the stage of a XlOO microscope. S. materials, as from small ponds, lakes, or streams. Incubate
volutans is often able to swim faster than the other bacteria the flasks at ca. 25C for 10 days.
222 GROWTH

Effective enrichments are indicated by a strong odor of small, motile and, preferably, flexible, it may be able to mi-
H2S and the development of a white film on the surface of grate through the filter pores into the agar below, where it
the medium and on the submerged upper walls of the flasks. can be isolated after growth by removing the filter and
Examine the film for the characteristic filaments of subculturing from the agar. This method is particularly
Beggiatoa organisms. To free the filaments of contaminants, useful for organisms that grow slowly and are easily over-
remove a tuft of filaments with a loop needle or micro- grown by contaminants during isolation attempts. The
forceps and place it into a vial of sterile basal salts solution principle is exemplified by enrichment procedures for
(section 11.6.10) plus 5 mM neutralized sodium sulfide. spirochetes such as cultivable, anaerobic Treponema spp.
Tease the tuft apart, swirl, and transfer to another wash and facultatively anaerobic Spirochaeta spp. and for thin,
bath. Repeat this process five times. Place the washed fila- flagellated organisms such as Aqwlspirillurri gracile and Serpens
ments on the dried surface of a 1.6% agar plate for about 1 flexibilis.
min to absorb excess fluid, and then place them on the sur-
face of plates containing a medium consisting of 0.1% yeast 11.1.6.1. Spirochetes
extract, 0.1% sodium acetate, and 2 mM sodium sulfide. The following method can be used for enrichment of cul-
View the plates after 24 h with a dissecting microscope, and tivable Treponem species from the oral cavity and intes-
use a fine sterile needle to transfer to a fresh plate those fil- tinal tract of humans and animals (86, 180). Prepare a petri
aments growing from the mass that are free of contami- dish of a suitable agar medium such as RGCA-SC medium
nants. The filaments that grow away from the central in- (section 11.6.58). Place a membrane filter (pore size, 0.15
oculum are cut out on agar blocks and are used as inoculum pm) on the surface of the agar. Place an O-ring (25 to 30
for new agar plates. These procedures have led to the isola- mm in diameter) that has been lightly coated with vacuum
tion of heterotrophic freshwater Begk.xtoa strains (162, grease on top of the filter. Place several drops of the diluted
187). specimen (sample from the oral cavity, intestinal contents,
Marine Beggiatoa strains require different enrichment or feces) onto the center of the O-ring. Incubate the petri
and isolation procedures (147). Plates for the isolation of dish in an anaerobic jar at 37C for 1 to 2 weeks.
marine Beggiatoa strains use a modified J3 basal medium, Treponemes are small enough to migrate through the filter
based on aged seawater (section 11.6.40). J3 basal medium pores and penetrate the underlying agar, where they grow as
is amended to produce an isolation medium (J-TS) by a haze in the medium. Remove the O-ring and membrane
adding the following sterile stocks, with final concentra- filter from the agar surface, and remove a plug of agar from
tions in parentheses: (i) 7.5 ml of 200 mM Na2S203 the hazy region with a Pasteur pipette. Examine the plug by
(2 mM); (ii) 3.75 ml of freshly neutralized 200 mM Na2S dark-field microscopy for treponemes and contaminants.
(1 mM); this is autoclaved as a basic solution, which is quite Subculture and purify as described in section 11.1.5.2. The
stable against auto-oxidation, and then neutralized with an inclusion of polymyxin B (800 U/ml) and nalidixic acid
equimolar quantity of sterile HC1 just prior to use; (iii) 15 (800 U/ml) in the agar medium often helps to suppress the
ml of 1 M NaHC03 (20 mM); to make this stock, autoclave growth of contaminants.
8.4 g of NaHC03 (dry) and add 100 ml of sterile water For enrichment of facultatively anaerobic Spirochaeta
when cool. strains (43), place a membrane filter with a pore size of 0.3
Immediately after solidification, agar plates are incu- or 0.45 p m on the surface of a suitable agar medium, such
bated in a bell jar for 24 h or more under anoxic conditions as GYPT medium (section 11.6.31). Place a drop of pond
(99.5% N2, 0.5% COz), with desiccant present to absorb water or water-mud slurry on the disc near the center.
water evaporating from the surface of the medium. The Incubate the plate aerobically at 22 to 30C for 12 to 18 h.
medium is buffered by the bicarbonate in conjunction with Remove the filter from the medium, and continue incubat-
the level of atmospheric C02.After inoculation with a tuft ing the plate. Within the agar medium, the spirochetes form
of Beggiatoa spp., plates are placed in a micro-oxic atmos- a subsurface veil-like growth that expands toward the pe-
phere (0.5% C 0 2 ; 0.2% 0,; balance N2). Exposing the riphery of the plate and away from colonies of contaminat-
medium and bacteria to full air for approximately 20 min ing bacteria that may also have passed through the filter
every day or two, as needed for inoculation or single- pores. Obtain pure cultures by serial dilution in deep tubes
filament isolations from agar plates, poses no problem to the of the GYPT medium: Spirochaeta colonies resemble a cot-
success of the technique. Pure cultures resulting from re- ton ball or veil-like growth with a denser center.
peated single-filament isolations are maintained in sulfide-
oxygen gradient media (section 11.2.14.2). 11.1.6.2. Thin, Flagellated Bacteria
Aquaspirillum gracile is the thinnest of the aerobic, fresh-
11.1.6. Filterability water spirilla, having a cell diameter of 0.2 to 0.3 pm. It can
The ability of some bacteria to pass through a membrane fil- be isolated by the following procedure (44). Place a mem-
ter has been used to advantage for enrichments. In oligo. brane filter with a pore size of 0.45 pm on the surface of the
trophic environments many bacteria are very small and may isolation agar (section 11.6.4), and deposit 0.5 ml of pond
be able to pass filters with 0.45-pm pore size. The initial fil- or stream water in the center of the filter. Incubate the
tration of samples containing Themtoplasm species has al+ plates for 1.5 to 2.0 h at room temperature. Remove the fil-
ready been mentioned (section 11.1.2.3). The small size ter, and continue the incubation for 3 days or longer. A.
and plastic properties of these wall-less cells allow them to gracile is thin enough to penetrate the pores of the filter into
pass through the pores of a 0.45-pm-pore-size membrane fil- the underlying agar. Look for spreading, semitransparent
ter. During enrichment of bdellovibrios, the samples are areas of growth within the agar medium, and subculture
filtered before being placed with potential prey cells (sec- from these areas. Other small bacteria (small vibrios, cocci,
tion 11.3.1). A somewhat different approach consists of or short rods) may also pass through the filter to form small
placing a sterile filter on top of a suitable agar medium in a colonies o n the surface of the isolation medium; these
petri dish and inoculating the surface of the filter with the colonies can be distinguished easily from the subsurface,
sample containing the desired organism. If the organism is spreading, semitransparent growth typical of A.gracile.
11. Enrichment and Isolation 223

Serpens flexibilis can be found in the sediment of eu- development of contaminants. In the laboratory, suspend
trophic freshwater ponds. The cells are rod shaped with a material scraped from the natural substrates in sterile liquid
diameter of 0.3 to 0.4 k m and move through agar gels in medium, and streak several plates directly from the suspen-
a characteristic serpentine manner. For enrichment, place a sion in addition to preparing a liquid enrichment culture. If
sterile membrane filter with a pore size of 0.3 to 0.45 pm on the suspension contains many contaminants, wash it re-
the surface of the isolation agar (85). Deposit a small peatedly in sterile medium by low-speed centrifugation to
amount of pond water-mud slurry on the center of the filter. reduce the level of contamination before streaking plates.
Incubate for 6 to 12 h at 30C, remove the filter, and incu- Confirm the purity of cultures of cyanobacteria by mi-
bate the plate for 2 to 4 days. The organism grows as a sub- croscopic observation and also by inoculating into complex
surface veil; remove a sample from the edge of this veil, and media and incubating in the dark at 30C, where the
streak onto a second plate of sterile medium to obtain a pure cyanobacteria do not grow.
culture. See section 11.1.12.1 for a further enrichment technique
for cyanobacteria.
11.1.7. Visible Illumination for Phototrophs
Unlike chemotrophic bacteria, phototrophic bacteria can 11.1.7.2. Purple Nonsulfur Bacteria (29)
use light as a source of energy. This ability has been used for Although termed purple nonsulfur bacteria, a number of
enrichment of these bacteria: with light as the sole energy these species can in fact use sulfide as an electron donor for
source, the growth of phototrophs will be favored over that growth, but they do so only when the sulfide is maintained
of chemotrophs. However, chemotrophs may still be able to at low, nontoxic concentrations. Consequently, the organ-
grow to some extent by using organic compounds produced isms are ordinarily cultured with organic electron donors.
by the phototrophs. Rather specific enrichment for particular species is possible,
depending on the carbon source provided. For enrichment,
11 .I .7.1. Unicellular Cyanobacteria only substrates that cannot be fermented by nonpho-
For freshwater cyanobacteria (formerly called blue-green totrophic organisms are provided in the enrichment
algae), inoculate samples from ponds, streams, or reservoirs medium.
into tubes of BG-11 medium (section 11.6.12). For The purple nonsulfur bacteria can be readily isolated
cyanobacteria from marine environments, use MN medium from freshwater and marine sediments and are less fre-
(section 11.6.44); some grow poorly in this medium, so quently isolated from field, lawn, or garden soils. Use a basal
ASN-111 medium (section 11.6.6) is sometimes preferable. enrichment medium (section 11.6.8), and supplement it
Wash all glassware well, and then rinse successively in tap with a suitable carbon source. (The particular carbon source
water, concentrated nitric acid, and deionized water. used depends on the genus and species desired; consult ref-
Incubate enrichment cultures in an illuminated water bath erences 3 and 8 for specific compounds.) Place 0.1 g of the
at 35C; this temperature inhibits the growth of most eu. specimen into a screw-cap tube, and fill the tube completely
caryotic algae. Use a light intensity of 2,000 to 3,000 lx with the enrichment medium, freshly made to minimize
( 186 to 279 ft-candles, or approximately the illumination oxygenation. Tighten the cap so that there is no air space at
provided by a 55-W incandescent desk lamp at a distance of the top, and incubate the culture at ca. 25C. Illuminate
10 to 12 in. [25 to 30 cm]); some cyanobacteria may require the culture continuously with incandescent light (not fluo-
lower intensities (500 lx or less). Examine the enrichment rescent light); use a 50- or 75-W lamp at a distance of 40 to
cultures periodically until there is evidence of development 60 cm. Make sure that the lamp does not heat the cultures.
of cyanobacteria. Then, streak onto media solidified with Look for development of turbidity with a brown, yellow, or
2% bacteriological-grade agar. Incubate plates under illumi- pink tinge in 3 to 7 days. Transfer a drop of culture to a sec-
nation ( d o 0 lx) at 25C in air or in an atmosphere slightly ond tube of medium for a secondary enrichment. For purifi-
enriched with C 0 2 ; clear plastic boxes such as household cation, use a n agar medium containing 1% yeast extract or
vegetable crispers make good chambers to help prevent peptone, 0.2 g sodium malate, and 1.5% agar. Use the shake
evaporation. Use a dissecting microscope to detect the com- tube method (section 11.4.2), or use pour plates and incu-
pact, deeply pigmented colonies of nonmotile cyanobacte- bate in an anaerobic jar. Illuminate the cultures during in-
ria. Restreaking several times may be necessary to eliminate cubation. Many of the purple nonsulfur bacteria also grow
nonphototrophic bacterial contaminants. For motile in the dark under an air atmosphere or under microaerobic
cyanobacteria, enrich by placing a small patch of culture conditions, although the colonies are less highly pigmented
material at one side of a petri dish of agar medium. than when grown anaerobically in the light.
Illuminate the dish from the opposite side. The cyanobac-
teria will respond phototactically by gliding across the agar 11.1.7.3. Green Sulfur Bacteria
toward the region of higher light intensity. When some of The green sulfur phototrophs are not as easy to cultivate as
the organisms have reached the opposite side, subculture the green nonsulfur forms, although massive developments
them to a new plate and repeat the procedure until non- (blooms) of them are often readily visible to the eye, par-
phototrophic bacterial contaminants have been eliminated. ticularly in marine and brackish environments. Unless in-
This technique has also been used effectively with some fil- ocula from such blooms are available, a useful enrichment
amentous cyanobacteria (183). approach is to set up a Winogradsky column. This method
For slow-growing cyanobacteria such as members of the provides anaerobic conditions and a long-lasting supply of
order Pleurocupsales,primary cultures are best established by H2S, and successive blooms of the sulfur phototrophs (and
direct isolation of colonies on solid media rather than by many other microorganisms as well) usually result.
preliminary cultivation in a liquid medium (207). During To prepare a Winogradsky column, obtain mud from
transport of rock chips, mollusk shells, or macroalgae from freshwater, brackish, or marine environments (for example,
intertidal zones to the laboratory, keep the samples in closed mud from the edge of a freshwater pond or stream lake or
bottles or tubes containing a damp piece of filter paper; do from a salt marsh). Mix 3 parts of mud with 1 part of CaS04
not submerge the samples in seawater, since this promotes * H20. Add some insoluble organic material such as finely
224 w GROWTH

shredded filter paper or small pieces of roots from aquatic colonies of the larger organism by plating; moreover, none
plants. If paper is used, also add a small amount of of a variety of selection methods was applicable. To enrich
NH,MgPO,. Pour the mixture into a tall glass cylinder (at for the larger spirillum, the smaller organism was isolated
least 5 cm in diameter) to a height of at least 15 cm, stirring and used to immunize a rabbit. When the resulting anti-
to avoid air pockets. Hold the cylinder in the dark for 2 to serum was added to the mixed culture, the smaller spirilla
3 days to minimize the development of oxygenic photosyn- agglutinated and settled to the bottom of the tube. The sue
thetic organisms. Expose the cylinder to incandescent light pernatant, now containing a high proportion of the large
or to diffuse daylight at 18 to 25C during subsequent incu- spirilla, was used to obtain isolated colonies.
bation. Anaerobic decomposition of the organic material in Magnetic beads coated with antibodies may be used to
the column (with concomitant production of COZ, alco- trap target organisms for which antibodies are available
hols, fatty acids, hydroxy acids, organic acids, and amines) (47). This technique was used to isolate Azospirillum species
and the formation of HZS from the CaS0+ will provide from natural soils (8 1). Polyclonal antibodies raised in rab-
an appropriate array of microhabitats in which the sulfur bits against whole cells of a mixture of five strains of
phototrophs can thrive and form distinctive purple, red, or AzospiriUum were used to coat magnetic beads. These were
green patches on the sides of the glass column or layers or then mixed with natural samples and used to capture the
bands in the water column above the sediment-water inter- beads and bacteria on a magnet. The samples were washed
face. before being used for cultivation experiments. The lectin
Use Pasteur pipettes to obtain organisms from the glass concavalin A was used to isolate coliform bacteria from
surfaces or from the distinctive layers for microscopic ex- freshwater and sewage (159).The immunomagnetic separa-
amination, isolation, or further enrichment in liquid cul- tion method was successfully combined with matrix-assisted
tures. Alternatively, sequentially remove portions of the laser desorption ionization-time of flight mass spectroscopy
sediment with a spoon or spatula to expose the various (127).
zones of growth.
Enrichment can also be accomplished in defined liquid 11.1.1 1. Cell Density
media. Select for specific sulfur phototrophs by varying the The ability of some bacteria to float or to sink in aqueous
wavelength and intensity of the illumination used, the tem- environments or to form a band during density gradient
perature of incubation, and/or the type and concentration centrifugation has been used to separate these organisms
of the electron donor. For a clear, detailed exposition of the from contaminants.
many intricacies of such enrichment and the subsequent
isolation of sulfur phototrophs into pure culture, see the 11 .I .I 1.I. Flotation on Water for Lampropedia
very useful essay by Van Niel (199). hyalina (140)
The strictly aerobic, tablet-forming coccus Lampropedia
11.1.8. Visible Illumination To Enhance Pigment hyalina forms a characteristic hydrophobic pellicle on the
Production surface of liquid media. If complex media are inoculated
Some bacteria respond to visible light by producing colored with material from habitats rich in organic matter, the lam-
pigments, which can greatly aid the differentiation and iso- propedias often form contaminated pellicles containing
lation of these organisms. For example, Brevibacterium spp. their very distinctive cells. If such pellicles develop, transfer
produce a distinctive orange pigment only when exposed to them with a Pasteur pipette to a plate containing soil
visible light. Incubate plates in the light during the period seeded with starch or wheat grains covered with water.
of active growth, not after colonies have developed fully. After incubation, pellicles containing the tablets of L.
hyalina (as shown by microscopy) can be transferred to agar
11.1.9. Sonic Oscillation Selection of media for purification.
Sporocytophaga myxococcoides
Enrichment of the microcyst-forming cellulolytic cytophaga 11 .I .I 1.2. Swirling for Achromatium
Sporocytophaga myxococcoides takes advantage of a sonica- oxaliferurn (1 1 1)
tion-resistant form in the organisms life cycle (112). Place The relatively high density of cells of Achromatium oxa-
100 ml of sporocytophaga medium (section 11.6.64) into a liferum is caused by the accumulation of intracellular
500-ml Erlenmeyer flask, and inoculate with ca. 0.1 g of CaC03, and this property can be exploited to concentrate
soil, mud, or plant material. After incubation at 30C for 7 the organism from natural samples. Place a small amount of
to 10 days with moderate agitation, remove 5 ml of culture, sediment from a suspected source in the bottom of a large
subject it to sonic oscillation for 15 to 30 s, and then use it beaker, and cover it with ca. 1 cm of slightly alkaline water.
to inoculate a secondary enrichment flask. After ca. 5 to 7 Tilt the beaker, and gently swirl the contents to separate the
days, look for a distinctive yellow hue in the flask, and use achromatia as a thin, white deposit directly above the sedi-
phase-contrast microscopy to look for both microcysts and ment. Transfer the cells to another beaker with a Pasteur
vegetative cells on and around the cellulose fibers. Subject pipette, and repeat the process until the achromatia are free
a portion of this culture to sonic oscillation, and then use of contaminants.
the pour plate method to obtain isolated colonies, as de-
scribed in section 11.2.12.5. 11 .I .I 1.3. Density Gradient Centrifugation
The buoyant density of bacteria in pure culture and in sam-
11.1.1 0. Use of Antiserum Agglutination or ples from natural aquatic environments has been studied by
Magnetic Beads Coated with Antibodies density gradient centrifugation in Percoll gradients, and the
The use of antiserum may enrich for the minority organism average density of a representative bacterium is 1.080
in mixed cultures. In a study of a culture in which two pg p,m-3 (78). There do not seem to be significant differ-
species of marine spirilla were present-a large and a small ences in density among bacteria; however, the density of a
organism-the smaller organism was found to be greatly given bacterium can change by as much as 7% from the av-
predominant. This made it impossible to obtain isolated erage as a result of the formation of inclusions such as sulfur
11. Enrichment and Isolation 225

and phosphorus storage materials or the formation of cap- taminating bacteria may survive, but if, before irradiation,
sules and/or gas vesicles. Such inclusions may be responsible the sample suspension is incubated for a time in a medium
for the occurrence of two or more bands of organisms when that allows endospores to germinate, this decreases the
samples from natural environments are subjected to density number of surviving sporeformers. After irradiation, plate
gradient centrifugation (78). Therefore, the density gradi- the samples onto a suitable medium such as TGYM medium
ent technique may be useful in enriching for organisms with (section 11.6.69) and incubate at 25 to 30C. Look for
different types of inclusions. colonies that are pink, orange+red,or red.
Another enrichment application of density gradient Deinococci can also be selected from soil by preparing a
centrifugation is the use of Urografin gradients to com- slurry of the soil and exposing the supernatant in a petri
pletely separate endospores from vegetative cells in sporu- dish to UV light at 600,900, and 1,200 J mP2 (142).
lating cultures of Bacillus spp. (191).
For further information on density gradient centrifuga- 11.1.1 3. Magnetic Field for Magnetotactic Bacteria
tion, see chapter 7.3. (138)
A number of aquatic bacteria contain small enveloped crys-
11 .I .1 1.4. Sampling the Neuston Layer for Nevskia tals of the iron oxide magnetite, which enable the cells to
rarnosa become oriented in a magnetic field. The bacteria are able
Nevskiu ramosa is abundant in the epineustic layer on the to move along the magnetic lines of force and, in natural
surface films on calm water bodies. To isolate Nevskia, sam- bodies of water, also move downward, since the Earth's geo-
ples are taken with a sterile loop needle from the surface magnetic field has a downward component. Magnetotactic
film and transferred to unshaken enrichment cultures with bacteria can be enriched from natural samples, such as pond
a medium free of combined nitrogen compounds (section water and sewage, by the following procedure. Place the water
11.6.50) (188). At room temperature (20 to 24C) Nevskia and sediment in a 2-liter beaker wrapped in opaque mate-
rumosu develops typical rosettes that consist of flat, binary rial and covered with plastic wrap. Attach a stirring-bar
branching polysaccharide stalks with cells in the tips. For magnet to the beaker, with the south pole (use the north
isolation the cells can be streaked out on agar. Addition of pole in the Southern hemisphere) touching the side of the
ammonium salts to the medium results in submersed beaker and positioned about 5 to 8 cm above the sediment.
growth. The magnetotactic cells will congregate near the magnet
and can be removed with a pipette.
11.1.I 2. Radiation Resistance A simple "racetrack" for selection of magnetotactic
Some bacteria are resistant to high doses of radiation such bacteria can be constructed (216). Briefly, prepare a capil-
as UV light, X-rays, and gamma-radiation, and this can be lary tube from a Pasteur pipette and seal the small end in
used for selection purposes by killing the less resistant con- a flame. Fill the sealed capillary with filter-sterilized water
taminants that may be present in a mixed culture. The fol- (preferably water collected from the surface layer of sedi-
lowing examples illustrate this principle. ment from which the inoculum is to be taken) by means
of a syringe and needle. The large, nonsealed end of the
11 .I .I 2.1. UV Irradiation for Cyanobacteria capillary forms a well whose bottom can be plugged with
It is often difficult to obtain cultures of cyanobacteria free a tiny wad of cotton. Place a drop of sediment contain.
from bacterial contaminants, which frequently penetrate ing magnetotactic bacteria into the well, and position a
and live in the gelatinous sheaths that surround the cells stirring-bar magnet near the opposite, sealed end of the
and filaments of cyanobacteria (section 11.1.7.1). However, capillary. Magnetotactic bacteria migrate from the well
by treating a suspension with UV light for an appropriate through the cotton plug, travel along the capillary, and ar-
period, it is possible to kill the contaminating bacteria and rive at the sealed end in a few minutes. The migration can
yet recover viable cyanobacteria. Success has been reported be followed by dark-field microscopy. Harvest the accu-
with the following method (75). Place a dilute suspension mulated cells by aseptically breaking the capillary near the
of cyanobacteria in a quartz chamber and irradiate with sealed end and removing the contents with a narrow ster-
275-nm UV light from a quartz-jacketed mercury vapor ile pipette.
lamp. Agitate the suspension by continuous stirring during Some magnetotactic bacteria have been isolated in pure
the incubation. At periodic intervals during the irradiation, culture (26,30, 130, 171).
remove samples and prepare a large number of dilution cul-
tures from each sample. In the dilution cultures that show 11.1.14. lsoelectric Focusing of Cells
growth of the cyanobacteria, test for bacterial contamina- Isoelectric focusing (IEF) can be used to separate cells based
tion by performing microscopic examination and by inocu- on their surface charges (97) (Fig. 1).In a pH gradient of 2
lating a variety of bacteriological media. One disadvantage to 10 and an electric field of 11.5 V cm-', cells are trans.
of this method is that the final pure culture may contain ported to the place where their net charge is compensated.
cyanobacteria in which mutations have occurred because of By this technique a mixture of cells from Chlorobium limicola
the UV light treatment. 6230, Pseudomom stutzeri DSM 50227, and Micrococcus lu-
teus DSM 20030 were successfully separated. A density gra-
11.1.1 2.2. X-Ray and Gamma-Radiation Resistance dient of Ficoll prevented convective currents in the system.
for Deinococci (141, 142) The method was also tested with a concentrated mixture of
Members of the family Deinococcaceae, although not spore- bacteria from a shallow eutrophic lake and yielded up to 10
formers, are highly resistant to X-rays and gamma-irradia- different bands. Species composition in each IEF band was
tion, and this property can be used to select for them. analyzed by PCR and DGGE. Each IEF band exhibited a
Samples suspected of containing deinococci (such as different species composition. After the separation of cells
ground meat, fish, fecal samples, and sawdust) can be irra- by IEF, three times more 1 6 s rRNA signals could be de-
diated with 1.0 to 1.5 Mrad of gamma-radiation, killing tected by DGGE than in the unfractionated natural bacte-
most or all other vegetative cells. A few endospores of con- rial community. At the same time, the IEF fractions were
226 m GROWTH

FIGURE 1 Isoelectric focusing. Schematic (A) and photograph (B) of apparatus for isoelectric
focusing of whole bacterial cells. The cathode (-) and anode (+) consist of platinum wires which
are connected to a power supply (97).

enriched for certain species and could be used in subsequent sputum samples, provided that aerosol-generating manipu-
cultivation experiments. lation of such specimens are conducted in a class I or I1
biological safety cabinet. Biosafety level 3 practices, con-
tainment equipment, and facilities are recommended for
11.2. BIOCHEMICAL ENRICHMENT activities involving the propagation and manipulation of
cultures that may contain Mycobucterium tuberculosis ( 164).
11.2.1. Alkali Treatment for Mycobacteria (24) See chapter 46 for biosafety methods.
Add a maximum of 10 ml of the suspected sputum sample
to a sterile, disposable, plastic 50-ml conical centrifuge tube 11.2.2. Incubation at Alkaline pH for Vibrios and
with a leakproof and aerosol-free plastic screw cap. Add an Sporosarcina ureae (24, 49)
equal volume of N-acetyl-L-cysteine-sodiumhydroxide so- To select for Vibrio cholerue from a fluid stool sample or rec-
lution (section 11.6.49). Tighten the screw cap, and mix tal swab, directly streak the specimen heavily onto a selec-
well in a Vortex mixer for a maximum of 30 s. Allow to tive medium such as TCBS agar (section 11.6.68). If there
stand at room temperature for 15 min. The N-acetyl-L- are only small numbers of V.cholerue in the specimen, inoc-
cysteine functions as a mucolytic agent; it converts the ulate 20-ml portions of alkaline peptone water (section
thick sputum to a thin, watery consistency. A n extra pinch 11.6.3), incubate for 5 h at 35"C, and then streak this cul-
of the powder may be needed to liquefy highly mucoid spu- ture heavily onto the TCBS agar. The alkalinity of the pep-
tum samples. The function of the sodium hydroxide is to de- tone water (pH 8.4) and the TCBS (pH 8.6) inhibits the
stroy many of the contaminants present in the sputum; the growth of most contaminants. On the TCBS agar, colonies
mycobacteria are relatively resistant to the alkaline treat- of V. chokrue are yellow (sucrose fermenting) and oxidase
ment. Fill the centrifuge tube to within 1 cm of the top with positive.
sterile 0.067 M phosphate buffer to neutralize the action of Sporosurcim ureue in soil samples can also be isolated on
the sodium hydroxide. Centrifuge at 3,600 X g for 15 min alkaline agar media (pH 8.5). Streak the medium (tryptic
to concentrate the mycobacteria. Carefully decant the su- soy-yeast extract agar with urea [section 11.6.751) with a di-
pernatant into a splash-proof container. Add 1 to 2 ml of lution of the soil, incubate at 30C, and check colonies
sterile water or buffer to suspend the sediment, and use this microscopically for the presence of packets or tetrads of
suspension to inoculate suitable culture media (for an ex- cocci, which may be motile. To test for endospore formation,
ample, see section 11.6.37). transfer suspected colonies to nutrient agar to which has
CAUTION: Biosafety level 2 practices, containment been added 20 ml of a 10% (wtlvol) solution of urea (previ-
equipment, and facilities are recommended for culturing ously sterilized by filtration) and 50 mg of MnS04 * HlO
11. Enrichment and Isolation 227

per liter. Incubate at a temperature below 25C. Endospores 11.2.5. Inhibition by Toxic Metals
are round and refractile by phase-contrast microscopy and The salts of heavy metals such as tellurium, thallium, and
are located centrally or laterally within the cocci. selenium can, at low concentrations, exert an inhibitory ef-
fect on many bacteria. The use of a particular metal salt at
11.2.3. Acid Treatment for legione//a Species an appropriate concentration can allow some bacteria to
Isolation of Legionella species, particularly those from the grow while inhibiting the growth of others. Some examples
environment, can sometimes be facilitated by acidification follow.
of samples to pH 2.2, which kills contaminants more
quickly than it does the Legionella species. The following 11.2.5.1. Tellurite Inhibition for Corynebacteria and
procedure is based on the isolation of Legionella shakespearei Certain Streptococci
from water taken from an evaporative cooling tower (201). Potassium tellurite inhibits gram-negative bacteria and
Pass 1 liter of water suspected of containing Legionella bac- most gram-positive bacteria when used at a suitable con-
teria through a 0.45-pm-pore-size membrane filter. Cut the centration. To select for corynebacteria such as Coryne-
filter into small pieces with sterile scissors, and suspend the bacterium diphtheriae, use potassium tellurite at a concentra-
pieces in a tube containing 20 ml of sterile distilled water. tion of 0.0375%, as in cystine tellurite blood agar (section
Cap the tube, and shake vigorously by hand for 1 min to 11.6.22). Colonies of corynebacteria are gray or black as a
suspend the bacteria. Centrifuge 10 ml of the suspension result of reduction of the tellurite. For selection of certain
at 4,000 rpm for 30 min, and then remove and discard 9 ml streptococci (Streptococcus mitis and S. salivarius) and en-
of the supernatant. Acidify the remaining 1 ml with 9 ml of terococci, use potassium tellurite at a concentration of
pH 2.2 KC1-HC1buffer (add 5.3 ml of 0.2 N HC1 and 25 ml 0.001%, as in mitis-salivarius agar (section 11.6.43). S. mitis
of 0.2 N KC1 to 100 ml of distilled water, adjust the pH to forms tiny blue colonies, S. salivarius forms larger blue
2.2, and sterilize by filtration). Allow to stand for 5 min. gumdrop colonies, and enterococci form small blue-black
Spread 0.1-ml portions onto the surface of BCYE agar plates colonies.
(section 11.6.11) supplemented with glycine (3 g/liter),
polymyxin B sulfate (79,200 IU/liter), vancomycin (5 11.2.5.2. Thallium Inhibition for Mycoplasmas
mg/liter), and cycloheximide (80 mg/liter). Incubate the and Enterococci (23, 105)
plates at 35C for 7 days in a vessel containing a highly
humid air atmosphere. Look for colonies that exhibit a Use thallous acetate at a concentration of 0.023%, as in E
ground-glass appearance when viewed under obliquely agar (section 11.6.24) and E broth (section 11.6.25), to se-
transmitted light. Presumptive identification of an isolate as lect for mycoplasmas such as Mycophma pneumoniae from
being a member of the genus Legionella is based on demon- the respiratory tract. Extract specimens collected on swabs
stration of small (diameter, 0.5 pm), aerobic, gram-negative into 2 ml of soybean-casein digest broth (section 11.6.62)
rods that require iron and cysteine for growth. containing 0.5% bovine serum albumin. Inoculate 0.1-ml
amounts of the suspension into biphasic E medium (section
11.2.4. Incubation at l o w pH 11.6.24) and also onto plates of E agar. Incubate the cul-
Most bacteria are inhibited by highly acidic conditions, but tures at 37C aerobically in sealed containers. Examine the
some are able to thrive, e.g., certain bacteria that live on plates at intervals for up to 30 days with a dissecting micro-
fruit, are used in cheese ripening, or oxidize reduced-sulfur scope (magnification, x20 to X60) for the appearance of
compounds to sulfuric acid. Incubation at low pH values minute colonies (diameter 10 to 100 pm) with a typical
can be very useful for the selection and isolation of these or- friedeegg appearance. Examine the biphasic cultures mi-
ganisms. croscopically by looking through the side of the tube for
spherules (fluid medium colonies); also observe the cultures
11.2.4.1. Lactobacilli (126) for a decrease in pH (yellowing of the phenol red indicator).
For lactobacilli in cheddar cheese, streak dilutions of the Inoculate E agar plates from the biphasic medium to obtain
cheese on modified Rogosas medium (section 11.6.46). isolated colonies. For principles of biphasic media, see chap-
This medium has a pH of 5.35 because of an acetic acid/ ter 12.1.
acetate buffer system. At this pH, lactobacilli such as Thallous acetate has also been used at a concentration of
Lactobacillus casei and L. planiurum form colonies, but the 0.1%, as in thallous acetate agar (section 11.6.70) to select
common dairy organism Luctococcus lactis does not. for enterococci.

11.2.4.2. Thiobacillus thiooxidans (203) 11.2.5.3. Selenite Inhibition for Salmonellae


Inoculate shallow layers of Thiobacillus thiooxldans medium Use sodium hydrogen selenite at a concentration of 0.4%,
(section 11.6.73) with samples from soil, mud, or water as in selenite F broth (section 11.6.60), to temporarily sup-
(marine mud is the most reliable source). Look for a drop in press the growth of coliforms while allowing salmonellae to
pH to 2.0 after 3 to 4 days or more, which virtually ensures grow. In addition to directly streaking stool samples onto se-
the predominance of this acid-tolerant organism. Purify by lective and nonselective agar media, inoculate selenite F
streaking on the solidified medium. broth heavily (ca. 1 g or 1 ml of stool specimen in 8 to 10
ml of broth). Incubate this enrichment at 35 to 37C for 12
11.2.4.3. Frateuria Species (190) to 16 h, and then streak onto the plating media.
Members of the genus Frateuria are commonly found on
fruit and can be enriched on Frateuria isolation medium 11.2.6. Phenylethanol Inhibition for
(section 11.6.26), which has an initial pH of 4.5. Inoculate Gram-Positive Cocci
the medium with fruit samples, and incubate at 30C. Use phenylethanol in agar media at a concentration of
Streak plates of GYC agar (section 11.6.30) with material 0.25%, as in phenylethyl alcohol agar (section 11.6.54), to
from the enrichment. GYC agar contains insoluble CaC03, inhibit the growth of gram-negative bacteria, particularly
and clear zones will form around the Frateuria colonies. Proteus species, when these occur in mixed culture with
228 GROWTH

gram-positive cocci. For example, one application is the 11.2.8.3. Halornonas Species (205)
isolation of coagulase-positive staphylococci from a stool Halomonads are eubacteria that tolerate both high and low
specimen. salt concentrations; all known species grow in NaCl con-
centrations from 0.2 to 25%, depending on the type of
11.2.7. Dye Inhibition for Cram-Negative Bacteria, medium used (205). They have been isolated from a wide
Mycobacteria, and Arthrobacters variety of saline environments, including solar salterns, the
Gram-positive bacteria are generally inhibited by lower Dead Sea, manganese nodules from the ocean, underground
concentrations of triphenylmethane dyes (such as crystal salt formations, and the Antarctic. They can be isolated on
violet, basic fuchsin, brilliant green, and malachite green) high-salt-containing media such as CAS medium (section
than are gram-negative bacteria. For example, the presence 11.6.19). They form white to yellow colonies, not pink or
of malachite green at a concentration of 1:4,000,000 in red as do the archaeal halophiles.
media inhibits the growth of Bacillus subtilis and at
1:1,000,000 inhibits the growth of staphylococci; however, 11.2.9. Bile Inhibition for Enteric Bacteria
concentrations of 1:30,000 to 1:40,000 are required to
Bile or bile salts are often incorporated into culture media
inhibit E. coli or Salmonella enterica serovar Typhi (72).
as selective agents for intestinal bacteria. There are some
Brilliant green is used in media for the confirmed test for co- exceptions to this selectivity; for example, the plague bacil-
liforms, as in brilliant green lactose bile broth (section
11.6.17), and in several selective media for members of the
lus Yersinia pestis can grow in pure bile even though it is not
an intestinal organism (146). However, Y. pestis is closely
Enterobucteriuceae,such as salmonella-shigella agar (section related to Y. pseudotuberculosis, an intestinal organism, so
11.6.59) and brilliant green agar (section 11.6.16). Crystal
perhaps the bile tolerance is not surprising. Nevertheless,
violet is also used for selection of members of the Entero-
the rule holds sufficiently well to make bile an important se-
bucteriaceue, as in violet red bile agar (section 11.6.78) and lective agent for gram-negative enteric rods and for entero-
MacConkey agar (section 11.6.38). Mycobacteria are very
cocci. For selection of members of the family Entero-
resistant to dyes; malachite green is often incorporated into
bacteriuceue, bile or bile salts are incorporated into such
media used for isolation of M. tuberculosis to inhibit the
media as MacConkey agar (section 11.6.38), salmonella-
growth of contaminants, as, for example, in Lowenstein-
shigella agar (section 11.6.59), violet red bile agar (section
Jensen medium (section 11.6.37). Methyl red has been used
11.6.78), and many others. For enterococci, bile esculin
to select for soil arthrobacters (section 11.6.5); the dye in-
agar (section 11.6.13) has proved to be an excellent selec-
hibits other gram-positive bacteria but not arthrobacters
tive and differential medium.
(79).
11.2.8. Salt inhibition 11.2.10. Antibiotic Inhibition
The growth of some bacteria is not inhibited by high con- Antibiotics provide one of the easiest ways to devise a se-
centrations of NaC1, and some bacteria actually require a lective medium for strains of a particular species or genus. It
high NaCl concentration for growth. For instance, the red makes no difference whether the organisms have medical
extreme halophiles such as Halobacterium species need at importance or not; for instance, they may merely be harm-
least 15% NaCl. Regardless of whether it is required or less soil or water organisms. Obtain several reference strains
merely tolerated, NaCl has proven to be useful as a selective of the bacterial taxon, and test them against a wide spec-
agent for certain groups of bacteria. trum of individual antibiotics. Identify antibiotics to which
all the strains are resistant, and incorporate several of these
11.2.8.1. Staphylococci antibiotics into an appropriate sterile agar medium. When
Most Staphylococcus species grow in media containing 10% samples from nature are streaked onto the medium, the de-
NaC1. For selection of staphylococci from foods and miscel- sired organisms, if present, will be able to grow, whereas
laneous environments, mannitol salt agar (section 11.6.39), many of the contaminant organisms will be suppressed by
which contains 7.5% NaC1, is a useful medium. The growth the antibiotics. A few specific examples follow.
of many other genera of bacteria is suppressed at this con- (i) Because mycoplasmas lack cell walls, they are resis-
centration of salt. tant to even very high concentrations of penicillin concen-
trations that inhibit most other bacteria. For instance, the
11.2.8.2. Halobacteria (76) antibiotic is used at a concentration of 194 U/ml in my-
The archaeobacterial genera Halobacterium and Halococcus coplasma isolation media such as E agar (section 11.6.24).
occur in heavily salted proteinaceous materials (such as (ii) Penicillin is useful in the isolation of Bordetella per-
salted fish), in salterns, and in the Dead Sea and other tussis from the nasopharynx because it helps to suppress the
highly saline lakes. They can often be isolated from samples growth of members of the normal flora while permitting B.
of solar salt. They require a high concentration of NaCl for pertussis to form characteristic pearl-gray (mercury-drop)
good growth (ca. 25% NaC1) and cannot grow with less colonies in ca. 4 days. The clinical specimen (obtained by
than ca. 15% NaC1. In fact, they are killed by even brief ex- means of a nasopharyngeal swab) is streaked onto plates of
posures to salt concentrations of less than ca. 15%. The use Bordet-Gengou agar (section 11.6.15) containing penicillin
of NaCl concentrations of 20% or greater makes their se- (0.5 U/ml).
lection from natural sources a simple matter. For an exam- (iii) Antibiotics are necessary when attempting to iso-
ple of a suitable enrichment medium, see section 11.6.32. late Campylobucter jejuni or other Campylobucter species
Incubate inoculated flasks at 37C with agitation, and sub- from stool specimens, in order to suppress the members of
sequently obtain isolated colonies by streaking from the en- the normal flora that otherwise would rapidly outgrow the
richment cultures onto plates of medium solidified with 2% campylobacters. One example of a medium suitable for iso-
agar. Incubate the plates at 37C for 3 to 14 days in plastic lation of C. jejuni is Modified Campy BAP (section
bags to prevent excessive drying. Look for the development 11.6.45), which contains cephalothin, polymyxin B,
of pink or red colonies. trimethoprim, vancomycin, and amphotericin B.
11. Enrichment and Isolation 229

(iv) Three antibiotics-vancomycin, colistin, and covered with paper or aluminum foil. Examine the surface
nystatin-aid in the selection of Neisseria rneningitidis from film daily by phase-contrast microscopy. When prosthecate
nasopharyngeal samples (to detect healthy carriers of the bacteria occur in a relative proportion of ca. 1 in 10 or 20
organism) and of Neisseria gonorrhoem from clinical speci- cells (usually in about 4 days), streak the surface film onto
mens (urethral exudates, cervical swabs, etc.). These an- plates of tap water agar containing 0.05% peptone and incu-
tibiotics are incorporated into Thayer-Martin agar (section bate at 10C. The low peptone level allows the caulobacters
11.6.7 1) and help to suppress the growth of members of the to form tiny colonies but does not allow heavy overgrowth
normal flora. The vancomycin suppresses gram-positive by other bacteria. After 4 days or more of incubation, select
contaminants, the colistin acts mainly against gram-nega- microcolonies of caulobacters under a dissecting microscope
tive contaminants, and the nystatin is an antifungal agent. and transfer as patches to a richer medium (0.2% peptone,
Table 1 lists some other examples of bacteria for which 0.1% yeast extract, 0.02% MgS04 . 7 H20, 1.0% agar).
antibiotics serve as selective agents. Do not overlook the After 2 days of incubation, prepare wet mounts from the
use of cycloheximide, nystatin, and other antifungal agents patches to detect caulobacters. Obtain isolated colonies by
as general inhibitors of fungi in selective media, especially streaking growth from the patches onto fresh plates. For ma-
when the source of the inoculum is the soil. rine caulobacters, add 0.01% peptone to samples of stored
seawater and incubate at 13C for ca. 7 days. When micro-
11.2.1 1. Dilute Media scopic observation indicates the development of a suitable
Many bacteria are residents of soil and aquatic habitats low proportion of prosthecate bacteria, streak the surface film
in nutrients and have difficulty growing in rich media. Also, onto plates of seawater agar containing 0.05% peptone and
many potential contaminants cannot compete in dilute incubate at 25C. Subculture microcolonies to fresh plates as
media, so that the shortage of nutrients becomes a selective patches, and later purify by streaking.
factor. Below are some examples of the use of dilute media
for isolation of various soil or water bacteria. 11.2.1 1.2. Aquatic Spirilla (213)
In dilute media, aerobic chemoheterotrophic spirilla (the
11.2.1 1 . I . Caulobacters (158) genera Aquapirillurn and Oceanospirillurn) can often com-
Caulobacters are prosthecate bacteria that can grow at lev- pete successfully with other bacteria for the nutrients pres-
els of nutrients that do not support good growth of many ent. For freshwater spirilla, add 1% peptone or yeast au-
contaminants. To samples of water from ponds, streams, or tolysate to samples of source water (e.g., water from
lakes or to samples of tap water add 0.01% peptone and in- stagnant ponds) and incubate at room temperature for ca. 7
cubate aerobically at 20 to 25C in bottles or flasks loosely days or until the spirilla become numerous. Then, add part

TABLE 1 Examples of the use of antibiotics in selective media


Bacterium Antibiotic Concn Medium (chapter section) Reference(s)
Agrobacterium biovar 1 Penicillin G 97,500 U/liter Agrobacterium biovar 1 isolation medium (11.6.1) 106
Streptomycin 30 mg/liter
Cycloheximide 250 mg/liter
Tyrothricin 1 mg/liter
Bacitracin 6,500 U/liter
Agrobacterium biovar 2 Cycloheximide 250 mg/liter Agrobacterium biovar 2 isolation medium (11.6.2) 106
Bacitracin 100 mg/liter
Tyrothricin 1 mg/liter
Brucella spp. Bacitracin 25 U/ml Brilliant green lactose bile broth ( 11.6.17) 53,65
plus antibiotics
Polymyxin B 5 U/ml
Cycloheximide 100 p,g/ml
Vancomycin 20 w/ml
Nalidixic acid 5 pg/ml
Nystatin 1 U/ml
Micrococm spp. Furazolidone 20 mdliter Frateuria enrichment medium (11.6.26) 56, 108,212
(from skin)
Nocardia spp. Demeclocycline" Diagnostic Sensitivity Test Agar (Oxoid) 150
plus antibiotics
or methacycline
Pseudomonas spp. Cycloheximide Pseudomom isolation medium (11.6.56) 77
Nitrofurantoin
Nalidixic acid
Spirochaeta spp. Rifampin Sphaerotilus medium (11.6.63) 44
'Formerly known as demethylchlortetracycline
230 rn GROWTH

of this initial culture to an equal part of the source water, days. Examine microscopically for evidence of filamentous
and sterilize by autoclaving. Inoculate this mixture from the growth after day 2. Obtain pure cultures by selecting a fila-
unsterilized portion of the original culture. After incubation ment from the enrichment broth and streaking it onto plates
and further development of the spirilla, dilute a portion of of a solid medium (0.05%meat extract, 1.5% agar). Incubate
the second culture with more source water, sterilize the mix- plates for 24 h at 25"C, and examine under a dissecting mi-
ture, and inoculate it from the unsterilized portion. croscope for the typical curling filaments of Sphae~otilus
Continue to deplete the nutrients in this manner until the species. Transfer isolates to a Trypticase-glycerol broth
spirilla predominate. Obtain colonies by streaking onto (Trypticase [BBL Microbiology Systems, Cockeysville, MD],
plates of MPSS agar (section 11.6.48). 5 g; glycerol, 5 g; distilled water, 1,000 ml [pH 7.0 to 7.2]),
Another approach depends on low levels of nitrogen and incubate for up to 2 weeks. Sphaerotilus species form a
sources (213). Supplement samples of source water with 1% heavy surface pellicle in 2 to 3 days, and the underlying
calcium malate or lactate, and incubate at room tempera- broth remains clear. If turbidity develops, contamination
ture for ca. 1 week. Make a serial transfer into sterile source has occurred. Even without development of turbidity,
water containing 1% of the carbon source, and incubate. Sphaerotilus species should be reisolated from the surface pel-
Continue in this manner (three or four serial transfers) licle by an additional streaking onto meat extract agar to en-
until the spirilla predominate. With this method it is im- sure purity. To confirm that the isolated organisms form a
portant not to add a nitrogen source such as NH&1 in sheath, place a small piece of slime growth on a slide in a
order to prevent overgrowth of the spirilla by contaminants. drop of water, apply a coverslip, and press down on the cov-
For marine spirilla, mix the seawater sample with an equal erslip with blotting paper. Place a very small drop of 1% crys-
volume of Giesberger base medium (section 11.6.28) sup- tal violet solution at the edge of the coverslip so that it flows
plemented with 1% calcium lactate (213). After incuba- into the preparation by capillary action. After 30 s, press
tion, remove a portion of the original culture and incubate. again with blotting paper to remove excess dye and observe
Continue to successively deplete the nitrogen content by with a bright-phase oil-immersion lens. Both the cells and
repeating this procedure until the spirilla become the pre- the sheath should be clearly visible.
dominating organisms. Obtain isolated colonies by streak-
ing onto MPSS agar (section 11.6.48). 11.2.1 1.4. Marine Oligotrophic Bacteria Growing
Another way to enrich for spirilla is to use a continuous- on Natural Seawater Medium
culture system that provides low levels of nutrients. For in- The natural dilute medium that sustains a great diversity of
stance, in chemostat experiments with a mixture of a ma- microbial life is natural freshwater or seawater. Novel oligo-
rine spirillum and a pseudomonad (95), the growth rate of trophic marine bacteria have been isolated on unamended
the spirillum exceeded that of the pseudomonad when the seawater as the medium (52). Freshly collected seawater is
dilution rate of the chemostat was decreased to the point at filtered through a 0.2-pm-pore-diameter Supor membrane
which the limiting carbon and energy source (lactate) fell and is immediately autoclaved. In order to restore the bi-
below 10 mg/liter. In other experiments the growth rate of carbonate buffer lost during autoclaving, the seawater is
the spirillum exceeded that of E. coli when the lactate con- sparged with sterile COZ for at least 6 h, followed by sterile
centration fell below 5 mg/liter (96). In similar experiments air for at least 12 h. Acid-washed polycarbonate containers
with a freshwater spirillum and a pseudomonad, when the are used for media whenever possible. Before use, the media
lactate concentration fell below ca. 0.09 mg/liter the pseu- are checked for sterility by directly counting cells stained
domonad was eliminated from the chemostat as a nongrow- with 4',6-diamidino-2-phenylindole(DAPI) as described
ing population (129). The more efficient scavenging ability earlier (198), except that 1% formaldehyde is used.
of the spirillum for lactate may be attributable to a lower K,,, Isolations on seawater medium often use dilution ap-
and a higher V,, of the transport system for lactate and proaches (extinction culturing; see section 11.4.3.2.), to ob-
also to a higher surface-to-volume ratio for the spirillum tain numerically dominant members of the bacterioplank-
(129). ton community. In this way, several cosmopolitan lineages
Under starvation conditions, spirilla appear to have a of oligotrophic marine bacteria, including the SARl 1 clus-
survival advantage (128). This may be related to their abil- ter within the alpha-proteobacteria, were brought into pure
ity to form intracellular reserves of poly-p-hydroxybutyrate culture years after their molecular detection by 16s rRNA
under conditions of prior growth with limiting levels of car- sequencing of bacterioplankton samples.
bon and energy sources. The role of poly- p-hydroxybutyrate
in bacterial survival has also been reported for other types 112.12. Special Substrates
of bacteria (60). Consequently, starvation conditions If a culture medium contains a sole carbon or nitrogen
should be considered as a possible way to select for bacteria source or a selective electron acceptor that can be used only
that form this polymer. by a particular species or group of bacteria, the growth of
that species or group will be favored over that of other or-
11.2.1 1.3. Sphaerotilus Species ganisms that may be present. However, be aware that other
The sheathed bacteria of the genus Sphaerotilus occur in organisms may be able to grow to some extent by using
streams contaminated with sewage or organic matter and products synthesized by the favored organisms; thus, this
form slimy tassels attached to submerged surfaces. They can method is not absolutely selective but may merely increase
also be isolated from rivers, open drains, or ditches where the proportion of the desired organism in the population.
there is no initial evidence of their presence. The enrich-
ment procedure takes advantage of the ability of Sphaerotilus 11.2.12.1. Tryptophan for Pseudomonads (1 17)
species to grow at very low nutrient concentrations. To To enrich for pseudomonads capable of using tryptophan as
50-ml volumes of Sphaerotilus medium (section 11.6.63), the sole carbon and nitrogen source, inoculate a 250-ml
add 25-ml volumes of the water sample or 1-, 5-, and 10-ml Erlenmeyer flask containing 40 ml of tryptophan medium
portions of settled sewage or the settled liquor from various (section 11.6.76) with ca. 0.1 g of soil. Incubate with shak-
stages of sewage treatment. Incubate at 22 to 25C for 5 ing at 25C for 5 to 7 days. Transfer 0.1 ml to a second flask
Next Page

1 1 . Enrichment and Isolation w 231

of medium, and incubate for 2 to 3 days. After a further se- to provide anoxic conditions. Fill the bottles completely
rial transfer, obtain pure cultures by streaking onto trypto- with the medium. Incubate in the dark at 30C. Be sure
phan medium solidified with 15 g of agar per liter. After 1 that the bottles remain completely filled by adding fresh
to 3 days of incubation, subculture isolated colonies to agar medium if necessary. Hyphomicrobia generally develop in
slants. ca. 8 days. Monitor their development by phase-contrast
microscopy: look for rod-shaped cells with pointed ends or
1 1.2 .I 2.2. N2 for Azospirillurn and Azotobacter oval, egg-shaped, or bean-shaped forms, which produce fil-
Species amentous outgrowths (prosthecae) that vary in length and
Azospirillum brasilense and A. lipoferum are microaerophilic may show branching. Prepare a secondary enrichment, this
nitrogen fixers associated with the roots of a variety of time with sterile medium. Finally, streak onto solidified
plants; they also occur in soil (61, 62). Place washed root Hyphomicrobium medium and incubate aerobically to obtain
pieces 5 to 8 mm long, macerated with a forceps, into ni- isolated colonies.
trogen-free semisolid medium (Nfb medium [section
11.6.5 I]). Alternatively, inoculate the medium with a loop- 11.2.12.5. Cellulose for Cellulolytic Cytophagas
ful of soil. Incubate with agitation for 40 h at 32C; then, Various methods and media have been devised for enrich-
test the enrichment culture for acetylene-reducing activity ment of cellulolytic cytophagas (163). A plate culture
(see chapter 15.3.11). Be careful not to disturb the dense method for enrichment is as follows. Place autoclaved,
subsurface pellicle that forms in the medium, since this may round, filter paper discs, such as are used in chemistry labo-
stop nitrogenase activity. If the culture reduces acetylene, ratories, on the surface of plates of ST6CX agar (section
enrich further by a serial transfer to fresh Nfk medium. 11.6.65). Inoculate the surface of the filter papers with soil,
Examine by phase-contrast microscopy, and look for plump, rotting plant material, or drops from water samples. For soil,
curved, motile rods ca. 1 Fm wide and filled with intracel- inoculate the filter papers in a regular pattern at different
lular granules. Then, streak onto plates of Nfb medium (so- places with a few grains of soil by tamping the soil down on
lidified with 1.5% agar) containing 20 mg of yeast extract the filter with a glass rod or distributing it with a swab.
per liter. After 1 week, look for small, white, dense colonies Incubate the plates at 25 to 30C. After 4 to 5 days and pe-
and transfer to Nfb semisolid medium. For final purifica- riodically up to 1 to 2 weeks, look for glassy, translucent,
tion, streak the Nfb culture onto BMS agar (section yellow to orange spots on the paper. Make transfers as early
11.6.14). Look for the development of typical pink, often as possible from the margins of the areas of cellulose de-
wrinkled colonies. composition to fresh plates of medium with several small
Unlike Arospirillum spp., which fix nitrogen only at low pieces of filter paper. Look for slender, flexible cytophaga
oxygen tension, Azotobacter spp. have mechanisms to pro- cells by phase-contrast microscopy in the areas of celluloly-
tect oxygen-labile nitrogenase from oxygen and can fix sis. Obtaining pure cultures from enrichments can be diffi-
nitrogen aerobically. Inoculate 0.1 g of soil into 100 ml of cult; for the various methods and their limitations, see ref-
nitrogen-free Arotobacter medium (section 11.6.7) con- erence 163.
tained in a 1-liter flask. Incubate at 30C on a shaking ma-
chine. Observe the culture microscopically at periodic inter- 11.2.12.6. Agar for Agarolytic Cytophagas
vals for the development of large, ovoid cells that are 2 pm Some facultatively anaerobic marine cytophagas are able to
or more in diameter. Prepare a secondary enrichment cul- hydrolyze and ferment agar; this trait is a valuable selective
ture, and purify by obtaining isolated colonies on nitrogen- feature that can be used for their enrichment. Fill glass-
free agar medium (Azotobacter medium solidified with stoppered bottles or screw-cap tubes to the top with non-
1.5% agar). sterile Veldkamp medium (section 11.6.77). Inoculate with
See also section 11.2.12.14 for a discussion of the use of marine mud from areas with decaying algae, stopper the
unusual carbon sources for isolation of nitrogen fixers. bottles, and incubate them in the dark at 30C. Look for de-
velopment of turbidity, gas formation, and a drop in pH in
11.2.1 2.3. H2 for Aquaspirillurn autotrophicurn (1 6) 3 to 7 days. Isolate colonies by the shake tube method (sec-
Collect bacteria on the surface of membrane filters (pore tion 11.4.2) with sterile Veldkamp medium containing 2%
size, 0.45 pm) by filtering water samples taken at different agar. Look for the development of colonies which, on mi-
depths (e.g., 3 and 7 m) from eutrophic lakes. Place the fil- croscopic examination, are composed of cells that exhibit
ters, bacterium side up, onto the surface of mineral agar the flexing movements characteristic of cytophagas.
plates (section 11.6.42). Incubate under a n atmosphere of Subculture the colonies to media containing 1% agar and
60% H2, 30% air, and 10% COz at 30C. Subculture 1% yeast extract to demonstrate softening or liquefaction of
growth that develops on the filter surface to mineral agar the agar.
plates. Obtain pure cultures by repeated streaking on min- For other methods of obtaining agarolytic cytophagas,
eral agar. Microscopically, look for spirilla 0.6 to 0.8 p m see reference 163.
wide with bipolar tufts of flagella. Confirm autotrophy by
demonstrating that growth is relying on C02 as the only 11.2.12.7. Lactate for Propionibacteria
carbon source. For most fermentative bacteria, lactate is an end product of
fermentation, not a beginning substrate. The relatively un-
11.2.12.4. Methanol for Hyphomicrobia (20) usual ability of propionibacteria to ferment lactate to propi-
Members of the genus Hyphomicrobium are able to use one- onate and COZ provides a basis for selection of these anaer-
carbon compounds such as methanol or methylamine as obic organisms. Swiss-type cheeses are a good source of
sole carbon sources. Add the inoculum (5.O ml of pond or propionibacteria because these bacteria are the ripening
ditch water, or 0.3 g of mud or soil-all preferably with a agents. Fill a screw-cap culture tube (25-ml capacity) with a
low organic content) to stoppered bottles (75- to 125-ml freshly boiled and cooled medium consisting of 4% sodium
capacity), and add nonsterile Hyphomicrobium medium lactate and 1% yeast extract. Add ca. 0.2 g of CaC03 to the
(section 11.6.33) through which nitrogen has been bubbled tube, and inoculate with a small piece of Swiss-type cheese.
12
Culture Techniquest
SYED A . HASHSHAM

12.1 SOLID. SEMISOLID. MEMBRANE SURFACE.


1MMOBILIZED.CELL. AND BIPHASIC CULTURES .............. 271
12.1.1. Solid Cultures and Solidifying Agents ...................... 271
12.1.1.1.Agar ....................................... 271
.2. Carrageenan ................................. 272
.
3. Silica Gel ................................... 272
.
4. Gellan Gum ................................. 272
.
5 . Pluronic Polyol ............................... 272
.
6. Gelatin ..................................... 272
.
7. Starch ...................................... 273
.
8. Isubgol ..................................... 273
12.1.2. Semisolid Culture and Applications ........................ 273
12.1.2.1. Microaerophiles ............................... 273
.
2. Motility .................................... 273
.
3. Chemotaxis .................................. 273
12.1.3. Membrane Surface Cultures ............................. 274
.
4. Bioautography ....................................... 274
.
5 . Immobilized-Cell Cultures .............................. 274
.
6. Biphasic Cultures .................................... 274
12.2. LABORATORY SCALE LIQUID CULTURES .................... 274
12.2.1. Range of Electron Acceptors and Donors and Carbon Sources ..... 274
.
2. Batch Culture Vessels ................................. 275
12.2.2.1. Culture Tubes ................................ 275
.
2. Shake Flasks ................................. 275
12.2.3. Batch Cultures ...................................... 276
12.2.3.1. pH-Controlled Batch Culture ..................... 276
.
2. Synchronous Batch Culture ...................... 277
.
3. Fed-Batch or Sequencing Batch Culture ............. 278
12.2.4. Turbidostats ........................................ 278
.
5. Chemostats ......................................... 278
12.2.5.1. Sterilization of Liquid Media ...................... 279
.
2. Sterilization of Air ............................. 279
.
3. Sampling. Back Contamination. and Temperature
Control ..................................... 279
.4. Aerobic Operation ............................. 280
.5 . Anaerobic Operation ........................... 280
12.2.6. Special Systems ...................................... 281
12.2.6.1. Dialysis Culture .............................. 281
.2. Product Removal Culture Systems ................. 281
.3. High-Density Batch Culture Strategies .............. 282
12.3. REFERENCES ............................................ 282

The purpose of this chapter is to describe the main culture growth sought (aerobic respiration. anaerobic respiration.
techniques for microbial growth. Many techniques are or fermentative). safety issues (e.g., production of aerosols).
available including solid. semisolid. biphasic. immobilized. and experimental objectives (protein production. model-
and liquid cultures. Factors governing the choice of the ing. observation. etc.). For most biotechnology objectives
technique used for microbial cell cultures include the mass (e.g., large-scale production of antibiotics). several culture
of microbial cells or their by-products desired. the mode of techniques are combined in series to accomplish the task.
This chapter first describes the solid. semisolid. biphasic.
membrane surface. and immobilized culture techniques
(section 12.1). This is followed by laboratory scale liquid-
'This chapter is a revised and combined version of previous chapter 9 (written
by Noel R . Krieg and Philipp Gerhardt) and previous chapter 10 (written by
culture techniques including specialized liquid cultures
Philipp Gerhardt and Stephen W. Drew) in Methods for General and Molecular such as synchronous and (section 12*2)*
Bacteriology (MGMB). References for further reading are also included.

270
12. CultureTechniques rn 271

12.1. SOLID, SEMISOLID, MEMBRANE P-D-galactose] and agaropectin (the ionic, nongelatinous
SURFACE, IMMOBILIZED-CELL, portion of agar [72]). Agaropectin may contain methylated
AND BlPHASlC CULTURES or unmethylated galactoside units, sulfate, and pyruvate in
varying amounts as well as D-glucuronic acid (72). Agarose
Culture media prepared in the solid state, in the form of firm makes up about 60% of the mixture (9). Agar is extracted
gels, have been used in bacteriology since adopted by Robert from certain red marine macroalgae. When first extracted,
Koch. The most important features of solidified media stem agar is contaminated by algal cell debris and various impu-
from their ability to enable separated colonies to arise from rities, most of which must be removed before the product is
individual cells in a population diluted into or onto a solid- suitable for microbiological purposes. The key properties of
ified medium. Thus, the streak plate is a simple but effective agar that make it a versatile gelling agent for microbiologi-
technique for isolating pure cultures of bacteria and the pour cal work are the following: it is not enzymatically degraded
plate, spread plate, and layered plate are similarly valuable by most bacterial species; agar gels are stable up to 85C or
for enumerating viable bacteria. Other single-cell or single- higher, yet molten agar does not gel until cooled to ca.
colony transfer techniques that rely on solid culture are 40C; and agar gels have a high degree of transparency.
multiple-point inoculation with velveteen and the auxano- Agar is available in various commercial grades, but for most
graphic method. Solid media are also used in mass culture, culturing purposes bacteriological grade is satisfactory.
bioautography, and physiological studies of bacterial cells. Lower grades of commercial agar may contain troublesome
Semisolid, biphasic, and immobilized cultures are variations impurities, e.g., starches, fatty acids, Cu2+, or bleaching
of the solid-culture method with respect to the consistency agents that are toxic; elevated levels of Ca2+and Mg2+ that
and type of the gelling agent used. The following sections can cause precipitates; hydrogen peroxide (85); and ther-
describe each of the above, their applications, and the main moduric spores that resist the usual autoclaving procedures.
type of solidifying agents currently in use. Special, Select, Noble, and Purified grades contain
decreased levels of impurities and thus are suitable for elec-
12.1 . I . Solid Cultures and Solidifying
. - Agents
- trophoretic, nutritional, enrichment and isolation, genetic,
Solid culture is one of the most useful techniques in the iso- recombinant DNA, serological, and other special applica-
lation and cultivation from single cells. The maximum den- tions. Whenever an agar is used for a special purpose,
sity of bacterial cells is obtained by growth as a colony on a pretest it to ensure effectiveness, check with the manufac-
solid surface. If the inoculum is highly diluted, each cell has turers technical service for suitability and analysis, and if
the potential to develop into a distinct colony; such a pro- possible, use a single control lot number. Trace amounts of
cedure may be desirable even for mass culture if uniformity agar can sometimes inhibit PCR and nucleic acid sequenc-
of colony characteristics is required before harvesting of the ing (31, 103, 104).
solid culture (for example, if one wants to harvest cells only To prepare an agar-solidified medium, first adjust the pH
when the colony texture becomes smooth). More fre- of the liquid medium to the desired value and then add the
quently, the inoculum is not diluted and is spread evenly granular agar. Bacteriological or higher grades of agar do not
over the surface so that confluent solid growth results. The alter the pH of the medium appreciably. For solid medium,
solid surface usually is that of an agar or otherwise solidified use 15 to 20 g of agar per liter; for isolating highly motile
medium. However, a number of techniques have been de. organisms, even higher concentrations of agar may be re-
scribed in which colonial growth is obtained on a mem- quired. For semisolid medium, use 1 to 4 g/liter, depending
brane over a reservoir of liquid or solidified medium (see on the consistency required. Different brands and grades of
section 12.1.3). In solid culture the cells are already con- agar may require different concentrations to achieve a par-
centrated, so there is no need to use a centrifuge or other ticular degree of firmness, and the instructions of the man-
means of harvesting the cells. Solid cultures are also rela- ufacturer should always be consulted in this regard.
tively free from macro- and micromolecular compounds of After adding the agar to a liquid medium, heat the mix-
the nutrient medium. Certain cell forms, e.g., fruiting bod- ture to boiling and dissolve the agar completely. If the
ies of myxobacteria and endospores of certain Bacillus medium is being heated over a flame or on a hot plate, stir
species, are better produced on a solid medium (sometimes it constantly during heating to prevent the agar from set-
this is the only medium from which they can be obtained). tling to the bottom of the container, where it can caramelize
Solid culture, however, is limited in the extent to which it and char; then, bring the medium to a rolling boil for 1 min
can be scaled up (e.g., one can effectively produce gram or so, being careful to avoid having it foam up and over the
amounts of cells, but dekagram amounts become difficult edge of the container. Alternatively, the agar can be dis-
and hectogram or kilogram amounts are impossible to solved by placing the mixture in a steam cabinet or mi-
achieve in the laboratory). Solid cultures are also not ho- crowave oven for an appropriate period. After the agar is
mogeneous in physiological properties of the cells. For ex- melted, mix to ensure uniformity in concentration through-
ample, the cells at the top of an aerobic colony (which is out the medium. Dispense the molten medium into tubes,
likely to be 1,000 cells deep) are nutrient starved but oxy- flasks, or bottles, and sterilize in an autoclave.
gen rich, whereas the opposite situation prevails at the bot- When an agar medium with a pH of 6.0 or less is re-
tom of the colony. In addition, solid cultures often yield a quired, initially prepare and sterilize the medium at a pH
small number of cells from a given amount of medium.The greater than 6.0; otherwise, the agar will be hydrolyzed dur-
following paragraphs describe some of the commonly used ing heating and will fail to solidify adequately when the
solidifying agents. medium is later cooled. Once the medium has been steril-
ized and cooled to 45 to 50C, add sufficient sterile acid
12.1 . I . I . Agar aseptically to achieve the final pH value desired. It is useful
Agar is the most commonly used solidifying agent for mi- to prepare an extra portion of medium to experiment with
crobiological media. It mainly consists of two polysaccha- in order to determine the correct amount of acid to add to
rides, agarose [the gelling component, a repeating sequence the main batch. Alternatively, autoclave agar separately at
of (1-4)-linked 3,6 anhydro-a-L-galactose and (1-3)-linked 2X strength in water (or a neutral mixture of some of the
272 GROWTH

medium components) and mix with the autoclaved, acidic molten carrageenan media by pipetting. For gels stable dur-
main medium components when cooled. ing incubation to 45"C, 2.0% carrageenan is used; for gels
For preparing tubes of slanted medium, place the hot stable to 60"C, 2.4% carrageenan is used (47). Variability
tubes from the autoclave in a tilted position and allow them may exist between lots of carrageenan (101). Some investi-
to cool and solidify. For preparing petri dishes of medium, gators have found carrageenan to be unsuitable for semi-
first cool the molten medium to 45 to 50C in a water bath solid media, whereas others have found it to be satisfactory
and then dispense the medium aseptically into the bottom (10, 47). In contrast to agar, carrageenan may cause alter-
halves of the sterile dishes (usually 15 to 20 ml per dish), ations in the pH of some media during preparation and ster-
taking care to replace the tops after each dish is poured. ilization; therefore, determine whether and to what magni-
Cover the dishes with newspaper or other insulation to pre- tude such pH changes occur for any given kind of medium.
vent condensation of moisture underneath their tops as the It may be necessary to compensate for such changes by
agar solidifies. Stacking the poured plates also discourages preparing the medium at a different initial pH value than
condensation under the lids. desired finally or by increasing the buffering capacity of the
medium. Media containing a phosphate buffering system
Notes seem to be particularly likely to exhibit a large decrease in
1. After the agar solidifies, drops of moisture (water of PH (47).
syneresis) may form on the surface. This moisture should be
evaporated before the plates are inoculated. Usually, storage 12.1 .I .3. Silica Gel
of the dishes in an inverted position overnight at room tem- Silica gel is an inorganic solidifying agent used in media for
perature results in sufficient drying of the agar. For more solid culture of autotrophic bacteria in the complete ab-
rapid drying, invert the dishes on the shelf of a 45C gravity- sence of organic substances (25,41,62,98). Such inorganic
convection incubator or in a sterile hood and adjust the media can be supplemented with various organic com-
agar-containing halves so that they are slightly ajar. pounds to study the ability of heterotrophic bacteria to use
Incubate in this manner until the water of syneresis disap- these compounds as sole carbon sources. Vitamin require-
pears. For some procedures, such as in the use of velveteen ments can also be determined by the use of silica gel media.
replicators, dishes that have been dried more extensively Silica gel media can be prepared according to the methods
may be required. of Funk and Krulwich (25) or Sommers and Harris (86) or
2. For storage of agar dishes under conditions that pre- more recent approaches (62, 98).
vent severe drying, place them inverted in closed polyeth-
ylene bags (the bags in which plastic petri dishes are pack- 12.1 .I .4. Gellan Gum
aged make excellent storage bags for media). Agar dishes Gellan gum is an exopolysaccharide produced by the gram-
can be stored in this way at room temperature for 4 to 5 negative bacterium Sphingomonas puucimobilis (5,30,38, 73,
weeks and under refrigeration for even longer periods. 92). After processing of the native product, gellan gum has
Media containing blood or other heat-labile components relatively better clarity than bacteriological-grade agar at
should always be refrigerated. much lower concentrations (91). Gelrite and Phytogel are
3. Store all culture media, liquid and solid, in the dark. some of its trade names (91). Initial applications demon-
Storage under illumination, especially sunlight (as near a strated a clear advantage of gellan gum for culturing and
window), may result in photochemical generation of hydro- quantifying thermophilic bacteria at temperatures to 120C
gen peroxide or other toxic forms of oxygen, which can and pressures to 265 atm (26,851 kPa) (15, 46, 83). It was
render the media inhibitory for the growth of bacteria (85, also shown to be noninhibitory in PCR applications (70,
100). Microaerophiles appear to be particularly sensitive to 76). Prepare solid media solidified with gellan gum in ac-
media that have been subjected to illumination (40). cordance with the manufacturer's or other published direc-
tions or after appropriate experimentation.
12.1 .I .2. Carrageenan
Carrageenan, long used in the food and dairy industry, has 12.1 .I .5. Pluronic Polyol
also been used as a solidifying agent for microbiological Pluronic polyol, also known as reverse agar, liquefies on
media. Also known as Irish moss or vegetable gelatin, car- cooling to ca. 4C and gels into a semirigid clear jelly at
rageenan is extracted from marine macroalgae (14, 20,44). temperatures of 11 to 32C depending on the polyol con-
There are several types of carrageenan: kappa, lambda, mu, centration (27). It is a block copolymer of polypropylene
and iota (94). All are complex polysaccharides composed of oxide and ethylene oxide and is a nontoxic solidifying
several types. Kappa carageenan, for example, consists of al- agent. Its unusual solidifying characteristics have been
ternating units of mostly sulfated 3-linked D-galactosyl and used to isolate heat-sensitive bacteria and to enrich for
4-linked D-galactosyl that may also be sulfated. The potas- denitrifying, sulfate-reducing, and methanogenic microbes
sium salt of kappa carrageenan is capable of forming rigid (27). It is especially useful for isolation of fungal colonies
transparent gels, which can be an effective substitute for (71).
agar in many microbiological media (47, 101). Carrageenan
is considerably less expensive than agar. Like agar gels, car- 12.1 .I .6. Gelatin
rageenan gels can be used for streaking or spreading inocula. Gelatin was the first solidifying agent used (by Robert
Carrageenan is not degraded by most species of bacteria, Koch) for microbiological media, but it was soon replaced
and gels stable to temperatures of 60C can be prepared. by agar because gelatin is liquefied by some species and
However, carrageenan does have certain limitations. One is melts at a relatively low temperature. At the 12% concen-
the high temperature at which liquid carrageenan media tration effective for solidification, it melts at 28 to 30C.
must be dispensed into petri dishes (55 to 60"C), which Gelatin presently is used mainly as a specific hydrolysis test
may preclude its use in pour plates for enumerating certain substrate in systematics. However, mixed with Noble agar it
species and precludes its use in making media containing was found to be more suitable for the isolation of oral spiro-
blood. Because of its high viscosity, it is difficult to dispense chete colonies than agar alone (12).
12. CultureTechniques 273

12.1.1.7. Starch poured or pipetted from the Roux bottle after incubation
Starch of the type used for gel electrophoresis has been used into a sterile vessel. Sterile phosphate buffer is then added
as a solidifying agent in culture media for thermoacidophilic to the vessel, and the cells are collected by centrifugation
Archueu such as Thennoplasma, Acidianus, and Sulfolobus and washed.
species (78, 79). For Thennoplasm species, starch plates
12.1.2.2. Motility
have been reported to be superior to agar and gellan gum
plates (78). Media containing 10 to 12% (wt/vol) starch are Semisolid media can also be used to determine whether a
boiled to dissolve the polymer, poured like agar into petri bacterial strain is motile. Inoculate a tube of motility
dishes, and allowed to solidify and to dry overnight at 4C medium (broth containing 0.4% agar) with a straight nee-
(79). Starch also plays a critical role in medium formula- dle to one-half the depth of the tube. During growth, motile
tions for the isolation of various pathogens and mammalian bacteria migrate from the line of inoculation to form diffuse
microflora (37, 61, 68). turbidity in the surrounding medium; nonmotile bacteria
grow only along the line of inoculation.
12.1.1.8. lsubgoi A modification of this method is to place a piece of
Isubgol is a mucilaginous husk derived from the seeds of open-ended glass tubing into the semisolid medium prior to
Plantago owuta (36). Its use as an economical gelling agent sterilization. Inoculate only the upper part of the medium
has been reported for both plant tissue culture (3, 7) and within the glass tubing. If the organism is motile, it will mi-
microbiological growth (36, 74). It requires a higher con- grate downward, emerge from the bottom of the tubing, and
centration than does agar. then migrate upward to the surface of the medium, where it
will grow extensively. This method is also helpful for select-
12.1.2. Semisolid Culture and Applications ing highly motile organisms for use in preparing H antigens
Semisolid media contain a low concentration of gelling for immunization. However, the method is suitable only for
agent (e.g., 0.1 to 0.4% agar) and have a soft, jellylike con- facultative organisms capable of metabolism under anaero-
sistency. Such media are useful for cultivating mi- bic conditions, such as members of the family Entero-
croaerophilic bacteria and for studying various aspects of bucteriuceue. Strictly aerobic organisms such as Pseudomom
motility and chemotaxis. These applications are briefly de- species do not migrate downward to the bottom of the glass
scribed below. tubing. Strategies to measure the rate of motility, e.g., by at-
taching latex particles to the cell, are also available (52).
12.1.2.1. Microaerophiles The subject of motility has been reviewed by Harshey (32).
Microaerophilic bacteria must be grown under low levels of
oxygen. They cannot grow under an atmosphere containing 12.1.2.3. Chemotaxis
high levels of oxygen, e.g., 21% oxygen present in air. Campy- Semisolid media are also useful in chemotaxis studies. For
lobucter jejuni, a microaerophile, grows best in a 6% oxygen at- example, a semisolid medium containing an oxidizable car-
mosphere. Helicobucter pylori living in mammalian or avian bon and energy source can be used to investigate positive
gut is also a microaerophile (39). Certain nitrogen-fixing bac- chemotaxis in Escherichiu coli (2), Vibrio cholerue (8),
teria that have a respiratory type of metabolism may grow as SalmoneUa entericu (87), and many other species. It is one of
aerobes when supplied with a source of fixed nitrogen (such as the most extensively studied subjects with reviews (2, 49,
ammonium sulfate) but can grow only as microaerophiles in 93), methods of measurements (60), and models (88).
nitrogen-deficient media. For example, Azospirillum species When a petri dish containing the medium is inoculated
grow best in a 1% oxygen atmosphere under nitrogen-fixing heavily at its center, the bacteria migrate outward as an ex-
conditions; the nitrogenase complex would be inactivated by panding ring of cells that consumes all of the oxidizable sub-
excess oxygen. The general subject of microaerophily has strate as the cells move. When two oxidizable carbon
been reviewed by Krieg and Hofhnan (40). sources are provided in the medium, two rings which mi-
The key factor for growing microaerophiles is the avail- grate at different rates are formed. In contrast to capillary-
ability of low concentrations of 02,as opposed to com- tube methods for studying chemotaxis, the oxygen supply
pletely aerobic conditions or total lack of oxygen. To obtain on the agar plate is never exhausted; thus, the migrating
a low concentration of 02, microaerophiles may sometimes rings form only in response to self-created substance gradi-
be grown deeper in a solid medium because oxygen diffusion ents and not in response to self-created oxygen gradients.
is limited under such conditions. Diffusivity of oxygen Negative chemotaxis also has been studied by using
through agar is re orted to be in the range of 7.60 X lo- semisolid media (95). E. coli cells are suspended in the
to 2.45 X mys-I at 30C (53). Thus, if tubes of semi- medium at a concentration sufficient to give visible turbid-
solid media are stored for long periods, oxygen may eventu- ity, and the inoculated medium is dispensed into a petri
ally diffuse deeply into the media. Placing such tubes in a dish. After the medium gels, a plug of hard agar (2% agar)
boiling-water bath for several minutes may help drive off containing the suspected repellent compound is inserted
the dissolved oxygen. into the medium. If the compound is a repellent, the sur-
The microaerophile C. jejuni can be easily cultivated in rounding bacteria migrate away from the plug and leave a
tubes of semisolid brucella medium (brucella broth contain- clear zone that becomes visible in ca. 30 min.
ing 0.15 to 0.3% agar). The nitrogemfixing Arospirillum The use of plates of semisolid media also makes it possi-
species grow well in a semisolid nitrogen-deficient malate ble to select nonchemotactic mutants (95). In the case of
medium, and large crops of these cells for physiological positive chemotaxis, nonchemotactic mutants (and also
studies can be obtained by using 1-liter Roux bottles con- nonmotile mutants) fail to respond to a self-created sub-
taining 150 ml of culture medium stratified with 0.05% agar strate gradient and remain near the center of the petri plate,
(59). This medium forms an 8-mm-deep layer on the wide, where they can be removed for subculturing and purifica-
flat bottom of the Roux bottle. The agar concentration is tion. In the case of negative chemotaxis, the nonchemotac-
high enough to allow the microaerophile to grow under aer- tic mutants can be isolated from the clear zone surrounding
obic conditions but low enough to permit the culture to be the hard agar plug.
274 GROWTH

12.1.3. Membrane Surface Cultures ble to any type of bacterium. Biphasic cultures are found to
Solid culture of bacteria can also be accomplished on the be more efficient in growing many pathogens (6,80) in-
surface of a dialysis or microfiltration membrane in contact cluding Mycobmterium (1) and Arcobacter spp. (17). To pre-
with an underlying reservoir of liquid or solidified medium. pare such a system, partially fill the container with hot
The principles and development of such colonial growth medium containing 2 to 3% agar. After the agar is solidi-
with membranes have been reviewed from time to time (26, fied, overlay it aseptically with a small volume of broth, in-
43, 66,77). This technique is particularly useful for viable oculate, and incubate. If the bacteria are aerobic, clamp the
counts of dilute concentrations of cells in air or water after container on a mechanical shaker to provide aeration and
membrane filtration, for studying conjugational transfer of agitation of the broth during incubation. The culture is
plasmids (97), and for studying microbial interactions (57). confined to the liquid overlay but has diffusional access to
the reservoir of nutrients in the solidified base, and conse-
12.1.4. Bioautography quently the culture becomes densely concentrated. Since
A n example of membrane surface culture is bioautography. the movement of nutrients is dependent on diffusion, the
It is a version of paper chromatography in which the growth agar base should be limited to about 5 cm in depth. The
of bacteria is used as a highly sensitive indicator for locating ratio of solid to liquid should be at least 4:land less than
the positions of certain compounds on a paper chro- 1O:l; the lower ratio provides a better yield, and the higher
matogram. The method has the advantage of specifically ratio provides a greater concentration of cells. A n
detecting compounds with biological activity, which chem- Erlenmeyer flask is a convenient container, but indenta-
ical or radioisotopic detection systems lack. The method is tions at its base are helpful to hold the agar in place if a me-
particularly applicable to locating the position of growth chanical shaker is used; rectangular containers also are use-
factors on chromatograms of spent culture media or cell ex- ful in this way. Also, use a greater percentage of agar if
tracts when the concentration of the growth factors is so breakup occurs during shaking.
low as to preclude the use of ordinary detection systems. For If all of the medium components are incorporated into
example, the location of as little as 5 to 10 ng of folic acid the agar base, the overlay can be distilled water. After an
on a chromatogram can be determined by use of bioautog overnight equilibration period, the resulting clear diffusate
raphy. For a good example of bioautography applied to the in the overlay can be inoculated. For bacteria such as gono-
detection of growth factors in cell extracts, see reference 84. cocci, which normally must be grown in a turbid medium
Bioautography is also widely used by the pharmaceutical in- enriched with blood and starch and which yield sparse pop-
dustry for the detection of antibiotic agents on paper chro- ulations, a relatively clean and dense population of cells can
matograms (69,102). For bioautography of antibiotic be harvested from such a diffusate overlay (28).A variety of
agents, the agar medium should be nutritionally complete bacteria have been preserved in the short term by subcul-
to allow the indicator organism to grow throughout the dish turing in a biphasic medium.
except at regions of the chromatogram where the antibiotic
substance is located. More recent approaches have disks 12.2. LABORATORY SCALE
with multiple antibiotics that are easier to handle and use.
LIQUID CULTURES
12.1.5. ImmobilizedCell Cultures Laboratory scale liquid cultures provide one of the most
Immobilization of cells is important in many areas including common techniques to grow and study the behavior of mi-
maximization of product formation, minimization of cell croorganisms. Containers for such cultures vary from simple
mass, water distribution systems, groundwater remediation, culture tubes to large carboys and reactors. The setup de-
and medical devices. Product maximization by immobilized pends on the need to maintain sterility, sampling, electron
cells has been practiced for many decades; e.g., in the acceptor conditions, substrate addition and product re-
quick vinegar process, acetic acid bacteria are immobi- moval, monitoring, and mixing regimes. The following sec-
lized as a colony film on a column of wood shavings, tions describe the basic types of systems used for liquid cul-
through which an ethanol solution is trickled downward tures both in batch and continuous systems. It also describes
while air is passed upward, yielding an acetic acid solution. some of the special rypes of liquid cultures that require dial-
Once the bacterial film is established, little further growth ysis bags or large animal hosts.
occurs and bacterial maintenance energy is derived from
the oxidation of ethanol to acetate. The process can be op- 12.2.1. Range of Electron Acceptors
erated continuously over periods of weeks or months. and Donors and Carbon Sources
Similar examples exist for the production of kojic acid, In all cases of microbial growth, it is important to ensure
using Aspergillus oryzae NRRL484 (99), or laccase from that the electron acceptor, electron donor, carbon source,
Coriolus versicolor IF04937, using more sophisticated mem- and trace element requirements are met as per the needs of
brane surface liquid reactors (34).Immobilized cell culture the particular type of microorganism. The section on ener-
technology has been reviewed for various applications (see getics and stoichiometry of chapter 13 describes the theo-
reference 58 and other articles in the same issue). retical aspects of microbial growth focusing on the electron
Laboratory scale studies often employ model surfaces and acceptor and donor and the carbon and nitrogen sources. In
reactors that mimic the relevant environmental surface addition, phosphorus and trace elements must also be added
characteristics and conditions and allow intrusive or non- in sufficient quantities. Phosphorus requirements are gener-
destructive observation of the immobilized cells. ally met by adding one-fifth of the amount of nitrogen re-
quired using the stoichiometric equation (see chapter
12.1.6. Biphasic Cultures 13.1.10). Requirements for media preparation and trace el-
Biphasic culture consists of a thick layer of solidified nutri- ements vary considerably among the range of microorgan-
ent medium overlaid with a thin layer of nutrient broth isms known today. For example, the range of electron ac-
(96). Populations in excess of 10lcells per ml can be ob- ceptors for Shewanella oneidensis MR-1 and Shewanella
tained in this way, and the technique is apparently applica- puwefuciens strain 200 includes 0 2 , N03-, NOz-, NO,
12. CultureTechniques 275

Fe3+, Mn4+, Mn3+, S032-, S2032-, S, dimethyl sulfoxide, machine to induce a vortex. Also increase the availability
arsenate, chromate, vanadate, neptunium, fumarate, sele- of air by using a small and loosely packed cotton plug, or a
nium, and technitium with the capability to use several or- plastic or stainless-steel cap.
ganic acids, sugars, and hydrogen as electron donors. Table
1 lists a number of common electron acceptors and donors 12.2.2.1.2. Anaerobic
for some of the common types of microorganisms. Estab- The use of thick-walled culture tubes (Fig. 1 [Bellco
lishing the media constituents for growing a given type of Biotechnology, Inc., Vineland, NJ]) is now common for
microorganism, especially if it is a relatively less studied or- many types of anaerobic applications ranging from isolation
ganism, should begin by a good literature search and evalu- of dehalorespiring microorganisms to methanogenic activ-
ation with respect to the objective. ity measurements to demonstration of photoautotrophic ac-
tivity (4). Larger-volume serum bottles (up to 1,000 ml)
12.2.2. Batch Culture Vessels may also be employed for ease of use and larger biomass and
sample needs. The use of anaerobe culture tubes and serum
12.2.2.1. Culture Tubes bottles requires (i) proper sealing by special butyl rubber
stoppers that can be punctured without causing leaks, (ii)
12.2.2.1.1. Aerobic purging of the headspace with appropriate oxygen-free
The lipless Pyrex glass culture tube (usually 16 by 150 gases, and (iii) proper sealing of the tubes or bottles with
mm), plugged with nonabsorbent cotton or plastic foam, is aluminum crimp caps. Laboratories engaged in anaerobic
the most convenient and widely used container for batch culture systems routinely assemble a system of gas cylinders,
liquid culture of bacteria. However, for aerobic cultivation, regulators, gassing and sparging stations, and cannulas for
test tubes usually provide only minimally effective condi- the preparation of media and sampling needs.
tions of oxygen supply. Fortunately, many bacteria are fac-
ultatively anaerobic and most microbiological uses of cul- 12.2.2.2. Shake Flasks
ture tubes do not require optimum growth conditions. To
improve oxygen supply, increase the surface-to-volume ratio 12.2.2.2.1. Aerobic
of the liquid medium by reducing the volume and slanting An Erlenmeyer flask (capacity, 100 to 2,000 ml) is com-
the tubes, or preferably, mount them on a rotary shaking monly used as a container for producing masses of cells in

TABLE 1 Electron acceptors and donors and carbon sources for selected microorganisms
Electron acceptor Electron donor Carbon source Examples Applications or context

0 2 Organic compounds Organic compounds Heterotrophs Wastewater treament,


remediation, many others
co2 Nitrosomonas NH4+oxidation to NO*-
co2 Nitrobucter NO2- oxidation to NO3-
co2 Thiobacilli Concrete corrosion
co2 HI-oxidizing bacteria NO3- removal (because
they can grow on NO3-)
0 2 Fe2+ co2 Thiobucillus ferrooxidans Iron oxidation
0 2 CH4 CH4 Methanotrophs TCE" remediation
NO3- Organic compounds, Organic compounds, Denitrifiers NO3- removal
some can use H2 co2
HS-/H2S, S co2 Thiobacillus deniwificans NO3- removal
HS-/H2S co2 Strain MLMS-1 Arsenic removal
Organic compounds, Organic compounds, Shewanella sp., Bacillus Selenium nanosphere
H2 co2 selenitireducens production
Lactate, acetate, H2 Organic compounds Sulfate-reducingbacteria Sewer, anaerobic processes
Fe3+ Acetate Acetate Desulfuromonas Acid mine drainage
Mn4+ Acetate Acetate Desulfuromonas
co2 H2 co2 Methanogens, acetogens Anaerobic digestion
Organic Organic compounds Organic compounds Clostridia, lactic acid Fermentation, probiotics
compounds bacteria
NO2- NH4+ Anaerobic ammonia Anammox process (NH4+
oxidizers and NO2-removal)
PCE~~TCE Acetate, formate, H2 Acetate, COZ Dehulococcoides Remediation
Cr6+ Lactate Lactate Shewunella Remediation
U6+ Organic compounds, Organic compounds, Geobacter sp., Uranium mobility
H2 co2 Shewanella sp.
"TCE, trichloroethene.
'PCE, tetrachloroethene.
276 GROWTH

- Aluminum crimD caD in the side and/or bottom surface, which act to break vortex
formation in flasks on a rotary shaking machine and to in-
duce turbulence. Baffled flasks with good design of the baf-
fles are available commercially (e.g., Wheaton Science
Products).

12.2.2.2.2. Anaerobic
Flasks or bottles of any other convenient shape are used
to cultivate anaerobic bacteria on an intermediate size
FIGURE 1 Thick glass culture tube with butyl rubber stop- scale. Two main principles prevail: the nutrient medium
pers and aluminum crimp caps useful in isolation and culturing must be prereduced to an Eh of - 150 mV or lower, and air
of anaerobic microorganisms. must be removed from the immediate environment of the
culture. Strict anaerobes require special precautions to re-
move all traces of oxygen. These are described more fully
elsewhere in this manual.
the laboratory or as the first stage in a scale-up study of an A few practical tips apply to mass cultivation of anaer-
industrial process involving batch liquid culture of bacteria. obes. Fill the container nearly full to minimize the effect of
Consequently, maximally effective conditions of air (oxy- the overlying gas phase. Use as large an inoculum as possi-
gen) supply are sought for obligately or facultatively anaer- ble, distributed from the bottom upward in a column of pre-
obic types of bacteria. Aerobic flask culture must be carried reduced medium. Displace the gas phase with oxygen-free
out with shaking of the flask to facilitate mass transfer of nitrogen or argon, plus 5 to 10% COZ, and allow growth to
oxygen (as well as other gases and nutrients) at two levels: begin with the medium quiescent. Once growth has pro-
from the gas phase across the gas-liquid interface into the gressed actively throughout the medium, provide agitation
liquid phase, and from the liquid phase across the liquid-cell by means of a magnetic stirring bar or by purging nitrogen
interface into the cell. Two major factors influence the abil- or an inert gas (e.g., helium or argon) through a tube with
ity of flask culture techniques to meet the oxygen require- its outlet at the bottom of the container. Use a water trap to
ments of the cells: gas exchange through the flask closure, permit the escape of introduced and metabolic gases with-
and the liquid surface area available for oxygen transport out the entrance of air. Caution: some anaerobic bacteria
from the gas phase. produce hydrogen or methane gas, which may cause flam-
The closure design for culture flasks must allow adequate mable or even explosive conditions, and most anaerobes
gas exchange between the external environment and the produce COz and other gases, which will increase the in-
flask interior, yet must maintain asepsis. Morton-type caps ternal pressure dangerously if adequate venting is not pro-
provide an adequate aseptic seal and good gas exchange, but vided.
they occasionally spin off a flask at high rotational speed.
Foam plastic plugs also are effective aseptic closures for 12.2.3. Batch Cultures
shake flask cultivation and provide moderate gas exchange, Growing microorganisms in batch culture vessels is one of
but the plugs must be sized to fit into the neck of the flask the most common techniques employed in microbiology
snugly yet without severe compression. Furthermore, foam and biotechnology. In batch cultures, substrate concentra-
plugs may contain plasticizers or other toxic chemicals, tion decreases from an initial value that is supplied to a final
which may require thorough washing of the plugs prior to concentration remaining after the growth has slowed down
their use as closures for biological culture. significantly. Concentration of biomass or cell increases
The rate at which dissolved oxygen is consumed in a liq- from an initial value that is determined by the inoculum
uid culture of bacteria is determined by the cell density and size and liquid volume to a final concentration after growth.
growth rate. The demand for dissolved oxygen is created by The volumes of batch cultures may vary from a few milli-
cell consumption and is met by continuous diffusion of oxy- liters in small test tubes to thousands of liters in very large
gen from the gas phase to the liquid phase and thence into fermentors. Batch cultures invariably serve as an intermedi-
the cell. The interfacial surface area between gas and liquid ate step between growth as a colony on a plate to small
often controls the flux or volumetric rate of oxygen transfer. flasks to carboys to large fermentors. It should be noted that
A large interfacial surface area results in high volumetric handling and manipulating large glass carboys during
rates of oxygen transfer and therefore allows rapid growth medium preparation, sterilization, and inoculation are haz-
without oxygen limitation. The interfacial surface area can ardous, and shaking of filled glass carboys for aerobic culti-
be maximized by maintaining low ratios of liquid volume to vation is risky, although the use of polypropylene or poly-
flask volume and by vigorously shaking the flasks. Oxygen carbonate plastic carboys lessens these concerns. Gas
limitation due to limited diffusion can usually be avoided by dispersion in any carboy is poor, even with stirrers, so that
preparing flasks with liquid volumes of no more than 20% adequate aeration is unlikely. Although preparation of
of flask volumes and by shaking the flasks at 200 to 350 rpm media in plastic carboys is sometimes necessary for large-
on a rotary shaker or at 150 to 250 strokes per min on a re- scale work, cultivation in carboys should be avoided.
ciprocating shaker. During incubation of the inoculated The following subsections describe some of the special
flasks at an appropriate temperature, rotary shaking should types of batch cultures that require better control of one or
cause the liquid meniscus to rise to approximately two- more parameters related to substrate characteristics, mi-
thirds of the flask height. Reciprocal shaking, such as is croorganism growth stages, or feeding patterns.
found in most water bath shakers, should be sufficiently vig-
orous to cause significant breaking (turbulence) of the liq- 12.2.3.1. pH-Controlled Batch Culture
uid wave as it moves from side to side in the flask, but not Controlled batch culture requires instrumentation that
so great as to wet the closure (24). Oxygen transfer is fur- monitors a n environmental parameter and triggers an addi-
ther enhanced by baffling. Baffled flasks have indentations tion to the fermentor so that the indicator parameter is
12. Culture Techniques 277

maintained at a steady value. A n example of environmen- will result in cell fractions in which the most dense fraction
tal parameter control in a fermentation system is the main- contains both young (daughter) cells and mature (ready-to-
tenance of pH at a constant value by the automatic addi- divide) cells, while the least dense fraction will contain a
tion of acid or base. The basic components of a pH control homogeneous population of cells that have progressed
system are the pH electrode(s) with its shielded connector through a common fraction of their cell cycles.
cable, the pH meter, an endpoint titrator, an acid or base Selection synchrony through density gradient centrifu-
reservoir with flexible tubing for connection to the fermen- gation cannot supply new daughter cells for direct study of
tor, and a single peristaltic pump or solenoid valve. All cell cycleelinked physiology, because these fractions will al-
components of the fermentor system, including pH and dis- ways be contaminated with mature cells ready to divide.
solved-oxygen electrodes, must be designed for steam steril- However, density gradient centrifugation can supply an
ization or be compatible with other sterilization methods, adequate inoculum for synchronous culture growth. Cell
such as ethylene oxide or formaldehyde-steam sparging. cycle-linked physiology may then be studied by direct Sam-
The pH meter and pH titrator must be interconnected and pling of the synchronous culture. The procedure described
therefore should be purchased from the same manufacturer below is presented as a general guideline for development of
to ensure equipment match. Although a peristaltic pump synchronous cultures and is based on the technique de-
can allow foolproof addition of acid or base against rela- scribed by Mitchison and Vincent (54). This technique may
tively high fermentor back-pressure, a simple tubing-pinch be used as an initial guide for development of a synchronous
solenoid valve for control of gravity feed of acid or base to cultivation technique specifically designed for the organism
the fermentor is usually sufficient for laboratory scale oper- in use (48).
ation. Fermentor manufacturers usually provide their own
Synchronization Procedure
pH control systems.
Delay synchronization experiments until reproducible
12.2.3.2. Synchronous Batch Culture batch cultivation conditions can be established, including
determination of asynchronous culture kinetics. When
Culture synchrony occurs when all of the cells divide at
these prerequisites are met, establish batch cultivation con-
nearly the same instant, thus mimicking individual cell
ditions so that two to five cell mass doublings occur during
growth. For synchronous cultures, a plot of the logarithm of
logarithmic growth.
cell numbers versus time resembles a stair step rather than a
straight line. Biochemical events associated with specific 1. Prepare 500 ml of sterilized medium in several culture
times in the individual cell cycle can be systematically stud- vessels (250-ml Erlenmeyer flasks are convenient).
ied in a synchronous culture. Although development of cul- Inoculate one half of the flasks, and incubate these under
ture synchrony is relatively straightforward, maintenance of appropriate conditions. Store the remaining sterile flasks
synchrony over long periods is a difficult task requiring pre- under identical conditions.
cise control. 2. Harvest the cells from the batch culture at the mid-
Culture synchrony can be achieved by periodically vary- exponential growth phase. Rapidly cool the culture to 0 to
ing a critical environmental condition. The technique 4C by swirling the flasks in an ice bath. Harvest the cells
forces the synchronization of cell multiplication by inter- by refrigerated centrifugation at 10,000 X g for 10 min.
rupting, promoting, or retarding metabolic function in a 3. Resuspend the sedimented cells in 2 ml of appropriate
cyclic manner. The population will gradually synchronize ice-cold buffer (0.1 M potassium phosphate; pH 7.0) by vig-
its response to these periodic disturbances of metabolic ac- orous agitation with a Vortex mixer. If the cells tend to ag-
tivity and will ultimately synchronize its growth pattern. gregate, use mild sonication or mild homogenization with a
The technique, however, requires severe disturbance of nor- blender or tissue grinder to prepare a suspension of discrete
mal metabolic activity and is therefore of limited use in the cells. For cultures that are particularly difficult to suspend,
study of growth cycle-linked bacterial physiology. One ex- use 0.01% (wtlvol) Tween 80 in the suspending buffer be-
ception to this limitation is bacterial spore germination as a fore mechanical or sonic treatment.
means of initiating culture synchrony. Step or pulse changes 4. Rapidly, but carefully, layer the suspended cells on a
in the culture environment or addition of a specific chemi- sterile, precooled density gradient prepared from Ficoll, su-
cal germinant can trigger spore germination and may crose, Percoll, or another appropriate medium as dictated by
closely model natural occurrences. the cell system. For example, to prepare an exponential su-
A generally useful method of synchronization is based on crose gradient in a discontinuous manner, layer sterile ice-
physical selection of a homogeneous fraction from a hetero- cold solutions of increasing sucrose concentration into pre-
geneous population of vegetative cells (21). This approach sterilized, precooled ( O O C ) centrifuge tubes. Place 10 ml of
is often termed selection synchrony and avoids most of the 35% (wtlvol) sucrose in phosphate buffer into the bottom
problems of metabolic disturbance during synchronization of a sterile centrifuge tube. Sequentially layer 10 ml each
by physically selecting cells that are in similar states of the of sucrose-buffer solutions containing 26.5, 25.5,24.5, 22.0,
cell growth cycle. 19.0, and 15.0% (wtlvol), respectively, onto the 35% sue
Kubitschek (42) and Poole (65) showed that the cell crose cushion (the buffers may all be prepared from the 35%
volume of E. coli increases linearly during the cell cycle sucrose stock solution). Filter sterilize the sucrose solutions
while its cell mass increases exponentially. The observation prior to use in forming the gradient. All solutions must be
that cell volume is lowest just after cell division suggests ice-cold and preaerated (for aerobic cells).
that centrifugation in a density gradient might allow recov- 5. Carefully centrifuge the cell-charged sealed centrifuge
ery of a population of new daughter cells from an asynchro- tubes in a precooled centrifuge at 2,500 x gfor 15 to 20 min
nous culture. However, the observation that the volume in- at 0C. For more precise separations, adjust the time and
crease is linear while the mass increase is exponential speed of centrifugation so that the optically dense band of
means that cells which are just ready to divide or cells bacteria moves no more than two-thirds of the way down
which have just divided will have the greatest cell density the centrifuge tube; speeds and times vary somewhat de.
despite cell size differences. Density gradient centrifugation pending on the culture being handled.
278 GROWTH

6. Inoculate prewarmed flasks of growth medium with by wiping the surface, but the many designs for this have
0.5 ml directly from the lightest-density fraction of cells. proved cumbersome and prone to failure.
7. Carefully monitor the optical density of the newly in-
oculated culture flasks for a definite stepwise increase in op- 12.2.5. Chemostats
tical density, an indication of synchronous growth. Chemostats are completely mixed reactors with inflow of
liquid containing the substrate and nutrients at a certain
Culture synchrony should be maintained for two to
rate (volume per unit of time) and outflow of liquid at the
three cycles. A study over longer periods requires the
same rate containing a much lower concentration of sub-
reestablishment of synchrony through the procedure de-
strate and nutrient left over after use, and by-products and
scribed above. Decay of synchrony occurs beyond a few cy-
biomass produced from the biochemical reaction. Mixing in
cles and should be carefully monitored and documented.
a chemostat is critical to minimize physical and chemical
concentration gradients and population heterogeneity.
12.2.3.3. Fed-Batch or Sequencing Batch Culture Hence, they are sometimes referred to as continuous stirred
In a batch culture, the transition from exponential growth tank reactors. Mixing or agitation is achieved by mechani-
to stationary phase may occur for a variety of reasons, in- cal means or by gas sparging. Inadequate agitation allows
cluding the depletion of an essential nutrient substrate or the development of regions of poor mixing (dead spots)
the buildup of a toxic metabolite product. When the tran- and results in uncontrolled local concentration gradients.
sition results from nutrient depletion, growth will con- The physiological result of poor mixing is a highly hetero-
tinue if fresh medium is added. The medium addition rate geneous cell population. For example, inadequate agitation
and the culture volume must be increased exponentially to of aerobic cultures can result in insufficient contact be-
maintain a constant rate of exponential growth. This tween the gas phase and the liquid medium. As a result,
technique for exponential growth maintenance (18,45) is some regions of the reactor or vessel may be anaerobic. If
usually called fed-batch culture. Periodic removal of cul- the organism is facultative, the cell population will have a
ture volume to allow additional feeding is called extended- wide range of respiratory characteristics and the cell yield
batch culture or repeated fed-batch culture. True fed- will be reduced. The operational volume of chemostats typ-
batch operation requires that the volume of the liquid ically ranges from a few milliliters to thousands of liters,
medium in the fermentor increase during the fermenta- with typical laboratory units having operating volumes of 1
tion. This requirement places an upper limit on the cul- to 20 liters. Aerobic chemostats require lines for aeration,
ture time based on feed rate and leads to changing con- feeding, sampling, and a mechanism to mix the liquid.
ditions of aeration and agitation effectiveness. T h e Often both aeration and mechanical mixing are practiced
fed-batch mode allows substantial improvements in cell to avoid foam and dead space. Anaerobic chemostats need
mass or product productivity over an ordinary batch oper- a water break at the effluent line to avoid exposure to air
ation. The technique of fed-batch culture may also be used (Fig. 2). To maintain sterility and growth in the feed, the
to supply large quantities of a potentially toxic substrate nutrient (nitrogen, phosphorus, and trace elements) line is
while maintaining a low concentration of the substrate in often separated from the main line supplying the fer-
the medium. mentable substrate and buffer. If intermittent feeding
and/or withdrawal is needed, the system could be automated
12.2.4. Turbidostats with the help of timers and automatic controllers.
In a turbidostat, cell concentration (biomass) is monitored Although it is possible to build a chemostat in a machine
and maintained at a constant level by adjusting the feed shop, the necessary attention to detail of design for mixing,
rate of fresh nutrients (50, 55). The substrate need not be aeration, sterilization, and maintenance of asepsis warrants
present in a limiting amount but, rather, is usually present the purchase of commercially available units. Chemostats
in excess. The system may be operated over a wide range of with better control may also be the best option for more so-
biomass concentrations near the critical dilution rate as phisticated studies related to, say, replication, gene expres-
long as all medium components are in excess. However, it is sion patterns, and stress response. Commercial fermentors
most stable when the specific growth rate of the culture is are capable of adequate aeration and agitation of relatively
near the maximum value, pmax, for the particular medium dense cultures and are usually equipped with a multiport
in use. The term turbidostat includes any technique that head plate through which pH probes, dissolved-oxygen
holds the cell concentration constant and includes moni- probes, and sampling or feed lines can be introduced into
toring techniques based on cellular metabolism. Since cer- the fermentor. They can be quite expensive depending on
tain metabolic functions (such as oxygen uptake, carbon the number of vessels, volume, probes, and desired controls.
dioxide evolution, and, in some cases, pH change) are inti- Alternatives to commercial fermentors are also abun-
mately linked to cell growth rate and ultimately to the spe- dant and are in more common use. Chemostats of sizes vary-
cific growth rate, these parameters may be used as control ing from a few milliliters to tens of liters can be built in-
variables for medium replacement. house by the appropriate choice of vessel, mixing and
The precision with which the biomass can be controlled aeration devices, peristaltic or syringe pumps for feeding
has historically been the weakest aspect of turbidostat culti- and withdrawal, monitoring probes, temperature control
vation. Most of the older methods of monitoring cell den- systems, sampling ports, and controls and timers for auto-
sity rely on optical monitoring with some type of photo- mated operation. Laboratory-built systems are generally less
electric sensor measuring scattered or transmitted light. sophisticated (unless the focus of building such a system is
These methods suffer from interference by bubbles and to push the limits of the existing commercially available
foam produced during aeration, wall growth on the fermen- systems) and may require somewhat more time for opera-
tor and, even more critically, the optical device. Bubble in- tion and maintenance. The main drawback of commercial
terference can be eliminated by using an external flow- units, however, is their high cost, requiring their purchase as
through optical cell. However, foaming remains a problem. specialized items instead of as routine laboratory equip-
Wall growth on optical surfaces can be partially remedied ment. Irrespective of the type of equipment or system used,
12. Culture Techniques 279

Nutrients +NaHCO,
I-+Gas

1
4 Liquid seal

Effluent
FIGURE 2 Schematic of an anaerobic continuous stirred tank reactor with separate pumps for
supplying substrate and nutrients, effluent pump with water seal, and gas collection line. For smaller
systems with multiple reactors, syringe pumps with multiple ports can be used.

the operation of a chemostat requires consideration of the Standard procedures for the sterilization of liquid media
following parameters in some form. are described elsewhere in this manual. Filter sterilization
Preparation and operation of the chemostat require that may be necessary to prevent destruction of heat-labile
all points of entry to or withdrawal from the system be de- medium components. However, most simple media for cul-
signed for aseptic operation to prevent contamination. tivation of bacteria can be autoclaved prior to use, but
Most chemostats have a sample and inoculation line that larger volumes of medium require longer autoclaving times.
penetrates the chemostat head plate and extends to within Vessels containing approximately 10 liters of liquid should
a few centimeters of the bottom of the vessel. Note that be autoclaved at 121C for 30 to 90 min, depending on
positive pressure on the fermentor must always be main- medium constituents. Media containing solids such as corn-
tained. Chemostat continuous cultivation is always pre- meal flour or soy flour may require up to 90 min for com-
ceded by transient batch cultivation, during which time the plete sterilization of a 10-liter volume. However, media
cell mass accumulates at the expense of the substrate. Start containing only dissolved components are usually sterile
the continuous culture while the batch cultivation is in the after 30 min at 121C. All vessels should be vented during
exponential phase of growth; this minimizes oscillations autoclaving to allow equilibration of pressure. The medium
due to nutritional step-up and avoids inadvertent washout volume in culture reservoirs should not exceed 75% of the
due to physiological lag. Start the dilution at a rate less than total reservoir volume, to minimize the chance of boil-over
the desired operational dilution rate, and then increase to during autoclaving. Furthermore, it is essential that the gas
the operational dilution rate within one residence time, the space in the reservoir be vented during autoclaving.
average time that a cell remains in the vessel. This mini-
mizes oscillations from toxic substrates. Chemostat response 12.2.5.2. Sterilization of Air
to toxic substrates is discussed in more detail by Pirt (63). The air or gas supply to sparged chemostats must be steril-
ized prior to injection into the vessel. This is most easily
12.2.5.1. Sterilization of Liquid Media done by physical removal of airborne microorganisms with
Continuous cultivation requires the use of rather large a fibrous-medium filter. The filtration medium must be
quantities of medium and produces equally large volumes of changed regularly to ensure adequate performance. A sup-
product culture. For daily maintenance, it is advisable to plementary or backup air sterilization filter may be needed
prepare enough medium for 20 h of operation to allow flex- for long-duration cultivation as a precaution against fouling
ibility in the scheduling of reservoir transfer. Since most of the duty filter.
laboratory scale operations require autoclaving of the
medium as the means of sterilization, the required volumes 12.2.5.3. Sampling, Back Contamination,
of medium will probably be most conveniently prepared in and Temperature Control
carboys of appropriate size. (Caution: exercise extreme care Sampling is best accomplished with an independent sample
in handling large volumes of hot liquids. Use autoclavable line system. Alternatively, samples may be taken from the
plastic carboys rather than glass ones.) For example, a 1- effluent line, although this procedure is not recommended
liter liquid working volume chemostat maintained so that when a small sample is needed, since the lumen of the ef-
the bacterial culture doubles every hour (dilution rate, 0.69 fluent tube may become coated with adherent bacteria
h-) would require 13.8 liters of fresh medium per 20-h in- which may break away and result in nonrepresentative Sam-
terval. Small volumes of medium may be easily autoclaved ples. If an external sampling system is used in place of Sam-
and stored in standard 20-liter plastic carboys. The prepara- ple collection from the effluent line, the sample size must be
tion of medium in small volumes (10 to 14 liters) allows ad- kept below 5% of the reactor working volume so that
equate heat sterilization without excessive deterioration. steady-state conditions will not be disturbed greatly. Finally,
The size of the chemostat to be operated is dictated by the the sample line must be purged of its entire contents before
need for sampling, cell mass, number of replicates or exper- samples are collected for analysis. Some chemostats are
imental reactors needed, and operational flexibility. equipped with a sterile air purge which displaces the sample
280 GROWTH

line contents back into the fermentor after a sample is control of both agitation and airflow rate in a sequential
taken. If the latter system is used, purging prior to sample manner.
collection is not necessary. Most well-aerated and agitated cultures produce rela-
The aeration and agitation conditions of continuous cul- tively stable foam at some point in the culture cycle. If
ture result in the production of an aerosol containing large foams are allowed to develop unchecked, they may wet the
numbers of the cultured bacterium. As a result, the medium air filters and lead to back contamination of the culture as
feed line is subject to back contamination and must be fit- well as to foam spillage. Foams may be controlled by me-
ted with a medium "break" tube, which acts as an aseptic chanical foam breakers or by the addition of chemical an-
seal between the culture vessel and the medium reservoir. tifoam agents. Many commercial units are equipped with
Most commercial chemostats for laboratory-scale opera- mechanical foam breakers as standard or optional equip-
tion are equipped with integral heat exchangers. The tem- ment. Current designs for mechanical foam breakers are ad-
perature is controlled by a temperature control unit to equate for all but a very few culture conditions.
maintain a constant temperature inside the vessel. In some Chemical antifoam agents provide a less expensive
cases, the vessel is jacketed or submerged in a constant- means of foam control when intermittent or light use is ex-
temperature bath as a means of temperature control when pected (11). However, antifoam agents must be added to
accuracy is required. the medium and therefore contribute to the overall medium
composition. Some antifoam agents such as vegetable (corn
12.2.5.4. Aerobic Operation oil or cottonseed oil) and animal (lard oil) ones may be me-
The agitation speed for adequate oxygen transfer in a tabolized by the culture and therefore contribute to the car-
chemostat is subject to change as the culture density (cell bon substrate pool. O n the other hand, nonmetabolizable
concentration) or operating conditions (volumetric airflow antifoam agents, such as the silicone antifoams, may be
rate, temperature, and liquid volume) change. The operator toxic at high concentrations. However, their action as sur-
should adjust the agitation speed and airflow rate so that factant materials requires their use in only very low con-
the dissolved-oxygen concentration never falls below 30% centrations. Effectiveness and toxicity should be deter-
of the initial saturated dissolved oxygen concentration mined on an individual basis. Antifoam agents can be
(82). Maintenance of dissolved oxygen concentrations added directly to the medium before sterilization or to the
above 30% of the saturation value at ambient temperatures fermentation system through a feed line in the head plate
allows growth of most bacterial cultures under conditions in (the antifoam agent, its reservoir, and the feed line must be
which oxygen is the growth-limiting substrate and keeps sterilized prior to use). Automatic control units for antifoam
aerobic cells growing exponentially. addition are usually available commercially.
Some basic properties of oxygen supply and demand
must be kept in mind when providing for adequate aeration 12.2.5.5. Anaerobic Operation
in the cultivation of aerobic bacteria whether in a fermen- The primary concern in system design for anaerobic culti-
tor, a shake flask, or a test tube. Oxygen is quite insoluble; vation in chemostats is the exclusion of oxygen. Use a large
for example, there are only about 9 ppm (0.0009%) in water inoculum (10 to 20% of total operating volume) to allow
at 20C in equilibrium with air. As the temperature is rapid establishment of the culture and to reduce the sensi-
raised, oxygen becomes even less soluble. Oxygen solubility tivity of the system to leaks of oxygen from the external en-
is directly proportional to the partial pressure of oxygen in vironment. If pH is to be controlled or nutrients are to be
the gas phase but is substantially independent of the total added in a continuous or semicontinuous fashion, keep the
pressure and the presence of other gases. Bacteria utilize flexible tubing connecting the reservoirs to the vessel as
only dissolved (not gaseous) oxygen. Oxygen is so insoluble short as possible, since oxygen permeates natural or silicone
that only a small reservoir of it exists in solution at any rubber tubing; use butyl rubber instead. Tygon tubing is also
given time. Consequently the rate of dissolved-oxygen sup- relatively oxygen impermeable but softens during autoclav-
ply must at least equal the rate of oxygen demand of the cul- ing, so it must be clamped or wired in place. Finally, fit the
ture. Fortunately, cell respiration proceeds at a rate that is gas exit of the fermentor with a water-gas trap to prevent
independent of the dissolved-oxygen concentration as long back diffusion of oxygen into the fermentor head space.
as it remains above a critical concentration, which is con- In all cases, prereduce the medium by adding a reducing
siderably below the saturation value. These basic properties agent and sparge the vessel with oxygen-free gas prior to in-
of oxygen supply and demand are further developed and ex- troducing the prereduced medium. Prepare and maintain
emplified in a classic review on aeration and agitation in acid, base, or nutrient solutions under oxygen-deficient
microbial culture by Finn (23) and in another excellent dis- conditions for addition to the vessel during operation.
cussion in the monograph by Pirt (63). Absolute exclusion of oxygen from acid, base, or other so-
Agitation requirements for adequate oxygen transfer are lutions to be added to the fermentor in small quantities is
almost always well in excess of the agitation requirements not necessary since the reducing characteristics of an active
for mixing of highly soluble nutrients. This rule also applies anaerobic culture adequately cope with very slight additions
to agitation conditions established for optimum dissolution of oxygen through the feed systems.
of other sparingly soluble substrates, such as immiscible hy- For the cultivation of anaerobic microbes, take special
drocarbons and steroids. For oxygen transfer, both agitation care to ensure establishment and maintenance of the chem-
speed and volumetric airflow rate can be varied to establish ically reduced state when autoclaving large volumes of pre-
the desired oxygen concentration in the fermentation reduced medium. Connect vessels containing prereduced
medium. Instrumentation for dissolved-oxygen monitoring media to an oxygen-free gas supply immediately upon re-
and for control of airflow rate is useful for the cultivation of moval from the autoclave. As the steam in the gas head-
aerobic organisms and allows the operator to control dis- space of the vessel begins to condense, a partial vacuum will
solved oxygen concentration as an independent parameter form. Sparging with oxygen-free gas allows release of the
( 19). Dissolved-oxygen control systems are available from vacuum formed during cooling without contamination of
all of the major fermentor manufacturers and usually allow the medium by oxygen in the air.
12. Culture Techniques w 281

12.2.6. Special Systems culture and dialysate (interface dialysis culture), such as in
a liquid/solid biophasic system.
12.2.6.1. Dialysis Culture
Dialysis is a process for separation of solute molecules by 12.2.6.1.1. In Vitro Systems
means of their unequal diffusion through a semipermeable Dialysis culture is often first attempted in the laboratory
membrane as a result of a concentration gradient (77, 89). by suspending a membrane sac containing the culture in a
The process is applied to the growth and maintenance of tube, flask, or carboy of medium. The simplest such arrange-
living cells by a technique called dialysis (or diffusion or ment is to use a length of dialysis membrane tubing that is
perfusion) culture. To use the technique of dialysis culture, intussuscepted so as to form a double-walled tube contain-
a membrane is positioned between a culture chamber and a ing culture in the annular space thus formed. The advan-
dialysate reservoir. For dialysis culture to be effective, the tages of shake flasks are combined with those of membrane
volume of the reservoir must be larger than that of the cul- dialysis culture in a unit assembled from flanged Pyrex glass
ture or else the reservoir must be replenishable. Further, the pipe. This and other flanged flask designs (89) enable the
permeability and area of the membrane must be sufficient to use of sheet membrane of any type. Scale-up of dialysis cul-
permit useful diffusion in rate and amount. Such a system of ture to fermentors is possible by carrying out growth in a
dialysis culture can be operated in vitro or in vivo and separate culture circuit that is connected with a separate
batchwise, continuously, or a combination of these modes. dialysate circuit by means of an intermediate dialyzer (26).
Specific advantages and reasons for using dialysis for Various types of hollow-fiber dialyzers have also been used
bacterial culture include (i) prolongation of the exponen- (67). The design of the system can be optimized for a given
tial growth phase in the batch cycle, which allows the at- culture situation. Stieber et al. (90) have developed and re-
tainment of very high densities of viable cells; (ii) extension viewed various modes of operation of an in vitro system.
of the maximum stationary phase in batch culture, thus per-
mitting increased production of secondary metabolites asso- 12.2.6.1.2. Animal Systems
ciated with this phase; (iii) relief of product inhibition con- The term diffusion chamber is often used to describe a
trol by removal of metabolite products, thus enabling their small dialysis culture unit that can be implanted within a
greater production in batch or continuous culture; (iv) es- living experimental animal (e.g., in the peritoneum or
tablishment of a steady-state population with mainly main- rumen or beneath the skin). Among a number of systems,
tenance metabolism, thus immobilizing the cells for pro- the best are made with a nondegradable filter membrane
longed production of a metabolite product; (v) production sealed on both sides of an inert plastic cylinder (like a
of metabolites free from cells and, conversely, production of drum) through which sampling access can be provided.
cells free from medium macromolecules; (vi) means to study Bacteria introduced into diluent within the chamber grow
and recover a cell population placed in an in situ or in vivo entirely on diffusible nutrients from the host. Such systems
environment, such as in an ecological or animal system; and have been used in bacteriology to study immune reactions
(vii) the capability for study of molecular interactions be- and the growth of fastidious pathogens, e.g., Mycobacterium
tween separated populations of cells. leprae, Treponema pallidurn, and Neisseria gonorrhoem.
There are two basic principles underlying dialysis cul- Although not common, examples do exist whereby a
ture. First, it provides a means for achieving substrate- microbial growth system is designed to be in contact with
limited growth, i.e., fed-batch culture. Substrate in the the bloodstream of a live animal (67). Such an ex vivo he-
dialysate reservoir diffuses through the membrane into the modialysis culture system enables the bacteria to grow en-
culture chamber, driven by a concentration gradient that tirely on the nutrients of the blood, yet separate from the
results as the substrate is used for metabolism and growth. macromolecular and cellular defense mechanisms, if a dial-
Second, dialysis culture provides a means for lowering the ysis type of membrane of appropriate molecular exclusion is
concentration of a diffusible metabolite product inhibitory used. The host-parasite reactions of many facultatively aer-
to growth; the product in the culture chamber diffuses obic (but no obligately anaerobic) bacteria were studied in
through the membrane and is diluted in the larger dialysate this manner (29).
reservoir, thus relieving the feedback inhibition by the
product that normally regulates its production. In the usual 12.2.6.2. Product Removal Culture Systems
dialysis culture system, nutrient supply and product with- The product removal and consequent cell-concentrating
drawal occur concurrently; i.e., exchange dialysis occurs. feature of dialysis culture can be accomplished much more
Three main types of membranes, differing in porosity, are efficiently by other systems involving membranes in which
applicable to dialysis culture. Dialysis membranes have a the driving force is greater than a concentration gradient,
nominal pore size on the order of 10 nm in diameter so that such as a pressure gradient for microfiltration and an elec-
they exclude cells and macromolecules but allow small mol- trical gradient for electrodialysis. However, these systems
ecules, such as the nutrients for bacterial growth, to pass. are more complicated in design, tend to foul the membrane
Filter membranes (or membrane filters) have a nominal more than dialysis does, and have specialized applicability
porosity on the order of 100 nm, so they also exclude cells, beyond the scope of this book. A n example of a continuous
but they allow macromolecules as well as small molecules to microfiltration culture system is given in reference 56, and
pass. Sheets of membrane are commercially available in a an example of an electrodialysis system is given in reference
wide range of porosities and materials. Factors to consider 33. Product removal and relief of product feedback inhibi-
for microbiological use include autoclavability, pore size, in- tion of bacterial growth can also be attained by a variety of
ertness, and permeability (which is governed by porosity, other means. These include extraction by use of nonaque-
void space, and thickness). Solution transport membranes ous solvents, biphasic aqueous systems, and catalytic mem-
have n o pores and pass gases because of their solubility in branes; sorption by solvents; vacuum evaporation and mem-
the membrane material itself (e.g., oxygen solubility in sili- brane pervaporation; and precipitation. These extractive
cone rubber or polycarbonate). A n interface between two culture systems are reviewed in a publication edited by
physically different phases may also be used to separate the Mattiasson and Holst (51).
282 w GROWTH

12.2.6.3. High-Density Batch Culture Strategies nate toxic-product formation, e.g., acetic acid by E. coli.
Ordinary liquid-culture methods produce relatively low The system must be monitored with a glucose or acetate
densities of bacterial cell mass. Usual maximum growth probe and controlled by a computer (to < 2 g of glucose per
densities of aerobic or facultatively aerobic bacteria in liter or <1.5 g of acetate per liter for E. coli).
batch systems are on the order of 0.1 g (dry weight) of cell Removing a toxic metabolic product as it is formed is
per liter in quiescent test tubes. Compared to this, shake another common strategy for attaining a high density of
flasks and aerated and agitated fermentors can achieve 10 bacterial cells. Although essential for anaerobes, product re-
and 100 times more biomass, respectively. Cell counts and moval alone should usually be a secondary strategy for aer-
other direct indices of growth increase similarly by decades. obes if bacterial biomass production is the objective: it is
Maximum densities of anaerobic bacteria are generally better to limit product formation than to correct it. In dial-
about 1 decade less than those of aerobes. ysis culture, product removal is coupled with nutrient sup-
Further large increases to still-higher cell densities can ply; this combination strategy is especially applicable for
be attained by strategically using one or more of the critical small-scale laboratory use by use of the biphasic shake flask
limiting factors in batch growth discussed earlier in this system.
chapter. High-density culture (concentrated culture) strate- Maintenance of pH at an optimum level is necessary for
gies for aerobic bacteria have resulted in concentrations as high-density culture and has become commonplace in fer-
high as 150 g (dry weight) of cells/liter (26), but the maxi- mentor systems with provision for a pH probe and either
mum practical concentration is limited by oxygen transfer manual or computer control by the addition of ammonium
capability to about 70 g (dry weight) of cells/liter. In- hydroxide, which also maintains the nitrogen supply neces-
creasing the cell density makes more sense than making sary for high-density culture.
replicates or increasing the vessel volume. Optimization of Control of temperature is also usual in fermentor sys-
a process for high cell density is desirable whether the pro- tems. For high-density culture, it is crucial not to exceed the
cess is for bacterial biomass production, metabolic-product optimum temperature, beyond which growth rates fall off
formation, or substrate consumption. precipitously. Indeed, a lower growth temperature (for E.
The key principle underlying high-density culture is to coli, 22C) has been used to lower the growth rate and so to
prevent the depletion of an essential nutrient and relieve extend the exponential phase (81), resulting in about a
the feedback inhibition of bacterial growth by limiting the doubled yield. A practical compromise between these con-
accumulation of toxic metabolic products. This is done by siderations is to use a moderately lower growth temperature,
(i) supplying essential nutrients in increasing amounts to e.g., 30C for E. coli.
meet but not exceed growth needs, (ii) removing toxic
products as they are formed, and (iii) controlling physico- 12.2.6.3.2. Anaerobes
chemical growth factors at optimum levels. These are best High-density cultures of anaerobic organisms are most
accomplished in a fermentor system and are usually applied common in two broad areas: production of probiotics (e.g.,
to aerobic bacteria, but they are also applicable to anaer- lactic acid bacteria [35, 751) and environmental biotech-
obes. A summary review of the theoretical and historical nology (e.g., anaerobic granules containing more than 10"
background of high-density culture was presented by cellslgram of granular sludge [16]). In addition, most high-
Shiloach et al. (82). density anaerobic cultures require specialized operation of
the reactor itself in addition to the removal of any in-
12.2.6.3.1. Aerobes hibitory products that may be produced. Membrane-based
High-density culture of aerobes is best exemplified with cell recycle systems (13), upflow anaerobic sludge blanket re-
E. coli and other enteric bacteria, which oxidize glucose and actors (64), and numerous attached-growth or immobilized-
other carbon and energy sources largely to COz via aerobic cell technologies are examples of increasing the cell density
respiration in the presence of an adequate oxygen supply, by reactor operation and modifications.
but incompletely to organic acids, particularly acetic acid,
via fermentation in the presence of an inadequate oxygen
supply. It is this accumulation of acetic acid by fermenta- 12.3. REFERENCES
tion as the result of oxygen depletion that mainly inhibits 1. Abe, C. 1997. Rapid diagnosis of tuberculosis. Kekkaku
growth. 72:659-672. (In Japanese.)
To keep aerobic cells growing exponentially in a fermen- 2. Adler, J. 1966. Chemotaxis in bacteria. Science 153:708-
tor, the dissolved-oxygen supply should be increased as 716.
growth increases and kept above 30% saturation; for a cul- 3 . Babbar, S. B., and N. Jain. 1998. 'Isubgol' as an alterna-
ture of 10 g (dry weight) of cells/liter, this amounts to 300 tive gelling agent in plant tissue culture media. Plant Cell
mmol of oxygen per liter per 30 min (82). This enormous Rep.-l7:3i8-322. -
oxygen demand can only partially be met by maximizing the 4. Balch, W. E., and R. S. Wolfe. 1976. New approach -- to
airflow and agitation rates in a fermentor. To further increase the cultivation of methanogenic bacteria: 2-mercap-
the dissolved-oxygen concentration, oxygen-enriched air toethanesulfonic acid (HS-CoM)-dependent growth of
Methanobacterium ruminantium in a pressurized atmos-
must be provided by adding pure oxygen from a liquid-oxygen phere. Appl. Enuiron. Microbiol. 32:781-791.
cylinder. Dissolved-oxygen concentration is monitored with 5. Banik, R. M., B. Kanari, and S. N. Upadhyay. 2000.
a probe (19). The increases in the oxygen-enriched airflow Exopolysaccharide of the gellan family: prospects and po-
and agitation rates corresponding to the increase in growth tential. World J. Microbiol. Biotechnol. 16:407-414.
are best controlled by a computer system (22). 6. Bannur, M., R. P. Fule, A. M. Saoji, and V. L.
Increasing the addition of essential nutrients as expo- Jahagirdar. 1995. Study of bacteraemia using conventional
nential growth progresses (fed-batch culture) is also usually and biphasic culture methods. Indian J. Pathol. Microbiol.
essential and a primary consideration for high-density cul- 38: 147-15 1.
ture of aerobes. Glucose (or another carbon and energy 7. Bhattacharya, P., S. Dey, and B. C. Bhattacharyya. 1994.
source) must usually be maintained at a low level to elimi- Use of low-cost gelling agents and support matrices for
12. Culture Techniques 283

industrial-scaleplant-tissue culture. Plant Cell Tissue Organ 29. Gerhardt, P., J. M. Quarles, T. C. Beaman, and R. C.
Cult. 37:15-23. Belding. 1977. Ex vivo hemodialysis culture of microbial
8. Boin, M. A,, M. J. Austin, and C. C. Hase. 2004. and mammalian cells. J. Infect. Dis.13542-50.
Chemotaxis in Vibno cholerae. FEMS Microbiol. Lett. 239: 30. Giavasis, I., L. M. Harvey, and B. McNeil. 2000. Gellan
1-8. gum. Crit. Rev. Biotechnol. 20:177-211.
9. Bridson, E. Y., and A. Brecker. 1970. Design and formu- 31. Gibb, A. P., and S. Wong. 1998. Inhibition of PCR by
lation of culture media, p. 229-295. In J. R. Norris and agar from bacteriologicaltransport media. J. Clin. Microbiol.
D. W. Ribbons (ed.), Methods in Microbiology, vol. 3A. 36: 275-276.
Academic Press, Inc., New York, NY. 32. Harshey, R. M. 2003. Bacterial motility on a surface:
10. Bromke, B. J., and M. Furiga. 1991. Carrageenan is a de- many ways to a common goal. Annu. Rev. Microbiol. 57:
sirable substitute for agar in media growing Trichomonus 249-2 73.
vaginalis. J. Microbiol. Methods 13:61-65. 33. Hongo, M., Y. Nomura, and M. Iwahara. 1986. Novel
11. Bryant, J. 1970. Anti foam agents, p. 187-203. In J. R. method of lactic acid production by electrodialysis fer-
Norris and D. W. Ribbons (ed.), Methods in Microbiology, mentation. Appl. Environ. Microbiol. 52:3 14-3 19.
vol. 2. Academic Press, Inc., New York, NY. 34. Hoshino, K., M. Yuzuriha, S. Morohashi, S. Kagaya, and
12. Chan, E. C. S., A. DeCiccio, R. McLaughlin, A. M. Taniguchi. 2002. Production of laccase by membrane.
Klitorinos, and R. Siboo. 1997. An inexpensive solid surface liquid culture with nonwoven fabric of Coriolus
medium for obtaining colony-forming units of oral spiro- versicolor. Biol. Syst. Eng. 830:108-120.
chetes. Oral Microbiol. Immunol. 12:372-376. 35. Ishizaki, A. 2003. Advanced continuous fermentation for
13. Chang, H. N., I. K. Yoo, and B. S. Kim. 1994. High- anaerobic microorganism. Ferment. Biotechnol. 862:2 1-35.
density cell-culture by membrane-based cell recycle. 36. Jain, N., S. Gupta, and S. B. Babbar. 1997. Isubgol as an
Biotechnol. Adv. 12467-487. alternative gelling agent for microbial culture media.
14. Chiovitti, A., G. T. Kraft, A. Bacic, D. J. Craik, S. L. A. J. Plant Biochem. Biotechnol. 6:129-131.
Munro, and M. L. Liao. 1998. Carrageenans from 37. Jenkins, J. A., and P. W. Taylor. 1995. An alternative
Australian representatives of the family Cystocloniaceae bacteriological medium for the isolation of Aeromonus spp.
(Gigartinales, Rhodophyta), with description of Calli- J. Wildl. Dis. 31:272-275.
blepharis celatospora sp. nov., and transfer of Austro- 38. ]in, H., N. K. Lee, M. K. Shin, S. K. Kim, D. L. Kaplan,
clonium to the family Areschougiaceae.J. Phycol. 34:5 15- and J. W. Lee. 2003. Production of gellan gum by
535. Sphingomm paucimobilis NK2000 with soybean pomace.
15. Deming, J. W., and J. A. Baross. 1986. Solid medium for Biochem. Eng. J. 16:357-360.
culturing black smoker bacteria at temperatures to 120C. 39. Kelly, D. J. 2001. The physiology and metabolism of
Appl. Environ. Microbiol. 5 1:238-243. Campylobacter jejuni and Helicobacter pylori. J. Appl.
16. Diaz, E., R. Amils, and J. Sanz. 2003. Molecular ecology Microbiol. 90: 16s-24s.
of anaerobic granular sludge grown at different conditions. 40. Krieg, N. R., and P. S. Hoffman. 1986. Microaerophily
Water Sci. Technol. 48:57-64. and oxygen toxicity. Annu. Rev. Microbiol. 40:107-130.
17. Dickson, J. S., T. R. Manke, I. V. Wesley, and A. L. 41. Kriukov, V. R. 1981. Development of hydrogen bacteria
Baetz. 1996. Biphasic culture of Arcobacter spp. Lett. Appl. on hard surfaces. Mikrobiologiia 50:299-304.
Microbiol. 22:195-198. 42. Kubitschek, H. E. 1987. Buoyant density variation during
18. Dunn, I. J., and J. R. Mor. 1975. Variable volume con- the cell cycle in microorganisms. Crit. Rev. Microbiol.
tinuous culture. Biotechnol. Bioeng. 17: 1805-1822. 14: 73-97.
19. Elsworth, R. 1972. The value and use of dissolved oxygen 43. Landwall, P., and T. Holme. 1977. Removal of inhibitors
measurement in deep culture. Chem. Eng. 29363-71. of bacterial growth by dialysis culture. J. Gen. Microbiol.
20. Epifanio, E. C., R. L. Veroy, F. Uyenco, G. J. B. Cajipe, 103:345-352.
and E. C. Laserna. 1981. Carrageenan from Eucheuma 44. Laserna, E. C., F. Uyenco, E. Epifanio, R. L. Veroy, and
striatum (Schmitz) in bacteriological media. Appl. Environ. G. J. B. Cajipe. 1981. Carrageenan from Eucheuma stria-
Microbiol. 41: 155-158. tum (Schmitz) in media for fungal and yeast cultures. Appl.
21. Evans, J. B. 1975. Preparation of synchronous cultures of Environ. Microbiol. 42:174-175.
Escherichia coli by continuous flow size selection. J. Gen. 45. Lim, H. C., B. J. Chen, and C. C. Creagan. 1977. An
Microbiol. 9 1:188-1 90. analysis of extended and exponentially fed batch cultures.
22. Fass, R., T. R. Clem, and J. Shiloach. 1989. Use of a Biotechnol. Bioeng. 19425433.
novel air separation system in a fed batch fermentative 46. Lin, C. C., and J. L. E. Casida. 1984. Gelrite as a gelling
culture of Escherichia coli. Appl. Environ. Microbiol. agent in media for growth of thermophilic microorgan-
55:1305-1307. isms. Appl. Environ. Microbiol. 47:427-429.
23. Finn, R. K. 1954. Agitation aeration in the laboratory and 47. Lines, A. D. 1977. Value of the K+ salt of carrageenan as
in industry. Bacteriol. Rev. 18:254-274. an agar substitute in routine bacteriological media. Appl.
24. Freedman, D. 1970. The shaker in bioengineering, p. 175- Environ. Microbiol. 34637-639.
185. In J. R. Norris and D. W. Ribbons (ed.), Methods in 48. Lloyd, D. L., J. C. Edwards, and A. H. Chagla. 1975.
Microbiology, vol. 2. Academic Press, Inc., New York, NY. Synchronous cultures of micro organisms: large scale
25. Funk, H. B., and T. A. Krulwich. 1964. Preparation of preparation by continuous flow size selection. J. Gen.
clear silica gels that can be streaked.J. Bacteriol. 88:1200- Microbiol. 88:153-158.
1201. 49. Lux, R., and W. Shi. 2004. Chemotaxis-guided move-
26. Gallup, D. M., and P. Gerhardt. 1963. Dialysis fermentor ments in bacteria. Crit. Rev. Oral. Biol. Med. 15207-
systems for concentrated culture of microorganisms.Appl. 220.
Microbiol. 11:506-512. 50. Markx, G. H., C. L. Davey, and D. B. Kell. 1991. The
27. Gardener, S., and J. G. Jones. 1984. A new solidifying permittistat: a novel type of turbidostat. J. Gen. Microbiol.
agent for culture media which liquefies on cooling. J. Gen. 137:735-743.
.~~ . .-
Microbiol . 130:73 1-733. 51. Mattiasson, B., and 0. Holst (ed.). 1991. Extractive
28. Gerhardt, P., and C. G. Hedn. 1960. Concentrated cul- Bioconoersions. Marcel Dekker, Inc., New York, NY.
ture of gonococci in clear liquid medium. Proc. SOC.Exp. 52. McBride, M. J. 2004. Cytophuga fivobacterium gliding
Biol. Med. 105:49-51. motility. J. Mol. Microbiol. Biotechnol. 7:63-71.
284 a GROWTH

53. Miller, C. W., M. H. Nguyen, M. Rooney, and K. 74. Sahay, S. 1999. The use of psyllium (isubgol) as an alter-
Kailasapathy. 2003. Novel apparatus to measure diffusion native gelling agent for microbial culture media. World
in gel type foods. Food Australia 9:432-435. J. Microbiol. Biotechnol. 15:733-735.
54. Mitchison, J. W., and W. S. Vincent. 1965. Preparation of 75. Schiraldi, C., V. Adduci, V. Valli, C. Maresca, M.
synchronous cell cultures by sedimentation. Nature Giuliano, M. Lamberti, M. Carteni, and M. De Rosa.
205:987-989. 2003. High cell density cultivation of probiotics and lactic
55. Munson, R. J. 1970. Turbidostats, p. 349-376. In J. R. acid production. Biotechnol. Bioeng. 82:213-222.
Norris and D. W. Ribbons (ed.), Methods in Microbiology, 76. Schmidt, D., and P. M. Rath. 2003. Faster genetic identi-
vol. 2. Academic Press, Inc., New York, NY. fication of medically important aspergilli by using gellan
56. Nipkow, A., J. G. Zeikus, and P. Gerhardt. 1989. gum as gelling agent in mycological media. J. Med.
Microfiltration cell recycle pilot system for continuous Microbiol. 52:653-655.
thermoanaerobic production of exoamylase. Biotechnol. 77. Schultz, J. S., and P. Gerhardt. 1969. Dialysis culture of
Bioeng. 34:1075-1084. microorganisms: design, theory, and results. Bucteriol. Rev.
57. Nordbring Hertz, B., M. Veenhuis, and W. Harder. 33:147.
1984. Dialysis membrane technique for ultrastructural 78. Segerer, A. H., and K. 0. Stetter. 1992. The genus
studies of microbial interactions. Appl. Environ. Microbiol. Thermoplasma, p. 712-718. In A. Balows, H. G.Thiper,
47: 195-197. M. Dworkin, W. Harder, and K. H. Schleifer (ed.), The
58. Norton, S., and J. C. Vuillemard. 1994. Food bioconver- Prokaryotes. A Handbook on the Biology of Bacteria:
sions and metabolite production using immobilized cell Ecophysiology, Isolation, Identification, Applications, 2nd ed.
technology. Crit. Rev. Biotechnol. 14: 193-224. Springer Verlag KG, Berlin, Germany.
59. Okon, Y., S. L. Albrecht, and R. H. Burris. 1976. 79. Segerer, A. H., and K. 0. Stetter. 1992. The order
Carbon and ammonia metabolism of Spirillum lipoferum. Sulfolobales, p. 684-701. In A. Balows, H. G. Thiper, M.
.IBucteriol.
. 128592-597. Dworkin, W. Harder, and K. H. Schleifer (ed.), The
60. Olson, M. S., R. M. Ford, J. A. Smith, and E. J. Prokaryotes. A Handbook on the Biology of Bacteria:
Fernandez. 2004. Quantification of bacterial chemotaxis Ecophysiology, Isolation, Identification, Applications, 2nd ed.
in porous media using magnetic resonance imaging. Springer Verlag KG, Berlin, Germany.
Environ. Sci. Technol. 38:3864-3870. 80. Shadowen, R. D., and C. V. Sciortino. 1989. Improved
61. Papapetropoulou, M., G. Rodopoulou, and E. growth of Campylobacterpylori in a biphasic system. J. Clin.
Giannoulaki. 1995. Improved glutaminate-starch-peni- Microbiol. 27: 1744-1 747.
cillin agar for the isolation and enumeration of Aeromonas 81. Shiloach, J., and S. Bauer. 1975. High yield growth of E.
hydrophila from seawater by membrane filtration. Pathol. coli at different temperatures in a bench scale fermentor.
Biol. (Paris) 43:622-627. Biotechnol. Bioeng. 17:227-239.
62. Parkinson, S. M., M. Wainwright, and K. Killham. 1989. 82. Shiloach, J., M. V. d. Walle, J. B. Kaufman, and R. Fass.
Observations on oligotrophic growth of fungi on silica-gel. 1991. High density growth of microorganisms for protein
Mycol. Res. 93529-534. production, p. 3 3 4 6 . In M. D. White, S. Reuveny, and
63. Pirt, J. S. 1975. Principles of Microbe and Cell Cultivation. A. Shafferman (ed.), Biologicals from Recombinant
John Wiley & Sons, Inc., New York, NY. Microorganisms and Animal Cells. VCH Publishers, New
64. Pol, L. W. H., S. I. D. Lopes, G. Lettinga, and P. N. L. York, NY.
Lens. 2004. Anaerobic sludge granulation. Water Res. 83. Shungu, D., M. Valiant, V. Tutlane, E. Weinberg, B.
38: 1376-13 89. Weissberger, L. Koupal, H. Gadebusch, and E. Stapley.
65. Poole, R. K. 1977. Fluctuations in buoyant density during 1983. Gelrite as an agar substitute in bacteriological
the cell cycle of Escherichia coli K12: significance for the media. Appl. Enuiron. Microbiol. 46:840-845.
preparation of synchronous cultures by age selection. 84. Sirotnak, F. M., G. J. Donati, and D. J. Hutchison. 1963.
J. Gen. Microbiol. 98:177-186. Folic acid derivatives synthesized during growth of
66. Portner, R., and I. H. Mark. 1998. Dialysis cultures. Appl. Diplococcus pneumonim. J. Bucteriol. 85:658-665.
Microbiol. Biotechnol. 5 0:403-4 14. 85. Sneath, P. H. A. 1955. Failure of Chromobacterium uio-
67. Quarles, J. M., R. C. Belding, T. C. Beaman, and P. laceum to grow on nutrient agar, attributed to hydrogen
Gerhardt. 1974. Hemodialysis culture of Serratia peroxide. J. Gen. Microbiol. 13:i.
marcescens in a goat artificial kidney fermentor system. 86. Sommers, L. E., and R. F. Harris. 1968. Routine prepara-
Infect. Immun. 9650-558. tion of silica gel media using silicate solutions of varying
68. Rabe, L. K., and S. L. Hillier. 2003. Optimization of pH. J.Bacteriol. 95:1174.
media for detection of hydrogen peroxide production by 87. Stecher, B., S. Hapfelmeier, C. Muller, M. Kremer, T.
Lactobacillus species. J. Clin. Microbiol. 41:3260-3264. Stallmach, and W. D. Hardt. 2004. Flagella and chemo-
69. Ramirez, A., R. Gutierrez, G. Diaz, C. Gonzalez, N. taxis are required for efficient induction of Salmonella
Perez, S. Vega, and M. Noa. 2003. High-performance enterica serovar Typhimurium colitis in streptomycin-
thin-layer chromatography-bioautography for multiple an- pretreated mice. Infect. Immun. 72:41384150.
tibiotic residues in cows milk. J. Chromatogr. B Anal. 88. Stelling, J. 2004. Mathematical models in microbial sys-
Technol. Biomed. Life Sci. 784:315-322. tems biology. Cum. Opin. Microbiol. 7513-518.
70. Rath, P. M., and D. Schmidt. 2001. Gellan gum as a suit- 89. Stieber, R. W. 1979. Dialysis Continuous Processes for
able gelling agent in microbiological media for PCR appli- Microbial Fermentations: Mathematical Models, Com-
cations. J. Med. Microbiol. 50:108-109. puter Simulations, and Experimental Tests. Ph.D. thesis.
71. Reeslev, M., and A. Kjoller. 1995. Comparison of biomass Michigan State University, East Lansing.
dry weights and radial growth-rates of fungal colonies on 90. Stieber, R. W., and P. Gerhardt. 1979. Dialysis continu-
media solidified with different gelling compounds. Appl. ous process for ammonium lactate fermentation: improved
Environ . Microbiol. 6 1:4236-423 9. mathematical model and use of deproteinized whey. Appl.
72. Roehrig, K. L. 1984. Carbohydrate Biochemistry and Environ. Microbiol. 37:487495.
Metabolism. The AVI Publishing Company, Westport, CT. 91. Sutherland, I. W. 1999. Microbial polysaccharide prod-
73. Rule, P. L., and A. D. Alexander. 1986. Gellan gum as a ucts. Biotechnol. Genet. Eng. Rev. 16:217-229.
substitute for agar in leptospiral media. J. Clin. Microbiol. 92. Sworn, G., G. R. Sanderson, and W. Gibson. 1995.
23500-504. Gellan gum fluid gels. Food Hydrocolloids 9:265-271.
12. CultureTechniques 285

93. Szurmant, H., and G. W. Ordal. 2004. Diversity in apparatus for membrane-surface liquid culture and pro-
chemotaxis mechanisms among the bacteria and archaea. duction of kojic acid using Aspergillus oryzue NRRL484.
Microbiol. Mot. Biol. Rev. 68:301-319. 1. Ferment. Bioeng. 85488494.
94. Towle, G. A., and R. L. Whistler. 1973. Hemicellulose 100. Waterworth, P. M. 1969. The action of light on culture
and gums, p. 198-248. In L. R. Miller (ed.), Phytochem- media. J. Clin. Pathol. 22:273-277.
istry, vol. 1. Van Nostrand Reinhold Co., New York, NY. 101. Watson, N., and D. Apirion. 1976. Substitute for agar in
95. Tso, W., and J. Adler. 1974. Negative chemotaxis in solid media for common usages in microbiology. Appl.
Escherichia coli. j . Bacteriol. 118:560-576. Enoiron. Microbiol. 31509-513.
96. Tyrrell, E. A., R. E. MacDonald, and P.Gerhardt. 1958. 102. Weinstein, M. J., and G. H. Wagman (ed.). 1978.
Biphasic system for growing bacteria in concentrated cul- Antibiotics. Isolation, Separation and Purification. Elsevier
ture. J. Bacteriol. 751-4. Scientific Publishing Co., New York, NY.
97. VanElsas, J. D., J. M. Govaert, and J. A. v. Veen. 1987. 103. Yamaguchi, Y., S. Nimbari, H. Obata, T. Ookawara,
Transfer of plasmid pFT30 between bacilli in soil as influ- H. Eguchi, T. Kurotsu, and K. Suzuki. 2002. Effects
enced by bacteria population dynamics and soil condi- of agarose and LB medium on dye-terminator DNA
tions. Soil Biol. Biochem. 19:639-647. sequencing. Yakugaku Zasshi-J. Pharm. SOC.Jpn. 122:
98. Wainwright, M.,and A. Al-Talhi. 1999. Selective isola- 495-498.
tion and oligotrophic growth of Candida on nutrient-free 104. Yamaguchi, Y., S. Nimbari, T. Ookawara, K. Oishi,
silica gel medium. 1. Med. Microbiol. 48:1130. H. Eguchi, and K. Suzuki. 2002. Inhibitory effects of
99. Wakisaka, Y., T. Segawa, K. Imamura, T. Sakiyama, agarose gel and LB medium on DNA sequencing.
and K. Nakanishi. 1998. Development of a cylindrical BioTechniques 33:2%2.
Energetics, Stoichiometry, and Kinetics of Microbial
G rowtht
SYED A. HASHSHAM AND SAM W. BAUSHKE

13.1. ENERGETICS A N D STOICHIOMETRY ........................ 287


13.1.1. Oxidation State of Reactants and Products .................. 288
13.1.1.1. Example 1 .................................. 290
13.1.2. Balanced Reduction Half-Reaction Normalized to 1 e- eq ....... 290
13.1.2.1. Example 2 .................................. 290
13.1.3. Gibbs Standard Free Energy at pH 7 for a Reduction
Half-Reaction (AGO ) ................................. 290
13.1.3.1. Example 3 .................................. 290
13.1.4. Commonly Used Electron Acceptor Half-Reactions (R,) ........ 292
.5. Commonly Used Electron Donor Half-Reactions ( R d ) .......... 294
.6. Electron Acceptor#DonorPair as Energy-Yielding Reaction ( R e ) . . 295
13.1.6.1. Example 4 .................................. 295
.2. Example 5 .................................. 295
13.1.7. Automated Assignment of Source of Carbon for Cell Synthesis ... 296
.8. Model Cell Synthesis Half-Reactions as a Function
of Nitrogen Source (R,) ............................... 296
.9. Assumed Efficiency of Energy Transfer ( E ) ................. 296
.lo. Fraction of electron donor used for cell synthesis ()f: .......... 296
13.1.10.1. Example 6 ................................. 297
13.1.1 1. The Overall Stoichiometric Equation (R) ................... 297
13.1.1 1.1. Example 7 ................................. 298
13.2. KINETICS OF MICROBIAL GROWTH ........................ 299
13.2.1. Modeling Batch Systems ............................... 302
13.2.1.1. Principles .................................. 302
13.2.2. Continuous-Culture Systems Modeling .................... 303
13.2.2.1. Chemostat .................................. 305
.2. Growth Yield Calculations ...................... 306
.3. Mean Cell Residence T i e , 8, versus Dilution Rate, D ... 307
13.3. REFERENCES ............................................ 307

Materials, electrons, and energy are three essential compo- duction of the materials and to polymerize the building
nents needed to support the growth of all microorganisms blocks together in the form of useful enzymes, structural ma-
(Fig. 1).Materials serve as building blocks for the additional terials, and information containing molecules such as nu-
cell mass and include carbon, nitrogen, phosphorus, and cleic acids. Energy is obtained by a transfer of electrons from
trace elements. Carbon is obtained from the vast number of the electron donor to an electron acceptor (a compound
organic compounds (by heterotrophic microorganisms) or that is electron deficient). Thus, there are two separate
from COZ (by autotrophic microorganisms). Nitrogen is ob- needs for the electron donor, one to provide electrons to re-
tained from ammonium, the most common nitrogen source, duce the materials and the other to transfer electrons to the
or from nitrate, nitrite, dinitrogen, or other nitrogen oxides. acceptor to release energy.
Electrons are needed to reduce most of these materials from The study of energy transactions in chemical or bio-
a relatively oxidized state that is commonly found in nature chemical reactions, e.g., during the transfer of electrons
to a reduced state that is characteristic of cell material. from an electron donor to an acceptor is known as energet-
Electrons come from an electron-rich compound, which is ics. The study of the quantitative relationships among the
termed electron donor. To quantitatively relate reactants electron donor, electron acceptor, cell mass, nitrogen, and
and products, it is often helpful to represent compounds other reactants and products is known as stoichiometry. In
in terms of their electron equivalents (e- eq). For example, addition to energetics and stoichiometry, it is also important
1 mol of glucose can be represented as 24 e- eq (see section to know the rate at which the enzymes present in the cell
13.1.1 for more details). Energy is needed to perform the re- mass may carry out the biological reactions. The study of
rates is known as kinetics. Kinetics plays an important role
in determining the size of the reaction vessel in which the
tThe section on kinetics in this chapter was previously written by Philipp process is being carried out or in determining the outcome
Gerhardt and Stephen W. Drew. of competition between two populations or guilds.

286
13. Energetics, Stoichiometry, and Kinetics w 287

Materials Electrons Energy Microbial Cell


FIGURE 1 Simplified depiction of the three essential components needed to support the growth
of all microorganisms.

The above description of energetics, stoichiometry, and Re, and the cell synthesis reaction, R, (Fig. 2 ) . A n energy-
kinetics highlights only the main aspects of microbial yielding reaction itself is constructed by combining an elec-
growth in a greatly simplified manner. In reality, the process tron donor half-reaction ( R d ) with an electron acceptor
of forming another microbial cell is extremely complex, es- half-reaction (R,) .
pecially at the molecular level of resolution. Reactions oc- For simplicity, all analysis is carried out with 1 e- eq of
curring within a single cell that ultimately enable the cell to donor. Considering that Rd represents the source of elec-
grow and divide are only beginning to be understood and trons, a fraction of the donor, represented by ,:f must be
modeled through some very large-scale efforts (for an ex- transferred to the acceptor reaction to yield energy. The
ample, see http://microbialcellproject.org/). Yet at a coarser remaining fraction, represented by :f (which also equals
resolution, the growth and performance of many different 1 -):f, is transferred to the synthesis reaction to reduce the
types of cells, populations, and guilds can be modeled to ob- materials used for cell synthesis. Thus, 1 e- eq of donor is
tain predictive equations for various applications. (A guild distributed between the acceptor half-reaction and cell syn-
is defined as a group of many different types of populations thesis half-reaction. The overall stoichiometric equation R
capable of utilizing similar substrates.) is written so that the donor half-reaction (Rd) seems to di-
This chapter introduces concepts related to energetics, vide the 1 e- eq between R, and R, in the ratio :f to.:f In
stoichiometry, and kinetics of microbial growth. It consists the form of an equation, the above statement can be writ-
of two parts: the first part focuses on energetics and stoi- ten as R = :f R, + Lo R, - R& This is depicted in Fig. 2 by
chiometry, and the second part focuses on kinetics of micro- arrows marked with ,:f representing the fraction of elec-
bial growth. The section on energetics and stoichiometry trons transferred from the donor to the acceptor for energy,
includes some basic concepts such as computation of oxida- and ,:f representing the electrons transferred from the
tion states and half-reactions that are critical to formulate a donor to the materials for cell synthesis. The total electron
stoichiometric equation. Illustrative examples, when appro- donor is equal to :f +,:f which is equal to 1 e- eq.
priate, are also presented. The section on kinetics presents A cell synthesis reaction represents the assembly of raw
equations governing cell growth, focusing on batch cultures materials from the environment into cell macromolecules.
and chemostats. All analyses and statements in this chapter In a cell synthesis reaction, the Gibbs standard free energy
assume the existence of a single cell (or spore that could at pH 7, AG;, obtained from the energy-yielding reaction
transform into a vegetative state) that contains the enzy- and electrons (f:) from the electron donor are used to re-
matic capacity and information necessary to put the building duce carbon, nitrogen, and other cell constituents into an
blocks, electrons, and energy into a copy of itself. oxidation state of the materials found in the cell and to as-
semble these constituents into cellular macromolecules. If
the donor is an organic compound, it may also serve as the
13.1. ENERGETICS AND STOICHIOMETRY source of carbon for cell synthesis. If the donor is an inor-
Quantitative studies related to growth of microorganisms ganic compound (e.g., NH4+, H2, HlS, etc.), the source of
often seek to determine the amount of cell mass that is ex- cell carbon is C02.In Fig. 2, the two sources of carbon are
pected from a given amount of material (electron donor, depicted by dotted lines emanating either from the donor or
electron acceptor, nitrogen, etc.) or aim to compute how from C02 and ending at the source of carbon for cell syn-
much donor (or acceptor) will be needed to consume a cer- thesis. The conversion of carbon to cell material is a com-
tain amount of acceptor (or donor). This requires writing a plex process. For simplicity of computing the energy re-
stoichiometric equation, R, that relates electron donor, quired to convert the carbon source to cell carbon, this
electron acceptor, carbon source, nitrogen source, cell mass conversion is assumed to take place in two steps via pyru-
produced, and other by-products of donor oxidation and ac- vate: (i) conversion of carbon source to pyruvate and (ii)
ceptor reduction in a quantitative manner. It combines conversion of pyruvate to cell carbon. The basis and need
principles from both redox chemistry and thermodynamics for this assumption are described in more detail in reference
and serves as the basis for evaluating the quantitative rela- 20.
tionships among the reactants and products. To write R, the Considering that approximately 13% of cell weight (dry
process of microbial growth is viewed as a set of two bio- weight) is nitrogen, the synthesis of cells requires a significant
chemical reactions, namely the energy-yielding reaction, amount of nitrogen. Depending on the source of nitrogen,
288 H GROWTH

Heterotrophy OR Autotrophy

aG,O,
ELECTRON DONOR .......................................... ................................................ .....~
....................................... co2
I I
+
Ca Ion
fe0 + fSO =1

Electrons: fso

feo

*em m
Energy: AGp
rn m i rn rn i ................ A%
..........
NH,
NO,-
NO,-
N2

1
AGa0 I ELECTRON ACCEPTOR I Pyruvate

P. Trace elements Vitrogen

CELL

1 ENERGY-YIELDING REACTION, Re
(Catabolism)
CELL SYNTliESlS REACTION, RC
(Anabolism)

FIGURE 2 The overall process of microbial growth in terms of electron donor, electron accep-
tor, carbon and nitrogen sources, and energy for synthesis reaction.

electrons may also be needed to reduce it. NH4+is the most common electron donor half-reactions (Rd)
common source of nitrogen. It is at the same oxidation state electron donor-acceptor pair as energy-yielding reactions
as cell nitrogen (i.e., -3). Hence, it does not require elec- (Re)
trons to be reduced to the oxidation state of cell nitrogen. model cell synthesis half-reactions as a function of ni-
Other more oxidized nitrogen sources (e.g., NO3-, NOz-, trogen source (R,)
and N2) must be reduced to the level of NH4+ by addition
of electrons from the donor. This is one of the reasons for a assumed efficiency of energy transfer (E )
somewhat lower cell yield on relatively oxidized nitrogen fraction of electron donor used for cell synthesis (f):
sources. The impact of nitrogen source on the process of automated assignment of source of carbon for cell syn-
writing the stoichiometric equation R is incorporated by thesis
having separate cell synthesis half-reactions for each nitro- the overall stoichiometric equation ( R )
gen source.
The overall process of writing a stoichiometric equation The discussion in these sections is intended to be an in-
R described above involves a number of concepts. These are troduction to the process of formulating an overall stoi-
listed below and described in the sections that follow chiometric equation. For further details, the reader is re-
(13.1.1 to 13.1.11). ferred to the original references (3, 20, 24).
oxidation state of reactants and products 13.1 . I . Oxidation State of Reactants and Products
balanced reduction half-reaction normalized to 1 e- eq During microbial growth, elements such as carbon, nitro-
Gibbs standard free energy at pH 7 for a reduction half- gen, oxygen, sulfur, etc., are often transformed from one ox-
reaction (AGO ) idation state to another. An electron donor gets oxidized,
common electron acceptor half-reactions (R,) and an electron acceptor gets reduced. Thus, the ability to
13. Energetics, Stoichiometry, and Kinetics 289

evaluate the oxidation states of reactants and products is average oxidation state of carbon in glucose as zero.
the first step in predicting the role of the compound as an Similarly, in COzthe oxidation state of carbon can be com-
electron donor or acceptor. Consider, for example, the fol- puted as +4 because each oxygen atom is at an oxidation
lowing transformations: glucose (C6H1206) to COq; state of -2. Thus, the transformation of C6H1206to CO2 is
trichloroethene (TCE, C2HC13) to ethene (C2H4);NH4 an oxidation involving a change in the oxidation state of
to NO3-; NO3- to NZ;O2 to H2O; and Cr6+ to Cr3'. All each carbon atom from 0 to +4. Because there are 6 carbon
the above transformations are known to support microbial atoms in glucose, a total of 24 e- eq may be donated by 1
growth. Their role as electron donor or acceptor can be ten- mol of glucose. This can also be readily seen by writing the
tatively assigned by computing the oxidation state of car- balanced half-reaction for complete glucose oxidation, as
bon, nitrogen, oxygen, or chromium, respectively. The oxi- +
follows: C6H1206 6 H 2 0 + +
6 COz 24 H+ + 24 e-.
dation state of a given element in a compound is A change in oxidation state combined with energy
determined by the following rules. yield and its utilization for growth determines the role of
the compound as a donor or acceptor. A substance being
All elements when combined with themselves (e.g., 0 2 , oxidized may have the potential to serve as an electron
Hz, and NJ have an oxidation state of 0. donor if it yields energy when combined with an acceptor
Except when combined with itself, oxygen has an oxida- to support microbial growth. Conversely, a substance
tion state of -2. However, in peroxides (HzOz and being reduced may have the potential to serve as an elec-
Na20z),the oxidation state of oxygen is - 1. tron acceptor if it yields energy when combined with an
Except when combined with itself, hydrogen has an ox- electron donor. Figure 3 depicts the oxidation states for a
+
idation state of 1. However, in metal hydrides (NaH, number of key elements present in various compounds of
LiH, etc.), the oxidation state of H is -1. interest.
The sum of oxidation states of all the elements in a Considering the above examples of transformations, it
species or group is the net charge on that group. can be easily shown that TCE transformation to ethene is a
Nitrogen and sulfur when present in an organic com- reductive process. Each carbon in TCE is at an oxidation
pound can be assumed to be in an oxidation state of -3 +
state of 1, which is computed from the knowledge that the
and -2, respectively, unless specified otherwise. This is oxidation state of C1 in TCE is -1 and that of H is +l.
only to simplify the calculation of the need for electrons Thus, the transformation of TCE to ethene has the poten-
and should not be used as a universal rule for the oxida- tial to serve as an electron acceptor. The transformation of
tion state of these compounds in microbial cells. NH,+ to NO3-, however, is an oxidation process because
nitrogen changes its oxidation state from -3 to +5. It in-
Following the above rules. the average oxidation state of volves a transfer of 8 electrons. Thus, the conversion of
each carbonvin C6HI2O6can be compG,ed as follows. We NH4+ to NO3- may potentially serve as a donor reaction.
begin by representing the unknown oxidation state, in this The remaining transformations (NO3- to Nz, O2to HzO,
case that of carbon, as x. As per the above rules, the oxida- and Cr6' to Cr3') are all reductive processes involving a
tion state of hydrogen is known to be + 1 and that of oxy- transfer of 5, 2, and 3 electrons, respectively. Each has the
gen is known to be -2. Thus, the sum of oxidation states of potential to serve as an electron acceptor. The following ex-
all elements in glucose can be written as 6x +
12(+1) + ample highlights the use of oxidation state in tentatively
6( -2). This must be equal to the charge on the compound, predicting the possible role of a given compound in an
which in the case of glucose is zero. Solving for x yields the energy-yielding reaction.

As

Mn Mn2+ MnO, MnO,,-


Cr Cr3+ Cr,O,,-

CI CI- CI, CIO; CIO, CIO,


H NaH H, H+

0 HZO H202 02

S HZS s so-; so42.


N NH,+ N2 NO; NO,-
C CH, co c6H1206 c2c14 COZ

-6 -5 -4 -3 -2 -1 0 +I +2 +3 +4 +5 +6
Oxidation
..............................
Reduction
..................................

FIGURE 3 Oxidation state of selected elements (in boldface) in various compounds of biologi-
cal interest.
290 GROWTH

13.1 .I . I . Example 1 13.1.2.1. Example 2


H2S is known to be transformed to SO:- and H+ by mi- Arsenic compounds in drinking water sources are a health
croorganisms that grow on the topmost surface of concrete hazard around the world. Both chemical and biological
inside sewer pipes and cause corrosion of concrete due to means of transforming and removing arsenic compounds
the acid production. How many electrons are transferred in from drinking water are of interest. Strain MLMS-1 is a
this process?What is the possible role of H2S in this micro- chemoautotrophic arsenate (AsO:-) respirer, which grows
bially mediated transformation process? by oxidizing sulfide to sulfate while reducing arsenate to ar-
The oxidation state of S in H2S must be -2 because H senite (HZAs03-) (7). Write a balanced reduction half-
is at + 1 and there is zero charge on the compound. The ox- reaction normalized to 1 eC eq for the electron acceptor
idation state of S in SO:- is +6 because 0 must be at an used by this organism.
oxidation state of -2 and the sum of oxidation states in Because 0 is at an oxidation state of -2 in both arsen-
SO:-, i.e., x + 4(-2), must be equal to the charge, -2. ate and arsenite, arsenic is at +5 oxidation state in arsenate
The change in the oxidation state of S from -2 to +6 in- and at + 3 oxidation state in arsenite. Hence, arsenate is the
dicates that the transformation of H2S to SO:- is an oxi- oxidized species and arsenite is the reduced species. The fol-
dation reaction involving 8 electrons. The transformation lowing seven steps follow the procedure listed above to ob-
is expected to serve as an electron donor reaction because S tain the reduction half-reaction normalized to 1 e- eq for
is getting oxidized. Further evidence for this role may be ob- the above transformation.
tained by computing the energy available when H2Sis com-
bined with an appropriate electron acceptor (0, in this 1. As0:- = H2As03-
case). 2. As0:- + e- = H2As03-
3. As0:- + e- = H2As03-
13.1.2. Balanced Reduction Half-Reaction 4. As0:- + e- + HzO
= H2As03-
Normalized to 1 e- eq 5. As0:- + 4H+ + e- = H2As03- + H 2 0
The information gathered from the above exercise related
6. As0:- + 4H+ + 2e- = H2As03- + H 2 0
7.1/2 AsOd- + 2H+ + e- = 1/2 H2As03- + 1/2 H2O
to oxidation states can be used to write half-reactions. Half-
reactions are reactions involving electrons as a substrate or
as a product. If a reaction does not contain electrons, it can- 13.1.3. Cibbs Standard Free Energy at p H 7
not be called a half-reaction. Each half-reaction can be for a Reduction Half-Reaction (AGO)
written either as a reduction half-reaction or an oxidation The Gibbs standard free energy of a half-reaction modified
half-reaction. Reduction half-reactions always have elec- for pH 7 (AGO ) for the donor and acceptor half-reacFions is
trons as reactants on the left-hand side of the equation. needed to compute the available free energy (AG,O) from
Similarly, oxidation half-reactions always have electrons as the energy-yieldingreaction (Re) and to determine the frac-
product on the right-hand side of the reaction. The avail- tion of electron donor,,:f used for the cell synthesis reac-
ability of balanced reduction half-reactions normalized to 1 tion. These values in turn are used to write the stoichio:
e- eq for the donor, acceptor, and cell synthesis greatly sim- metric equation, R. Tables 1 and 2 list the associaged AGO
plifies the process of writing the stoichiometric equation R for many half-reactions of interest. Values of AGO for any
and subsequent quantitative analysis. Normalization of the reaction can be computed from the free energies of forma-
half-reactions to 1 e- eq simplifies the procedure of obtain- tion (Gfo) of reactants and products by using the following
ing R to a simple combination of the half-reactions for equation (10).
donor (Rd), acceptor (RJ,and cell synthesis (R,) in a man-
ner governed by .:f All half-reactions, by convention, are AGO = CGP (products) - CGfo (reactants) ( 1 )
written as reduction half-reactions. This is critical for cor- Excellent resources to obtain GP values exist, e.g., refer-
rect use of the approach for obtaining R and :f described ences 20 and 24, Brock Biology of Microorganism (lo), the
here (20). A number of half-reactions are listed in Table 1 online version of the CRCs Handbook of Chemistry and
(organic) and Table 2 (inorganic) with their associated Physics (http://www.hbcpnetbase.com/), and Thermodyn
Gibbs standard free energies (adopted from various sources (see above). Some of these values are reproduced here for
such as references 20 and 24 and Thermodyn, an XL-based easy reference (Table 3).
program on energetics [K. Hanselmann {http://www
.microeco.unizh.ch~], often with some modifications to 13.1.3.1. Example 3
present them as reduction half-reactions normalized to
1,1,l-Trichloroethane (l,l,l-TCA) is a chlorinated solvent
1 e- eq as needed in the approach described here). Any bal-
and a groundwater contaminant in the United States (14).
anced reduction half-reaction normalized to l e- eq can be
Recently, it has been shown to support microbial growth,
obtained by following the steps described below.
serving as an electron acceptor (22). You are interested in
1. Write the oxidized chemical species on the left-hand computing the Gibbs standard free energy (AGO ) for the
side and the reduced chemical species on the right-hand side. transformation of l,l,l-TCA to chloroethane so that the
2. Add electrons on the left-hand side because the half- computed value can be used with the donor half-reaction to
reaction is a reduction half-reaction. determine the amount of energy available. You have written
3. Add other species as needed to balance the carbon, ni- the reduction half-reaction normalized to 1 e- eq for the
trogen, phosphorus, etc. transformation of l,l,l-TCA to chloroethane. Calculate
4. Balance the oxygen by adding water (and not 02). the AGO for the half-reaction representing the conversion
5. Balance the hydrogen with H+. of 1,1,1-TCA to chloroethane.
6. Balance the charge by changing the number of e-. First we write the balanced reduction half-reaction of
7. In the final step, the balanced half-reaction is divided 1,1,1-TCA transformation to chloroethane normalized to 1
by an integer (the multiplication coefficient of the eC) to e- eq, following the procedure described in sections 13.1.1
convert the half-reaction to one electron equivalent. and 13.1.2.
G
N (a3vd ixau uo panuuuo3)
~~
so0 01'0 05'0 15'0 85'0 65'0 9Z'lZ O'H Z6/1E + -003b'(ZH3)'H3 Z6/1 = + +H + -'03H Z6/1 + '03 6I/SI Of
ln SO0 01'0 oso LSO 85'0 65'0 bC1Z O'H Of/El + -003'H93 Of/I = -3 + +H + -'03H OdI + '03S/I 62
.-U
Y
a, so0 01'0 05'0 85'0 85'0 65'0 b'1Z O'H 8/E + -003'H3 8/1 = -a + +H + -'03H 8/1 + '03 8/1 82
(I:
2 so0 01'0 05'0 85'0 85'0 65'0 E9'1Z O'H bI/S + _003'H3'H3 bI/I = -a + +H + -'03H bI/1 + '03 1/1 1z
-0 so0 11'0 05'0 85'0 85'0 65'0 bL'1Z O'H OZ/L + -003'H3'H3'H3 OZ/I = -a + +H + -'03H OZ/1 + '03 OZ/E 9z
C
m
900 11.0 05'0 8S'O 65'0 09'0 28'12 O'H Zf/11 + _003'('H3)'H3 Zf/I = -a + +H + -'03H ZE/I + '03 Z d S SZ
E
Y
100 1'0 ZS'O 090 09'0 19'0 6062 O'H 1/E + '(~003)'('H3)bI/1 = -2 + +H + -'03H 1/1 + '03 1/1 bZ
a,
.-
: 01'0 51'0 bS'O 290 29'0 E9'0 z L'OE OZH8/E + - Z 0 3 Z H 3 0 3 E H91/13 = -a + +H+ -'03H 91/1+ '03 91/f EZ
L 01'0 91'0 bS'0 29'0 29'0 E9'0 E6'OE OZH6/b +-003ZHNH3ZH3ZH3H00381/1 = -a + +H + ++HN81/1 + -'03H 6/1 + '03 911 ZZ
.-U
0 11.0 91'0 SS'O Z9'0 Z90 E9'0 IOIE O'H d1 + -003'H3HOH3'H3 81/1 = + +H + -'03H 81/1 + '03 81/E IZ
5;
v; 11'0 91'0 SS'O Z9'0 f90 b9'0 81'1E O'H +/I + HOZH3'H3 Z1/1 = -2 + +H + '03 911 oz
.-cU 11.0 91'0 SS'O Z9'0 E90 b9'0 1fIE O'H ZI/S + -003'HNH3'H3 Z1/1 = -a + +H ZI/II + +'HN Z1/1 +
a,
53 -'03H ZI/1 + '03 911 61
a,
K EI'O 81'0 9S'O b9'0 b90 59'0 6Z'ZE O'H d1 + -003HOH3'H3 ZI/1 = -2 + +H + -'03H ZI/1 + '03 911 81
w
m EI'O 81'0 LS'O b90 b90 S9'0 Eb'ZE O'H Z/I + -00303'H3'H3 91/1 = -2 + +H + -'03H 91/1 + '03 8/1 11
7
b1.O 61'0 85'0 590 590 990 80EE O'H 6/b +
~ ~ 3 ' H 3 ~ ~ 0 0 3 ~ H 0 3 ' H81/1 3 ~=~-30 +0 +H
3 ~+ -'03H 9/1 + '03 911 91
51'0 OZ'O 85'0 59'0 S90 99'0 Sb'f E O'H 6/b +
- 0 0 3 H O H 3 - 0 0 3 H 3 ' H 3 - 0 0 3 81/1 = -9 + +H + -'03H 81/ + '03 81/E ST
SI'O OZ'O 85'0 S9'0 990 19'0 9'fE O'H OI/E + OH3'H3 OI/I = -3 + +H+ '03 S/I bI
91'0 ZZ'O 65'0 99'0 99'0 19'0 11'tf O'H ZI/S + - 0 0 3 ' H 3 H O H 3 - 0 0 3 Z/I = -a + +H + -'03H 9/1 + '03 9/1 El
91'0 ZZ'O 65'0 99'0 19'0 19'0 SZ'bE O'H 91/1 + 0 0 3 0 3 ' H 3 ' H 3 ~ 0 0 3 91/1 = -a + +H + -'03H 8/1 + '03 91/f ZI
81'0 EZ'O 19'0 19'0 89'0 69'0 60SE O'H S/Z+ - 0 0 3 0 3 ' H 3 01/1 = -a + +H+ -'03H OI/I+ '03 S/I I1
IZ'O 9Z'O Z9'0 89'0 69'0 01'0 t8'9E O'H 9/1 + HO'H3 911 = -9 + +H + '03 911 01
ZZ'O 1Z'O E9'0 69'0 690 01'0 E8'1E OZHZ/I + - 0 0 3 ' H 3 0 3 - 0 0 3 O l / l = + +H + -'03H S/I + '03 S/I 6
EZ'O 82'0 E9'0 69'0 01'0 01'0 SZ'8E O'H S/1 + HOZH3HOZH3OI/I = -2 + +H + '03 S/I 8
bZ'0 62'0 E9'0 01'0 01'0 11.0 88'8E O'H bI/E + HOZH3HOH3HOZH3bI/I = -9 + +H + '03 bI/E 1
SZ'O 62'0 $9'0 01'0 01'0 11.0 61'6E O'H Z/I + - 0 0 3 H Z/I = -a + +H + -'03H Z/I 9
91'0 o('0 b9'O 01'0 11.0 11.0 K6E O'H Z/I + H003'HNZH3 911 = -a + +H + +bHN911 + -'03H 911 + '03 911 S
82'0 ECO 59'0 11.0 21.0 Z1'0 Sc'Ib OZH+/I + 9 0 ~ 1 ~ bz/r 9 3 = -a + +H + 2 0 3 +/I b
oc'o b('0 99'0 Z1'0 Z1'0 EL'O 99Zb -J
O'H +/I + OH3HOH3HOZH3Z/I = + +H + '03 +/I E
9c'O Ob'O 69.0 tL'O 51.0 SL'O 99t O'H b/I + O'H3 +/I = -a + +H + '03 b/I Z
Ib'O bb'O 11.0 91'0 11.0 11.0 lV6t O'H Z/I + - 0 0 3 H 0 3 H +/I = -2 + +H + -'03H b/I + '03 +/I I
292 H GROWTH

i +
1/4 C2H3C13 1/2 H+ + e-
z = 1/4 C2HSCl + 1/2 C1- (2)
Then, we obtain the free energies of formation (Gfo)for
$
I-
C2H3C13, Hf, e-, C2H5C1,and C1- from reference 20. The
following values (in kilojoules/mole)were obtained from ref-
erence 20: -69.04 for l,l,l-TCA a ueous state [aq], -39.87
for H+ (as) at pH 7, 0 for e- (Gf 'b for e- is always zero),
-36.83 for chloroethane (aq), and -131.26 for C1- (aq).
Note that unless a reactant or product is known to participate
in the reaction in other forms, the values selected for Gfo
should be for the aqueous state.
Using equation 1, the value of AGO' for the above half-
reaction is computed as:

AGO' = CG: (products) - C G: (reactants)


- [1/4 (-36.83) + 1/2 (-131.26)] - [1/4(-69.04)
+ 1/2 (-39.87) + (O)]
= -37.64 kJ/e- eq
It should be noted that the superscript "0" in AG,"' im-
plies that the free energy is computed for standard conditions
and the prime in AG? implies that the Gibbs standard free
energy (originally AG,") has been corrected for pH 7 (i.e.,
H+ concentration is assumed to be lop7M instead of 1 M).
This is to compare the energies at a pH that is relevant for
most, but not all, biological reactions. There may be in-
stances where this correction may be necessary for other
values of pH (e.g., in biological reactions occurring at much
lower pH, as in acid mine drainage). In such cases, the value
of G: for Hf corresponding to the pH of interest should
be used. Because G: for Hf at pH 0 is 0, the change in G:
per pH unit is -5.69 kJ/mol.

13.1.4. Commonly Used Electron Acceptor


m
c)
Half-Reactions (R,)
a
3
As mentioned previously, the compound that accepts the
%
a electrons from the donor in an energy-yielding reaction is
m I
II
I
I1
I
II
I
II II
I I
1
I
I
II
I
II
I
II
I
II known as the electron acceptor. It is the compound that the
3 ~ ~ a J a J a J a J a J a J a J a J organism is said to "breathe." All aerobes, including most
+ + + + + + + + + + chemo-organotrophs and chemolithotrophs, can use oxygen
d as an electron acceptor. In the absence of oxygen, nitrate-
rL
reducing organisms or denitrifiers use NO3- and other ni-
trogen oxides as their electron acceptor; SO:- is the major
electron acceptor for sulfate-reducing organisms; and C02
is the electron acceptor for most methanogens and aceto-
gens. These compounds can serve only as electron accep-
tors, because the corresponding elements are at their most
oxidized states (0 for 02,+5 for N, +6 for S, and +4 for
C02).In addition, organic matter can be used by fermenters
as a source of electron donor as well as a compound that can
accept electrons, because carbon in organic compounds can
exist at oxidation states ranging from -4 to less than +4.
Besides the above well-known acceptors, there are many
other relatively less studied electron acceptors in nature, e.g.,
Fe3+,Mn3+,Cr6+,etc. Over the past 2 decades, many more
compounds have been added to the list of electron accep-
tors. These include several chlorinated compounds, such as
chlorobenzoate (4), perchloroethene, TCE, dichloroethene,
vinyl chloride ( l l ) , l , l , l - T C A (22), chloropropanes,
halophenols, etc., and inorganic ions, such as chlorate, per-
chlorate, arsenate, selenate, uranium(IV), etc. The half-
reactions for many of these acceptors are listed in Tables 1
and 2.
Note that the term electron acceptor is reserved for the
compounds whose reduction releases energy which must be
TABLE 2 Reduction half-reactions normalized to 1 e- eq for selected inorganic compounds and the associated values of AGO' and theoretical values of fp

Reaction Reduced-oxidized Substrates and products


no. compounds

1 Sulfite-sulfate 1/2 S04-2 + H+ + e- = 1/2 SO3-' + 1/2 H2O 50.30 0.26 0.25 0.25 0.21 0.08 0.07
2 Hydrogen-H' H+ + e- = 1/2 H2 39.87 0.24 0.24 0.23 0.19 0.05 0.04
3 Ammonium-nitrogen 1/6 N2 + 4/3 H' + e- = 1/6 NH: 26.70 0.22 0.22 0.21 0.16 0.02 0.01
4 Sulfide-sulfur 1/2 S + 1/2 H+ + e- = 1/2 HS- 26.05 0.22 0.21 0.21 0.16 0.02 0.01
5 Thiosulfate-sulfate 1/4 + 5/4 H' + e- = 1/8 S Z O ~+- ~5/8 H2O 23.58 0.22 0.21 0.21 0.16 0.01 0.00
6 Methane-carbon 1/8 C 0 2 + H+ + e- = 1/8 CH4 + 1/4 H2O 23.12 0.22 0.21 0.21 0.16 0.01
dioxide
7 Sulfide-sulfate +
1/8 S04-2+ 9/8 H' e- = 1/8 HS- + 1/2 H2O 21.23 0.21 0.21 0.20 0.15 0.01
8 Sulfide-sulfate 1/8 S04-2 + 19/16 H' + e- = 1/16 HZS + 1/16 HS- + 1/2 H2O 20.85 0.21 0.21 0.20 0.15 0.00
9 Sulfide-thiosulfate 1/8 S203-2+ H+ + e- = 1/4 HS- + 3/8 H2O 20.26 0.21 0.20 0.20 0.15 0.00
10 Sulfur-sulfate 1/6 S04-2 + 4/3 H+ + e- = 1/6 S + 2/3 H2O 19.15 0.21 0.20 0.20 0.15 N/A
11 Sulfide-sulfite 1/6 + 5/4 H+ + e- = 1/12 H2S + 1/12 HS- + 1/2 HzO 11.03 0.20 0.19 0.18 0.13
12 Arsenite-arsenate 1/2 A s O ~ -+~2 H+ + e- = 1/2 H2As03- + 1/2 H2O - 14.47 0.15 0.14 0.14 0.08
13 Cuprous-cupric cu+' + e- = c u + - 15.44 0.15 0.14 0.13 0.08
14 Te-Te(OH), 1/4 Te(OH)3 + 3/4 H+ + e- = 1/4 Te + 3/4 H2O -24.12 0.13 0.12 0.12 0.05
15 Selenium-selenite 1/4 Se03-2 + 3/2 H+ e- = 1/4 Se + 3/4 H2O -26.05 0.13 0.12 0.11 0.05
16 Selenium-selenate 1/6 Se04-' + 4/3 H+ + e- = 1/6 Se + 2/3 H2O -31.84 0.11 0.10 0.10 0.04
17 Ammonium-nitrite 1/6 NO2- + 4/3 H+ + e- = 1/6 NH4' + 1/3 H2O -32.93 0.11 0.10 0.10 0.03
18 Ammonium-nitrate +
1/8 NO3- 5/4 H++ e- = 1/8 NH4' + 3/8 H2O -35.11 0.11 0.10 0.09 0.03
19 Nitrite-nitrate 1/2 NO3- + H+ + e- = 1/2 NO*- + 1/2 H2O -41.65 0.09 0.08 0.08 0.01
20 Selenite-selenate 1/2 Se04-* + H+ e- = 1/2 Se03-2 + 1/2 H2O -43.42 0.09 0.08 0.07 0.01
21 Manganous-manganese 1/2 Mn02 + 2H+ e- = 1/2 Mn'2 + H2O -45.35 0.08 0.07 0.07 N/A
dioxide
22 Nitrogen-nitrate 1/5 NO3- + 6/5 H+ + e- = 1/10 Nz + 3/5 HzO -72.20 0.02 0.01 N/A
23 Ferrous-ferric Fe+3 + e- = Fe" -74.27 0.01 N/A
24 Mercury-mercuric 1/2 Hg" + e- = 1/2 Hg -77.19 0.00
25 Water-oxygen 1/4 O2 + H+ + e- = 1/2 H20 -78.72 N/A w
3
26 Nitrogen-nitrite 1/3 NO2-+ 4/3 H+ + e- = 1/6 Nz + 2/3 H2O -92.56 Q

27 Chloride-chlorate 1/6 C103- + H+ + e- = 1/6 C1- + 1/2 H2O - 100.34


x-
2

28 -
Nitroeen-nitrous oxide . N -7 0 + H+ + e-
1/2 = 1/2 N, + 1/2 H,O - 126.40 3.
G!
"Computed assuming NH4+as nitrogen source, C5H,O,N as cell formula, and E = 0.6. Note that the values off: are provided to save time in computation. They do not necessarily imply the existence of an organism that can
use the corresponding electron acceptor-acceptor pair. Data from references 3, 20, and 24.
294 GROWTH

TABLE 3 Free energies of formation, GP, for selected substances


Substance G: (kJmol-') Substance G: (kJmol-')
co2 -394.4 P
o
:- - 1026.55
co -137.34 H2PO4- - 1130.2
0 2 0 Seo 0
H2 0 SeOt- -441.3
H2O -237.17 H2Se -77.09
HC03- -586.85 scot- -439.95
co32- -527.90 MgSO4 - 1199.5
H2C03 -623.16 PbS -92.59
N2 0 cus -49.02
NH3 -26.57 cu2+ +64.94
NH4+ -79.37 cu+ +50.28
NO +86.57 MoSz -225.42
NO2 +51.95 Moot- -836.3
NO3- - 111.34 ZnS -198.60
NO2- -37.2 Zn+' -147.1
N20 +104.18 Mn3+ -82.12
SO 0 Mn+2 -227.93
~ 0 ~ ~ - -486.6 MnOz -456.71
s
o
:- -744.6 Mn04- -506.57
HS03- -527.7 MnS04 -955.32
HSO4- -755.9
H202 - 134.1 Aspartate -700.4
~203'- -513.4 Crotonate -277.4
H2S -27.87 Cysteine -339.8
HS- +12.05 Fructose -915.38
S=- +85.8 Fumarate -604.2 1
c1- -131.2 Gluconate - 1128.3
CD- 17.2 Glycerate -658.1
(210,- -8.5 Guanine +46.99
C103- -8.0 Glycolate -530.95
c10- -36.8 Lactose -1515.24
~r207~- -1301.1 Mannitol -942.61
Cat- -727.8 Methane -50.75
Fe2+ -78.87 0xa1ate -674.04
Fe3+ -4.6 Phenol -47.6
FeCO3 -673.23 n-Propanol -175.81
FeS04 -829.62 Ribose -757.3
FeS2 - 150.84 Sucrose -370.90
u+4 -531.9 Urea -203.76
u+3 -476.2 Valerate -344.34
Electron (e-) 0, always
H+ 0 at pH 0; -39.83 at pH 7 (-5.69 per pH unit)
OH- 57.3 at pH 14; -198.76 at pH 7; -237.57 at pH 0

captured by the enzymatic machinery of the microorgan- thesis reaction defines the term electron acceptor and not
isms to support growth. Many other compounds may use the acceptance of electrons alone.
electrons (e.g., for the reduction of materials for cell syn-
thesis or cometabolic reductive transformation of contami- 3.1 Commonh'
s5. Electron
nants), but those compounds are not referred to as electron Donor Half-Reactions (Rd)
acceptors. Thus, availability of energy when combined with Similar to the electron acceptor, the compound that do-
a donor and the subsequent use of energy in the cell syn- nates the electrons to the acceptor in a n energy-yielding
13. Energetics, Stoichiometry, and Kinetics 295

reaction is known as the electron donor. It is the com- reaction is also reversed. The reversed donor half-reaction is
pound that is said to be the energy source for the microor- added to equation 3 as follows to obtain equation 5:
ganisms (although it is more accurate to state that energy
is provided by the donor-acceptor pair). The array of com- -Rd: 1/24 CGH1206 + 1/4 H2O
pounds that can donate electrons is much larger (in thou- R,: 1/4 Oz+ H++ e-
sands) than the number of electron acceptors, which is
probably less than 100 at present. Many organic com- Re: 1/24 C~H1206+ 1/4 0 2
pounds (e.g., sugars, proteins, acids, and alcohols) and
some inorganic compounds (e.g., NH4+, HZ, Fez+, Mn2+, = 1/4 COz + H+ + e- -41.35 kJ/e- eq (4a)
S, etc.) can serve as electron donors. During heterotrophic = 1/2 HzO -78.72 kJ/e- eq (3)
growth (i.e., when the donor is organic), the donor also
= 1/4 COz + 1/4 HzO -120.07 kJ/e- eq (5)
serves as the carbon source. However, there are many de-
nitrifying and sulfate-reducing bacteria that can oxidize an Note that the energie? for the two half-reactions are also
inorganic donor, especially Hz. In such cases, COZ serves as added up. Because AG, (equation 5) is negative, indicat-
the carbon source. COzis also the carbon source during au- ing release of energy, the reaction can serve as an energy-
totrophic growth (i.e., when the donor is an inorganic yielding reaction and the assumed roles for glucose as elec-
compound) because the donor can only serve as a source of tron donor and Ozas electron acceptor were correct. In an
electrons. Nitrifiers are an example of autotrophic bacteria energy-yielding reaction, both donor and acceptor must end
using NH4+ as donor and Oz as acceptor. Similarly, iron- up on the left-hand side and the products of their oxidation
and sulfur-oxidizing bacteria, or the arsenate-respiring bac- (COzfrom glucose) or reduction (HzO from 0 2 ) must end
teria that use sulfide as the donor (see Example 2) are all up on the right-hand side of the reaction.
autotrophic, using COz as a carbon source.
13.1.6. Electron Acceptor-Donor Pair 13.1.6.2. Example 5
as Energy-Yielding Reaction (Re) Anaerobic ammonia oxidation (anammox) is a recently oh-
served chemolithotrophic process whereby ammonia is oxi-
The energy-yielding reaction can be obtained by subtract-
dized to dinitrogen gas anaerobically with nitrite as an electron
ing the donor half-reaction (%) from the acceptor half-
acceptor. The potential for existence of such a process was pre-
reaction (R,). In the form of an equation, this is written as
dicted a long time ago based on energetics. At present, many
Re = R, - Rb Because the identity of donor or acceptor species including C u ~ t uScalindua
s wagneri, Candidatus
may not always be known a priori, evaluation of their role
Scalindua brodae, Candidatus Scalindua sorokinii, Can-
proceeds by assuming that one of the half-reactions is a
didatus Brocadia anammoxidans, and Candidatus Kuenenia
donor (Rd) and the other is, an acceptor (R,), The AG: is
computed as AG: - AG: under the assumed roles. If the stuttgartiensis are known to obtain energy for growth by
anaerobic ammonia oxidation. Compute the amount of energy
computed AG, is negative, the assumed roles are said to be
available for the above donor-acceptor pair.
correct and it is concluded that energy is available and can
First, the half-reactions for NO2- and NH4+ (both trans-
potentially be used to support growth. A positive AG,, on
the other hand, implies that the assumed roles are incorrect formjng to Nz) are selected from Table 1. The associated
and energy is not available for growth from the pair under
AGO value for each half-reaction is also written as such.
Then, the known roles are assigned to each half-reaction (R,
the conditions tested. When the donor half-reaction is re-
versed to obtain -Rd, the sign of the Gibbs standard free to NOz- half-reaction and R d to NH4+ half-reaction).
energy for the donor half-reaction also reverses (i.e., it be- R,: 1/3 NOz- + 4/3 H++ e- = 1/6 N2 + 2/3 H 2 0
comes the negative of whatever the value was for AG:). In
addition, in an energy-yielding reaction (Re), electrons -92.56 kJ/e- eq (6)
must cancel out each other. Under no circumstances will an Rd: 1/6 Nz + 4/3 H+ + e- = 1/3 NH4+
energy-yielding reaction have electrons as the substrate or
product. Only half-reactions can have electrons. The fol- 26.70 kJ/e- eq (7)
lowing examples illustrate the process of writing an energy- The donor reaction is then reversed to obtain equation
yielding reaction. 7a, which is added to equation 6 as follows:
13.1.6.1. Example 4 -Rd: 1/3 NH4+ = 1/6 Nz + 4/3 H+ + e-
Evaluate if the oxygen half-reaction (Oz/H20)and the glue -26.70 kJ/e- eq (7a)
cose half-reaction (glucose/COz) can be combined to serve
as a source of energy for microbial growth. R,: 1/3 NOz- + 4/3 H + + e- = 1/6 Nz + 213 HzO
First, the half-reactions for O2 and glucose are written -92.56 kJ/e- eq (6)
as they are listed in Tables 1 and 2, i.e., reduction half-
reactions normalized to 1 e- eq. The associated AGO for Re: 1/3 NH4+ + 1/3 NOz- = 1/3 Nz + 2/3 HzO
each half-reaction is also written as such. A tentative role -118.26 kJ/e- eq (8)
to each half-reaction is assigned (say, R, to Ozhalf-reaction
and R d to glucose half-reaction). Note that the availability of energy alone does not guar-
antee that microorganisms will be able to use the released
R,: 1/4 0 2 + H+ + e- = 1/2 HzO -78.72 kJ/e- eq ( 3 ) energy for growth. Metabolic enzymes must also be available
Rd: 1/4 COZ + H+ + e- = 1/24 C6H1206 + 1/4 HzO to capture the released energy. For less commonly known
donor-acceptor pairs, the availability of enzymes to capture
+41.35 kJ/e- eq (4) the available energy can be shown only experimentally. For
Then, the assumed donor half-reactiqn is reversed to ob- example, the use of energy for growth from the C-C1 bond
tain -Rd (equation 4a). The sign of AGO for the donor half- breakage was unknown until 1987 when Dolfing and Tiedje
296 m GROWTH

(4) demonstrated that the energy released during the re- dry cell mass. In the cell, it is generally present in the aver-
duction of chlorobenzoate to benzoate can be captured for age oxidation state of -3 (as NH3/NH4+).Nitrogen in the
growth. Since then, many more microorganisms including environment may exist at an oxidation state varying from
Dehalobucter restrictus (8), Dehalococcoides ethenogenes (12), -3 (NH3/NH4+)to +5 (NO3-). Thus, if the nitrogen
and Dehukxpirillum multivorum have been shown to respire source available to the cells is other than NH3/NH4+,addi-
halogenated compounds. The subject of dehalorespiration tional electrons will be needed to reduce it to the oxidation
was recently reviewed by Smidt and de Vos (21). state of NH3/NH4+.NO3- and NO - may be used as nitro-
gen sources in the absence of NH4+z, and N2 may also serve
13.1.7. Automated Assignment of Source as a source of nitrogen if a mechanism to fix N2 is available
of Carbon for Cell Synthesis to the microorganism. Other oxides of nitrogen can also be
In nature, carbon is available as reduced organic carbon used as a source of nitrogen. From the perspective of the
with an average oxidation state varying from -4 to +3 or need for electrons to reduce the nitrogen source, NO3- is
higher but not +4 (all organic compounds) or as COz (in- the most expensive and NH3 is the least expensive among
cluding HC03-, C03-), which is the most oxidized state the four nitrogen sources. Similar to the lower yield ob-
of carbon (+4). Most compounds in the cell providing served when COz is used as the carbon source, a somewhat
structure, function, or information and especially carbon- lower growth on the more oxidized nitrogen sources is also
and nitrogen-containing compounds are found in the re- expected and observed.
duced form compared to the environment. Consider for ex- It is much easier to account for the variability in cell
ample, the elemental formula for cell material of Escherichiu yield due to various nitrogen sources by writing separate cell
coli (17), which has 50 to 54% C, 8% H, 20% 0,and 14% synthesis half-reactions (R,), one for each type of nitrogen
N (all dry weight basis). This gives a formula for cell mate- source as shown in Table 4. These equations are written
rial as CH1.900,3N0,2. From this formula, the oxidation state along the lines of R, and Rd. When writing R, one of these
of cell carbon can be computed as -0.7, which is relatively four cell synthesis half-reactions is chosen corresponding to
reduced compared to that in a typical substrate such as glu- the nitrogen source available for growth. From the coeffi-
cose, where the average oxidation state of C is 0. Because cients associated with the nitrogen source, it can be seen
carbon is also the most abundant element in microbial cells, that the number of moles of electrons needed to make 1 mol
constituting up to 50 to 55% of the dry cell mass, the need of cell material will be 28 for NO,-, 26 for NOz-, and 23
for electrons to reduce carbon is also substantial. This is es- for Nz compared to only 20 for NH4+,illustrating the effect
pecially true when the carbon source is more oxidized, e.g., of nitrogen source on cell yield.
C02/HC03-. This is why for the same amount of donor,
the cell yield is much lower when C02is used as the carbon 13.1.9. Assumed Efficiency of Energy Transfer (E)
source (autotrophy) than when an organic compound is One of the key assumptions in the process of writing R is
used as the carbon source (heterotrophy). related to the efficiency of energy transfer, 8, by the cellu-
The incorporation of the carbon source into cell carbon lar machinery, i.e., how much of the AG,O can really be
and its effect on cell synthesis are taken into account by utilized and how much of it is lost as heat. The value of E
computing:f for each carbon source and by writing the half- is assumed to be 0.6 (i.e., 40% of the energy is lost), unless
reactions in a manner that automatically reflects the source demonstrated otherwise. This assumption has important
of carbon in the overall stoichiometric equation. If the implications for the cell yield. An efficiency much lower
donor is an organic carbon source, the half-reaction for the than 0.6 (which is possible in many cases) will result in a
donor combined with the energy and synthesis reactions en- much lower cell yield. For many compounds, often the ef-
sures that the organic donor appears on the left-hand side of ficiency, E , is unknown. The value of E combined with a
the stoichiometric equation R as a source of electrons as well quantitative evaluation of the electrons and free energy
as cell carbon. If the donor is an inorganic compound, the also allows the calculation of the fraction of electrons used
combination of half-reactions ensures that COz and/or for cell synthesis (f:) and those needed for energy genera-
HCO3- appear on the left-hand side of R as the source of tion (f:) as described below. For a detailed description of
cell carbon. Thus, in the process of writing R, no active de- the rationale for using 0.6, the reader is referred to refer-
cision is required to correctly portray the carbon source. ence 20.
Exceptions may exist to this generalization, for which an in-
organic donor (e.g., Hz) and an organic carbon source (e.g., 13.1 .I 0. Fraction of Electron Donor
acetate or formate) are necessary for growth. Dependence of Used for Cell Synthesis (f,)
f> on the source of carbon is described in section 13.1.10. The fraction of electron donor that is used by the cell syn-
thesis reaction (f:) must be known to write the stoichio-
13.1.8. Model Cell Synthesis Half-Reactions metric equation. The value off: depends upon the choice
as a Function of Nitrogen Source (R& of electron donor and acceptor, carbon and nitrogen
After carbon, nitrogen is the next most abundant element sources, and the efficiency of the enzymes that the microor-
in microbial cells, constituting approximately 13% of the ganism has. The role of each one of these components in

TABLE 4 Half-reactions representing cell synthesis using NH4+,NO3-, NOz-, or NL as N source


N source Cell synthesis half-reaction,R,
NH4+ ........................... 1/20 NH4+ + 1/5 C02 + 1/20 HC03- + H+ + e- = 1/20 CjH702N + 9/20 H I 0
NO3- ........................... 1/28 NO3- + 5/28 C 0 2 + 29/28 H+ + e- = 1/28 C5H702N+ 11/28 H 2 0
NOz- ........................... 1/26 NO2- + 5/26 C02 + 27/26 H f e- = 1/26 CjH702N+ 10/26 H20
Nz .............................. 1/46 N2 + 5/23 C02 + H++ e- = 1/23 CjH702N+ 8/23 H 2 0
13. Energetics, Stoichiometry, and Kinetics 297

determining :f is incorporated in the following equation, AGp = (35.09 - AG,"')


which is obtained by considering the amount of energy = 35.09 - 36.84
needed to synthesize an equivalent of cells, the amount of
energy available from the donor-acceptor pair, and the -1.75 kJ/e- eq
=
losses. To compute,:f Rittmann and McCarty (20) assumed Because AGp is negative, n = - 1. AGO,is dictated by the
pyruvate as the key intermediate to which all carbon nitrogen source, which in this case is NO3 . Therefore, the
sources must be converted (albeit theoretically in many value of AG,, is equal to 13.5 kJ/e- eq.
cases) before being transformed to macromolecules of the Next, we obtain AG, by computing AG': - AGF. The
cell. The following paragraphs present the procedure to acceptor reaction is NO3- to N2, implying that AG': is
compute.:f For the rationale behind this assumption and -72.20 kJ/e- eq. The donor reaction is the same as above,
the use of terms in this equation, the original reference and AG," is 36.84 kJ/e- eq. Therefore,
should be consulted (20).
The fraction of electron donor (in terms of e- eq) used AG, = (-72.20 - 36.84)
for cell synthesis (f:) is computed as follows: = - 109.04 kJ/reaction
0 1 Using an E of 0.6 in equation 10, we can obtain the
f, =-l + A (9) value of A as follows:

where A is computed as follows: --1.75 13.5


A = - 0.6-' + -
Oh = 0.33
0.6(-109.04)
The value off: is computed using equation 9 as:
0 1
All the factors needed to compute A are related to the f, =-1+ 0.33 = 0.75
free energies associated with the donor, acceptor, or cell
synthesis half-reactions and the related efficiency of trans- This implies that :f which is equal to (1 - f:), is 0.25.
fer in the following manner. Figure 4 depicts the variability in :f for the organic and
inorganic donors listed in Tables 1 and 2 when combined
AGp is the difference between the free energy of the pyru- with a number of common electron acceptors. Several points
vate half-reaction and that of the carbon source. It is are worth noting including the following: (i) the value off:
equal to 113.8 kJ/e- eq for autotrophic reactions, i.e., is generally much lower for autotrophic growth than for het-
when COZ is, the carbon source and is computed as erotrophic growth; (ii) oxygen, iron, and nitrate have higher
(35.09 -AG," ) for heterotrophic reactions where donor :f than sulfate and C02,with manganese dioxide havingf:
and carbon source are the same. closer to the former set of acceptors; and (iii) certain chlori-
AG,, is the amount of energy needed to convert pyruvate to nated compounds, even when acting as donor with high-
cell carbon, estimated by McCarty (13). It also includes energy-yieldingacceptors, have relatively low :f values.
the effects of various nitrogen sources by using a differ-
ent value for each nitrogen source: 18.8 kJ/e- eq for 13.1.I 1. The Overall Stoichiometric Equation ( R )
NH4+,13.5 kJ/e- eq for NO3-, 14.5 kJ/e- eq for NOz-, The stoichiomteric equation, R, serves many needs. Using
and 16.4 kJ/e- eq for N2 . In all cases, the cell formula is this equation we can calculate (i) the amount of cells ex-
assumed to be C5H702N.Combining the effects of dif- pected from a given substrate, (ii) the amount of oxygen
ferent nitrogen sources into AG,, amounts to an as- or any other electron acceptor needed to accomplish the
sumption that electrons to reduce the oxidized nitrogen conversion, (iii) the amount of nitrogen required to make
sources come from pyruvate. sure only the donor is limiting and not other key nutri-
AG, is the energy available fro,m the electron donor-accep- ents, and (iv) the amount of other products generated, etc.
tor pair, i.e., AG: - AG," , Under standard conditions As mentioned earlier, the available electrons from the
and pH 7, this is the same as AG,"'. donor are apportioned into the energg-yielding reaction
E is the efficiency of energy transfer, assumed to be 0.6 (but (f:) and the cell synthesis reaction (f, ). For the purpose
it may vary fordifferent organisms and substrates). of writing the stoichiometric equation, it is easier to start
n is a factor that addresses the fact that some carbon sources with 1 e- eq of the electron donor and divide it into two
may actually release energy when converted to pyruvate portions: one portion is used by the acceptor to yield en-
and others may require energy. Thus, it is + 1 when AGp ergy, and the other portion is used in the cell synthesis re-
is positive and - 1 when AGOis negative. action to reduce carbon and nitrogen (if it is other than
NH4+/NH3)and synthesize the macromolecules. In other
The following example illustrates the use of the above words, the electron donor half-reaction (Rd) supplies 1 e-
equation for computing.:f eq, a fraction of which (f:) goes to the acceptor half-reac-
tion (R,) and the remaining fraction (f:) goes to the syn-
13.1 .I 0.1. Example 6 thesis half-reaction (R,). Using Rd, R,, and R,, this is writ-
You are interested in removing NO3- from groundwater ten mathematically as
using denitrifying bacteria and decide to use methanol,
which is also the carbon source, as the donor. Assume that R = :f R, + :f R, - R d (11)
no NH4+ is present, so NO3- is also the nitrogen source. To obtain R, the acceptor half-reaction is multiplied by
Compute the value off: expected for the denitrifying or- ,:f the cell synthesis half-reaction is multiplied by ,:f the
ganisms during the above process. donor half-reaction is multiplied by -1, and the three re-
First, we compute AG, usingAG,"' of the methanol half- sulting equations are added algebraically. The following ex-
reaction. ample illustrates the main steps.
298 H GROWTH

13.1 .I 1 . I . Example 7 The last term (8 g of COD in 1 e- eq of donor) is a


Write the overall stoichiometric equation for the oxidation constant which is true for all donors because 1 e- eq of
of methanol to C02 (methanol acts as an electron donor any organic donor requires 8 g of oxygen to be completely
and also as a carbon source), with NO3- as an electron ac- oxidized to C02.The above equation can be translated to
ceptor and also serving as the nitrogen source. Use the the following four equations, one for each nitrogen
value off: as 0.75 (thus, :f = 0.25) computed above. source.
The half-reactions needed to write the stoichiometric
equation are methanol to C02 half-reaction, NO3- to Nz
Y = 0.706 :f : NH4+as nitrogen source
half-reaction, and cell synthesis half-reaction with NO3- as = 0.504 f> : NO3- as nitrogen source
the nitrogen source. They are reproduced below:
= 0.543 :f : NO2- as nitrogen source
R,: 1/5 NO3- + 6/5 H+ + e- = 1/10 N2 + 3/5 HzO = 0.614 :f : N2 as nitrogen source
R,: 1/28 NO3- + 5/28 C02 + 29/28 H+ + e- Using the :f values for each nitrogen source and the above
= 1/28 C5H702N + 11/28 H2O
equations, the yield coefficient Y has the following rela-
Rd: 1/6 COz + H+ + e- = 1/6 CH30H + 1/6 H 2 0 tionship:
We multiply R, by 0.25 (f:), R, by 0.75 (f:), and R d by - 1; Ynirrace = (0.79 2 0.06) Yammnnium
rewrite the resulting equations; and add them as shown
below: Ynittate = (0.83 2 0.05)Yammonium
:f R,: 0.05 NO3- + 0.3 H+ + 0.25 e- Ydinitrogen = (0.91 2 0.05)Yammnnium
= + 0.15 H2O
0.025 N2 Thus, the yield coefficient Y for denitrifiers growing on
fR
: :, 0.027 NO3- + 0.134 C02 + 0.777 H++ 0.75 e- methanol with ammonium as nitrogen source will be ap-
= 0.027 C5H702N + 0.29 H2O proximately 0.72 (obtained by dividing the value of Y on ni-
trate, which is 0.57, by 0.79, the ratio between the yield co-
-Rd: 0,167 CH,OH + 0.167 H20 efficients for nitrate and ammonium). The above
= 0.167 COz + H+ + e- relationships were obtained by computing f> and Y for the
0.167 CH30H + 0.077 NO3- + 0.077 H+ = 0.025 N2 four nitrogen sources over a range of donors (those listed in
+ 0.033 C02 + 0.278 H2O + 0.027 C5H70zN (12) Tables 1 and 2) and acceptors (those shown in Fig. 4). It can
be used to obtain approximate values. These relationships
Note that no electrons remain in the final equation be- also highlight the effect of nitrogen source on cell yield.
cause 1 e- eq from the donor is exactly divided between the 2. Energy is also needed to maintain the cellular func-
synthesis and the energy-yielding reactions in the ratio tions. This is known as maintenance energy and comes from
0.75:0.25. the energy-yieldingreaction. It is modeled by increasing the
Equation 12 is the stoichiometric equation, R. It may :f by a fraction, m, and re resenting it by fe (thus, :f + m
be used to compute the quantitative relationship among B
= fe) and decreasing the fs by the same fraction and repre-
the reactants and products including cell mass. It is help- senting it by fs (thus :f - m = fs). The fraction m is com-
ful to first calculate the molecular mass of each reactant puted from an equation that is a function of the rate of cell
or product. Thus, the molecular mass of methanol is decay and cell age. The fraction m effectively reduces the
+ +
(1 x 12 4 x 1 1 x 16) = 32 and that of the cell of amount of donor used for cell synthesis by an amount that
+
the given formula is (12 x 5) + (1 X 7) (16 X 2) 14 + is equal to the amount of donor required for maintenance
= 113. Using the above equation, it can be computed that (20).
3.05 g (= 0.027 x 113) of cells are expected from 5.34 g 3. In the reduction half-reactions used in the above
(= 0.167 x 32) of methanol oxidized to C02. Expressed analysis, all reactants and products except H+ are at 1 M
as a ratio (of grams of cells produced per grams of substrate concentration. In practice, this is generally not the case; the
consumed), this is also known as the true yield coefficient reactants and products may be at concentrations other than
Y. A value of 0.57 for the yield coefficient Yon the basis 1 M, and their concentrations may be continuously chang-
of methanol is typical of the values observed for aerobic ing with time due to microbial activity. In most cases, the
and denitrifying processes. Equation 12 also indicates that change in AGO due to a change in reactant or product con-
for each 5.34 g of methanol oxidized, 4.77 g of NO3- centration will not be significant enough to change the
(0.077 X 62) or 1.08 g of N03--N (0.077 X 14) will be conclusions about the potential for growth. However, in
consumed. certain instances, it is possible that the reaction is thermo-
dynamically favorable (meaning negative AGO ) under one
Notes: set of reactant and product concentrations but unfavorable
1. The true yield, Y in grams of cells per gram of organic (meaning positive AGO) under another. The effect of
matter expressed as chemical oxygen demand can also be change in concentration is taken into account by the
computed from :f using the formula: Nernst equation (20) whose general form is
Y = :f (M, g of cells/mol of cells)/ AG = AGO' + RT In Q (13)
[(n,e- eq/mol cells)(8 g of COD/e- eq donor)]
where R, T, and Q are the gas constant (8.314 J mol-'
where COD is chemical oxygen demand. It is defined as K-'), temperature (in kelvin), and reaction quotient, re-
the amount of oxygen required to oxidize 1 g of any spectively. The reaction quotient, Q, depends on the reac-
substrate, M, is the empirical formula weight of cells (113 tion of interest (20).
for C5H702N),and n, is the number of e- eq per mol 4. If the source of nitrogen is other than those mentioned
of cells (20 for NH4+, 28 for NO3-, 26 for NO2-, and 23 above (NH4+,NO3-, NOz-, and N2), a separate cell syn-
for N2). thesis equation may be written corresponding to that source
13. Energetics, Stoichiometry, and Kinetics 299
-
I
I
--I--
n no:
Glucose
Formate d
7
ii?
7
Acetate

Dichlorophenol - / en

-
Vinyl Chloride-

lonor i d SI ate a cept

0.00 0.05 0.10 0.15 0.20 0.25 0.30 0.35 0.40 0.45 0.50 0.55 0.60 0.65 0.70 0.75 0.80

fS0

FIGURE 4 Trends in the computed value off: as a function of selected electron acceptors and
electron donors for NH4+ as the nitrogen sources and C5H702N as the cell formula. (Part 1)
Organic donors (heterotrophy); (Part 2) Inorganic donors (autotrophy).

of nitrogen. Similarly, the cell synthesis equation R, in the balance. Experimentally, it is of course critical to ensure that
above analysis uses C5H702Nas the cell formula. Significant these elements are present in excess.
variability in cell formula exists due to the variability in the
type of microorganism and growth substrates. If a better esti-
mate for the cell formula is known and such a theoretical 13.2. KINETICS OF MICROBIAL GROWTH
analysis is still useful, writing a new R, corresponding to the A single cell grows into two cells with the input of raw ma-
known cell formula may be more appropriate. terials (carbon, nitrogen, phosphorus, nutrients, etc.), elec-
5. The effect of source of phosphorus, sulfur, and other trons, and energy. For the purpose of modeling, it is impor-
trace elements on electron donor requirement is neglected tant to assume that only one of the raw materials is limiting,
in this analysis. Phosphorus constitutes only up to 2.5% of called the limiting substrate (or just substrate) and that all
the cell mass under normal conditions, and most of it is pres- other required substrates and trace elements are in excess. In
ent as phosphate, the oxidized state of phosphorus. Sulfur practice, it is possible that more than one substrate is limit-
and other trace elements which may be present in a reduced ing. Such a scenario requires a more complex modeling ap-
state constitute even lower fractions of the cell mass. These proach that is outside the scope of this chapter. The limiting
are generally excluded from the quantitative analyses related substrate is generally the electron donor, but it could be any
to assess the need for electrons and the electron donor be- one of the raw materials. The concentration of this limiting
cause the complexity of equations is increased without sig- substrate can be represented as S (in milligrams per liter) at
nificant improvement in the energy, electrons, and material any time t, while the initial concentration can be represented
300 GROWTH

3r ar I as i .on d lor a
:cep 0.22

0.00 0.05 0.10 0.15 0.20 0.25 0.30 0.35 0.40 0.45 0.50 0.55 0.60 0.65 0.70 0.75 0.80

fS0

FIGURE 4 (Continued)

as So at the start of the reaction, to. The initial concentration types of substrates (e.g., in the case of aerobic bacteria uti-
of cells can be represented as (in milligrams per liter) lizing complex carbon compounds as the donor substrate
while the cell concentration at any time t can be represented and oxygen as the acceptor substrate, only one of them will
as X. Some materials are utilized during the time the mi- be assumed to be limiting either the donor or the acceptor).
croorganisms grew, implying the existence of a rate at which Thus, the growth of a single species on a single substrate be-
substrate was utilized. This is called the rate of substrate uti- comes a special case of the more complex situations. Note
lization, rsu.The concentration of the substrate (S), biomass also that there are some special cases that require additional
(X), change in the concentration of substrate (a), change in steps to obtain appropriate modeling equations. These in-
the concentration of biomass (a), change in time (dt), rate clude diauxic growth, synchronous culture growth, and ef-
of change of biomass concentration (dX/dt), and rate of fect of toxic compounds. When a single species is growing
change of substrate concentration (&/dt) need to be related on multiple substrates, there is a chance that the substrates
to each other for a certain system (reactor vessel, plate will become limiting one after another; this is called diauxic
colony, biofilm, etc.) in order to predict a target unknown pa- growth, the modeling of which requires additional consid-
rameter when all others are known. erations beyond the scope of the current discussion.
Such predictive equations can be written for a single Similarly, there is also a scenario in which the growth of the
substrate, a group of microorganisms using a similar sub- cells is synchronized to divide at the same time. This is
strate (known as a guild, e.g., in fermentation with mixed known as synchronous culture growth and has to be mod-
population), or a collection of guilds each utilizing a differ- eled differently from the growth discussed above, which is
ent substrate but together performing an overall function for asynchronous growth. These exercises are introductory
(e.g., in anaerobic digestion). Often the same approach is because most models may need to be developed much be-
applied for modeling even if a large number of different yond what is presented here. Many books and journal arti-
types of microorganisms utilize a collection of different cles are available on this subject.
13. Energetics, Stoichiometry, and Kinetics 301

It so happens that a batch system is easier to operate but unit amount of biomass and having dimensions of recipro-
more complex to model. The reason is that in a batch sys- cal time (l/t)I

tem, substrate concentration keeps changing. Figure 5 If p is constant, integration of equation 14 shows that X
shows that the specific growth rate ( p ) is a function of the will increase exponentially with time as follows:
substrate concentration (S). Thus, in a batch system, unless
the substrate is in great excess, microorganisms grow at x
In -= p t
different specific growth rates because S is changing with x
0
time. To calculate the total amount of growth (and thus
the concentration of biomass after time t) or the new sub- where In is the natural logarithm (to the base e = 2.303)
strate concentration after some of it has been utilized, the and X o is the amount of biomass when t = 0.
total growth needs to be integrated mathematically. This is It follows by rearrangement of equation 15 that the final
in contrast to a chemostat where a single substrate concen- biomass concentration X is
tration, S, exists at all times, resulting in a constant specific X = Xoepf (16)
growth rate ( p ) at all times, no integration is required, and
the analytical solution is much simpler. Bacterial growth that follows this relationship is called ex-
The techniques used to measure growth monitor average ponential or logarithmic growth.
values and obscure the fact that all bacterial cultures are The relationship between the biomass doubling time (td)
grossly heterogeneous. They describe the growth of a popu- and the specific growth rate is found by letting X = 2x0 at
lation rather than of individual cells. Although all members t = td in equation 15 and solving the equation as follows:
of a particular population may be genetically identical, the In 2 0.693
individual members vary with respect to doubling time, age, t --=- (17)
d -
composition, metabolic characteristics, and size. The mag- c1 CL
nitudes of these variations are influenced by the environ- Equations 14 to 17 predict the growth of bacteria in sim-
ment and can often be minimized by careful design and de- ple systems in which the factors influencing growth are con-
velopment of the system for cultivation. Most of the systems stant but do not allow prediction of deviation from constant-
described here are designed to minimize heterogeneity growth (steady-state) conditions. The techniques for
within the population of a pure culture. continuous cultivation very nearly establish steady-state
In such systems, the growth behavior of a bacterial pop- conditions of theoretically infinite duration, whereas the
ulation can be predicted by the simple relationship: techniques for batch cultivation allow significant changes
ax
-=px
in the environment during the time course of cultivation.
In batch culture, equations 14 to 17 will apply without ad-
dt justment to the value of p only during the portion of the
where d X is the increase in amount of cell mass (biomass), growth cycle in which the changes in the growth environ-
dt is the time interval, X is the amount of biomass, and p is ment have n o influence on population growth (i.e., during
the specific growth rate, representing the rate of growth per the exponential growth phase).

Pmax

K S Substrate concentration, s
t
Concentration at which the sp. growth is half the maximum

FIGURE 5 Specific growth rate and half velocity constant, the basis for Monod kinetics.
302 GROWTH

13.2.1. Modeling Batch Systems ume of fresh medium may result in outward diffusion of vi-
tamins, cofactors, and ions which are required for many in-
13.2.1 . I . Principles tracellular enzyme activities. If cells are inoculated from a
A batch culture system is one in which nothing (with the rich medium to a minimum medium, the lag time may be af-
frequent exception of the gas phase) is added to or removed fected by inoculum size as a result of carryover of trace nu-
from the culture vessel after a medium of appropriate com- trients from the original medium.
position is inoculated with living cells; this is called a closed The age of the inoculum influences the lag in a fresh
system. It follows that a batch system can support cell mul- medium as a result of toxic materials accumulated and es-
tiplication for only a limited time and with progressive sential nutrients depleted within the cells during their prior
changes in the original medium and environment. growth. Both a positive and a negative effect on the length
of the lag phase in fresh medium can occur. In general, an
13.2.1.1.1. Normal Growth Cycle increasing inoculum age lengthens the lag phase, especially
Figure 6 shows an idealized normal growth cycle (curve) when cells are inoculated from a nutritionally complex
for a simple, homogeneous batch culture of bacteria. The medium into a simple one.
normal growth cycle assumes asynchronous binary fission of Finally, changes in nutrient composition and concentra-
the cells. Growth proceeds through a lag phase, during tion between the inoculum culture and the fresh medium
which cell mass increases but cell numbers do not, and into may trigger the control and regulation of enzyme activities
a growth phase, which usually is characterized by an expo- within the cells or of morphological differentiation, such as
nential increase in numbers and follows the relationships of spore formation. If the cells are transferred from a simple
equations 14 to 17. Ultimately, changes in the chemical or medium to a richer medium, both time and nutrients will be
physical environment result in a phase of no net increase in expended to allow an increase in the concentration of en-
numbers, the maximum stationary phase. Cells in the sta- zymes essential for metabolism. When cells are inoculated
tionary phase still require an energy source for the mainte- from a rich to a less rich medium, the cells may resume ex-
nance of viability. If the availability of an energy source in ponential growth immediately but at a lower rate.
a batch culture is limited, there ensues a death phase, which The fact that constant exponential growth can occur for
is often characterized by an exponential decrease in the even a limited time in batch culture shows that the growth
number of living cells. rate can be virtually unaffected by changes in substrate con-
The lag phase may be brought about by the shock of centrations over wide ranges. Under these conditions the cul-
rapid change in the culture environment. In fresh medium, ture is said to be in balanced growth, whose rate can be de-
the length of the lag phase depends on the size of the in- scribed by a single numerical value, p.Eventually, the culture
oculum, the age of the inoculum, and the changes in nutri- will deviate from constant exponential growth and can no
ent composition and concentration experienced by the longer be described only by the value of p, even though it
cells. A small volume of inoculum transferred to a large vol- is possible to calculate this value for the case of limitation

Lag phase Growth phase Maximum stationary phase Death phase


4 H

/-

Time
FIGURE 6 Idealized normal growth cycle for a bacterial population in a batch culture system.
13. Energetics, Stoichiometry, and Kinetics 303

by a single nutrient substrate. In a classic article (15), the growth cycle if growth is determined by optical mea-
Monod described the relationship between substrate con- surement. For example, stationary-phase bacterial cells are
centration and bacterial growth in simple systems at steady often more transparent than those growing in the exponen-
state as follows: tial phase. As a result, a growth curve plotted as the loga-
rithm of absorbance versus time may show an apparent de-
crease in stationary-phase cell concentration compared
with that attained at the end of exponential growth. In this
case, the aberrant growth cycle represents a change in the
where p is the specific growth rate, pmax is the maximum
morphological characteristics of the cell rather than a
value of p obtained when S is much greater than K,, and K,
change in the number of cells.
is the saturation constant equivalent to a Michaelis-
Growth curves plotting the log of cell number versus time
Menten constant. K, is the substrate concentration at
occasionally show an unusually rapid increase in cell number
which the specific growth rate is one-half the maximum
just after the lag phase, which then settles into a slower
growth rate, pmax. S is the instantaneous or steady-state sub- increase. The unusual burst of growth may in fact be an indi-
strate concentration of a single limiting nutrient. Both K,
cation of partial or complete culture synchrony. This syn-
and S have the units of milligrams per liter (or mass per unit
chronized growth may result, for example, from culture ac-
volume) because both are concentrations.
climation to a new nutritional environment. Such culture
Several alternative models for growth response to sub-
synchrony degenerates rapidly, and asynchronous growth usu-
strate concentration exist (15). Equation 18 may be used to
ally dominates within two generations. However, special tech-
describe growth response to substrate limitation only under
niques have been devised to synchronize cell divisions in a
steady-state conditions with uncomplicated bacterial sys-
growing population as a way to mimic individual cell growth.
tems. When exponential growth ceases as a result of sub-
Arithmetic (linear) growth occurs when the supply of a
strate limitation, conditions are no longer steady state for
critical nutrient is regulated by an arithmetic process (such
cell mass accumulation or substrate concentration, and
as by dropwise addition or by diffusion) and this process
equation 14 must be used with equations 18 and 19 to ade-
becomes limiting. For example, limited diffusion of air
quately model the response of the culture to diminishing
through the cotton plug in a test tube or of nutrients
substrate. Simultaneous solution of equations 14, 18, and 19
through a membrane in a dialysis culture (chapter 12) may
yields a somewhat bulky model for batch growth, which is
cause a shift from exponential to arithmetic growth.
at best a rough estimation for very simple systems. This is
Although most bacterial cultures reproduce by binary fis-
why models of batch reactors are more complex than those
sion as individual cells, some (e.g., actinomycetes) repro-
of chemostats.
duce as filamentous extensions. If growth occurs primarily
The maximum population in a batch culture can be es- through extension of the tips of the hyphae, the increase in
timated from experimental data relating the increase in cell
cell mass with respect to time will be arithmetic.
number or mass to the corresponding decrease in substrate
Filamentous bacteria often grow as pellets in liquid cul-
concentration (assuming that concentration of substrate is
ture; when this occurs, biomass increases more slowly than
the only thing that limits growth):
the classical exponential rate and is proportional to the
cube of time. Microbial growth in pellets may be severely af-
fected by diffusion of nutrients into the pellet and by diffu-
sion of metabolic products out from the pellet, a possibility
where X and S are the cell and substrate concentrations at that is not taken into account by equation 18.
time t, Xo and So are the cell and substrate concentrations, Bacterial cells in complex environments often metabo-
respectively, at an earlier time, to, and Y is the overall yield lize usable substrates in a sequential manner. That is, the
coefficient. Equation 19 accurately describes the relation- presence of certain substrates may lead to repression of the
ship between cell concentration and substrate concentra- enzymes for metabolism of other substrates. In this instance,
tion during exponential growth. If exponential growth con- only when the concentration of the repressing substrate has
tinues unabated until the stationary phase is reached and been reduced through bacterial consumption can the en-
the substrate is completely consumed during exponential zymes for metabolism of other substrates be elaborated. This
growth, the maximum cell number or concentration will be regulation of bacterial physiology leads to an aberrant
given by equation 19. This estimation assumes that the growth cycle that shows one or more intermediate but tran-
yield coefficient is constant throughout the growth cycle sient stationary phases. This response to a changing envi-
and neglects substrate consumption during the lag and sta- ronment is termed diauxic growth. A classical example of
tionary phases. In fact, the yield coefficient cannot be as- diauxic growth is that of E. coli growing in the presence of
sumed to be constant for other than constant exponential both glucose and lactose (Fig. 7). Rapid growth on glucose
growth conditions at a single specific growth rate. It follows occurs first. At the point of glucose exhaustion, an inflec-
that the prediction of a maximum population from equation tion in the biomass curve occurs and there may even be a
19 will lead to an overestimation. The concept of yield co- decline in biomass. A new enzyme system for metabolism of
efficient is discussed in greater detail later in this chapter. lactose is induced during this lag, and biomass accumulation
Prediction of the time required to attain maximum popula- at the expense of lactose then continues.
tion density in batch culture requires simplifying assump-
tions (1, 5). 13.2.2. Continuous-Culture Systems Modeling
Continuous cultivation differs from batch cultivation in
13.2.1.1.2. Aberrant Growth Cycles that a fresh supply of medium is added continuously at the
Aberrations from the normal batch growth cycle are same rate that culture is withdrawn (an open system). If
common. Morphological change in the culture (such as an the culture is well mixed, a sample that is representative of
increase in opacity, cellular refractive index, individual cell both the cell population and the substrate concentration
size, or cell aggregation) can lead to an apparent change in within the fermentor can be withdrawn. The technique of
304 w GROWTH

Time v

FIGURE 7 Idealized diauxic growth of a bacterial population (biomass) in a batch culture system
with two usable substrates (glucose and lactose).

continuous cultivation theoretically allows continuous ex- feed in grams per liter (Xois generally zero for most labora-
ponential growth of the culture in a system requiring con- tory scale systems, but for the sake of completeness of mass
stant addition of fresh medium and constant withdrawal of balance, it is included here); X is the cell concentration in
culture so that the culture volume and cell concentration the fermentor, again in grams per liter; p is the s ecific
remain constant with time. In its broadest sense, continu- growth rate, b is the specific death rate (in hours- P), and
ous cultivation does not require a constant cell concentra- dX/dt is the rate of change in cell mass (in grams per liter-
tion, but most literature accounts of quantitative study of hour).
continuous cultivation have dealt with such a situation At steady state, dX/dt = 0. The volume of a true con-
and the principles developed below deal only with this sit- tinuous culture is fixed in theory and undergoes negligible
uation. variation in practice. Therefore, the flow rates to and from
The term steady state is often applied to continuous the fermentor must be identical. Finally, the specific death
cultivation and means literally that no change in status oc- rate is almost always much lower than the specific growth
curs during the time span studied. In reality, this definition rate, so the death term ( b X ) may be ignored. (Exceptions
is too broad, since practical application of the continuous- to this rule may occur at very low growth rates, in the pres-
cultivation theory often results in changes in some parame- ence of toxic substances, or in an extreme biophysical en-
ters while others remain constant. A continuous culture is vironment.) Since the feed to the fermentor is usually
therefore defined at steady state by an invariant biomass sterile such that X o = 0, at steady state, the following
concentration with respect to the time span of observation. applies:
In contrast, a batch culture may have a steady-state dis-
solved oxygen concentration maintained by constant re- Q
placement, but it is not a continuous culture because the
biomass concentration changes with time.
p=v=D
Continuous cultivation at steady state is possible only That is, the specific rate of growth of the population within
when all factors contributing to the accumulation of bio- the fermentor is determined by the dilution rate, D, where
mass are exactly balanced by all factors contributing to the D = Q/V.
loss of biomass from the system. This is shown by the fol- Many types of continuous cultures are possible (1, 5).
lowing general material balance on the bacterial cells: The following discussion is limited to one of the most
widely used types of continuous culture, the chemostat,
Cells accumulated within the system = cells added to the which achieves steady state by precise control of the avail-
+
system - cells removed from the system cells produced ability of a single growth-limiting substrate. The mathemat-
through growth - cells consumed through death ical principles underlying the chemostat also apply to the
The balance is shown mathematically as follows: turbidostat, which achieves steady state by actually remov-
ing the cell mass and replacing it with fresh medium at the
same rate as cell growth. However, turbidostats are better
suited to continuous-cultivation studies of growth at or near
the maximum specific growth rate, pmax.
where Q is the medium flow rate to and from the fermen- Although many references on continuous cultures now
tor (in liters per hour); V is the liquid volume within the exist, background reading on continuous culture should
fermentor (in liters); X o is the cell concentration in the start with the classic papers published in 1950 by Monod
13. Energetics, Stoichiometry, and Kinetics 305

(16) and Novick and Szilard (18). Luedeking (9) has also Substitution of equation 21 into equation 23 gives
presented a very useful discussion of continuous-culture
theory and practice. The article describes graphical design x = Yx/s(so- S) (24)
and analysis of continuous-culture systems, avoiding the re- Note that equation 24 was presented earlier without der-
quirement for an accurate model of growth response to ivation for use with batch systems (equation 19). The over-
changing environmental properties (such as the Monod all growth yield is assumed to be dependent only on the lim-
model). A n empirical approach to continuous cultivation iting nutrient concentration and independent of specific
based on batch data has its pitfalls, which were discussed by growth rate. Equations 21 and 24 are the steady-state equations
Luedeking; however, the approach is still useful for obtain- for usual chemostut continuous cultivation.
ing approximate design criteria. A model expressing the specific growth rate as a func-
tion of substrate concentration must be assumed before bio-
13.2.2.1. Chemostat mass concentration, substrate concentration, and specific
Deviations from simple chemostat theory abound, but most growth rate can be related to define a stable set of operating
are the result of (i) product formation, which may require a conditions. Equation 18 provides an adequate model for
sophisticated model of yield as a function of p; (ii) imper- many situations. Substitution of equation 21 into equation
fect mixing in the fermentor, which may allow a stable di- 18 yields
lution rate that is higher than the critical dilution rate; (iii)
wall growth, which has an effect similar to imperfect mix-
ing but is more pronounced; and (iv) idiosyncrasies of phys-
iological response. Pirt (19) has discussed the first three of
these deviations. where D, is the critical dilution rate corresponding to the
In its simplest form, chemostat continuous cultivation maximum specific growth rate, pmax. Operation of the chemo-
can be described as a collection of reaction steps in which stat at dilution rates above D, will result in complete washout of
the rate of growth of a culture is determined by the lowest the culture.
rate of nutrient metabolism. Although a growing culture Equation 25 can be arranged to give
requires the metabolism of many different nutrients, the
growth rate of an ideal culture at any given instant is de-
termined by the rate of metabolism of a single limiting nu-
trient. Monod (15, 16) found that growth rate dependence Substitution of equation 26 into equation 23 results in
on substrate concentration could be predicted by an equa-
tion whose form is essentially a Michaelis-Menten-type
function, as shown in equation 18. Monod's equation is
most easily applied to experimental conditions of steady
x = Y,/, so- Z ( ~

D )
state. For usual purposes, a steady state exists when the sub- Equation 27 relates the steady-state biomass concentra-
strate concentration does not fluctuate with time and the tion to the dilution rate. Figure 8 depicts the generalized
resulting cell population in the continuous-culture vessel is system response to changes in the dilution rate, as predicted
constant with time. When a chemostat is operated with a by equations 26 and 27.
sterile feed (Xo= 0) and without recycle, the specific
growth rate is numerically equal to the dilution rate (equa-
tion 21). This identity is forced by controlling the avail-
ability of a limiting nutrient through the addition of fresh
Biomass
medium.
A limiting nutrient balance can be written for a chem-
ostat:
input - output - consumed = accumulation
The mathematical expression for this, similar to equation
20 for cell mass, is as follows:

ds = DSo - DS - -
- clx
dt YX,$
where D is the dilution rate (D = QW);So and S are the
limiting substrate concentrations in the feed and chemo-
stat, respectively; p is the specific growth rate of the culture
in the chemostat; X is the cell mass (dry weight) in the
fermentor; YxIs is the overall yield coefficient (cells
formed/substrate consumed); and dS/dt is the rate of change
of substrate concentration in the fermentor. The overall
yield term is a composite that includes contributions for
both growth and maintenance. No product? other than '.* .................................... ..*...
cells are assumed. If product formation does occur, an addi-
tional consumption term must be added. At steady 'state, Dilution rate Washout
equation 22 then becomes
FIGURE 8 Generalized effects of changes in the dilution rate
on culture variables in a chemostat system for continuous culti-
vation, where the dilution rate approaches
, , , ,p at washout.
306 w GROWTH

The Monod model (equation 18) is one of several mod- growth. This can be easily accomplished by ensuring that all
els relating growth rate and substrate concentration. The nutrients, other than the one on which yields will be based,
assumptions implicit with this model are quite simple and are in excess. An insight into this medium design can be ob-
do not adequately describe all systems. An excellent discus- tained from an elemental analysis of the bacteria to be
sion of the fundamental theory of chemostat continuous grown in the chemostat. The medium must supply all of the
culture is presented by Tempest (23) components of the cell and can therefore be based on ele-
mental analysis. The ultimate level of growth will be deter-
13.2.2.2. Growth Yield Calculations mined by the concentration of the limiting nutrient. Since
The concept of growth yield is developed from a material cell concentration in a chemostat will automatically adjust
balance of a stable cultivation system. Monod originally de+ to match the rate of limiting-nutrient addition, the ratios of
fined the yield constant, Y, in terms of mass units: medium components are more important than their ab-
solute amounts. Media for the determination of overall
grams (dry weight) of cells formed growth yield should therefore be designed such that the ra-
Y= (28)
grams of substrate consumed tios of elements supplied by the medium to the element
The yield from substrate is often indicated by Yds, indi- chosen as the limiting nutrient are considerably higher than
cating a ratio of the respective concentrations. The con. predicted by taking the ratio of elements from elemental
vention of mass ratios for expressing the yield of cells on the analysis.
basis of carbon substrate consumption is fairly well ac- The calculations described below require the establish-
cepted. Yields can be easily calculated by determining the ment of steady-state conditions in terms of cell concentra-
production of cell mass (dry weight) over a given period of tion and substrate concentration. Make sure that this con-
the cultivation and dividing by the mass of carbon substrate dition is met by monitoring them both over a long enough
consumed during the same period. period to allow displacement of one reactor volume of
If the yield is to be expressed on the basis of ATP pro- medium. Whenever conditions are changed (such as in-
duced or oxygen consumed during growth, the cell yield can creasing or decreasing the rate of medium addition to a
be defined as follows: chemostat), allow sufficient time for a return to steady state.
This can be ensured by allowing at least four turnovers of
grams (dry weight) of cells formed fermentor liquid volume. Since a continuous reactor oper-
YX,.4TP = (29) ates on a dilution principle, a step of pulse change in oper-
moles of ATP formed
ating conditions would cause deviation from the preexistent
- grams (dry weight) of cells formed steady state. This would approach a new steady-state value
- (30)
yx/oz moles of oxygen consumed at a rate predicted by the dilution of the culture; that is, four
mean residence times would be necessary for a close asymp-
Under many conditions, the yield with respect to ATP totic approach to the new steady-state equilibrium value.
synthesis or oxygen consumption is relatively constant for For example, consider a 2-liter chemostat in which the rate
many organisms. For example, the yield of cells per mole of of medium addition and withdrawal is 0.5 liter/h with a di-
ATP synthesized under conditions of energy substrate limi- lution rate of FIV = D = 0.25 h-I. The time necessary for
tation and fairly high growth rate is approximately 10 2 2 four turnovers of medium volume is calculated by dividing
g of cells (dry weight) formed per mol of ATP (2). 4 by the dilution rate. In this case, the reactor medium
However, the yield of cells per mole of ATP is not con- volume will have been completely replaced four times after
stant for all bacteria and is generally variable if the energy 16 h (4/0.25 h-'). That is, at least 16 h must elapse after
substrate does not limit growth or if the growth rate of the changing the growth conditions of a continuous culture
culture is significantly lower than the maximum rate. Also, prior to the achievement of steady-state conditions.
carbon and energy sources may be consumed not only for In reality, the time necessary to obtain a close asymp
cellular growth but also for product formation. Furthermore, totic approach to the new steady-state value may be de-
energy substrates are involved in cell maintenance as well pendent on the way in which operating conditions have
as in cell growth. Since cells can use energy substrates for been changed. If the change in operating conditions in-
endogenous respiration without growth, maintenance en- volves a nutritional step-down, for instance, four mean res-
ergy requirements will contribute to the consumption of idence times should be adequate. If the change involved a
energy substrates without a concomitant increase in cell dry nutritional step-up under conditions of rapid growth, the
weight. Although the influence of endogenous metabolism system may require more than four mean residence times to
on the calculation of cell growth yield is minor when the achieve steady state. In the latter case the rapidly growing
culture growth rate is near its maximum value with the en- cells may require elaboration of new enzymes for metabo-
ergy substrate as the limiting nutrient, consumption of en- lism of the added nutrients.
ergy substrates for maintenance can be quite significant at Once steady-state continuous cultivation is achieved,
growth rates lower than the maximum or in an extreme the overall growth yield is determined as shown below:
growth environment.
Since meaningful calculation of yield values requires (X- X o )-
-
grams (dry weight) of cell produced
careful and precise control of the cultivation conditions, Yxl, = (31)
(So - 3) grams of substrate used
yield data are best obtained from continuous-culture sys-
tems. In this way, the growth-limiting substrate and the spe- where X is grams of cells (dry weight) per liter of effluent
cific rate of growth, p, can be specified and controlled. medium, Xo is grams of cells (dry weight) per liter of influ-
Calculation of the overall growth yield from continuous- ent medium (usually zero), S is grams of substrate per liter of
culture data assumes steady-state substrate and cell concen- effluent medium, and So is grams of substrate per liter of in-
trations and therefore is most easily accomplished in a fluent medium. If the medium added to the fermentor is
chemostat. The medium for chemostat operation must be sterile, the value of Xo is zero and the yield becomes Yds =
designed so that a single nutrient limits the rate of cell W S O - S).
13. Energetics, Stoichiometry, and Kinetics 307

To determine the overall growth yield, first determine system divided by total biomass wasted daily still applies.
the cell dry weight by quantitatively filtering a precisely It is also important to note that at steady state, the total
measured volume of culture effluent through a preweighed biomass wasted must be equal to the total biomass being
0.2-pm-pore-size membrane filter. Wash the collected cells produced, or else biomass accumulation or washout will
with buffer or distilled water to remove excess medium. Dry occur.
the membrane filter and cells to constant weight in an oven
whose temperature is not greater than 105C. Lower oven
temperatures may be required to prevent extensive brown- 13.3. REFERENCES
ing and therefore weight loss of the sample. Caution: many 1. Bailey, J. E., and D. F. Ollis. 1986. Biochemical Engineering
filters lose weight to a variable extent after washing and dry- Fundamentah, 2nd ed. McGraw Hill Book Co., New York,
ing. Therefore, several samples should be filtered NY.
and weighed, and several filters should be washed, dried, and 2. Bauchop, T., and S. R. Elsden. 1960. The growth of
weighed as controls. It is more accurate to centrifuge micro-organisms in relation to their energy supply. 1.Gen.
and wash cells as described above from a larger volume of Microbiol. 23 :45 7-469.
medium and then to resuspend in a small volume of water, 3. Criddle, C. S., L. M. Alvarez, and P. L. McCarty. 1991.
dry, and weigh. Microbial processes in porous media, p. 639-691. In J. Bear
Overall growth yield is related to maintenance and and M. Y. Corapcioglu (ed.), Transport Processes in Porous
Media. Kluwer Academic Publishers, Dordrecht, The
growth requirements for limiting substrates that act as en- Netherlands.
ergy sources according to the following equation: 4. Dolfing, J., and J. M. Tiedje. 1987. Growth yield increase
linked to ATP production and growth in an anerobic bac-
terium, strain DCB-1. Arch. Microbiol. 153:264-266.
5. Drew, S. W. 1981. Liquid culture, p. 151-178. In P.
Gerhardt, R. G. E. Murray, R. N. Costilow, E. W. Nester,
where p is the specific growth rate (in reciprocal hours), m W. A. Wood, N. R. Krieg, and G. B. Phillips (ed.), Manual
is the specific rate of substrate uptake for cellular mainte- of Methods for General Bacteriology. American Society for
nance (in reciprocal hours), Yds is the overall yield, and YG Microbiology, Washington, DC.
is the growth-specific yield. The values of m and Yc can be 6. Herbert, D. 1976. Stoichiometric aspects of microbial
estimated by plotting l/Yds versus l/p. (Remember that p growth, p. 1-30. In A. C. R. Dean, D. C. Ellwood, C. G. T.
= D only for simple continuous culture without cell recy- Evans, and J. Melling (ed.), Continuous Culture 6:
cle.) If the data form a straight-line relationship, the inter- Applications and New Fields. Ellis Horwood Ltd.,
cept will be the value of 1/YG and the slope of the line will Ch-ichester, England.
be the value of p. 7. Hoeft, S. E., T. R. Kulp, J. F. Stolz, J. T. Hollibaugh, and
Although growth yields are conventionally expressed in R. S. Oremland. 2004. Dissimilatory arsenate reduction
terms of mass of cells formed per mass or moles of substrate with sulfide as electron donor: experiments with Mono
used, this mixture of terms is often confusing. Herbert (6) lake water and isolation of strain MLMS-1, a chemoau-
has suggested that yields should be expressed in terms of totrophic arsenate respirer. Appl. Enuiron. Microbiol. 70:
gram-atoms of cellular carbon formed per gram-atom of el- 2741-2747.
ement consumed from the limiting substrate. Adoption of 8. Holliger, C., D. Hahn, H. Harmsen, W. Ludwig, W.
this convention would standardize reporting procedures and
Schumacher, B. Xndall, F. Vazquez, N. Weiss, and A. J.
B. Zehnder. 1998. Dehalobacter restrictus gen. nov. and sp.
eliminate much of the confusion in interpreting yield data. nov., a strictly anaerobic bacterium that reductively
Expression of growth in terms of gram-atoms of cellular car- dechlorinates tetra- and trichloroethene in an anaerobic
bon formed does not require a determination of the com- respiration. Arch. Microbiol. 169:313-321.
plete elementary composition of the cells. Rather, only the 9. Luedeking, R. (ed.). 1976. Fermentation Process Kinetics,
carbon content of the cells need be determined to allow use vol. 1. Academic Press, Inc., New York, NY.
of this convention. 10. Madigan, M. T., J. M. Martinko, and J. Parker. 2002.
Brock Biology of Microorganisms, 10th ed. Prentice Hall,
13.2.2.3. Mean Cell Residence Time, Upper Saddle River, NJ.
8, Versus Dilution Rate, D 11. Mayrn6-Gatel1, X., Y. Chien, J. M. Gossett, and S. H.
Mean cell residence time signifies the amount of time the Zinder. 1997. Isolation of a bacterium that reductively
microorganisms spend in a given vessel, on an average. It is dechlorinates tetrachloroethene to ethene. Science
represented by 8 and is defined as the total biomass in the 276:1568-1571.
12. Maym6-Gatell, X., I. Nijenhuis, and S. H. Zinder. 2001.
system divided by the total biomass wasted daily, intention-
Reductive dechlorination of cis-1,2-dichloroethene and
ally or otherwise. Mathematically, assuming Q as the flow vinyl chloride by Dehalococcoides e thenogenes. Environ.
rate in liters or cubic meters per day, X as the biomass con- Sci. Technol. 35516-521.
centration in milligrams per liter, and V as the liquid vol- 13. McCarty, P. L. 1971. Energetics and bacterial growth. In
ume of the vessel in liters, the total biomass in the system S. D. Faust and J. V.Hunter (ed.), Organic Compounds in
can be represented as V * X and the total biomass wasted Aquatic Environments. Marcel Dekker, New York, NY.
daily can be represented by Q * X.Thus, 14. McCarty, P. L. 1997. Microbiology: breathing with chlo-
rinated solvents. Science 276: 1521-1522.
e=- vx (33) 15. Monod, J. 1949. The growth of bacterial cultures. Annu.
QX Rev. Microbiol. 3:371-394.
16. Monod, J. 1950. La technique de culture continue: theo-
which for chemostat, is the same as V/Q or the inverse of rie et applications. Ann. Inst. Pasteur (Paris) 79:390-
D (because D is Q/V). When cell mass is recycled or 410.
added to a vessel, then the equation takes a different 17. Neidhardt, F. C., R. Curtiss 111, J. L. Ingraham, E. C. C.
form, although the basic definition of total biomass in the Lin, K. B. Low, B. Magasanik, W. S. Reznikoff,
308 GROWTH

M. Riley, M. A. Schaechter, and E. H. Umbarger (ed.). 21. Smidt, H., and W. M. de Vos. 2004. Anaerobic microbial
1996. Escherichia coli and Salmonella: Cellular and Mokc- dehalogenation. Annu. Rev. Microbiol. 58:43-73.
ular Biology. ASM Press, Washington, DC. 22. Sun, B. L., B. M. Griffin, H. L. Ayala-del-Rio, S. A.
18. Novick, A,, and L. Szilard. 1950. Experiments with the Hashsham, and J. M. Xedje. 2002. Microbial dehalores-
chemostat on spontaneous mutations of bacteria. Proc. piration with l,l,l-trichloroethane. Science 298:1023-1025.
Natl. Acad. Sci. USA 36:708-719. 23. Tempest, D. W. (ed.). 1970. The Continuous Cultivation of
19. Pirt, J. S. 1975. Principles of Microbe and Cell Cultivation. Microorganisms. I . Theory of the Chemostat, vol. 2.
John Wiley & Sons, Inc., New York, NY. Academic Press, Inc., New York, NY.
20. Rittmann, B. E., and P. L. McCarty. 2001. Environmental 24. Thauer, R. K., K. Jungermann, and K. Decker. 1977.
Biotechnology: Principles and Applications, 1st ed. McGraw- Energy conservation in chemotrophic anaerobic bacteria.
Hill Higher Education, New York, NY. Bacteriol. Rev. 41:lOO-180.
Physicochemical Factors in Growth
JOHN A . BREZNAK AND RALPH N . COSTILOWt

14.1. HYDROGEN ION ACTIVITY (pH) ........................... 309


14.1.1. pH Measurement ..................................... 310
.
2. pH Buffers ......................................... 310
.
3. Continuous Control of pH .............................. 314
14.2. WATER ACTIVITY AND OSMOTIC PRESSURE ................. 314
14.2.1. Measurement of Water Activity .......................... 314
.
2. Measurement of Osmotic Pressure ........................ 314
14.3. TEMPERATURE .......................................... 315
14.4. PRESSURE .............................................. 316
14.5. OXYGEN AND OTHER GASES .............................. 316
14.5.1. Dissolved-Oxygen Measurement .......................... 317
.
2. Measurement of Oxygen Absorption Rate ................... 317
.3.'Hypoxia and Microaerophiles ............................ 317
.4. C02 and Capnophiles .................................. 319
14.6. ANAEROBIOSIS .......................................... 319
14.6.1. Oxidation-Reduction Potential (Eh) ........................ 319
14.6.1.1. Electrometric Measurement of .................. 320
Eh
.2. Dyes Sensitive to ........................... 320
Eh
14.6.2. Reducing Agents ..................................... 320
.3. Techniques for Nonstringent Anaerobes .................... 321
.4. Hungate Technique for Stringent Anaerobes ................. 322
14.6.4.1. Removal of Oxygen from Gases ................... 322
.2. Preparation of Prereduced Media .................. 322
.3. Inoculation and Transfer ........................ 323
.4. Roll Tubes, Shake Tubes, and Bottle Plates ........... 324
14.6.5. Anoxic Chambers for Stringent Anaerobes.................. 324
14.7. LIGHT ................................................. 325
14.7.1. Light Measurement ................................... 325
. ...............................
2. Light Sources and Filters 326
14.8. DIFFUSION GRADIENTS .................................. 326
14.9. MAGNETIC FIELDS ....................................... 326
14.10. VISCOSITY .............................................. 327
.
14.1 1 REFERENCES ............................................ 327

Some physicochemical factors affecting microbial growth 14.1. HYDROGEN ION ACTIVITY (pH)
are controlled by the constituents of the culture medium pH is a measure of the mean hydrogen ion activity of a so-
(hydrogen ion activity. water activity. osmotic pressure. lution ( a H + ) and is defined as follows: pH = -log.. aH+.
and viscosity). Others are controlled by the external envi- The activity of the Hf ion is the product of its molar con-
ronment (temperature. oxygen. light. hydrostatic pressure. centration and its activity coefficient. which. in turn. is a
and magnetic-field strength) . The oxidation-reduction po- function of the total ionic strength of the solution (p,) . In
tential is controlled by both the medium and the environ- dilute solutions. where p ranges from 0 to 0.25, the activity
ment . All of these factors can influence the growth rate. coefficient of the H' ion (at 25OC) ranges from 1 to 0.85,
cell yield. metabolic pattern. and chemical composition of respectively. Most routinely used microbiological media
bacteria. The control of hydrogen ion activity. tempera- can be considered ionically dilute solutions. For example.
ture. and oxygen supply is critical with every bacterial cul- in a typical glucose salts medium. p, is about 0.02.
ture. whereas the control of oxidation-reduction potential Therefore. the activity coefficient of the H+ ion is close to
is of major importance in culturing obligate anaerobes. unity. and hence a H + is essentially equal to the hydrogen
Light quality and quantity are critical for growth of photo- ion concentration. [H'] . Consequently. pH = -log [Hf]
trophs. This chapter addresses practical aspects of applica- for most microbiological media.
tion and control of physicochemical factors. The pH scale is typically thought of as ranging from 0
([H'] = 1 M) to 14 ([H+] = M). although values
outside this range are possible . Most known microbes grow
tDeceased . over a relatively narrow range of pH. usually best near

309
310 GROWTH

neutrality (pH 7.0). However, an ever-increasing number of control is desirable, so calibration with standard buffer need
extremophiles continue to be recognized and isolated from be done at only one temperature. In any case, keep in mind
nature (78, 79). Many of these grow optimally at very low that an average error of only 0.003 pH unit per degree
pH (acidophiles) or very high pH (alkaliphiles). Some of Celsius exists at 1 pH unit from standardization; this is often
these organisms also grow at high temperatures (thermo- less than the required accuracy for measurements.
and hyperthermophiles) or high salinities (halophiles) or 2. Standardize the pH meter with a buffer that is near
both. Others grow optimally at low temperatures (psy- the expected pH of the sample and of similar ionic strength.
chrophiles). Accordingly, for precise definition of the pH If accurate readings are to be made across a fairly wide range
optimum for growth of such bacteria, it is important to keep of pH values (e.g., 3 pH units), a meter with a slope control
in mind the factors that affect the activity coefficient of the is desirable and is used after two-point calibration. In this
hydrogen ion and hence the aH+.Among these factors are procedure, the pH meter is standardized with a buffer, the
temperature, ionic strength, ion charge, dielectric constant, pH of which is at or just outside one end of the expected
and the physical size of various ions in the solution. In prac- range; the electrode(s) is placed in a second standard buffer
tice, the influence of such factors on pH is taken into ac- representing the other end of the expected range; and then
count by standardizing the pH meter with a pH reference the pH meter readout is adjusted to the appropriate pH by
solution whose composition and temperature are as close as using the slope control. A frequently encountered sample of
possible to those of the solution being measured (e.g., the relatively high ionic strength is seawater. The pH of such
microbial culture fluid). samples should be made after standardization with a buffer
consisting of 0.4186 M NaC1, 0.0596 M MgC12, 0.02856
14.1 . I . pH Measurement M Na2S04,0.005 M CaC12,0.02 M Tris, and 0.02M Tris-
For accurate pH measurements of virtually all types of solu- HC1. The pH of this buffer is 8.835 at 5C, 8.517 at 15C,
tions, use a pH meter with a glass electrode. Two types of 8.224 at 25C, and 7.953 at 35C (100).
glass electrode systems may be used. One consists of a pH- 3. If the sample must be stirred (e.g., with a magnetic
measuring electrode paired with a separate pH reference stirrer) during measurement, stir the calibrating buffer in
electrode (Hg/Hg2C12or AglAgCl), both of which are put the same manner, since stirring often alters the signal of the
into the solution being measured. Another system consists pH electrode.
of a single electrode containing elements of both the pH- 4. Determine the pH of media after sterilization, not be-
measuring and reference electrode, referred to as a pH com- fore. Autoclaving of media frequently results in a significant
bination electrode. This type of electrode is popular, be- change in the pH owing to the escape of C 0 2 or the pre-
cause its generally compact size facilitates the measurement cipitation of alkaline-earth phosphates. Even filtration may
of small volumes contained in test tubes. Epoxy electrodes have a significant effect on the pH of media.
(pH-measuring, pH reference, and pH combination types) 5. Just prior to use, check the pHs of media that have
are also available and are generally more durable than glass been stored.
electrodes. However, epoxy electrodes are not recom- 6. Some pH electrodes (those with linen fiber junctions)
mended for use with certain organic solvents; consult with do not give accurate pH readings with Tris buffers. Be sure
the manufacturer for the solvent tolerance of such elec- that the electrodes used with Tris buffers are recommended
trodes if this is a concern. So-called microelectrodes, with for such use by the manufacturer, e.g., Orion no.71-03 Tris
tip diameters in the micrometer range, have become widely calomel pH electrodes with ceramic junctions. In addition,
used for making measurements on tiny samples (e.g., insect reference electrodes may be especially sensitive toward
guts) or for fine-scale resolution of physicochemical gradi- chelators (e.g., EDTA) or sulfides. For such instances,
ents present in natural samples (e.g., sediments). probes with salt-bridged reference electrodes should be
Microelectrodes capable of measuring the pH, Eh, certain used.
ions other than protons (e.g., NO3-), various dissolved
gases (e.g., 0 2 , HI, NzO, and others), and other physico- In instances when accuracy is not required, such as in
chemical parameters (e.g., temperature, flow velocity, etc.) the preparation of routine media, the pH may be measured
are available from Unisense, Aarhus, Denmark (www by use of pH indicator dye solutions or pH paper. By proper
.unisense.com) and Diamond General Development Corp., selection of either, the pH can be estimated to within 0.2
Ann Arbor, MI (www.diamondgeneral.com). See chapter pH unit. Indicator solutions and papers, and their corre-
17.2 for a further description of hydrogen ion and other ion- sponding color standards for various pH ranges, are avail-
selective electrode systems, procedures used for calibration able from most scientific supply companies. Common pH
of pH meters, and proper care of electrodes. Also see refer- indicators and their useful pH ranges are listed in Table 1.
ences 3 1,68, and 100 for details on measurement and con- Also see chapter 15.5.15.
trol of pH. A number of pH indicators are frequently incorporated
When accurate pH measurements are desired, observe into culture media to demonstrate pH changes during
the following precautions. growth of microbes. Select the appropriate indicator for the
pH range of interest, and make sure that it does not inhibit
pH Measurement growth of the organism. All of these pH indicators are
poorly soluble in water but are effective at very low con-
1. Standardize the pH meter with a buffer (a substance
centrations (less than 0.01%) in media.
that resists change in pH; see below) whose temperature is
similar to that of the sample. The pH of buffers changes
with temperature; e.g., the pH of standard phosphate buffer 14.1.2. pH Buffers
is 6.98 at OC, 6.88 at ZOC, and 6.84 at 37C. See the Many organisms consume or produce significant amounts of
Handbook of Chemistry and Physics (99) for pH values of acidic or basic ions during growth. This commonly occurs
standard buffers at different temperatures. If pH mea- during anaerobic fermentation and during aerobic oxida-
surements are to be made on numerous samples at different tion of the salts of organic acids. Unless acid production and
temperatures, a pH meter with a temperature compensation consumption are balanced, large changes in pH may occur
14. Physicochernical Factors in Growth m 311

TABLE 1 pH indicators and their useful pH range groups of organic molecules (such as proteins, peptides, and
amino acids) provide some buffering capacity. Owing to the
Color variety of compounds present in such media, there may be a
Indicatora pHrange pK,
Acid Alkaline fair degree of buffering action over a wide range of pH.
However, the buffering capacity at any given pH varies
Thymol blue 8.0-9.6 8.9 Yellow Blue widely with the types and concentrations of organic mole-
Metacresol purple 7.4-9 .O 8.3 Yellow Purple cules present. Consider the following in selecting a buffer
for use: (i) the pH desired; (ii) possible inhibitory or toxic
Cresol red 7.2-8.8 8.3 Yellow Red
effects of the buffer; (iii) possible utilization of the buffer by
Phenol red 6.8-8.4 7.9 Yellow Red the culture; and (iv) possible binding of di- and trivalent
Bromthymol blue 6.0-7.6 7.0 Yellow Blue metal ions by the buffer.
Bromcresol purple 5.2-6.8 6.3 Yellow Purple Weak acids and bases buffer most effectively at the pH
Methyl red 4.4-6.0 5.2 Yellow Red where they are 50% dissociated. This pH is equal to the pK,
Yellow Blue
(the negative logarithm of the dissociation constant) of the
Bromcresol green 3.8-5.4 4.7 acid or base. The effective range of a buffer is within 1 pH
Bromphenol blue 3.0-4.9 4.0 Yellow Blue unit of its pK,. A number of compounds that have been
Thymol blue 1.2-2.8 1.5 Red Yellow used as buffers in growth media are listed in Table 2, along
Prepare 0.04% solutions by solubilizing 0.1 g in the smallest possible volume
with their pK, values. The Good buffers, originally devel-
(10 to 30 ml) of 0.01 N NaOH, and then dilute to 250 ml. oped by N. E. Good et al. (33), have greatly expanded the
range of buffers useful in biological systems. The Good
buffers have the advantages of a pK, between 6.0 and 8.5,
high aqueous solubility, impermeability by biological mem-
during growth, and growth may be significantly inhibited. branes, minimum interaction with mineral cations, enzy-
The pH of cultures may be controlled to some extent by in- matic and hydrolytic stability, little or no participation in
corporating pH buffers into the medium. Continuous pH biochemical reactions, minimum absorbance between 240
control may be achieved by the automatic addition of acid and 700 nm, and usually no toxicity. In addition, they show
or base (section 14.1.3). minimum effects due to ionic composition of solutions, con-
pH buffers usually are mixtures of weak acids and their centration, and temperature. A disadvantage is that they
conjugate bases. In complex media, the acidic and basic are slightly more expensive than some of the other buffers.

TABLE 2 pK, values of chemical compounds used in buffers


pK, at:
Compound
25C 20C
~~~~ ~ ~~~~ ~

Citric acid 3.13, 4.77, 6.40


Phthalic acid 2.89, 5.51
Barbituric acid 4.01
Oxalic acid 4.19
Succinic acid 4.16,5.61
Acetic acid 4.75
Monobasic phosphate 7.21
Tris(hydroxymethy1)aminomethane 8.08
Boric acid 9.24
Glycine 9.87
The Good buffers:
MES [2-(N-morpholino)-ethanesulfonic acid] 6.15
ADA (N+2-acetamidoiminodiaceticacid) 6.60
PIPES [piperazine-N,N-bis(2-ethanesulfonic acid)] 6.80
ACES (N-2-acetamido-2-aminoethanesulfonic acid) 6.90
BES [N,N-bis-(2-hydroxyethyl)2-aminoethanesulfonicacid] 7.15
MOPS [3-(N-morpholino)propanesulfonic acid] 7.20
TES [3-tris(hydroxymethyl)methyl-2-aminoethanesulfonicacid] 7.50
HEPES (N-2-hydroxyethylpiperazine-N-2-ethanesulfonic acid) 7.55
TAPSO (N-[tris(hydroxymethyl)methyl]3-amino-2-hydroxy-propanesulfonic
acid] 7.60
HEPPS (N-2-hydroxyethylpiperazine-N-3-propanesulfonic acid) 8.00
TRICINE [N-tris(hydroxymethyl)rnethylglycine] 8.15
BICINE [N,N-bis(2-hydroxyethyl)glycine] 8.30
TAPS (N-[tris(hydroxymethyl)methyl]-3-arninopropanesulfonicacid] 8.40
312 n GROWTH

Note that the salts of citric, phthalic, and succinic acids ratio of the volume of acidic solution to basic solution is
buffer over a wide range of pH because these acids have about 1:l; if it is 1 pH unit above the pK,, the ratio is about
more than one dissociable hydrogen ion and hence more 1:lO; and if it is 1 pH unit below the pK,, the ratio is
than one pK,. about 1O:l. For example, to prepare about 100 ml of 0.1 M
Buffers containing the compounds in Table 2 are pre- sodium phosphate buffer at pH 7.0, mix solutions of 0.1 M
pared by one of the following two general procedures. NaH2P04 (monobasic) and 0.1 M Na2HP04 (dibasic) in
approximately equal proportions and then adjust to pH 7.0
1. Prepare a solution of the buffer salt at twice the de- by adding more of the appropriate solution as determined
sired concentration, and while the solution is being stirred,
with a pH meter. To prepare 100 ml of the phosphate buffer
titrate it with either NaOH or HC1 to the desired pH by
at pH 6.0, titrate 95 ml of the monobasic solution with the
using a pH meter. Dilute to final volume with distilled or
dibasic solution.
deionized water. For example, to prepare 1 liter of 0.1 M
Tris-hydrochloride buffer at pH 7.6, make 500 ml of a 0.2 M Although the Good buffers are becoming increasingly
solution of Tris, titrate with HC1 to pH 7.6, and then dilute popular, the most commonly used buffers in microbiological
to 1 liter. Dilution has only a small effect on the final pH. If growth media are phosphate, Tris-hydrochloride, citrate,
this effect is important, make a final correction before and acetate. These four systems buffer over almost the en-
adding the last few milliliters of water during dilution. tire range of pH permitting bacterial growth. Solutions used
2. Prepare equimolar concentrations of the acidic and for their preparation and the appropriate volumes required
basic components of the buffer system. With the aid of a to buffer at various pH values are given in Table 3. As noted
magnetic stirrer and a pH meter, titrate one solution with above, the actual pH of any buffer is temperature depen-
the other until the desired pH is obtained. If the desired pH dent. Tables for preparing buffers of other types and at dif-
of the buffer is near the pK, of the acid component, the ferent temperatures are available elsewhere (22,32,68,99).

TABLE 3 Formulas for buffers frequently used in microbiological mediaa


Value of x to be used in buffer formulas
PH Acetate bufferb Citrate-phosphate buffer Phosphate bufferd Tris-hydrochloridebuffere
4.0 41.0 30.7
4.2 36.8 29.4
4.4 30.5 27.8
4.6 25.5 26.7
4.8 20.0 25.2
5.0 14.8 24.3
5.2 10.5 23.3
5.4 8.8 22.2
5.6 4.8 21.0
5.8 19.7 46.0
6.0 17.9 43.85
6.2 16.9 40.75
6.4 15.4 36.75
6.6 13.6 3 1.25
6.8 9.1 25.5
7.0 6.5 19.5
7.2 14.0 44.2
7.4 9.5 41.4
7.6 6.5 38.4
7.8 4.25 32.5
8.0 2.65 26.8
8.2 21.9
8.4 16.5
8.6 12.2
8.8 8.1
9.0 5.0
Data adapted from reference 32.
bFormula: x ml of 0.2 M acetic acid + (50 - x) ml of 0.2 M sodium acetate, diluted to 100 ml.
Formula: x ml of 0.1 M citric acid + (50 - x) ml of 0.2 M Na2HP04, diluted to 100 ml.
dFormula: x ml of 0.2 M NaH2P04 + (50 - x) ml of 0.2 M Na2HP04,diluted to 100 ml.
Formula: 50 ml of 0.2 M Tris + x ml of 0.2 M HCI. diluted to 100 ml.
14. Physicochemical Factors in Growth w 313

When cultures are incubated in atmospheres enriched (in this case water) vapor pressure to the total gas pressure
with C 0 2 , the C02-bicarbonate equilibrium is an important of 760 mm Hg; otherwise, a small error will be introduced
consideration and, in fact, can be exploited as a buffer sys- in the calculation. This can be seen from Table 4, which
tem itself at a pH between 6.0 and 8.0. C02 dissolves in and lists values for a that do (column B) and do not (column A)
reacts with water to form carbonic acid, which readily ion- consider the vapor pressure of water and which also lists
izes to form a proton and a bicarbonate anion as follows. values for pK' at various temperatures. As an example, the
following is a calculation of the concentration of NaHC03
KS +HzO Ka1 required to buffer a medium at an initial pH of 7.0 at 30C
C02(gas) t)C02 (dissolved)t)H2C03t)H+ + HC03- under a gas phase containing 20% COz:
-HZ0
log [HC03-] = pH - pK'
The formation of carbonic acid and, hence, the acidity + log [a. ( % c o 2 ). (4.35 x
created by it are dependent on the concentration of dis-
solved C 0 2 ,which, in turn, is dependent on the concen- log [HC03-] = 7.0 - 6.348
tration of COz in the gas phase in accord with Henry's law. +
log [(0.642) . (20) . (4.35 X = -1.601
Ks is the solubility constant of C02 (which is temperature [HCO3-] =z 0.025 M
dependent). Inasmuch as the concentration of H2C03in
solution is usually very low, the dissociation constant Kal However, values for a and pK' are also influenced by the
usually refers to the reaction, presence of other ions in solution. Therefore, in practice
slight adjustments may have to be made in the amount of
C02 (dissolved) + H 2 0 t)H+ + HC03- HC03- included in the liquid phase, or in the concentra-
The Henderson-Hasselbach equation for this system is tion of C02used in the gas phase, to achieve the desired pH
of the medium before inoculation. Adjustment of the pH
pH = pK' + log [HC03-]/[C02 (dissolved)] before inoculation can also be made by addition of sterile
Na2C03or HC1. For growth of alkaliphiles, advantage can
where pK' (the negative logarithm of Kal for the reaction be taken of the second dissociation of bicarbonate to yield
shown above) normally takes into account the activity of a proton and the carbonate anion:
H2O (which is 1 by definition and is close to 1 for dilute so-
lutions), which can itself be considered a constant at a Kaz
given temperature. The pK' value is about 6.4 at 25C. HC03- H H+ + C03'
Therefore, the molar ratio of dissolved C02 to HC03- is The pK of this reaction at 25C is 10.33 and ranges from
about 1:l at this pH. C02-bicarbonate buffers are effective 10.56 at 5C to 10.16 at 100C. Hence, a buffer system of
over a pH range of about 5.4 to 7.4. If the pH of the medium NaHC03-NaZCO3 can be prepared, as described above,
is below 5.0, essentially no bicarbonate is present, but an in- that will facilitate control of pH between 9 and 11.5. In this
crease in COz concentration in the atmosphere will still re- case, inclusion of C 0 2 in the gas phase is unnecessary, as
sult in a decrease in pH as a result of the formation of car- virtually all of the relevant carbonate species will be present
bonic acid. In an atmosphere of 50 to 100% COz, the in solution. More detailed information on COz-bicarbonate-
medium must contain a substantial amount of bicarbonate carbonate equilibria and buffering capacity can be found in
(e.g., NaHC03) to maintain a pH level near neutrality. The the classic manual for manometric methods (94) and in
high salt concentration that results may be toxic to some Stumm and Morgan (87). The latter includes detailed dis-
bacteria. cussion of the effect of other ions on the equilibrium and
After appropriate substitution, the Henderson- the solubility of the carbonate species.
Hasselbach equation can be solved to determine the molar- The pH of media used for growing microbes that produce
ity of bicarbonate needed in a medium for buffering at a large amounts of acid (e.g., lactic acid bacteria) may be very
given pH when the atmosphere contains a certain percent-
age of COz. For this calculation, use the following equation:
log [HC03-] = pH - pK'
+ log [P . a . (%coz)(5.87 x TABLE 4 Values of (Y (C02 solubility) and pK' at various
where P is the atmospheric pressure in millimeters of Hg; a temperatures
(often referred to as the Bunsen absorption coefficient) is aa
the volume of gas (normalized to the volume it would oc- Temp ("C) pK'b
cupy at a 0C and 760 mm Hg) that is absorbed by a unit A B
volume of water (at the temperature of the measurement)
under a gas pressure of 760 mm Hg; %COz is the volume of 20 0.878 0.859 6.392
C02 per unit volume of gas phase X 100; and 5.87 x lop7 25 0.759 0.738 6.365
is a factor that converts a to moles per liter at 760 mm Hg 30 0.665 0.642 6.348
(94). If atmospheric pressure varies between 720 and 760 35 0.592 0.565 6.328
mm Hg (96.0 and 101.3 kPa), the variation has little effect
on the calculated [HC03-]. Therefore, in most instances 40 0.530 0.494 6.3 12
the following simplified equation can be used (calculated a Milliliters of C02 (reduced to 0C and 760 mm Hg) that will dissolve in
for P = 740 mm Hg): 1 ml of pure water at a gas pressure of 1 atm (760 mm Hg) at the stated temper-
ature. The values for a in column A do not consider the contribution of water
log [HC03-] = pH - pK' vapor pressure t o the total gas pressure ( 2 3 , 5 5 ) . The values in column B have
+ log [a. ( % c o z ) . (4.35 x been recalculated from q values in references 23 and 55 and do consider the con-
tribution of water vapor pressure to the total gas pressure.
K. I
When consulting published values for a,use tabulated pK' = -log K,, for the reaction: CO2 (dissolved) + H20 c) H' +
values that take into account the contribution of solvent HC03-. Data from reference 94.
314 GROWTH

difficult to control with soluble buffers. However, the acid The osmotic pressure (IT) of a solution, expressed in at-
can be neutralized as fast as it is formed by inclusion of mospheres, is related to a, as follows:
finely ground C a C 0 3 (chalk) in the medium. In fact, a good
way of visualizing acid production by colonies growing on 7~ = [RT/v,] In a,,,
agar plates is to include 0.3% finely ground chalk in the where R is the gas constant (0.0821 liter atm mol-' K-'), T
medium. Make sure that the chalk is well suspended in the is the absolute temperature in kelvins (K) (= degrees
molten agar when the plate is poured. Alternatively, pour a +
Celsius 273), and V, is the volume of 1 mol of water. For
chalk-free base layer first and, when solidified, pour a rela- more-detailed presentations of these relationships, see refer-
tively thin chalk-containing top layer; this keeps the sus- ences 14, 36, 75, 76,80, and 92.
pended C a C 0 3 close to colonies developing on the surface. The minimum a, values at which microbes grow vary
Colonies that produce acid will be surrounded by a clear widely, but the optimum values for nonmarine species is typ-
zone. The acid produced in liquid cultures can also be with- ically greater than 0.99. By contrast, most marine bacteria
drawn by use of a dialysis or other product removal culture are somewhat halophilic and require sodium ions; they grow
system (see chapter 12.2.6.1 and 12.2.6.2) or can be neu- best in the presence of 0.5to 1.0 M NaC1, at which a, is ap-
tralized by the addition of alkali with a system under auto- proximately equal to 0.98. Extreme halophiles are largely
matic pH control (see below). represented by some Archaea and grow best in 3 to 5 M NaCl
(a, = 0.8), which is near saturation. Species of so-called xe-
14.1.3. Continuous Control of pH rophilic fungi are capable of growth on relatively dry sub-
In instances when the pH of a culture must be controlled strates such as dried fruits, candy, and cereal, whose a, is
within a narrow range, an automatic pH control system about 0.7. For any given microbe, however, variations in a,
should be used. Such a system should include a pH meter, will almost certainly affect growth rates, cell composition,
steam-sterilizable pH electrodes, a controller and controls and metabolic activity. Many solutes have very specific ef-
for setting the desired pH limit, and pumps for the addition fects at concentrations below those required to limit the
of acid and/or base. Most such systems. are linked to a availability of water. Therefore, determinations of growth-
recorder to provide a continuous record. Automatic sys- limiting a, values should be conducted by using media in
tems, including steam-sterilizable pH electrodes, are avail- which a, has been adjusted with more than one solute. The
able from a number of sources (e.g., New Brunswick a, and osmotic pressure of microbiological media are best
Scientific Co., Inc., Edison, NJ [www.nbsc.com]; The VirTis controlled by adding nonnutrient solutes such as sodium
Co., Inc., Gardiner, NY [www.virtis.com];and Cole-Parmer chloride, potassium chloride, sodium sulfate, or mixtures of
Instrument Co., Vernon Hills, IL [www.coleparmer.com]. such salts. Scott (80) has detailed methods for calculating
Check pH control systems at frequent intervals for ac- the a, of nutrient media and for calculating the required
curacy by using a separate pH meter. Problems that may be concentrations of salt(s) to attain a given a, value.
encountered are (i) improper or inadequate grounding of For considerations of osmotic pressure relative to bacte-
the system; (ii) establishment of pH control points (i.e., rial permeability and transport, see chapter 20.4. See refer-
upper and lower limits) that are too narrow, resulting in al- ences 12, 14, 80, and 92 for reviews of the physiological ef-
most constant addition of acid and base and hence signifi- fects of a, on bacteria.
cant dilution of the culture; (iii) aging and deterioration of
the pH glass electrode as a result of repeated steam sterili- 14.2.1. Measurement of Water Activity
zation; and (iv) contamination of the reference electrode A number of methods have been developed for the mea.
diaphragm by continuous exposure to medium constituents surement of relative humidity or a,,, directly. Special instru-
or metabolic products of the bacteria. The most common ments and probes for this purpose are available from
contaminants of the diaphragms are proteins and silver sul- Omnimark Instrument Corp., Tempe, AZ [www.omniwww
fide precipitate. The latter is formed by reaction of the sil- .corn]; Rotronic Instrument Corp., Huntington, NY
ver chloride in the reference electrode with sulfur-contain- [www.rotronic-usa.com];Omega Engineering Inc., Stamford,
ing substances in the medium. Remove the proteins by CT [www.omega.com]; GE General Eastern Instruments,
soaking the electrode in a protease (e.g., pronase) solution, Billerica, MA [www.globalspec.com/Supplier/Catalog/
and remove the silver sulfide deposits with an acidic solu- GEGeneralEastern]; and Newport Scientific Inc., Jessup, MD
tion of thiourea. For further discussion of automatic pH [www.newport-scientific.com]. The advantages and disad-
control and sterilization of electrodes, see chapter 12.2.3.1 vantages of these instruments have been reviewed (76,92).
and reference 68. The results of a study (54) of methods for determining a,
indicated that there is considerable variability among the
values obtained by different procedures and that the signif-
14.2. WATER ACTIVITY AND OSMOTIC icance of values reported beyond the second decimal place
PRESSURE is doubtful. A statistical analysis (91) of measurements
Water must be available to cells for metabolism and growth. made with the electronic Sina-Scope instrument (now
However, the mere presence of water in a medium does not Novasina and marketed through Omnimark, above) over
ensure its availability, which is determined by the water ac- an a,,, range of 0.755 to 0.967 indicated a variation of less
tivity (a,) of the medium. The a, represents the mole frac- than t0.01.The accuracy for such instruments is currently
tion of the total water molecules that are available and is stated to be 2 0.01.
equal to the ratio of the vapor pressure of the solution to
that of pure water(p/p,). This ratio is equal to the fractional 14.2.2. Measurement of Osmotic Pressure
relative humidity (%RH/100)of the atmosphere above the The most commonly used instruments for measuring osmotic
medium at equilibrium. Temperature variation within the pressure are osmometers and are based on measurement of
range in which most bacteria grow has little effect on the a, vapor pressure (e.g., Discovery Diagnostics, Claremont,
of the medium. Ontario, Canada [www.discovery-diagnostics.com])or
14. Physicochernical Factors in Growth m 315

freezing point depression (e.g., Advanced Instruments Inc., temperature for growth is complicated by the fact that a
Norwood, MA [www.aicompanies.com]).Although some of shift of cells to lower temperatures is often accompanied by
these instruments are calibrated for direct readout, several a period of no growth, whose duration is extended the lower
standard solutions should be run simultaneously with the the temperature. Hence, it is frequently necessary to incu-
measured solutions. bate a bacterial culture for a prolonged period to measure
the growth rate near the minimum temperature. For exam-
ple, one Escherichia coli culture required over 41 h for one
14.3. TEMPERATURE generation at 8Ccompared to 21 min at the optimum tem-
The temperature of incubation dramatically affects the perature plateau of 36 to 42C (45). The minimum temper-
growth rate of microbes, because it affects the rates of all ature for growth is best determined by growing a culture
cellular reactions. In addition, temperature may affect the somewhat above the minimum and then shifting it to pro-
metabolic pattern, nutritional requirements, and composi- gressively lower temperatures (46).
tion of bacterial cells. Over a limited range of temperature below the optimum,
The growth rates of most microbes respond to tempera- the changes in growth rates of a microbe are comparable to
ture in a manner similar to that shown in Fig. 1. The useful the responses of chemical reaction rates to temperature.
range of temperature at or near the optimum is usually quite Therefore, if the logarithm of the growth rate is plotted
narrow, and the maximum temperature for growth is only a against the reciprocal of the absolute temperature (i.e., an
few degrees ( 3 to 5C) above the optimum. By contrast, the Arrhenius plot [Fig. l]),a linear slope is observed in a limited
minimum temperature for growth may be 20 to 40C below range (45, 74, 75). For most bacteria, the temperature coeffi-
the optimum. However, determination of the minimum cient (Qlo)for the growth rate in this limited range is about
2; i.e., the growth rate doubles with a 10C increase in tem-
perature. The slope of such an Arrhenius plot becomes essen-
tially zero around the optimum growth temperatures and ap-
3.0 proaches infinity near the maximum temperature for growth.
Generally, mrcrobes not only grow more slowly but also
die more rapidly at temperatures above the maximum for
2.6
growth. Consequently, incubation of a culture at tempera-
tures above the microbe's optimum requires precise temper-
ature control. To be safe, incubate a culture below its opti-
f 2o mum temperature to an extent determined by the
.-5 variability of the incubator.
The temperature within ordinary convection-type air in-
1.6
L
Q
cubators may vary by several degrees Celsius, even more if
E opened frequently. Incubators equipped with circulating
fans or water jackets maintain a more constant temperature,
1.0
but variations in temperature of 1C are not uncommon.
Nevertheless, for routine cultivation of microbes, an array
0.6
of air incubators, including models designed for low-
temperature incubation, are commercially available from
scientific supply companies.
n I I For accurate measurements of the effect of temperature
0 10 2 0 3 0 40 w on bacterial growth rate, however, use a stirred water bath
*C whose temperature is controlled by a thermostat with a
sensing element that is immersed in the circulating water.
Such baths are available commercially and include models
for high- and low-temperature incubation. For accurate
0.5 J measurements, also be sure to use an accurately calibrated
thermometer. To assess the temperature response of a mi-
crobe, measure growth rates at a number of carefully con-
f 0.0 trolled temperatures. Plot these data as illustrated in Fig. 1.
If the general shapes of the resulting curves vary greatly
C
.-
0
CI from the normal pattern, repeat the determinations while
f6 6 closely monitoring the stability of the temperature of the
water bath.
8 Practical procedures and precautions relative to temper-
5m -1.0 ature control of bacterial growth are as follows.
Temperature Control
-1.6 Measure temperatures at various positions in air incubators
I I I I
by using thermometers inserted into stoppered water-
31 32 33 34 35 36 filled flasks or tubes.
10"l'K Air convection incubators may vary widely in temperature.
Use circulation fans, and check to see that they are op-
FIGURE 1 Effect of temperature on the generation time of erating efficiently. Position cultures in the incubators so
E. coli, as depicted by an arithmetic plot (top) and by a semi- that "dead" air spaces are not created. Do not open in-
logarithmic Arrhenius plot (bottom). Data from reference 45. cubators more frequently than absolutely necessary.
316 GROWTH

Surface cooling due to evaporation of media in air incuba- 14.5. OXYGEN AND OTHER GASES
tors must be controlled in critical studies of temperature Gases frequently constitute substrates for bacteria, whether
effects. When possible, use tightly stoppered culture ves- serving as a n oxidizable energy source (e.g., H2, CH,, and
sels. Where free air exchange is necessary, as for aerobic CO), a terminal electron acceptor of aerobic respiration
bacteria, use incubators with provision to maintain high (e.g., 02), or a source of nitrogen (e.g., N2). Consequently,
relative humidities. This is exceedingly important at the metabolism and growth rates of bacteria are often de-
high incubation temperatures, at which loss in culture pendent on the concentration of gas in solution. Among
volume could present a serious problem, particularly the most prevalent concerns in this regard are aerobic and
with shaken cultures. facultative bacteria, whose growth rate and yield may de-
Use 50% polyethylene glycol antifreeze or light silicone oil pend critically on the concentration of oxygen in solution.
in baths used for high-temperature or low-temperature Hence, the following discussion focuses on oxygen as an ex-
incubation. ample, although the principles are also true for virtually any
Temperature gradient incubators have been reviewed by gas that dissolves in water.
Patching and Rose (74). These incubators provide temper- Unlike most nutrients, oxygen is relatively insoluble in
atures that exceed the entire temperature range of growth water ( < l o mg/liter) and may quickly become limiting in
for a bacterium. Different designs allow for incubation of liquid bacterial cultures unless special precautions are taken
organisms with various oxygen requirements. If large num- to ensure that it is supplied and dissolved continuously dur-
bers of determinations are to be made, the construction ing growth. Principal factors to consider in supplying dis-
and use of a gradient incubator may be desirable. A com- solved oxygen to liquid cultures (aeration) are as follows.
mercial source for a temperature gradient incubator is not Aeration of Liquid Cultures
known to us, but one can be constructed in a machine
shop. A recent design has been described by Elsgaard and For availability of gaseous oxygen to the gas-liquid inter-
JBrgensen (27). face, the opening of the culture vessel must be suffi-
ciently large and stoppered or covered with porous ma-
terial so as to allow maximum exchange of the
14.4. PRESSURE atmospheres inside and outside the vessel.
High hydrostatic pressure forces volume changes and can Large gaseous surface-to-liquid volume ratios in static cul-
have deleterious effects on cell macromolecules. High pres- tures can be maintained only by use of very shallow liq-
sure can also inhibit or accelerate enzymatic reactions that uid volumes in flat-bottom vessels such as Erlenmeyer or
are accompanied by an increase or decrease in volume, Fernbach flasks. Either rotary or reciprocal shaking of
respectively (1). Little or no effect on most species has flasks greatly increases the surface area of a given volume
been observed at pressures ranging from 0.101 to 5 MPa of medium and hence increases the efficiency of mass
(0.101 MPa = 1 atm = 15 lb/in2 = 101,325 N/m2 = 760 transfer. This effect can be further enhanced by creation
mm Hg = 1.01325 bar). However, at 5 MPa a detectable of additional turbulence in the flask, either by using
diminution in the growth rate of E. coli is observed (61). At flasks with dimples or baffles in the side of the flask
69 MPa, protein synthesis in E. coli ceases owing to inhibi- or by placing a stainless-steel coil spring in the flask. The
tion of polysome formation and translocation (46). O n the oxygen-solution rate obtained in a shaken flask de-
other hand, with increased exploration of the deep sea, creases rapidly as the volume of culture increases and as
barophilic (also known as piezophilic [l]) Bacteria and the density of cells increases.
Archaea have been recognized that show optimum growth With large volumes of cultures (>0.5 liter), it is usually
at hydrostatic pressures significantly greater than that of at- necessary to force air through (sparge) the liquid. The ef-
mospheric (107). Some bacteria isolated from the Mariana ficiency of oxygen dissolution in sparged cultures de-
Trench, the deepest ocean bottom in the world, have even pends primarily on the number and smallness of the air
proven to be obligate barophiles. For example, strain MT41, bubbles and on the time of contact between the bubbles
a y-proteobacterium related to Colwellia, grows optimally at and the liquid. The use of gas spargers with small pores
69 MPa and also grows at 103 MPa, a pressure close to that results in small bubbles but may induce foaming. The
at the depth of its origin (10 km). However, this strain does time of contact can be maximized by using culture ves-
not grow at 35 MPa or less, which are pressures found at av- sels with high height-to-diameter ratios, by vigorously
erage depths of the sea (24, 108). Other obligately stirring the cultures, and by using baffles in the vessel.
barophilic y-proteobacteria have been isolated from this Finally it should be remembered that, according to Henrys
same habitat (42). law, the mass of gas that dissolves in a definite mass of
Studies of bacteria growing under high pressures are lim- liquid at a given temperature is directly proportional to
ited and currently represent a rather specialized domain of the partial pressure of that gas. Therefore, if agitated liq-
research, so details on the methods used are not described uid cultures are incubated under (or sparged with) a gas
here. However, Marquis (60) has described experimental phase containing 0 2 at concentrations greater than at-
procedures for applying high pressures to cultures, and mospheric (i.e., >21% [vol/vol]), the amount of dis-
Jannasch and colleagues (49,89) have described equipment solved oxygen that can be provided to growing cells is
that allows sampling, manipulation, and even isolation of increased. Gas solubilities are often expressed in terms of
cultures under high hydrostatic pressure without decom- an Ostwald coefficient, L, which is the ratio of the vol-
pression. Recently, Nauhaus et al. (69) described a device ume of gas absorbed to the volume of the absorbing liq-
for incubating marine sediments containing anaerobic uid, both measured at the same temperature. The partial
methane oxidizing consortia under high hydrostatic pres- pressure of the gas must be designated, and tabulated
sures. The effects of hydrostatic pressure on the physiology values are usually reported for the pure gas at 1 atm
and metabolism of bacteria are discussed in references 48, (101.325 kPa). Multiplication of L by the decimal frac-
60, and 61. tion of a particular gas in the gas phase (e.g., 0.21 for 0 2
14. Physicochemical Factors in Growth H 317

in air) yields the equilibrium concentration of that gas in tion by air. If partial freezing occurs, warm the sample before
water, as milliliters of gas per milliliter of H20, at the completing the titration.
specified temperature, T. Further multiplication by 3. Add a few drops of a 10% starch solution, and titrate
(273.15)( 1,OOO)/( 22.4)(absolute temperature in kelvins) to a permanent blue end point with a freshly standardized
expresses the equilibrium concentration as micromoles solution of iodine. Standardize the iodine solution against a
of gas per milliliter of water, or millimolar. standard sodium thiosulfate solution. The normality of the
iodine solution should be about one-fifth that of the sulfite
Table 5 gives Ostwald coefficients for various gases at
solution.
five different temperatures. Using this table and the infor-
mation above, the concentration of O2in water in equilib-
4. Calculate the OAR as follows.
rium with air at 1 atm and 25C is calculated to be 0.0065 (ml of titration difference x normality of iodine)
ml of 02/ml of H20, or 0.267 mM. A more extensive tabu- OAR =
lation of Ostwald coefficients and other constants for vari- 4
ous gases is given by Wilhelm et al. (103). See chapter 12 1,000 ml 1
X-
for more details on liquid cultures. The principles of aera- 5ml 'min
tion are also reviewed in references 75 and 96.
The concentrations of sulfite and iodine most appropri-
14.5.1. Dissolved-OxygenMeasurement ate for any given efficiency of aeration may be calculated by
Dissolved oxygen is most conveniently measured polaro- substitution in this equation. For example, if the OAR is
graphically with an oxygen electrode. The principles and about 3.0, the use of a 0.15 N iodine solution would result
operating procedures involved are described in chapter in a titration difference of about 4 ml for a 10-min interval
17.2.3. Procedures for using such an instrument to deter- of oxygen absorption by the solution. The initial sodium
mine the rates of oxygen solution in cultures are described sulfite concentration should be about five times the iodine
in detail in references 40, 75, and 96. concentration, or 0.75 N.

14.5.2. Measurement of Oxygen Absorption Rate


14.5.3. Hypoxia and Microaerophiles
Oxygen absorption rate (OAR) is expressed in millimoles of
O2 per liter per minute and provides information on the
A number of microorganisms capable of respiring with 0 2 as
terminal electron accceptor grow better (or will only grow)
oxygenation efficiency of an aeration system. Such rates can
under hypoxic atmospheres, in which oxygen is present at
be measured polarographically by using an oxygen electrode
concentrations substantially less than atmospheric, e.g.,
as mentioned above. In the absence of such equipment, the
from 1 to 10% O2 (vol/vol). Such microbes are termed mi-
OAR can also be determined chemically by measuring the
croaerophiles. The physiological basis for this phenotype
rate of oxidation of sulfite to sulfate by dissolved oxygen in
appears to reside in the sensitivity of cells to reactive oxy-
the presence of a copper catalyst. However, the actual OAR
gen species (ROS) (e.g., superoxide anion, hydrogen perox-
in broth cultures may be quite different from that observed
ide, and hydroxyl radical) generated by their own metabo-
in sodium sulfite solutions. Procedures for measuring OAR in
lism in the presence of O2 or formed abiotically in culture
media in the presence or absence of biomass are described
media. Endogenous sources of ROS can be diminished by
elsewhere (15, 75, 96). The following is a simple method
incubation of cultures under hypoxia. Formation of exoge-
(20) for measuring OAR by sulfite oxidation. nous ROS can be diminished by preparation of media under
1. Instead of a growth medium, use the appropriate vol- hypoxic (or anoxic) conditions and by avoiding exposure of
ume of a sodium sulfite solution containing 0.001 M copper media to light; and residual ROS can be minimized by in-
sulfate, and operate the system at the same temperature and clusion in media of agents that quench ROS, e.g., catalase,
under the same conditions of aeration to be used for cul- superoxide dismutase, pyruvate, and sodium metabisulfite in
tures. The appropriate concentration of sodium sulfite to combination with an iron salt. See reference 53 for a more
use depends on the aeration conditions under study (see detailed discussion of this issue. Examples of mi-
below). croaerophiles include species of Campylobacter (98),
2. At various intervals after initiating aeration, pipette Helicobucter (84), Spin'llum ( 5 2 , 73), and Magnetospin'llum
duplicate 5-ml samples of the sulfite solution into 2.5- by (85). Some bacteria may be considered conditional micro-
25-cm test tubes each containing a small pellet of dry ice. aerophiles in that they exhibit a microaerophilic phenotype
The rising C02 stirs the sample during titration and blan- when grown under certain conditions, e.g., Teredinibmter
kets the sample with gaseous C02to prevent further oxida- tumerae and Azospirillum species, when growing under

TABLE 5 Ostwald coefficients for various gases


Ostwald coefficient"
Temp ("C)
0 2 H2 N2 co CH4 C2H2 C2H4 CZH6
20 0.0334 0.0194 0.0169 0.0249 0.0367 1.108 0.1275 0.0514
25 0.03 11 0.0191 0.0159 0.0233 0.0340 1.013 0.1162 0.0453
30 0.0293 0.0190 0.0151 0.0221 0.0316 0.935 0.1068 0.0405
35 0.0277 0.0189 0.0145 0.021 1 0.0296 0.870 0.0990 0.0367
40 0.0265 0.0189 0.0140 0.0202 0.0280 0.816 0.0925 0.0337
'Values are in milliliters per milliliter of HZOat a partial pressure of 1 atm of the pure gas and at the specified temperature
Next Page

318 GROWTH

N2-fixing conditions, to prevent 02-inactivation of nitroge- flow rate desired. Therefore, one should have some idea of
nase (25,37). A number of Pseudomonas and Bacillus species the proportions in which the gases are likely to be mixed
also exhibit a microaerophilic phenotype when growing in and the total combined gas flow rate desired before pur-
low-substrate media (62). chasing such a device. This will depend on the rate of O2
Considerations relating to the cultivation of microbes consumption per milliliter of culture, the volume of cul-
under hypoxia include not merely creating a hypoxic gas tures, and the anticipated number of cultures to be grown at
mixture or a liquid medium containing a low amount of dis- any one time. Keep in mind that outflow from a single gas
solved O2 (both of which are fairly easy to accomplish), but proportioner can be connected to a manifold capable of de-
continually replenishing the O2 as it is used up by cells. livery to several culture vessels, with optional additional
Taken together, these considerations have led to three gen- flow controllers regulating the flow rate to the individual
eral strategies for cultivation of microbes in hypoxia, as fol- vessels. A n alternative to the tube-float-type gas propor-
lows. (i) For motile microaerophiles that can tolerate O2to tioners are mass flow blenders. These are electronically con-
the extent that they can initiate growth in air-equilibrated trolled and give digital readouts of individual and combined
medium (ca. 250 to 300 p M dissolved 02), cells can be in- gas flow rates. Mass flow controllers and blenders are less
oculated into unshaken flasks or serum bottles of liquid susceptible to variations in temperature and back-pressure
medium or soft agar medium under a n atmosphere of air. than are the tube-float units, but they are more expensive.
This allows cells to migrate to, and grow within, their pre- Gas proportioners and mass flow blenders are available from
ferred position below the air-medium interface, within the a number of commercial sources, including Matheson Gases
O2 gradient that is eventually created by the downwardly & Equipment, Chicago, IL (www.matheson-trigas.com);
diffusing O2and their consumption of it. If cells cannot tol- Cole-Parmer Instrument Co., Vernon Hills, IL (www
erate air-equilibrated media, then media can be deoxy- .coleparmer.com); and Sable Systems International, Inc.,
genated by placing tubes in a boiling water bath for about Henderson, NV (www.sablesys.com).
10 min (or using them soon after being removed from an au- Whatever device is used for mixing, it should be cali-
toclave) and quickly cooling them prior to inoculation. (ii) brated (or its precalibration should be checked) at a stated
A gas mixture containing a low concentration of O2can be back-pressure of source gas (5 or 10 lb/in2 back-pressure is
prepared and sparged through, or swept over, a liquid cul- typical). A fairly constant back-pressure can be achieved by
ture that is shaken or stirred. (iii) Cells can be grown under using a good quality, two-stage regulator for the compressed
a hypoxic gas mixture of sufficient volume that the O2con- gas tanks, or by using in-line pressure regulators between
centration is not significantly changed by the microbes the compressed gas tank and the gas proportioner/blender.
consumption of it during growth. Individual or combined gas outflows are then easily mea-
Strategy (i) has the advantage of simplicity and may sured by using mass flow detectors or simple soap-bubble-
even facilitate harvesting of cells (by aspiration of cells ac- type glass flowmeters. The latter are available in sizes of up
cumulated within a band). However, the growth rate of to 1,000 ml from Alltech Associates, Inc., Deerfield, IL
cells may be slow and cell yields may be limited, because al- (www. alltechWEB.com); and calibration involves measur-
though the cells may position themselves in a band at a ing (with a stopwatch) the rate at which a bubble film is
zone of optimum dissolved 02, a band of cells in a static cul- pushed up the graduated column by gas issuing from the gas
ture is likely to become diffusion limited for oxidizable proportioner/blender. A bubble solution can be purchased
nutrients. Periodic swirling to disperse cells in the band (e.g., Snoop gas leak detector, from Alltech) or prepared
may overcome this diffusion limitation, if tolerated by the by making a dilute solution of dishwashing detergent in
culture. water. Sable Systems (above) also sells equipment for mea-
Strategy (ii) requires only an appropriate gas mixture suring the 0 2 and C02 concentration of gas mixtures down
and sealed culture vessels equipped with ports for inflow to 1 ppm by volume (ppmv) (0.1 Pa). Low concentrations
and outflow of the (filter-sterilized) gas mixture. A gas mix- of O2(10 to 1,000 ppmv [l to 100 Pa]) can also be measured
ture containing a low concentration of O2can be custom- colorimetrically, by using the pyrogallol method (6).
prepared by most commercial suppliers of compressed gases. Strategy (iii) can be achieved a number of ways. A n old-
These are convenient but are considerably more expensive fashioned device to accomplish this is the candle jar,
than preparing the desired mixture in the laboratory-espe- which consists of a gas-tight jar containing the plates and/or
cially so if mixtures containing different O2concentrations tubes to be incubated and a small candle. The candle is lit
are being used. Gas mixtures containing variable amounts just before the jar is sealed and allowed to burn until extin-
of 0 2 are easily prepared by diluting compressed air with N2 guished, a process that consumes some of the oxygen in the
by means of a gas proportioner. For example, compressed air jar and also produces C02,the latter often required by, or
contains about 21% (vol/vol) O2 (balance, almost entirely stimulatory to, microaerophiles. Although somewhat useful
N2): hence, a mixture of compressed air flowing at a rate of for aerotolerant microaerophiles, the candle jar leaves an
50 mL/min with pure NL flowing at a rate of 450ml/min will atmosphere that is still quite rich in O2(ca. 17% O2and 3%
yield a blended gas containing 2.1% O2 delivered at a rate C02 [97]). Hence, it may not be suitable for micro-
of 500 ml/min. aerophiles like Campylobacter, which grow best at 5 to 6%
Gas proportioners are units fitted with two or more grad- 02,except perhaps during primary isolation from feces
uated glass tubes through which individual gases flow, each when accompanied by other 02-consuming colonies on
containing a glass or stainless steel spherical float. The rate plates (97). A better method involves the use of large des-
of flow of gas through the tubes is adjustable by needle iccator jars, aluminum pressure cookers, or polycarbonate
valves and is indicated by the height of the float. The indi- GasPakB jars (BD Diagnostic Systems, Sparks, MD
vidual gases are then merged in a common mixing tube [www.bd.com]) containing, or retrofitted with, at least one
(usually an empty metal tube containing a wire brush or a port that can be used to vacuum evacuate and fill the gas
coiled pipe cleaner to facilitate mixing) before issuing from phase with (or dilute the ambient gas phase to) a defined
the outflow port of the unit. The glass tubes and companion hypoxic atmosphere. A simple procedure for doing this is
floats come in different sizes, depending on the range of gas described in chapter 15.2.65. A n alternative to evacuation
15
Phenotypic Characterization and the Principles
of Comparative Systematics
BRIAN J . TINDALL. JOHANNES SIKORSKI. ROBERT A SMIBERT. AND NOEL R . KRIEG

15.1. AN INTRODUCTION TO SYSTEMATICS ...................... 335


.2. ROUTINE TESTS ......................................... 338
................................. 338
15.2.1. Acetamide Hydrolysis
. ................................... 338
2. Acid-Fast Reaction
. .......................... 338
3. Acidification of Carbohydrates
................................. 338
15.2.3.1. Method 1
. ................................. 339
2. Method 2
........................... 339
15.2.4. Agar Corrosion and Digestion
. 5. Ammonia from Arginine............................... 339
. ........................... 339
6. Arginine Dihydrolase Activity
................................. 339
15.2.6.1. Method 1
. ................................. 339
2. Method 2
. ................................. 339
3. Method 3
15.2.7. Aromatic Ring Cleavage............................... 340
. ................................. 340
8. Arylsulfatase Activity
. 9. Autotrophic Growth on Hydrogen........................ 340
................................. 340
.10. Bacitracin Sensitivity
...................................... 340
.1 1. Bile Solubility
...................................... 340
.12. Bile Tolerance
................................. 340
15.2.12.1. Method 1
. ................................. 340
2.Method2
15.2.13. CAMP Test (Beta-Hemolysis Accentuation) ................ 340
........................... 341
.14. Carbon Dioxide Requirement
................................... 341
.15. Casein Hydrolysis
.................................... 341
.16. Catalase Activity
................................. 341
15.2.16.1.Method 1
. ................................. 341
2. Method 2
. ................................. 341
3. Method 3
................................. 341
15.2.17. Cellular Pigmentation
................................. 341
15.2.17.1. Method 1
. ................................. 342
2. Method 2
. ................................. 342
3. Method 3
................................... 342
15.2.18. Cellulase Activity
................................. 342
15.2.18.1. Method 1
. ................................. 342
2. Method 2
................................... 342
15.2.19. Citrate Utilization
................................. 342
15.2.19.1. Method 1
. ................................. 342
2. Method 2
................................... 342
15.2.20. Coagulase Activity
. . ..................................... 342
2 1 Coccoid Bodies
.......................................... 342
.2 2. Colonies
................................. 342
.2 3. Cysts and Microcysts
................................. 343
15.2.23.1. Method 1
. ................................. 343
2. Method 2
...................................... 343
15.2.24. Dye Tolerance
................................... 343
.2 5. Esculin Hydrolysis
....................................
.2 6. Esterase Activity 343
................................. 344
15.2.26.1. Method 1
. ................................. 344
2. Method 2
........................................... 344
15.2.27. Flagella
.................................. 344
.2 8. Fluorescent Pigment
......................... 344
.2 9. Formate-Fumarate Requirement
............................ 344
.3 0. Gas Production from Sugars
................................. 344
15.2.30.1.Method 1

330
15. Phenotypic Characterization 331

.2.Method2 ................................. 344


.3. Method 3 ................................. 345
15.2.31. Gas Vacuole Production ............................... 345
15.2.31.1.Method 1 ................................. 345
.2. Method 2 ................................. 345
15.2.32. Gelatin Hydrolysis ................................... 345
15.2.32.1. Method 1 ................................. 345
.2. Method 2 ................................. 345
.3. Method 3 ................................. 345
15.2.33. Glycosidase Activity .................................. 345
15.2.33.1. Method 1 ................................. 345
.2. Method 2 ................................. 346
.3. Method 3 ................................. 347
15.2.34. Gram Reaction ..................................... 347
15.2.34.1. KOH Test ................................. 347
.2. Aminopeptidase Test ......................... 347
15.2.35. Hemolysis ......................................... 347
15.2.35.1. Method 1 ................................. 347
.2. Method 2 ................................. 347
15.2.36. Hippurate Hydrolysis ................................. 347
15.2.36.1. Method 1 ................................. 347
.2. Method 2 ................................. 348
15.2.37. Hydrogen Sulfide Production from Thiosulfate .............. 348
15.2.37.1. Method 1 ................................. 348
.2. Method 2 ................................. 348
15.2.38. Hydrogen Sulfide Production from Cysteine ................ 348
.3 9. Indole Production ................................... 348
15.2.39.1. Method 1 ................................. 348
.2.Method2 ................................. 348
.3. Method 3 ................................. 348
15.2.40. Indoxyl Acetate Hydrolysis ............................. 348
.41.Iron-Porphyrin Compounds ............................ 349
.42. 2-Ketogluconate from Glucose Oxidation ................... 349
.4 3. 3-Ketolactose from Lactose Oxidation ..................... 349
.44. Lactic Acid Optical Rotation ............................ 349
.4 5. P-Lactamase Activity ................................. 349
.4 6. Lecithinase Activity .................................. 350
.4 7. Lipase Activity ..................................... 350
15.2.47.1. Method 1 ................................. 350
.2. Method2 ................................. 350
.3. Method 3 ................................. 350
15.2.48. Lysine Decarboxylase Activity .......................... 350
.4 9. Lysozyme Resistance ................................. 350
5 0.Malonate Utilization .................................. 350
.5 1. Meat Digestion by Anaerobes ........................... 350
.5 2. Methyl Red Reaction ................................. 350
.53. Milk Reactions ..................................... 350
15.2.53.1. Method 1 ................................. 350
.2. Method 2 ................................. 350
15.2.54. Motility: Swimming .................................. 350
.5 5. Motility: Twitching .................................. 351
.5 6. Nalidixic Acid Susceptibility ............................ 351
.5 7. Nitrate Reduction and Denitrification ..................... 351
15.2.57.1.Method 1 ................................. 351
.2. Method 2 ................................. 351
.3. Method 3 ................................. 351
15.2.58. Nitrite Reduction .................................... 352
.59. Nucleic Acid Hydrolysis ............................... 352
15.2.59.1. Method 1 ................................. 352
.2. Method 2 ................................. 352
.3. Method 3 ................................. 352
15.2.60. 0/129 Antibacterial Susceptibility ........................ 352
.6 1. Optochin Susceptibility ............................... 352
.6 2. Ornithine Decarboxylase Activity ........................ 352
.63. Oxidase Activity (268) ................................ 352
.64. 0xidation.Fermentation Catabolism ....................... 352
332 w GROWTH

.6 5. Oxygen Relationships................................. 353


15.2.65.1. Aerobes .................................. 353
. ................................. 353
2. Anaerobes
. ........................ 353
3. Facultative Anaerobes
. ............................. 353
4. Microaerophiles
15.2.66. Peptidase Activity ................................... 354
15.2.66.1. Method 1 ................................. 355
.2. Method 2................................. 355
15.2.67. pH Optima ........................................ 355
.68. Phenylalanine Deaminase Activity ....................... 355
.6 9. Phosphatase Activity................................. 355
15.2.69.1. Method 1 ................................. 355
.2. Method 2................................. 356
.3. Method 3
15.2.70. PHB Presence
................................. 356
...................................... 356
..........................
15.2.70.1. Staining Procedures 356
. ........................... 356
2. Chemical Analysis
15.2.71. PHB Hydrolysis .................................... 356
.7 2. Proteinase Activity.................................. 356
.7 3. Pyocyanin and Other Phenazine Pigments .................. 357
.74. Pyrazinamidase Activity ............................... 357
...... 357
15.2.74.1. Method 1 (as Applied to Corynebacterium spp.)
.
15.2.75. Sodium Chloride Optima
............ 357
2. Method 2 (as Applied to Yersinia spp.)
.............................. 357
.7 6. Spores (Endospores).................................. 357
.7 7. Starch Hydrolysis ................................... 357
.7 8. Sulfur Production by Microorganisms ..................... 357
.7 9. Temperature Optima ................................. 358
.8 0. Triple Sugar Iron Agar Reactions........................ 358
15.2.80.1. Sugar Fermentations......................... 358
. ............................. 358
2. Gas Production
.
15.2.81. Urease Activity .....................................
...... 358
3. Hydrogen Sulfide Production from Thiosulfate
358
15.2.81.1. Method 1 ................................. 358
.2. Method 2................................. 358
15.2.82. Voges-Proskauer Reaction ............................. 358
.8 3. X and V Factor Requirements ........................... 358
15.3. SPECIAL TESTS AND METHODS ............................ 358
15.3.1. Fermentation Products ................................ 358
15.3.1.1. Organic Acids and Alcohols by Gas Chromatography... 358
. 2. Organic Acids and Alcohols by HPLC ............. 359
. 3. Hydrogen and Methane by Gas Chromatography
15.3.2. Substrate Utilization Patterns
...... 359
........................... 360
15.3.2.1. Auxanographic Nutritional Method ............... 360
. ......................... 360
2. Multipoint Inoculators
. ................ 361
3. Velveteen for Multiple Inoculation
. ..... 361
4. Commercial System Based on Nutritional Patterns
15.3.3. PAGE Patterns ..................................... 361
. ....... 362
4. Fatty Acid (and Other Hydrophobic Side Chain) Profiles
15.3.4.1. Gas Chromatography of Cellular Long-Chain
................................. 362
Fatty Acids
. 2. MIDI Microbial Identification System for
Identification of Bacteria by Gas Chromatography
......................... 362
of Cellular Fatty Acids
. .. 363
3. Simple Methods for Distinguishing Unresolved Peaks
. ................... 363
4. Release of Ether-Linked Lipids
. ......... 364
5. Thin-Layer Chromatography of Ether Lipids
. .............. 364
6. Gas Chromatography of Ether Lipids
. ................... 364
7. Analysis of Longschain Bases
15.3.5. Lipids ............................................ 364
....................... 364
15.3.5.1. Respiratory Lipoquinones
. ................................ 365
2. Polar Lipids
15.3.6. Analysis of Pigments Soluble in Organic Solvents ............. 365
. 7. Polyamines ........................................ 366
...........................
15.3.7.1. Method 1. Modified 366
. 2. Method 2.................................. 366
15. Phenotypic Characterization rn 333

15.3.8. FTIR ............................................ 366


. 9. MALDLTOF Analysis of Whole Cells .................... 366
15.3.9.1. Whole-Cell MALDI-TOF Analysis: Lower-
..... 366
Molecular-Weight Region (500 Da to 3. 500 Da)
. 2. Whole-Cell MALDI-TOF Analysis: Higher-
... 367
Molecular-Weight Region (2.000 Da to 20. 000 Da)
. 3. MALDI-TOF Analysis of DNA or RNA.......... 367
15.3.10. Multitest Systems.................................... 367
. .
11 Nitrogenase Activity and Nitrogen Fixation ................. 367
15.3.1 1.1. Aerobic Nitrogen Fixers....................... 367
.
.2 Facultative and Anaerobic Nitrogen Fixers......... 367
. ................. 370
3. Microaerophilic Nitrogen Fixers
. 4. Gas Chromatography ......................... 370
15.3.12. Antigen-Antibody Reactions ............................ 370
15.3.12.1. Direct Bacterial Agglutination .................. 370
. ............................. 371
2. Coagglutination
. .......................... 371
3. Latex Agglutination
. 4. Hemagglutination........................... 371
. ........................... 372
5. Quellung Reaction
. 6. Fluorescent-Antibody Tests.................... 372
. .... 372
7. Precipitin Test (for Identification of Streptococci)
15.3.13. RAPD-PCR ....................................... 373
15.3.13.1. PCR Protocol .............................. 373
. 2. PCR Machines and DNA Polymerases............ 373
. ........................... 373
3. Choice of Primers
. 4. Template DNA Concentration and Preparation ...... 373
. 5. Ensuring Reproducibility of the RAPDSPCR Protocol . 373
. .................... 374
6. Agarose Gel Electrophoresis
. .......... 374
7. Scoring of Bands for Raw-Data Extraction
. .............................. 374
8. Data Analyses
15.3.14. Gene Probes ....................................... 374
15.4. MEDIA ................................................. 374
15.4.1. A Medium of King et a1.............................. 374
. 2. Acetamide Agar ..................................... 374
. ........................... 375
3. Arginine Broth of Niven et a1
. ....................... 375
4. Arginine Broth of Evans and Niven
. 5. Autotrophic Growth on Hydrogen ........................ 375
. .......... 375
6. &ospirillum Semisolid Nitrogen-Free Malate Medium
. .............................. 375
7. B Medium of King et a1
. 8. Bile Agar.......................................... 375
. 9. Blood Agar ........................................ 375
.l0. Board and Holding Medium ............................ 376
.11. Burk Medium. Modified............................... 376
.12. Chopped-Meat Broth. Prereduced........................ 376
.............................. 376
.13. Christensen Citrate Agar
................................
.14. Christensen Urea Agar 376
............376
.l 5. Clostridium pasteurianum Nitrogen-Free Medium
.16. Egg Yolk Agar...................................... 377
.17. Haynes Broth...................................... 377
.l8. Hino and Wilson Nitrogen-Free Medium for Bacillus
...........................................
Species 377
.19. Hugh and Leifson Oxidation-Fermentation Medium........... 377
................................ 377
.2 0. Hutners Mineral Base
. . ............ 377
2 1 Leifson Modified Oxidation-Fermentation Medium
.2 2. Litmus Milk....................................... 378
....................... 378
.2 3. Lysozyme Resistance Test Medium
.2 4. Malonate Broth ..................................... 378
.2 5. Milk Medium. Prereduced ............................. 378
..................................
.2 6. Mqillers Broth Base 378
.2 7. Methyl Red-Voges-Proskauer Broth ...................... 378
.2 8. Nutrient Agar ...................................... 378
.2 9. Phenylalanine Agar .................................. 378
.3 0. PY Agar. Prereduced ................................. 378
.3 1. PY Broth. Prereduced ................................ 379
.3 2. Pyrazinamide Medium ................................ 379
.3 3. Seawater. Artificial.................................. 379
334 GROWTH

.34. Simmons Citrate Agar ................................ 379


.35. Soybean-Casein Digest Agar ............................ 380
.36. Thornleys Semisolid Arginine Medium .................... 380
.3 7. Todd-Hewitt Broth. Modified ........................... 380
.38. Triple Sugar Iron Agar ................................ 380
.39. Yoch and Pengra Nitrogen-Free Medium for Klebsiella
Species ........................................... 380
15.5. REAGENTS .............................................. 380
15.5.1. Benedicts Reagent ................................... 380
.2. Buffered Glycerol Mounting Fluid ........................ 380
.3. Charcoal-Gelatin Disks ................................ 380
.4. Developing Solution for Arginine ........................ 381
.5. Ehrlichs Reagent .................................... 381
.6. Formate-Fumarate Solution ............................. 381
.7. Kovfics Reagent ..................................... 381
.8. Lactic Acid Reagents for Optical Rotation .................. 381
.9. McFarland Turbidity Standards .......................... 381
.1 0. Methyl Green for DNase Test ........................... 381
.1 1. Nesslers Solution ................................... 381
.1 2. Nile Blue Sulfate Fat ................................. 382
.1 3. Nitrite Test Reagents ................................. 382
.14. Peptidase-Developing Reagents .......................... 382
.15. pH Indicators for Culture Media ......................... 382
.1 6.PBS ............................................. 382
. l 7. PHB Granule Suspension ............................. 382
.18. Potassium Phosphate Buffer ............................ 383
.1 9. Standard Solutions for Gas Chromatography ................ 383
.20. Williamson and Wilkinson Hypochlorite Reagent ............. 384
15.6. SPRAY REAGENTS FOR THIN-LAYER CHROMATOGRAPHY ..... 384
15.6.1. Sugars ............................................ 384
15.6.1.1. Anisaldehyde-Sulfuric Acid ..................... 384
.2. a-Naphthol-Sulfuric Acid ...................... 384
15.6.2. Ninhydrin (for Lipids Containing Free Amino Groups) ......... 384
.3. Secondary Amines ................................... 384
.4. PeriodateSchiff ..................................... 384
.5 . Dragendorffs Reagent ................................ 384
.6. Molybdophosphoric Acid .............................. 385
.7. Phosphate Reagents .................................. 385
15.7. REFERENCES ............................................ 385
15.7.1. General References .................................. 385
.2. Specific References .................................. 386

During a long-term study of a group of strains. stock cultures variety of strains in liquid nitrogen (106) has shown that
may be subject to genetic variation. Therefore. the cultures this method works well for both aerobes and anaerobes. In
of an organism that are used near the end of a study may not addition. a single (20-p1-culture-volume) capillary is
contain quite the same organism as those used earlier. If cul- opened per strain. and this practice further avoids the risk
tures are properly preserved at the beginning of the study. of contamination. which may be encountered when re-
one can always have a source of the organism with its orig- opening vials.
inal characteristics. It is also advantageous to have a work- In the past decades. our approach to characterizing or
ing culture of a strain as well as a seed stock. which may be identifying prokaryotes has changed considerably. The ap-
drawn upon should the working culture become contami- proach which one takes is often dependent on the nature of
nated . Chapter 47 of this volume outlines a number of the strain(s) being characterized. If it is suspected that a
preservation methods. In practice. however. only a few strain is novel. it may be prudent to initially determine the
methods would be routinely used in a research laboratory. 16s rRNA gene sequence. Based on this information. it is
with freezing probably being the most popular. When freez- usually possible to establish which known genera and
ing cultures. it is worthwhile remembering that standard species show a degree of similarity to the new isolate. It
flip-top (Eppendorf) microcentrifuge tubes are not suitable. should be remembered that 16s rRNA gene sequences
due to the difficulty of opening them without contamina- alone do not allow one to assign a new strain to one of sev-
tion . Avoiding plastic altogether in mechanical freezers is to eral species in which the 16s rRNA gene sequence similar-
be recommended. Small glass vials (such as those used in ities among the strains approach 100%. On the other hand.
autosamplers) with screw caps can easily be sterilized and when dealing with environments where familiar strains may
opened with a minimal risk of contamination . Experience be encountered (and where the taxon is well characterized).
in the Deutsche Sammlung von Mikroorganismen und then it is often possible to rely on the results of the pheno-
Zellkulturen with a glass capillary system for storing a wide typic tests. Irrespective of the approach. the scope of the
15. Phenotypic Characterization 335

methods used should be greater the more comprehensive tests are often not absolutely specific, and additional physi-
the characterization or identification of the organism needs ological tests are usually required for confirmation of the
to be. Despite the advent of molecular methods (including identification. Agglutination of gram-negative enteric rods,
whole-genome sequencing), it is also evident that prokary- for example, by polyvalent Salmonella serum is presumptive
otes do not always give up their secrets easily and a (phe- evidence for a Salmonella strain, but this identification must
notypically or epigenetically) well-characterized strain is always be confirmed by a number of biochemical tests.
almost a prerequisite for helping to make sense of the ge- In performing phenotypic characterization tests, the or-
nomic data. In characterizing or identifying prokaryotes at ganisms used for the inoculation of test media should be
the phenotypic level, certain general characteristics are of from fresh transfers and in good physiological condition.
primary importance for determining the major group to Old stock cultures, which may contain mostly dead or dying
which the new isolate is most likely to belong. The investi- cells, are not satisfactory. Incubate the inoculated test
gator should determine whether the organism is pho- media under conditions that are optimum for the organisms
totrophic, chemoautotrophic, or chemoheterotrophic and or for the characteristic being tested, e.g., optimum temper-
whether it is aerobic, anaerobic, microaerophilic, or faculta- ature, pH, gaseous conditions, and ionic conditions.
tive. Similarly, certain morphological features should be de- Furthermore, apply characterization methods not only to
termined: the Gram reaction and the cell shape (e.g., rod, the organism to be tested but, wherever possible, also in
coccus, vibrioid, helix, or other). The presence of special parallel to strains known to be positive and negative for the
morphological features (e.g., endospores, exospores, true characteristic, as a check on the reliability of the reagents
branching, acid fastness, sheaths, cysts, stalks or other ap- and media.
pendages, fruiting bodies, gliding motility, or budding divi- The methods described in this chapter are based pri-
sion) is valuable. The arrangement of the cells, e.g., cocci oc- marily on methods developed for the characterization and
curring in chains or in irregular clusters, may be helpful. identification of organisms which have usually been iso-
Three physiological tests are of primary importance: the oxi- lated on nutrient-rich media (and often at neutral pH).
dase test, the catalase test, and the determination of whether There is increasing evidence that the number of strains
sugars are oxidized or fermented. The presence of special which may be isolated from a given environment may in-
physiological characteristics (e.g., nitrogen-fixing ability, the crease if lessmutrient-rich media are used (37, 220). Some
ability to degrade cellulose, bioluminescence, the require- of these strains may not grow in the media developed for the
ment for seawater for growth, the production of pigments, or tests described in this chapter, and due consideration should
a heme requirement) may narrow the field of possibilities be given to working with lower concentrations of substrates
considerably. The increasing interest in unusual environ- and/or other components in the media, as well as other ef-
ments means that determining the ranges of temperatures, fects (289). Other problems which may be encountered in-
pHs, and salinities for growth as well as the optima should clude the isolation of obligate acidophiles or alkaliphiles, in
also be a routine part of comprehensive characterization. which shifts in pH required to detect a positive reaction
It is desirable to use an orderly approach to the charac- may be minimal due to the buffering capacity of the growth
terization and identification of a strain, one that is based on medium. Naturally, at extremes of pH the standard indica-
common sense and uses only the tests that are pertinent. tors given in the individual recipes will also not work, and
Avoid a shotgun approach, in which all sorts of tests are alternatives are to be found in chapter 11, Table 1. Despite
performed in the desperate hope that some of them may be tremendous advances in genomics, proteomics, and tran-
helpful. Studies of complete genomes indicate that organ- scriptomics, there appears to be no easy molecular tool for
isms with larger genomes may be more versatile than those determining the optimum temperature, salt concentration,
with smaller genomes (134). The consequences of these ob- or pH, although specific changes in the gene sequences of
servations may also have an impact on the scope of tests proteins may reflect adaptation to extremes of any of these
used in characterizing a novel strain. The table of contents three environmental parameters. In this sense, there is still
and the major keys given in Bergeys Manual of Systematic a need to isolate (see chapter 11) and characterize prokary-
Bacteriology (4, 10, 17, 18, 20) will be of great assistance, as otes from the diversity of natural environments in which
will the general descriptions of the various genera and they can be found (210). Combining the information
species of prokaryotes. Clearly, this type of approach is gained using the methods described in this chapter together
helped by access to the 1 6 s rRNA gene data. However, it with other methods of analysis described in other chapters
should be remembered that some groups of taxa with a low in this book and appreciating prokaryote diversity at differ-
level of 16s rRNA gene similarity may show a relatively ent levels are some of the major goals of prokaryote system-
low degree of physiological diversity (e.g., the anaerobic atics.
clostridia) while others may show greater physiological di- At the end of this chapter is listed a selection of general
versity with a relatively small degree of difference in 16s references ( 1-20) that provide identification schemes for
rRNA genes (e.g., the genera and Nitrobacter, Brady- various groups of bacteria, further information on method-
rhizobium, Afipia, and Rhodopseudomonas). After the initial ology, and additional characterization methods. During the
possibilities have become more restricted, consult the de- coming years the publication of the newer volumes of
tailed keys and tables from various sources (many of which Bergeys Manual of Systematic Bacteriology will certainly pro-
are listed in references 1-4, 7, 8, 10-12, and 15-20) for the vide additional information. A section dealing with charac-
identification of genera and species. In the final steps terization and systematics would not be complete without a
of identification, compare the new strain with, preferably, brief overview of the objectives of systematics.
either the type strain of the suspected species or some other
named strain whose inclusion in the species has been firmly
established. 15.1. AN INTRODUCTIONTO SYSTEMATICS
In many cases, especially with pathogenic prokaryotes, Systematics is not always fully appreciated. While there is
one may be able to make a rapid presumptive identification often a tendency to equate systematics with taxonomy, it is
of a genus or species by the use of antisera. However, such more appropriate to regard the latter as a part of the former.
336 GROWTH

In simple terms, systematics may be considered to be one of physics has reference points for the meter or the kilogram,
the most elementary and comprehensive sciences, because so too does biology, with the nomenclatural types being the
it is an essential aspect in comparing any two elements in reference points for a taxon. In the cases of the species and
biology, whether it be genes, proteins, biochemical path- subspecies, these reference points are represented by type
ways, or organisms themselves. Thus, systematics continues strains (147). Given the central importance of type strains,
to play an important role in an age when it is becoming in- it is important that they be made available as widely as pos-
creasingly easy to sequence whole genomes or to character- sible. The best course of action would appear to be to de-
ize the genes of populations of organisms. posit type strains in a suitable culture collection, which
Systematics has been defined in many ways but may be should be able to maintain the distribution of the strain in
succinctly described as the cradle of comparative biology. the future (83, 141). Clearly, depositing a type strain in
Taxonomy can be clearly defined as encompassing charac- such a way that it is not easy to access is counterproductive
terization, classification, and nomenclature. The character- to the principle behind the deposit of type strains, that of
ization of an organism is no longer bounded by method- making them widely and easily available for comparative
ological barriers, and it is now possible to fully sequence the purposes.
whole genome of a strain, to study individual genes, or to Organisms were initially described by a variety of obser-
examine the genetic information by using amplified frag- vations, although the only way of observing organisms in
ment length polymorphism, random amplification of poly- natural samples was either by studying mass accumulations
morphic DNA (RAPD), and G + C content analysis. How- (as in Winogradsky columns), by using microscopy, by
ever, genes do not exist on their own, and it is becoming studying the results of spoilage, or by studying organisms in
increasingly clear that the study of the biochemical path- connection with a disease. Pure-culture techniques for aer-
ways of an organism, the roles structural elements (proteins obes became available at the end of the 19th century, with
and lipopolysaccharides, etc.) may play in morphology, or pure cultures of anaerobes being available at the beginning
the chemical composition of the cell should be correlated of the 20th century. Staining methods were also developed
with the underlying genetic information. Classification is at this time, with Christian Gram (88) developing the
the arrangement of prokaryotes into groups. Different forms Gram stain. The concept of the significance of the bio-
of classification may have different goals. Organisms may be chemical properties of a microorganism was formulated by
grouped according to their pathogenic potentials (biologi- Orla-Jensen (203, 204), and den Dooren de long (63) was
cal safety levels) or in an arrangement based on more com- also instrumental in developing this concept further (208).
plex theories (i.e., the course of evolution). Nomenclature Thus, until the 1960s most microbiologists relied on mor-
is the naming of those groups. When the groups are species, phological aspects together with biochemical and physio-
genera, and families, etc., then the way they are named or logical properties. The discovery of the structure of DNA
the links between names which have been used in the past during the 1950s was also paralleled by the development of
are governed by an International Code of Nomenclature methods for the study of the chemical compositions of cells
(147). Identification is often considered to be a part of tax- (gas chromatography, nuclear magnetic resonance [NMR],
onomy but is concerned with comparing unknown organ- mass spectrometry, and gel electrophoresis, etc.). This de-
isms with organisms which have already been classified. As velopment laid the basis for the further development, over
such, identification can be carried out only once a taxon- the next SO years, of the modern spectrum of methods
omy has been established. Typically, identification proto- which we know today, many of which are covered in this
cols have the goal of quickly assigning an organism to a book. While the immediate question to most is what meth-
known group by using the minimum number of methods. In ods do we use, one must first examine what we try to
contrast, a novel organism should be characterized as fully achieve in characterizing and classifying prokaryotes.
as possible in order for subsequent identification systems to The ultimate goal of taxonomy is to find the correct
have a reliable basis on which to work. The more reliable order which is present in nature. Although this is often
the characterization and classification, the greater chance called the natural system, the concept of natural has,
one will have of being able to pick identification methods over the centuries, meant different things to different peo-
which both are accurate and have a long-term future. ple (41). Prior to Darwin, the term natural would have
Nomenclature is regulated in prokaryote systematics by referred to the order which was a result of the act of cre-
an official system of registering (or indexing) those names ation. During the 1940s, this term became associated with
which may be used in prokaryote taxonomy via a central- a particular philosophical approach outlined by J. S.L.
ized system. The formal term for registering a name is valid Gilmour, later to become known by the term phenetic, and
publication [of a name]. This system was unique to the finally, the term natural was associated with evolution. Un-
International Code of Nomenclature of Bacteria (but virol- fortunately, some confusion has arisen around the term phe-
ogy has followed this principle) and was introduced in 1980 netic because of the popular belief that it has nothing to do
to combat uncertainties in the application of some 40,000 with evolution and that it deals only with phenotypic data.
names, which had accumulated over the previous -200 This is not so, and the proper definition by Cain and
years. With the introduction of the Approved Lists of Harrison (41) makes clear and unambiguous reference to the
Bacterial Names (240), only 2,000+ names made the grade fact that it deals with overall similarity and may include
into the modern system. While nomenclature is formally both phenotypic and genotypic data. Harrison (97) noted
regulated, it is important to be aware that taxonomy is not that the analogy of genotypic to genetic versus phenotypic
regulated by the code. Thus, the code recognizes a valid to phenetic was unfortunate and misleading. This miscon-
published species name, but the term validly published ception persists most strongly in prokaryotic systematics.
species has no meaning, despite being frequently met with Based on the conceptual work of Gilmour, which has been
in some of the literature. In order for a name to be validly elaborated by Sneath and Sokal (246, 249, E l ) , the phe-
published, the proposal for the name must be accompanied netic approach made a clear distinction between the exact
by a number of criteria (147, 247, 248). Among these cri- ancestor-descendant relationships (phyletic lineages) and
teria is the designation of a nomenclatural type. Just as those groups which have arisen as the result of evolution
15. Phenotypic Characterization w 337

(phyletic groups). Under certain circumstances (largely iar with the principles of numerical taxonomy will have no-
those in which homoplasy, convergent evolution, paral- ticed an interesting trend as full genome sequences became
lelism, gene transfer, or gene conversion does not play a available, with the sheer flood of data being taken as the so-
role), a phenetic system may also reflect the true course of lution. However, that this is not the case is now fully evi-
evolution. In contrast to the phenetic approach (which has dent, and it is becoming increasingly difficult to find more
flavored much of modern microbial systematics), the cladis- than about 100 genes which can be used to cover all taxa
tic approach has been much favored in botany and zoology, (154). When different genes give different results, gene
where bitter and often acrimonious debates have raged over transfer is the simple answer (65), although this may well be
the years on the merits of one system over another. Briefly, an oversimplification ( 140). While zoologists may be con-
the cladistic approach (also known as phylogenetic system- tent to classify all insects or all vertebrates, prokaryote sys-
atics) evaluates the significance of individual characters tematics generally deals with all prokaryotes and in an evo-
and their contribution to delineating the evolutionary his- lutionary context attempts to extend this study back to the
tory of the organisms concerned. The term clade was origin of life itself. It is the latter, the origin of life and the
coined by Huxley (112), but the major contribution to root of the tree of life, which may well be the most elusive
cladistic evaluation is usually attributed to Hennig (who element of all (27). In the absence of a root, it becomes dif-
used the term Phylogenetische Systematik [102]). This ficult to determine whether major groups such as the
methodology centered largely on the use of morphological Bacteria and the Archaea are monophyletic, monophyletic
data. Debates over the advantages of one method over an- groups being the product of rooted trees.
other included topics such as the naturalness of taxa and A central element in any comparative work is the com-
the information content of the classifications (73, 74). parison of like with like. This concept has as much a bio-
One of the present problems is that the term phyloge- logical as a philosophical basis. The idea of like with like
netic has also come to be associated with a number of mean- benefited from a formal definition proposed by Owen, the
ings. Cowan (54) noted different meanings being associated concept of homology. Despite the widespread usage of the
with the terms phylogeny and phylogenetic classification. term homology in the post-Darwinian era, Owen was a firm
However, he refers to Constance (51), who, writing in opponent of Darwins theory and his concept of homology
1964, had not been caught up by the tendency to equate was not formulated with an evolutionary basis. Never-
cladistics with Hennigian phylogenetic systematics. In a theless, this concept has become incorporated into an evo-
modern context, phylogenetics has become associated with lutionary context, although there is continued and regular
the study of gene or protein sequences. The paradox arose debate on the exact use, meaning, and definition of the
in the early days of the use of Sab values for evaluating 16s term. Certainly, in gene or protein sequence-based work
rRNA gene catalogues that this method was taken to be there is a tendency to equate similarity (of the aligned se-
phylogenetic (301,302), whereas Sneath (245) maintained quences) with homology. While it is true that homologous
that it was not cladistic and primarily phenetic. It is inter- proteins will show a degree of similarity, it is also true that
esting that one author makes the distinction between phe- some proteins may be similar but are not the same (i.e., ho-
netic and phylogenetic methods based on whether a pro- mologues). Until recently, BLAST searches were called
gram will allow one to mask a sequence (66). Sneath (245) homology searches. In reality, they determine the most
has indicated that it is sometimes not easy to determine similar sequence(s) in the database without being able to
when phylogenetic means evolutionary, cladistic, or sim- confirm to what extent the sequences being composed rep-
ply genomic. The confusion over how a particular term was, resent the same proteins or genes (i.e., homologues). This
is, or will be used will no doubt continue, so it is probably problem was highlighted by Margoliash et al. (161), but
best to avoid those terms which cause confusion altogether the debate about the misinterpretation of similarity as
and simply divide the methods of classification into three: equating with homology regularly resurfaces. Patterson
overall similarity, character analysis (273), and a combi- (212, 213) has outlined the problem, and Fitch (77) has
nation of both (164). Hillis et al. (104) have also provided also recently aired his views on this topic. It is also becom-
a good overview of how some of the methods used for infer- ing increasingly clear how difficult it is to decide exactly
ring phylogenies from sequences may be classified. what is orthologous, parologous, or xenologous at the gene
Numerical taxonomy is, in its simplest form, the use of or protein sequence level. Lerat et al. (154) have indicated
computers in the taxonomic process. In microbiology, the that it is not easy to unambiguously distinguish the three in
term is often (and very mistakenly) taken to mean the use closely related taxa and have proposed the term synologue.
of computers in the evaluation of phenotypic data, based on This term has also been used by Gogarten (85) in another
phenetic philosophy. In areas such as gene or protein se- context. The problem of identifying homology was clearly
quence analysis, the basic elements of numerical taxonomy articulated by Sneath and Sokal(251), who proposed using
are evident in many alignment programs and in the princi- the term isologue and developed the concept of opera-
ple underlying simple BLAST searches. One of the basic tional homology. It is likely that the problem will continue
principles of numerical taxonomy is that given enough data, to be with us for some time to come. Irrespective of how one
the correct natural groups will be found. Certainly in mi- sees the problem, homology may have slightly different in-
crobiology the use of a wide range of biochemical and phys- terpretations at different levels. However, two elements
iological test data has not proven to be the optimal solu- should always be taken into consideration: those of com-
tion, particularly across higher taxa. However, this should mon evolutionary history and structure and function
not be taken to mean that such data do not reflect evolu- (77). The latter is clearly not a one-dimensional concept,
tion. If we mistakenly assume that all cocci are a mono- and the determination of homology should not be reduced
phyletic group, then it is not the data that are incorrect but to a simple linear, statistical relationship (301,302). Clearly
rather our assumptions that led us to these conclusions. the correct use of terminology in the literature as well as
Siefert and Fox (236) have indicated how complex the in- correctly communicating these concepts to younger scien-
terrelationships are between rods and cocci in relation to tists, yet to be tainted by misuse, would help to reduce the
16s rRNA gene sequence-based groupings. Anyone famil- confusion.
338 GROWTH

It is often claimed that sequence data are the only means a better appreciation of its significance. Simpson advocated
of establishing any evolutionary dimension for prokaryotes, taking a similar course in zoology (239). In this respect, the
with the work of Zuckerkandl and Pauling (308) being the methods employed in prokaryote systematics will continue
quoted source. However, these authors were well aware that to expand, and some methods may fall by the wayside (186,
certain aspects of the phenotype may be useful in determin- 288). However, just as understanding the functioning of the
ing at least parts of the evolutionary tree while also ac- ribosome requires an understanding of the three-dimen-
knowledging that one must consider the results from indi- sional interactions of the various RNAs and proteins (177),
vidual sequences in the context of the evolutionary history so too is there a need for the continued reference to the
of the whole organism. It is this history which may not be so wider range of properties of an organism.
easy to decipher. In other cases, one gene may not be in ac- The methods described in this chapter deal primarily
cord with others (25) or different treatments of the raw data with the biochemical, physiological, and chemical proper-
may give different results (152), and it is evident that the ties of prokaryotes. Genetic aspects are covered in chapter
original goal to test an evolutionary hypothesis (302) is 11.5 and sections IV and V. One chapter deals specifically
often forgotten. Clearly, grouping all anoxygenic pho- with accessing data gained by genomic methods (chapter
totrophs into a single (monophyletic) group is not consis- 35), and another deals with the thorny question of phylo-
tent with 16s rRNA gene groupings. However, the distribu- genetic trees (chapter 36). The reader is also referred to
tion of genes coding for (bacterio)chlorophyll is proving to more comprehensive reference works, such as The
be a tantalizing evolutionary puzzle, with some groups of Prokaryotes, Bergeys Manual of Systematic Bacteriology, and
organisms consistently having such genes (cyanobacteria, specific chapters in these volumes (10, 17, 18, 20, 36, 118,
heliobacteria, and chlorobiaceae) while others, such as the 137, 156, 257). The International Journal of Systematic Bac-
alpha-, beta-, and gammaproteobacteria, seem to have a teriology, now renamed the International Journal of Systematic
patchwork distribution. Clearly, in some groups photo- and Evolutionary Microbiology, is one of the most important
trophic growth is a characteristic of all of the members and journals dealing with modern prokaryotic systematics. The
in others it is not. Morphology is as unpredictable; spiro- reader is also referred to the List of Prokaryotic Names with
chete morphology is group specific, but cocci may be found Standing in Nomenclature, compiled by Jean EuzCby
across the breadth of modern taxa. The presence of various (www.bacterio.cict.fr/), which covers all validly published
biochemical pathways such as methanogenesis may define names and includes references to the original publications,
fairly large, coherent groups, while the ability for nitrate re- as well as a wealth of additional information.
duction seems to surface at random. Given the fact that the
phenotype of an organism is the result of the underlying ge-
netic information, such problems will also be reflected at the 15.2. ROUTINE TESTS
genetic level. In some cases genes may be present but ex-
pressed either only to a low degree or not at all. 15.2.1. Acetamide Hydrolysis
Wilson et al. (300) stated: To test for acetamide hydrolysis, streak the surface of an
acetamide agar slant (section 15.4.2) with a sample from a
We have already alluded to the observation that amino acid dilute suspension. Incubate for up to 7 days. Red or magenta
and nucleotide sequences evolve at fairly steady rates that
seem virtually independent of rates of organismal evolution.
signals a positive test; no color change signals a negative
Molecular evolutionists were slow to recognise this surpris- test.
ing and intriguing fact. They had assumed that organismal Examples: positive, Pseudomonas ueruginosa; negative,
evolution depends on sequence evolution in proteins and Pseudomonas jluorescens.
expected a simple relationship between the two types of
evolution. In particular it was expected that morphologi- 15.2.2. Acid-Fast Reaction
cally conservative organisms should have experienced See chapter 2.3.6 for two staining methods for the acid-fast
slower macromolecular evolution than organisms that had reaction. A positive reaction indicates the presence of a
evolved unusually rapidly at the morphological level. To
date, however, there is no convincing evidence that pro-
member of the genus Mycobacterium (which is strongly acid
teins or nucleic acids of conservative creatures are conser- fast), Nocardia, or Rhodococcus (members of the last two
vative in regard to their amino acid or nucleotide se- genera are partially acid fast) (17).
quences.
15.2.3. Acidification of Carbohydrates
It is the regularity of biological systems which has arisen as
the result of evolution that remains to be documented. 15.2.3.1. Method 1
Tracking those patterns in nature at one level can help to Incorporate a pH indicator into the medium during its
reinforce the information gathered at another. In the ab- preparation. For a list of the most commonly used indicators
sence of a wealth of data on biochemical pathways and their and their properties, see Table 8. In cases in which an indi-
end products or the structural components of prokaryotes, cator may be toxic to the bacteria or may be reduced during
little progress could have been made in elucidating prokary- their growth, add drops of the indicator (0.02 to 0.04% al-
ote genomes. Using data from annotated genomes can, in coholic solution) to the culture after it has grown to maxi-
return, help to locate postulated new pathways or to con- mum turbidity.
firm the molecular infrastructures underlying the results of A relatively high concentration of sugars, e.g., 1 to 2%
simple biochemical tests. The presence of genes for bacteri- (wtlvol), is usually added to the basal medium. O n the
ochlorophyll synthesis does not confirm that they are ex- other hand, the peptone concentration in the basal medium
pressed to any appreciable degree. Systems biology seeks to should be kept as low as possible while still allowing good
integrate the whole and steers a course similar to a broadly growth to minimize buffering and alkali formation. This is
based approach to systematics, in which information from particularly important for characterizing aerobic organisms,
different levels (gene sequence, protein sequence, biochem- which produce comparatively little acidity when oxidizing
ical pathway, and end product) can be integrated to provide sugars. For instance, in determining the acidification of
15. Phenotypic Characterization 339

sugar media by various Aquapirillurn species, the peptone 15.2.4. Agar Corrosion and Digestion
level in the medium should not exceed 0.2% and a pH in- Colonies of agar-corroding or agar-pitting bacteria appear as
dicator, such as phenol red, that will change color with only if they are in a shallow pit in the surface of the agar
slight acidity should be used (114). medium. The term corrosion is used rather than digestion
In general, sugar-containing media to be used for testing because the agar seems not to be softened or liquefied.
acidification can be sterilized by autoclaving at 115 to Examples are Eikenella corrodens, Bacteroides ureolyticus (this
118C for 10 to 15 min. However, heating is known to alter organism is related to members of the genus Campylobacter
some sugars, especially in an alkaline solution, or to convert [280]), Kingella kingue, and various Moraxella species.
one sugar to another (169). Consequently, when using xy- Several species of gliding bacteria are able to digest agar
lose, lactose, sucrose, arabinose, trehalose, rhamnose, or sa- by means of agarolytic enzymes. For example, the colonies
licin, it is prudent to sterilize the complete sugar-containing of Nannocystis exedens may deeply corrode the surface of
medium by filtration or to sterilize a 5 or 10% stock solution the medium and may even penetrate the medium, produc-
of the sugar separately by filtration and then add appropri- ing holes or tunnels in the agar. Chondromyces and Poly-
ate amounts aseptically to portions of the autoclaved, angium species can pit, erode, and penetrate agar media.
cooled basal medium (1). Vibrio alginolyticus and some Cytophga species can soften
Some organisms, such as Neisseria spp., show acidity best or totally liquefy agar gels, whereas others, such as Cyto-
when grown in a semisolid medium. phga fermentans, merely produce gelase fields and craters
Care should be taken to ensure that the basal medium to on agar plates.
which the carbohydrates are added does not contain fer-
mentable or oxidizable sugars, and control media without 15.2.5. Ammonia from Arginine
any added carbohydrates should always be included during Inoculate arginine broth (see sections 15.4.3 and 15.4.4).
testing of cultures. Moreover, basal media should not con- Also inoculate a control lacking arginine. After incubation
tain the oxidizable salts of organic acids, such as malate, for 2 to 3 days, test samples of the culture on a spot plate
pyruvate, or succinate, because oxidation of these sub- with Nesslers solution (section 15.5.11). A positive test is
stances by the test organism will cause an alkaline reaction indicated by yellow or orange compared with the control.
that may obscure acidification due to carbohydrate oxida- Examples: positive, Enterococcus fuecalis; negative, Strep-
tion or fermentation. tococcus salivurius.
With cultures grown in defined media with an ammo-
nium salt or nitrate as the major or sole nitrogen source, the 15.2.6. Arginine Dihydrolase Activity
utilization of the ammonium salt will cause a decrease in pH
and the utilization of the nitrate will cause an increase in pH 15.2.6.1. Method 1
(168). This effect may lead to incorrect conclusions about The following method is widely used to distinguish among
the acidification of sugar media. For instance, almost all of members of the family Enterobucteriaceae. Inoculate MGllers
the acidity produced in cultures of the aerobe Azospirillum broth base (section 15.4.26) supplemented with 1% L-arginine
lipoferum grown in a defined fructose-containing medium is monohydrochloride (or 2% of the DL form). Also inoculate
due to the utilization of ammonium sulfate and not to the a control lacking arginine. After inoculation, overlay the
production of organic acids from the fructose (84). broth with 10 mm of sterile mineral (paraffin) oil. Examine
daily for 4 days. A positive test is indicated by violet or red-
15.2.3.2. Method 2 dish violet due to an increase in pH. Weak reactions are in-
To provide a more quantitative measurement of acidification, dicated by bluish gray.
use a pH meter to measure the pH of the culture after it has Examples: positive, Enterobacter cloacae; negative, Proteus
grown to maximum turbidity. Use a long, thin, combination vulgaris.
pH electrode, which can be inserted directly into a culture
tube. Affix a flat rubber washer with a diameter larger than 15.2.6.2. Method 2
that of the culture tube to the upper portion of the electrode The following method is suitable for a wide variety of facul-
to prevent the electrode tip from striking the bottom of the tatively anaerobic bacteria. Inoculate Thornleys semisolid
culture tube when the electrode is inserted. When determin- medium containing arginine (see section 15.4.36) and also
ing the pHs of a number of cultures of pathogenic bacteria, a control lacking arginine. After stab inoculation, seal the
dip the electrode into a beaker of distilled water between cul- medium with a layer of sterile melted petrolatum and incu-
tures. This water should later be autoclaved. When finished bate. A positive test is indicated by a change from yellow-
with the electrode, rinse it with a suitable disinfectant (e.g., orange to red within 7 days due to an increase in pH.
3% hydrogen peroxide or 70% ethanol).
For anaerobes cultured in prereduced PY broth (section 15.2.6.3. Method 3 (258)
15.4.31) containing carbohydrates, the oxygen-free carbon The following method is suitable for aerobic and facultative
dioxide used to purge the tube during inoculation will usu- bacteria and is based on the direct measurement of the dis-
ally lower the pH of the medium to 6.2 to 6.4. Con- appearance of arginine. Make a dense suspension of the bac-
sequently, pHs of PY carbohydrate broth cultures are usually teria in 0.033 M phosphate buffer (pH 6.8) (see section
interpreted to indicate acidification when they are 5.5 to 15.5.18). Purge 4 ml of the suspension by bubbling nitrogen
6.0 (weak acid) or below 5.5 (strong acid). For PY broth through the suspension for several minutes, and add 1 ml of
cultures containing arabinose, ribose, or xylose, a pH of 5.7 0.001 M L-arginine monohydrochloride. After purging
or below usually indicates acidification, because the sterile again, stopper the tubes, incubate them for 2 h, and heat
medium purged with carbon dioxide may have a pH of 5.9 them at 100Cfor 15 min. After removing the cells by cen-
after 1 to 2 days of incubation (3). In any case, the pH of trifugation, determine the concentration of arginine in the
cultures in PY broth lacking any carbohydrate should be de- supernatant by the method of Rosenberg et al. (224) as fol-
termined as a control, because acids may be formed from lows. Mix 1 ml of the sample with 1 ml of 3 N NaOH, 2 ml
peptones in certain cases. of developing solution (see section 15.5.4), and 6 ml of
340 H GROWTH

water; read the tubes at 30 min against a blank prepared growth on nitrogen-free medium, omit NH4C1and incubate
without arginine by using a colorimeter equipped with a the culture under an atmosphere of 2% (vol/vol) 02,10%
green filter (540 nm); and compare the readings with those C02, 10% H2, and 78% N2 or heterotrophically under 2%
obtained with an uninoculated control containing arginine. (vol/vol) O2and 98% N2 (160).
A positive test is indicated by the disappearance of some or Examples: positive, Xanthobacter jluuus; negative, Esche-
all of the arginine. richia coli.
15.2.7. Aromatic Ring Cleavage (207) 15.2.1 0. Bacitracin Sensitivity
Grow the culture in chemically defined medium containing Use sterile commercially available differentiation (not sen-
an aromatic substrate such as 0.1% sodium p-hydroxyben- sitivity) disks (Difco Laboratories, Detroit, MI, and BD
zoate as the carbon source. (Also grow it on a yeast extract [BBL] Microbiology Systems, Cockeysville, MD) or sterile
agar without the aromatic substrate to determine whether filter paper disks impregnated with 0.04 U of bacitracin
the enzymes are constitutive or inducible.) Scrape growth (Sigma Chemical Co., St. Louis, MO). Place a disk on an
from the agar, and suspend it in 2 ml of 0.02 M Tris buffer inoculated blood agar plate (see section 15.4.9), and incu-
(pH 8.0). Shake the tubes with 0.5 ml of toluene, and add bate for 24 h. A positive test is indicated by a zone of
0.2 ml of either 0.1 M catechol or 0.1 M sodium protocate- growth inhibition around the disk.
chuate. A bright yellow color appearing in a few minutes Examples: positive, Streptococcus pyogenes; negative,
indicates meta-type cleavage. If no color appears, shake the other beta-hemolytic streptococci.
tubes for 1 h at 30C. Add solid (NH4)2S04to saturation,
adjust the pH to approximately 10 by adding 2 drops of 5 N 15.2.1 1. Bile Solubility
ammonium hydroxide, and then add 1 drop of freshly pre- Centrifuge a 24-h culture grown in 10 ml of Todd-Hewitt
pared 25% (wtlvol) sodium nitroprusside (nitroferri- broth (see section 15.4.37), and discard the supernatant
cyanide). Deep purple indicates ortho-type cleavage. into a flask of disinfectant. Suspend the cells in 0.5 ml of
Examples: meta-type cleavage, Delftia acidovorans 0.067 M phosphate buffer (pH 7.0) (see section 15.5.18).
(Pseudomonas acidouorans); ortho-type cleavage, Pseudo- Add 0.5 ml of 10% sodium deoxycholate, and incubate the
moms fluorescens; negative, Eschen'chia coli. mixture at 37C for 15 to 30 min. If the suspension becomes
clear, the test is positive.
15.2.8. Arylsulfatase Activity Examples: positive, Streptococcus pneumonia; negative,
Aseptically add a sufficient amount of a filter-sterilized 0.08 other alpha-hemolytic streptococci.
M solution of tripotassium phenolphthalein disulfate to a
sterile liquid medium to give a final concentration of 0.001 15.2.1 2. Bile Tolerance
M. Media containing methionine as the sole source of sul-
fur are best for the synthesis of arylsulfatase; sulfur sources 15.2.12.1. Method 1
such as sulfate, sulfite, thiosulfate, or cysteine may repress Streak a plate of bile agar (section 15.4.8). Compare the
synthesis (20a, 170). Inoculate the medium, incubate it for growth on the plates with that occurring in the absence of
7 days, and add 1 N NaOH or 1 M Na2C03,drop by drop. the ox gall.
A positive test is indicated by faint pink to light red. A n Examples: positive (10 and 40% bile), Enterococcus fae-
uninoculated control similarly treated should remain color- calis; positive (10 but not 40% bile), Streptococcus saliuarius;
less. negative (neither 10 nor 40% bile), Streptococcus dysgalactiae.
Examples: positive, Proteus rettgeri; negative, Chromo-
bacterium uiolaceum. 15.2.12.2. Method 2
Use method 2 for anaerobes (7). Inoculate PY broth (sec-
15.2.9. Autotrophic Growth on Hydrogen (159) tion 15.4.31) containing 2% ox gall and 1% glucose.
Prepare the medium (see section 15.4.5) in either large- Inoculate tubes with a Pasteur pipette while flushing them
volume bottles, which can be fitted with aluminium seals with oxygen-free carbon dioxide. Stopper the tubes, and in-
and a rubber stopper, or in suitable small conical flasks (100 cubate. Observe the growth response, and compare it with
ml), which can be placed in an anaerobic jar (see section that in the absence of the ox gall.
15.2.65). The gas phase may be mixed either externally (in Examples: growth, Prewotella oralis; no growth, Prevotella
the case of aluminum seal type bottles) or internally by melaninogenica.
evacuation and flushing with gas as described in section
15.2.65. The principal gases are H2, 02, C02,and N2, and 15.2.1 3. CAMP Test (Beta-Hemolysis Accentuation)
they may be mixed in their relative proportions as outlined The CAMP test (the acronym derives from the originating
in section 15.2.65 (see also Table 4). CAUTION: hydrogen authors' names) has been used mainly in the identification
and oxygen are flammable gases, and the hydrogen-oxygen of Streptococcus agaluctiae and of Listeria monocytogenes and
mixtures should not be brought into contact with open flames Listeria seeligeri, but it might be useful for other bacteria as
or sparks. When using conical flasks in an anaerobic jar, the well.
mouths of the flasks must be stoppered with a gas-permeable Make a single streak of a strain of beta-hemolysin-
material, such as commercially available silicon stoppers. producing Staphylococcus aureus (e.g., ATCC 25923) across
Other forms of stoppers may encourage the growth of the middle of a plate of blood agar (containing sheep eryth-
bacteria and fungi under the damp conditions generated in rocytes that have been washed to remove natural antibod-
the jar. ies to hemolysins). Make a single streak of each test organ-
For the chemolithotrophic growth of aerobes, incubate ism perpendicular to but not quite touching the
the culture under an atmosphere of 10% (vol/vol) 02, 10% Staphylococcus aureus streak (about 3 to 4 mm away). For a
C02,60% HI, and 20% N2.For heterotrophic growth, sup- positive control, make a similar streak of a reference strain
plement the mineral medium with an appropriate carbon of Streptococcus agalactiae, Listeria monocytogenes, or Listeria
source (0.2% carbohydrate or 0.1% organic acid). For seeligeri, and for a negative control, make a streak of a refer-
15. Phenotypic Characterization w 341

ence strain of Streptococcus pyogenes or Listeria innocua (32). may occur at or near the surfaces of semisolid media. For
Incubate the plate aerobically overnight. A positive test is broth cultures, add 0.5 ml of 3% hydrogen peroxide to 0.5
indicated by the appearance of a crescent- or arrowhead- ml of culture and observe for continuous bubbling. Note:
shaped zone of enhanced hemolysis at the juncture of the media containing blood must not be used for catalase tests,
test organism and the Staphylococcus aureus streak. because blood contains catalase activity unless the blood
The test has also been used in the identification of has been heated (see section 15.2.16.3). Also note that
Listeria ivanovii; in this case, however, a strain of some bacteria (e.g., certain lactic acid organisms) make a
Rhodococcus equi (e.g., NCTC 1621) is substituted for nonheme pseudocatalase in media containing little or no
Staphylococcus aureus. Listeria ivanovii gives a positive reac- glucose (293). Pseudocatalase production can be prevented
tion, whereas Listeria monocytogenes and Listeria seeligeri give by incorporating 1% glucose into the medium. When
a negative reaction (32). anaerobes are tested for catalase activity, it is important to
expose the cultures to air for 30 min before adding peroxide
15.2.1 4. Carbon Dioxide Requirement (7).
Some aerobic, microaerophilic, or facultatively anaerobic Examples: positive, Staphylococcus epidemidis; negative,
chemoheterotrophs are characterized by a requirement for Lactococcus lactis.
3 to 15% C02 (vol/vol) in the gaseous atmosphere in the
culture vessel. Such bacteria are called capnophilic bacte- 15.2.1 6.2. Method 2
ria. The COz is required to initiate growth; after the organ- The following method eliminates problems of penetration
isms have begun to grow in a liquid or semisolid medium, that might occur when peroxide is added to colonies on a
they usually can provide themselves with COZ from their plate or to growth on a slant. Scrape the growth from a slant
own metabolism. However, when growing in direct contact or plate with a nonmetallic instrument, and suspend it in a
with the gaseous atmosphere, as on the surface of a solid drop of 3% hydrogen peroxide on a slide. Examine immedi-
medium, they may continue to require a supply of exoge- ately and at 5 min for bubbles, either macroscopically or
nous COz. with a low-power microscope (or hand lens). If the catalase
Some examples of capnophilic bacteria are as follows: activity is weak, a coverslip placed over the wet-mount
Capnocytophga ochrucea, facultative anaerobe that requires preparation can help to capture the bubbles.
5% COz (109); Campylobacmjejuni and other Campylobacter
species, microaerophiles that require 3 to 5% C02 (241); 15.2.1 6.3. Method 3
Neisseria gonmhoeae, aerobe that requires 3 to 10% COz, Use the following method for certain bacteria that can
(283); BweUa ovis and some strains of Brucella abortus, aer- make catalase only if grown on a heme-containing medium,
obes that require 5 to 10% COz (52) (note that Bwella ovis e.g., certain lactic acid bacteria (293). To sterile blood agar
and Brucella abortus may be treated as biovars within the base (section 15.4.35) containing 1% glucose to inhibit
species Bwella melitensis, although the International Com- pseudocatalase formation, add 5% (vol/vol) of a 1:l mixture
mittee on Systematics of Prokaryotes subcommittee dealing of defibrinated blood (section 15.4.9) and sterile water.
with this genus is not in full agreement with this solution); Heat the medium at 100Cfor 15 min to inactivate blood
some strains of Streptococcuspneumnim,Streptococcusmutans, catalase, cool to 45 to 50C, and dispense into plates. Test
and Streptococcusanginosus (which includes Streptococcusmil- growth on the plates directly with 3% hydrogen peroxide,
leri), facultative anaerobes that require 5% C02(94). or use method 2.
There are some bacteria that do not have an absolute re-
quirement for exogenous C02but whose growth is never- 15.2.1 7. Cellular Pigmentation
theless markedly stimulated by C02.For instance, Neisseria Cells of many prokaryotes are strongly pigmented, with col-
meningitidis grows much better when 5 to 8% C02 is pro- ors ranging from black, red, and yellow to green. Defining a
vided (283). Similarly, many streptococci grow better in the simple absorption spectrum for the living cells may provide
presence of an atmosphere enriched with COz (94). useful information on the nature of the pigments. Most
dual-beam scanning spectrophotometers, which measure
15.2.1 5. Casein Hydrolysis light in the UV to visible range, cover the wavelengths
Combine sterile (autoclaved) skim milk at 50C with an from about 200 to 900 nm. In some cases, namely, those of
equal volume of double-strength nutrient agar (see section bacteriochlorophyll b-producing members of the anoxy-
15.4.28) or other carbohydrate-free agar medium at 50 to genic phototrophic bacteria, a spectrophotometer which
55C. Incubate streaked plates for up to 14 days, and look measures to about 1,200 nm is needed in order to record the
for clear zones surrounding the growth. Confirm by flooding peak that occurs at about 1,015 to 1,035 nm. A number of
the plates with 10% HC1. Note: acid production from the methods have been described for measuring the absorption
lactose in the milk may in rare instances inhibit the casein spectra of cells. One of the problems which needs to be
hydrolysis and necessitate prior dialysis of the skim milk. countered is the light-scattering effect of intact cells.
Examples: positive, Paenibacillus polymyxa (Bacillus poly-
myxa); negative, Paenibacillus maceruns (Bacillus mucerans). 15.2.1 7.1. Method 1
Take a liquid culture, or make a suspension of the cells in a
15.2.1 6. Catalase Activity suitable medium. Filter the cell suspension through a glass
microfiber filter (Whatman GF/C). Filter either water or
15.2.1 6.1. Method 1 the medium used for suspending the organisms through a
Inoculate a nutrient agar slant (see section 15.4.28) or second, blank filter. Either alter a cuvette (plastic dispos-
other medium lacking blood. After incubation, trickle 1 ml able cuvettes are usually suitable) to take a strip of the fil-
of 3% hydrogen peroxide down the slant. Examine immedi- ter, or cut the filters so that they fit inside of an intact cu-
ately and after 5 min for the evolution of bubbles, which in- vette. Place the blank filter in the reference beam of the
dicates a positive test. Alternatively, add a few drops of 3% spectrophotometer and the filter with the bacterial suspen-
peroxide to colonies on a plate or to the heavy growth that sion in the measuring beam.
342 GROWTH

15.2.17.2. Method 2 Examples: positive, Enterobacter aerogenes; negative,


Suspend a small volume of a liquid culture or colonies from Escherichia coli.
a plate in a 50% solution of sucrose. Suspend the cells well
in order to avoid aggregates. Use 50% sucrose as the refer-
15.2.19.2. Method 2
ence blank. From a dilute suspension, streak the entire surface of a slant
of Christensen citrate agar (section 15.4.13). Incubate for
15.2.1 7.3. Method 3 up to 7 days. A positive test is indicated by red or magenta.
If an ultrasonic unit is available, this unit may be used to Note: a positive reaction indicates that citrate is used but
disrupt the cells. Be careful not to overheat the cells, which not necessarily as the sole carbon source; i.e., an organism
may have an adverse effect on some of the natural pigment could give a positive reaction on Christensen agar and a
protein complexes. negative reaction on Simmons' citrate agar.
Various pigments absorb at characteristic wavelengths.
Carotenoids typically absorb in the region of 400 to 600 nm 15.2.20. Coagulase Activity
(31,230). Various bacteriochlorophylls (in intact cells) absorb Mix one loopful of growth from an agar slant, 0.1 ml of broth
over the range from 590 to 1,100 nm. Flexirubins are charac- culture, or a single colony from an agar plate with 0.5 ml of
terized by the fact that they undergo a reversible shift in the undiluted rabbit plasma or plasma diluted 1:4 with saline.
absorption spectrum at different pHs (230). At neutral pH, Incubate at 37"C, and examine at 4 and 24 h. A positive test
flexirubins are yellow, but at alkaline pHs they are blue. This is indicated by a solid clot or a loose clot suspended in the
method should not be confused with a test for carotenoid-like plasma. Granular or ropey formations are inconclusive.
pigments first described by Molisch (174). In the presence of Examples: positive, Staphylococcus aureus; negative,
concentrated sulfuric acid, carotenoid pigments become blue. Stuphylococcus epidemidis.
More information can be obtained by extracting the pigments
and subjecting them to more-detailed analysis. Methods suit- 15.2.21. Coccoid Bodies
able for the extraction of pigments that are soluble in organic Use phase-contrast microscopy to examine cultures that are
solvents are given below (see section 15.3.5). held static (not shaken during growth) for up to 4 weeks.
Coccoid bodies may occur at as early as 2 or 3 days. A pos-
15.2.1 8. Cellulase Activity itive test is indicated by a predominance of round, refractile
forms which have diameters greater than those of the origi-
15.2.18.1. Method 1 (111) nal cells and which lack a thickened cell wall. Many of the
The following method of preparing cellulose gives the best forms have a discrete, dark peripheral region of cytoplasm in
form of native cellulose for testing cellulolytic activity; for an otherwise empty-appearing cell. Coccoid bodies occur
other methods, see references 15 and 16. Incorporate finely among certain spirilla, vibrios, and campylobacters.
divided cellulose into appropriate carbohydrate-free agar Examples: positive, Aquaspirillum itersonii; negative,
media. To prepare the cellulose, wet grind 3% (wtlvol) Aqwlspirillum serpens.
Whatman no. 1 filter paper in a pebble mill as follows. Place
30 g of the paper (torn into small pieces) into a porcelain 15.2.22. Colonies
jar (ca. +liter capacity) with 1 liter of water; add enough Measure the colony diameter in millimeters, describe the
flint pebbles (porcelain balls are not as satisfactory) that the pigmentation, and describe the form, elevation, and margin
liquid just covers them; roll the jar for 24 h at 74 rpm or as indicated in Fig. 1. Low-power microscopy (or a hand
until a very fine state of suspension has been achieved and lens) may be necessary for observation of the margin. Also
the suspension has become viscous; stop before the viscos- indicate whether the colonies are smooth (shiny, glistening
ity decreases and copper-reducing substances appear. (To surface), rough (dull, bumpy, granular, or matte surface), or
test for the latter, remove 1 ml of the suspension and test as mucoid (slimy or gummy appearance). Record the opacity
described in section 15.2.42 with 1 ml of Benedict's reagent of the colonies (transparent, translucent, or opaque) and
[section 15.5.11.) A positive test is indicated by the appear- their texture when tested with a needle: butyrous (butter-
ance of clear zones around the colonies on the cellulose like texture), viscous (gummy), or dry (brittle or powdery).
agar. Long periods of incubation may be required. Describe the colonies from both young and old cultures.
Examples: positive, Cellulomonas species; negative, Esch- The medium, age of the culture, gaseous conditions, expo-
erichia coli. sure to illumination, and other culture conditions may af-
fect the colony characteristics.
15.2.18.2. Method 2
Method 2 is less sensitive than method 1. Place a strip of 15.2.23. Cysts and Microcysts
Whatman no. 1 filter paper in tubes of carbohydrate-free Examine cultures daily by phase-contrast microscopy.
broth before autoclaving. A portion of the strip should ex- Initially rod-shaped cells become spherical or ovoid with
tend above the level of the broth. A positive test is indi- thickened walls in older cultures. These forms may be opti-
cated by partial or complete disintegration of the paper strip cally dense, or they may be refractile. They do not have the
during the growth of the culture. heat resistance of endospores (section 15.2.76), except for
the cyst-like forms made by certain Nocardia species, but
15.2.1 9. Citrate Utilization they are extremely resistant to desiccation. These forms of
Arotobacter strains are termed cysts. Those in the mycobac-
15.2.1 9.1. Method 1 teria (order Myxococcales) are termed microcysts; here, the
Prepare a dilute suspension of the organisms in sterile water term cyst refers to the sporangium, if any, which contains
or saline. Make a single streak up a slant of Simmons' cit- the microcysts. These forms of Nocardia strains are termed
rate agar (section 15.4.34). Incubate for up to 7 days. Blue microcysts or chlamydospores.
indicates the utilization of citrate as the sole carbon source The following methods can be used to compare the lev-
(i.e., a positive test). els of desiccation resistance of cyst-enriched cultures of an
15. Phenotypic Characterization 343

FORM

Punctiform Circular Filamentous Irregular Rhizoid Spindle

ELEVATION - 0

Flat Raised Convex Pulvinate Umbonate

MARGIN

Entire Undulate Lobate Erose Filamentous Curled


FIGURE 1 Diagram illustrating the various forms, elevations, and margins of bacterial colonies.
(Adapted by permission of the McGraw-Hill Book Co. from reference 214.)

organism to those of cultures lacking cysts. See also chapter of Bordetella species have used paper strips soaked in basic
2.3.8 for methods of staining cysts. fuchsin (0.0005%), methyl violet (0.00025%), pyronin G
(0.000125%), safranin 0 (0.000125%), and thionine
15.2.23.1. Method 1 (143) (0.000125%) (117).
Spread suspensions of the cells onto plates of media con-
taining 2% agar, and incubate until the agar has dried com- 15.2.25. Esculin Hydrolysis
pletely to a thin film. Cut the agar film into pieces with Grow cultures in broth or agar medium supplemented with
sterile scissors, and transfer each piece aseptically to a 30-ml 0.01% esculin and 0.05% ferric citrate. A positive test oc-
screw-cap vial half full of silica gel desiccant. Each vial curs when the medium becomes brownish black as a result
should contain a sterile paper disk or funnel to separate the of a reaction between 6,7-dihydroxycoumarin and Fe3+.
agar film from the desiccant. Store vials in the dark at room Examples: positive, Enterococcus faecalis ; negative,
temperature. At various periods, remove a piece of agar film Saeptococcus mitis.
from a vial, place it upon fresh agar culture medium, and in-
cubate for up to 7 days to see whether growth occurs. 15.2.26. Esterase Activity
See also Lipase Activity (section 15.2.47).
15.2.23.2. Method 2 (226) The hydrolysis of ester linkages between fatty acids and
Prepare a suspension of the cells with sterile distilled water, 4-methylumbelliferone results in the liberation of the latter
and then prepare a series of 10-fold dilutions of this suspen- compound, which is fluorescent when exposed to UV light
sion. Transfer 100-pl portions of each dilution into sterile,
1.5-ml Eppendorf microcentrifuge tubes, Place the open
tubes in a sterile petri dish, and incubate at 30C until dry. TABLE 1 Some dyes that have been used in dye tolerance
Immediately add 100 p l of sterile distilled water to some of tests for characterizing bacteria
the tubes, then agitate vigorously to suspend the cells, and
plate onto an appropriate agar medium to determine the Dye % Concn (wtlvol) Reference
initial colony count. After various storage periods, repeat
the hydration and plating procedure with the remaining Alizarin red 0.032 267
tubes to estimate the percentage of surviving cells. Azure I1 0.0025 266
Basic fuchsin 0.005 266
15.2.24. Dye Tolerance
0.002 52
The ability to grow in the presence of low concentrations of
Brilliant green 0.001 62
various dyes added to culture media has been used to char-
acterize and differentiate bacteria, especially those that Crystal violet 0.0005 266
have few other readily determinable phenotypic character- 0.0001 62
istics. For example, Wolinella cuwa can be differentiated Indulin scarlet 0.05 266
from Wolinella recta by its ability to grow in the presence of Janus green 0.01 266
Janus green, basic fuchsin, methyl orange, indulin scarlet,
Malachite green 0.001 62
and safranin (266, 267).
Some dyes that have been used in dye tolerance tests for Methyl orange 0.032 266
characterizing bacteria are listed in Table 1. In some in- Methylene blue 0.1 184
stances, dye tolerance has been determined by soaking 0.3 185
paper strips in a dye solution, placing the strips on inocu- Safranin 0.05 266
lated agar plates, and noting whether growth occurs around
Thioninc 0.002 52
the strips after incubation. For instance, taxonomic studies
Next Page

344 GROWTH

at 362 to 365 nm. Commercially available substrates in- at 24,48, and 72 h under UV light (below 260 nm). A short-
clude 4-methylumbelliferyl (4-MU) acetate, 4-MU propi- wave lamp used for the examination of mineral specimens is
onate, 4-MU butyrate, 4-MU heptanoate, 4-MU nonanoate, suitable. Plates should not be reincubated after examination,
4-MU oleate, 4-MU elaidate, and 4-MU palmitate. because the UV illumination may be bactericidal. A positive
test is indicated by a fluorescent zone in the agar surrounding
15.2.26.1. Method 1 (282) the growth. Fluorescent pseudomonads produce a yellow-
A rapid spot test for the detection of butyrate esterase can green pigment; certain Azotobucter, Azomonas, and Beijer-
be done as follows. Dissolve 100 mg of 4-MU butyrate inckia species may form green or white fluorescent pigments.
(Sigma) in 10 ml of dimethyl sulfoxide (DMSO) and 100 pl Examples: positive, Pseudomonas fluorescens; negative,
of Triton X-100 detergent (Sigma). Dilute this stock solu- Escherichia coli.
tion 1:lO in citrate buffer (0.1 M citric acid, 0.05 M Methanogenic members of the Archaea also fluoresce
Na2HP04 . 12H20 [pH 5.011). This diluted reagent can be under UV light. The fluorescence is strong enough in ac-
stored in 250-yl portions at -70C for at least 1 month. To tively growing cells to allow individual cells to be seen with
test a culture, place a few drops of the reagent onto a piece an epifluorescence microscope. The cofactor F420 is one of
of filter paper. Streak two or three colonies from a culture of the major components responsible for this autofluores-
the test organism onto the paper. After 30 s, expose the cence. Freshly prepared wet mounts are best examined im-
paper to a UV source (362 to 365 mn, or the long wave- mediately under a suitable epifluorescence microscope using
length of a Woods lamp). A positive test is indicated by a 40X or lOOX objective. Cells will fluoresce bright green
light blue fluorescence. A negative test has no fluorescence. when excited and viewed with an appropriate filter.
Examples: positive, Moraxella catarrhalis (Branhamella Members of the methanogenic genus Methanothrix (Meth-
catarrhalis), Staphylococcus aureus; negative, Neisseria sicca, anosueta) do not produce strong fluorescence. In natural
Neisseria lactamica. samples, algae may also fluoresce red while some natural
minerals may also show up as bright fluorescent objects.
15.2.26.2. Method 2 (89)
The following quantitative method for esterases has been 15.2.29. Formate-Fumarate Requirement
applied to mycobacteria but can be adapted for use with Use the formate-fumarate requirement test to distinguish
other bacteria. Prepare 0.04 M stock solutions of 4-MU ac- certain bacteria that, when grown anaerobically, oxidize
etate, 4-MU propionate, 4-MU butyrate, and 4-MU hep- formate and concomitantly reduce fumarate to succinate.
tanoate in DMSO. Prepare 0.02 M stock solutions of 4-MU Prepare formate-fumarate stock solution (section 15.5.6),
oleate, 4-MU elaidate, and 4-MU palmitate in DMSO. and add 1 drop of the solution per ml of prereduced PY
Store at -20C until needed. Immediately before use, add broth (section 15.4.31) when inoculating the medium
0.2 to 9.8 ml of 0.2 M phosphate buffer. The pH of the buffer under oxygen-free carbon dioxide. Compare the growth re-
depends on the esterase and the species. For mycobacterial sponse with that occurring in medium lacking the formate-
esterases, the optimum pH is 8.0 to 8.5; however, sponta- fumarate. If the growth is greatly stimulated by the formate-
neous hydrolysis of the substrates is reported to occur at fumarate, the test is positive. In some cases, growth may not
these pHs. Therefore, buffer with a pH of 7.3 has been used occur at all unless the supplement is added.
instead (89). Reaction mixtures contain 0.1 ml of a suspen- Examples: positive, Bacteroides ureolyticus (this organism
sion of the bacteria to be tested, 0.9 ml of phosphate buffer, is related to members of the genus Campylobacter [280]);
and 1.Oml of the buffered 4-MU ester. (Also prepare a blank negative, Prevotella oralis.
containing 0.1 ml of distilled water instead of bacteria.)
Incubate the mixtures at 37C for the desired period, and 15.2.30. Gas Production from Sugars
then stop the reaction by adding 1.0 ml of glycine buffer (0.2
M solution of glycine, adjusted to pH 10.5 with 1.0 N 15.2.30.1. Method 1
NaOH). Measure the fluorescence in quartz cuvettes at Before autoclaving the sugar broth, place a small vial (ca.
37C with a spectrofluorimeter using an excitation wave- 10 by 75 mm; also known as a Durham tube) in an inverted
length of 362 nm and an emission wavelength of 450 nm. position in the tube. After autoclaving, the vial will become
completely filled with medium. For thermolabile sugars, add
15.2.27. Flagella filter-sterilized stock solutions aseptically after the base
Use flagellum staining with light microscopy (see chapter medium has been autoclaved and cooled; allow the tubes to
2.3.10) or electron microscopy (see chapter 4.2.2 and stand for a day or so to allow diffusion of the sugar into the
4.2.5.1). Agitate the bacteria as little as possible during their inverted vials. Inoculate the medium and incubate. A posi-
preparation, because flagella may be easily broken off the tive test is indicated by the accumulation of gas within the
cells. Flagellar stains with light microscopy are occasionally vial. Note: if media are stored in a refrigerator before use and
misleading when applied to polar-flagellated bacteria, be- then are inoculated and incubated, dissolved gases in the
cause a fascicle of several flagella sometimes seems to be a sin- media may be liberated and accumulate in the vials, giving
gle flagellum (298). Culture conditions may be important; false-positive reactions. Sterile controls should be used to
e.g., some bacteria, such as Vibrio parahaemolyticw, form a sin- detect this occurrence. Also, if the organism forms only
gle polar flagellum when grown in liquid media but also form small amounts of carbon dioxide, detection may be difficult
lateral flagella of shorter wavelengths when grown on solid because of the great solubility of carbon dioxide and be-
media (235). Some bacteria, such as Listeria monocytogenes, cause of rapid diffusion into the air. Methods 2 and 3 are de-
may not form flagella at certain temperatures but do form signed to eliminate this difficulty.
them at lower temperatures (128). In some instances, glucose-
containing media inhibit the formation of flagella (21). 15.2.30.2. Method 2
Method 2 is the same as method 1 but with the following
15.2.28. Fluorescent Pigment change. After inoculation, add ca. 1 ml of sterile mineral
Make a single streak across a plate of Kings medium B (see (paraffin) oil to prevent the diffusion of carbon dioxide into
section 15.4.7). Examine the plates with the covers removed the air. Alternatively, use sugar-containing agar medium
16
General Methods To Investigate Microbial Symbioses
TODD A . CICHE AND SHANA K . GOFFREDI

16.1. INTRODUCTION ......................................... 394


16.1.1. Symbiosis .......................................... 394
.
2. Kochs Postulates Applied to Symbiosis ..................... 395
.
3. Diversity and Significance of Symbiosis ..................... 397
16.1.3.1. Bacteria-Phage ................................ 397
.
2. Microbe-Microbe .............................. 397
.
3. Bacteria-Protists .............................. 397
.
4. Rhizobium-Legume Symbiosis ..................... 397
.5. BuchneraeAphids .............................. 398
.6. Termites .................................... 398
.7. Riftia pachyptila .............................. 398
.8. Epibiotic Associations .......................... 398
16.1.4. Methodological Dissection of Symbiosis ..................... 398
16.2. TECHNIQUES USED TO STUDY ENVIRONMENTAL
SYMBIOSES ............................................. 398
16.2.1. Environmental Symbioses ............................... 398
16.2.1.1. The Phallodrine Oligochaete-Microbial Symbiosis ...... 399
16.2.2. Microscopy ......................................... 399
.
3. Molecular Dissection of Symbiosis ........................ 400
16.2.3.1. PCR Fingerprinting ............................ 400
.
2. FISH ...................................... 401
16.2.4. Physiological Dissection of Symbiosis...................... 402
16.2.4.1. Direct Measures of Enzyme Activity ................ 402
.
2. Measures of Bacterial-Specific Biomarkers and Stable
Isotopes .................................... 403
.3. Physiological Studies ........................... 404
.4. Tissue Autoradiography ......................... 405
16.3. METHODS TO STUDY SYMBIOSIS I N MODEL SYSTEMS ......... 406
16.3.1. Model Systems ....................................... 406
16.3.1.1. Monoxenic or Binary Symbioses ................... 406
.2. Polyxenic or Consortia1 Symbioses ................. 407
16.3.2. Colonization Assays ................................... 407
.3. Molecular Genetic Analysis of Symbiotic Bacteria ............. 408
16.3.3.1. Use of Reporters for Symbiont Detection ............ 408
.2. Labeling of Symbiotic Bacteria with GFP ............. 408
.3. Transposon Mutagenesis of Symbiotic Bacteria ......... 410
.4. Targeted Gene Disruption by Allelic Exchange ......... 411
.5. Strand-Overlap Extension (SOE) PCR and Allelic
Exchange for Targeted Gene Disruption ............. 411
16.3.4. Genetic. Cellular. and Molecular Analysis of Symbiosis
in the Host ......................................... 413
......................... 413
16.3.4.1. Cell Biological Analysis
.
2. Molecular and Proteomic Analysis................. 413
16.4. REFERENCES ........................................ 414

16.1. INTRODUCTION fungal mycorrhizae and rhizobia for the procurement of


phosphate and reduced nitrogen. respectively. Symbiosis.
.
16.1 1. Symbiosis in sum. is a powerful evolutionary mechanism that has re-
Symbiosis. defined by de Bary as the nontransient inter- sulted in novel characteristics and allowed for the ex-
action between dissimilar organisms (38).has dramatically ploitation of diverse environmental niches.
affected life on Earth. For example. eukaryotic cells in our All macroorganisms and microorganisms are surrounded
own bodies have resulted from symbiotic mergers between by a diverse microbiota. with which they have evolved to
microbes millions of years ago (111). Entire biomes. in- interact for part or all of their lives. Organisms tend to re-
cluding forests and grasslands. rely upon symbiosis for sta- sist pathogenic or parasitic interactions (i.e., immunity)
bility and efficiency;for example. roots require infection by while fostering beneficial interactions . I t has become

394
16. Methods To Investigate Microbial Symbioses w 395

increasingly apparent that components of the innate im- a gradient of interactions ranging from parasitism to com-
mune system are required for animals and plants to interact mensalism and mutualism whereby the host fitness is de-
both in mutualism and pathogenesis (125). How then does creased, unaffected, or increased, respectively, as a result of
the host differentiate between benign or beneficial micro- the interaction. It is also often assumed that the host is a eu-
organisms and those that are pathogenic? Understanding karyotic organism, but the definition from de Bary does not
the molecular mechanisms of the establishment and main- exclude bacterial-bacterial or bacterial-phage interactions as
tenance of symbiosis will shed light on the factors affecting being symbiotic (examples of these are shown in Table 1).
the stability of beneficial microbial interactions that are im- In this chapter, methods are presented that highlight
portant for most plant and animal life. two general areas of symbiosis research: (i) the detection
Unlike parasitic and pathogenic interactions that cause and characterization of environmental symbioses and (ii)
disease, symbiotic associations are usually operating incognito elucidation of the molecular and cellular mechanisms of
in healthy hosts and only become apparent upon disruption of symbiosis in model systems. We describe here methods to
the association and close examination by the researcher. demonstrate that an organism is symbiotic and examples of
Although different in effect on host fitness, mutualism and techniques used to elucidate the molecular, physiological,
pathogenesis share many characteristics that have recently cellular, and evolutionary basis of symbiosis. Research in
been revealed by genomics, genetics, and molecular biology these areas is in its infancy and should provide many more
(78). We use the general definition of symbiosis by de Bary, interesting discoveries in the future, taking advantage of
acknowledging similarities between pathogenic and mutual some of the techniques described here. These studies should
host-bacterial interactions, as long as they are nontransient, result in an increasing knowledge about the role of symbi-
which result in a great range of fitness to the host. We also use otic microbes to host biology and ecology and should indi-
similarities between pathogenesis and symbiosis to develop a cate that symbiotic associations involve sophisticated
suite of criteria, described below, that one can use to infer sym- mechanisms for host-bacterial interactions, somewhat com-
biotic relationships involving bacteria. parable to those of host-pathogen interactions.
A common conception of symbiosis is of two organisms
cooperating for their common benefit. However, the fitness 16.1.2. Kochs Postulates Applied to Symbiosis
of each organism resulting from symbiosis is difficult to If we want to identify a potential symbiosis involving two or
quantify and is usually dynamic, influenced by environmen- more organisms, we must first demonstrate a nontransient
tal and genetic variables. Thus, de Bary defined symbiosis as interaction, distinguish the residents (i.e., the symbionts)

TABLE 1 Examples of microbial symbioses


Symbiosis Type Reference(s)
Bacteria-phage
Vibrio cholerae-CTXQ phage Broadened host range 169
Shigella flemri-Sf(1-V) phage Broadened host range 1
Microbe-microbe
Chlorochromatium Phototrophic consortia 60,70
Archaea-bacteria
Anaerobic methane-oxidizing-sulfate-reducing consortium Syntrophy 13, 129; Color Plate 9A
Archaea-archaea
Nanoarchaeum equitans-Ignicoccus Parasitism 173
Protozoa-bacteria
S tuurojoenina-Ves tibaculum Unknown 158
Amoeba poteus D strain-X symbiont S-adenosylmethionine dependence 87
Plant-bacteria
Legume-rhizobia Diazotrophy (N, fixation) 166; Color Plate 9F
Plant-growth-promotingbacteria Nutrition, defense 142
Animal-bacteria
Aphid-Buchnera Nutrition, essential amino acids 9,42
Euprymna scolopes-Vibrio fischeri Bioluminescence 125; Color Plate 9E
Riftia pachyptila-sulfur-oxidizingautotrophic bacteria Nutrition-carbonenergy 26; Color Plate 9 C
Nematodes/polychaetes/shrimp-sulfide-oxidizing Nutrition-epibionts 76, 140, 141; Color
8-Proteobacteria Plate 9B
Heterorhabditis bacteriophora-Photorhabdus luminescens Insect pathogenicity-nutrition 33, 54; Color Plate 9G
Animal-polymicrobial
Termite-bacteria, archea, protozoa, fungi Cellulose degradation-nitrogen fixation 18; Color Plate 9D
Phallodriline worms-a-, 7 - ,6-Proteobacteria Nutrition, unknown 12,45,49, 94; Color Plates
9H, 10
Human gastrointestinal tract-bacteria, archaea Nutrition, other unknown 80
396 m GROWTH

from the tourists (i.e., free-living contaminants), and as- isolate and cultivate symbiotic bacteria and the inability to
sess the effect of the symbiosis on the fitness of the host. selectively eliminate the symbiont or generate an aposym-
Since the broad definition of symbiosis includes parasitism biotic host. These challenges are similar to those encoun-
(i.e., pathogenesis), criteria for determining the causative tered when investigating pathogenesis caused by uncultured
agent of disease (Kochs postulates) can be applied to sym- bacteria, viruses, or chronic infections. In 1996, Relman
biosis where the characteristics of the symbiosis (e.g., colo- adapted Kochs postulates for criteria to determine the
nization, tissue structure, and fitness) can be substituted for causative agent of disease by use of sequence-based detec-
the symptoms of disease. tion methods for pathogens (58). Similar sequence-based
The contributions of many scientists, including Koch criteria can be adapted to symbiosis to provide evidence
and Pasteur in the late 19th century, on the ability of bac- that an organism is symbiotic (listed below). These criteria
teria to cause disease are useful for our understanding of are generally listed from strongest (i.e., by definition, a non-
host-bacterial interactions in general. Robert Koch pre- transient association between symbiont nucleic acid and
sented four postulates that, when fulfilled, establish causal host is required for symbiosis) to weakest (the specific na-
evidence for the microbe causing a given disease, although ture of the association may be unknown). Even though
failure to meet these criteria does not prove otherwise. many symbioses only partially meet these criteria, they are
Kochs postulates include the following. presented here to aid the researcher when examining sym-
biosis from a molecular perspective.
1. The pathogen must be present in each diseased host.
2. The causative pathogen must be isolated and grown in Biochemical and Molecular Criteria for Symbiosis
pure culture.
3. The pure culture must cause disease in the new host. 1. A nucleic acid sequence (or ribotype) belonging to
4. The causative pathogen must be reisolated from the the putative symbiont should be present in most cases of
new host in pure culture and identified. symbiosis and found preferentially in organs, tissues, or cells
known to be involved in symbiosis (based on other
Symbiotic microorganisms do not fit easily into Kochs
anatomic, histologic, or chemical evidence of symbiosis).
postulates, because the resulting change in observable fit.
2. Fewer or no copy numbers of symbiont-associated nu-
ness of the host is usually less obvious than acute disease
cleic acid sequences should occur in aposymbiotic hosts or
caused by a pathogenic bacterium. Because symbiosis en-
tissues not involved in housing symbionts.
compasses a wide range of interactions, the effect of the as-
3. Efforts should be made to demonstrate the microbial
sociation on host fitness will vary from unobservable (come
ribotype in situ (via fluorescence in situ hybridization
mensalisms) to obvious or even essential to host and/or
[FISH] microscopy on tissues or cellular level; see section
symbiont fitness (mutualism). However, symbiosis is often
16.2.3.1 below).
accompanied by novel characteristics not observed in apo-
(or non-) symbiotic organisms. Even for commensal rela-
4. These sequence-based forms of evidence for microbial
causation should be reproducible.
tionships, a nontransient presence (i.e., colonization) of
symbionts on or in host tissues can be reliably observed.
5. After elimination of symbionts (i.e., by antibiotic
treatment), the symbiont-associated ribotype should de-
These characteristics can be substituted for disease to adapt
crease or become nondetectable, manifested in loss of sym-
Kochs postulates to symbiosis as given below.
biont-derived characteristics.
Kochs postulates have been adopted for symbiosis, and
6. When nucleic acid sequence detection predates sym-
these include the following.
biosis (e.g., during ontogenetic development), or sequence
1. Identify symbiont from the host. copy number correlates with symbiosis (e.g., health of the
2. Eliminate the symbiont from the host. host for mutualism), the sequence-symbiosis association is
3. Observe difference(s) between symbiotic and aposym- more likely to be a causal relationship. The term onto-
biotic host. genetic refers to the origin and development of an individ-
4. Add the symbiont to a new aposymbiotic host and ob- ual organism from embryo to adult.
serve symbiotic state. 7. The nature of the microorganism inferred from the
5. Identify the symbiont from the new host. available sequence should be consistent with the known
characteristics for that group of organisms. When pheno-
An example of the fulfillment of these modified postu- type (e.g., microbial morphology and physiology) is pre-
lates is the symbiosis between the Hawaiian bobtail squid,
dicted by sequence-based phylogenetic relationships, the
Eup-ymna scolopes, and the bioluminescent bacterium, Vibrio
meaningfulness of the sequence is enhanced but should be
fischeri, which resides in the squids light organ (Color Plate
supported by other evidence (e.g., molecular or physiologi-
9, described in section 16.3): symbiotic bacteria are readily
cal; see section 16.2).
isolated and identified from the light organs of symbiotic
hosts (postulate 1); hatchling squid are aposymbiotic in the These criteria have been met for many of the symbioses
absence of symbiotic bacteria, and symbiotic bacteria can be discussed in section 16.2, including the chemoautotrophic
eliminated from adult squid by antibiotic treatment (postu- symbiosis involving the giant tube worm Riftiu pachyptila,
late 2); aposymbiotic hosts are not bioluminescent (pos- which inhabits hydrothermal vents. For example, the sym-
tulate 3); addition of symbiotic bacteria to aposymbiotic biont nucleic acid (or ribotype) and bacterial-specific en-
hosts reestablishes the symbiotic function (i.e., biolumines- zymes are always found in symbiotic tube worms (criterion
cence of the light organ) (postulate 4); and symbiotic V. fischeri l ) , the nonsymbiotic tissues of R . pachyptila are devoid of
can be reisolated from symbiotic hosts (postulate 5). These symbionts (criterion 2), the direct identification and local-
postulates have been met for the majority of symbioses de- ization of the symbiont has been observed via fluorescence
scribed in the introduction to model symbioses (section in situ microscopy of nucleic acids and immunohistochem-
16.3.1 below). istry of intracellular bacteria-specific enzymes (criterion 3),
Many symbioses, however, present additional challenges these results are reproducible from worm to worm (criterion
to fulfilling Kochs postulates. These include the inability to 4), symbiont nucleic acid is absent from R. pachyptila larvae
16. Methods To Investigate Microbial Symbioses 397

(criterion 6), and sulfide oxidation and COZ fixation are surrounded by a shell of Desulfosarcina cells. The Archaea
metabolic properties consistent with certain members of the are thought to mediate methane oxidation, while the bac-
y-Proteobacteria, with which the symbiont is phylogeneti- terial partner is presumed to consume some reduced inter-
cally related (criterion 7). mediate for the reduction of sulfate, thereby creating a fa-
vorable energy gradient for the conversion of methane to
16.1.3. Diversity and Significance of Symbiosis carbon dioxide. Similarly, a likely syntrophy involves the
Symbiosis is prevalent in almost all types of organisms, from phototrophic consortium Chlorochromutium aggregatum,
bacteria to protists, plants, and animals (Color Plate 9). originally thought to be a single organism. This consortium
In fact, among plants and animals, symbioses with micro- can be very abundant in freshwater lakes and is thought to
organisms are the norm, rather than the exception. be important in sulfur and carbon cycles. This consortium
Historically, the important and often essential role of sym- was recently isolated for the first time (59) and found to
biotic organisms to the lives of their hosts has been over- consist of a single motile nonpigmented central p-
looked. For example, biologists studied for decades the sym- proteobacterium surrounded by green sulfur photosynthetic
biosis between leaf-cutting Attine ants and their fungal epibionts (60). It is hypothesized that the consortium uti-
symbionts without realizing that a conspicuous patch of lizes a closed sulfur cycle in which the epibionts oxidize sul-
waxy bloom on the ants thorax was an actinomycete sym- fide during anaerobic photolithotrophy and the central
biont (36). The symbiotic actinomycete produces anti- symbionts reduce sulfur while oxidizing carbon compounds
biotics specific to the parasitic fungus Escouopsis, which can from the host. The consortium may also interact during
infect the symbiotic fungal garden, and is preferentially phototaxis (60) and coordinate cell division. A draft
found in ant castes that groom the primary fungal symbiont. genome sequence of this consortium is available (htpp://
Thus, even symbiosis researchers can overlook symbiosis. genome.jgi-psf.org/finished_microbes/chlag/chlag.home
There is an enormous diversity in the number of organ- .html), further adding to the utility of this model phototro-
isms that function as hosts, as well as the types of microbes phic consortium for the study of bacteria-bacteria symbiosis.
that become symbionts. Examples of the range of bacterial-
host interactions are listed in Table 1 (see also Color Plate 16.1.3.3. Bacteria-Protists
9). We mention only a few examples here to illustrate the Many diverse protists, including at least three types of flagel-
breadth and diversity of symbioses, focusing on bacterial lates that occur as symbionts within the termite gut, are asso-
symbioses despite the abundance of many other host- ciated with symbiotic bacteria. One recent example of this is
eukaryotic associations (i.e., algae, protists, and fungi). a cellulose-degradinghypermastigote, Staurojoenina sp., found
within the gut of the termite Neotermes cubanus (158).
16.1.3.1. Bacteria-Phage Electron microscopy revealed a dense coat of epibiotic rod-
Viruses and phages, which by de Barys definition can be shaped bacteria (up to 3,500 per flagellate) that interact with
considered symbiotic, affect their hosts in a variety of ways, the cuticle via special attachment sites on the cell envelope
including acute virulence, nonvirulence, or even benefit. of the host. The nature and function of this relationship is
Microbial genomes are littered with genes that confer added unknown but is thought to be different from the motility-
capabilities to the host organism and in many cases have conferring epibionts of other gut flagellates (e.g., Treponem
been shown to be transmitted by phages. This is most evi- [spirochetes] found on Mixocricha sp.) (34, 162, 174).
dent in pathogens where toxins and/or virulence factors are
encoded on a lysogenic phage (19), i.e., the cholera toxin 16.1.3.4. Rhizobium-Legume Symbiosis
genes, ctxAB, are carried and transmitted by CTXphi, The symbioses between plants in the family Leguminosae
which itself utilizes the colonization factor toxin coregu- and the a-Proteobucteria Rhizobium, Mesorhizobium, Sinur-
lated pilus (TCP), encoded on another phage-like element, hizobium, Brudyrhizobium, and Azorhizobium, collectively
for its infection (50). Another type of bacteria-phage sym- called rhizobia, are among the best understood symbioses
biosis is the gene transfer agents found in several diverse (Color Plate 9F). Only a general introduction to the detailed
prokaryotes, including Rhodobucter cupsulatus (99) and knowledge of this complex symbiosis is given here. The rhi-
Bruchyspiru hyodysenteriae (1 14). These phages appear to be zobium-legume symbiosis is one in which the symbiont fixes
obligate symbionts of the host cell and function mainly as dinitrogen in return for photosynthate and a protected niche
gene transfer agents by indiscriminately packaging host (root nodule) from the host. The establishment of this sym-
DNA. The study of bacteria-phage symbioses is important biosis is well understood and involves a complex develop-
for our understanding of bacterial evolution and emergence mental program of binding, signaling, and growth between
of disease as well as providing powerful experimental mod- symbiont and plant (reviewed in reference 62). The rhizobia
els for the study of symbiosis in a test tube. likely bind to root hairs, at first weakly, in a Ca2+-dependent
manner partially through rhicadhesin. The rhizobia sense
16.1.3.2. Microbe-Microbe plant flavonoids and produce signaling molecules called nod
Bacteria-bacteria syntrophies are one type of symbiosis in factors or lipo-chito-oligosaccharides that trigger root hair
which energetically unfavorable reactions are made more curling and infection thread formation. A further dialogue
favorable by the close association of two or more micro- ensues when the rhizobia migrate through the infection
organisms. In marine cold seeps and other subseafloor diffu- thread, which in Sinorhizobium meliloti requires exopolysac-
sion-limited environments, for example, there are cell ag- charide production (reviewed in reference 57) as bacteria
gregations of Archuea belonging to the Methanosarcinales, grow from a few colonizing cells in the infection thread (61).
surrounded by, and often in direct physical contact with, Several lines of evidence suggest that the plants control the
sulfate-reducing bacteria related to the Desulfosarcina genus infection by producing reactive oxygen and nitrogen species,
(13). These organisms work together to achieve the ther- phenolics, etc., factors similar to a hypersensitive response
modynamically unfavorable anaerobic oxidation of (reviewed in reference 7). After passing through the gaunt-
methane. In this association, the Archaea are commonly lo- let, the rhizobia then differentiate into bacterioids, capable of
cated in the central core of each microbial aggregate and are nitrogen fixation, while the plant develops a mature nodule.
398 GROWTH

The legume maintains a suitable environment for nitrogen anoxic basins. R. puchyptila houses symbiotic y-proteobacte-
fixation by producing leghemoglobin to protect the sym- ria in a unique internal organ known as the trophosome,
biont nitrogenase that is irreversibly inactivated by oxygen which can account for up to 50% of the host volume and is
and by providing photosynthate to fuel the high energy de- highly vascularized (23, 97). First observed via transmission
mands of symbiont nitrogen fixation. electron microscopy (23, 26), these intracellular bacteria
were discovered to be sulfide-oxidizing and carbon dioxide-
16.1.3.5. Buchnera-Aphids fixing symbionts solely responsible for the nutrition of the
The aphid symbiosis with Buchnera aphidicola is one of the worm (51). Many important studies have been conducted
most well-understood bacterial-animal endosymbioses. on the metabolic needs of both partners as well as the nu-
The symbionts provide the insect host (e.g., Schizaphis tritional dialogue between them (see section 16.2.4). For
graminurn) with essential amino acids which are lacking example, it is now known that the host has evolved many
in the plant phloem on which it feeds. Aphids are not able biochemical adaptations for symbiont accommodation, in-
to meet their nutritional requirements without Buchnera, cluding effective mechanisms to concentrate inorganic car-
and Buchnera has so far not been found outside the aphid bon internally, sulfide acquisition from the environment
hosts. The capacity for synthesizing certain essential using specialized hemoglobin molecules, as well as unprece-
amino acids, such as tryptophan and leucine, has been rel- dented pH and ion regulation. The symbiont, in turn, pro-
egated to symbiont plasmids, thus allowing for indepen- vides the host with organic carbon, which enables the worm
dent regulation of these genes in relation to the rest of the to grow rapidly, quickly dominating communities around
genome. Buchnera organisms inhabit vesicles inside spe- newly established hydrothermal vents.
cialized aphid cells called bacteriocytes and are vertically
transmitted by infection of aphid gametes during ovula- 16.1.3.8. Epibiotic Associations
tion (42). The strict vertical transmission of symbionts is There are many examples of commensal or mutual associa-
manifested in the coevolution of Buchnera symbiont and tions between host organisms and symbionts on the external
aphid host (113), which has significant consequences to surface of the host (epibiont). For instance, a diverse num-
both partners, including dramatic genome reduction of ber of marine hosts, including the Stilbonematinae (nema-
the symbiont (123). todes [Color Plate 9B]), the Alvinellidae (polychaete worms),
and the Bresiliidae (shrimp) have dense growth of epibionts
16.1.3.6. Termites often consisting of highly ordered sulfide-oxidizing, -pro-
Termites are now regarded as one of the classic examples of teobacteria (39, 132, 165). In all cases, the hosts physically
an obligate mutualistic symbiosis (Color Plate 9D).Early ob- bridge the gap between oxic and anoxic waters via movement,
servations suggested that nearly all of the termite species thereby exposing the symbionts to all necessary metabolites.
lived almost exclusively on cellulose-rich materials, a difficult The hosts are thought to graze on the symbionts as a source of
task for most animals. The secret to their success is the pres. nutrition. Epibiotic associations are also abundant on nonma-
ence of a diverse microbiota located in the hindgut region of rine organisms and can affect the host in a variety of ways, for
the alimentary tract, the major site of nutrient absorption by example in disease suppression in plants (142).
the host (85). Over the past century, much has been learned
about the obligate dependence of termites on protozoans and 16.1.4. Methodological Dissection of Symbiosis
bacteria for the supply of carbon compounds resulting from Within the last decade, two main areas of symbiosis re-
cellulose digestion and on bacteria for nitrogen fixation and search have emerged, mostly as a result of the development
recycling. The tremendous diversity of termite symbionts has of molecular biological tools: (i) discovering and elucidat-
been elucidated by molecular surveys as well as cultivation ing the important role of symbiotic microorganisms to life
studies. In fact, this is one instance where cultivation has and (ii) dissecting the dialogue required to establish and
been reasonably successful (see section 16.2.4.2), even maintain a symbiosis. Because there are increasing numbers
though many of the potentially hundreds of symbiont species of described symbioses, we have adapted Kochs postulates
within the gut have not been isolated in the lab. Bacteria and molecular Kochs postulates as criteria in which to
associated with termites include methanogens, acetogens, judge the degree of evidence for a particular symbiosis and
fermenters, sulfate reducers, and nitrogen fixers (95, 127). A a causal relationship of the symbiont to one or many host
portion of the dinitrogenase reductase gene (nijH) was di- attributes. In section 16.2, we describe some examples of
rectly amplified from DNA extracted from the mixed micro- the techniques used to identify and characterize novel or
bial population in the termite gut (128). Sequences from the poorly understood symbioses from the environment, from
termites included four groups of microbial nijH sequences, insects to deep-sea worms. Most of the techniques described
some distinct from those previously recognized in studies are independent of the ability to cultivate the symbiont or
using classical microbiological techniques, indicating the host. We present a case study involving the dual symbionts
presence of diverse nitrogen-fixing microbial assemblages in of phallodrine oligochaetes, for which much has been
the guts of termites. Some of the termite gut microbes are as- learned despite the inability to specifically manipulate ei-
sociated directly with the termite, while others are symbionts ther partner. In section 16.3, we describe established and
on protists within termites, and some are involved in specific emerging model systems and genetic and molecular tools
bacterial-bacterial interactions. Many of the termite sym- used to elucidate symbiotic factors in both the symbiont
bionts are endemic to termite guts. and host at the cellular, molecular, and evolutionary level.
16.1.3.7. Riftia pachyptila
R. pachyptila, the gutless giant hydrothermal vent tube 16.2. TECHNIQUES USED TO STUDY
worm, was the first demonstration of a chemoautotrophic ENVIRONMENTAL SYMBIOSES
bacteria-marine invertebrate association (Color Plate 9C).
Since its discovery in 1977, a number of other, similar asso- 16.2.1. Environmental Symbioses
ciations have been discovered in a variety of habitats in- The study of presently uncultured symbioses, for which ei-
cluding vents, cold seeps, sewage outfalls, eelgrass beds, and ther cultivation of the symbiont or live maintenance of the
16. Methods To Investigate Microbial Symbioses w 399

host is intractable or has not been attempted, can be ac- (made fresh) or 4% formalin (both in l x phosphate-
complished using many recent advances in molecular biol- buffered saline [PBS]; 1X PBS = 130 mM NaC1, 10 mM
ogy and biochemistry, as well as more traditional method- Na2HP04, pH 7.2, 0.2-pm-pore-size filter sterilized), but
ologies. We now know a great deal about hundreds of these specimens preserved in 70 to 100% ethanol may be suffi-
uncultured symbioses, most of which have been discovered cient and can even be postfixed in 4% formalin.
only within the past 40 years. The following section sum- 2. Preserved specimens for light microscopy should be
marizes many of the strategies that have helped to under- rinsed and dehydrated before embedding. The benefit of
stand some of these remarkable host-microbial systems and sectioning is the retention of tissue and cell morphology
will undoubtedly be useful for the continued investigation and a better understanding of symbiont localization within
of potential symbioses. the host.
3. Embedding in Steedmans polyester wax is described;
16.2.1 . I . The Phallodrine Oligochaete-Microbial however, a number of embedding resins are available.
Symbiosis Steedmans wax has a lower melting point than other resins
A fascinating host-microbe symbiotic system is that found in and, thus, reduces heat-induced artifacts such as tissue
many members of the annelid subfamily Phallodrilinae, ma- shrinkage. Melt 90 g of polyethylene glycol distearate at
rine relatives of earthworms (Color Plate 9H). First discov- 60C. Add 10 g of 1-hexadecanol (cetyl alcohol) and shake
ered and identified in 1979 (64), there are now over 100 until dissolved. All tissue incubation steps are performed at
species known worldwide today, all belonging to the genera 37C for 1 h; 3:l EtOH (100%):wax; then, 2:l Et0H:wax;
Inanidrilus and Olaoius (49). All known species of these gen- then, 1:l Et0H:wax. Incubate tissue three times in 100%
era lack digestive and excretory systems, and at least 20 wax and place in Peel-A-Way mold with fresh 100% wax.
species possess multiple symbionts belonging to a variety of Let harden at room temperature for 3 to 4 h. Store at
bacterial groups (63, 147). The worms are thought to de- -20C until sectioning.
pend entirely on bacterial symbionts for nutrition, as well as 4. Sections should be cut (6 to 8 pm thick with a micro-
potential recycling and elimination of nitrogenous com- tome) from wax-embedded specimens and mounted on glass
pounds. Many of the techniques described in this section slides. After wax removal via ethanol (three rinses in 100%
have been carried out with phallodriline worms, illustrating EtOH for 5 min each), sections can be stained with either
a successful attempt to characterize an uncultured symbiosis. hematoxylin-eosin (suitable for tissues fixed in formalin or
alcohol) or toluidine blue.
16.2.2. Microscopy 5a. Hematoxylin: dissolve ethylene glycol (25% final
concentration) in distilled water. Add hematoxylin (0.6%,
Microscopy is a traditional, and very reliable, method for wtlvol), then sodium iodate (0.06%, wtlvol). Add alu-
examining the ultrastructure of host tissues to look for evi- minum sulfate (8%, wtlvol) and glacial acetic acid. Let sit
dence of bacteria-like cells within (intracellular) or be- 1 h at room temperature. Eosin: add eosin Y (0.5%, wtlvol)
tween (extracellular) host cells or on external surfaces to 96% ethanol with several drops of glacial acetic acid.
(epibiotic). Light and electron (both transmission and Procedure: stain in hematoxylin for 5 to 15 min. Wash in
scanning) microscopy have played very significant roles in tap water for 10 min. Rinse with acid alcohol (1% HC1,
the initial characterization of many symbioses known to 70% EtOH) followed by tap water rinse. Soak twice in dis-
date. In some of the earliest microscopic observations of tilled water 2 min. Counterstain with eosin for 2 to 5 min.
symbiotic systems, parasites were discovered in the intes- Dehydrate using 96 and 100% ethanol.
tine of the termite by Leidy and Hungate in the late 1800s 5b. Toluidine blue: place a drop of toluidine blue on the
and early 1900s using light microscopy (85, 104). Buchner, section for 30 to 60 s. Drain and wash in distilled water for
in the 1960s, continued to pioneer the use of microscopy 5 s. Drain and dry thoroughly on a hot plate at 80C for
and documented a surprising array of microbial symbionts 2 min before using a coverslip with a permanent mounting
in insects (20). medium.
Light microscopy (detailed below) can reveal much 6. View with a compound microscope at x 100 magnifi-
about the morphology of cells and tissues. It is probably the cation to look for bacteria, including cells smaller (1 to 10
most commonly used research tool in microbiology, with pm) than eukaryotic cells (10 to 500 pm), sometimes en-
continuing advances in techniques and staining protocols closed within secondary vacuoles.
allowing for more precise observations. Embedding of sam-
ples and staining of eukaryotic tissue allows for better ori- Note: electron microscopy (not described here) allows for
entation within complex samples. Both are described below, magnification greater than x 1,000 and resolution greater
including toluidine blue and hematoxylin, which allow for than 0.2 pm, the approximate limits of light microscopy.
enhanced visualization of bacteria. Electron microscopy can be used to examine objects on a
very fine scale to learn more about tissue, cellular, and sub-
light Microscopy cellular morphology and even elemental composition. For
In almost all cases, depending on the level of integration scanning electron microscopy (4), whole samples are dried
between the two partners, symbionts are restricted to a spe- and sputter coated and the electrons that are produced as a
cific region or tissue within the host. As evidence for sym- beam hits the sample reveal information about the three-
biosis, one should generally look for (i) the presence of bac- dimensional external structure and surface features. For
terial cells, (ii) unusual or novel structures, (iii) lack of transmission electron microscopy (TEM), tissues are infil-
typical structures or organs, (iv) evidence for extreme vas- trated with embedding medium (plastics such as Spurrs
cularization, (v) precipitation of minerals or other sub- epoxy resin, Embed 812, or araldite resin) and sliced into
stances, or (vi) unusual color patterns within the tissues. ultrathin sections (50 to 100 nm). Electrons that pass
All of these features are common anatomical characteristics through the sample reveal information about the internal
of many hosts involved in mutual symbiotic arrangements. structure of the mecimen.
1. Specimens for routine histology should be preserved Microscopic investigation (including light microscopy,
(i.e., fixed) quickly, preferably in 2 to 4% paraformaldehyde TEM, and scanning electron microscopy) of phallodrine
400 GROWTH

oligochaetes provided evidence about the nature of these rectly, tissues can be preserved using any number of DNA-
associations, including the presence of multiple symbionts stabilizing methods. Tissue frozen at -20C is best, but
and the mode of transmission from one generation to the preservation in ethanol, RNALater (Ambion), or guani-
next. Bacteria-like cells were observed just below the cuti- dine-containing solutions like DNAzol (GibCo) may also be
cle between extensions of epidermal cells (Color Plate sufficient. Once DNA extraction is complete, DNA frag-
lOA), suggesting that up to 25% of the host volume was ments can be PCR amplified using bacteria-specific 16s
comprised of symbionts, for which there was significant dif- rRNA primers (usually 27F and 1492R [98]). Provided that
ference in appearance (including cytoplasm and cell mem- caution has been taken to avoid contamination during tissue
brane) and size between two distinct morphotypes (66,67). dissection, a positive bacterial amplification with general
Via microscopy, Giere and Langheld (66) discovered that PCR primers may be evidence of the presence of a potential
the symbionts could penetrate a developing egg while it was symbiont. Usually, however, subsequent PCR amplification
still adhered to the adult female. This external intrusion is undertaken using symbiont-specific primer sets (see, for
was observed via TEM on cultured eggs and developing em- example, the extracellular endosymbionts of Bugula sp. [bry-
bryos in various ontogenetic stages, suggesting that there is zoan] larvae [77, 106]), particularly in order to screen addi-
an initial infection by the symbiont. tional individuals for the presence of the symbiont (bio-
chemical and molecular criterion for symbiosis no. 4).
16.2.3. Molecular Dissection of Symbiosis Whether one (monoxenic) or many (polyxenic) sym-
bionts are present can be determined by digesting an aliquot
16.2.3.1. PCR Fingerprinting of the -1.5-kb PCR product with any number of restriction
The enormous diversity of bacterial symbionts has only re- endonucleases (e.g., RsaI, HaeIII) and analysis of the di-
cently been appreciated, in large part due to the introduc- gested products by agarose gel electrophoresis. If the restric-
tion of molecular techniques and the ability to construct tion fragment lengths add to multiples of 1.5 kb, this indi-
molecular phylogenies of microorganisms. Prior to this, lit- cates the likelihood of a polyxenic symbiosis. To analyze
tle was understood of the identity and function of bacterial polyxenic symbioses, the 1 6 s sequences are usually isolated
symbionts, since limited morphology did not allow for by denaturing gradient gel electrophoresis (DGGE) or by
meaningful interpretations of relatedness or function, and cloning. In DGGE, the two strands of a DNA molecule sep-
most, to this day, remain uncultured (although some ad- arate upon exposure to heat and/or a chemical denaturant.
vances in that area are described in section 16.2.4.3). When separated by electrophoresis through a gradient of
Nucleic acid techniques not only allow for the identifica- increasing chemical denaturant (usually formamide and
tion of microbes potentially associated with host organisms urea), the mobility of the molecule is affected by the num-
(fulfilling biochemical and molecular criterion for symbiosis ber of GC-rich regions, which require higher temperatures
no. 1) but also may reveal insight into their function. or denaturant concentrations to separate than AT-rich re-
There are many studies in which the phylogenetic iden- gions. During DGGE, an artificial G C clamp is added to
tity of symbionts within a host has been determined via 1 6 s one end of the molecule (i.e., PCR amplification using a
ribosomal sequences, long recognized as a suitable molecu- PCR primer with a 5 tail consisting of a sequence of 40 G C
lar marker for identifying organisms (44, 72, 77, 106, 127, bp); thus, the molecule becomes entangled in the gel as the
134). In some cases, including many monospecific (monox- branched structure separates into a semisingle stranded
enic) chemoautotrophic symbioses, direct bacteria-specific moiety. In this way, even DNA molecules which differ by
PCR amplification from symbiont-containing tissues of the only one nucleotide can be distinguished (12).
host results in only one unambiguous symbiont sequence.
For example, Distel and Felbeck (40) used direct sequenc- DNA Extraction: Dissection
ing of bacterial 16s rRNA to characterize the symbionts 1. Careful dissection is critical in order to reduce the
from the tissues of six sulfur-oxidizing polychaete and bi- risk of recovering bacterial contaminants by mistake.
valve hosts. All of the sulfur-oxidizingsymbionts examined Usually symbiont-containing tissues are separated from
clustered within two groups of the y-Proteobacteria, with symbiont-free tissues prior to analysis or preservation.
Thiomicrospira as the closest free-living genera. In other 2. Tissues should be rinsed with 90% ethanol or dilute
polyxenic cases, especially hosts with dual symbionts (for bleach (0.1% sodium hypochlorite) solution to remove sur-
example, many insects, oligochaetes, and deep-sea mussels), face contaminants, especially with tissues that are typically
microbial consortia (e.g., termites), or in systems with high in contact with the environment (e.g., bivalve gills).
potential for contamination by external bacteria (for exam-
ple, bivalve gills, sponges, or epibiotic associations), it is
DNA Extraction Using CTAB
necessary to construct bacterial clone libraries for adequate
(Modified from references 28 and 176.)
sampling of possible microbial variability (see surface steril-
ization considerations during the DNA extraction protocol 3. Add -300 mg of sample to 567 pl of TE buffer (10
below). In all of these cases, additional FISH assays are nec- mM Tris, 1 mM EDTA, pH 8.0)-gently homogenize or
essary to show definitively which recovered ribotype belongs vortex.
to an actual symbiont (see the section on FISH microscopy 4. Add 30 pl of 10% SDS (sodium dodecyl sulfate) and
below). 3 pl of 20 mg/ml pronase E, a mixture of endo- and exopro-
For DNA extraction, many standard kits (Qiagen teinases (final concentration of 100 pg/ml in 0.5% SDS).
DNAeasy, MoBio, Isoquick, etc.) or protocols (CTAB Mix thoroughly and incubate 1 to 4 h (or overnight when
[hexadecyltrimethyl ammonium bromide], GITC [guanidine required) at 56C.
thiocyanate], traditional phenol:chloroform, etc.) are avail- 5. Add 100 p1of 5 M NaCl and mix thoroughly.
able and work well, even with very small amounts of tissue. 6. Add 80 p1 of CTAB-NaC1 solution (0.7 M NaC1,
The CTAB protocol for DNA extraction is particularly ef- 10% CTAB; first dissolve NaCl in water, then slowly add
fective at removing exopolysaccharides from cells and is de- CTAB, while heating). Mix thoroughly and incubate 10
tailed below. If it is not possible to process fresh tissue di- min at 65C.
16. Methods To Investigate Microbial Symbioses w 401

7. Add an equal volume (0.7 to 0.8 ml) of chloroform/ tentially providing simultaneous symbiont identity and host
isoamyl alcohol, vortex. ultrastructure. The detection of specific rRNA sequences is
8. Centrifuge at 16,000 X g for 5 min. performed by first hybridizing a fluorescently labeled probe
9. Remove aqueous, viscous supernatant to a fresh with the target rRNA, in this case that of the putative sym-
microcentrifuge tube, leaving the interface behind. Add biont. Usually one starts with probes that target particular
an equal volume of phenol/chloroform/isoamyl alcohol, bacterial subdivisions (for examde, group-wecific Drobes
vortex. include Gam42a [5- gCCTTCCkACATCg?lT*3 I-[1101;
10. Centrifuge at 16,000 X g for 5 min. alf968 [5-ggTAAggTTCTgCgCgTT-3] [124], and
11. Transfer the supernatant to a fresh tube. Add 0.6 vol- Delta495a [5-AgTTAgCCggTgCTTCCT-3]
- __ - [ 1091 for the
ume isopropanol to precipitate the nucleic acids. Gently y-, a- and- 6-Prkobacteria, respectively). In addition to
vortex. confirming the presence of a particular ribotype in the host
12. Pellet the precipitate by brief centrifugation at (biochemical and molecular criterion for symbiosis no. 3),
16,000 X gfor 1 min. FISH can also provide specific ultrastructural integration
13. Wash the DNA with 70% ethanol to remove resid- between the bacteria and host (e.g., intra- versus extra-
ual CTAB. cellular, nonrandom distribution within tissues, etc.). For
14. Centrifuge at 16,000 X g for 5 min at room temper- example, FISH examination of Alvinella pompejunu, a hydro-
ature. Air dry pellet. thermal vent polychaete annelid, revealed the identity and
15. Suspend pellet in 100 p-1 of TE buffer (up to 1 h). localization of two filamentous bacterial phylotypes at-
*Alternatively, the supernatant from step no. 11 can be tached to the dorsal integument of the host (22). These E-
added to a CentriconB centrifugal filter unit (Millipore) proteobacteria are thought to play a role in detoxification of
and spun to a volume of 100 p l (in place of steps 12 to 15). high levels of poisonous sulfide and metals within the hosts
environment.
Once sequences are generated, the molecular identity of Similarly, Sipe et al. (154) investigated the mode of
a potential symbiont can then be assigned via comparison transmission of symbionts within two shipworm species,
of new sequences to those in existing databases (including Lyrodus pedicelatus and Bunkia setacea, using specifically de-
GenBank and RDPII). When examining symbionts, a prob- signed FISH probes, citing evidence for vertical transmis-
lem occasionally arises when appropriately similar se- sion based on the microscopic presence of the symbiont
quences (from close relatives) do not exist in public se- ribotype in eggs and ovarial tissue, as well as gill tissue (bio-
quence databases. At the very least, a bacterial subdivision chemical and molecular criteria for symbiosis no. 2 and 3).
(and likely family) can be inferred by immediate compari- There are many other examples, too numerous to include in
son to known databases. detail, so the reader is encouraged to explore recent exam-
One can also take a nucleic acid approach to infer meta- ples including Thiothrix epibionts on amphipods (68),
bolic pathways that may be utilized by a symbiont. The spe- Bacteroides epibionts on flagellate protists ( 158), Mollicutes
cific physiological capabilities of the symbiont, whether iso- gut symbionts in isopods (170), Acidovorux symbionts in the
lated or not, can be explored through methods such as nephridia of earthworms (15l ) , and extracellular symbionts
DNA amplification and Southern blotting for specific cod- of bryzoan larvae (106), to name a few. Two very good ref-
ing regions, RNA amplification (via reverse transcription- erences for general FISH protocols are Pernthaler et al.
PCR and Northern blotting), and simple PCR amplifica- (136, 138).
tion for genes that encode proteins with certain functions. The primary considerations for performing FISH assays,
For example, Millikan and colleagues cloned and character- besides those detailed in the FISH protocol below, include
ized a flagellin gene, one of the subunits that form the fla- probe design, probe labeling, and tissue preparation. Using
gellum in a variety of bacterial species, from the Riftia variable regions of the 1 6 s rRNA sequence, symbiont-
pachyptila symbiont (120). Degenerate primers were de- specific probes can be designed to selectively label a sym-
signed by aligning conserved sequences of known enteric biont even in complex microbial communities. Probe de-
fliC genes. The symbiont protein was expressed in non- velopment should be based on the following criteria: desired
motile Escherichia coli, and flagella were observed by elec- specificity for a particular target bacterium or group of bac-
tron microscopy (120), suggesting the retention of motility teria and adequate in situ accessibility. For maximum pene-
by the symbiont. Similarly, many genes involved in essen- tration into the highly structured ribosome during hy-
tial amino acid biosynthesis (e.g., tryptophan and leucine) bridization, it is best to target regions of the rRNA that
have been detected within the Buchneru symbiont genome have been empirically demonstrated to be accessible by
(8). In phallodrine oligochaetes, the gene that codes for dis- oligonucleotide probes (10). Probes can be labeled (com-
similatory sulfite reductase, an essential and key enzyme of mercially or in-house) with any number of fluorescent dyes.
sulfate reduction in free-living microbes, was amplified from Fluorescein is commonly used; however, cyanine dyes (Cy3
the symbiont-containing tissues of Olavius ulgarvensis (45). and Cy5) are brighter, pH-independent alternatives that
The dissimilatory sulfite reductase sequence of the 0. algur- avoid shorter-wavelength tissue autofluorescence. Probe
vensis symbiont was most closely related to that of the sul- modifications (e.g., labeling with heat-stabilized horse-
fate-reducing 6-proteobacterium Desulfosarcina, the same radish peroxidase) such as those for CARD (catalyzed re-
closest relative to the secondary symbiont observed during porter deposition)-FISH can also increase the sensitivity of
16s ribosomal surveys (45). the assay (137).
16.2.3.2. FISH Protocol for FISH
Many symbioses have been characterized by a combination 1. Preserve tissue for 3 h in 4% paraformaldehyde
of the above molecular methods followed by FISH mi- (made fresh in 1X PBS) at 4C. Rinse twice with 1X PBS.
croscopy. FISH microscopy was developed in the late 1980s Transfer to 1:l 1X PBS:100% ethanol, store at 4C.
for environmental bacterial applications, and for symbiosis 2. Homogenize and gently sonicate sample (in 1 X
research it complements other types of microscopy by po- PBS:ethanol). *Whole tissue can also be embedded in
402 GROWTH

Steedmans wax (see section 16.2.2, Light Microscopy, step vealed the presence of particulate methane monooxygenase
3). If so, proceed to step 4. transcript (pmoA), encoding a bacterial enzyme responsible
3. Spot -50 ~1 of homogenate onto slide (e.g., coated for conversion of methane to methanol, suggesting this ca-
SuperFrost Plus slides; Fisher Scientific). Let dry. pability in one of the two mussel symbionts (A. Pernthaler,
4. Take slide through ethanol series (50, 75, and 100% Max Planck Institute, personal communication [Color
EtOH; 5 min for each). Let dry (can store at -20C). Plate 111). The successful visualization of other expressed
5. Remove tissue autofluorescence with 10-min rinse in genes within symbiotic arrangements is a very real and ex-
0.1 M triethanolamine (pH 8.0) with 0.5% acetic anhy- citing possibility to link symbiont phylogeny and location
dride (vol/vol). Repeat three times. *Alternatively, a 30- within the host with metabolism and physiology.
min dark rinse in 50 mM sodium borohydride (made in Molecular characterization of the phallodrine oligo-
lOOmM Tris-HC1, pH 8.0) is also effective at removing tis- chaete Inanidrilus leukodematus symbiosis, using 16s rRNA
sue autofluorescence. (PCR) characterization and FISH microscopy, determined
6. Make hybridization buffer (0.9 M NaCl, 20 mM Tris- that the primary symbiont was a unique member of the
HCL, 0.01% SDS, 35% formamide). y-Proteobacteria and clustered with other known chemo-
7. Add 3 pl of Cy3-labeled (or other fluorescently autotrophic symbionts (94). FISH microscopy showed a flu-
labeled) oligonucleotide probe (50 pg/ml, specific to the orescent signal in a region between the cuticle and epider-
endosymbiont; see text for design tips) to 30 pl of hy- mis, confirming that the recovered 16s rRNA originated
bridization buffer. Add to sample on slide. from an endosymbiont.
8. Hybridize at 46C for 2 h to overnight in a chamber The possibility of multiple symbionts in these oligo-
to prevent drying out (such as a capped 50-ml Falcon tube). chaetes was explored again in 1999 when Dubilier and col-
9. Make wash buffer (e.g., 50 ml per slide; 70 mM leagues used TEM and 16s rRNA characterization, includ-
NaC1, 20 mM Tris-HC1, 0.01% SDS, 5 mM EDTA). Heat ing PCR fingerprinting, FISH microscopy, and DGGE to
to 48C before proceeding to step 10. compare the symbionts of Olavius loisae to known bacteria
10. Remove slide from hybridization chamber and place (12). PCR fingerprinting resulted in a heterogeneous pat-
in wash buffer for 15 min at 48C. tern, with two dominant families detected as a result of
11. Remove slide from wash buffer and rinse in distilled cloning efforts. Again one symbiont clustered with known
water. y-proteobacterial symbionts; however, the other symbiont
12. Stain with a dilute 46-diamidino-2-phenylindole clustered within the a-Proteobacteria. FISH probes designed
(DAPI) solution (5 Fg ml-) for 1 min. for each of the clone families confirmed the presence of two
13. Rinse in distilled water, let slide dry completely, add ribotypes within the worm host, the larger y-proteobacteria
antifade mounting compound (such as Citifluor [Ted Pella, and the generally smaller a-proteobacteria distributed
Inc.] or VectaShieldB [Vector Laboratories]), add coverslip, evenly between the cuticle and epidermis. DGGE patterns
and examine under epifluorescence microscopy. from individual worms showed the presence of both sym-
biont types, resulting in a fast method for screening other
The biggest challenge when using FISH for investigation oligochaetes for the presence of these symbionts as well as
of potential symbioses is background interference caused by the distribution of symbionts within different tissue regions
the inherent autofluorescence of many animal and plant tis- within one host. Phylogenetic screening (PCR fingerprint-
sues. Certain reagents, such as glutaraldehyde fixative, are ad- ing and FISH microscopy) of 0. algarvensis and 0. crassitu-
ditional sources of autofluorescence. Methods to reduce auto- nicatus (Color Plate lOB), on the other hand, revealed one
fluorescence, before hybridization, include acetylation or dominant clone group related to known sulfide-oxidizing
sodium borohydride treatment (described above in step 5). y-proteobacteria and the other, surprisingly, a 8-proteo-
Confocal laser scanning microscopy (and the alterna- bacterium most closely related to sulfate-reducing bacteria
tive, deconvolution microscopy) has also been established within the Desulfosarcina cluster (12, 45). Thus, it appears
as a valuable tool for the study of symbiosis. Confocal mi- that most phallodrine oligochaetes possess a common verti-
croscopy, in contrast to conventional epifluorescence cally transmitted primary symbiont (a y-proteobacterium),
microscopy, allows for the possibility to distinguish interior yet they form interactions with a diverse group of poten-
detail, by obtaining high-resolution images and three- tially horizontally transmitted secondary symbionts (includ-
dimensional reconstructions via serial optical sectioning. ing 6- and a-proteobacteria), with some reports of addi-
Confocal imaging and deconvolution can be applied to all tional symbionts as well, including spirochetes (N. Dubilier,
fluorescence microscopy applications and thus proves to be Max Planck Institute, personal communication).
an invaluable tool with which to study the structural intri-
cacies between host and symbiont. 16.2.4. Physiological Dissection of Symbiosis
Many other modifications to the FISH methodology
have great potential for application to symbiotic systems. 16.2.4.1. Direct Measures of Enzyme Activity
For example, a method known as mRNA-FISH allows for The presence and physiological capabilities of symbionts
the simultaneous detection of functional gene transcripts can be inferred from rRNA similarity as described above;
and traditional rRNA FISH cell identification (136). This however, it is not necessarily proof that they share similar
technique has been used extensively with eukaryotes; how- function with close relatives. Direct evidence for potential
ever, decreased mRNA stability and copy number and de- pathways utilized by a symbiont involves the measure of ac-
creased permeability of bacterial cell membranes has made tual enzyme activity. In many cases, the presence of a par-
this technique underutilized in terms of symbiosis. Recently, ticular prokaryote enzyme within animal, plant, or proto-
these issues have been overcome by increased amplification zoan tissues can provide evidence for a bacterial
of the mRNA signal and rigorous permeabilization steps, al- symbiont-host arrangement and fulfill biochemical and mo-
lowing for the investigation of symbiont function without lecular criteria no. 2 and 3 (see section 16.1.2).
the requirement of symbiont isolation. In the vent mussel For example, microbes can potentially use at least four
Bathymodiolus puteoserpentis, for example, mRNA-FISH re- major pathways for C02 fixation, including the Calvin
16. Methods To Investigate Microbial Symbioses 403

Benson cycle, the reverse tricarboxylic acid cycle, the Coupled enzyme method for measuring RuBisCO
acetyl-coenzyme A CoA pathway, and the 3-hydroxypropi- activity (modified from reference 143)
onate cycle. All of these involve specific enzymes, which
1-3. Follow steps as described in the protocol above.
have been demonstrated by a number of studies investigat-
ing the function of symbionts. In particular, one of the first
4. Make reaction mixture (usually 0.5 ml) containing 50
pmol of Tris-HC1 (pH 7.8), 12 pmol of ATP, 10 pmol of
demonstrations of a chemoautotrophic bacteria-invertebrate
MgC12, 75 pmol of NaHC03 (or KHC03), 0.03 pmol of
symbiosis was based on an enzyme assay for ribulose 1,5-
EDTA (pH 6.5), 10 pmol of glutathione, 1.0 pmol of
bisphosphate carboxylase/oxygenase (RuBisCO), a key en-
D-ribulose 1,5-bisphosphate, 0.15 pmol of reduced NADH,
zyme in the Calvin Benson cycle (51). This enzyme, which
30,000 U of glyceraldehyde 3-phosphate dehydrogenase,
has been measured enzymatically (described below), im-
and 30,000 U of 3-phosphoglycerate kinase.
munochemically, and molecularly (via PCR, sequencing,
5. Add sample (20 to 100 1.1) to reaction mixture and
and expression), is now known to be critical to the inter-
measure change in absorbance at 340 nm, after an initial lag
action between hosts and symbionts in many nutritional as-
of several minutes.
sociations (25, 94, 152). In phallodrine oligochaetes, the
presence of RuBisCO was enzymatically determined in There are a number of other enzymes and biochemical
whole animal homogenates from two host species, suggest- pathways that may be diagnostic for the presence of bacter-
ing production of organic carbon by the symbionts via the ial symbionts. For example, evidence of the reverse tri-
Calvin Benson cycle (53). Similarly, the possibility of au- carboxylic acid cycle has recently been demonstrated
totrophic C02fixation via RuBisCO in the oligochaete I. in epibionts of alvinellid polychaetes (21) and the
leukodermatus was measured, at the ultrastructural level, s-proteobacterial endosymbiont of a snail, Alvinoconcha sp.
using an antibody against a specific form of RuBisCO found (161; S. K. Goffredi, unpublished results), by the positive
in many symbiotic bacteria (94). PCR amplification of ATP-dependent citrate lyase in cer-
tain tissues. Methodologies for measuring many prokaryotic
Enzymatic Technique for Measuring
enzymes in animal (and plant) tissues are well established.
RuBisCO (EC 4.1 .I .39)
General concerns when applying these techniques to the
RuBisCO catalyzes the irreversible carboxylation of ribulose
study of potential symbioses include appropriate negative
1,5-diphosphate to two molecules of 3-phosphoglycerate.
controls (e.g., boiled tissues) and positive controls (or stan-
It is a good indicator for the presence of bacteria and eu- dards).
karyotic algae in animal tissue as it has been found only in
autotrophic organisms. RuBisCO enzyme activity can be 16.2.4.2. Measures of Bacterial-Specific Biomarkers
determined by measuring incorporation of I4C-labeled C02 and Stable Isotopes
(usually in the form of HC03-) into phosphoglycerate, or
There are many bacteria-specific compounds that, if posi-
following the rate of COz-dependent NADH oxidation in a
tively detected within eukaryote tissues, may indicate that
nonradioactive enzymatic assay that couples production of
symbiotic bacteria are responsible for their production. These
3-phosphoglycerate to its phosphorylation and reduction to
include vaccenic acid, a fatty acid produced only by bacteria
glyceraldehyde-3-phosphate.
(86, 149); poly-P-hydroxybutyrate (93, 115), a bacteria-
Protocol for radioactive isotopic method for specific polyester storage product; elemental sulfur or polysul-
measuring RuBisCO activity (modified from fides (135, 150, 167), energy storage products not known to
reference 171) be produced by metazoans and likely too difficult to take up
across membranes from the environment; lipopolysaccharide
1. Make homogenization buffer: lOmM Tris-HC1 (pH
(26); and cell components (e.g., N-acetylglucosamine and N-
7.8 at 25C), 0.1 mM EDTA, 10 mM P-mercaptoethanol.
acetyl muramic acid) and the Vibrio fischeri-derived tracheal
2. Gently homogenize tissue in ground glass homoge- cytotoxin component of petidoglycan required for morpho-
nizer and sonicate (5 X 30 s on ice). Sonication is reported
genesis of the Eupymna scolopes light organ (see section
to cause a threefold increase in RuBisCO activity (24).
16.3). In phallodrine oligochaetes, structures resembling
3 . To remove cell debris, centrifuge homogenate at
poly-P-hydroxybutyrate were observed microscopically and
27,000 X g for 10 to 30 min (at 4C).
later confirmed, via biochemical assays, to account for up to
4. Make reaction mixture (usually 0.5 ml) containing -10% (dry weight) of worm tissues. Additionally, energy-
100 pmol Tris-HC1 (pH 7.6), 5 pmol of MgC12, 25 pmol of
dispersive X-ray analysis indicated the presence of elemental
NaC03, 0.03 pmol of EDTA (pH 6.5), 3 pmol of gluta-
sulfur in the symbiont-containing tissues, suggesting the pres-
thione, 1 pmol of D-ribulose 1,5-bis hos hate, and -2 pCi
P P
of NaHI4CO3. *Alternatively, KH 4c03can be added at
ence of chemosynthetic sulfide-oxidizing bacterial symbionts
(66). Certain body segments of the worm even appeared
the same specific activity.
white, presumably due to sulfur inclusions produced by the
5. Add sample (20 to 100 1.1) to the reaction mixture symbionts themselves, and reflected the distribution of bac-
and incubate for 5 to 10 min at 20 to 30C.
teria on the oligochaete body.
6. Stop reaction with 0.2 ml of 6N HC1 and heat at 90C
Additionally, natural stable isotope measurements can be
for 60 min.
useful in determining the presence of bacteria within eukary-
7. Aliquot 0.1 ml into glass vial. Dry for 1 h at 95C. otic tissues. Many factors can affect the stable isotope value
Add 0.3 ml of water followed by 3 ml of liquid scintillant
within an organism; however, many bacteria possess a dis-
(many are available on the market, e.g., Kodak or Packard
tinct isotopic signature that gives clues to their metabolic
Instruments).
pathways. Isotopic compositions are typically reported as
8. Measure incorporation of radioactivity into the acidi- delta values, defined as follows: 6 (per mil) = 1,000 x
fied samples by using a liquid scintillation counter.
[(Rsampie/Rstandard)- 11 where R is the ratio between two iso-
Units: one unit of RuBisCO is defined as that amount of topes (e.g., l3C/l2c)and the Rstanddrd used is the Vienna
enzyme which catalyzes the carboxylation of 1.O pmol of PeeDee Belemnite (VPDB). Thus, the abundance of the
ribulose diphosphate per min. rare, heavy isotope is always expressed relative to the more
404 w GROWTH

common light isotope. For instance, bacteria that consume B. Density gradients:
methane have 813Cisotopic values that are very light (-40 i. Percoll - 60% (in l x PBS). Establish a gradi-
to -60%0), whereas chemoautotro hic bacteria that fix C02 ent by centrifugation at 13,000 X g (1 h). Overlay
via the Calvin Benson cycle have 6?3Cvalues typically in the the homogenate and centrifuge at 13,000 X g (1 h).
-20 to -30%0 range (with some exceptions, of course). Remove layers and wash by low-speed (750 X g) cen-
Giere et al. measured stable carbon isotopes in phallodrine trifugation (1 h) in 1X PBS, or a buffer solution that
oligochaetes and determined the symbiotic bacteria to be simulates host fluids, for example, imidazole buffer so-
chemoautotrophic, based on similarity of the 8l3C value lution (50 mM imidazole, 500 mM NaC1, 20 mM
( -26%0) with free-living chemoautotrophic microbes (65). MgS04, 10 mM KC1, 10 mM CaC12, pH 7.1) (40).
Stable isotope ratios provide not only clues as to the ii. Nycodenz (also known as Histodenz), 16 to
functioning of the bacterial symbiont but also the potential 28% (usually made up in 1X PBS). Establish the gra-
nutritional integration between the symbiont and host. dient at 4C overnight. Overlay homogenate and
Recently several innovative techniques have been used to centrifuge at 10,000 x g (1 h). Remove layers and
measure stable isotope signatures associated with symbiotic bring each to -2 ml with l x PBS. Wash, via cen-
associations. One such method combines the techniques of trifugation at 10,000 x g for 30 min, and collect pel-
FISH microscopy with secondary ion mass spectrometry let, if present (72).
(130, 131) to measure simultaneously the metabolic func- 2. To confirm successful separation of symbionts from
tion and the identity of individual microbes. This technique host cells, bacterial cell counts can be made via DAPI stain-
was used to investigate the utilization and alteration of spe- ing and epifluorescence microscopy, by acridine orange
cific compounds by syntrophic cooperation of Archaea be- staining, or by hemocytometer and/or light microscopy
longing to the Methanosarcinales, and sulfate-reducing bac- counting.
teria related to the Desulfosarcina genus, described in 3. Suitability of the enriched bacteria for physiological
section 16.1.3. Both members of the consortia had isotopic studies can be assessed via trypan blue or Hoechst staining
signatures that reflected the initial assimilation of isotopi- to observe cell membrane integrity. For trypan blue, mix
cally light methane by the archaeal partner and transfer of cells 1:l with trypan blue solution (0.8 mM in 1 X PBS) for
this carbon to the sulfate-reducing partner (131). less than 30 min. Dead cells stain blue due to trypan blue
uptake. For Hoechst stain, pellet cells briefly (8 to 10 s)
- Studies
16.2.4.3. Physiological at high speed (approximately 16,000 X g) in a micro-
Physiological investigation of currently uncultured sym- centrifuge. Pour off supernatant and resuspend cells in 50 pl
bionts is often difficult due to poorly understood require- of freshly made Hoescht-33342 (100 pg/ml in 1X PBS).
ments for survival and a fragile balance between symbiotic Incubate on ice for 5 min. The Hoescht-33342 stain (which
partners. The physiological dissection of these symbioses is permeable and can stain both fixed and nonfixed cells) is
has been successful, nonetheless, and can take many forms prepared from a 10 mg/ml solution. Cells can be examined
including investigations of symbiont enrichments, pure immediately by fluorescence microscopy or fixed and stored
symbiont cultures (which is rarely achieved), and whole in- for later examination. If cells are fixed in 10% formalin,
tact associations. they can be stored for up to 1 week at 4C without signifi-
cant loss of staining. Live cells have evenly stained nucleo-
16.2.4.3.1. Symbiont Enrichments somes. Dead cells show fragmented nuclei.
In the absence of pure symbiont cultures, much can still 4. Physiological experiments should be initiated imme-
be learned about the physiology, metabolism, and biochem- diately following isolation, if not fixed.
istry of a symbiont if a bacterial enrichment, separated from
host tissues, cellular debris, etc., is achieved. The procedure 16.2.4.3.2. Pure Symbiont Cultures
for initial separation of symbionts from host tissue is de- Improvements in culture methods over the last few
scribed below. Bacterial symbionts have been successfully decades have made the isolation of symbiotic microbes pos-
purified from the vent tube worm Riftia pachyptila and were sible, especially for those that are heterotrophic in nature
observed to excrete succinate and glutamate into the cul- (e.g., shipworm symbionts, described below). Many physio-
ture medium (52). Researchers attributed this microbial re- logical properties of symbionts, including favorable envi-
lease of carbon as the likely source of nutrition for the worm ronmental conditions, can be observed only through the
host. Likewise, Wilmot and Vetter (175) incubated a puri- study of cultured isolates. Although it is an important
fied bacterial fraction from R. pachyptila trophosome with method for the investigation of symbiotic host-bacterial in-
radiolabeled sulfur compounds and discovered the strict uti- teractions, we provide only a brief discussion on the culti-
lization of 35S-sulfide,and not other sulfur compounds, by vation of symbionts, with emphasis on two cases in which
the symbionts. In some cases, metabolic activity of purified symbiont cultures have been obtained.
bacterial fractions was observed to be higher than crude ho- In 1983, Waterbury and colleagues successfully cultured
mogenates (11); thus, the additional step of removing host the symbionts from six species of bivalve shipworm (in-
cellular debris may be advantageous. cluding Lyrodus pedicellatus) by serial dilution ( lo7 to lo8)
Protocol for Symbiont Enrichments of homogenate from the symbiont-containing tissue, the
gland of Deshayes (172). A single species of bacterium was
1. To achieve a preparation from host tissue that is en- isolated from all species with the ability to grow on cellu-
riched in symbiont cells, gently homogenize tissue in 1 x lose as a sole carbon source without a fixed nitrogen source.
PBS in a ground glass homogenizer (keep cold), prefilter Semisolid media (0.2 to 0.9% agar in tubes and plates)
through coarse (i.e., >lO-pm mesh) NitexB (optional), were used to create a gradient of both oxygen and cellulose,
and separate by: allowing the microbe to choose the most suitable envi-
A. Differential centrifugation: spin at 3,000 X g ronment within the isolation chamber, a technique that
(twice) to remove large cellular debris, followed by a alleviates the need for complete knowledge of the in vivo
spin at 15,000 X g to pellet the symbiont cells (11). habitat.
Next Page

16. Methods To Investigate Microbial Symbioses m 405

Similarly, many microbial symbionts, including spiro- and contributes to the nutrition of the host. Douglas et al.
chetes, methanogens, and other microbial representatives (43) demonstrated that only 8% of aphids experimentally
within termite guts, have been cultivated. In the 1990s, stripped of Buchnera by antibiotic treatment survived (com-
Breznak and colleagues achieved pure cultures of termite pared to 72% in nonmanipulated aphids). Aphids cannot
methanogens by using long incubations (>8 weeks), dilu- synthesize essential amino acids and developed normally
tion-to-extinction methods, and antibiotics to which without symbionts only if the diet was supplemented
methanogens were naturally resistant (100, 101). They ex- with specific essential amino acids, based on incorporation
amined oxygen tolerance, pH, and temperature optima, as of 14C-labeled compounds. This provides evidence for the
well as energy sources and nutritional requirements of the utilization of symbiont-produced essential amino acids by
symbiotic methanogens. Acetogenic, nitrogen-fixing spiro- the host (43), thus fulfilling biochemical and molecular cri-
chetes from the termite gut were isolated, for the first time, terion for symbiosis no. 5.
shortly thereafter (74, 102, 105). This isolation of spiro- For successful radiolabeling experiments, one must first
chetes, in an anoxic dilution series that contained anti- choose a labeled substrate that is likely to be utilized by the
microbial compounds (102), facilitated the observation of symbiont (i.e., COz and CH4 for organic carbon produc-
metabolic capabilities that were previously unknown for tion, acetate incorporation into fatty acids, etc.). The spe-
spirochetes, including Hz/CO2acetogenesis and nitrogen cific activity often has to be determined empirically. Ex-
fixation (74, 102, 105). posure of the intact association, maintained in a healthy
state, to the label is usually for a short time period (0 to 6 h).
16.2.4.3.3. Intact Associations Samples are taken at intervals to monitor the movement of
The most direct way to investigate physiological or bio- the label throughout the symbiont and/or host. Samples,
chemical interactions between symbiotic partners is to including symbiont/host tissues or the surrounding
monitor intact associations. This, in the case of many newly medium, can be taken and separated into specific carbon
discovered symbioses, is a difficult prospect when one con- (or other) compounds via liquid or thin-layer chromatog-
siders the lack of knowledge regarding environmental re- raphy, followed by liquid scintillation counting or autora-
quirements for both partners. Nevertheless, many attempts diography.
to manipulate and/or characterize intact symbiotic associa- This technique can also be performed on purified sym-
tions have resulted in a wealth of information, including or- bionts. For example, when incubated with 14C-acetate and
ganismal studies of metabolite production and transfer. 14C-glucose,purified symbionts of termites were found to
Methods for physiological experiments on intact sym- excrete amino acids that were eventually absorbed by the
bioses are too diverse to cover in detail, and creativity is en- host (14). Similarly, Felbeck et al. (53) used autoradiogra-
couraged as most nonmodel associations are somewhat dif- phy with labeled substrates to demonstrate the uptake of
ficult with which to work. Many physiological experiments COz and glucose and production of organic acids by sym-
using respiration chambers and, techniques to measure biont enrichments from two phallodrine oligochaete host
fluxes of gaseous or ionic metabolites have also contributed species, further confirming the potentially autotrophic na-
to our understanding of symbiont and host metabolism in ture of the symbionts.
many symbiotic associations. For example, Girguis et al.
(69) conducted the first live animal experiments detailing 16.2.4.4. Tissue Autoradiography
proton exchange in the deep-sea tube worm host, Riftia
This procedure involves the microscopic examination of
pmhyptila. This symbiosis relies on the ability of the worm visible reduction of silver halide by incorporation of a ra-
to rapidly remove symbiont waste products internally, the
dioactive substance, most commonly a 14C-,3H-, or 35S-la-
absence of which would result in unsuitable conditions for
beled compound, into living tissue through the use of a pho-
both symbiont survival and host protein function. By in-
tographic emulsion (modified from reference 15).
hibiting specific biochemical properties of both partners,
they observed an increase in host elimination of protons as 1. Incubate living (freshly collected) tissue for 1 h in
a result of increased symbiont productivity. Exposure to spe- O.Z-p,m-pore-size filtered sea water (for marine animals)
cific inhibitors of symbiont and/or host function is another containing 40 pCi ml-' of NaH14C03.
effective way to tease apart the functioning of either part- 2. Fix tissue immediately in phosphate-buffered 3% glu-
ner in whole association experiments. Examples of these in- taraldehyde (0.1 M, pH 7.4, made fresh in l x PBS), for 1
clude inhibitors of host ion elimination (e.g., N-methyl- to 2 h.
maleimide and vanadate) (71) and inhibitors of autotrophic 3. Cut semithin (1.5-pm) sections and mount on slides.
COZ fixation via RuBisCO (e.g., D,t-glyceraldehyde) (30). *It may be necessary to embed in paraffin, with a subse-
Successful organismal studies rely on good controls, usually quent procedure to deparafhize, and rehydrate as described
an unmanipulated association (positive control) or incuba- above in section 16.2.3.1.
tion chambers without the organisms present (negative 4. For autoradiography, dip in liquid emulsion (e.g.,
control). Similarly, it is important to have good a priori 50% Kodak Autoradiography Emulsion Type NTB, melted
knowledge of the in situ environmental conditions, such in a water bath at 42C) in darkness, allow to dry.
that environmentally relevant conditions are simulated and 5. Expose for 5 to 14 days in darkness, desiccate at 4C.
healthy individuals are maintained. During exposure, trial slides can be developed. Examine the
One common physiological examination of intact sym- slide at a magnification of X 10 to X40 with a bright-field
bioses includes radiolabeling/autoradiography experiments microscope and look for the presence of black silver grains
(detailed below). These studies mostly examined partition- located over the cells.
ing and movement of radiolabeled tracers and were able to 6. Develop in, for example, Kodak D-19 Developer at
quantify the nutritional importance of bacterially produced 20C for 3 min.
compounds (160). Odelson and Breznak (126), for exam- 7. Wash for 10 s in distilled water.
ple, used 14C-labeledsubstrates to show that acetate results 8. Fix with Kodak Fixer for 3 min.
from the breakdown of cellulose in the intact termite gut 9. Wash in running water for 15 min.
Introduction to Metabolism 423 22. Carbohydrate Fermentations 558
C. A. REDDY, Editor R. MEGANATHAN, YAMINI RANGANATHAN,
AND C. A. REDDY
17. Physical Analysis and Purification
Methods 424 23. Metabolism of Aromatic
SCOTT B. MULROONEY, WILLIS A. WOOD, Compounds 586
AND J. R. PATEREK JEROME J. KUKOR, BORIS WAWRIK,
AND GERBEN J. ZYLSTRA
18. Chemical Analysis 462
LACY DANIELS, RICHARD S. HANSON, 24. Cellulases, Hemicellulases, and
AND JANE A. PHILLIPS Pectinases 596
MICHAEL E. HIMMEL, JOHN 0. BAKER,
19. Enzymatic Activity 504 WILLIAM S. ADNEY, AND STEPHEN R. DECKER
ROBERT P. HAUSINGER AND
ALLEN T. PHILLIPS 25. Lignin and Lignin-Modifying
Enzymes 611
20. Permeability and Transport 527 CARLOS G. DOSORETZ AND C. A. REDDY
ROBERT E. MARQUIS

21. Bacterial Respiration 539


ROBERT P. GUNSALUS, GARY CECCHINI,
AND IMKE SCHRODER
Introduction to Metabolism
C. A. REDDY

Metabolism is central to the question of how microbes rearranged, and updated substantially by Robert P. Hausinger
make a living. The metabolic systems that are involved in and Phillips for this edition. The chapter authored by
generating energy for growth, motion, maintenance, and Marquis on permeability and transport (chapter 20) is essen-
cell division are studied through physical, chemical, and tially intact from the previous edition, with minor revisions.
enzymatic methods. Methods are also needed to study ox- Chapters 21, 22, and 23, Bacterial Respiration,
idative and fermentation mechanisms for producing ATP Carbohydrate Fermentations, and Metabolism of
and biosynthetic intermediates needed by the cell. Aromatic Compounds, are completely new to this edition.
Furthermore, there is a growing interest in studying plant A n understanding of the oxidation and fermentation path-
biopolymers, as exemplified by cellulose, hemicellulose, ways as sources of energy to a microbial cell is of funda-
and lignin, the most abundant sources of organic carbon mental interest to microbiologists. The chapter on bacter-
and energy in the biosphere. There is a great deal of current ial respiration, authored by Gunsalus et al., presents
interest in the economic conversion of these plant poly- methods for studying localization of respiratory enzymes
mers as sources of food, feed, fuels, and chemicals. Methods and related assays as well as basic methods used for study-
to study the above-mentioned topics in metabolism consti- ing aerobic and anaerobic respiratory enzymes.
tute the main focus of this section, which has been signifi- Carbohydrate Fermentations, by Meganathan et al., is
cantly expanded with nine chapters, compared to four one of the most comprehensive presentations of its kind,
chapters in the previous edition. with detailed schematics depicting most of the major fer-
Chapters 17 to 20, dealing with physical, chemical, and mentation pathways, including details on relative ATP
enzymatic analysis and with permeability and transport, are yields and key oxidation/reduction steps in each of the
retained from previous edition of this manual, Methods fur pathways and details of assay procedures for various en-
General and Molecular Bacteriology (MGMB). Mulrooney re- zymes involved in these pathways. In chapter 23, Kukor et
vised and updated chapter 17, previously authored by Wood al. have done an excellent job of presenting succinct and
and Paterek. This chapter deals with the basic physical ana- current information on methods for studying metabolism of
lytical methods and separation procedures such as spec- aromatic compounds.
trophotometry, electrodes, chromatography, radioactivity, Chapters 24 and 25 focus on methods for studying key
and electrophoresis. Chapter 18 on chemical analysis, au- enzymes involved in the degradation of plant polymers.
thored by Daniels et al. as chapter 22 in the previous edition, Himmel et al. present useful information related to the
required relatively little revision and is largely intact in this study of cellulases, hemicellulases, and pectinases, while
edition. Chapter 19 on enzymatic activity, authored by Dosoretz and Reddy discuss current approaches for studying
Phillips as chapter 23 in the previous edition, was revised, fungal lignin-modifying enzymes.

423
Physical Analysis and Purification Methods
SCOTT B. MULROONEY. WILLIS A. WOOD. AND J . R . PATEREK

17.1. SPECTROPHOTOMETRY ................................... 426


17.1.1. Absorbance Spectroscopy ............................... 426
17.1.1.1. Spectrophotometer Components: Dispersive and Diode
Array Instruments............................. 426
. 2. Performance Factors for Dispersive Spectrophotometers .. 426
. 3. Performance Factors for Diode-Array
............................ 428
Spectrophotometers
. .............. 428
4. Determination of Solute Concentration
. ............. 428
5. Determination of Particle Concentration
. .................. 428
6. Determination of Reaction Rates
17.1.2. Fluorescence Spectroscopy.............................. 428
.................... 428
17.1.2.1. Types of Fluorescence Spectra
. .............. 429
2. Common Applications of Fluorescence
. ...................... 429
3. Factors Affecting Accuracy
17.2. ELECTRODES ............................................ 430
17.2.1. Hydrogen Ion Electrode................................ 430
...................... 430
17.2.1.1. Calibration of the pH Meter
. .................................... 430
2. Problems
. ................................. 431
3. Effect of Salt
17.2.2. Other Ion-Specific Electrodes ............................ 431
17.2.2.1. Ammonia Electrode ............................ 431
. .............................
2. Fluoride Electrode 431
17.2.3. Oxygen Polarograph ................................... 432
..........................
17.2.3.1. Effects of Temperature 432
. ......................
2. Calibration and Calculations 432
. 3. Operating Information .......................... 432
. 4. Assembly of Electrode Membrane .................. 432
17.3. CHROMATOGRAPHY ..................................... 432
17.3.1. Ion-Exchange Chromatography ........................... 433
17.3.1.1. Resin Exchangers ............................. 433

..2. Cellulose, Dextran, and Agarose Exchangers


3. Chromatofocusing
..........
.............................
434
434
17.3.2. Adsorption Chromatography ............................. 435
17.3.2.1. Types of Adsorbant ............................
............ 435
435
17.3.3. Reversed-Phase and Hydrophobic Chromatography
.
4. Liquid-Liquid Partition Chromatography .................... 435
.
5. Gas-Liquid Chromatography ............................. 435
17.3.5.1. Principles ................................... 435
. ............................... 436
2. Instrumentation
. .............. 437
3. Sample Preparation and Derivatization
. 4. Metabolic Products............................ 437
. 5. Gas Production and Consumption.................. 437
. 6. Membrane Lipid Analysis for Identification of Bacteria .. 438
. 7. Biomineralization of Xenobiotic Organic Compounds
.................. 438
.... 438
17.3.6. High-Performance Liquid Chromatography
17.3.6.1. Instrumentation ............................... 439
. 2. Guanine-Plus-Cytosine Content of DNA ............ 439
. 3. Separation and Quantitation of Bacterial Photosynthetic
Pigments.................................... 440
17.3.7. Ion Chromatography .................................. 440
17.3.7.1. Instrumentation............................... 440
. 2. Ions Determined.............................. 440
. 3. Eluants..................................... 440
. ...................................
4. Sensitivity 440
. 5. Ion Chromatography Coupled with Atomic
Spectrometry ................................. 440

424
1 7. Physical Analysis and Purification Methods 425

17.3.8. Affinity Chromatography............................... 441


17.3.8.1. Preparation of a Custom Affinity Matrix: Coupling
................................... 441
of Ligand
. ......... 441
2. Special Considerations for Coupling Reactions
. 3. Affinity Chromatography of Tagged Recombinant
.................................... 441
Proteins
17.3.9. Gel Permeation Chromatography.......................... 442
.......................... 442
17.3.9.1. Gel Permeation Media
. ........................ 442
2. Multicomponent Analysis
. 3. Fractionation of Proteins and Determination of Native
............................. 442
Molecular Weight
. ......................... 443
4. Practical Considerations
17.4. RADIOACTIVITY ........................................ 444
.................................. 445
17.4.1. Safety Considerations
. ..................... 445
2. Purity of Isotopically Labeled Materials
. ............................. 446
3. Isotope Dilution Techniques
. .................. 446
4. Calculation of Amount of Isotope Required
. ............................ 447
5. Enzyme Assays with Isotopes
. ................... 448
6. Special Problems with Tritiated Substrates
. ............................ 448
7. Liquid Scintillation Counting
............................... 448
17.4.7.1. Solid Materials
. ............................ 449
2. Polyacrylamide Gels
. ............................... 449
3. Carbon Dioxide
. ....................... 449
4. LSC Instrument Operation
. ............................ 450
5. Counting Solutions
.................................. 451
17.4.8. Statistics of Counting
17.5. ELECTROPHORESIS ...................................... 451
................................... 451
17.5.1. Gel Electrophoresis
.... 452
17.5.1.2. SDS-Polyacrylamide Gel Electrophoresis of Proteins
.................. 453
17.5.2 Other Types of Polyacrylamide Gel Analysis
. ............................... 454
3. Capillary Electrophoresis
. ................................... 454
4. Isoelectric Focusing
. ................. 454
5. Immunoelectrophoresis and Western Blotting
17.6. ENZYME PURIFICATION .................................. 454
.............................. 455
17.6.1. Preparation of Cell Extract
. .............................. 455
2. Removal of Nucleic Acids
. ............................ 455
3. Fractionation by Precipitation
. ........................... 456
4. Fractionation by Denaturation
. .......................... 456
5. Purification by Chromatography
. ........................... 456
6. Integrated Purification Systems
. ........................... 457
7. Affinity Elution with Substrate
. 8. Dye Ligand Chromatography ............................ 457
. ....................................... 457
9. Crystallization
17.7. CONCLUDING COMMENTS AND FUTURE TRENDS ............ 457
17.8. REFERENCES ............................................ 458
17.8.1 Photometry ......................................... 458
............................ 458
17.8.1.1. General References
. ............................. 458
2 Specific References
....................................... 458
17.8.2. Ion Electrodes
............................ 458
17.8.2.1. General References
. ............................. 458
2 Specific References
17.8.3. Chromatography ..................................... 458
............................ 458
17.8.3.1. General References
. ............................ 459
2. Specific References
........................................ 460
17.8.4. Radioactivity
............................ 460
17.8.4.1. General References
. ............................ 460
2. Specific References
................................... 460
17.8.5. Gel Electrophoresis
............................ 460
17.8.5.1. General References
. ............................. 461
2 Specific References
................................... 461
17.8.6. Enzyme Purification
............................ 461
17.8.6.1. General References
. ............................. 461
2 Specific References
17.8.7. Concluding Comments and Future Trends ................... 461
17.8.7.1. General References ............................ 461
. 2 Specific References............................. 461
426 METABOLISM

This chapter deals with the basic physical analytical meth- ods, circular dichroism is the most popular; it can be used to
ods and separation procedures of spectrophotometry, elec- examine structural features of nucleic acids and proteins as
trodes, chromatography, radioactivity, and electrophoresis. well as observing stereoisomer preference of an enzymatic
Application of these methods in microbiology has become reaction by substrate-monitored turnover. More details can
routine. The approach in this manual is to give a general de- be found in reference 3.
scription along with strengths and weaknesses of a method,
specific techniques and preparations where appropriate, and 17.1 .I .I. Spectrophotometer Components:
examples of application to investigations with bacteria. The Dispersive and Diode Array Instruments
purification of enzyme proteins is particularly exemplified. All absorbance spectrophotometers have several common
In this chapter, especially, it has been necessary to con- components. These are described briefly below along with
fine the scope to the simple procedures which are appropri- how they fit into the overall instrument design. The first el-
ate for laboratory classes or are routinely used in a research ement, the light source, is typically a tungsten-halogen
laboratory. In-depth treatment of the methods presented lamp for visible-light wavelengths of 350 to 800 nm, and a
here can be found in books specifically dedicated to indi- deuterium arc lamp for UV wavelengths of 190 to 350 nm.
vidual techniques, some of which are listed in section 17.8. Another type of light source is a xenon lamp that emits a
In addition, catalogs and commercial literature from manu- broad range of 200 to 900 nm; thus, a single lamp can be
facturers and suppliers contain a wealth of information that used instead of deuterium and tungsten lamps. Xenon lamps
is highly valuable to the investigator. Some specialized have the added advantage of providing higher-energy emis-
techniques such as thin-layer chromatography and capillary sions in the UV range than deuterium. Furthermore, new
electrophoresis are mentioned only briefly. designs incorporate a flash xenon lamp that is energized
only when a reading is being taken.
Energy emitted from the light source then passes through
17.1. SPECTROPHOTOMETRY a very narrow entrance slit. The light passing through the
A wide variety of photometric methods have found impor- slit is usually collimated by a mirror or lens with the overall
tant uses in bacteriological research and applications. goal of presenting nearly parallel light rays to the sample.
Absorbance spectroscopy and spectrofluorimetry (1-6) (The location of the slit differs in typical diode-array instru-
have been particularly useful. These have facilitated identi- ments, where the light is passed first through the sample and
fication of compounds, determination of structure, estima- then through the slit.) In dispersive spectrophotometers, the
tion of concentrations, and measurement of enzymatic re- light is next directed to an element that separates different
action rates. For details of photometric measurement of wavelengths. This element, typically a diffraction grating
bacterial growth, see chapter 9.6. (but some instruments use a prism), is mounted on an as-
sembly connected to a step motor that allows the spectrum
17.1.I. Absorbance Spectroscopy to be scanned at various rates.
UV-visible spectrophotometers can be classified into two The light is next directed through the sample compart-
broad categories: those containing a wavelength-dispersive ment which contains a cuvette holder. Dispersive spec-
monochromator that sends monochromatic light through a trophotometers can pass the light through a single sample
sample, and diode-array instruments that pass a broad spec- (single beam), or the light is first split into two beams (dou-
trum through the sample and subsequently measure light at ble beam) where one passes through a reference cell and the
individual wavelengths. No matter what the design, the second passes through the cell being measured. The double
basic principles of light absorption are common to all spec- beam design allows for correction of any fluctuation in lamp
trophotometers. Light from a radiant source is directly or in- energy by always measuring the difference between the refer-
directly (through a monochromator) passed through the ence and sample beams. After leaving the sample compart-
sample to a detector, which quantitates the energy received ment, the light is quantitated by a detector that converts the
and expresses it as the ratio of the transmitted light (IT) to energy to a voltage. Typical dispersive spectrophotometers
the incident light (lo).When I. is set at 100, the instrument use photomultiplier tubes while the photodiode array instru-
response to light passing through the sample is percent ment projects the light off a zero-order grating onto the array,
transmittance (%T). This can be related to the molar con- where each diode responds to a discrete wavelength. The in-
centration (C) of an absorbing solute through the Beer- strument contains circuitry that polls the voltage of each
Lambert law: diode. One advantage of the diode-array design is that all of
the diodes can be scanned in as little as a few milliseconds,
-log %T = E ~ C
=A
making data collection less time-consuming.
where E is the molar extinction coefficient of the absorbing Finally, most moderate to high-end spectrophotometers
species at a specified wavelength and 1 is the length of the produced today have some type of computer interface. This
light path in centimeters. Since both E and 1 are constant in allows the researcher complete control of the instrument
practice, C is proportional to the negative logarithm of %T from the computer and allows more rapid saving, display,
or directly proportional to absorbance (A). For this reason, and preparation of presentation quality results.
newer spectrophotometers reading linearly in A are more
convenient and more accurate for samples of moderate to 17.1 .I .2. Performance Factors for Dispersive
high absorbance. Spectrophotometers
Other spectroscopic methods that are not discussed in For laboratory spectrophotometers, quality is determined by
detail here are measurement of the ability of an optically ac- the purity of the light presented to the sample, the intensity
tive chromophore to rotate polarized light (optical rotatory and area of the beam (numerical aperture), wavelength ac-
dispersion) or the differential absorption of right- and left- curacy, the accuracy of photometer calibration, the linear-
polarized light (circular dichroism). The spectropolarimeter ity of response, signal-to-noise ratio, and short- and long-
instrumentation is complex and expensive, and is not rou- term drift. Manufacturers of some instruments go to great
tinely found in the microbiology laboratory. Of these meth- lengths and expense to achieve high quality.
17. Physical Analysis and Purification Methods w 427

Effect of spectral bandwidth. Spectral purity of light from a wavelength region at which determinations are to be
monochromator, or spectral bandwidth, is the width in made.
nanometers at one-half the height of the emission en- Newer spectrophotometers in wide use can make ac-
ergy peak at a specified nominal wavelength (Fig. 1B). curate absorbance measurements with an upper range of
This bandwidth is specified for the instrument and is 1.5 A, although some designs can go as high as 2.0 to 5.0
not readily determined in the laboratory. Bandwidth is A. The ability to maintain linearity at such high ab-
reasonably constant across the wavelength range in sorbance is the result of newer designs that incorporate a
monochromators in which a grating is the dispersing el- premonochromator in the light path between the light
ement but varies with wavelength for prism monochro- source and the monochromator. This results in decreased
mators. It varies with slit width in both cases. The ac- stray light and better sensitivity to the minute amounts
curacy of a measured absorbance, assuming no stray of light impinging on the detector. Furthermore, many
light (see below), depends on the ratio of the spectral spectrophotometers are now designed to be operated
bandwidth (a property of the instrument) to the natural with sample compartments open while eliminating most
bandwidth (the width in nanometers at one-half the of the potential for room light that would otherwise in-
peak height) of the peak of the solute in the light beam terfere with the optical measurements.
(a property of the chromophore in solution) (Fig. 1A). Photometric accuracy and linearity. For certain spectropho-
Therefore, in an inexpensive spectrophotometer with a tometers, electronic adjustment of photometric calibra-
spectral bandwidth at 340 nm of 8 nm, a peak with an tion is not available and linearity of response is assumed.
80-nm natural bandwidth (giving a ratio of 0.1) allows It is a good policy to routinely check both absorbance ac-
absorbance measurements with 99.5% accuracy (9). curacy and linearity of absorbance measurement. This
Similarly, with an 8-nm bandwidth, 99% accuracy is allows the researcher to have confidence in mea-
achieved with a chromophore with a 50-nm natural surements for a particular series of experiments and can
bandwidth. The most commonly measured material at serve as an early indicator that some component of the
340 nm, NADH, has a natural bandwidth of 58 nm at instrument is failing. For instruments for which ab-
that wavelength. Therefore, it is readily apparent that sorbance calibration is possible, neutral density filter
absorbing compounds such as NADH can be measured standards of known absorbance at specified wavelengths
at better than 99% accuracy in this inexpensive spec- are supplied and are used to adjust the photometer re-
trophotometer (4, 9). sponse to give the same absorbance value as the stan-
Effect of stray light. Another consequence of wide spectral dards at one or a few absorbance values. With linearity
bandwidth, or low spectral purity, is an increased con- between these points assumed, the total absorbance
tent of stray light (i.e., energies at wavelengths removed range is calibrated. Most spectrophotometer manufac-
from the nominal value and not part of the spectral turers provide protocols to check for instrument per-
bandwidth wavelengths). Since spectral bandwidth for formance and linearity.
prism monochromators is related to both slit width and Effect of slit width. For prism monochromators working at a
wavelength, so also is stray-light content. Stray light de- fixed slit setting, there is a very marked nonlinear de-
rives from incomplete removal of unwanted wave- pendency of band pass on wavelength. Operating manu-
lengths owing to faulty design or dirty optics. The stray- als from spectrophotometer manufacturers and books on
light content of the exit beam diminishes the linear spectrophotometry contain graphs of this function.
range of response to chromophore concentration, i.e., Thus, to make measurements at constant band pass with
the range where the Beer-Lambert law applies. The op- prism optics, take the slit setting at the wavelength of
erator should determine the useful linear range in the measurement from the graph. To be rigorous, determine
the solute concentration at the slit width setting used
for the determination of its extinction coefficient. Con-
versely, when reporting an extinction coefficient, state
2 I I the band pass used. For prism instruments this involves
I I

e. both the slit width and the graph of band pass versus
h

-I Natural Band ----.+I


>\ Width wavelength for that instrument.
c
rn
C
Similarly, spectra should be acquired at constant
a,
c band pass, not constant slit width. The absorbance val-
-
c ues at fixed wavelength or the absorbance spectra deter-
a- 1
h

v
mined at two different slit widths seldom are identical.
The error is magnified when the absorbance peaks of the
8C solutes are sharp and may be negligible when peaks are
m broad. Therefore, it is prudent to use the minimum slit
4? width. Slit width also is a function of the sensitivity of
0
rn the photometer, the spectral characteristics of the pho-
2 todetector, and the source. More importantly, the inten-
0 sity of the lamp, cleanliness of optical surfaces, and crit-
270 290 310 330 350 370 390 ical focusing of the source on the slit greatly affect slit
Wavelength (nm) width. As a rule of thumb, the average spectrophotome-
ter should be capable of nulling by opening the slit to 2
FIGURE 1 (A) Natural bandwidth diagram of NADH; the mm at 210 nm or lower. If this is not possible, one or
bandwidth is 58 nm. (B) Spectral bandwidth diagram more of the above factors may require attention.
(schematic) of spectrophotometer exit beam; the spectral Single-beam versw double-beam spectrophotometers. Double-
bandwidth is 8 nm. beam instruments are constructed so that there are two
428 rn METABOLISM

light beams giving a simultaneous or parallel comparison 17.1.1.6. Determination of Reaction Rates
between I. and IT. Single-beam units make these two When either the product or the reactant of a reaction is a
measurements serially, whereas double-beam spec- chromophore, the rate of chromophore production or uti-
trophotometers split the light beam so that part of the lization can be determined at fixed wavelength by reading
beam passes through the solution used to set the instru- the absorbance at time intervals or by monitoring the ab-
ment at I. = %T = 100or A. = 0, while the second part sorbance change versus time ( 2 , 8). A plot of absorbance
passes through the sample to measure %T or Al. For this versus time permits determination of velocity from the
approach to be valid, the parallel optical systems and slope of the line. Specialized spectrophotometer attach-
their associated electronics must perform identically. In ments that allow for temperature control and/or stirring of
single-beam units, the same end is achieved by first set- the sample cuvettes have been developed for this purpose.
ting %T = 100 or A. = 0 with the blank and then read- Automatic sample changers can be used to assay several
ing %T or Al when the sample is introduced into the simultaneous reactions and record readings at intervals as
light beam. short as 10 to 20 s, and spectrophotometers are available to
record kinetic results from 96-well plates. Spectrophoto-
17.1.1.3. Performance Factors for Diode-Array meters equipped with a computer interface usually have
Spectrophotometers software available to rapidly calculate and plot reaction pa-
Many of the performance factors mentioned above also rameters.
apply to diode-array instruments. One disadvantage of this
design is that the photodiode array has lower sensitivity to 17.1.2. Fluorescence Spectroscopy
light compared to photomultipliers. Instrument manufac- Fluorescence is the absorbance of light by a chromophore
turers compensate for this by increasing the intensity of (fluorophore) with concomitant emission of light at a
light that is presented to the sample; however, this can have longer wavelength (lower energy) (2, 5). Since different
a deleterious effect on samples that are light sensitive. This wavelengths are involved in excitation and fluorescent
shortcoming is to some extent ameliorated by the ability to emission, the instrument must be designed to prevent exci-
scan the entire spectrum rapidly, minimizing the exposure tation energy from reaching the photodetector. This is usu-
time of the sample to the intense light. Most diode-array ally accomplished by placing the detector at 90" to the
spectrophotometers are single-beam instruments and usu- excitation beam. Thus, the measurement involves the ap-
ally have motor-driven cuvette holders that place the de- pearance of radiant energy against the very low level of
sired sample in the light path. Instruments should be background light from the blank and the signal-to-noise
checked at regular intervals for precise alignment of the cell ratio is very high, so fluorimetric methods are inherently
holders. In some cases, black-masked cuvettes provide im- more sensitive than photometric methods that are based on
proved spectral data by reducing the amount of stray light absorption of light. For example, the fluorescent method for
that enters the detector. determination of NADH is as much as 1,000 times more
sensitive than the spectrophotometric method. Fluo-
17.1.1.4. Determination of Solute Concentration rescence intensity depends on the concentration of fluo-
Direct concentration determination of a chromophore is rophore and, unlike absorption, also depends on the inten-
straightforward as long as there is no significant interfer- sity of the excitation beam. Stray light arising from
ence by other absorbing species. With a properly calibrated scattering both in the solution and from surfaces can be a
spectrophotometer and standard l-cm light path cuvettes, major problem, but this can be minimized by placing before
absorbance at the wavelength specified for the molar ex- the photodetector a sharp cutoff filter, which passes light at
tinction coefficient ( E ) of a chromophore is measured. the wavelength of fluorescent emission but blocks excita-
Calculation of A/& yields molar concentration. If the ex- tion energy.
tinction coefficient for a particular chromophore is not Since fluorescence intensity is dependent on excitation
known, it can be easily measured. In that case, prepare a intensity, lamp emission characteristics and stability are of
standard regression line which relates absorbance to con- major importance. Some instruments monitor beam inten-
centration for a set of concentration standards that are mea- sity through a second photodetector and display the ratio of
sured directly. The plot will show the linear region and the fluorescence to excitation intensities. In other spectrofluo-
variability of individual determinations. Standard statistical rimeters, the excitation beam is split, with one going to the
considerations apply, i.e., replication, least-square fits of the sample and the other going to a cuvette containing a refer-
data to straight lines, determination of standard deviation ence dye such as rhodamine B, resulting in a ratio spectrum
from the mean, and standard error. If a particular substance (see below). At low emission intensities, photodetector out-
to be measured has no significant absorbance, then second- put voltage is nearly linear with intensity; hence, concen-
ary color-development reactions will be needed. In all cases, tration can be determined by measuring fluorescence at
care must be taken to ensure that the absorbance readings wavelengths characteristic for a particular fluorophore. The
are within the range of linearity for the instrument. process basically involves preparation of a solvent blank
and a set of standards, construction of a standard curve, and
17.1.1.5. Determination of Particle Concentration comparison with unknowns.
Spectrophotometers and colorimeters can also be used to 17.1.2.1. Types of Fluorescence Spectra
determine the concentration of microbial cells. The
method depends on attenuation of the transmitted beam as Excitation and emission spectra. Most spectrofluorimeters
a result of light scattering rather than absorbance. The pho- possess two wavelength dispersive elements; an excita-
tometric response does not follow the Beer-Lambert law; tion monochromator that directs single-wavelength
however, a calibration curve relating instrument response light to the sample cuvette, and an emission monochro-
to bacterial cell concentration, for instance, gives satisfac- mator positioned at a right angle relative to the excita-
tory results (see chapter 9.6). tion beam that scans the spectrum of light emitted by
17. Physical Analysis and Purification Methods 429

the sample. One monochromator can be used to scan a example of corrected excitation and emission fluores-
spectrum while the second monochromator is held at a cence spectra compared to an absorbance spectrum is
fixed wavelength. A n excitation spectrum is taken by shown in Fig. 2, where panel A is an absorbance spec-
scanning the excitation monochromator and recording trum of the flavin adenine dinucleotide-containing
the emitted light at a wavelength that is maximal for thioredoxin reductase (expressed as extinction coeffi-
that particular sample. For example, the excitation spec- cient) and panel B shows the corrected fluorescence ex-
trum of NADH is taken by scanning the excitation citation spectrum on the left and the fluorescence emis-
monochromator while monitoring the emission at a sion spectrum on the right. Note how the shape and
fixed wavelength of 470 nm. A n emission spectrum is peak locations of the corrected fluorescence excitation
taken by holding the excitation monochromator wave- and absorbance spectra are nearly identical. Computer
length of peak absorbance (340 nm for NADH) while interfaces available on many instrument models make
scanning the emission monochromator. this procedure simple. It is important that researchers
Corrected and uncorrected specma. Spectrofluorimeters pro- state in published spectra whether they are corrected or
duce spectra that are inherently distorted due to varia- uncorrected and what instrument model and settings
tion of lamp intensity across the spectrum, variation in were used.
detector sensitivity at different wavelengths, and other
optical and electronic artifacts. For many purposes such 17.1.2.2. Common Applications of Fluorescence
as single-wavelength reaction assays or a series of exper- Fluorescence spectroscopy can be a useful tool for a wide va-
iments all done on the same instrument, direct, or un- riety of analytical situations. In kinetic assays such as en-
corrected, spectral data are sufficient. If, however, the zyme reactions utilizing NAD(P)H, there is much greater
spectra are to be compared with those obtained on dif- sensitivity than in absorbance-based monitoring. Intrinsic
ferent instruments or compared with absorbance spectra fluorescence of proteins or enzymes containing tryptophan
as shown in Fig. 2, then some correction must be made can be observed to obtain data on unfolding, substrate bind-
for the instrument artifacts. A corrected spectrum is re- ing, and conformational changes because of the extreme
corded on a double-excitation beam spectrofluorimeter sensitivity of this fluorophore to small changes in its envi-
where the instrument aberrations are subtracted out, ronment (5, 10). Various fluorescent dyes (fluorescent
usually by multiplying the data by a calibration spectrum donor), used in conjunction with dyes that absorb light
file. This calibration is typically performed by placing emitted by the donor (acceptor), can be used to estimate
two rhodamine B-containing cuvettes in the reference intermolecular and intramolecular distances by resonance
and sample compartments and simultaneously scanning energy transfer (5). Green fluorescent protein, a commonly
both the excitation and emission monochromators. A n used reporter for gene expression studies, is quantitated
using fluorescence-based methods like UV microscopy,
multiwell high-throughput screening, and cell sorting (7).

y
- 12000 17.1.2.3. Factors Affecting Accuracy
Instrument variation and drift. Care should be taken to cor-
rect for or prevent fluctuations in excitation intensity as
a result of variations in lamp intensity. It may be neces-
sary to introduce the solvent blank and standards fre-
w .o quently. It is best to use a known standard that has fluo-
5
0
3000 rescence properties similar to what is being measured
and to check that an identical reading is obtained
throughout the length of the experiment. If readings
begin to drift, then signal gain can be adjusted to main-
tain accuracy.
Absorbance of solutions. High absorbance of solutions pro-
duces two kinds of errors: (i) reabsorption of fluorescent
energy to give an apparently lower fluorescence yield,
and (ii) excessive absorbance of excitation light due to
high absorbance by the sample at this wavele2gth. For
this reason, absorbance should not exceed 0.2 A, and in-
dependent absorbance measurements on standards and
unknowns should be made at the same excitation and
300 400 500 600 700 emission wavelengths.
Wavelength (nm) Fluorescence of impurities. The limiting sensitivity is deter-
mined by background fluorescence. This is minimized by
FIGURE 2 Comparison of absorbance and corrected fluores- use of pure reagents. In addition, errors can result from
cence spectra. (A) Absorbance spectrum of the flavin-contain- nonuniform contamination of samples and glassware by a
ing enzyme thioredoxin reductase. (B) Corrected fluorescence variety of fluorescent materials, including rubber, grease,
spectra: the excitation spectrum (excitation with a scanned fingerprints, and extracts of filter paper or dialysis tub-
spectrum and recorded at 530 nm emission) is on the left ing. Turbidity contributes to error through scattering,
(solid line) and the emission spectrum (recorded by exciting at which, unless an excitation energy-blocking filter is pres-
455 nm and recording the spectrum of emitted light) is on the ent, contributes to the measured fluorescence intensity.
right (dashed line). Note the similar shapes and peak locations Therefore, maintenance of scrupulously clean glassware
of the absorbance and fluorescence excitation spectra. for measurement and storage of solutions is necessary.
430 METABOLISM

Many of the above effects can be minimized by the use evolved or consumed. In this application, alkali or acid is
of blocking filters at the photomultiplier. added to maintain constant pH; a plot of acid or alkali con-
Effects of temperature and ionic strength. Since fluorescent sumption versus time expresses the reaction progress. For
phenomena are temperature dependent, all mea- instance, the oxidation of glucose and glucose 6-phosphate
surements should be made at constant temperature, either enzymatically or chemically, and the phosphorylation
preferably in a thermostated cell. Fluorescence is also ex- of substrates with adenosine triphosphate by kinases, are re-
tremely sensitive to ionic strength, and care must be actions that can be tracked with the glass electrode.
taken when performing experiments that will result in
changes in the ion concentration of the sample. 17.2.1 . I . Calibration of the pH Meter
The system must be calibrated by using buffers of known
pH. A popular but expensive approach to calibration is to
17.2. ELECTRODES buy prepackaged liquid or solid buffers of specified pH, but
this is by no means necessary or superior. Table 1 lists some
The Nernst equation, standard buffers (13) which have dependable pH values.
RT [ox] With linear electrode behavior, a single standard buffer
E = E, + -1n- should serve to calibrate across the useful range. However,
nF [red]
electrode behavior is seldom ideal; hence, it is wise to use
can be used to measure the concentration (activity), or the two standard buffers (for instance, pH 4 and 6.88) and to
ratio of concentrations, of certain molecules or ions. E is bracket the pH region of the unknown.
the measured potential, Eo is the standard electrode poten-
tial, R is 8.315 J/T, T is the absolute temperature, n is the 17.2.1.2. Problems
number of electrons, F (Faraday) is 96,500 C, and [ox] and The major problems in measurement of pH come from non-
[red] are the concentrations of oxidized and reduced mem- linearity in the alkaline region, absorption of C02 in neu-
bers of the half-cell pair, respectively. tral and alkaline samples and in standards, and slow evapo-
This basic behavior has made possible the determination ration of standards. Additional problems of unsatisfactory
of a number of biologically important molecules, e.g., pro- instrument behavior commonly arise from poor mainte-
tons, HI, 02,ammonium, and chloride. Incorporation of nance of electrodes (16). The glass electrode needs an equi-
these principles into a usable instrument involves (i) the librium-hydrated silica gel layer for accurate and constant
availability of a measurement electrode capable of sensing pH measurement. Electrode manufacturers recommend that
the potential generated by the internal oxidant-reductant a new or cleaned electrode be soaked in water, or in buffer
system, (ii) a standard half-cell of known potential, and (iii) followed by water, for up to several days to establish the gel
an electrometer or potentiometer which measures the volt- layer. They also recommend that the electrodes be main-
age between the two half-cells (11-13). Solution of the tained in distilled water between measurements. Electrodes
Nernst equation for the measuring half-cell results from the must be cleaned after use by washing and careful wiping to
relationship remove adhering materials.
Unfortunately, incomplete cleaning plus storage in water
E (measuring half-cell) = E -t EO can result in a buildup of contaminants and fouling of the
The ability to measure a specific molecule is dependent on glass surface. Lipid and protein contribute to the nonlinear-
the construction of the measuring electrode. For instance, a ity of electrode behavior and produce a slow response. In ad-
glass electrode develops a potential proportional to hydrogen dition, bacterial growth may occur, with both fouling of the
ion concentration, and several ion-selective electrodes de- glass surface and plugging of the asbestos fiber which func-
velop potentials for specific ions. An ion-specific oxidation- tions as a salt bridge between the standard electrode and the
reduction occurs in the coating material that isolates the solution being measured. The KC1 level in the calomel elec-
electrode from the medium. There are, in addition, many trode should be maintained, and the cap should be removed
other strategies for determination of concentration of various during measurement so that KC1 drainage through the fiber
ions on the basis of electrode behavior. The oxygen electrode prevents impurities from lodging in the fiber.
device, for instance, is a form of polarograph, because there Once the glass electrode is fouled, the temptation is to
must be a polarizing voltage to ionize the oxygen. replace it with a new one. However, it can often be cleaned
17.2.1. Hydrogen Ion Electrode
The glass electrode is the only practical means of measuring
hydrogen-ion activity (pH). A thin membrane of special TABLE 1 Standard buffers for calibration of pH meters
glass isolates a silver-silver chloride half-cell from the mea- at 25C"
sured solution (12-14). The electrical potential of the glass PH Buffer
electrode varies linearly with pH over a wide range, pH 1 to ~~

11. In the region of pH 11 and above, there is nonlinearity 1.10.. . . . . . . . . 0.1 M HCl
unless a special alkali-resistant glass is used. The reference 2.16 . . . . . . . . . . 0.1 M potassium tetroxalate
is a standard calomel (Hg/Hg2C12)or Ag/AgCl half-cell,
3.56 . . . . . . . . . . Saturated potassium hydrogen tartrate
which is connected to the measured solution by a salt bridge
of KCL. Both the glass electrode and the standard electrode 4.00 . . . . . . . . . . 0.05 M potassium hydrogen phthalate
are immersed in the sample or assembled into a single, com- 6.88 . . . . . . . . , . 0.025 M KH2P04+ 0.025 M Na2HP04
bination electrode. The latter can be constructed to enter 7.41 . . . . . . . . . . 0.008695 M KH2P04+ 0.03043 M Na2HP04
long test tubes or to accommodate very small samples. 9.22 . . . . . . . . . . 0.01 M sodium borate decahydrate (borax)
In addition to the static determination of the pH of sam-
12.45 . . . . . . . . . . Saturated calcium hydroxide
ples, the pH meter or an automated version can be used to
monitor over time the reactions in which a hydrogen ion is "Data from reference 1
17. Physical Analysis and Purification Methods w 431

effectively by soaking alternately in 0.1 N HC1 and 0.1 N lutions to provide a stable activity coefficient for the par-
NaOH at 50C for several cycles, followed by reestablish- ticular ion to be measured. Reagents such as Total Ionic
ment of the gel layer by soaking. In difficult cases, specific Strength Adjustment Buffers can be used to bring the ionic
methods recommended by the manufacturers should be em- strength within proper parameters for an ion-specific elec-
ployed for more aggressive cleaning. trode (18).
17.2.1.3. Effect of Salt Sensitivity
A high ionic strength of the solution being measured results With respect to sensitivity, various manufacturers' specifica-
in errors in pH measurement by as much as 0.5 pH unit. tions show that measurements can be made over the range
This problem is commonly encountered in preparing high of lop6 to lop8 M to 1 M for certain ions. However, at
molarity ammonium sulfate solutions in the neutral and al- lower concentrations some time is required to establish
kaline pH region. I t is common practice to dilute such sam- equilibrium with the electrode. Seldom is the sensor highly
ples 120 before pH measurement. Dilution of solutions of specific. In fact, interference with other ions is quite com-
completely dissociated salts has little effect on pH because mon. For instance, a calcium electrode gives 10% res onse
the ratio of ions remains nearly constant. The magnitude of to equivalent amounts of Pb2+, Zn", CU", and Fc?' in
the salt effect for specific circumstances is ascertained by the 10-5 to M range. Chloride electrodes respond sim-
making up standards in the salt solutions involved. ilarly to iodide and bromide, i.e., 10% response at 10-5
to M. O n the other hand, the K' electrode is quite in-
17.2.2. Other Ion-Specific Electrodes sensitive to Na', Li', Cs', and H'. The specific details of
Ion-specific electrodes are available for electrometric deter- sensitivity and selectivity depend on the particular elec-
mination of a large number of ions and gases (Table 2) (12). trode being used. Refer to the manufacturer's literature for
Three examples of commonly used electrodes, ammonium, detailed information.
fluoride, and oxygen, are briefly described below. Although
17.2.2.1. Ammonia Electrode
most of these have applications in industrial processes, sev-
eral measure biologically important ions with sufficient sen- The ammonia electrode uses a gas-permeable membrane
sitivity and selectivity to be of use in bacteriological inves- (12, 18, 19) through which ammonia diffuses to react with
tigations. Ion-specific membrane probes usually have a solid the filling solution:
crystalline or compressed crystalline pellet, or a plastic poly- NH3 + HzO = NH, + OH-
mer impregnated with ion-carrier organic molecules. In ad-
dition, there are now many proprietary designs in use. The The hydroxide level of the filling solution is then measured
majority of these electrodes are constructed of polyvinyl against a reference electrode. The electrode is specific for
chloride, glass, or epoxy. Electrode response is, broadly ammonia; however, the ammonium ion concentration can
speaking, the result of an ion-exchange process, and poten- be measured after deprotonation by addition of NaOH to
tials follow the Nernst equation or one of its expanded pH 11. Only volatile amines interfere, in the range from 1
forms. The response times for many ion-specific electrodes X lop7to 5 x lop7M. Nitrate and nitrite can also be mea-
are much slower than for pH electrodes; it can take from sured after reduction to ammonia, and urea-N can also be
30 s to 2 min for a stable reading to be obtained, especially measured in a special adaptation of the ammonia electrode
at very low concentration ranges. in which the enzyme urease is immobilized on the mem-
brane (17). Alternatively, the samples may be pretreated
Calibration with urease and the normal ammonia electrode may be used
Since there is no system for absolute calibration of probes, for measurement.
as there is for pH with the hydrogen electrode, it has been
common practice to use dilutions of stock solution stan- 17.2.2.2. Fluoride Electrode
dards made up of a completely dissociating salt (18). Many The fluoride electrode is typical of a solid-state type of sen-
concentration standard solutions are commercially avail- sor. The membrane consists of a single lanthanum fluoride
able for a wide variety of ions. These stock solutions are crystal that is doped with europium fluoride to increase the
tested by more expensive methods to verify the specific ion conductivity. Some interference from hydroxide anion oc-
in each production lot. curs, but this can be minimized by maintaining the pH
Ion-specific electrodes generate a potential in response within the 5 to 8 range and using Ionic Strength Ad-
to the activity of a particular ion, not necessarily its con- justment Buffers where appropriate. When measuring F- at
centration. Thus, for many types of electrodes when mea- very low concentrations, it may be necessary to reduce in-
suring within certain concentration ranges, it is necessary to terference by taking measurements in plastic or Teflon con-
adjust the total ionic strength of standard and unknown so- tainers instead of glass.

TABLE 2 Ion-selective electrodes"


Type Examples of ions and gases detected
Glass membrane. ........................ H f , Na+
Solid state. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Ag+, Br-, Cdzf, C1-, CuZf,CN-, F-, I-, Pb2+,S2-
Gas sensing. ............................ NH3, C02,SO2
Polymer membrane. ...................... NH,,Ca2+,BF4-, Li+, NO,-, K+
' Data compiled from manufacturers' specifications.
432 rn METABOLISM

17.2.3. Oxygen Polarograph two times or obtained from the slope of plots of percent full
A platinum electrode at -0.5 to -0.8 V consumes oxygen scale versus time involving several readings or continuously
according to the equation recorded in a real-time assay.
Unfortunately, it is unlikely that reaction mixtures
O2 + 2 H 2 0 + 4e- + 40H under study have the same composition as the oxygen stan-
The equation shows that the process occurs with the dard, and hence, there will be some error. However, the
flow of electrons or current toward the electrode. This cur- error can be minimized by maintaining a low total solute
rent flow through a resistor appears as a voltage across the concentration. When solutions of greater oxygen concen-
resistor. The potential of such a half-cell, when used with tration are needed, increased partial pressures can be gener-
a standard Hg/Hg2Cl2 or Ag/AgCl (silver wire in chloride- ated by saturation of standards and working solutions with
containing solution) half-cell, can be measured if the two oxygen or oxygen-nitrogen mixtures.
are connected by a salt bridge. Alternatively, the current The oxygen content of standards and reagents is influ-
may be measured with a galvanometer or the voltage can be enced by temperature and altitude. Barometric pressure
measured after amplification with a potentiometer, and fluctuations seldom change oxygen concentration by more
these do not require a reference electrode. than 3%. Correction of the oxygen solubility coefficient for
The platinum electrode may be stationary, rotated, or os- barometric change involves the relationship
cillated to minimize diffusion gradients. However, such observed barometric pressure
electrodes tend to be poisoned in biological measurements.
A popular solution to this problem is the Clark oxygen elec- 760
trode ( Z O ) , in which the platinum electrode is covered with x solubility at 760 mm Hg (101.3 kPa)
a gas-permeable membrane. Whereas the platinum elec-
trode measures the number of oxygen molecules present at 17.2.3.3. Operating Information
its surface as governed mostly by collision theory, the Clark At the beginning of an experiment, the probe should be
electrode response is proportional to the rate of diffusion tested for (i) plateau setting of polarizing voltage, (ii) re-
across the membrane isolating the electrode. In this sponse time for a step change in polarizing voltage, (iii)
arrangement a gradient across the membrane results from noise, and (iv) drift. The lack of horizontal plateau indi-
the zero oxygen concentration at the electrode surface cates a dirty electrode; however, some slope can be tolerated
where oxygen consumption occurs. Fortunately, the rate of in many instances. The response time for a step change in
diffusion is linearly dependent on oxygen concentration or polarizing voltage should be 1 to 1.5 min. Noise may result
partial pressure. The current output is also dependent on from poor connections and grounding, a damaged mem-
the platinum cathode area. brane (folds, holes, KC1 crystals, or drying), or poor contact
A plot of current versus polarizing voltage shows a flat or with the silver electrode, in which case cleaning with am-
nearly constant response in the region of -0.5 to -0.8 V. monia is recommended.
Therefore, there should be a polarizing voltage adjusted to A properly functioning probe should have less than 2%
the center of the constant-current region. The behavior in drift per h. Drift may be caused by atmospheric changes or
response to polarizing voltage is the major distinction of the by contamination with organic material. Thus, proper care
oxygen electrode. and cleaning of the probe are essential. In one study, repro-
ducible results were obtained when the probe and recepta-
17.2.3.1. Effects of Temperature cle were washed with reagents used in the test.
The sensor current is quite sensitive to temperature because
the membrane material permeability has a high temperature 17.2.3.4. Assembly of Electrode Membrane
coefficient (about 3 to 59%/C). In addition, oxygen solu- The demanding aspect of the Clark electrode is attaching
bility varies with temperature. For this reason, good tem- the membrane. Directions and spare membranes are fur-
perature control and adequate equilibration are required. nished by manufacturers of the probe. Care must be taken
in assembly to ensure that the membrane is smoothly
17.2.3.2. Calibration and Calculations stretched over the probe, that there is a true seal at all
Calibration is based on the known oxygen solubility of a so- points, and that no air bubbles are trapped on the electrode
lution under defined conditions (20). Zero oxygen level or side of the membrane. To obtain good wetting in assembly,
bleed current can be defined as the response in the pres- add 3 or 4 drops of Kodak Photoflow per eyedropper bottle
ence of added sodium dithionite. Full response is set at of KC1 used in assembly.
100% with an oxygen-saturated solution. The oxygen con-
centration at the operating temperature is obtained from
oxygen solubility tables in the Handbook of Chemistry and 17.3. CHROMATOGRAPHY
Physics (15) or a similar reference publication. The oxygen Chromatography and related separation techniques depend
content of solutions to be used as working standards can on many kinds of distribution processes, which take place
then be measured. For most work the actual oxygen con- repeatedly in a small local environment (22, 33, 34).
centration of standards is not needed; rather, a reproducible Therefore, the operations of ion-exchange, solvent parti-
concentration is necessary to set the instrument. The mea- tion (including reversed-phase), adsorption-desorption,
surements needed are the relative differences between ex- hydrophobic-interaction, ion pair and hydrogen-bonding,
perimental samples, or assays can be monitored over time in and affinity (association-dissociation) chromatography, as
a continuous manner. For example, if Ringers solution con- well as the operationally related gel permeation (size exclu-
tains 5 ~1 of OJml when air saturated, 3 ml of solution giv- sion) chromatography based on a sieving principle, can all
ing an instrument response of 92% saturation contains 13.8 be carried out with the same laboratory equipment to make
~1 of 0 2 (3 X 5.0 X 0.92). Rates of oxygen consumption or separations based on widely different properties of the
evolution are either calculated by taking measurements at solute molecules.
17. Physical Analysis and Purification Methods w 433

17.3.1. Ion-Exchange Chromatography ber. For weak acids and bases, strength of binding is related
Sorption of solutes is based on ionic charge, although other to the pK of the ionizing group if the pH of operation is in
types of bonding may have some influence on separation. the dissociation range.
Charged groups (-X) on a permeable matrix are able to ex- Exchange of counterions and deionization. Since ion exchang-
change sorbed ions with those in solution. ers are charged with a counterion (for instance, for
-(X)S03Na+ + KCI- -(X)SO3-K+ + Na+C1 Dowex-1 this might be OH-, C1-, or formate), it is pos-
sible to exchange virtually completely an ion in the sol-
Matrix materials include cross-linked, substituted, or de- vent with the charged counterion. For instance, chloride
rivatized resins such as polystyrene or polymethacrylate, as can be exchanged for OH- with Dowex-1 and H+ can
well as derivatized carbohydrate containing natural poly- be exchanged for Na+ with Dowex-50. Therefore, a
mers such as cellulose, dextran, or agarose. major application of exchangers is the deionization of
Exchangers are available with a great variety of func- uncharged solutes such as urea and polyols. This can be
tional groups, support materials, purity, porosity, capacity, accomplished by serial passages of the sample through
size, shape, selectivity, and stability (34-36). It is therefore cationic and anionic exchangers or through a mixed bed.
necessary to consult technical literature from manufacturers However, the mixed bed is more difficult to regenerate
for the specific information needed, for instance, Dow because the cationic and anionic exchangers must first
Chemical Co. for Dowex resins, Rohm and Haas Co. for be separated on the basis of density difference. Alter-
Amberlites, Bio-Rad Laboratories for a wide variety of ex- natively, the spent exchanger is discarded.
changers, and Amersham Biosciences for Sephadexes and Selective removal of divalent and trivalent cations is
Sepharoses. A more extensive listing can be found in refer- accomplished by specialized resins which have EDTA,
ence 35. iminodiacetate, or related groups to chelate the metal
17.3.1 . I . Resin Exchangers ion. Chelex (Bio-Rad), Prosep chelating columns
v
(Millipore, Bedford, MA), and other products perform
Many synthetic resins are available with strong and weak this function.
acidic and basic exchange groups as well as with different
degrees of cross-linking. Lower cross-linking results in Concentration. As a consequence of the high affinity for
higher permeability and water uptake and an ability to in- ions plus the fact that fresh resin is encountered with
teract with larger molecules. For instance, for Bio-Rad movement of the sample through the column, it is pos-
(Hercules, CA) resins of the AG 1 series, 2% cross-linking sible to bind ions from a large volume of dilute solution.
produces an exclusion limit of 2,700 Da whereas 8% cross- The sorbed ions can then be recovered in a small volume
linked resin excludes 1,000-Da molecules. Thus, high cross- of eluant containing an ion of higher affinity or of higher
linking favors sorption of substrates and products of enzyme concentration. In this way, any number of common and
reactions while discriminating against sorption of the en- trace inorganic elements as well as radionuclides and
zyme involved. Ion-exchange wet-volume capacity and trace organic compounds have been concentrated for
selectivity increase with higher cross-linking and also in- analysis.
crease as the particle size decreases. Thus, fine-mesh ex- Purification and fractionation. If two ions have different
changers have increased resolution per bed length simply affinities for an exchanger at a given pH, they can be
because of the increased number of exchange cycles taking separated by finding conditions which sorb and/or elute
place. However, the increased packing of small particles de- one ion but not the other. In such extreme cases, there
creases the flow rate, which in turn requires increased oper- is a frontal elution of each ion. In cases when the affin-
ating pressures. Newer, more rigid polymer resins, ceramic ity is similar, separation is still possible by finding condi-
encapsulation of hydrophilic gels, polyacrylamide-based tions which selectively elute the ion. Development of
gels, or other proprietary designs can help to overcome this the column with eluants then leads to a gradual separa-
flow restriction by making the column bed less compressi- tion. If the column length is sufficient, separation can be
ble. In addition, strong anion, cation, and chelating resin attained. However, diffusion of ions limits the ability of
beads have been cast into membranes with a pore size of increased column length to resolve the components.
0.45 pm. Such membranes, when fabricated into conven- Changing pH or ionic strength elutes sorbed ions of dif-
ient plastic housings, offer the advantages of speed and ease ferent pK values or of increasing affinities.
of use for certain applications. Gradient elution. Elution of sorbed molecules by continuous
Sorption, purification, and fractionation are major ap- change of eluant composition is a useful way to maximize
plications of exchangers. In sorption, a desired ion is selec- the separation potential of a chromatographic system.
tively removed from extraneous material by ion exchange. Rather than making step changes in concentration by
In purification, the ion desired is selectively sorbed and application of different eluants, the change is made to be
eluted while other ions are not sorbed or are retained. continuous within given limits. Devices and procedures
Fractionation is the process of separating several similarly range from the very complicated using expensive micro-
charged ions by introduction of solvent-solute systems processor-controlled pumps that produce complex gradi-
which progressively elute sorbed ions of increasing charge. ents, to the relatively simple linear concentration gradi-
The charge state is often a function of the pK of dissociat- ent involving mixing of two components. In recent years
ing groups and the pH of the environment. the simple linear gradient has been used most frequently.
As a rule of thumb, resolution is related to bed length, For this purpose, two reservoirs are connected so that the
whereas for a given resolution, capacity is related to bed outlet of one is fed into the other. Another outlet of the
cross-section. Since binding of soluble ions is stoichiomet- second reservoir feeds the column. A mixer in the sec-
ric and reversible, the amount of resin needed can be calcu- ond vessel keeps the eluant composition homogeneous.
lated from its published capacity. In general, the strength of When both vessels are open to the atmosphere and each
binding of inorganic ions is directly related to valence vessel has the same cross-sectional area, the eluant com-
(number of charges) and inversely related to atomic num- position will change linearly from the limit in the second
Next Page

434 rn METABOLISM

(mixing) reservoir to the limit in the first. The steepness pH; for the system they include the column dimensions,
of the gradient is determined by the volume in the two flow rate, and elution program. In addition, attention must
reservoirs relative to the column bed. Many varieties of be paid to exchanger packing, sample addition, mainte-
specially designed simple gradient formers are available nance of low sample viscosity, and column design, since
and consist of two reservoirs connected through a valve- these also determine the degree of resolution. Treatment of
containing tube or orifice with an outlet from one side these aspects in adequate detail is beyond the space avail-
going to the column. able here, and hence, only general guides for each are given.
Publications by Amersham Biosciences (22) for exchanger
17.3.1.2. Cellulose, Dextran, and Agarose column media provide more details.
Exchangers In general, uniform spherical dextran beads (e.g.,
For separation of macromolecules, natural polymers have Sephadex and Sepharose) give higher flow rates, although
been modified to contain weak acid or base functional the cellulose derivatives now available also perform well.
groups (22, 27). These matrices also have capacities and Smaller bead or particle sizes give lower flow rates and in-
other properties generally more suitable than resins for frac- creased resolution. Greater porosity increases the capacity
tionation of biomolecules. With dextrans and agaroses, for a given molecule and is particularly important for work
cross-linking and particle size distribution can be con- with macromolecules. The choice of exchanger is based on
trolled, and this minimizes shrinkage and swelling and pro- the ionic forms to be separated as described above. The pH
duces exchangers with high capacity for macromolecules as chosen should be at least 1 pH unit removed from the iso-
well as high and uniform flow rates. Generally, agaroses bet- electric point or the pK of the dissociating group. When
ter maintain bed volume when ionic strength increases and possible, cationic buffers (alkylamines, ammonia, barbital,
when the pH is outside the neutral region. imidazole, and Tris) should be used with anionic exchang-
Since proteins are amphoteric and have a large number ers; anionic buffers (acetate, citrate, glycine, and phos-
of charges, often in clusters, it is possible to treat them as phate) are used with cationic exchangers. The recom-
cations or anions by choosing a pH for ion-exchange sepa- mended pH range for DEAE-Sephadex is 2 to 9, and that
rations that is at least l unit above or below the isoionic for carboxymethyl-Sephadex is 6 to 10. Ionic strength in-
point or the PI value. Cycles of binding and elution make fluences the binding and therefore the capacity for ions. A
use of a pH change which can cause disappearance of ionic relatively high ionic strength of ca. 0.1 M is recommended,
groups, e.g., carboxyl or amino groups on the protein or on but the concentration should be somewhat lower than is
the exchanger. Alternatively, elution is effected by dis- needed to elute the ions desired.
placement with another ion. Column height determines the resolution achieved. When
The DEAE derivative of cellulose, dextran, or agarose is the zone of absorbed ions is 1 to 2 cm, a 20-cm bed height is
a weak anion exchanger (pK of about 9.5) (Table 3). recommended as a starting point. At fixed height, the col-
Hence, in the neutral or lower pH region, the amino group umn capacity is linearly related to the cross-sectional area.
is protonated and binds proteins and peptides if the pH is Elution is effected either by changing the pH to cause
near or above their PI values. The carboxymethyl deriva- the ionic charge to diminish or disappear or by increasing
tives of the same supports are cationic exchangers with a pK the ionic strength to the point at which the buffer ion dis-
of about 3.5. Separations are generally carried out above pH places the ion of interest. These can be done by either step-
4. When the pH of the separation is below the isoelectric wise or gradient change. For anion exchangers the pH is
point of the protein and above pH 4, sorption is efficient. decreased or the ionic strength is increased; for cation ex-
Other cellulose and dextran derivatives used in ion- changers the elution is effected by increasing the pH or the
exchange chromatography are listed in Table 3. ionic strength.
Either the sample total ionic content or column size must
Practical Considerations be determined first, because available capacity is the prop-
Successful use of exchangers rests on the judicious choice of erty of a specified volume of exchanger for each exchanger
a number of parameters. For the exchanger these include type. The ionic composition of the sample ideally should be
the type of charged group, matrix, particle size and shape, that of the starting buffer. When necessary, the ionic com-
and porosity; for the eluant they include the buffer type and position is changed to that desired by means of gel filtration
on Sephadex G-25, dialysis, or dilution. The sample volume
is of minor importance for all elution programs except when
using only the starting buffer for development. The sample
TABLE 3 Chemical groups used in ion-exchange must be added without disturbing the bed and without be-
chromatography coming distributed in the oncoming developing buffer.
Ion exchanger Chemical name PK As with other types of chromatography, it is essential to
use glassware designed for ease of packing, addition of elu-
TMAE Trimethylammoniumethyl >13 ant and sample, application of pressure (if needed), regula-
QAE Die th yl-2 -hydroxypropyl 12 tion of flow rate, prevention from running dry, and low
aminoethyl holdup volume above and below the exchanger bed. It is
also necessary to establish criteria for achieving the objec-
DEAE Diethylaminoethyl 9.5 tive in mind and to analyze the effluent to determine the
TEAE Triethylaminoethyl 9.5 performance of the column.
DMAE Dimethylaminoethyl 8-9
CM Carboxymethyl 3.54 17.3.1.3. Chromatofocusing
P Orthophosphate 3,6 Isoelectric focusing (IEF) by ion exchange, or chromatofo-
cusing, is a specialized type of ion-exchange chromatography
SP Sulfopropyl 2-2.5
in which a pH gradient is used to elute bound proteins in-
Data from various manufacturers specifications. stead of a salt gradient (52). A pH gradient along the length
18
Chemical Analysis
LACY DANIELS. RICHARD S. HANSON. AND JANE A . PHILLIPS

18.1. FRACTIONATION OF MAIN CHEMICAL COMPONENTS ....... 463


18.1.1. Reagents and Materials ............................... 464
.
.2 Radioactive Labeling ................................ 464
. 3. Small Molecules .................................... 464
. 4. Lipids ........................................... 465
. 5. RNA ........................................... 465
. 6. DNA ........................................... 465
. 7. Proteins ......................................... 466
. 8. Applications ...................................... 466
18.2. SMALL-MOLECULE EXTRACTION AND ANALYSIS ............ 466
18.3. CARBOHYDRATES ...................................... 467
18.3.1. Total Carbohydrates by Anthrone Reaction ................ 467
. 2. Total Carbohydrates by Phenol Reaction .................. 468
. 3. D.Glucose, Galactose, Lactose, and Sucrose................ 468
. 4. Glycogen ......................................... 469
. 5. Starch ........................................... 469
. 6. Hexosamines in Purified Peptidoglycans .................. 469
. 7. Hexosamines in Complex Solutions ...................... 470
. 8. Muramic Acid ..................................... 471
. 9. 2-Keto-3-Deoxyoctanoic Acid .......................... 471
.l 0. Heptoses ......................................... 472
. .
11 Polysaccharides and Complex Carbohydrates ............... 472
18.4. LIPIDS ................................................ 473
18.4.1. Crude Total Lipids .................................. 473
.
.2 Purified Total Lipids ................................. 474
.3. Lipid Classes ......................................
............................ 474
..4. Phospholipid Fractionation
5. Phosphate in Phospholipids ............................
475
475
. 6. Phospholipid Identification ............................ 476
. 7. Archaeal Lipids .................................... 477
18.4.7.1. Extraction................................. 477
. .................................
2. Separation 477
. 3. Analysis .................................. 477
18.4.8. Fatty Acids ....................................... 478
. 9. Poly.P-Hydroxybutyrate .............................. 478
. 10. Poly-P-Hydroxyalkanoate Polymers ...................... 479
18.5. CELL WALL POLYMERS ................................... 480
18.5.1. Peptidoglycan ..................................... 481
. 2. Total Lipopolysaccharides ............................. 482
. .........
3. Lipid A and Polysaccharide from Lipopolysaccharides 482
. 4. Archaeal Cell Wall Components ........................ 483
18.6. NUCLEIC ACIDS ........................................ 483
18.6.1. Total DNA by Diphenylamine Reaction ................... 483
. 2. Total DNA by Fluorescence Spectroscopy ................. 484
........
18.6.2.1. Bisbenzimide (Hoechst 33258 Dye) Binding 484
. .....................
2. Ethidium Bromide Binding 485
18.6.3. Total RNA by Orcinol Reaction ........................ 485
. 4. DNA and RNA by Other Methods ...................... 485
18.7. NITROGEN COMPOUNDS ................................ 486
18.7.1. Nitrite ........................................... 486
.
.2 Nitrate .......................................... 489
18.7.2.1. Sampling.................................. 489
. .....................
2. Zinc Reduction Procedure 489
. ..................
3. Cadmium Reduction Procedure 489
18.7.3. Ammonium by Indophenol Blue Reaction ................. 490
. 4. Ammonium by Nessler Reaction ........................ 490

462
18. Chemical Analysis 463

.5.
The Challenge of Protein Analysis ..................... 491
.6.
Protein by the Lowry Assay (Folin Reaction) .............. 491
.7.
Protein by Biuret Reaction ........................... 492
.8.
Protein by Coomassie Blue Reaction (Bradford Assay) ....... 492
.9.
Protein by Bicinchoninic Acid-Copper Reaction
(BCA Assay) ..................................... 493
.10. Protein by UV Absorption ........................... 493
.1 1. Total Nitrogen (Kjeldahl Digestion) ..................... 494
.1 2. Molecular Nitrogen ................................ 494
.13. Specific Nitrogenous Compounds ...................... 494
.14. Total Amino Acids ................................. 494
.l5. meso-DiaminopimelicAcid ........................... 495
.16. Dipicolinic Acid ................................... 495
18.8. ORGANIC ACIDS AND ALCOHOLS ......................... 496
18.9. MINERALS ............................................. 496
18.10. HAZARDS AND PRECAUTIONS ........................... 496
18.11. REFERENCES ........................................... 498
18.11.1. Fractionation and Radioactivity ........................ 498
.
2. Small Molecules ................................... 498
.
3. Carbohydrates .................................... 498
18.1 1.3.1. General References........................ 498
. .........................
2. Specific References 498
18.11.4. Lipids .......................................... 500
18.11.4.1. General References ........................ 500
. .........................
2. Specific References 500
18.11.5. Cell Wall Polymers ................................. 501
18.11.5.1. General References ........................ 501
. .........................
2. Specific References 501
18.11.6. Nucleic Acids .................................... 501
18.11.6.1. General References ........................ 501
. .........................
2. Specific References 502
18.11.7. Nitrogen Components .............................. 502
18.11.7.1. General References ........................ 502
. .........................
2. Specific References 502
18.11.8. Minerals ........................................ 503
18.1 1.8.1. General References ........................ 503
. .........................
2. Specific References 503

Analysis to estimate amounts of the major and minor com- how rapidly such components are synthesized .The incorpo-
ponents of microorganisms and their metabolites is often re- ration of radioactively labeled compounds into bacterial
quired to address certain research questions. In other in- cells is used as a means of estimating the rates and amounts
stances. a microbial component may need to be purified to of synthesis as well as the distribution of the main small-
study its properties. e.g., a labeling pattern to ascertain a molecule and macromolecule fractions of the cells (2).
metabolic pathway. The methods selected for this chapter After the cells have been incubated with the labeled sub-
are most commonly used in research relating to microbial strate and the various molecular components have been
components and metabolism. but most are also suited to ad- fractionated. the radioactivity in each fraction can be de-
vanced undergraduate and graduate teaching laboratories. termined.
Many other methods and more detailed descriptions of Macromolecules are precipitated by cold dilute solu-
those selected are presented in publications listed at the end tions of trichloroacetic acid. The (denatured) macromole-
of the chapter. Techniques that require costly instrumenta- cules are separated from the small molecules by centrifuga-
tion or significant training are not described in detail. tion . The supernatant fraction of small molecules contains
Specialized methods (which vary with the nature of the the pooled metabolites of sugars. sugar derivatives. organic
samples. equipment. and expertise) are treated by providing acids. amino acids. nucleotides. and coenzymes; oligonu-
references to other sources of information rather than by cleotides and basic proteins with fewer than 20 nucleotide
describing experimental details. As with virtually all labo- or amino acid residues are also contained in this fraction
ratory methods. some steps in the described methods re- (2) . (Note that some coenzymes are destroyed by acid
quire the use of dangerous chemicals or equipment. and ap- extraction; see section 18.2 for alternate methods of co-
propriate attention to safety issues is essential. enzyme measurements.) The precipitate fraction of
macromolecules is sequentially extracted with organic sol-
vents for lipids. alkali for RNAs. and hot trichloroacetic
18.1. FRACTIONATION OF M A I N CHEMICAL acid for DNAs; the precipitate remaining after these ex-
COMPONENTS tractions contains the cell proteins and peptidoglycan. A
It is sometimes useful to know the amounts of major bacte- flow diagram of this fractionation procedure is shown in
rial cell components present at a given time and to know Fig. 1.
464 METABOLISM

MICROBIAL CELLS
f . .
Cold trichloroacetic acid

SMALL MOLECULES MACROM 0LECULES


f

LIPIDS
a RNA + DNA
f
+ PROTEINS
NaOH

RNA DNA + PROTEIN


f
Hot trichloroacetic acid

DNA PROTEINS

FIGURE 1 Flow diagram of fractionation procedure.

No single procedure gives complete separation of all of Scintillation counter


the chemical components and total recovery of a given type Vortex mixer
of macromolecule in a single fraction. The procedures de-
scribed by Kennel1 (1) are presented below. They are mod- 18.1.2. Radioactive labeling
ifications of the methods of Roberts et al. (2). (CAUTION: See section 18.10.) The use of radioactive iso-
topes can be hazardous and is thus strictly regulated. All
18.1 .I. Reagents and Materials personnel involved in their use must be familiar with safety
Saline solution: 0.85% (wtlvol) NaCl in distilled water issues and regulations regarding their use (see also chapter
Trichloroacetic acid solutions (CAUTION: See section 17.4.13).
18.10): 50, 20, 10, and 5% (wtlvol) trichloroacetic acid The culture volume to be used will vary with the organ-
in distilled water ism and growth conditions. For example, 30 ml of a mid-
Ethanol (CAUTION: See section 18.10): 70% (vol/vol) in exponential-phase culture of Escherichia coli is adequate to
distilled water provide the 10 mg of cells required.
If the purpose of the experiment is to determine cell
Diethyl ether (CAUTION: See section 18.10) composition, a uniformly labeled substrate such as [14C]glu-
Sodium hydroxide (0.5 N ) (CAUTION: See section cose (5 pCi/30 ml of a culture containing 1% glucose as the
18.10). Dissolve 2.0 g in 100 ml of distilled water. only carbon source) is suitable (1 FCi = 37,000 Bq). The
Bovine serum albumin (BSA). Dissolve 0.1 g in 10 ml of incorporation of an amino acid into protein can be mea-
distilled water. Store at 4C. sured by adding 2 to 5 pCi of a labeled amino acid to 30 ml
Scintillation fluid (available from a variety of vendors) of a culture growing on a mineral salts medium plus glucose.
Glassware: 20-ml Pyrex glass beakers, 10-ml Erlenmeyer Ideally, an auxotrophic mutant that is unable to synthesize
flasks, 25-ml volumetric flasks, capped 15- and 30-ml the amino acid should be used when incorporation into pro-
centrifuge tubes tein is measured. In this case, 10 pg of the unlabeled amino
acid per ml and 2 to 5 pCi of the labeled amino acid should
Filters: fiberglass or synthetic membrane filters (pore size, be added to the medium. It is important to use a nonmeta-
0.45 pm), usually 25 mm in diameter. The filters must bolizable amino acid as a labeled protein precursor. Leucine,
be acid and solvent resistant. Pyrex filter holders with lysine, phenylalanine, and tyrosine are not significantly me-
fritted-glass filter supports. tabolized by most bacteria. When labeled thymidine is used
Vacuum filtering flask: 125-ml flask and test tubes cut to fit as a precursor of DNA, and when uracil or uridine is used as
below the filter holder inside the flask. The test tubes are a precursor of RNA, auxotrophs should also be used to ob-
used to collect filtrates. tain incorporation of a larger fraction of the label added to
Heating lamp: 250-W infrared heat lamp, and a suitable re- the medium. The cultures (inoculated with healthy, mid-
flector for sample drying; alternatively, an oven heated exponential-phase cultures) should be harvested during ex-
at 50 to 80C is suitable but should not be used with sig- ponential growth when cell composition studies are per-
nificant amounts (> 1 ml) of ether. formed. The cell composition changes during the lag and
Ice-water bath stationary phases of growth.
Water bath, adjustable to 80C
18.1.3. Small Molecules
Refrigerated centrifuge, capable of achieving 10,000 X g,
with a centrifuge head that holds 30-ml centrifuge 1. Cool the culture rapidly by pouring it over ice.
tubes Centrifuge the radioactively labeled cells (approximately
18. Chemical Analysis 465

10 mg [dry weight]) from the growth medium at 5,000X g a scintillation vial under a stream of filtered air or nitrogen
for 10 min at 5"C, and wash the sedimented cells twice by at 40C. Gas filtration (to remove oil and particulates) can
centrifugation with ice-cold (5C) 0.85% saline (one-fifth be accomplished by passing compressed gas through glass
of the culture volume). wool or an in-line 0.45-km-pore-size bacterial filter.
2. Suspend the cells with 10 ml of distilled water in the 6. Determine the radioactivity of the dried lipid sample
centrifuge tube. Place the suspension in an ice bath until by use of a suitable scintillation cocktail (see chapter 17.4).
cold, and add 10 ml of ice-cold 20% trichloroacetic acid.
Cap the centrifuge tube, and mix the contents by inversion. 18.1.5. RNA
Place the tube in the ice bath for 5 min to allow complete 1. Separate RNA from the precipitated fraction of
precipitation. macromolecules, with the lipids now removed. Remove the
3. Centrifuge (5,000 to 10,000 X g for 15 min) at 0 to filter containing the precipitate from the filter holder (step
4C. Carefully decant the supernatant fraction, without dis- 4 above), and place upside down in a 20-ml beaker. Allow
turbing the precipitate, into a 25-ml volumetric flask. the filter to air dry in a hood (about 10 min).
4. Add 4 ml of ice-cold 10% trichloroacetic acid to the 2. Add 2 ml of 0.5N NaOH (warmed to 37C) to the
precipitate, suspend it, and centrifuge again. Decant the su- beaker, and incubate with occasional shaking at 37C for
pernatant fraction into that obtained after the first cen- precisely 40 min. Make sure the filter is completely covered
trifugation. Adjust the volume of the combined supernatant with the NaOH solution. Longer incubation causes hydrol-
fractions to 25 ml with distilled water. ysis of some proteins (1,3).
5. Determine the radioactivity of a sample of the com- 3. Transfer the beaker to an ice bath, and cool rapidly.
bined supernatant fraction containing the small molecules Add 0.6 ml of ice-cold 50% trichloroacetic acid, mix, and
(metabolite pools) directly in a solution used for deter- leave the beaker in the ice bath for about 30 min.
mining radioactivity in aqueous samples, as described else- 4. Transfer the liquid in the beaker to a new filter held
where in this book (see chapter 17.4). Internal or external in a filter holder, apply a vacuum, and collect the acid-
standards should be used to correct for quenching by soluble products in a test tube. Rinse the beaker and original
trichloroacetic acid. Alternatively, trichloroacetic acid can filter with 3 ml of ice-cold 5% trichloroacetic acid, transfer
be removed by three extractions with equal volumes of the liquid to the same filter, and collect the filtrate in the
ether. However, ether extraction will remove organic acids same test tube. Adjust the volume of the combined filtrates
from the small-molecule fraction. Samples of the ether- to 15 ml in a volumetric flask with distilled water. The fil-
extracted portion can be counted separately in a counting trate contains about 95% of the ribonucleotides in RNA.
solution used for aqueous solvents or can be dried in vials 5. Determine the radioactivity of the hydrolyzed RNA
with the assistance of a heat lamp. Large volumes (>1 ml) fraction in the same way as for the small-molecule fraction
of ether solutions should not be dried in an oven because of (see above and chapter 17.4).
the explosion hazard, but it is possible to evaporate most of
the ether at room temperature, preferably in a fume hood, 18.1.6. D N A
and then place the container in the oven. After drying, the 1. After removing lipids and RNA from the macromo-
residue can be counted in a suitable scintillation cocktail lecular fraction, separate the DNA from the precipitated
(see chapter 17.4 and section 18.2). fraction of macromolecules by treatment with hot trichlo-
roacetic acid. Use a duplicate sample from the suspended
18.1.4. lipids sediment in cold trichloroacetic acid, as described in the
1. Separate lipids from the precipitated fraction of first steps of the lipid fractionation (section 18.1.4). Repeat
macromolecules in the centrifuge tube (see above). Suspend steps 1 through 4 for removal of lipids. After washing the
the sediment in 10 ml of ice-cold 10% trichloroacetic acid filter with ethanol and ethanol-ether to remove lipids, place
with the aid of a vortex mixer. (CAUTION: Cap the tube to the filter in a beaker, add 2 ml of 0.5N NaOH, mix, and in-
avoid trichloroacetic acid and radioisotope aerosols.) cubate for 90 min at 37C. The incubation time with
2. Filter 1 ml of the homogeneous suspension, and wash NaOH is increased to give total hydrolysis of RNA. DNA is
the filter twice with 2 ml of ice-cold 10% trichloroacetic present in smaller amounts than RNA, and traces of unhy-
acid. Wash the filter with 3 ml of ice-cold 70% ethanol to re- drolyzed RNA can significantly increase estimates of ra-
move residual trichloroacetic acid, which may cause solubi- dioactivity in DNA. The solubilization of some proteins
lization of some protein in the next step. Discard the filtrate. will not affect the estimations of DNA.
3. Place the filter holder (with the filter and suction 2. Cool the sample on ice, add 0.6 ml of cold 50%
flask) and a test tube for collecting the filtrate in an incu- (wtlvol) trichloroacetic acid, and keep it on ice for 30 min;
bator at 45"C, and allow the assembly to warm to tempera- then, pour the liquid in the beaker onto a new filter that has
ture. Then, remove the filter flask from the incubator, been presoaked in cold 5% trichloroacetic acid. Apply a
quickly add 10 ml of 70% ethanol (warmed in a water bath vacuum to facilitate filtration. Wash the original filter twice
to 45"C), and allow the solvent to pass slowly through the with 3 ml of ice-cold 5% trichloroacetic acid, and discard
filter. Collect the filtrate in a test tube. The filtration the filtrate.
should take approximately 10 min to effect the extraction. 3. Place the two filters in a 10-ml Erlenmeyer flask, add 3
4. Wash the filter in a hood with 2- to 5-ml volumes of ml of 5% trichloroacetic acid, and heat for 30 min in a water
ethanol-diethyl ether ( l : l , vol/vol) warmed to 45C in a bath at 80C; this step hydrolyzes purines from the DNA.
water bath by allowing the solvents to pass slowly through Make sure the filters are entirely immersed in the liquid.
the filter (10 min). Collect the filtrate in a test tube, and 4. Add 0.1 ml of a solution (1.0 mg/ml) of BSA, mix rap-
combine it with the 45C ethanol wash. Retain the filter idly, cool the flask for 30 min in an ice bath, and filter the
containing the residual precipitated fraction of macromole- entire contents of the Erlenmeyer flask through a new filter.
cules for further fractionation. The BSA ensures precipitation of minute amounts of mac-
5. Adjust the volume of the combined filtrates (which romolecules that may otherwise remain in suspension. Use
represents the lipid fraction) to 25 ml, and dry a sample in a slight vacuum to increase the filtration rate. Collect the
466 METABOLISM

filtrate in a test tube placed inside the vacuum flask. Wash are not accounted for. Products of hydrolysis of teichoic
the Erlenmeyer flask and filters twice with 3 ml of cold 5% acids and polyphosphates will appear in the RNA fraction,
trichloroacetic acid. Collect the filtrates in the same test and peptidoglycan will be precipitated with protein.
tube. Adjust the volume of the combined filtrates to 15 ml Poly-P-hydroxybutyrate, if present, can be extracted
with water in a volumetric flask. Discard the filters. with chloroform-methanol (2:1, vol/vol) at 60C after the
5. Determine the radioactivity of the hydrolyzed DNA ether-ethanol washes (see also section 18.4.9). If this sol-
fraction in the same way as for the small-molecule fraction vent is used, care should be taken to use filters that are sta-
(see above and chapter 17.4). ble in methanol and chloroform. (CAUTION: The extrac-
tion should be performed in a hood [see section 18.101.)
18.1.7. Proteins Polysaccharides, if present, are partially extracted by
1. Separate proteins from the precipitated fraction of cold trichloroacetic acid and can significantly affect esti-
macromolecules. Use a duplicate sample from the sus- mates of the small molecules. When present in large
pended sediment in cold trichloroacetic acid, as described amounts, polysaccharides present a variety of troublesome
in the first step of the lipid fractionation (section 18.1.4). problems if nonspecific precursors of macromolecules such
Transfer 1 ml into a test tube (15 by 100 mm). Dilute with as radioactive glucose are used. When these problems occur,
1 ml of water to give a trichloroacetic acid concentration of these procedures must be modified ( 3 ) .
5% (wtlvol). Some bacterial cells leak part of their pools of small mol-
2. Heat the diluted sample for 30 min at 80C to hy- ecules when chilled during centrifugation and washing.
drolyze both RNA and DNA. Some cells have high activities of nucleases or proteases
3. Cool the hydrolyzed sample on ice for 30 min to pre- that may be activated during harvesting and washing. If
cipitate proteins. Collect the precipitate on a filter, and dis- cells are frozen and then thawed, even more small mole-
card the filtrate. Wash the test tube twice with 2 ml of ice- cules and enzymes will leak.
cold 10% trichloroacetic acid, and apply both washes to a Significant losses of macromolecules occur when smaller
filter; wash the filter once with 3 ml of ice-cold 70% amounts of material than indicated are used in the first
ethanol to remove residual trichloroacetic acid that would steps. It is possible to add unlabeled bacterial cells as a car-
otherwise solubilize some protein in the next step. rier to facilitate the precipitation of macromolecules.
4. Wash the filter twice with 5 ml of 70% ethanol Precipitation of DNA and protein can also be facilitated by
(45"C), twice with 5.0 ml of ethanol-diethyl ether (1:l) at addition of serum albumin to the solutions.
45"C, and once with 5 ml of diethyl ether. Each organism potentially presents a unique problem in
5. Air dry the filter, place it in a scintillation vial, and fractionation. Although the procedures described above
dry it under a heat lamp. have been widely used, the data obtained should be inter-
6. Determine the radioactivity of the dried protein by preted with proper regard for the problems of obtaining total
use of a suitable scintillation cocktail (see chapter 17.4). separation of the small molecules and macromolecules (3).
Sequential methods for treating a single sample have
been described ( 2 , 3 ) . However, they are likely to result in
18.1.8. Applications greater cross-contamination of fractions than the procedure
The techniques described above are usually applied to the described here.
fractionation of bacterial cells labeled with a medium com-
ponent that is nonspecifically incorporated into all of the
cell molecules. When bacterial cells are incubated in the 18.2. SMALL-MOLECULE EXTRACTION
presence of a specific precursor of a macromolecule to mea- A N D ANALYSIS
sure its rate of synthesis, it is important to determine that the In some cases the small molecules of interest are unstable to
macromolecule of interest contains all of the radioactivity. acid treatment or require further fractionation or analysis
This can be determined by fractionating the cells and esti- for quantitation. Also, coenzyme analysis may be needed
mating the radioactivity of each fraction by the techniques without the need for any of the other analyses described
described above. If all the radioactivity occurs in the fraction above for macromolecules. For example, coenzymes could
of interest, subsequent experiments can be simplified. For be extracted under quite different conditions from those de-
example, if radioactive thymidine is used to measure the scribed above, from either labeled or unlabeled cells, and
amount of DNA synthesis and preliminary experiments in- analyzed further. Detailed extraction procedures for a wide
dicate that it is not found in other fractions, the cells in their range of coenzymes has been provided for a wide range of
culture medium can be cooled and treated with ice-cold coenzymes in Methods in Enzymology (4, 11-14). In some
10% trichloroacetic acid. The precipitate containing the cases, anaerobic methods are required to maintain the sta-
DNA can then be collected on filters, washed with cold bility of oxygen-sensitive components, e.g., by conducting
ethanol, and dried, and the radioactivity can be counted. extraction under nitrogen or argon gas as is done for
This procedure is suitable for measuring the amount of DNA tetrahydromethanopterin from methanogens (7). In other
synthesis. The same technique can be applied to measure the cases, some coenzymes (e.g., BIZ)are sensitive to light, and
amount of protein or RNA synthesis if the precursor appears procedures should be conducted under low-light conditions.
only in the one fraction. Other factors must be considered in When a small molecule is stable to heat, boiling or briefly
calculating the rates of synthesis. A primary concern is the autoclaving cells is generally one of the easiest and most ef-
dilution of exogenously added substrates by unlabeled pre- fective extraction methods. One type of more gentle ex-
cursors in the cell pools. In kinetic experiments, it is neces- traction is the use of 70% aqueous ethanol or 50% aqueous
sary to determine the rate of saturation of the pool by ra- acetone at 4C to resuspend the original cell pellet (15, 16).
dioactive precursors. (CAUTION: See section 18.10.) The suspension is stirred
Although the methods described above provide a means at 4C for 5 to 15 h and centrifuged, and the supernatant is
of obtaining an estimate of the distribution of carbon from a saved; the pellet is then extracted one more time with fresh
14C-labeledcompound in bacteria, several cell components solvent. The combined supernatants contain virtually all
18. Chemical Analysis w 467

the water-soluble, free coenzymes. Extraction and analysis acids, etc.). Some macromolecules contain both carbohy-
of coenzyme F420 illustrates the importance of understand- drate and noncarbohydrate components (lipopolysaccha-
ing the physiology of the organism being examined for rides, peptidoglycans, teichoic acids, lipoteichoic acids, tei-
coenzyme content. F420 is found in methanogenic archaea churonic acids, nucleic acids, glycoproteins, etc.).
and some aerobic actinomycetes but is absent from most Colorimetric methods based on the Molisch test for car-
other microbes (5,6,8, 10). F420 is chemically stable to oxy- bohydrates are useful for estimating the amount of simple
gen, i.e., pure F420 is unaffected by exposure to 0 2 . sugars and their polymers (17, 29, 30, 46). Specific assays
However, if methanogens are extracted under conditions for pentoses in nucleic acids (ribose and deoxyribose) are
that allow enzyme activity to proceed in the presence of air described below in the sections that deal with nucleic acids
while cells are being heated relatively slowly to lyse them to (sections 18.6.1 and 18.6.2). Specific enzymatic assay ex-
release F420, then an oxygen-dependent enzyme called F390 amples are given for glucose, galactose, lactose, and sucrose.
synthase will convert F420 into F390,resulting in significant More complex descriptions of analytical methods for the
loss of F420 (9, 15). In contrast, aerobic actinomycetes do analysis of carbohydrates have been presented (17-20, 28,
not have this enzyme, and extracting by heating in the air 33, 35, 38, 41). Details of automated enzymatic methods
causes no loss of F420 (10). can be obtained from the suppliers of enzyme kits or from
Some coenzymes are not very water soluble, and thus, in instrument manufacturers.
these cases extraction with a suitable nonaqueous solvent is
required, e.g., menaquinones can be extracted with chloro- 18.3.1. Total Carbohydrates by Anthrone Reaction
form-methanol, acetone:methanol, or ethanol ( 14). The most convenient assays for total carbohydrates involve
After extraction, analysis must be conducted to measure heating media, cells, or isolated carbohydrates with sulfuric
the coenzyme content. In some cases, the extract (diluted acid to hydrolyze the polysaccharides and dehydrate the
or evaporated to remove possibly inhibitory solvents in monosaccharides to form furfural from pentoses and hy-
some cases) can be examined by using bioassay techniques. droxymethylfurfural from hexoses. The solutions of furfural
However, more commonly the supernatant is fractionated and hydroxymethylfurfural are then treated with a reagent
by chromatography on suitable columns, e.g., DEAE- (an aromatic compound or phenol) to produce a colored
Sephadex, or C18reverse-phase high-pressure liquid chro- compound, which is measured in a spectrophotometer.
matography (HPLC). Coenzyme elution can be monitored Many variations of this Molisch test have been described
by absorbance at an appropriate UV or visible wavelength (17, 29, 30, 46). Pentoses, hexoses, heptoses, and their de-
or by fluorescence. In some cases, if the material is suffi- rivatives (except the amino sugars) yield colored products,
ciently pure after one chromatographic step, the coenzyme but trioses and tetroses do not.
can be quantitated by measuring the absorbance or the flu- No method accurately measures all the carbohydrate in
orescence of the pooled column fractions. Alternatively, bacteria, because different sugars give different color inten-
the area under the HPLC chromatographic profile can be sities at any selected wavelength (19). The anthrone and
measured; care should be taken to ensure that the proper the phenol methods (described below) are presented be-
extinction coefficients are used for the pH and solvent in cause of their simplicity, relative insensitivity to interfer-
which the material is dissolved as it passes through the de- ence by other cell components, and similar response to most
tector, since organic solvents and pH can greatly affect the common hexoses in cells, thus providing a reliable index of
spectra of some small molecules (see chapter 17.1.1.4). A total carbohydrates.
variety of coenzymes can be quantitated at one time by Either the anthrone or the phenol procedure is suitable
HPLC methods if retention times are known and if there for estimation of total carbohydrate in bacteria that contain
are no interfering peaks; the use of a rapid spectral scan of 10%or more hexose polymers. The pentoses in nucleic acid
HPLC peaks while the material passes through the detector interfere with these essays when the hexose content is low.
is an excellent tool to examine the purity of peaks and thus Two notable disadvantages of these methods are that indi-
confirm the validity of the quantitation and the purity of vidual sugars are not identified and that working with con-
the material. Examples of HPLC methods for quantitation centrated sulfuric acid and phenol is dangerous.
of ascorbic acid, thiamine, biotin, NADH, flavins, de-
azaflavins such as F420, folates, and methanopterin species Reagents and Equipment
have been described (8).
If labeled material has been used to feed the cells prior to Saline solution: 0.85% (wtlvol) NaCl in distilled water
coenzyme analysis, the specific radioactivity of the pure Stock glucose solutions. Dissolve 50 mg of glucose in 50 ml
coenzymes can be determined by comparing the molar of 0.15% (wtlvol) benzoic acid, which is added as a pre-
quantity of coenzyme and the radioactivity measured by servative. Store at 5C. This solution is stable for sev-
scintillation counting. This approach may be useful in the eral months. Dilute 1:lO in distilled water just before
study of precursors of coenzyme production. use to give a solution containing 100 kg/ml. Prepare
In addition to coenzymes, other small molecules of in- standard solutions (10 to 100 pg/ml) from the diluted
terest, including amino acids and sugars, can be extracted solution.
by this general method and analyzed as described below Sulfuric acid solution (75%, vol/vol). Add 150 ml of
(sections 18.3 and 18.7). reagent-grade concentrated sulfuric acid to 50 ml of dis-
tilled water (CAUTION: Sulfuric acid is very dangerous
[see section 18.101). Also, avoid contact of paper with
18.3. CARBOHYDRATES either of the sulfuric acid-containing reagents, since cel-
A large number of different carbohydrates can be found in lulose will be hydrolyzed to sugars, resulting in a high
bacteria and archaea: free sugars, sugar derivatives, simple background; one way in which this contamination can
polysaccharides (polymers of a single sugar or sugar deriva- occur is to wipe acid off a pipette tip with a paper towel
tive, such as glycogen), and complex polysaccharides (poly- and then to continue using the pipette. Also, the con-
mers of more than one different sugar, amino sugars, uronic centrated sulfuric acid should be of good quality and
468 METABOLISM

free of excess water; excess water in the assay causes tur- anthrone reaction, for a discussion of the disadvantages of
bidity. these two assays, and see below for more-specific assays.)
Anthrone reagent. Add 100 mg of anthrone to 2.5 ml of ab-
solute ethanol, and make up to 50 ml with 75% H2S04 18.3.3. D-Glucose, Galactose, lactose, and Sucrose
(CAUTION: See section 18.10). Stir until dissolved. A wide variety of monosaccharides are liberated by the hy-
Prepare fresh daily, and store in a refrigerator. drolysis of simple and complex polysaccharides in bacterial
Thick-walled Pyrex boiling tubes cells. Because the polymers exist as mixtures, except in ho-
mopolymers such as glycogen, the estimation of specific
Glass marbles to fit boiling tubes (to avoid water dropping sugars sometimes presents difficult analytical problems that
in, and to avoid evaporation; a drop of water can cause can be best solved with HPLC, ion chromatography, or gas-
turbidity) liquid chromatography procedures (18, 24, 33, 35, 52, 55,
Spectrophotometer or colorimeter 62). However, some sugars can be estimated by specific col-
Glass cuvettes or tubes orimetric reactions or enzymatic methods (17, 32, 47, 57,
Gogg1es 58); two good examples are the hexoses glucose and galac-
tose, for which a variety of enzymatic methods are available.
Procedure A specific example is the use of glucose oxidase to assay glu-
Wash samples of bacterial cells free from medium compo- cose, but a very similar system works for galactose. Glucose
nents by centrifugation at 5,000 X g for 10 min. Resuspend oxidase catalyzes the following reaction:
in saline solution, and centrifuge again. Resuspend the sed-
imented cells in distilled water, and pipette samples (0.06 to
P-D-glucose + o2-+ D-glucono-6-lactone + H2O2
0.6 ml) of the cell suspension into the boiling tubes. The hydrogen peroxide, in the presence of peroxidase and a
(Alternatively, culture medium can be assayed after its sep- reduced chromogen, causes the production of a brown or
aration from cells by centrifugation.) Adjust the volume of blue oxidized chromogen that can be measured spectropho-
all samples to 0.6 ml with distilled water. Prepare a reagent tometrically. Different chromogenic compounds are used in
blank of distilled water (0.6 ml) and a standard curve by commercially available kits, which also contain the needed
adding 0.6 ml of standard solutions containing 10 to 100 pg enzymes and sometimes glucose standards. The glucono-6-
of glucose per ml. Chill the tubes and the anthrone reagent lactone can be converted into gluconic acid under basic
in an ice-water bath until cold. Add 3.0 ml of cold anthrone conditions, but this is not needed for the assay. The glucose
reagent, mix rapidly by swirling the tubes in ice water, and oxidase assay is specific, sensitive, and easily performed.
continue mixing in the ice-water bath for 5 min. Then, Only glucose and 2-deoxyglucose are known to produce the
transfer the tubes to a bath of boiling water for precisely 10 colored products.
min. Return the tubes to the ice water. Measure the Galactose oxidase assay kits are also available from com-
A625 of each solution. Determine the concentration of sug- mercial sources. The kits contain galactose oxidase, chro-
ars in the samples from a standard curve prepared by plot- mogen, and standards. Since galactose oxidase oxidizes the
ting the absorbances of the standards versus the concentra- C-6-hydroxymethyl group of galactose to produce galact-
tion of glucose. uronic acid, the enzyme will also oxidize some galactosides.
Commercial kits and/or the constituent enzymes are
18.3.2. Total Carbohydrates by Phenol Reaction available from many sources, but a few examples are Sigma
Chemical Co., St. Louis, MO; Worthington Biochemical
Reagents and Equipment
Co., Freehold, NJ; and Boehringer Mannheim Biochem-
Phenol reagent. Dissolve 5 g of reagent-grade phenol in 100 icals, Indianapolis, IN.
ml of distilled water (CAUTION: See section 18.10) A variation on the glucose oxidase assays has been de-
Reagent-grade concentrated sulfuric acid (CAUTION: See veloped for use in an automated system, in which hydrogen
section 18.10) peroxide is detected electrochemically (44, 49, 50, 63;
Glucose standard (see anthrone procedure above) Yellow Springs Instrument Co. [YSI], Immobilized Enzyme
Biosensor, 2001 [http://www.ysi.com/lifesciences.htm]). This
Thick-walled Pyrex boiling tubes (15 by 2.5 cm) method is particularly attractive in classroom settings,
Spectrophotometer or colorimeter where a large number of students have to do assays in a lim-
Glass cuvettes or tubes ited period and with a higher degree of safety than is possi-
Goggles ble with the anthrone and phenol assays; it is also cost-
effective in an industrial setting, where many assays are
Procedure done routinely.
Pipette the samples to be analyzed into thick-walled Pyrex A convenient method for glucose analysis of some types
tubes. Adjust the volume of all samples to 0.5 ml with dis- of samples (e.g., liquid medium containing growing mi-
tilled water. Prepare a reagent blank by adding 0.5 ml of dis- crobes) is the use of strips that are sold for glucose determi-
tilled water, and prepare a standard curve by adding 0.5 ml nation in urine, e.g., for diabetic patients. These are readily
of solutions containing 10 to 100 pg of glucose per ml. Add available from drug stores, are inexpensive, and require no
0.5 ml of the phenol reagent; mix rapidly and thoroughly. additional equipment, making them ideal for many class-
Add 2.5 ml of concentrated sulfuric acid, mix rapidly, and room and research settings. Proper dilution of samples to an
let stand for 10 min. (CAUTION: Addition of sulfuric acid appropriate glucose concentration and appropriate controls
to aqueous solutions causes rapid heating and occasionally with no added glucose are required.
boiling; see also section 18.10.) Place the tubes in a water Several other sugars can also be assayed by a system very
bath at 25C for 15 min. Read the A488of each tube against similar to those above with an enzyme that generates hy-
the blank prepared without glucose. Determine the concen- drogen peroxide, sometimes assisted by other immobilized
tration of sugars in the sample from a standard curve pre- enzymes. Lactose, galactose, and sucrose are three examples
pared by plotting the absorbances of standards versus the of additional sugars that can be determined with the auto-
concentration of glucose. (See also section 18.3.1, on the mated analyzer from Yellow Springs Instrument Co. (YSI),
18. Chemical Analysis 469

Yellow Springs, OH (see above). The same system can also Glycogen, like other polysaccharides, is resistant to hydrol-
estimate ethanol, methanol, L-glutamate, L-glutamine, and ysis by alkali, but it is readily soluble in water and insoluble
L-lactate by using an enzymatic approach. in ethanol. The quantitative isolation of the polymer takes
A n alternative to the automated glucose oxidase ap- advantage of these properties. After isolation, and after hy-
proach is an automated glucose dehydrogenase reaction, as drolysis for 6 h in 4 N HC1 at 100C in a sealed ampoule,
used in the HemoCue Betaglucose analyzer (61). the amount of glucose can be estimated by the anthrone or
A n example of the use of glucose oxidase in a manual phenol reaction or by enzymatic assay.
spectroscopic system is as follows. The assay for galactose
would be very similar. Reagents and Equipment
Absolute ethanol (CAUTION: See section 18.10)
Reagents Potassium hydroxide (30%, wt/vol) in distilled water
Glucose stock solution. Dissolve 300 mg of D-glucose in 100 (CAUTION: See section 18.10)
ml of 0.15% (wtlvol) benzoic acid. Store in a refrigera- Refrigerated centrifuge, capable of achieving 10,000 X g
tor. Dilute 1:lO with distilled water, and adjust to pH 7.0
before use. 30-ml glass centrifuge tubes
Commercial glucose oxidase-peroxidase preparation. Vacuum desiccator or lyophilizer
Prepare as specified by the manufacturer. Procedure
4 M HC1 (CAUTION: See section 18.10) Harvest bacterial cells by centrifugation at 5,000 to 10,000
Chromogen: available as part of the commercial kits. X g for 15 min, resuspend the cells in an equal volume of
Prepare as specified by the manufacturer. 0.85% (wtlvol) NaC1, and centrifuge again. Wash the cells
by centrifugation once more, and then lyophilize (see chap-
Procedure ter 47.2.1). Treat 100 mg of dry cells in a Pyrex screw-cap
Dilute samples with distilled water so that they contain 50 test tube with a Teflon-lined cap with 1 ml of 30% KOH at
to 300 kg of glucose per ml. Adjust the pH to between 6.8 100C in a steamer or a boiling-water bath for 3 h. If
and 7.2 with 0.1 M KOH or 0.1 M HCI. A pH indicator lyophilization is not practical, suspend 500 mg of packed
paper is adequate. Prepare a blank and a set of standards wet cells in 0.6 ml of 50% (wtlvol) KOH and heat for 3 h
containing 50 to 300 kg of glucose in 1.0 ml of distilled at 100C to solubilize the glycogen. Cool without opening
water. Add the enzyme and chromogen, and incubate the the tube, and then add 3 ml of distilled water and 8 ml of
mixture as specified by the manufacturer. Add 1 drop of 4 M ethanol to precipitate the glycogen. Centrifuge at 10,000 x
HCl with a Pasteur pipette, and read the A400.Determine g for 15 min, and wash the precipitate with 8 ml of 60%
the concentration of glucose in the samples from a standard (vol/vol in distilled water) ice-cold ethanol by centrifuga-
curve prepared by plotting the absorbances of the standards tion. Dry the precipitate in a vacuum desiccator or
versus the concentration of glucose. lyophilize. The washed precipitate can be analyzed for glu-
cose by the anthrone or phenol method (see section 18.3.1
Applications or 18.3.2) or by enzymatic methods (see section 18.3.3).
The oxidase tests for glucose, galactose, lactose, and sucrose Mechanical disruption of gram-positive cells is some-
are suitable for estimating levels of each in growth media, in times necessary to obtain complete extraction of glycogen
neutralized hydrolysates of polysaccharides, and in enzyme (31). If cells have a polysaccharide capsule, the capsular
reaction mixtures. For example, the enzymatic hydrolysis of material will contaminate the glycogen preparation since,
cellulose can be monitored by measurement of glucose re- in most cases, it has similar solubility properties in ethanol.
leased, and the metabolism of sucrose in a fermentor con-
taining molasses can be monitored by measurement of su- 18.3.5. Starch
crose disappearance. A particularly attractive monitoring Starch is not a common component of microbial cells, but
system is found in the YSI analyzer equipped for simultane- it is used as a substrate for a variety of bacteria and yeasts. It
ous measurement of both glucose consumption and either is therefore sometimes of value to know the level of starch
ethanol or lactate production due to metabolizing microor- in a microbial culture. Soluble starch can be obtained free
ganisms, e.g., yeasts or lactic acid bacteria, respectively (see of cells by use of selective centrifugation, and some organic
YSI website above). solvents can be of value in its precipitation and purification.
One of the major attractions of enzymatic methods for It is easily hydrolyzed by acid and heat, or by using enzymes
sugar analyses is their relative specificity. In general, there (e.g., glucoamylase), and the resultant glucose can be esti-
are many fewer interferences than with chemical methods. mated by any of the methods above in sections 18.3.1,
However, caution is always a good practice in analysis, and 18.3.2, and 18.3.3. A n automated enzymatic method is also
appropriate controls should always be used. It is wise to add available in the YSI analyzer discussed above. Insoluble
a standard to each type of unknown solution to check for crude starch is more problematic, because of the difficulty of
interference by salt and other possible inhibitors of oxi- purification separate from the growing cells.
dases.
18.3.6. Hexosamines in Purified Peptidoglycans
18.3.4. Glycogen Hexosamines are found in lipopolysaccharides, peptidogly-
Glycogen, in contrast with many bacterially produced poly- cans, and teichoic acids of bacteria (19, 20, 22, 30, 31, 37,
saccharides, is easy to quantify by unsophisticated methods. 40, 56). The most common hexosamines are glucosamine,
The method for isolation of glycogen is presented because galactosamine, and muramic acid. All N-acetylated hex-
this polymer functions as a reserve material in many bacte- osamines react with pdimethylaminobenzaldehyde to pro-
ria and can account for a significant fraction of the biomass duce a red product. The test is very sensitive and specific for
in cultures. Methods for isolation of other commonly oc- hexosamines but does not distinguish among glucosamine,
curring complex polysaccharides (capsules, lipopolysaccha- muramic acid, and galactosamine. Muramic acid can be es-
rides, and peptidoglycan) are presented in separate sections. timated separately by other procedures (section 18.3.8).
470 rn METABOLISM

The following procedure modified by Ghuysen et al. Applications


(37) is more precise and convenient than the Morgan-Elson As mentioned above, this procedure is not suited for analy-
procedure as originally described and is applicable to the es- ses of hexosamines in complex mixtures of sugars and sugar
timation of total hexosamines in purified peptidoglycans. derivatives. Hexosamine derivatives such as N-acetylmu-
A n alternative set of procedures (not described here) which ramic acid substituted with peptides produced by the action
can distinguish between glucosamine and galactosamine, of lysozyme on cell walls yield the same colored products.
and which can also estimate total hexosamine, has been The precise neutralization of the HC1 after hydrolysis of
described by Blumenkrantz and Asboe-Hansen (22). samples is critical to the reaction. Therefore, it is important
However, estimation of hexosamines in complex polymers to prepare the HC1 and NaOH carefully and to be sure that
other than peptidoglycan or bacterial cells requires separa- the ampoules or tubes are tightly sealed during hydrolysis.
tion of hexosamines from neutral sugars by ion-exchange Glucosamine, galactosamine, and muramic acid can be
chromatography, as described in the method in the next separately estimated by other methods (26, 60). A n enzy-
section (Hexosamines in Complex Solutions). matic assay for galactosamine has been described previously
(58). Other assays for muramic acid are described in section
Reagents, Equipment, and Supplies 18.3.8.
Morgan-Elson reagents. Dissolve 16 g of p-dimethyl-
aminobenzaldehyde in sufficient acetic acid to give a 18.3.7. Hexosamines in Complex Solutions
volume of 95 ml. Add 5 ml of concentrated HC1. Dilute Hexosamines in complex solutions and those produced by
1:5 with acetic acid to prepare the color reagent. (CAU- the hydrolysis of mixed polymers can be separated from
TION: See section 18.10.) neutral sugars that interfere with the Morgan-Elson reac-
3 N HC1 (CAUTION: See section 18.10.) tion by ion-exchange chromatography. The purified hex-
osamines can then be quantified by the modified reaction
3 N NaOH (CAUTION: See section 18.10.)
described above. Alternative procedures (not described
Saturated solution of sodium bicarbonate. Add ca. 15 g of here) that can distinguish between glucosamine and galac-
NaHC03 to 100 ml of distilled water, and stir at room tosamine and can also estimate total hexosamine have been
temperature for 20 min. described by Blumenkrantz and Asboe-Hansen (22).
5% (vol/vol) acetic anhydride in distilled water. Prepare
just before use, and keep on ice until use. (CAUTION: Reagents, Equipment, and Supplies
See section 18.10.) 4 N HC1 (CAUTION: See section 18.10)
5% Potassium tetraborate in distilled water 1 N NaOH (CAUTION: See section 18.10)
Glucosamine standard. Dissolve 17.92 mg of glucosamine in Lyophilization ampoules or Pyrex screw-cap test tubes with
10 ml of 3 N HC1; store this stock solution in the refrig- Teflon-lined caps
erator (for no longer than 1 week). This standard con-
Dowex-50 (H) columns:
tains 0.2 pmol in 20 p1. Dilute with 3 N HC1 to obtain
standards containing 0.01 to 0.2 pmo1/20 pl. Preparation (precycling) procedure
Ampoules: 1-ml lyophilization ampoules or 1-ml screw-cap
test tubes with Teflon-lined caps Add Dowex-50 resin (1 g for each sample to be analyzed)
to 2 N NaOH (20 ml/g of resin). Mix for ca. 8 min,
Pyrex test tubes, 3 ml and filter onto a Buchner funnel. Wash the resin with
Spectrophotometer or colorimeter distilled water (50 ml/g), and slowly stir the resin
Glass cuvettes or tubes for spectrophotometry, 1 ml with 3 N HC1 (20 ml/g). Remove the HC1 by filtra-
Lyophilizer or vacuum desiccator tion on a Buchner funnel, and wash the resin on the
funnel with ca. 2 liters of distilled water to remove
Procedure the acid. Remove the water by applying suction, and
Lyophilize, or dry in a vacuum desiccator, samples contain- suspend the resin in distilled water (1 g/5 ml). This
ing between 0.01 and 0.1 pmol of hexosamine. Add 20 p l preparation procedure is important for removal of im-
of 3 N HC1 to each sample. Prepare standards of glu- purities in the Dowex resin.
cosamine (0.01 to 0.2 p,mo1/20 pl). Heat the samples and
standards at 95C for 4 h in a sealed ampoule or tube. Cool Use
and neutralize the hydrolysates with 20 pl of 3 M NaOH. Transfer 5 ml of the suspended resin to a Pasteur pipette
Transfer a 30-pl sample of each hydrolysate and standard to or a 3-ml disposable syringe with a ca. 18-gauge nee-
separate 3-ml test tubes. Add 10 p l of a saturated solution dle, plugged at the bottom with glass wool, or to a
of NaHC03 and 10 p l of freshly prepared 5% acetic anhy- commercially prepared minicolumn. Attach rubber
dride. Allow the tubes to stand at room temperature for 10 or silicon tubing and a clamp at the bottom to con-
min, during which N acetylation of the hexosamines oc- trol effluent flow. Do not allow the liquid surface to
curs. Then, immerse them in boiling water for precisely 3 pass below the top of the resin bed.
min to destroy the unreacted acetic anhydride. Add 50 p l Vacuum desiccator containing NaOH pellets
of 5% KZB407, mix, and heat in a boiling-water bath for 7
min. Cool, and add 0.7 ml of the color reagent (a 1:5 dilu- Nitrogen gas supply (CAUTION: See section 18.10)
tion of the Morgan-Elson reagent). Mix again, and incubate Sintered-glass funnel, fine porosity
at 37C for 20 min. Read the A585 of each tube. Buchner funnel
Determine the amount of total hexosamine in each
sample from a standard curve prepared by plotting the A585 Procedure
versus the amount of hexosamine in each standard. Mix the sample to be assayed with 4 N HC1 in a small
Reaction products of glucosamine, galactosamine, and mu- lyophilization ampoule. The amount of material and vol-
ramic acids have the same extinction coefficients. ume of acid will vary with the material used. Samples
18. Chemical Analysis 471

should contain 50 to 100 mg (dry weight) of bacterial cells mation of the 0 acetylation of muramic acid may be of in-
and lesser amounts of purified material in 3 ml of 4 N HC1. terest. This can be accomplished using a modified peptido-
Each sample should contain 0.15 to 0.50 mg of hexosamine glycan protocol, NaOH to hydrolyze the ether-linked ac-
in 3 ml. Hydrolyze the preparation for 6 h at 100C in a vial etate, and HPLC or enzymatic analysis of the acetate (54).
which has been flushed with nitrogen gas and sealed.
Hydrolysis times should be optimized by using several Reagents, Equipment, and Glassware
subsamples treated for different times. The amount of hex- Copper sulfate solution. Dissolve 4 g of CuSO4 in 100 ml of
osamine will increase to a maximum and then decrease with distilled water.
time of hydrolysis because of hexosamine destruction. Cor- p-Hydroxydiphenyl reagent. Dissolve 1.5 g of phydroxy-
rections for losses can be made by adding a known amount diphenyl in 100 ml of 95% (vol/vol) ethanol (CAU-
of hexosamine to one sample. TION: See section 18.10).
Dry the hydrolysate in vacuo over NaOH to remove
HC1. Drying can be accomplished in a vacuum desiccator Muramic acid stock solution. Dissolve muramic acid (Sigma
containing NaOH pellets; add water and redry. Dissolve the Chemical Co.) in water to give 0.25 mg/ml. Muramic
hydrolysate in water, and filter to remove insoluble mate- acid contains 30% by weight of 0-ether-linked lactic
rial. Add the filtered hydrolysate to a small column con- acid.
taining 1 g of Dowex-50 resin. Wash the column with 15 ml Screw-cap test tubes, 20 ml with Teflon-lined screw caps
of water, and discard the eluate. Elute the hexosamines with Spectrophotometer and glass cuvettes or tubes. All glass-
10 ml of 2 N HCI, and dry the eluate in a vacuum desicca- ware must be carefully cleaned, rinsed with sulfuric acid,
tor containing NaOH pellets. Add 2 ml of water, and redry. and then rinsed very well with high-quality distilled
Drying of the hydrolysates and column eluates causes water. After cleaning, contamination with dust and fin-
considerable destruction of muramic acid as a result of con- gerprints must be avoided.
centration of HC1. The losses of this compound are variable; Concentrated sulfuric acid (CAUTION: See section
this procedure tends to underestimate total hexosamines. 18.10)
Dissolve the dried hexosamine fraction in 0.1 ml of
water, and proceed with the estimation of hexosamines as Procedure
described above (section 18.3.6). Because hydrolysis has Neutralize samples of hydrolyzed peptidoglycan in 3 N HCl
been completed, treatment with 3 N HC1 and subsequent (see section 18.3.6) containing 5 to 50 p,g of muramic acid
neutralization can be eliminated. in screw-cap test tubes with Teflon-lined caps by adding an
equal volume of 3 N NaOH. Prepare standards containing
Applications 1 to 100 pg of lactic acid from the muramic acid stock so-
This procedure for purification of hexosamines is necessary lution. Also prepare a reagent blank. Adjust the volume of
when complex polymers other than peptidoglycan (e.g., the samples, standards, and the reagent blank to 1.0 ml with
lipopolysaccharides) are examined. It can be applied to distilled water. Add an additional 0.5 ml of 1.0 N NaOH,
whole bacterial cells with the knowledge that muramic acid and incubate the solution for 30 min at 37C. Add 10 ml of
recoveries will be variable. It is also applicable to estimation concentrated sulfuric acid, seal the tubes, and heat them in
of hexosamines in culture media. For galactosamine analy- boiling water for 10 min. After cooling, add 0.1 ml of the
sis, an alternative to separation of the components of com- copper sulfate reagent and 0.2 ml of the p-hydroxydiphenyl
plex polymers is possible when galactose oxidase is used in reagent and mix rapidly. Incubate the tubes at 30C for 30
an enzymatic assay (58). min in a water bath, and read the A560. Determine the con-
centration of muramic acid in the samples from a standard
18.3.8. Muramic Acid curve prepared by plotting the absorbance of each standard
Muramic acid is found only as a component of the cell wall versus the concentration of muramic acid in the standard.
of bacteria and is not found in any of the archaea (34, 37,
119). The amount of muramic acid in a bacterial cell sus- Applications
pension is a measure of the presence and the amount of pep- The method described above is a simple, rapid, inexpensive
tidoglycan. procedure for detection of muramic acid when metabolic
The most convenient and specific assays for muramic lactic acid is not present. Other components of the mu-
acid involve treatment of acid hydrolysates of cell walls ramic acid do not interfere, nor do uronic acids and neutral
with alkali to remove D-lactate from the 3-position of mu- sugars up to 250 pg per sample.
ramic acid. The D-lactate is then estimated by an enzymatic
method (64) involving lactate dehydrogenase or, as men. 18.3.9. 2-Keto-3-Deoxyoctanoic Acid
tioned above (section 18.3.3), in an automatic enzymatic The 2-keto-3-deoxyoctanoic acid (KDO) in lipopolysac-
method, or by the colorimetric method of Hadzija (40) (see charide can be determined by the method of Weisbach and
below). (Some archaea contain a peptidoglycan cell wall Hurwitz (66) as modified by Osborn (53). When oxidized
material called pseudomurein, but although it contains with periodate, KDO (as well as other 2-keto-3-deoxy car-
hexosamines, it does not contain D-lactate or muramic acid bohydrates) yields formylpyruvic acid, which reacts with
and thus would be negative in the assay described here thiobarbituric acid to yield a chromogen with an absorption
[1191.) maximum at 550 nm. Deoxy sugars other than 2-keto-3-
Recently there has been an increased interest in a struc- deoxy sugar acids produce different products, which form
tural variation in muramic acid, where 0 acetylation occurs chromogens that absorb light maximally outside the 545- to
specifically at the C-6 hydroxyl of muramoyl residues. This 550-nm spectral range.
variation occurs in a least 10 genera of gram-positive and However, some researchers choose to use more than one
gram-negative bacteria (54,65). It is thought that 0 acety- type of KDO assay and average the results in an effort to in-
lation could help some bacteria resist lysozyme action and crease the accuracy, since KDO is generally present at low
thus could be important for virulence (21, 54). Thus, esti- levels (<2% of lipopolysaccharide [dry weight]) compared
472 METABOLISM

to other sugars. For example, Gillespie et al. (38) used an well. Read the A505 and A545 2 h after adding cysteine
average of three different KDO assays (23, 26, 43) to de- reagent. Use a blank containing the same reagents de-
monstrate that KDO accounted for 0.6% of the dry weight scribed above, but lacking the cysteine, to zero the spec-
of Wolinella recta lipopolysaccharide. More recently a fluo- trophotometer at each wavelength.
rometric HPLC method has been described which estimates Subtract the A545from the A505. After this subtraction,
KDO in sialic acid, which should be adaptable to lipo- 1 Fmol of L-glycero-D-mannoheptose in 1 ml gives an ab-
polysaccharide analysis (41). sorbance of 1.07 in a 1-cm cuvette.
Reagents
18.3.1 1. Polysaccharides and Complex
Periodate reagent. Dissolve HI04 in 0.125 N H2S04to give Carbohydrates
a concentration of 0.025 N. (CAUTION: Iodic acid and
periodate are dangerous acids and should be handled in In some cases, experiments require analysis of multicompo-
the hood, using precautions as described for other nent carbohydrates containing a variety of possible sugars
volatile acids in section 18.10) and sometimes organic acids, proteins, and lipids. Analysis
of this type of material is considerably more complex than
Dilute sulfuric acid, 0.02 N (CAUTION: See section that of simple polymers of monosaccharides, and often the
18.10) structural information required may involve the use of nu-
Sodium arsenite reagent: 0.2 N sodium arsenite dissolved in clear magnetic resonance (NMR) and mass spectral data,
0.5 N HCI (CAUTION: See section 18.10) both of which are beyond the scope of this chapter. How-
Thiobarbituric acid reagent: 0.3% thiobarbituric acid in ever, a variety of capsule or slime polysaccharides provide
high-quality distilled water; adjust to pH 2 examples of the utility of techniques available in a large
number of well-equipped laboratories and departments. In
Procedure some cases, noncarbohydrate components, if they are lim-
Dissolve lyophilized polysaccharide containing 0.01 to 0.25 ited in number, can be analyzed by chromatography systems
pmol of KDO in 0.1 ml of 0.02 N H2S04,and heat at identical to or not too dissimilar from those already being
100C for 20 min to release KDO from the polymer. used for sugar analysis.
Hydrolysis needs will vary somewhat, depending on the
Adjust samples of the hydrolysate to 0.2 ml with 0.02 N material, and thus, it is advisable to conduct hydrolysis
H2S04.Add 0.25 ml of the periodate reagent, mix, and under several conditions (in some cases with internal
let stand for 20 min at room temperature. Add 0.5 ml of sugar controls to estimate sugar destruction) and to exam-
0.2 N sodium arsenite solution. Incubate for 2 min at ine the products by relevant and informative methods,
room temperature, and add 2 ml of the thiobarbituric e.g., HPLC or ion chromatography. Both of these chro-
acid reagent. Mix, and heat at 100C for 20 min. Cool, matographic techniques allow the separation of a number
and read the A548.One micromole of KDO per ml gives of compounds and the use of variable detection methods,
an A548of 19.0 in a 1-cm cuvette. e.g., refractive index, pulsed amperometric detection, or
absorbance at various wavelengths. Evaluation of polysac-
Applications charides and complex carbohydrates will be greatly
The procedure described above is suitable for the determi- strengthened by additional confirmatory work, e.g., by en-
nation of KDO in purified lipopolysaccharide or cell wall zymatic assays for glucose or galactose, so that chromato-
preparations. graphic information can be verified when multiple and
sometimes overlapping peaks are present in elution pro-
18.3.1 0. Heptoses files. Likewise, thin-layer chromatography can play a valu-
The product formed when a heptose is heated with sulfuric able confirmatory role in identification.
acid reacts with cysteine hydrochloride to produce a chro- Although exocellular polysaccharides vary considerably
mogen with an intense orange color. This chromogen in their sugar content, most can be harvested and partly pu-
changes to a pink compound which absorbs at 505 nm (53). rified by solvent precipitation, and the component sugars
Reagents, Supplies, and Equipment can be determined by HPLC or ion chromatography.
Described below is an example of a procedure that would be
Sulfuric acid reagent. Add 6 volumes of concentrated suitable for recovery and analysis of exopolysaccharides
HZS04 to 1 volume of water (CAUTION: See section from Acinetobacter calcoaceticus (24,42),Klebsiella spp. (24),
18.10). Rhodotorula glutinis (36), and (possibly with a slight modifi-
Cysteine hydrochloride reagent: 0.3 g of cysteine hy- cation to use ethanol or isopropanol instead of acetone)
drochloride in 10 ml of distilled water Xanthomonas campestris (25), Pseudomonas mendocina (39),
Water bath at 20C and Azotobacter vinelandii (27). The general problem of
microbial polysaccharide recovery has been discussed by
Ice-water bath
Smith and Pace (59).
Boiling-water bath
Spectrophotometer
Reagents, Materials, and Equipment
Cuvettes or tubes for spectrophotometer
Sodium chloride
Procedure Acetone (CAUTION: See section 18.10)
Adjust sample volume to 0.5 ml. Place in an ice bath. Add Sodium azide (CAUTION: See section 18.10)
2.25 ml of the sulfuric acid reagent to samples. Mix the
tubes in the ice-water bath for 3 min. Transfer to the 20C Incubator at 4C
water bath for 3 min, and then heat in boiling water for 10 Dialysis tubing
min. Cool, add 0.05 ml of the cysteine reagent, and mix Screw-cap tubes with Teflon-lined caps
Next Page

18. Chemical Analysis 473

Trifluoroacetic acid (8 N aqueous solution) (CAUTION: Cytoplasmic membranes of all living cells contain lipids.
See section 18.10) Bacterial membranes are much less complex than those of
Boiling-water bath eukaryotic cells. However, bacteria contain a wide variety
Vacuum desiccator or rotary evaporator of lipids in addition to phospholipids. These include sphin-
golipids, neutral lipids, glycolipids, and a variety of unusual
Suitable analysis instrument (e.g., HPLC or ion chromato- lipids associated with organisms of specific genera such
graph) and supplies for its use as Corynebmterium, Nocurdia, and Mycobucterium (69).
Centrifuge, refrigerated Prokaryotes were believed not to contain sterols; however,
Magnetic stirrer it is now known that some bacteria can contain squalene
Lyophilizer and a variety of sterols (70, 91). Gram-negative bacteria
contain unique lipid constituents in lipopolysaccharide
Procedure complexes as part of their outer membrane (69). Poly-p-
Grow cells in 50-ml volumes in a 250-ml flask with an ap- hydroxybutyrate is accumulated in large amounts in the
propriate medium for polysaccharide production (for exam- cytoplasm of several bacteria and serves as a carbon and en-
ples, see above references). Remove cells by centrifugation ergy reserve (88, 89).
at 5,000X g for 20 min. To the supernatant of ca. 45 ml add Archaeal lipids are quite different from those found in
0.05g of NaCL, and mix. Pour this mixture into 200 ml of bacteria (81, 83-87, 96, 97). Archaeal lipids have an ether
acetone, and stir well (acetone can be replaced with 95% bond linking the hydrophobic polyisoprenoid group to the
ethanol or neat isopropanol in most cases). Allow this to sit hydrophilic glycerol, whereas bacterial lipids have an ester
without stirring overnight at 4"C, and the following day bond between the glycerol and an alkane chain. A diversity
centrifuge the material at 10,000 x g for 20 min (at 4C) of novel variations on this ether-linkage and polyisoprenoid
and decant most of the supernatant, leaving the precipitate structural motif can be found among the archaea. Since no
undisturbed. Resuspend the precipitate in a minimum of the archaea have yet been demonstrated to have an outer mem-
remaining solution, and place the slurry in a dialysis bag. (If brane, the major location of these lipids is the cytoplasmic
necessary, the polysaccharide could be washed one or more membrane. In general, total archaeal lipids account for
times with ethanol or another solvent, as described above, about 2 to 5% of the cell (dry weight).
before starting dialysis.) Dialyze overnight at 4C in dis- Because of the complexity of microbial lipids, a descrip-
tilled water containing 0.01% sodium azide. Replace the tion of procedures that would provide a total analysis of
water with pure distilled water, and dialyze for an additional each class is impractical here. For variations of the proce-
3 to 4 h. Lyophilize the remaining slurry to make a dry poly- dures described below, and for detailed techniques required
saccharide powder. for a more complete analysis of bacterial lipids, the reader
Place 4 mg of polysaccharide powder in a Teflon-lined should consult one of the specialized articles on the subject
screw-top glass tube, add 1 ml of 8 N trifluoroacetic acid, (67,68, 71, 76-87,90-101).
and tightly cap the tube. Heat the tube at 100C for 2 h.
Cool the tube to room temperature, uncap, and dry under 18.4.1. Crude Total lipids
reduced pressure (e.g., in a vacuum desiccator containing The procedure for extraction of total crude lipids described
NaOH pellets, under continuous vacuum) for ca. 48 h until by Bligh and Dyer (71) is less time-consuming and requires
only a residue remains (24, 52). less sample manipulation than other methods. The proce-
The residue can then be examined for sugar content by dure has been widely used with bacteria. The bacterial cells
any of a variety of methods including HPLC (18; also see are mixed with chloroform and methanol in amounts that
informational literature from HPLC suppliers), gas-liquid yield a monophasic solution when combined with water in
chromatography (51, 52) (see chapter 17.3.5), or ion the tissue (75). Upon dilution with water or chloroform, a
chromatography (24, 45, 55) (see chapter 17.3.7; e.g., biphasic solution results. The lipids are retained in the chlo-
with a Dionex Q1C analysis system equipped with an roform phase, and nonlipid materials are retained in the
HPIC-AS6 anion-exchange column). In some cases gas- methanol-water phase. The chloroform layer is isolated,
liquid chromatography can be useful when coupled with and the lipids can be purified and separated into classes as
mass spectrometry (28, 48) The enzymatic methods de- described below. The recovery of total crude lipids after sep-
scribed above (section 18.3) are also excellent and specific aration from the nonlipid contaminants should exceed
methods for some possible component sugars. See also 95%.
chapter 17.3 for a description of types of chromatographic Hara and Radin (76) have described an alternative to
analysis. this procedure which does not involve chloroform use and
which has the advantage that centrifugation can be used in-
stead of the filtration steps in the Bligh and Dyer procedure.
18.4. LIPIDS A hexane-isopropanol mixture is used to extract lipids, and
Lipids are a chemically diverse group of biological mole- nonlipid contaminants are removed by washing the extract
cules that are insoluble in water but soluble in organic sol- with aqueous sodium sulfate. The lipid extract can be frac-
vents and that commonly contain long-chain hydrocarbon tionated on silica gel columns by use of hexane, iso-
groups. Lipids include long-chain hydrocarbons, alcohols, propanol, and water mixtures.
aldehydes, fatty acids, and their derivatives including glyc-
erides, phospholipids, glycolipids, and sulfolipids. Sterols, Reagents, Materials, and Equipment
fatty acid esters of sterols, vitamins A, D, E, and K, Methanol (CAUTION: See section 18.10)
carotenoids, and other polyisoprenoids often are considered Chloroform (CAUTION: See section 18.10)
in this class because they, like lipids, are associated with
membranes and can be extracted by lipid solvents. Surveys Filter paper, Whatman no. 1
of the types and composition of microbial and archaeal Porcelain Buchner funnel, ca. 43-mm diameter
lipids have been compiled (69, 81, 85, 96,97). Suction flask: glass, 125 ml
Enzymatic Activity
ROBERT P. HAUSINGER AND ALLEN T. PHILLIPS

19.1. PRINCIPLES ............................................. 505


19.1.1. Units and Specific Activity .............................. 505
. .............................. 505
2. Purification Tables and k...
.3. Reaction Classification and Nomenclature ................... 505
.4. Cofactors. Coenzymes. and Metal Ions ...................... 506
.5. Unknown Substrates .................................. 506
19.2. TYPES OF ENZYME PREPARATIONS ......................... 507
19.2.1. Secreted Enzymes .................................... 507
.2. Intact and Permeabilized Cells ........................... 507
.3. Soluble Extracts ...................................... 507
.4. Membrane Fractions ................................... 508
.5 . Purified Enzymes ..................................... 508
19.3. PRACTICAL ASSAY CONSIDERATIONS ....................... 508
19.3.1. General Comments ................................... 508
.2. Linearity........................................... 509
.3. Enzyme Concentration................................. 510
. ................................
4. Substrate Concentration 510
.5. Coupling Enzymes .................................... 510
. ................................
6. Choice of Assay Method 510
.............................
19.3.6.1. Spectrophotometry 510
. .................................
2. Fluorescence 511
. ............................
3. Selective Electrodes 512
. 4. Chromatography .............................. 512
. .................................
5. Radioactivity 513
. .........................
6. Chemical Derivatization 513
. 7. Other Assay Approaches ........................ 514
19.3.7. Optimization ........................................ 514
19.3.7.1. Effects of pH and Buffer........................ 514
. ..........................
2. Effects of Temperature 515
. .............................
3. Stabilizing Agents 515
. ...............................
4. Oxygen Lability 516
19.4. BASIC ENZYME KINETICS ................................. 516
19.4.1. Single Substrate ...................................... 516
19.4.1.1. Michaelis-Menten Kinetics....................... 516
. 2. Graphical Analysis Methods...................... 517
. ............................
3. Substrate Inhibition 518
. ...........................
4. Cooperative Behavior 518
. ...........................
.5 Reversible Inhibitors 519
. ..................................
6. Inactivators 521
19.4.2. Bisubstrate Reactions .................................. 521
19.4.2.1. Experimental Design ........................... 521
. 2. Substituted Enzymes ........................... 522
. 3. Sequential Enzymes ............................ 522
. 4. Analysis for Ordered Binding ..................... 524
.
.5 Analysis for Random Binding ..................... 524
19.4.3. Regulation of Enzyme Activity ........................... 524
19.4.3.1. Allosteric Regulators ........................... 524
. 2. Covalent Regulation ........................... 525
19.5. CONCLUDING COMMENTS ................................ 525
19.6. REFERENCES ............................................ 526

The study of enzymes and enzyme-catalyzed reactions has at the molecular level. Nevertheless. there remain enor-
contributed greatly to our understanding of microbial me mous gaps in our knowledge regarding the biochemistry
tabolism. just as current advances in protein structure and physiology of all but the best-studied bacteria. and
promise new insights into the detailed workings of enzymes thus. a thorough analysis of enzymes. their action. and their

504
19. Enzymatic Activity 505

properties continues to be an important effort for bacteriol- ent is the active enzyme. For reporting the results of en-
ogists. This chapter is intended to provide a brief descrip- zyme isolation, data should be presented in a purification
tion of the most important principles and approaches used table that includes total units and specific activities at
in enzyme activity measurements. In addition, the discus- each stage of purification. Typically, such a table also in-
sion presents practical information useful to investigators cludes values for enzyme recovery and increase in purifica-
who seek to design enzyme assays for performing meaning- tion (fold purification) for each step. The percent re-
ful and accurate evaluations of catalytic activities. covery is usually calculated on the basis of remaining total
units versus the total units measured in crude cell extracts,
whereas the fold purification is calculated by dividing the
19.1. PRINCIPLES specific activity at each step by the specific activity of the
initial sample. These values are useful in identifying, and
19.1 .I. Units and Specific Activity overcoming, steps that cause substantial losses of enzyme
Although most enzymes are proteins (with the exception of activity (e.g., due to protein instability, loss of a cofactor,
catalytic RNA species) and can be quantified by protein- or other reason). In addition, the final fold purification
specific methods, it is almost invariably useful to express the can give insight into the cellular abundance of the partic-
quantity of an enzyme present in terms of its catalytic activ- ular enzyme of interest. For example, a highly overpro-
ity rather than the content of protein. This has the advan- duced enzyme comprising 10% of the total cell protein
tage of relative specificity, assuming no interfering activities correspondingly would have a maximum of 10-fold purifi-
are present, and measurements can usually be done in im- cation. By contrast, most enzymes are present at much
pure systems containing numerous other protein species as lower abundance and may require several hundred fold, or
well as on purified preparations. The most common defini- higher, purification.
tion of one unit (U) of enzyme activity is the amount of en- A n important term for describing the activity of an iso-
zyme which catalyzes the conversion of 1 pmol of substrate lated enzyme is k,,,, the molecular activity of the enzyme
to product in 1 min under a prescribed set of assay condi- (also referred to as its turnover number). This number de-
tions, i.e., at a stated pH, temperature, buffer type and con- scribes the molecules of substrate(s) consumed per minute
centration, and substrate and cofactor concentrations. This per molecule of enzyme and represents a composite of rate
definition is widely used and is applicable to most situations. constants that limit reaction velocity. It has units of recip-
When the resulting numbers are inconveniently large or rocal minutes and is calculated from the specific activity at
small, multipliers such as kilounits or milliunits are prefer- saturating substrate concentration(s) divided by the molec-
able to defining a nonstandard unit. A n exception to this ular weight of the enzyme. The reciprocal of k,,, is the time
general practice exists for the case when the molecular required for one catalytic cycle. For example, an enzyme
weight of a substrate is unknown, and thus, a micromole with an M, of 30,000 (30 mglpmol) and a measured specific
quantity cannot be specified. In this instance it is acceptable activity of 8,000 U/mg has a k,,, of 2.4 x lo5 min- and
to use some convenient mass of substrate converted to prod- thus completes each catalytic cycle in 250 ps.
uct per minute as the basis for the unit definition. (Another
unit occasionally encountered is the katal, the amount of
enzyme that converts 1 mol of substrate to product in 1 s.
19.1.3. Reaction Classification and Nomenclature
One enzyme unit as described above is equal to 16.67 nkat.) The current system of enzyme nomenclature recognizes six
Whereas an enzyme unit is a measure of quantity ex- classes of enzymes and defines the basis for a systematic
pressed in catalytic terms, the relative purity of an enzyme name for each enzyme. A listing of enzymes along with their
is specified by its specific activity. Specific activity typically recommended names and numerical enzyme codes (EC
is indicated as the number of enzyme units present per mil- numbers) is periodically updated by the International
ligram of protein. Since estimates of protein content are Union of Biochemistry and Molecular Biology (http://www
highly influenced by the method used for their determina- .chem.qmul.ac.uk/iubmb/enzyme/)and should be consulted
tion, values of specific activity should always be accompa- when preparing results of enzyme studies for publication.
nied by information about how the protein is quantified The EC number (e.g., 3.2.1.23) is made up of four parts: the
(both the method and the reference protein used) as well as first decimal indicates the enzymes class; the next two dec-
how an enzyme unit is defined. For cell extracts and other imals classify the enzyme according to type of bonds broken,
impure preparations, simple colorimetric protein assays (e.g., coenzymes used, and other important characteristics; and
Lowry, Bradford, and bicinchoninic acid methods; see refer- the final decimal is a serial number used to distinguish
ence 4) are suitable. These methods yield widely variable among enzymes otherwise classified together. Because EC
extinction coefficients for different purified protein samples; numbers provide generally unambiguous identification of
hence, it is necessary to determine the appropriate correction enzymes, they are of increasing importance in assisting the
factor for use with the isolated protein of interest. Precise cataloging of sequence data for computer searches. A brief
measurements of purified protein concentrations are ob- explanation of the six enzyme classes follows.
tained by quantitative amino acid analysis or by using the Class 1: Oxidoreductases. Enzymes in class 1 catalyze oxida-
molar absorption at 280 nm calculated on the basis of the tion-reduction reactions and are assigned systematic
numbers of tryptophan, tyrosine, and cystine residues in names based on the form hydrogen donor:acceptor oxi-
the unfolded protein (26). Comparisons of enzyme specific doreductase. The name dehydrogenase is recommended
activities among cells grown under different medium condi- for normal use, but reductase is acceptable in cases
tions can provide important information about the relative where this more correctly describes the usual reaction di-
levels of biosynthesis of the enzyme of interest. rection; oxygenase or oxidase is used when O2is the ac-
ceptor species. Thus, the NAD+-dependent isocitrate
19.1.2. Purification Tables and kcat dehydrogenase of the tricarboxylic acid cycle has the as-
For a given enzyme, the specific activity increases during signed systematic name isocitrate:NAD+ oxidoreductase
purification up to a limit value at which all protein pres- (decarboxylating) , EC 1.1.141.
506 METABOLISM

Class 2: Tramferases. Class 2 enzymes catalyze group transfer 19.1.4. Cofactors, Coenzymes, and Metal Ions
from a donor to an acceptor. Accordingly, the systematic Many enzyme reactions require substances other than the
designation for the enzyme catalyzing the ATP-dependent protein and substrate for optimal activity. These accessory
phosphorylation of adenosine to yield AMP + ADP is materials, termed cofactors, are in most cases unchanged at
ATP:adenosine 5'-phosphotransferase, EC 2.7.1.20, the end of a catalytic cycle and are regarded as important
more commonly known as adenosine kinase. Enzymes (often essential) parts of the reaction mechanism. One im-
catalyzing transamination (aminotransferases) and hex- portant class of cofactors involves coenzymes, organic mol-
ose transfer (e.g., glycogen synthase, with its systematic ecules that have a precisely defined role in the reaction. In
name UDP-g1ucose:glycogen 4-a-D-g~ucosyltransferase, some cases, these coenzymes are tightly bound (e.g., flavin
EC 2.4.1.11) are also included in this class. and cobalamin) or even covalently bonded (e.g., biotin and
Class 3: Hydrolases. Class 3 is a broad category that covers lipoic acid) to the enzyme protein and are known as pros-
enzymes catalyzing the hydrolytic cleavage of a substrate. thetic groups. Alternatively, the coenzyme may be more
These enzymes comprise a special group of transferases loosely associated with the enzyme of interest (e.g., pyri-
in which the acceptor group is water. In cases such as doxal phosphate) and can be lost during purification. Many
proteolytic enzymes (carboxypeptidase or collagenase) investigators refer to compounds such as ATP, NAD+, and
or phosphatases, specificity is usually directed toward a CoA as cellular coenzymes, since they are regenerated by
family of related substrates containing common struc- other enzyme systems within the cell, even though they are
tural features rather than specific compounds. Although modified during the specific enzyme reaction. When work-
their systematic names always include the term hydro- ing with the isolated enzymes, such compounds must be
lase, as in urea amidohydrolase (EC 3.5.1.5), members of treated as substrates unless a regenerating system is pro-
this class are frequently referred to by the name of their vided. Metal ions comprise a second large group of cofac-
substrate with the suffix -ase (e.g., urease, penicillinase, tors. It has been estimated that more than one-third of all
and P-galactosidase). Also found in this class are pro- enzymes possess bound metal ions, often with the metals
teinases (proteases), many with nonstandard but still playing a critical role in substrate binding, electron transfer,
widely used names, such as trypsin and papain. or protein stabilization. Although substitution with related
Class 4 : Lyases. Class 4 enzymes catalyze cleavage of C-C, metal ions often is possible, metal-containing enzymes typ-
C-0, and C-N bonds (plus a few others) with elimina- ically are highly specific to a particular metal ion due to
tion of a group to leave an unsaturated residue; the con- structural constraints on the size or allowed coordination
verse addition of groups to double bonds is included as geometry. Metalloenzymes include examples with a wide
well. Most lyase reactions are referred to as uni-bi type, variety of mononuclear, dinuclear, and polynuclear metallo-
meaning that they involve conversion of a single sub- centers (10). Additional cellular substances also can act as
strate to two products. Their systematic names are based enzyme activators by enhancing catalysis or stabilizing en-
on the form substrate group-lyase, as in S-adenosylme- zyme structure. Cellular control of the levels of these sub-
thionine carboxy-lyase, EC 4.1.1.50 (also known as a de- stances may allow for allosteric regulation of selected en-
carboxylase), and L-malate hydro-lyase, commonly zymes (see section 19.4.3.1).
called fumarase or fumarate hydratase. Several enzymes 19.1.5. Unknown Substrates
involving coenzyme A (CoA)-dependent cleavages, as
Microbiologists increasingly are faced with the challenge of
in citrate synthase, or addition of another element con-
characterizing gene products suspected of being enzymes,
comitant with substrate cleavage (e.g., anthranilate syn-
but for which the reactions catalyzed are unknown. An-
thase) are less obvious members of the lyase family.
notations derived from genomics studies often are flawed or
Class 5: Isomerases. Class 5 enzymes catalyze geometric or misleading, and even protein structures obtained from pro-
structural changes within one molecule. Class 5 includes teomics studies can fail to assign proper function. Never-
enzymes variously known as racemases, epimerases, iso- theless, the results of such studies should be carefully scruti-
merases, tautomerases, and mutases. There is no single nized as a first step to identifying the true substrate. A target
form for the systematic name, and thus, one finds ala- gene sequence may be related to those of family members
nine racemase (interconverting D- and L-alanine, EC that catalyze mechanistically similar reactions or that use
5.1.1.1 ) as well as ~-ribulose-5-phosphate3-epimerase structurally similar substrates (7). Examination of flanking
(EC 5.1.3.1 ) and ~-glyceraldehyde-3-phosphate ketol- DNA regions may reveal sequences encoding enzymes cat-
isomerase (EC 5.3.1.1) (better recognized as triosephos- alyzing other steps in a pathway and thus help to define the
phate isomerase). role of the gene of interest. Structural information about a
Class 6: Ligases (Synthetases) . Ligases catalyze the joining of gene product should be compared to known three-dimen-
two molecules, coupled with the hydrolysis of a py- sional structures, again with the goal of seeking features that
rophosphate bond from ATP or similar triphosphate. provide mechanistic insight or substrate preferences. Gene
Their systematic names are based on the convention array or other approaches to assess the effects of varied
substrate 1:substrate 2 ligase (ADP-forming). The rec- growth conditions on gene expression can yield additional
ommended class name is synthetase, not to be confused hints about function. After compiling such information,
with synthase, which simply describes an enzyme that reasonable guesses of possible substrates will need to be
joins parts of two molecules without involvement of py- tested by appropriately designed assays. If transformation of
rophosphate bond cleavage. Examples of synthetases are any compound is detected, structural variations of that
pyruvate carboxylase (pyruvate:carbon-dioxide ligase compound can be tested. Comparison of possible substrates
[ADP-forming], EC 6.4.1.1), glutamine synthetase [(L- typically involves analysis of the catalytic efficiency
g1utamate:ammonia ligase [ADP-forming], EC 6.3.1.2), (Vmax/Km), a ratio of kinetic constants described in section
and leucyl-tRNA synthetase (L-leucine:tRNALeUligase 19.4, that accounts for both the rate of the reaction and the
[AMP-forming], EC 6.1.1.4). affinity of the enzyme for the test substrate. This value is
19. Enzymatic Activity m 507

also known as the specificity constant and is the second- reached high intracellular concentrations, or alternatively,
order rate constant for a reaction at low substrate concen- the product formed may be consumed by subsequent reac-
trations. The enzyme often, but not always, exhibits the tions catalyzed by other oxygenases, thus producing an
highest catalytic efficiency/specificity constant with the erroneous rate of oxygen consumption. Nuclear magnetic
true substrate of a reaction. resonance (NMR) analysis is an approach that partially
A different situation is encountered in many oxidation- overcomes these limitations in studies of in vivo activities.
reduction reactions in which the cellular electron carrier is For example, NMR using 31P- and '3C-containing sub-
unknown. Common electron carriers include a variety of strates enables whole-cell measurements of the rates of spe-
cytochromes, ferredoxins, quinones, and other components, cific enzymes in cells maintained under various conditions
many of which are distinct for different microorganisms. In (2, 34). The low sensitivity of NMR necessitates high cell
some cases, a commercially available, easily purified, or densities in order to have suffkient concentrations of meta-
more soluble surrogate electron carrier can be used, such bolic intermediates; for many analyses, 10" cells per ml are
as horse heart cytochrome c, spinach ferredoxin, or required, and this presents some difficulty if oxygenation is
menaquinone; however, caution must be used in the inter- important. Due to the above concerns, quantitative assays
pretation of kinetic studies in which the natural electron are not generally recommended with whole cells.
carrier is replaced by these reagents. In addition, some en- One approach to overcome problems involving substrate
zyme activities are monitored by using artificial electron ac- or product transport across the membrane is to permeabilize
ceptors such as synthetic dyes (e.g., methyl or benzyl violo- the cells by chemical or enzymatic treatment. Among the
gen). These compounds undergo a large change in color gentlest permeabilization methods is Tris-EDTA treatment.
intensity depending on their oxidation-reduction state, al- Solutions containing moderate cell densities (1 mg [dry
lowing for convenient and sensitive assays, but again the weightllml or less) are adjusted to 0.1 M Tris-HC1 (pH 7.5)
relevance to the authentic biological electron carrier can be and 1 mM EDTA, which alters the permeability of the
questioned. lipopolysaccharide-containing outer membrane of gram-
negative bacteria by a combination of chelating Mg2+ ions
and some incompletely understood effects of Tris buffer
19.2. TYPES OF ENZYME PREPARATIONS (1 1). Toluene is another widely used permeabilization agent
that probably disrupts membrane lipid regions, permitting a
19.2.1. Secreted Enzymes wide variety of low-molecular-weight materials access to the
Particular microorganisms are adept at degrading various cell interior while also allowing many metabolites to diffuse
biopolymers by secreting the corresponding biodegradative out. For example, 1 drop (50 1.1) in a 1-ml assay volume is
enzymes, including proteases, glycosidases, lipases, and nu- sufficient to render Escherichia coli cells permeable to vari-
cleases. In addition, a secreted enzyme can result from use ous amino acids and carbohydrates. Chloroform with deter-
of any of several commercially available cloning vectors gent added (e.g., 15 pl of chloroform and 50 p1 of a 1%
that are designed to export the product of any selected gene. [wt/vol] sodium deoxycholate solution per ml of cell sus-
Analysis of such secreted enzymes simply requires removal pension) also is effective for permeabilizing enteric organ-
of the intact cells by standard centrifugation or filtration isms. Detergents alone are used as permeabilization agents,
methods and use of an appropriate assay. If desired, addi- although in many instances it is unclear whether the cells
tional enrichment steps can be carried out by traditional remain intact during the subsequent assay. The cationic de-
purification methods prior to characterization of the en- tergent cetyltrimethylammonium bromide (CTAB) (used
zyme activity. at a final concentration of 0.05 to 0.1 mg/ml) is representa-
tive of these compounds. In all applications with permeabi-
19.2.2. Intact and Permeabilized Cells lized cells, there may be some variation in results as a func-
Intact cells are often used to provide qualitative informa- tion of the concentration of cells used or the time allowed
tion related to an enzyme activity. For example, whole-cell for the permeabilizer to function. Standardization of cell
assays are very useful for identifying particular isolates pos- density in the assay is generally desirable for maximum re-
sessing high levels of a desired activity or for screening re- producibility. Finally, it is important to include controls
combinants to identify those that express a desired gene. In with no permeabilizing agent to ascertain that the treat-
contrast, in only a few cases is it possible to obtain reason- ment is effective.
ably quantitative enzyme activity results by using intact
cells. For example, the enzyme nitrogenase typically is as- 19.2.3. Soluble Extracts
sayed by monitoring conversion of acetylene (an alternate The great majority of enzyme assays are conducted on cell-
substrate) to ethylene using gas chromatography (see sec- free preparations. This approach allows for careful control
tion 19.3.6.4). Both of these gases readily diffuse through of substrate and cofactor concentrations, reaction condi-
the cellular membrane so the substrate can be delivered in tions such as pH and ionic strength, and the use of coupled
known and nonlimiting quantities while the product does assays where desired. Selective enrichment of periplasmic
not collect within the cell. Other gases (e.g., 02, COz,and enzymes is possible by use of methods that lyse the outer
Hz) and some nongaseous substrates also diffuse readily membrane while keeping the inner membrane intact. The
through membranes and can be delivered in a controlled remaining cellular material is simply removed by centrifu-
fashion; however, assays involving these compounds must gation, and the soluble periplasmic material is further char-
also take into consideration whether cosubstrates or prod- acterized. This approach is especially useful for analysis of
ucts also have the ability to cross the membrane. In the the products of genes that are cloned into commercially avail-
assay of a specific oxygenase, an oxygen electrode can mea- able plasmid vectors designed for recombinant protein export
sure oxygen consumption activity in a suspension of whole to the periplasmic region. For characterization of cytoplasmic
cells (see section 19.3.6.3), but the second substrate under- enzymes, a wide variety of physical methods is available to
going oxidation may be rate limiting because it has not disrupt cells including those based on hydrodynamic shear,
508 METABOLISM

ultrasonic disruption, and homogenization with glass beads. or gel filtration can be used to remove contaminating low-
In addition, release of cytoplasmic proteins can be achieved molecular-weight components. A reasonable selection of
using enzymatic lysis and commercially available cell lysis protease inhibitors is available, including metal chelators
reagents. Membranes and other cellular debris are readily for metalloproteases, serine protease inhibitors such as
removed by ultracentrifugation, leaving a soluble solution phenylmethylsulfonyl fluoride, and inhibitory peptides such
of cell extracts. Further information on cell disruption tech- as leupeptin. These inhibitors may be unnecessary for the
niques is found in chapter 7. high protein concentration of most crude extracts unless
extracts are to be stored prior to assay, an inadvisable op-
19.2.4. Membrane Fractions tion. The problem of competing activities, e.g., contami-
Some enzymes are membrane associated, leading to numer- nating ATPase activity in assays which measure kinase-
ous complications in their characterization. The prepara- dependent production of ADP from ATP, is clearly a major
tion of a membrane fraction begins with one of the cell dis- limitation on the conduct of assays and one for which there
ruption methods mentioned above. Various centrifugation is no general solution. Fortunately, innovative assay meth-
methods are available (e.g., standard ultracentrifugation as ods or a modified design can help in some instances, but
well as sucrose gradient and isopycnic methods) to further when that is not possible, partial or complete purification is
enrich a general membrane fraction or to isolate a particu- the only recourse. Summation of the numerous detailed
lar type of membrane, as described in chapter 7. The result- procedures available for enzyme purification (for an exam-
ing preparation generally contains membrane vesicles, ple, see reference 4) is beyond the scope of this chapter.
which are problematic for enzyme studies. Light refraction
by such vesicles interferes with many common enzyme as-
says. Also, the enzyme of interest may be positioned ran- 19.3. PRACTICAL ASSAY CONSIDERATIONS
domly toward each face or selectively facing either the
inner or outer surface depending on the physical method 19.3.1. General Comments
used to disrupt the cells; the orientation can impact sub- Depending on the needs of the investigator, assays are per-
strate delivery to, and product release from, the enzyme. formed to ascertain whether a particular activity is present,
Finally, the presence of other membrane-associated proteins assess the activity level under precisely defined conditions,
complicates efforts at enzyme isolation. In particular situa- or monitor the effects of various conditions on enzymatic
tions, enzyme characterization studies can be carried out rates. In the most simple situation, samples are incubated
with such vesicles (e.g., examining cytochrome c oxidase or with a given amount (usually a near saturating level) of sub-
other enzymes involved in bioenergetics where an intact strate( s) and any additional required components (cofac-
membrane is needed), but in general further solubilization tors, metal ions, stabilizing agents, or allosteric effectors) in
is desired. For loosely associated proteins, the membrane in- a specific buffer (and pH) at a chosen temperature and for a
teraction often can be disrupted by high ionic strength (e.g., particular time, and then the extent of product formation or
inclusion of 0.5 to 5 M KC1 or other salt). In other cases, it substrate consumption is monitored. The amount of trans-
is possible by careful use of proteases to separate a soluble formation is compared to the extent of nonenzymatic con-
domain associated with activity from its attachment to a version to assess activities (e.g., in cell extracts derived from
hydrophobic domain inserted into the membrane. More various isolates or from a single strain grown under a variety
typically, solubilization requires the selective use of low of growth conditions). The activity values obtained from
concentrations (e.g., 0.1 to 3%, wtlvol) of a mild ionic or these measurements allow reasonable comparisons to be
nonionic detergent such as sodium deoxycholate or Triton made among the various samples for the given experimen-
(see chapter 7). In each of these cases, the solubilized pro- tal conditions. For more detailed characterization of an en-
tein can be further purified by most routine methods (al- zyme activity, the approach usually involves precise deter-
though it may be necessary to retain detergent in all mination of initial rates of the enzyme activity in an
buffers). If necessary, purified and solubilized membrane optimized assay system, often using varied substrate concen-
proteins can be reconstituted into artificially created lipid tration(s). Key criteria in developing an assay for an enzyme
vesicles in order to assay reactions that require intact mem- of interest include its specificity, sensitivity, precision, and
branes. convenience.
Two choices in developing an assay involve use of a con-
19.2.5. Purified Enzymes tinuous or discontinuous method and whether to measure
Serious kinetic studies and documentation of most enzyme product formation or substrate disappearance. Only certain
properties are best conducted with purified preparations, assay procedures (e.g., spectrophotometry, fluorescence
but there are situations in which quantitative assays of en- methods, and various types of electrodes) allow continuous
zymes in unpurified extracts are desirable and even prefer- data collection, whereas others require the taking of time
able. For example, some important enzymatic properties can points. The former approach generally requires little effort
be altered by purification, such as the removal of a key al- by an investigator during a single assay and provides imme-
losteric effector (see section 19.4.3.1). More commonly, as- diate information; however, instrument design may limit
says are performed with crude extracts simply because only monitoring to only one or a few samples at a time.
comparative activities are sought among samples in a group Although discontinuous methods do not provide immedi-
and pure enzymes are not needed for the desired compar- ate feedback, automated sampling systems have increased
isons. This might be the case when conducting a physiolog- the ease of analysis for multiple samples and allow extensive
ical study, such as the effect of a growth condition on en- sets of data to be collected on previously quenched samples
zyme production, or when assaying mutants. (in which the reaction has been stopped). Thus, both meth-
The principal limitations on the use of crude extracts for ods are commonly employed in enzyme assays. For either
assays are the presence of competing activities, endogenous assay approach, the product concentrations typically un-
inhibitors, endogenous substrate, and proteolytic enzymes dergo dramatic increases during the initial stages of an en-
that might reduce the assayable activity. Frequently, dialysis zyme assay while substrate concentration changes may be
19. Enzymatic Activity m 509

minimal; thus, analysis of product formation often offers the 160 I I


greatest sensitivity for measurements of initial rates.
Nevertheless, it is possible to obtain initial rates by follow- 140
ing changes in substrate concentration if the detection lim-
its allow precise measurements in samples that retain satu- 120
rating substrate levels.
The following sections provide additional guidelines for I, 100
development and use of quantitative assays. As described in
sections 19.3.2 and 19.3.3, it is important that the enzy-
matic rate (also known as velocity) be linear for an accept-
able time period and is directly proportional to the amount
of added enzyme. Substrate concentrations and other assay 40
conditions must be carefully controlled, as detailed in sec-
tions 19.3.4 and 19.3.7. Furthermore, one must choose the 20
best assay procedure (several specific examples are described
in section 19.3.6) based on its sensitivity, ease of use, equip- 0
ment available, or other factors related to an enzyme of in- 0 5 10 15 20 25 30
terest. In some assays, the immediate product of the target Time, min
enzyme is converted to another species for ease of detection;
use of such coupling enzymes can introduce additional con- FIGURE 1 Comparison of progress curves for enzymes that
cerns as described in section 19.3.5. Regardless of the assay are stable, activated, or inactivated during the assay. For the fa-
method, it is critical to calibrate the procedure with stan- vorable situation of a stable enzyme (a), the concentration of
dards of known purity. For example, in assays that monitor product formed per unit of time increases in a linear fashion
an absorbance change it may be necessary to determine the (shown here as w = 5 pM/min). For a particular enzyme, the
extinction coefficient of a product or substrate. More gen- activity may increase during the assay to reach a steady-state
erally, it is advisable to perform mock assays that contain all level ( A ) . This situation is observed for some coupled enzyme
ingredients but one (e.g., no substrate or no enzyme) and systems, or it may be associated with enzyme disaggregation,
then add known amounts of the material to be analyzed. It dissociation of a slow-binding inhibitor, or other type of acti-
is also important to emphasize that development of an assay vation that occurs under assay conditions. Progress curve data
is an iterative procedure in which changes made in one pa- for an enzyme exhibiting activation can be fit to equation 12
rameter may affect the enzyme behavior toward other pa- (with vg = 0) to obtain the final rate (wt) and the apparent
rameters. For a much more complete description of specific first-order rate constant for establishment of equilibrium (kapp).
assays, multivolume collective works as Methods in En- In the opposite situation (m), the rate of product formation may
zymology and Methods of Enzymatic Analysis should be con- decrease over time and eventually result in complete enzyme
sulted. inactivation. Progress curve data for such a labile enzyme can
be fit to equation 1 to obtain the initial rate and the inactiva-
19.3.2. Linearity tion rate constant (w, = 5 pM/min and k,,,,, = 0.1 min-' for
An important criterion in any enzyme assay is establishing the situation shown). Attempts to estimate v, by taking the
that the velocity is constant throughout the incubation pe- tangent to the first few points of such data generally result in
riod. To evaluate the constancy of velocity, a series of iden- an underestimation of the true value. By proper choice of assay
tical assays are set up (with the same enzyme concentration conditions (inclusion of reductant, addition or removal of
throughout) and terminated at different times. The amount metal chelators, change in ionic strength, provision of glycerol
of product formed (or substrate consumed) should yield, in or other additives, etc.), it may be possible to sufficiently en-
a favorable situation, a linear plot as shown by the circles in hance the stability of an enzyme so that a linear response is ob-
Fig. 1. If the data are linear over a reasonable time period, a tained, thus facilitating further characterization.
single-time-point assay taken within the linear range may
suffice for further enzyme characterization.
Nonlinearity of the data can take two forms: a lag phase
followed by a linear plot (Fig. 1, triangles) or the more com-
mon situation of decreasing velocity with time (Fig. 1, aggregation during the assay or the slow binding of an acti-
squares). A lag phase observed during the assay may be at- vator in the mixture, the enzyme can be diluted into the
tributed to a change in the state of aggregation of the en- assay mix and allowed to equilibrate prior to adding a small
zyme upon dilution into the assay mixture (for example, a volume of concentrated substrate to initiate the reaction.
monomeric species may exhibit higher activity than a poly- Problems related to dissociation of a tightly bound inhibitor
mer), dissociation of a tight-binding inhibitor (present also may be alleviated by extensive dialysis of the sample
within the cell or added during a stage of purification; see prior to doing the assay.
section 19.4.1.5), slow activation by some component in The opposite situation, a loss of activity over time, is de-
the assay mix, stimulation of activity by the product, or any picted in Fig. 1 (squares) and can arise from many possible
of several other reasons. In addition, this behavior is often types of inactivation reactions. One situation giving rise to
observed in coupled enzyme systems as described in section this type of behavior involves substrate depletion; the use of
19.3.5. Simply measuring the enzymatic rate during the lin- higher initial substrate concentrations may eliminate this
ear phase generally provides suitable values for maximal concern, provided substrate inhibition is not a problem. In
rates; however, this approach may be inadequate for more other cases, the enzyme can be stabilized, thus increasing
complete kinetic analyses. It may be worthwhile to examine the assay linearity, by suitable adjustment of pH, decreasing
modifications of the assay in an effort to eliminate the lag the temperature, adding stabilizing agents, or removing oxy-
phase. To reduce the possible complications of enzyme dis- gen, as described in section 19.3.7. It is generally advisable
510 METABOLISM

to expend effort at an early stage of investigation to elimi- substrate concentrations range from about 20% of this value
nate or minimize nonlinearity of an assay in order to ensure to at least fivefold this amount. Further discussion of the
that meaningful results are obtained during enzyme charac- methods used to abstract kinetic constants by studies in-
terization. volving variation of substrate concentration is found in sec-
Nonlinear assays cannot be eliminated for some enzymes tion 19.4.
due to various types of autoinactivation events. For exam-
ple, some nonheme iron oxygenases react with oxygen to 19.3.5. Coupling Enzymes
yield an activated oxygenating intermediate that partitions Measurement of reaction products is often made easier by
between two pathways: catalysis and enzyme inactivation. adding an auxiliary enzyme that converts the primary prod-
The latter pathway may simply involve metal oxidation (a uct to a new material while simultaneously providing a use-
reversible process) or reaction with an active-site protein ful detection system, such as a convenient absorbance
side chain, thus inactivating the enzyme (28). In addition, change. The assay for phosphoglucomutase nicely illustrates
some enzymes must be chemically derivatized in order to this concept. Phosphoglucomutase catalyzes the intercon-
achieve an activated state (e.g., citrate lyase is acetylated) version of glucose-1-phosphate (glucose-1-P) and glucose-
and this modification may be lost during the assay. In such 6-phosphate (glucose-6-P), two compounds that are not
cases, the plot of product formation as a function of time easily distinguished by typical assay methods.
(also known as a progress curve) can be analyzed by use of
equation 1.
Glucose-1-P @ glucose-6-P
. (1 - e-f41nact)t )/k( inact) (1)
The enzyme is assayed in a reaction that initially contains
p, = only glucose- 1-P, with conversion to glucose-6-P followed
In this equation, P, is the accumulated product at time t, pi, spectrophotometrically by use of purified glucose-6-P dehy-
is the initial velocity, and k(inact) is the inactivation rate drogenase and NADPf. Glucose-6-P generated by phos-
constant. As illustrated in Fig. 1 (squares) the product con- phoglucomutase catalysis is immediately oxidized by the
centration increases over time but levels off at longer time dehydrogenase concomitant with NADP' reduction to
points. Data should be fit (e.g., using a commercially avail- NADPH, which is easily monitored in a continuous man-
able fitting program) to equation 1 to obtain an inactiva- ner as a change in A340(see section 19.3.6.1).
tion rate constant and the initial rates that are needed for Several conditions must be met for such coupling reac-
kinetic analysis. By contrast, efforts to obtain p,i by estima- tions to provide valid measurements of the rate for the first
tion of a tangent to the initial points will almost certainly enzyme. These are as follows: (i) the substrate(s) for the first
yield a low value. reaction must be in large excess (known as saturating con-
ditions and associated with zero-order reactions); (ii) the
19.3.3. Enzyme Concentration velocity of the second reaction must be linear with the con-
Fundamental to any assay is demonstrating a linear depend- centration of its substrate that is provided by the first reac-
ence of the observed reaction velocity on the concentration tion (resulting in what is termed a first-order reaction); and
of enzyme present. To assess whether the chosen assay con- (iii) neither reaction should be significantly reversible
ditions meet this criterion, varied amounts of enzyme are under the assay conditions. In the example cited above, this
added to otherwise identical assay solutions and the reac- would mean that excess glucose-1-P would have to be sup-
tion velocities are monitored. Typically, one observes a re- plied for the phosphoglucomutase reaction, that excess
gion showing direct proportionality between the measured NADP+ is provided for the coupling reaction, and that suf-
velocity and enzyme supplied, followed by a region of de- ficient glucose-6-P dehydrogenase is available so that the
clining increments in velocity with further enzyme in- velocity of this step is a direct function only of the amount
creases. The nonlinearity may arise from substrate deple- of glucose-6-P being produced by phosphoglucomutase.
tion, product inhibition, pH shifts, or other changes during Functional irreversibility of the two reactions is achieved by
the assay. Routine assays must be carried out in the linear having the dehydrogenase remove glucose-6-P as it is
region; thus, the enzyme solution must be diluted if this lin- formed and by monitoring the overall velocity before the
ear region is exceeded and a reassay must be performed. final products have accumulated appreciably. The require-
ment that the coupling enzyme be in excess can be met by
19.3.4. Substrate Concentration a trial-and-error process in which its concentration is in-
For normal assessment of enzyme activity and for calcula- creased until further additions have no effect on the mea-
tion of related kinetic constants, initial velocity conditions sured velocity of the overall reaction. If sufficient coupling
are essential and there should be a negligible decrease in the enzyme is not provided in the assay, especially if it exhibits
starting substrate concentration. In practice, substrate con- low affinity for the substrate produced by the first reaction,
sumption should not exceed 5% over the course of the mea- long lag phases may obscure the true enzymatic rate (see
sured assay period. This means that short time points cou- Fig. 1, triangles). O n the other hand, a large excess of cou-
pled to exquisite detection sensitivity may be needed for pling enzyme may be disadvantageous due to its expense
assays involving low substrate concentrations. High con- and to possible interference with the reaction of interest
centrations of substrate are utilized to measure the maxi- (e.g., by introducing an inhibitory substance along with the
mum velocities of the desired enzyme; however, inhibition coupling enzyme). A detailed discussion of the kinetic con-
by excess substrate concentration is not uncommon (see sequences of multiply coupled enzymes is found in refer-
section 19.4.1.3) so it is important to monitor the effects of ences 6 and 20.
a wide range of substrate concentrations. Examination of
the effect of substrate concentration also is required to 19.3.6. Choice of Assay Method
determine the affinity of an enzyme for a substrate.
Preliminary studies can define the approximate substrate 19.3.6.1. Spectrophotometry
concentration leading to approximately half-maximal rates; The sensitivity, wide availability, and ease of use of UV-visible
a series of assays should then be carried out in which the spectrophotometers have resulted in their prominence as a
19. Enzymatic Activity w 51 1

versatile tool for enzymatic analysis. As a general precau- NADH, phosphoenolpyruvate, and two coupling enzymes:
tion, assays must be performed under conditions where the pyruvate kinase and lactate dehydrogenase. The ADP gen-
light absorption is directly proportional to the concentra- erated by phosphofructokinase is cycled back to ATP by
tion of absorbing species. Optimal sensitivity and reliability pyruvate kinase, and the pyruvate formed in this reaction is
typically are achieved by designing assays to measure values reduced by lactate dehydrogenase. Contamination with
in the range of 0.1 to 1.5 absorption units, although some ATPase or NADH oxidase interferes with this spectropho-
instruments can extend this range. Several common types tometric analysis.
of assays based on spectrophotometry are illustrated below, Spectrophotometric assays also are useful for many non-
and additional details about the use of absorption spec- oxidoreductase reactions, as exemplified by an assay for the
troscopy are found in chapter 17 simple sugar hydrolase P-galactosidase (EC 3.2.1.23 [22]).
The reaction catalyzed by glucose-6-Pdehydrogenase (EC This is one of the most frequently assayed enzymes, albeit
1.1.1.49 ref. [ 191) shows why oxidoreductases are among the often qualitatively, because of the many lac2 gene fusions
easiest enzymes to assay by continuous spectrophotometric constructed. However, it is also easily assayed by a quite pre-
methods. With only minor variations, the following com- cise spectrophotometric procedure using the chromogenic
ments also apply to almost any dehydrogenase using substrate o-nitrophenyl-P-D-galactoside (ONPG) that is
NAD(P)+ as a substrate. The reaction measured is converted by the enzyme to galactose and o-nitrophenol.
The latter compound is faintly yellow, and at high pH, at
+
P-~-Glucose-6-P NAD(P)+ @
which the nitrophenolate ion dominates, a more intense
D-gluconolactone-6-P + NAD(P)H + H+ yellow is produced and can be measured at 420 nm. Quartz
When examined in the forward [NAD(P)+ reduction] di- cells are not required to monitor absorption changes at this
rection, assays are usually run at relatively high pH to con- wavelength, so glass or plastic cells can be utilized. An en-
sume protons and shift the equilibrium to the right. By con- zyme solution is incubated with a buffered solution con-
trast, the oxidation of NAD(P)H is normally run at a lower taining ONPG and other additives (e.g., 0.1 M sodium
pH since H+ is a substrate in that reaction. This adjustment phosphate [pH 7.01, 10 mM KC1, 1 mM MgS04, and 50
of the assay pH with reaction direction is particularly nec- mM mercaptoethanol), aliquots are removed at timed in-
essary when assays reveal a very short reaction period of lin- tervals, and 0.5 M Na2C03is added to adjust the pH to -1 1
ear product formation, indicative of an unfavorable equilib- and inactivate the @-galactosidase.The activity is deter-
rium; however, the choice of pH must also be dictated by mined by monitoring absorption changes at 420 nm and by
the stability of the enzyme (section 19.3.7.1). Assays run in using the extinction coefficient of 4.5 mM-'cm-' for o-
the forward direction typically use a quartz spectropho- nitrophenol at alkaline pH. It is important to include con-
tometer cell (preferably using a thermojacketed holder for trol reactions lacking ONPG (to correct for color due to the
precise temperature control) with a l-cm light path and in- sample) and without enzyme (to correct for the sponta-
volve the monitoring of absorption increases at 340 nm neous hydrolysis of ONPG). The ONPG assay can be
where the extinction coefficient of NADH or NADPH is adapted for use in permeabilized whole cells by adding 2
6.22 mM-' cm-'. Using this approach, a change of 0.10 op- drops of CHC13 and 1 drop of 0.1% sodium dodecyl sulfate
tical density (OD) unit/min corresponds to the production per ml of cell suspension. It is important to note that
of 16 FM NAD(P)H per min. Analysis in the reverse di- ONPG and related chromogenic compounds are not the
rection is facilitated by use of a 0.2-cm-path-length cell to natural substrates of the enzymes; however, data obtained
increase the allowable concentration of this substrate. For with these substances are generally useful for routine analy-
example, a 1 mM solution of NADH possesses an out-of- ses during protein purification and, depending on the sub-
range absorption of 6.22 in a l-cm cell, but can be moni- strate specificity of the enzyme, may be relevant to catalysis
tored in a 0.2-cm cell yielding an absorption of 1.24. When using the authentic substrates.
examined in this direction, a substrate-free control should Although artificial chromogenic substrates that yield
be included to detect background NAD(P)H oxidase activ- easily monitored colored products are very conveniently
ity. Solutions of NAD' and NADP+ should not be stored used (ONPG is just one of many examples), assays can also
at pH values over 7, since they are unstable under this con- be based on spectroscopic changes accompanying enzymatic
dition. Stock NAD( P)H solutions should be stored in alka- conversion of a substrate to product in reactions that do not
line solutions for enhanced stability. involve nicotinamide coenzymes. One such example is an
Oxidoreductases are often used in coupled assays, as al- assay for histidine ammonia-lyase (EC 4.3.1.3; usually
ready indicated in section 19.3.5, due to the ease of moni- called histidase), an enzyme that catalyzes the nonoxidative
toring the associated spectroscopic changes. To further re- deamination of L-histidine to form urocanate (imidazole
inforce this point, two coupled-enzyme methods are acrylate) and ammonia (27). Because it is unsaturated, uro-
described to assay phosphofructokinase (EC 2.7.1.1 l ) , canate production can be followed on the basis of its
which catalyzes the ATP-dependent phosphorylation of absorption at 277 nm, using an extinction coefficient of
fructose-6-phosphate to produce fructose-l,6-bisphosphate 18.8 rnM-'cm-'. Thus, an OD change of O.lO/min corre-
and ADP. In one approach, fructose-l,6-bisphosphatefor- sponds to a urocanate formation rate of 5.3 pM/min. A sim-
mation is coupled to NADH oxidation via three linking en- ilar approach can be used with other enzymes producing
zymes: fructose bisphosphate aldolase, triosephosphate iso- unsaturated products (e.g., fumarate or cinnamate) or con-
merase, and a-glycerolphosphate dehydrogenase ( 18). suming unsaturated substrates (e.g., protocatechuate and
Calculation of the units of phosphofructokinase present in other aromatic compounds).
the assay is based on the decrease in 340 nm absorption of
NADH and must take into account that for each mole of 19.3.6.2. Fluorescence
hexose-phosphate consumed, 2 mol of triosephosphate are Fluorescence spectroscopy can be used to monitor an enzyme
formed in the assay and thus 2 mol of NADH are oxidized. reaction, either continuously or discontinuously, as long
A second system to monitor phosphofructokinase activity as the substrate and product exhibit distinct fluorescence
requires the two substrates of the target enzyme as well as properties. For example, tryptophan hydroxylase converts
512 METABOLISM

tryptophan to hydroxytryptophan. Hydroxytryptophans flu- For oxidoreductases that catalyze oxygen uptake, polaro-
orescence excitation extends out to 310 nm, whereas that of graphic procedures are an excellent approach to measure
tryptophan is only at a wavelength of 280 nm. By monitoring activity. As an example, protocatechuate-3,4-dioxygenase
the 330-nm fluorescence emission properties using 300-nm (EC 1.13.11.3 [35]) can be assayed using a Clark-type oxy-
light, the enzymes formation of hydroxytryptophan can be gen electrode. This Fe-dependent enzyme catalyzes intra-
monitored (24). Direct detection of nicotinamide coenzyme diol cleavage of protocatechuate to form P-carboxy-&,cis-
reduction also is possible on the basis of its blue fluorescence. muconate. The assay simply involves filling a sample
Similarly, the decrease in yellow-green fluorescence of the chamber of a polarograph (containing a Clark electrode)
methanogen coenzyme F420,a 5-deazaflavin, can be used to with buffer that has been well equilibrated with air, addition
monitor its reduction. Analogous to the chromogenic assay of the substrate protocatechuate, recording of the O2uptake
approach described above involving such compounds as until a stable baseline reading is obtained, addition of a pre-
ONPG, fluorogenic substrates can be utilized. For example, cise volume of the dioxygenase, and measurement of the
4-methylumbelliferyl-linkedsubstrates may be used to assay initial rate of oxygen uptake. The rate (measured as
the cognate enzymes on the basis of the released fluorescent recorder units per minute) is converted to micromoles of O2
compound. Four drawbacks to the use of fluorescence include consumed per minute by conducting a calibration reaction
the more limited availability of the necessary instrumenta- in a similar fashion, but with a limiting and known amount
tion, interference by other compounds that absorb the inci- of protocatechuate. Variations of this procedure can be used
dent or fluorescently emitted light, complications introduced with a wide variety of oxygenases; however, it is important
by membranes or denatured proteins that scatter the incident to note that uncoupling reactions are observed in some en-
radiation, and lack of a quantitative unit of fluorescence zymes. Thus, the amount of oxygen consumed may be
comparable to an absorbance unit. Nevertheless, fluores- greater than the amount of primary substrate oxidized.
cence assays are widely used because of their ease and sensi- Control experiments examining oxygen consumption in
tivity, and they provide useful quantitative values if directly the absence of substrate are important to perform, but these
compared to an appropriate standard curve. may not account for all uncoupling activity since this side
reaction is sometimes stimulated by substrate.
19.3.6.3. Selective Electrodes
A variety of commercially available electrodes exhibit se- 19.3.6.4. Chromatography
lective responses to particular inorganic ions or gases and Many chromatographic approaches are available to allow
can be utilized in enzyme assays. For example, chloride ion- the separation of product(s) from initial reactant(s) for use
selective electrodes allow ease of monitoring dehalogenase in enzyme assays. Gas chromatography (GC) is the method
activities and an ammonia gas-sensing electrode facilitates of choice for volatile substances or compounds that are
assays involving deamination reactions. Electrodes are readily converted to volatile compounds by simple chemical
available specific for many compounds of interest (includ- steps. The already mentioned assay for nitrogenase utilizes
ing protons, oxygen, hydrogen, carbon dioxide, nitrate, ni- this approach to separate the two gases acetylene and eth-
trite, potassium, sulfide, and sodium). Each has a particular ylene from one another. Because high temperatures can be
working concentration range and interferences; thus, it is used during the separation, this technique also can be ap-
imperative to carefully study the literature accompanying plied to monitoring the transformations of less volatile sub-
the electrode of interest. One important consideration is re- stances such polyaromatic hydrocarbons or polychlorinated
sponse time, i.e., whether the electrode senses the rapid biphenyls during biodegradation studies. Quantification of
changes in target compound concentration during the reac- substances separated by GC can occur by flame ionization
tion (allowing use in a continuous assay) or whether a slow detectors or other means. More details about the principles
response dictates use in a discontinuous fashion. Additional of the method, instrumentation, and sample preparation
comments about the use of selective electrodes are found in and derivatization are found in chapter 17.
chapter 17. Two widely used electrode-based assays are de- In contrast to the gas-liquid partitioning of substances
scribed in the following paragraphs. associated with G C methods, thin-layer chromatography
Many biochemical reactions result in net proton uptake and various types of column chromatography separate com-
or release; thus, potentiometric measurement of pH is a pounds on the basis of their differential interactions be-
valuable assay approach. High-quality instruments permit a tween solid and liquid (either constant [isocratic] or vari-
resolution of 0.005pH units, so the pH of the assay solution able [gradient]) phases. The solid support can encompass a
need not shift appreciably during the assay; however, any wide range of compounds, with a corresponding wide range
significant change in pH can alter the activity of an en- of properties. Similarly, the liquid phases are carefully cho-
zyme. Another concern is that the observed pH changes sen to provide the best resolution. For example, the sub-
will be highly dependent on the concentration of buffer strate(s) and product(s) may be separated on the basis of
present. Yet another disadvantage of this assay is that the differential hydrophobicity by using a highly nonpolar CIS
readout of a pH meter isnot linear with proton concentra- column (a resin with bound alkyl groups) and eluting with
tion (recall that pH is the negative log of the hydrogen ion solvent of decreasing polarity, a process known as reverse-
concentration), so the data must be converted prior to ini- phase chromatography. Alternatively, separation may focus
tial rate determination. A n approach to at least partially on differences in charge (ion-exchange chromatography),
overcome these concerns involves continuous titration size (gel permeation chromatography), or more specific in-
while maintaining constant pH. In such a pH-stat system, teractions (affinity chromatography), with appropriate
the rate of added acid or base is monitored and reflects the changes in solvent as needed to enhance separation while
concentration of protons consumed or released. A n exam- allowing timely elution. Visualization of components sepa-
ple of this approach is found in the assay of urease (EC rated by thin-layer chromatography often utilizes quench-
3.5.1.5 [l]), an enzyme that hydrolyzes urea to yield two ing of a fluorophore that is impregnated in the support;
molecules of ammonia and one of carbonate, resulting in al- however, many alternative procedures are available such as
kalinization of the solution. those that specifically stain the samples based on selective
Next Page

19. Enzymatic Activity 513

chemistry of their reactive groups. Recent developments in autoradiogram. It is also possible to scrape the appropriate
scanning instruments have facilitated ease of quantification band of chloramphenicol-3-acetate from the plate and
of such results. Detection of components eluting from a col- quantify the product by scintillation counting. This assay is
umn may be based on UV or visible light absorption, often used for studying on/off regulation of a promoterless
changes in refractive index, fluorescence, or reactivity with CAT gene to detect the function of a suspected promoter
chemical reagents. Additional comments about the instru- sequence.
mentation available for these approaches are provided in Another method to measure CAT activity illustrates
chapter 17, and examples of methods for chemical analysis how radiolabeled samples can be used in coupled assays. In
are found in chapter 18. this case, the assay uses ['4C]acetyl-CoA and chloram-
phenicol. Because ['4C]acetyl-CoA is unstable, a cycling
19.3.6.5. Radioactivity system composed of ATP, CoA, phosphoenolpyruvate, ac-
Some assays utilize radiolabeled substrates that contain ra- etate kinase, phosphotransacetylase, and pyruvate kinase is
dioisotopes such as I4C,3H, or 32P. Each of these isotopes necessary to maintain its excess throughout the reaction
undergoes radioactive decomposition to release a p particle (30). Thus, acetate released by hydrolysis is activated by
(alternative types of radioactive decay occur for some other phosphorylation and transferred onto CoA in a process
isotopes), and this event can be very precisely quantified. driven by ATP, which is replenished by pyruvate kinase
By mixing aliquots of such radiolabeled samples with a using phosphoenolpyruvate. The labeled chloramphenicol-
"counting cocktail" containing scintillants (chemicals that 3-acetate is monitored after extraction into an organic sol-
emit light after colliding with a p particle), the number of vent (toluene) under conditions where ['4C]acetyl-CoA or
counts per minute (cpm) can be monitored by use of a scin- [I4C]acetate is not extracted.
tillation counter. O n the basis of the instrument's counting Radiolabeling studies are particularly well suited to as-
efficiencies for each radioisotope, the number of disintegra- says involving C 0 2 fixation by enzymes such as ribulose-
tions per minute (dpm) can be calculated. Investigators 1,5-bisphosphate carboxylase (EC 4.1.1.39 [lS]), which car-
using radioactive material generally undergo rigorous train- boxylates this substrate to form two molecules of
ing in the special procedures needed to ensure their safety 3-phosphoglycerate. A buffered ribulose bisphosphate solu-
and proper handling of materials and equipment during tion and radiolabeled bicarbonate are sealed within stop-
the experiment and to learn proper disposal methods. Ad- pered vials, enzyme is injected through the stoppers, and
ditional details about the types of scintillants available, in- the reactions are allowed to proceed for precisely measured
strument operation, statistics, problems involving proton time periods. The reactions are quenched by addition of a
exchange with solvent, and other complications are given 10% volume of 6 M acetic acid, which inactivates the en-
in chapter 17. zyme and converts the bicarbonate to gaseous COZ. The
The simplest types of assays using radioisotopes involve vial caps are removed in a chemical hood, and the vials are
selective extraction or precipitation of a radiolabeled sub- allowed to stand open for at least 30 min to remove unre-
strate or product. Such a procedure is commonly used to assay acted radiolabeled substrate. The remaining radioactivity,
aminoacyl-tRNA synthetases, enzymes catalyzing essential associated with product, is measured by addition of scintil-
ligase reactions that activate amino acids for protein synthe- lation fluid and counting.
sis. For example, the assay for glutaminyl-tRNA synthetase Complementary procedures are available to monitor
(EC 6.1.1.18 [9]) involves incubation of the ''C-labeled C 0 2release by using radiolabeled substrate. In this case, the
amino acid with tRNA (a mixture or the specific species), substrate must be purchased or synthesized with I4C incor-
MgATP, buffer, and sample containing the enzyme for varied porated into the carbon that is lost as C02. Assays are run
time periods. The mixtures are treated with 5% trichlor- using multiple enclosed vials, each of which contains a sep-
oacetic acid, resulting in precipitation of the tRNA species arate chamber containing a piece of filter paper soaked in
(both free and containing covalently bound glutamine), and basic solution. The reactions are quenched at predeter-
the precipitate is collected by filtration, washed, dried, and mined time points by injection of acid; acidification of the
counted. A time course should exhibit linear increases in the assay mixture causes the generated I4CO2to transfer to the
amount of radioactivity collected on the filters. basic filter paper that subsequently is mixed with scintilla-
Use of radioisotopes also can increase dramatically the tion fluid and counted. This approach is illustrated in chap-
sensitivity of some chromatography-based assays. Typically, ter 17.
tracer levels of the specifically labeled substrate (commer-
cially available or synthesized to contain a known amount 19.3.6.6. Chemical Derivatization
of radioisotope per known amount of sample) are mixed Discontinuous colorimetric assays are widely used to moni-
with unlabeled material prior to use. Labeled substrate is in- tor enzyme reactions in microbiology. In this approach,
cubated with the enzyme of interest, aliquots are removed at aliquots of a reaction mixture are removed at timed inter-
timed intervals, the product is separated from the remaining vals and the product (or remaining substrate) is chemically
substrate (e.g., using one of the chromatographic methods derivatized to form a colored species. Specific methods to
described above), and the radioactivity associated with one quantify inorganic phosphate, ammonia, nitrate, nitrite,
or both of the components is measured. A n assay for chlor- and several other compounds are described in chapter 18
amphenicol acetyltransferase (CAT) (EC 2.3.1.28) pro- and can be incorporated into enzyme assays. Two specific
vides an example of this approach. This enzyme, responsible examples involving chemical derivatization are described
for the resistance of many bacteria to chloramphenicol, below, one that strictly requires a discontinuous assay and a
acetylates the substrate by using acetyl-CoA to form chlor- second that can be used in a continuous assay.
amphenicol-3-acetate plus free C o A thiol. Thin-layer A n assay for ribosephosphate isomerase (EC 5.3.1.6), a
silica gel chromatography separates ['4C]chloramphenicol key enzyme of the pentose phosphate pathway and a com-
from the labeled acetylated product. The thin-layer support ponent of the Calvin carbon reduction cycle, makes use of
can be exposed to X-ray film for autoradiographic analysis, the reddish purple color derived by reaction of ribulose-5-
using a densitometer to quantify the results by scanning the phosphate with a cysteine-carbazole reagent (17). Enzyme
Permeability and Transport
ROBERT E. MARQUIS

20.1. PERMEABILITY .......................................... 528


20.1.1. Assay Procedures..................................... 528
.............................
20.1.1.1. Washing of Cells 528
. .........................
2. Centrifugation of Cells 528
.
3. Measurement of Pellet Weight and Volume .......... 528
.4.
Mixing and Incubation of Cells with Solute .......... 529
.
5. Recentrifugation ............................. 529
.
6. Determination of Solute Concentration ............. 529
.
7. Determination of Interstitial Space ................ 529
.
8. Expression of Results .......................... 530
.
9. Cell Volume Determination ..................... 530
.10. Cell Water Content Determinations ............... 530
. . ................
11 Live-Dead Stains and Permeability 530
20.2. TRANSPORT ............................................ 530
20.2.1. Assay Procedures ..................................... 530
20.2.1.1. Preparation of Cells ........................... 530
.
2. Mixing and Incubating Cells with Solute ............ 531
.
3. Sampling and Separation ........................ 531
.
4. Washing of Cells ............................. 531
.
5. Assay of Cells for Content of Transported Solute ...... 531
.
6. Expression of Results .......................... 531
.
7. General Considerations ........................ 532
20.3. SPECIAL METHODS ...................................... 532
20.3.1. Proton Motive Force. Intracellular pH, and Proton Permeability... 532
.
2. Buffer Capacities of Cells ............................... 534
20.4. OSMOTICALLY SENSITIVE CELLS ........................... 534
20.5. SOLUTE UPTAKE BY BIOFILMS ............................. 535
20.6. MEMBRANE VESICLES AND LIPOSOMES ..................... 536
20.7. PERMEABILIZED CELLS ................................... 537
20.8. REFERENCES ............................................ 537

Two general types of processes for entry and exit of solutes curve is parabolic with a plateau at Cin= C.... i.e., when
across cell membranes can be distinguished. Permeation the internal concentration equals the external concentra-
denotes passive movement of a solute into (uptake) or out tion or. more correctly. when the electrochemical activity
of (efflux) a cell by diffusion to diminish the electrochem- of the solute is the same on both sides of the membrane.
ical potential of the specific solute across the cell mem- For such a curve. Cin= (maximum Cin ) (1 - Ckt). where
brane . Solute passage may be facilitated by chemical inter- k is a constant and t is the time . Diffusion can often be
actions with cell structures. say. within the cell membrane. speeded up by interaction of solutes with membrane struc-
Permeability is a property of the cell and not of the solute. tures. and the process is then called facilitated diffusion .
The Fick diffusion equation can be applied to permeation However. even for facilitated diffusion the electrochemi-
of a solute through the membrane of a cell: cal potentials inside and outside become equal at equi-
librium.
dsldt = PA(AC)
Transport denotes active movement of a solute into or
where ds/dt is the change in amount of internal solute per out of a cell or organelle by means of specific transport sys-
unit of time. P is the permeability coefficient (which has tems which are coupled to metabolism and involves ener-
units of distance divided by time). A is the surface area of gized carriers. Two well-studied general types of microbial
the membrane or other structure across which diffusion oc- transport systems are the phospheno1pyruvate:sugar phos-
curs. and AC is the difference in solute concentration (or photransferase system (PTS) (53) and various permease
electrochemical potential if the solute is ionized) between systems (17.43). The PTS functions in facultatively anaer-
the interior and the exterior of the cell or membrane- obic bacteria for transport of sugars. while essentially all
enclosed organelle. If the internal concentration (Cin) is bacteria have permease systems for transport of mineral
plotted as a function of time after exposure of the cell to a cations. inorganic anions. amino acids. inorganic acids. nu-
higher environmental level of the solute. the resulting cleic acid precursors. sugars. and a wide variety of other

527
528 METABOLISM

solutes. The general regulatory roles of the PTS have be- than for energized transport coupled to ATP hydrolysis or
come more and more appreciated, and in some organisms Ap across the cell membrane.
its functions are more for regulation of metabolism than The space technique is designed specifically to give an
for transport. Permease systems may be energized by ATP, estimate of the fractional space or volume of cells pene-
for example, ATP binding cassette systems, or by the pro- trated by the test solute in the absence of energized trans-
ton motive force across the cell membrane. In addition, port. The technique is based simply on determination of the
P-ATPases may act as solute transporters, for example, degree of dilution of a solution containing the solute as a re-
Ca-ATPase, and some F-ATPases may transport Naf sult of mixing with a pellet of centrifuged cells. The follow-
rather than H'. ing information is needed: the initial volume and concen-
A curve relating solute uptake to time for a specific trans- tration of the test solution, the volume of the cell pellet,
port process does not differ qualitatively from one for a gen- and the final concentration of test solute in the extracellu-
eral permeation process. However, active or energized trans- lar phase of the mixture. Cell pellets contain not only cells
port does not stop when Ci, = C,,,but continues often until but also interstitial (intercellular) space. The fractional vol-
a substantial concentration gradient has developed and the ume of the interstitial space is generally estimated by use of
process has become one of concentrative transport. Max- high-molecular-weight polymeric solutes, which can pene-
imum uptake is then determined by the rates of the inward trate the interstitium but not the cell wall.
and outward flows, but any maintained gradient of electro- The steps involved in the space technique are described
chemical potential requires continued energy coupling. below.
As indicated, transport results in net movement of
solutes into or out of cells, that is, uptake or secretion. For 20.1 .I . I . Washing of Cells
I

many solutes, a transport process of so-called exchange dif- After harvest, cells generally need to be washed before use
fusion occurs in which there is a one-for-one exchange of for permeability assays. A single wash with a large volume
internal and external solutes across the membrane. This ex- of fluid is generally sufficient to remove medium con-
change is generally assessed by use of radioactively labeled stituents without depleting most types of bacteria, archaea,
solutes; details of the technique are presented by Maloney or fungi of internal solutes. Two washings may be done, but
et al. (35). Exchange may involve antiporters and heterolo- normally not three. Gram-positive bacteria are osmotically
gous exchange. For example, Driessen et al. (16) have char- robust and can be washed with water or dilute buffers with-
acterized the arginine/ornithine antiporter for the arginine out harm, especially when they are chilled. Other organ-
deiminase system of Lactococcus lactis. The antiporter cat- isms, particularly gram-negative bacteria and many archaea,
alyzes exchange of ornithine, the product of the system for are more sensitive and can be damaged by water washing.
arginine, the substrate of the system, without need for ex- Commonly used wash solutions for gram-negative bacteria
penditure of ATP. Concentrative solute uptake does not contain magnesium ions (1 to 10 mM), a buffer, sometimes
necessarily require a membrane with transport catalysts. a n osmotic stabilizer (e.g., 0.4 M KC1 or 0.5 M sucrose), and
Ion-exchange resins, for example, can take up and concen- sometimes an agent protective against denaturation, such as
trate ions selectively. Solutes, then, can be taken up and re- glycerol or another compatible solute. Maloney et al. (35)
tained as they would be by an ion-exchange resin. In fact, recommended using growth medium 63 with deletion of the
most of the mineral ions in cells are counterions for charged carbon source for washing Escherichia coli cells. With any
groups of macromolecules in solid phases, e.g., cell walls, previously untested organism, it is worthwhile to test a
membranes, ribosomes, nuclear bodies, organelles, etc. number of washing solutions to determine the best one. The
criteria for efficacy are that the cells should be freed of
medium constituents but retain internal pools of potassium,
20.1. PERMEABILITY amino acids, or Pi. No wash solution is perfect, and some
compromise must be accepted. When dense suspensions or
20.1 . I . Assay Procedures biofilms are used, the composition of the wash fluid is less
A standard technique for assessing solute permeation into critical than when dilute suspensions of cells are used.
bacterial cells or cell structures is the space (or thick-sus-
pension) technique described by Conway and Downey (10) 20.1 .I .2. Centrifugation of Cells
and modified by Mitchelland Mgyle (40) and MacDonald Harvested cells need to be centrifuged to obtain a tight pel-
and Gerliardt (33) and others. Large masses of cells are used let, and the details of centrifugation depend on the particu-
for the assay-generally 3 g, wet weight, or more per assay lar organism. A packing curve of the type shown in Fig. 1
tube. Obtaining this quantity of cells can be difficult but can be prepared by plotting pellet weight against centrifu-
not when common, easily grown organisms are used. gation time, using a series of cell suspensions to obtain the
Techniques that require less biomass and are based on use of curve. The centrifugation time chosen should be well out in
membrane filters or a high-speed microcentrifuge are de- the plateau region so that minor variations in centrifuga-
scribed in the section on transport (section 20.2). For any tion time do not affect pellet weight. The final centrifuga-
procedure, careful attention should be paid to details of tion before solute addition and mixing is generally done
growth and harvest because permeability may change dur- with a tared tube so that pellet wet weight can be obtained
ing the culture cycle in batch cultures or as a result of wash- by assessing change in weight. Prior to weighing, super-
ing procedures used to rid cells of medium constituents. natant fluids should be decanted carefully, the tubes in-
There can be an advantage in using cells obtained from verted to allow drainage before careful wiping with ab-
continuous cultures. It is usually best to take large harvests sorbent tissue. Attempts to blot the pellet surface are best
from the main vessel rather than using so-called runoff cells, avoided because cells can be removed with the liquid.
which may have altered permeability because of storage on
ice during collection. Changes in growth medium, growth 20.1 .I .3. Measurement of Pellet Weight and Volume
temperature, pH, etc., can also be expected to alter perme- Pellet wet weight is usually an adequate indicator of pellet
ability, although alterations will generally be less severe volume and may be used instead of volume in calculations
20. Permeability and Transport w 529

ogy. The cytocrit method gives useful volume estimates, but


it has difficulties in practice and requires use of a swinging-
bucket centrifuge head.
20.1 .I .4. Mixing and Incubation of Cells with Solute
The cell pellet to be used for a permeability assay is brought
to the chosen experimental temperature and mixed with
test solute in solution. Mixing can be enhanced by vortex-
ing the material. However, care must be taken when work-
ing with samples of sensitive organisms, such as anaerobes
or spheroplasts. The volume of solution should, in general,
be about equal to the pellet volume because there should be
sufficient dilution of the solution to allow changes in con-
centration to be readily assessed. After mixing, the suspen-
sion should be incubated for a time sufficient to allow diffu-
sion equilibrium to be reached even in clumps of cells, say,
5 to 15 min depending on the tendency of the cells to
clump. Vortex mixing may help to reduce clumping.
20.1 .I .5. Recentrifugation
After appropriate incubation, the mixture should be cen-
trifuged sufficiently to obtain a cell-free supernatant fluid
for solute assay. The supernatant fluid may require an addi-
tional centrifugation in the absence of the main pellet.

--
20.1 .I .6. Determination of Solute Concentration
The concentration of solutes should be assayed for the ini-
tial solution and for the supernatant fluid after recentrifu-
gation. Sometimes the assay can be a gravimetric one based
simply on weighing the material dried from known volumes
Time of centrifugation of the solutions or, alternatively, by means of refractometry.
Unfortunately, these techniques are not chemically specific
FIGURE 1 Typical packing curve for a cell pellet. and material leached from the cells may interfere.
Therefore, it is usually necessary to use specific chemical or
radiochemical assays to assess solute concentration.
because wet densities of vegetative cells are generally about
1.02 to 1.04 g/ml. However, bacterial spores have signifi- 20.1 .I .7. Determination of Interstitial Space
cantly higher wet densities, as high as 1.22 g/ml for Since cell pellets contain intercellular or interstitial space
Geobacillus stearothemophilus spores, and pellet weight is into which solutes can diffuse readily, there is a need to as-
not a good substitute for pellet volume. Certain inclusion sess the volume of this space. The assessment is usually
granules, e.g., poly-P-hydroxybutyrate, of vegetative cells made with solutes of high molecular weight, which are able
also may have high wet densities and may increase the dif- to penetrate the interstitiurn but cannot pass through the
ference between pellet weight and volume. cell wall or outer membrane. Dextrans with average molec-
Pellet volumes can be estimated also with the use of a ular weights of 2,000,000 (Dextran T2000; Pharmacia-
standard pycnometer or a volumetric flask. For example, to Pfizer Fine Chemicals, Piscataway, NJ) serve the purpose.
assess the volume of a 3-g wet-weight pellet, one can mix the Dextrans are polydispersed, but even the smallest molecules
pellet with water, transfer it to a 50-ml, tared pycnometer in the distribution of this large dextran preparation do not
vessel or volumetric flask, and add water until the mixture penetrate even relatively porous cell walls. Dextran con-
comes up to the mark. Suppose the weight of the suspension centrations can be determined gravimetrically, by means of
determined with an analytical balance is 50.110 g (weight refractometry, or chemically by the phenol-sulfuric acid
mixture plus pycnometer minus weight of pycnometer) and method (18).
the pycnometer has a calibrated volume of exactly 50.000 ml A number of other polymeric materials have been used
at 25"C, the temperature of weighing. Suppose that the pel- instead of dextrans. Proteins, such as serum albumin, have
let was found to weigh exactly 3.000 g. Then, the weight of an advantage in being nearly monodispersed and easy to
water added to the pellet must be 50.1 10 -3.000, or 47.1 10 assay. However, they have a disadvantage in that they may
g. A t 25"C, this weight of water would have a volume of bind to cells. Inulin has also been used often but may par-
47.249 ml based on water density at that temperature ob- tially penetrate bacterial cell walls. The major need is for an
tained from tables in a standard chemical handbook. easily assayed polymer that does not bind to cells.
Therefore, the pellet volume must be 50.000 -47.249, or The interstitial volume fraction may be an appreciable
2.752 ml. The density of the pellet is then 3.000 g/2.752 ml, part of the total pellet volume. For closely packed, nonde-
or 1.090 g/ml. Once the density is known, and if standard formable spheres, the interstitium makes up some 27% of the
centrifugation conditions are used, the pellet volume can be total volume. For bacterial cells packed by centrifugation to
readily calculated from the pellet weight. essentially constant volume, the interstitium may represent
Pellet volumes also have been estimated directly by use only some 10 to 15% of the pellet volume because of irregu-
of Calibrated centrifuge tubes, which yield cytocrit data sim- lar shapes and sizes of the cells and their deformability. If
ilar to the hematocrit data obtained routinely in hematol- solute uptake is not great, uptake into the interstitium may
530 METABOLISM

be a large part of the total uptake. However, for solutes that from knowledge of the specific volume of water at the tem-
are concentrated by cells, uptake into the interstitium will perature of the measurements, generally, about 1 ml/g.
be a smaller part of the total uptake. Since the volume of interstitial water in centrifuged pellets
can be estimated in terms, say, of dextran-permeable vol-
20.1 . I .8. Expression of Results ume, the amount of cell water in the pellet can also be esti-
A commonly used permeability index is the space value, mated.
or S value, which can be calculated by use of the formula. Sometimes, it is convenient to estimate the amount of
cell water directly from the R value for solutes such as 3 H z 0
or [14C]ureaor glycerol, which readily permeate but are not
where V, is the volume of solution added to the pellet, V,, is normally concentrated by resting cells, although many cells
the volume of the pellet or, commonly, the pellet wet do have energized transport systems for urea or glycerol.
weight (W,,), C,is the initial concentration of solute, and Tritiated water has an advantage in terms of its speed of
C,is the final concentration. S values indicate the fraction entry into cells, but during prolonged incubations, tritium
of the cell pellet penetrated by the solute. However, the de- exchange can occur between labeled water and cell compo-
sired information is the fraction of the cell volume pene- nents. There is value for many studies in estimating the vol-
trated. The cell volume fraction penetrated is called the R ume of cell wall water, which is outside the cell membrane
value. It is calculated by use of the formula but within what would ordinarily be considered the outer
wall boundary of the cell. Small solutes penetrate readily
into the heteroporous cell walls of most gram-positive or-
where Ssolis the corrected space value for the solute being ganisms (51) but may not penetrate the outer, cell wall
tested and Sinr is the volume of the interstitial space; if membrane of gram-negative bacteria. Cell wall water is gen-
dextran is used to measure the volume, Sdextran is used in- erally assessed in terms of the cell volume or space perme-
stead of s,,,.A zero value for R indicates that the cells are able to small solutes not transported across the cell mem-
impermeable to the solute. Values of zero to one indicate brane of the particular cells used minus the interstitial
various degrees of penetration, and values greater than space. Sucrose and raffinose have been used for this purpose
one indicate that the cells actually concentrate the solute. for cells without sucrose or raffinose transport systems or
It is often desirable to express results in terms of cell dry with nonmetabolizing, chilled cells. Radioactively labeled
weight or cell water, e.g., sucrose-permeable space per taurine also has often been used, for example, by Tran and
gram of cell dry weight or per milliliter of cell water. The Unden (56),for assessing the cell wall water volumes of
pellet volume impermeable to small solutes such as sucrose cells of E. coli. The only requirements for a useful probe are
can also be readily estimated, and if the cell membrane is that it should be small and able to penetrate the wall water
impermeable to the small solute, then the, say, sucrose-im- volume, that it is not strongly bound by wall components,
permeable volume is approximately equal to the protoplast and that it is not transported across the cell membrane.
volume.
20.1 .I .I 1. Live-Dead Stains and Permeability
20.1 .I .9. Cell Volume Determination The use of live-dead stains has become increasingly popular,
Useful cytological information can be gained from space especially for microscopic estimations of cell viability under
values. For example, the technique offers the most accurate stress conditions or in biofilms. The most commonly used
and direct means available for determining average cell size. stain is propidium iodide, which can be obtained from Mo-
The pellet volume minus the interstitial volume is equal to lecular Probes (Eugene, OR). The differentiation between
the volume of cells. If the pellet is resuspended in water to live and dead cells is based on live cells with intact mem-
some known volume, say, 50 ml, in a volumetric flask, the branes being impermeable to propidium iodide, while dead
number of cells can be assessed with use of a Petroff-Hausser cells with damaged membranes are permeable to the dye.
counter, and the average volume of a cell can then be calcu- There are questions regarding the relationship of increased
lated. The technique can also be used to estimate the vol- cell permeability and capacity to, say, grow on agar plates to
ume of the cell protoplast if the cell wall volume or the space yield a colony. However, live-dead staining methods do
impermeable to a small solute that cannot cross the cell offer a convenient way for obtaining microscopic assess-
membrane is known. ments of cell viability.
Cell counting with the Petroff-Hausser chamber has
high levels of inherent error, as does plate counting, because
of the small numbers of cells or colonies counted and the 20.2. TRANSPORT
Poisson distribution of possible counts (28). Electronic
counters capable of counting large numbers of cells per sam- 20.2.1. Assay Procedures
ple can be used to obtain more accurate estimates of cell
numbers. Cells are counted by electronic counters on the 20.2.1 . I . Preparation of Cells
basis of differences in electrical conductivity between cells For studies of rates of solute uptake, some means of rapidly
and their suspending medium. Manufacturers of electronic separating cells from their suspending medium is required.
cell counters include Beckman Coulter (http://www Separation is often accomplished by filtration through
.beckman.com/). membrane filters, and therefore, it is necessary to use dilute
cell suspensions, generally with less than 200 Fg (dry
20.1 .I .lo. Cell Water Content Determinations weight) per ml, if 1-ml samples are to be filtered through
The resuspended cells can also be used for dry-weight de- standard 22-mm-diameter filters. The use of large filters al-
terminations from which one can estimate the dry weight lows for use of more concentrated cell suspensions but can
per cell. The weight of cell water is then equal to the dif- present problems in terms of rapid manipulations.
ference between the wet weight of the cell and its dry As with permeability assays, care must be taken in trans-
weight, and the weight can be converted to volume simply port assays to control closely the conditions of cell growth
20. Permeability and Transport 531

and harvest. Wash and resuspension media tend to be more It is often possible to stop transport processes by adding
important in preparing cell suspensions for transport assays samples directly to iced or chilled wash medium.
because cells in dilute versus concentrated suspensions are Centrifugation can then be carried out in the cold. For this
more liable to be harmed by unfavorable environmental method to be useful, chilling must stop influx but not
conditions, including those leading to leaching of internal induce efflux. Satisfactory results are more likely to be ob-
solutes from the cells. tained with gram-positive than with gram-negative organ-
For transport studies, cells often have to be precondi- isms.
tioned before addition of the substrate to be transported. Sometimes, it is not necessary to separate the cells from
For example, they may be starved to deplete endogenous the test solution. For example, ion-specific or solute-specific
pools of substrate, which may be of advantage when ra- electrodes can allow for continuous monitoring of pH, par-
dioactive substrates are used to reduce levels of endogenous tial pressure of oxygen (pOz),potassium, or other metal ions
unlabeled substrate and the need to make major corrections or organic solutes. In studies of salt uptake, it may be possi-
in estimated specific radioactivity. Alternatively, they may ble to assess uptake simply by assessing changes in conduc-
be fed to allow for synthesis of ATP and establishment of Ap tivity of the suspension measured with a conventional con-
(the proton motive force) across the cell membrane. The ductivity cell and meter.
physiology of many microbial cells is set to maintain pools
of energy transfer substrates such as ATP or phospho- 20.2.1.4. Washing of Cells
enolpyruvate. Cells retained on filters or pelleted must generally be
washed to remove adherent or interstitial suspending
20.2.1.2. Mixing and incubating Cells with Solute medium. For gram-negative organisms particularly, the
The cell suspension and test solution are brought to the ex- wash liquid should be osmotically buffered to avoid leach-
perimental temperature separately and then mixed at zero ing of solutes from the cytoplasm. Concentrated sugar solu-
time. Care has to be taken to avoid cells being severely tions, e.g., 0.5 M sucrose solution, are often used for wash-
shocked during mixing. For example, there should not be ing. Magnesium ions, usually 1 to 10 mM, may also be
major changes in osmolality or ionic strength associated added as chloride or sulfate salts. A n ideal wash solution
with mixing arising from differences between the suspend- should remove test solute from interstitial and adherent sus-
ing medium for cells and the solution containing the test pending medium during the first wash but not appreciably
solute. Rapid mixing is often required because of the rapid- in subsequent washes. Some compromise generally has to be
ity of transport processes, and incubation of the mixtures is accepted. As indicated above, washing can be avoided by
generally of short duration, seconds or minutes rather than centrifuging cells through silicone oils.
hours.
20.2.1.5. Assay of Cells for Content
20.2.1.3. Sampling and Separation of Transported Solute
Samples of the reaction mixture are withdrawn shortly after Cells may be eluted from membrane filters, resuspended in
mixing. The cells are rapidly separated from the suspending a known volume of fluid, and assayed for the test solute by
medium, for example, by filtration through membrane fil- chemical, radiometric, or other methods. When radioactive
ters with 0.45-pm-diameter pores, previously wetted with solutes are used, the membrane filters can be placed directly
wash solution. Lower-porosity filters may be used, but they into scintillation fluid for counting. The amount of solute
have more resistance to fluid flow with resultant slower fil- incorporated into polymeric materials can be estimated by
tration. The 0.45-pm-pore-size filters are effective for most treating the cells on the filter with cold, 5% (wtlvol)
bacterial cells. Vacuum from laboratory lines is often suffi- trichloroacetic acid solution and then washing them with
cient for rapid filtration, although vacuum pumps offer an water before counting. The acid treatment damages the cell
advantage for developing higher levels of vacuum. The membrane so that small solutes but not macromolecules are
common, clip-on, vacuum-funnel attachment can be re- released from the cells. The acidification results also in pro-
placed by custom-made, heavy brass rings to hold the filter tonation of carboxyl and phosphate groups with release of
membrane in place and allow for more rapid processing of bound cations.
samples. Also, convenient manifold filter units can be pur- Membrane filters may bind test solutes, especially ion-
chased if many samples have to be processed in a short time. ized ones, and corrections to uptake values may be needed
There may be an advantage to mixing assay components because of this binding. A n estimate of the extent of bind-
in a syringe with a Swinney filter attachment from which ing can be made by passing some of the test solution
samples of cell-free suspending medium can be expressed. through a membrane filter, washing the filter in the same
The obvious disadvantage of the method is that calculation way the cells are washed, and then assaying the filter for the
of solute uptake depends on estimation of changes in solute retention of the test solute.
concentration in the suspending fluid rather than changes
in solute levels in the cells. Moreover, repeated removal of 20.2.1.6. Expression of Results
suspending fluid changes the ratio of cells to suspending Transport is usually expressed in terms of uptake or excre-
medium in the remaining mixture. tion in molar amounts per unit of biomass, e.g., cell dry
Often, rapid centrifugation can be used to separate cells weight. It may also be expressed per unit of cell water, espe-
and suspending medium, even when rate data are required. cially if there is a desire to determine if transport is concen-
Microcentrifuges, which accommodate small tubes of 250- trative and, therefore, requires some type of energetic cou-
to 1,500-pl capacity, can attain centrifugal forces of 12,000 pling. Results may also be expressed per cell. However,
x g in only a few seconds. Another commonly used means there is then the problem reviewed earlier of obtaining an
to separate cells from their suspending medium is to cen- accurate cell count. If cell numbers are determined by plate
trifuge them through silicone oils (22). A n advantage of counting, transport can be expressed in terms of CFU,
this method is that very little interstitial aqueous solution is which may or may not be the same as total cell count.
pelleted with the cells and washing is less of a problem. When using radioactive solutes, data should be converted
532 METABOLISM

from counts per minute, curies, or becquerels to molar suspensions in which cells may be highly sensitive to
equivalents. changes in environmental parameters.
Transport processes are generally viewed as a subclass of
enzyme-catalyzed reactions, and the principles and prac-
tices of enzyme kinetics can be applied to transport kinet- 20.3. SPECIAL METHODS
ics. For example, Lineweaver-Burk, double-reciprocal plots
of the inverse of transport rate versus the inverse of solute 20.3.1. Proton Motive Force, lntracellular pH, and
concentration often yield straight lines from which one can Proton Permeability
calculate values for K,, (Michaelis constant) and V,, The proton motive force is considered to be a major pa-
(maximum velocity). Alternatively, Eadie-Hofstee plots of rameter related to energy transfer in biological systems. I t is
velocity of transport versus the velocity divided by substrate defined as
concentration can be used for estimating kinetic parameters
and substrate affinities. The complications of kinetics that
AP = A$ - 2.3(RT/F) (ApH)
arise in the study of enzyme reactions arise also in studies of where AD is the proton motive force, A+ is the membrane
transport. potential, R is the gas constant (8.3 18 volt-coulomb/K
It is not uncommon for cells to have more than one charge equivalent), T is the temperature (kelvin), F is the
transport system for a particular solute. For example, many faraday (96,500 kcal/charge equivalent), and ApH is the
bacteria have a high-affinity system that is highly specific difference in pH between the interior and the exterior of
for an individual aromatic amino acid, in addition to a low- the cell. At 25C, the value of 2.3 RT/F (which is often
affinity system that can catalyze transport of a variety of given the symbol z) is approximately 59 mV. Thus, to esti-
amino acids. Methods for assessing these multiple systems mate AD, it is necessary to estimate A+ and ApH separately.
are described by Ames (2). Techniques for estimating these parameters have been re-
In addition, many solutes can enter cells passively by dif- viewed critically by Kashket (26).
fusion (permeation) as well as actively by energized trans- A$ for giant cells of E. coli has been assessed directly by
port. The diffusion component generally does not show sat- means of microelectrodes (19), but the method is not ap-
uration at high substrate concentrations. Its extent can be plicable to ordinary bacterial cells. A+ is usually assessed in-
estimated from plots such as the one shown in Fig. 2. With directly from determinations of the distribution of permeant
intact bacteria, the diffusion component may include diffu- cations between the interior and the exterior of cells.
sion of solute into the water of the cell wall (36), which can Harold and Altendorf (21) indicated that a useful indicator
account for some 50% of the cell volume of organisms such of A$ must diffuse rapidly across the membrane . . . be
as Micrococcus luteus. fully dissociated by physiological pH, metabolically innocu-
ous and not subject to translocation by a biological trans-
20.2.1.7. General Considerations port system. These criteria can be met by Kf or Rb+ in
A major advantage of the use of dilute compared with dense cells treated with 1 to 10 p M valinomycin to render the cy-
suspensions of cells is the ease with which kinetic data can toplasmic membrane permeable to the cations. The mea-
be obtained. Also, with dilute suspensions, closer control of sured membrane potential of bacterial cells is generally
solute concentration, pol, pH, ionic strength, etc., can be about 180 mV with the interior negative, so the K+ con-
maintained. The disadvantages include problems associated centration in the cytoplasm would be some 20 times that in
with solute leaching from cells during initial suspension or the suspending medium. Potassium concentrations can be
washing, the general need to assess solute uptake or loss determined by flame photometry or with ion-specific elec-
from changes in intracellular rather than extracellular con- trodes. The uptake of Kf can be assessed by use of the
centrations, and the inherent vagaries in the use of dilute space-value method, and the concentration can be ex-
pressed in terms of moles of K+ taken up per unit of cell
water. 86Rb+can be used for estimation of Rb+ uptake.
A$ has often been assessed by determining the distribu-
I 1 tion of permeant, lipophilic organic cations or fluorescent
dyes. Kashket (26) recommended tetraphenylphosphonium
(TPP+) for assessing A+ and describes basic procedures for
its use. There have been concerns about the use of TPPf and
similar compounds in many organisms since Midgley (39)
pointed out that E. coli has an efflux system with broad
specificity for complex organic cations, including TPP+. In
fact, these sorts of multidrug transporters occur widely
among microbes and cause concerns about the uses of dyes
for A$ estimates. The use of K+ or Rbf with valinomycin,
then, has an advantage for studies of organisms with mul-
tidrug transporters. Improved methods to determine both
ApH and A$ for Methanobacterium thennoautomophicum have
been developed (11) based on the pH-dependent fluores-
ExtracelI uIar solute Concentration cence of coenzyme F420to assess ApH and the fluorescent
probe bis-( 1,3-dibutylbarbituric acid)trimethine oxonol to
FIGURE 2 Graphic estimation of the diffusion component assess A$. Fluorescent dyes from Molecular Probes have also
in the total uptake of a solute by microbial cells. The transport been used (9) for assessing A$, but again, there is a need to
uptake is the total uptake minus the uptake due to passive dif- know if they can be transported by multidrug transporters.
fusion. The uptake measure can be either the rate or extent of For determinations of ApH, a weak acid or base to which
uptake. the cell is permeable is used. The compound chosen must
20. Permeability and Transport H 533

dissociate at physiologic pH values so that its distribution A flow dialysis method that does not require separation
between the cell and its environment will depend on the of cells and suspending medium has been described in detail
pH difference between the two phases. When a weak acid is by Ramos et al. (45,46) and applied in studies of membrane
used, the membrane of the cell should be permeable to the vesicles. Methods have been described by Avison et al. (3)
undissociated form but impermeable to the ionized or disso- for estimation of intracellular pH for bacterial cells by use of
ciated form. Compounds that are commonly used for ApH magnetic resonance techniques. pH, has been assessed by
estimates include acetate, benzoate, 5,5-dimethyl-2,4- using fluorescent dyes such as 9-aminoacridine (26). When
oxazolidinedione, or salicylate when the cytoplasm is alka- the value of pHi is known, ApH can be calculated directly
line relative to the exterior pH. When the cytoplasm is more from knowledge of the pH of the suspending medium.
acidic than the environment, weak bases such as methy- Internal pH values can also be determined by a so-called
lamine, hexylamine, or benzylamine are used. As Kashket null-point method. For this method, cells are suspended in
(26), emphasized, the ApH calculation is valid only when thick suspensions in a set of dilute buffers at various pH val-
the pK of the probe is one pH unit or more different than ues. A membrane-damaging agent, such as butanol, toluene
the exterior pH value. In other words, one probe does not or, say, nigericin, is added to render the cells fully proton
do for all cells or for any one cell type under different con- permeable, and changes in suspension pH as a result of dam-
ditions. Fluorescent probes such as 2',7'-bis(carboxyethy1)- age are recorded. The changes are then plotted against the
4- or 5-carboxyfluorescein have been used extensively, for initial pH values. The initial pH value at which there was
example, by Iwami et al. (23) to assess intracellular pH. In no change in pH induced by membrane damage, obtained
the unhydrolyzed form, they can cross the cell membrane to by extrapolation, can be taken as the internal pH of the
be hydrolyzed by esterases in the cytoplasm to yield the ac- cells. The method is labor-intensive and requires large
tual probe. quantities of cells.
The Henderson-Hasselbalch equation is used for esti- Assessments of Ap, A+, and ApH are subject to many in-
mating internal pH values. In the equation, A stands for the accuracies, as reviewed in some detail by Kashket (26).
anionic form of the weak acid used, H A stands for the pro- There is also the problem of pH, and ApH being very sensi-
tonated form, and subscripts i and o refer to concentrations tive to manipulations of the cells, for example, centrifuging
inside and outside the protoplast, or outside the cell if no or filtering them. Moreover, there are questions about the
correction has been made for the cell wall water volume. capacities of bacterial cells to maintain internal pH.
Therefore, Bacteria such as E. coli are considered to have homeostatic
mechanisms for maintaining internal pH, and a set point
pHi = pKi + log [Ai]/[HAJ pH for maintenance. Other bacteria, such as the strepto-
PH, = PK, + log "4oI/[H%I cocci, do not appear to have a set point pH for homeostasis
but appear to do the best they can to maintain ApH across
It is generally assumed that pKi = pK,, although they are the cell membrane of about one pH unit. As the environ-
not exactly equal because of differences in ionic strength on mental pH drops due to, say, acid production by the gly-
the two sides of the membrane but are sufficiently close for colytic system, the internal pH falls as well. In fact, it ap-
most purposes. Since the membrane is considered to be pears that the fall in internal pH may have a desirable
highly permeable to the undissociated form, [HA,] = function in slowing metabolism under unfavorable condi-
[HA,]. Therefore, tions (50).
Because of the many problems in estimating internal pH
values and ApH, in the past few years, we have more and
more chosen to assess permeabilities of cells to protons
Since rather than ApH (42). In fact, when studying membrane-
active agents, changes in proton permeabilities induced by
the agents are the most pertinent parameters. Methods for
and assessing proton permeability are described by Bender et al.
(5) based on procedures described earlier by Scholes and
Mitchell ( 5 2 ) and Maloney (34). Total flux of protons
then across the membrane per cell or per unit of biomass is read-
ily calculated, usually from initial slopes of pH-versus-time
curves of the types shown in Fig. 3. To obtain the data, pro-
and ton permeabilities of cells in suspensions or in biofilms were
assessed by the procedures described previously (32).
Basically, the suspensions or biofilms immersed in liquid
ApH can then be assessed from estimates of the distribu- medium were brought to constant pH values, and then HCl
tion of, say, benzoate between cells and suspending medium was added to drop the pH value, usually by 0.1 to 0.2 unit.
by the methods described above for permeability or trans- The subsequent rise in pH value as protons moved into the
port assays. From the external benzoate concentration and cells was recorded with a glass electrode in the suspending
the measured external pH, one can calculate the external medium. Initial rates of proton entry were estimated from
concentration of HA or, here, protonated benzoate, which pH changes during the first few minutes after acidification.
is equal to the internal concentration. The internal con- The cells used had buffer capacities of approximately 330
centration of A, here the unprotonated form of benzoate, is Fmol H+ per pH unit change per gram of cell dry weight.
equal to the total internal concentration minus the con- (8). However, cell buffering would not have much effect on
centration of HA. From the ratio [Ai]/[HAi] and a knowl- the calculation of proton flux into the cells because the pH
edge of pKi (assumed to be equal to pK,) the internal changes were small initially. Internal buffering by cell con-
pH value can be estimated by use of the Henderson- stituents would mean that the change in pH in the cyto-
Hasselbalch equation. plasm would not necessarily be the same as the change in
534 METABOLISM

reaction is extremely rapid. Matts and Knowles (38) used a


stopped-flow spectrophotometer to show that the osmotic

5.25 1 movement of water from E. coli cells transferred into a 0.4


M MgClz solution was complete within 50 ms at 37C.
Since water movement is so rapid, the rate of swelling ac-
companying solute uptake, indicated by decreased cell re-
5.00 fractive index and light scattering, can be used as an indi-
cator of transport in studies of protoplasts or spheroplasts
with damaged cell walls. However, the walls of intact bac-
% 4.75 terial cells have sufficient elasticity to resist swelling (14).
This elasticity of the wall results in buildup of tension in the
wall and hydrostatic pressure within the cell, called turgor
4.50 pressure. The turgor pressure (Pm - Pout)can be calculated
by use of the equation

4.25 Pin - Pout= (2.3 RT/v)(log~,-&lif/~in)


where R is the gas constant, T is the temperature (kelvin),
4.00 I V is the molar volume of water, aoutis the water activity out-
side the cell, and amis the water activity within the cell. At
0 20 40 60 80 100 120 a temperature of 25"C, turgor pressure is equal to (311
MPa) X (log [aou'/u'"]).The symbol T is generally used for
Minutes turgor pressure.
The swelling and shrinking of an ideal osmometer can
FIGURE 3 Proton permeability of biofilms of Actinomyces be predicted from the van't Hoff-Boyle equation,
naeslundii in the presence of 0 (open squares), 0.5 (closed tri- V - b = a/n
angles), 1.0 (closed squares), or 5.0 (closed inverted triangles)
mM NaF, a weak acid enhancer of proton movements across where V is the volume of the cell, b is the so-called osmot-
cell membranes. Butanol was added (5% [vol/vol]) at the indi- ically dead space, which generally is approximately equal to
cated time to damage the cell membrane and render it totally the calculated volume of dry matter in the cell, and T is the
permeable to protons. osmolality of the suspending medium. For an ideal 0s-
mometer, a plot of V versus l / yields~ a straight line with
intercept b and slope a. The actual responses of bacterial
cells lead to curved lines (Fig. 4). Clearly, bacterial cells are
pH in the environment. Rates of proton movement de- not ideal osmometers.
creased with time, but this decrease is expected because of Osmotic responsiveness is markedly muted at high 0s-
reductions in ApH across the cell membrane with time. molality, and a value for b can be obtained only by extrapo-
However, the cells continued to have some capacity to lation (Fig. 4). It is not clear why the cell would be so poorly
maintain ApH as indicated by the rise in pH that occurred
when cells nearly equilibrated in terms of pH change with
the suspending medium were treated with butanol, which
opens the membrane to proton movements. Still, it seems
that changes in initial rates of proton uptake give a good in-
dication of the extent of effects of membrane-active agents
on proton permeability of membranes and on their capaci-
ties to maintain ApH between the interior and exterior of
the cell protoplast.
20.3.2. Buffer Capacities of Cells
Buffer capacities of cells can be estimated by the methods
described by Krulwich et al. (31) for assessing external
buffering (B,,), total buffering (BJ, and internal buffering
( B i ) on the basis of acid-base titrations of thick suspensions
of intact or permeabilized cells and the differences between
the two. As expected, buffering capacities vary with pH and
tend to be dominated by carboxyl, phosphate, and amino
groups. Capacity is generally expressed in nanomoles of H+
per pH unit per unit of cell weight or cell protein, in other
words, the numbers of protons required to change the pH of
a suspension containing a unit of cells by one pH unit.
~

Osrnolaiity-1,-*
20.4. OSMOTICALLY SENSITIVE CELLS
When a solute is taken up by a cell, the process results in a FIGURE 4 Osmotic volume changes for microbial cells. The
lowering of water activity in the interior and a consequent dashed line indicates the ideal behavior predicted by the van't
influx of water with resultant swelling of the cell. Biological Hoff-Boyle equation. The solid curve indicates the actual be-
membranes are highly permeable to water, and the swelling havior.
20. Permeability and Transport w 535

responsive in media of high osmolality. The behavior may Bacterial cells require turgor for growth, cell division,
have to do with bound water or with gel-like states of the and certain other functions. How membrane proteins sense
cytoplasm, or, possibly, with reduced permeability of the water stress has been reviewed by Poolman et al. (44).
collapsed membrane to water. Bacterial cells have well developed means to maintain tur-
When placed in media of low osmolality, bacterial cells gor when placed in hypertonic media through pooling of
are resistant to osmotic swelling primarily because they solutes, including KC, glycine betaine, proline, and so-
have elastic cell walls. When placed in concentrated media, called compatible solutes. Osmosensing and osmoregulation
they become plasmolyzed; that is, the protoplast shrinks so have been reviewed in papers by Wood (61), Roessler and
that the membrane is pulled away from the wall, and plas- Muller (49), and Morbach and Kramer (41).
molysis vacuoles are formed between the wall and the pro-
toplast membrane. These vacuoles can often be seen micro-
scopically, especially in gram-negative bacteria. However, 20.5. SOLUTE UPTAKE BY BlOFlLMS
even if they cannot be seen, their presence can be detected Over the past few years, a greater appreciation for the com-
by permeability determinations. Thus, one can assess the munal lives of most microbes in nature has developed. In
total cell volume in terms of the dextran-impermeable fact, most microorganisms are members of biofilm commu-
space. The protoplast volume is essentially the raffinose- nities, especially at solid-liquid interfaces but also at liquid-
impermeable volume under conditions in which raffinose gas interfaces. The growing view is that microbiologists who
cannot cross the cell membrane. The aqueous volume of the study pure cultures are studying peculiar cells without the
cell wall and the plasmolysis vacuoles together is then equal normal range of intercellular interactions of normal cells
to the dextran-impermeable volume minus the raffinose- living in association with a diverse community. Pure cul-
impermeable volume. A reasonable estimate of the aqueous tures can show intercellular phenomena such as quorum
wall volume alone can be made from the raffinose- sensing but do not have to deal with interactions in a di-
impermeable volume of the unplasmolyzed cell. verse community. Biofilms allow for a wide range of biodi-
If plasmolyzed cells are returned to more dilute media, versity. For example, anaerobes and aerobic or facultative
water flows into the protoplast across the water-permeable organisms can live together in biofilms because the aerobes
cell membrane, the protoplast swells to occupy the volume or facultative organisms can protect anaerobes from oxida-
previously occupied by the plasmolysis vacuoles, and the tive damage. Because of the usual slow growth in biofilm
optical density of the suspension increases. There is a di- communities, slowly growing organisms can exist in associ-
rect relationship between the protoplast volume and the ation with organisms capable of very rapid growth. The re-
inverse of the optical density (36). Initially when the pro- sult is that slowly growing organisms are not eliminated
toplast swells, there is little resistance to swelling, and the from biofilms, as they are from suspension cultures.
process is nearly ideal; i.e., it follows the vant Hoff-Boyle Moreover, slow growth can also protect organisms from
relationship. However, when the protoplast abuts against damage by antibiotics, such as the p-lactams, which require
the cell wall, the elasticity of the wall resists further that cells grow rapidly for maximum effect.
swelling, and major deviation from the ideal behavior oc- Methods for producing biofilms in the laboratory have
curs. Now water uptake involves an increase not only in been described in multiple publications and involve a wide
the volume of the protoplast but also in that of the whole variety of variations and specific designs for biofilm reactors.
cell, with stretching of the wall. A break in the curve for V Good sources for information of growth and manipulation of
versus l/n occurs at what is called the point of incipient biofilms are the three volumes of Methods in En.tymology, vol-
plasmolysis. Further reductions in medium osmolality re- umes 310,336, and 337, edited by R. J. Doyle (12,13), as well
sult in the buildup of turgor pressure. It is considered that as multiple other books on the subject of biofilms. Mixed
the medium osmolality at the point of incipient plasmoly- populations with multiple organisms can be maintained in
sis is essentially equal to the internal osmolality of the cell, biofilms under conditions of continuous culture. For exam-
as discussed primarily for plant cells by Stadelmann (54). ple, with oral microbes, it has been possible to develop sys-
One can obtain another estimate of internal osmolality tems for mixed, continuous cultures with as many as 10 sep-
from the value of the constant a (the number of ideal os- arate species in the biofilm consortium (6). Growth of
momolar equivalents within the cell) and a knowledge of biofilms is often carried out with continuous feeding, al-
the protoplast volume. though batch, or more commonly, fed-batch culture is widely
The elasticity of the cell wall does not affect swelling of used. Also, the films can be grown in constant-depth fer-
isolated protoplasts, which can be used for transport studies. mentors or without control of film depth, which allows for
If protoplasts or spheroplasts are made to swell very slowly, sloughing of cells or parts of the film and recruitment of
their membranes do not undergo the sort of brittle fracture newer organisms from the planktonic phase. In general,
that occurs when they are rapidly shocked osmotically. biofilms have associated planktonic organisms in the suspen-
However, the membrane can then become taut, and the sion around the film. Although the planktonic cells are not
magnitude of swelling is not predicted by the vant Hoff- technically part of the biofilm, they can interact with it and
Boyle equation (36). may even become incorporated into the film. A need in
In a study of regulation of turgor pressure by Bacillus sub- working with biofilms is to take account of the age of the
tilis, Whatmore and Reed (59) used a Coulter Counter films used. Biofilms do age, even those grown in the labora-
(Beckmans Coulter) to assess changes in cell volume, and tory with a continuous supply of nutrients. The transport and
these changes were used to construct a vant Hoff-Boyle other physiological capacities of the films can change signifi-
plot. The authors found that spectrophotometric analyses of cantly with age, so there is need for fairly rigid standardiza-
volume changes gave reasonable but somewhat low esti- tion of growth and processing procedures if fairly uniform re-
mates for turgor. For bacteria with internal gas vesicles, sults are needed. Still, the usual finding is that biofilm
changes in light scattering as a result of collapse of the vesi- cultures are more variable than suspension cultures, and
cles under pressure can be used to estimate cell turgor, as de- greater numbers of replicates of experiments are required to
scribed by Reed and Walsby (47). obtain data with acceptable levels of statistical significance.
536 w METABOLISM

Solute transport into biofilms has a number of peculiari- actual biofilm cells may not be. For solute uptake studies,
ties, which currently are not fully understood. It is generally cell water or protoplast water is probably the best base to
felt that growth in biofilms is often diffusion limited and use. There could be an advantage in using cell or cytoplas-
that solute diffusion into biofilms is slow. The topic of dif- mic water as a base for considering solute uptake or excre-
fusion in biofilms has been reviewed by Stewart (55). The tion. The need, then, for estimating cytoplasmic water in
general finding is that life is at a slow pace in biofilms. biofilms is for a solute that can penetrate readily the poly-
Certainly, growth is slow, and generation times of a day or mer matrix of the biofilm and the cell wall but cannot pen-
more are common even for organisms that are able to grow etrate the inner cell membrane. The best candidates are
with generation times of less than an hour in suspension again the ones used with suspension cells, small solutes such
cultures. Catabolism as well is generally slowed in biofilms, as sucrose or raffinose for which the cells under study do not
say, to about one-third the rate for cells in suspensions. have significant permeability or transport systems, or non-
Moreover, there is ample evidence that cells can undergo transported analogues of known substrates. The total water
physiological adaptations peculiar to growth in biofilms. of the biofilm can be assessed with tritiated water or with
Much of the current work on biofilm biology involves use of solutes such as labeled glycerol or urea, although many cells
single-organism biofilms. However, ultimately, the need is actually do have transport systems for urea or glycerol.
for information with multiorganism biofilms and natural
biofilms in the human or animal body or in the environ-
ment. The perspective will then be one of study of perme- 20.6. M E M B R A N E VESICLES AND L I P O S O M E S
ability and solute uptake of a community rather than a sin- Membrane vesicles have been useful for physiological stud-
gle species of organism. ies since the early use by Kaback and Stadtman (25) of cy-
A major orientation currently with biofilms is to do in toplasmic membrane preparations from E. coli cells to study
situ measurements, for example, of pH. One seemingly sim- proline uptake. Generally, the first step in preparing vesicles
ple way to measure pH in biofilms is to use tiny pH elec- is to convert the test organism into an osmotically sensitive
trodes, for example, the Beetrode pH electrode from World form, which is then easily disrupted osmotically or mechan-
Precision Instruments (New Haven, CT), and to place the ically under conditions that allow for resealing of mem-
tip directly into the biofilm, as described by Burne and brane fragments to form vesicles. Spheroplasts of gram-
Marquis (7). More elaborate methods involve two-photon negative bacteria such as E. coli can be prepared by the
excitation microscopy and use of fluorescent dyes such as lysozyme-EDTAmethod (24), which leaves the outer mem-
carboxyfluorescein, as described by Vroom et al. (58). brane of the cell wall associated with the spheroplast.
Biofilms are actually thin films, at least as viewed by a Protoplasts of gram-positive bacteria can often be com-
chemical engineer, and diffusion of small molecules into the pletely freed from cell wall structures by treatment with
films can be rapid. Diffusion of larger molecules can be wall-lytic enzymes, such as lysozyme for cells of Bacillus
greatly slowed, and for organisms dining on, say, proteins, megateriurn, B. subtilis, Enterococcus hirae (Streptococcus fae-
access to substrate within the biofilm can be highly re- calk), or Micrococcus luteus; lysostaphin for Staphylococcus
stricted. Of course, cells at the surface of the biofilm can se- aureus; or mutanolysin for Streptococcus rnutans (29).
crete enzymes to hydrolyze large molecules outside the com- Kobayashi et al. (27) isolated vesicles from E. hirae
munity so that the digested pieces can diffuse into the ATCC 9790, and their procedure is illustrative. They first
biofilm community. Biofilms commonly have channels that suspended cells in an osmotic stabilizing solution of 500 mM
penetrate the films, and these channels can be conduits for glycylglycine plus 2 mM MgS04 (pH 7.2), added 0.5 mg of
solutes to reach the bottom parts of the film. They have lysozyme per ml, and incubated the mixture at 37C for 1 h.
been visualized by means of confocal laser scanning mi- (Protoplasts can also be stabilized with sucrose instead of gly-
croscopy (60),especially in Pseudomonas biofilms, but also cylglycine.) The protoplast suspension was centrifuged; re-
for complex dental-plaque biofilms grown in the human suspended in a hypotonic solution of 50 mM Tris-maleate
mouth on enamel pieces (62). However, most biofilms are (pH 7.4), 250 mM sucrose, and 5 mM MgS04; and then
stratified. For example, the more aerobic organisms may be treated with DNase I, which requires magnesium ions for
positioned at the liquid interface, where O2is more abun- activity. The suspension was chilled, homogenized with a tis-
dant, and obligate anaerobes may be in the depth of the sue homogenizer, centrifuged at 5,000 x g for 10 min to re-
film, where very little O2can penetrate without being me- move debris, and then centrifuged at 27,000 x g for 10 min
tabolized. Often studies of solute transport involve use of to pellet the membranes. The membranes were resuspended
entire biofilms, so that an average value is obtained for what in the buffer solution of Tris-maleate plus sucrose plus
may be a very heterogeneous set of uptakes by individual MgS04 and then passed through a French press (10,000
cells. However, more and more, techniques such as confocal lb/in2, or 68 MPa). The resulting suspension was centrifuged
microscopy and use of fluorescent tags are being exploited twice, once at 27,000 x g to remove large particles and once
to analyze particular solutes in particular parts of the at 48,000 X g for 90 min to pellet the vesicles.
biofilm. In a very direct approach, Robinson et al. (48) de- There are many possible variants of this procedure, and
veloped a system in which they froze intact biofilms, sec- some preliminary experimentation is needed to determine
tioned them with a microtome, and then measured previ- optimal procedures for a particular organism. A shortened,
ously added metabolites or chemicals in the slices as a one-step procedure for use with gram-positive bacteria, no-
function of depth into the biofilm. tably B. subtilis, was described by Konings et al. (30).
There can be a problem in comparing biofilms to cells The vesicles in these preparations tend to be heteroge-
from suspension cultures in relation to the base for compar- neous in size, and the preparations by Kobayashi et al. (27)
ison. Often, biomass is used as a base. However, biofilms are contained both right-side-out and wrong-side-out vesicles,
generally very rich in polysaccharides because the biofilm as indicated by their appearance in electron micrographs
matrix is mainly polysaccharide. Protein content can be after freeze-fracturing. With E.coli, it is possible to prepare
used as a base, but again, there can be problems in that populations of vesicles all in the right-side-out conforma-
biofilms are relatively low in protein content, although the tion. For example, Kaback (24) described a procedure in-
20. Permeability and Transport w 537

volving osmotic lysis without passage through a French 4. Belli, W. A., and R. E. Marquis. 1991. Adaptation of
pressure cell. E. coli vesicles prepared with use of the French Streptococcus mutuns and Enterococcus hirae to acid stress
pressure cell are mainly wrong-side-out and, for example, in continuous culture. Appl. Enuiron. Microbiol. 57:1134-
will not extrude Ca2+ or take up proline (1). Both right- 1138.
side-out and wrong-side-out vesicles can be useful for phys- 5. Bender, G. R., S. V. W. Sutton, andR. E. Marquis. 1986.
iological studies. However, it is best to have preparations Acid tolerance, proton permeabilities, and membrane
nearly all in one conformation or the other. Membranes of ATPases of oral streptococci. Infect. Immun. 53:331-338.
many organisms are very resistant to resealing after rupture, 6. Bradshaw, D. J., P. D. Marsh, K. M. Schilling, and
D. Cummins. 1996. A modified chemostat system to study
so it is nearly impossible to prepare vesicles from organisms the ecology of oral biofilms. J. Appl. Bacteriol. 80:124-
such as S. mutans and only very small numbers of sealed 130.
vesicles from large amounts of isolated membrane. 7. Burne, R. A., and R. E. Marquis. 2001. Biofilm acidbase
The membrane continuity of vesicles is best tested by as- physiology and gene expression in oral bacteria. Methods
sessing the extent of swelling or shrinking in response to En&nol~337:403415.-
changes in osmolality of the suspending medium. The pro- 8. Burne, R. A., R. G. Quivey, Jr., and R. E. Marquis.
cedures described in the section on osmotically sensitive 1999. Physiologic homeostasis and stress response in oral
cells can be adapted for use with vesicles, especially those biofilms. Methods Enqmol. 310:441460.
involving measurements of changes in light scattering (tur- 9. Chung, H-J., T. J. Montville, and M. L. Chikindas.
bidity) associated with swelling (decreased optical density) 2000. Nisin depletes ATP and proton motive force in my-
and shrinking (increased optical density) of vesicles. The cobacteria. Lett. Appl. Microbiol. 31:416420.
functionality of vesicles is determined in terms of their ca- 10. Conway, E. J., and M. Downey. 1950. An outer meta-
pacities to transport solutes. bolic region of the yeast cell. Biochem. J. 47:347-355.
The workings of individual transport components, such 11. de Poorter, L. M. I., and J. T. Keltjens. 2001.
as permease proteins, antiporters, symporters, or ion- Convenient fluorescence-basedmethods to measure mem-
translocating ATPases, can often best be studied by isolat- brane potential and intracellular pH in the Archaeon
Methanobacterium thermoautotrophicum. J. Microbiol.
ing the catalysts from cell membranes and incorporating
Methods 47:233-241.
them into liposomes or proteoliposomes. The test system is 12. Doyle, R. J. (ed.). 1999. Methods in Enzymology, vol. 310.
then somewhat simpler than the intact cell, but there com- Biofilms. Academic Press, Inc., San Diego, CA.
monly is a need to set up some sort of membrane potential 13. Doyle, R. J. (ed.). 2001. Methods in Enzymology, vol.
or ApH or to incorporate ATP or phosphoenolpyruvate into 336/337. Microbial Growth in Biofilms. Part A, Develop-
the liposomes or proteoliposomes to energize the transloca- mental and Molecular Aspects. Part B, Special Environments
tors. A thorough description of the use of proteoliposomes and Physicochemical Aspects. Academic Press, Inc., San
containing E. coli phospholipids and Lac permease is given Diego, CA.
by Viitanen et al. (57). In a more elaborate system, other 14. Doyle, R. J., and R. E. Marquis. 1994. Elastic, flexible
proteins can be incorporated into proteoliposomes so that peptidoglycan and bacterial cell wall -properties.
- Trends
Ap can be generated by the structures themselves. For ex- M&obiil.' 2:57-60.
ample, Fristedt et al. (20) used proteoliposomes with incor- 15. Driessen, A. J. M., and W. N. Konings. 1993. Insertion
porated purified cytochrome c oxidase from beef heart mi- of lipids and proteins into bacterial membranes by fusion
tochondria to study the functioning of the proton-coupled with liposomes. Methods Enzymol. 221:394408.
Pho84 phosphate permease of Saccharomyces cereuisiue. The 16. Driessen, A. J. M., D. Molenaar, and W. N. Konings.
cytochrome oxidase acted to generate Ap across the vesicle 1989. Kinetic mechanism and specificity of the arginine-
membrane to drive uptake of phosphate. Fusion of the com- ornithine antiporter of Lactococcus lactis. J. Biol. Chem.
264: 10361-103 70.
ponents of the proteolysosomes was accomplished by a 17. Driessen, A. J. M., B. P. Rosen, and W. N. Konings.
freeze-thaw-sonication method (15). 2000. Diversity of transport mechanisms: common struc-
tural principles. Trends Biochem. Sci. 25:397401.
20.7. PERMEABlLlZED CELLS 18. Dubois, M., K. A. Gilles, J. K. Hamilton, P. A. Rebers,
and F. Smith. 1956. Colorimetric method for determina-
Methods to render cells permeable to solutes by chemical or tion of sugars and related substances.Anal. Chem. 28:350-
enzymatic treatment are used widely. Perhaps the most 356.
common is that involving treatment of cells with toluene in 19. Felle, H., J. S. Porter, C. L. Slayman, and H. R. Kaback.
standard assays of P-galactosidase in bacteria such as E. coli. 1980. Quantitative measurements of membrane potential
Gram-positive bacteria may require more rigorous proce- in Escherichia coli. Biochemistry 19:3585-3590.
dures; for example, permeabilization of cells of S. mutans re- 20. Fristedt, U., M. van der Rest, B. Poolman, W. N.
quires treatment with 10% (vol/vol) toluene (which ex- Konings, and B. L. Persson. 1999. Studies of cytochrome
ceeds the solubility of toluene in water) followed by two c oxidase-driven H+-coupled phosphate transport cat-
rounds of freezing and thawing (4). alyzed by the Saccharomyces cerevisiue Ph084 permease in
coreconstituted vesicles. Biochemiscry 38: 16010-1 60 15.
21. Harold, F. M., and K. Altendorf. 1974. Cation transport
20.8. REFERENCES in bacteria: K+,Na+, and H+. Curr. Top. Membr. Tramp.
1. Altendorf, K. H., and L. A. Staehelin. 1974. Orientation 5:l-50.
of membrane vesicles from Escherichia coli as detected by 22. Hurwitz, C., C. B. Braun, and R. A. Peabody. 1965.
freeze-cleave electron microscopy. J. Bacteriol. 117:888- Washing bacteria by centrifugation through a water-im-
899. miscible layer of silicone. J. Bacteriol. 90:1692-1695.
2. Ames, G. F.-L. 1974. Two methods for the assay of amino 23. Iwami, Y., S. Hata, C. F. Schachtele, and T. Yamada.
acid transport in bacteria. Methods Enqmol. 32:843-849. 1995. Simultaneous monitoring of intracellular pH and
3. Avison, M. J., H. P. Hetherington, and R. G . Shulman. proton excretion during glycolysis by Screptococcus mutans
1984. Applications of NMR to studies of tissue metabo- and Streptococcus sanguis: effect of low pH and fluoride.
lism. Annu. Rev. Biophys. Biophys. Chem. 15:377-402. Oral Microbiol. Immunol. 10:35 5-3 59.
538 w METABOLISM

24. Kaback, H. R. 1971. Bacterial membranes. Methods 45. Ramos, S., and H. R. Kaback. 1977. The electrochemical
Enzymol. 22:99-120. proton gradient in Escherichia coli membrane vesicles.
25. Kaback, H. R., and E. R. Stadtman. 1966. Proline uptake Biochemistry 16:848-854.
by an isolated cytoplasmic membrane preparation of Esch- 46. Ramos, S., S. Schuldiner, and H. R. Kaback. 1976. The
erichiu coli. Proc. Natl. Acad. Sci. USA 55:920-927. electrochemical gradient of protons and its relationship to
26. Kashket, E. R. 1985. The proton motive force in bacteria: active transport in Escherichia coli membrane vesicles. PrOC.
a critical assessment of methods. Annu. Rev. Microbiol. Natl. Acad. Sci. USA 73:1892-1896.
39:219-242. 47. Reed, R. H., and A. E. Walsby. 1985. Changes in turgor
27. Kobayashi, H., J. Van Brunt, and E M. Harold. 1978. pressure in response to increases in external NaCl concen-
ATP-linked calcium transport in cells and membrane vesi- tration in the gas-vacuolate cyanobacteriumMicrocystis sp.
cles of Streptococcus faecalis. J. Biol. Chem. 253:2085- Arch. Microbiol. 143:290-296.
2092. 48. Robinson, C., J.Kirkham, R. Percival, R. C. Shore,
28. Koch, A. L. 1994. Growth measurement, p. 248-277. In W. A. Bonass, S. J. Brookes, L. Kusa, H. Nakagaki, K.
P. Gerhardt (ed.), Methods for General and Molecular Kato, and B. Nattress. 1997. A method for the quantita-
Bacteriology. ASM Press, Washington, DC. tive, site-specific study of the biochemistry within dental
29. Konings, W. N. 1977. Active transport of solutes in bacte- plaque biofilms in vivo. Caries Res. 31:194-200.
rial membrane vesicles. Adv. Microb. Physiol. 15:175-251. 49. Roessler, M., and V. Muller. 2001. Osmoadaptation in
30. Konings, W. N., A. Bisschop, M. Veenhuis, and C. A. bacteria and archaea: common principles and differences.
Vermeulen. 1973. New procedure for the isolation of Environ. Microbiol, 3:743-754.
membrane vesicles of Bacillus subtilis and an electron mi- 50. Russell, J. B., and F. Diez-Gonzales. 1998. The effects
croscopic study of their ultrastructure. J. Bacteriol. 116: of fermentation acids on bacterial growth. Adu. Microb.
1456-1465. Physiol. 39~205-234.
31. Krulwich, T. A., R. Agus, M. Schneier, and A. A. 51. Scherrer, R., and P. Gerhardt. 1971. Molecular sieving by
Guffanti. 1985. Buffering capacity of bacilli that grow at the Bacillus megaterium cell wall and protoplast. J. Bac-
different pH ranges. J. Bacteriol. 162:768-772. teriol. 107:7 18-735.
32. Ma, Y., T. M. Curran, and R. E. Marquis. 1997. Rapid 52. Scholes, P., and P. Mitchell. 1970. Acid-base titration
procedures for acid adaptation of oral lactic-acid bacteria across the plasma membrane of Micrococcus denitrificans:
and further characterization of the response. Can. J. factors affecting the effective proton conductance and the
Microbiol. 43: 143-148. respiratory rate. J. Bioenerg. 1:61-72.
33. MacDonald, R. E., and P. Gerhardt. 1958. Bacterial per- 53. Siebold, C., K. Flukiger, R. Beutler, and B. Erni. 2001.
meability: the uptake and oxidation of citrate by Esche- Carbohydrate transporters of the bacterial phospho-
richia coli. Can. J. Microbiol. 4:109-124. eno1pyruvate:sugar phosphotransferase system (PTS).
34. Maloney, P. C. 1979. Membrane H+ conductance of FEBS Lett. 504:104-111.
Streptococcus lactis. J. Bacteriol. 140:197-205. 54. Stadelmann, E. J. 1966. Evaluation of turgidity, plasmoly-
35. Maloney, P. C., E. R. Kashket, and T. H. Wilson. 1975. sis and deplasmolysis of plant cells. Methods Cell Physiol.
Methods for studying transport in bacteria. Methods 2: 143-2 16.
Membr. Biol. 5:1-49. 55. Stewart, P. S. 2003. Diffusion in biofilms. J. Bacteriol.
36. Marquis, R. E. 1967. Osmotic sensitivity of bacterial pro- 185:1485-1491.
toplasts and the response of their limiting membrane to 56. Tran, Q. H., and G. Unden. 1998. Changes in the proton
stretching. Arch. Biochem. Biophys. 118:323-331. potential and the cellular energetics of Escherichia coli dur-
37. Marquis, R. E., and P. Gerhardt. 1964. Respiration- ing growth by aerobic and anaerobic respiration or by fer-
coupled and passive uptake of a-aminoisobutyric acid, a mentation. Eur. J. Biochem. 25 1:538-543.
metabolically inert transport analogue, by Bacillus mega- 57. Viitanen, P. M., J. Newman, D. L. Foster, T. H. Wilson,
terium. ]. Biol. Chem. 239:3361-3371. and H. R. Kaback. 1986. Purification, reconstitution, and
38. Matts, T. C., and C. J. Knowles. 1971. Stopped-flow characterization of the lac permease of Escherichia coli.
studies of salt-induced turbidity changes of Escherichia coli. Methods Enzymol. 125429-452.
Biochim. Biophys. Acta 249583-587. 58. Vroom, J. M., K. J. de Grauw, H. C. Gerritsen, D. J.
39. Midgley, M. 1987. An efflux system for cationic dyes and Bradshaw, P. D. Marsh, G. K. Watson, J. J. Birmingham,
related compounds in Escherichia coli. Microbiol. Sci. 4: and C. Allison. 1999. Depth penetration and detection
125-127. of pH gradients in biofilms by two-photon excitation mi-
40. Mitchell, P., and J. Moyle. 1959. Permeability of the en- croscopy. A@. Environ. Microbiol. 65:3502-3511.
velopes of Staphylococcus aureus to some salts, amino acids, 59. Whatmore, A. M., and R. H. Reed. 1990. Determination
and non-electrolytes. J. Gen. Microbiol. 20:434441. of turgor pressure in Bacillus subtilis: a possible role for K+
41. Morbach, S., and R. Kramer. 2002. Body shaping under in turgor regulation. J. Gen. Microbiol. 136:2521-2526.
water stress: osmosensing and osmoregulation of solute 60. White, N. S. 1995. Visualization systems for multidimen-
transport in bacteria. ChemBioChemistry 3:384-397. sional CLSM images, p. 211-254. In J. B. Pawley (ed.),
42. Phan, T..N., J. S. Reidmiller, and R. E. Marquis. 2000. Handbook of Biological Confocal Microscopy. Plenum Press,
Sensitization of Actinomyces nueslundii and Streptococcus New York, NY.
sanguis in biofilms and suspensions to acid damage by fluo- 61. Wood, J. M. 1999. Osmosensing by bacteria. Signals and
ride and other weak acids. Arch. Microbiol. 174:248-255. membrane-based sensors. Microbiol. Mol. Biol. Rev. 63:
43. Poolman, B. 2002. Transporters and their roles in LAB 230-262.
cell physiology. Antonie Leeuwenhoek 82:147-164. 62. Wood, S. R., J. Kirkham, P. D. Marsh, R. C . Shore, B.
44. Poolman, B., P. Blout, J. H. A. Folgering, R. H. E. Friesen, Nattress, and C. Robinson. 2000. Architecture of intact
P. Moe, and T. der Heide. 2002. How do membrane pro- natural human plaque biofilms studied by confocal laser
teins sense water stress?Mol. Microbiol. 44:889-902. scanning microscopy. J. Dent. Res. 79:21-27.
Bacterial Respiration
ROBERT P. GUNSALUS. GARY CECCHINI. AND IMKE SCHRODER

21.1. ENZYME ASSAYS ......................................... 540


21.1.1. Reaction Classification and Nomenclature ................... 540
.
2. Units and Specific Activity .............................. 541
.
3. Types of Activity Assays ............................... 541
21.1.3.1. Qualitative Assays ............................. 541
.
2. Quantitative Assays ............................ 542
2 1.2. ENZYME LOCALIZATION .................................. 543
21.2.1. Intact Cells ......................................... 543
.
2. Permeabilized Cells ................................... 543
.
3. Spheroplasts and the Periplasmic Fraction ................... 544
.4. Cell Extracts ........................................ 544
.5. Membrane and Cytoplasmic Protein Fractions ................ 544
.6. Location(s) of Enzyme Activity in Cell Compartments .......... 544
.7. Determining the Orientation of the Enzyme's Substrate Site ...... 545
2 1.3. AEROBIC RESPIRATORY ENZYMES .......................... 545
2 1.3.1. NADH:Q Oxidoreductase .............................. 545
.2. Succinate Dehydrogenase ............................... 546
21.3.2.1. Activation of Succinate Dehydrogenase .............. 546
.2. Succinate Oxidation with PES or PMS .............. 546
.3. Succinate Oxidation with Ferricyanide .............. 546
.4. Succinate Oxidation with Ubiquinone Analogs ........ 546
2 1.3.3. Cytochrome Oxidase .................................. 546
21.3.3.1. Oxygen Reduction with Ubiquinol as Electron Donor ... 547
.2. Cytochrome c as Electron Donor .................. 547
21.4. ANAEROBIC RESPIRATORY ENZYMES ....................... 547
21.4.1. Anaerobic Enzyme Assays .............................. 548
21.4.1.1. Fumarate Reduction ............................ 548
.2. Nitrate Reduction (Denitrification. Ammonification.
and Assimilation) ............................. 549
.3. Anaerobic Reduction of Amine N-Oxides and
Sulfoxides ................................... 550
.4. Reduction of Metal Oxides (Selenate, Tellurate.
Arsenate, and Iron) ............................ 551
.5. Sulfur Oxide Reduction ......................... 552
.6. Formate Dehydrogenase ......................... 552
.7. Hydrogenase ................................. 553
21.5. REFERENCES ............................................. 554

Prokaryotes as a group exhibit vast respiratory diversity. anaerobically (8. 26). This respiratory diversity among all
This is manifested in the large range of electron donors and life forms thus allows many different strategies to obtain
electron acceptors that may be redox coupled for energy energy and to sustain cell growth and survival. The respira-
generation. Respiring microbes include obligate aerobes. tory ability is predicated by the genetic makeup of the
obligate anaerobes. microaerophiles. and facultative anaer- organism. Associated with this prokaryotic respiratory di-
obes. Only some types of fermentative bacteria are inca- versity is a general ability to use the energetically more fa-
pable of any respiration. Depending on the species and vorable electron acceptor-donor combinations in lieu of
strain. a microbe may possess a relatively simple electron lower-energy releasing reactions (26. 34. 91).
transport pathway (e.g., oxidation of NADH coupled to For all respiring microbes. the paired oxidation-reduc-
di-oxygen reduction to water) as in the mitochondria of tion reactions are coupled to ion transport and to ATP syn-
yeasts. while other microbes possess multiple branching res- thesis. The maximal energy released depends on the mid-
piratory pathways for energy generation from a large vari- point potential difference between the electron donor and
ety of electron donors and electron acceptors. For example. electron acceptor pair (Table 1). while the energy yield de-
Eschen'chia coli can utilize over 20 combinations of electron pends on the efficiency of the specific types of respiratory
donors and electron acceptors to respire aerobically or complexes made by the organism. Thus. the overall energy

539
540 METABOLISM

yield achieved for a given electron donor and electron ac- Anaerobic respiration
ceptor pair may differ between species.
The oxidation-reduction midpoint potentials for many NO3- + NADH + H+ + NO2- + NADf + H 2 0
aerobic and anaerobic respiratory substrates are presented in AG" = -143 kJ/mol
Table 1. Common bacterial electron acceptors include oxy-
gen, nitrate, nitrite, nitrous oxide, nitric oxide, methyl sulf- TMAO + NADH + H+ -+TMA + NADf + H 2 0
oxides, methyl amine-N-oxides, fumarate, sulfate, sulfite, AG" = -86 kJ/mol
elemental sulfur, ferric ion, and carbon dioxide plus less Fumarate + NADH + Hf + succinate + NADf
well-studied acceptors like selenate, arsenate, tellurate, AG'' = -67 kJ/mol
manganous ion, and uranium(V1). Commonly used bacter-
ial electron donors include hydrogen gas, formate, glycerol, NO3- + formate + NO2- + C02 + HlO
ethanol, acetate, lactate, and succinate (7, 26, 40, 91, 93). AG" = -167 kJ/mol
Several examples of aerobic and anaerobic oxidation-
reduction reactions are presented below along with the en- The Gibbs free energy (AG") is determined under stan-
ergy released under standard reaction conditions. dard biochemical conditions.
General scheme: AG'O = - n ~ ~ ~ r o
Acceptor + donor + products free energy released where AE" = [E" (for A/AH) -E'O (for D/DH)] (E is in
(AG" = -nF AE")
volts [the redox values are in Table l]), AE'' is the differ-
Aerobic respiration ence in redox potentials between the acceptor and donor
pair, n is the number of electrons transferred, and F is the
O2+ 2NADH + 2H+ -+ 2 H 2 0 + 2NADf Faraday constant (96.5 kJ/mol/V)
AG" = -438 kJ
Depending on the genetic blueprint of a microbe, it may
0 2 + 2H2 + 2H20 + 2NAD' only produce a few respiratory enzymes or it may be capable
AG" = -474 kJ of synthesizing many alternative respiratory complexes. For
example, E. coli possesses two distinct cytochrome oxidases,
three nitrate reductases, two nitrite reductases, two
TABLE 1 Standard redox potentials of selected electron trimethylamine-N-oxide (TMAO) reductases, a dimethyl
donors and acceptors involved in bacterial oxidation-reduc- sulfoxide (DMSO) reductase, and a fumarate reductase.
tion reactions Most enzymes are differentially synthesized in response to
oxygen and nitrate availability and, for some species, in re-
Redox pair (oxlred) Elo (mV)" sponse to the presence of TMAO or fumarate. The level of
SO.,-/ HS03- .............................. -516 a given enzyme in an organism may also vary depending on
the conditions used for cell growth. Thus, prior to cell har-
C 0 2 /formate .*. -432 vest and enzyme assay, consideration should be given to the
H+/H2 .................................... -414 composition of the culture medium used (oxygen, nitrate,
S2032-/HS- + HS03- ....................... -402 formate, etc.). One may also consider other environmental
NAD+/NADH + H+ ........................ -320 parameters for cell growth (e.g., pH, salinity, and tempera-
ture). Finally, if the genomic DNA sequence of the organ-
ism is known, inspection of the predicted gene and thus
protein content may provide additional insight to the po-
tential for cell respiration.
Acetaldehyde/ethanol ........................ - 197 The goal of this chapter is to provide a brief introduction
Pyruvate/lactate . . . . . . . . . . . . to assay the activity of respiratory enzymes. Of the large
Dihydroxyacetone-P/glycerol-P . . . . . . . . . . . . . . . . . - 190
range of organic and/or inorganic electron donors and ac-
ceptors used by microbes ( Y l ) , a subset of these respiratory
HS03-/S302- . . . . . . . . . . substrates is considered here. For more in-depth informa-
HSO,-/HS- . . . . . . . . . . . . tion or specific enzyme assays, the reader is directed to the
MQ ox/red ........................... literature.
APS/AMP + HS03- ......................... -60
Fumarate/succinate .. ......................... +33
Ubiquinone ox/red . . ..... . . . . . . . . . . +110 21 .l.ENZYME ASSAYS
TMAO/TMA ............................... + 130 Assays for many commonly encountered respiratory en-
zymes are presented below, while literature citations are
DMSO/DMS + 160 presented for others. Assays of several anaerobic respiratory
N02-/NO ................................. +350 enzymes associated with fermenting conditions (e.g., lac-
N03-/N02-. ..... +433 tate dehydrogenase, hydrogenase, and formate dehydroge-
S ~ O ; - / S ~ O ~ ~.............................
- +475 nase) are described in a companion chapter in this book
Fe3+/Fe2+ +772 (chapter 22).
O2/H20 ................................... +818
21.I .I. Reaction Classification and Nomenclature
NOIN20 .... .... +1,175
Enzyme nomenclature as defined by the International
N2O/N, ................................... +1,355
Union of Biochemistry and Molecular Biology is not dis-
Values derived from reference 91 cussed here but may be explored at http://www.chem.qmul
21. Bacterial Respiration 541

.ac.uk/iubmb/enzyme/. Additional enzyme information is mM BV, plus the protein sample to be assayed. After equil-
available at BRENDA, the Comprehensive Enzyme ibrating the tubes at the desired temperature, 5 mM sodium
Information System at http://www.brenda.uni-koeln.de/, dithionite or less (Na2S204dissolved in 0.1 M NaHC03,
and chapter 19 of this volume. stored on ice) is added to reduce the viologen dye. The ap-
pearance of a blue-purple color indicates the presence of re-
21 .I .2. Units and Specific Activity duced viologen. The amount of dithionite used in the spot
Unless indicated otherwise, one unit of enzyme activity is assay is typically sufficient to reduce any oxygen present in
defined as micromoles of substrate reduced or oxidized per the tube since reduced viologen is readily autoxidized by
minute. The specific activity refers to units per milligram of oxygen. However, excess dithionite will also inhibit nitrate
protein (see also chapter 19 of this volume and reference reductase activity, so care should be taken in this step of the
69). procedure. The nitrate reductase assay is then initiated by
the addition of 10 mM sodium nitrate (i.e., from a concen-
21 .I .3. Types of Activity Assays trated aerobic solution), and the microcentrifuge tube con-
In general, quantitative enzyme assays are routinely per- taining the assay mixture is incubated at the desired tem-
formed to calculate an enzymes specific activity and other perature for a fixed time period (e.g., about 10 min)
kinetic properties. However, qualitative enzyme assays may depending on the level of nitrate reductase present in the
be conveniently applied for some studies to screen for the sample. The disappearance of purple color indicates the
presence of a desired enzyme. These methods include in situ nitrate-dependent oxidation of reduced BV dye where the
activity stains of native polyacrylamide gels (i.e., zymo- degree of color change is roughly correlated to the amount
grams), or qualitative or semiquantitative spot plate as- of enzyme activity present. It is important to include a
says. The investigator is encouraged to improvise specific nonenzyme control tube to monitor autoxidation of re-
applications for different enzymes and/or strains based on duced viologen dye. Activity is scored by the relative differ-
the examples described below. ence in color across the range of assay tubes. Following
completion of the assay as described above, the protein sam-
21 .I .3.1. Qualitative Assays ples are either evaluated immediately for nitrite formation
In exploratory studies to screen for the presence of a given or frozen at -20C. In the latter case, the samples should
enzyme activity, it is often convenient to use qualitative have a neutral pH (e.g., not preserved with acid).
screening methods. Applications employ the enzymes spe- Method B. Since it is often problematic to rely on decol-
cific substrate and a redox dye that changes color as a result oration of reduced viologen dye, the detection of nitrite
of the reaction catalyzed by the enzyme. Examples follow for presents a more reliable method. Formation of nitrite is
detection of nitrate reductase. based on the reaction of nitrite with sulfanilamide under
acidic conditions to form a diazo compound (32; see also
21.1.3.1.1. Use of Spot Assays or Color chapter 18 in this volume). N-( 1-Naphthy1)-ethylenedi-
Indicator Assays amine is a colorless compound which in the presence of ni-
Spot assays can facilitate the rapid detection of enzyme trite forms a purple compound with absorbance at 543 nm.
activities in whole or permeabilized cells, in subcellular The sensitivity of this assay ranges from 1 to 180 nmol of
samples, or in chromatography fractions to follow protein N02-/ml. Certain substances interfere with the assay, in-
purification. Compared to quantitative cuvette assays that cluding colored or particulate materials that may be present
may need to be performed under controlled conditions, spot in the protein sample, or the presence of C1-, Fe3+,or Cu3+
assays may be done in microtiter dishes or in microcen- ions. To prepare the color reagent, 1 part 0.08% N-(l-
trifuge tubes under less rigorous conditions. The tube or mi- naphty1)-ethylenediamine,dissolved in H20,is mixed with
crotiter dish containing the assay mixture and enzyme sam- 2 parts of 4% sulfanilamide (p-aminobenzene sulfonamide,
ple is mixed with a colorimetric indicator reagent. dissolved in 25% HC1). Note that the 0.08% N-(l-
Typically, an increase or decrease in color change indicates naphty1)-ethylenediaminesolution should be stored in the
the presence and relative amount of enzyme activity pres- dark and discarded when it turns brown. To initiate the ni-
ent. The degree of color change can be visually estimated, trite detection reaction, 0.3 ml of color reagent is added to
or semiquantified by spectral methods to determine what a 1-ml nitrate reductase spot assay from method A. A solu-
fraction(s) contain the most abundant enzyme activity. tion of the diluted enzyme sample is mixed and incubated
However, since no clear attempt is made to validate exper- for 10 min at room temperature. Formation of a purple color
imental conditions with respect to assay linearity, substrate indicates the formation of nitrite ions where the color in-
limitation, or presence of interfering materials, this ap- tensity is proportional to the amount of nitrite present.
proach is not suitable for enzyme rate determinations. Rate Remaining reduced viologen dye typically oxidizes during
determinations require the measurement of a color-based this procedure and does not interfere with the nitrite detec-
activity change with time under carefully established assay tion assay. Note: the presence of any nitrite reductase activ-
conditions. ity in the protein sample may interfere with the assay.

21.1.3.1.1.1. Nitrate reductase spot assay. Nitrate re- 21.1.3.1.2. Zymograms, or In Gel
ductase catalyzes the reduction of nitrate to nitrite and can Polyacrylamide Activity Stains
be assayed by following the color change of a redox dye or In-gel activity stains, or zymograms, offer an alternative
by detection of end product formed (i.e., nitrite). approach to detect oxidoreductase enzymes, especially
Method A. Nitrate reduction in this assay is monitored by when the level of enzyme activity is at or below the sensi-
following the oxidation of the artificial electron donor, re- tivity limit of a standardized quantitative enzyme assay. Low
duced benzyl viologen (BV) or methyl viologen (MV) in enzyme activity may be due to a low cellular abundance of
1.5-ml microcentrifuge tubes. The tubes are filled with an the enzyme in the sample, or due to suboptimal conditions
aerobic solution of 0.1 M potassium phosphate (pH 7.2), 2 used for the enzyme assay. Zymograms can also be used to
542 METABOLISM

detect isozymes that differ in molecular mass when multiple performed prior to gassing. The investigator is encouraged
enzymes with similar activities are present in a cell extract. to improvise on and optimize assay conditions for hisher
For extensive information regarding the principles and ap- specific application.
plications of enzyme analysis using zymograms,see reference
59. One specific application to detect soluble nitrate reduc- 21 .I .3.2. Quantitative Assays
tase in cell extracts is provided below.
Quantitative assay methods are applied to first detect en-
zyme activity in cells or cell fractions and then, more im-
21.1.3.1.2.1. Nitrate reducase zymogram assay. The portantly, to obtain a detailed understanding of the en-
(ha1o)alkaliphilic sulfur-oxidizing bacterium Thioalkalivibrio zymes kinetic properties. As noted above, both require
nitratireducens contains a soluble nitrate reductase (5). optimization of the enzyme assay conditions and require-
Following cell harvest, cells are broken by sonication (see ments of the specific enzyme being studied. The enzyme as-
section 21.2). The soluble protein fraction is isolated via says described in the following subsections are based on
centrifugation and separated by native polyacrylamide elec- specific examples from the literature. Therefore, the inves-
trophoresis. The polyacrylamide gel lanes are then cut into tigator may need to optimize the assay conditions depend-
vertical strips and placed in a shallow tray containing the ing on the source of the enzyme (i.e., the microorganism
enzyme assay solution (0.2 M sodium phosphate buffer [pH that makes it) and the enzymes requirements. The investi-
7.01, 5 mM MV, and 10 mM KN03). A sodium dithionite gator is directed to the microbiological and biochemical lit-
solution (10 mM) is then slowly added to reduce the MV, erature for additional details. Also, see chapters 19 and 20
which in turn provides electrons for the nitrate reduction in this volume for further details on enzymatic activity, bac-
reaction. The immersed gels strips are then incubated with terial transport, and membrane permeability.
gentle shaking at the preferred temperature (e.g., room tem- The choices of electron donors or acceptors include the
perature) until transparent bands appear in the gel strip physiological electron donors or acceptors such as
against the blue background of reduced viologen dye. The ubiquinone, menaquinone (MQ), NAD(P)H, c-type cyto-
transparent band(s) in the gel correspond to the location(s) chromes, or iron-sulfur proteins that provide or accept re-
of the nitrate reductase activity. The gel is subsequently ducing potential to the oxidoreductase complex. In addi-
washed with 0.05% (wtlvol) triphenyltetrazolium chloride tion, many respiratory enzymes are commonly assayed with
and fixed in a 5% (vol/vol) acetic acid solution. The gel is artificial electron donors or acceptors that are coupled with
then washed in water and stored in 50% ethanol before dry- the reduction or oxidation of their specific substrate. This is
ing for archival purposes. because many membrane-associated enzymes interact with
quinones that are difficult to monitor by UV-visible spec-
troscopy because of protein absorption at the same wave-
21.1.3.1.2.2. Detection of other respiratory enzymes length. Properties of commonly used artificial redox dyes are
by zymogram assay. A variety of other substrate-specific
listed in Table 2. Assays employing membrane-impermeative
oxidoreductase enzymes may be detected by using the above
artificial dyes may be performed to determine the orienta-
zymogram approach where nitrate is replaced by the desired
tion of the oxidoreductase enzyme active site on the inside
alternative substrate. For example, nitrate is replaced by 1
or outside of the cell membrane (see section 21.2.6 below).
mM N a N 0 2 to detect nitrite reductase, by 2 mM Na2Se04
Finally, since anaerobic assays require specialized tech-
for selenate reductase, by 2 mM AsHNa204for arsenate re-
niques, not all respiratory enzyme assays are equally easy to
ductase, or by 2 mM Na2S03 for sulfite reductase. Other
master.
substrates can also be tested in place of nitrate at concen-
trations in the range of 2 to 10 mM to screen for the pres-
ence of the corresponding oxidoreductase activity. 2 1.1.3.2.1. General Consideration of
Membrane-bound oxidoreductases can be identified in Enzyme Stability
an analogous fashion. However, the enzyme must first be The general assay methods described below apply to most
extracted from the cytoplasmic membranes using detergents neutrophilic gram-negative bacteria such as E. coli. The
prior to protein separation by native polyacrylamide gel assays may require adjustment of the pH, temperature,
electrophoresis. Typically, the cytoplasmic membrane frac- salinity, and other parameters for application to different
tion (section 21.2) is incubated with 2% (or higher) Triton microbes. For general methods of cell growth, harvest,
X-100 for 60 min. The sample is then subjected to ultra- breakage, and fractionation, see reference 69. Note that,
centrifugation to remove the lipid fraction prior to separa- while many respiratory enzymes are relatively stable, other
tion of the solubilized proteins by polyacrylamide gel elec- enzymes can be extremely labile. Therefore, before em-
trophoresis. The extraction efficiency of an enzyme is barking on a major study it is useful to compare enzyme ac-
dependent on its hydrophobicity and charge; therefore, the tivities in cells grown to stationary versus exponential
extraction protocol for each protein must be addressed em- phase, to evaluate methods used for cell breakage and sen-
pirically (99). For improved separation performance a de- sitivity to oxygen. Furthermore, if stability varies, compare
tergent can he included in the polyacrylamide gel (99). enzyme activities in freshly harvested cells versus frozen
The zymogram-based enzyme assay can also be per- cell material.
formed under oxygen-limiting conditions to improve the For certain detergent-solubilized preparations of mem-
assay efficiency since reduced MV is readily oxidized by any brane enzymes, lipids such as soybean asolectin or native E.
O2that has diffused into the assay chamber during incuba- coli lipids can be added to the assay mixtures to stimulate ac-
tion. To reduce autoxidation of reduced MV, it is possible to tivity (29, 80). The increase in activity can be significant
incubate the gel slices within a closed plastic container that (up to eightfold). Therefore, conditions of solubilization
has been perforated with a gas inlet and outlet port to allow and lipid addition to the assay must be considered for accu-
a constant flow of oxygen-free N2 or argon gas over the gel rate activity determinations. General methods for cell dis-
slices during the assay. To further reduce dye autoxidation, ruption by French press or cell sonication are described in
careful titration of dithionite into the assay mixture may be chapter 7 in this volume.
21. Bacterial Respiration 543

TABLE 2 Standard redox potentials of artificial and physiologicial redox substrates commonly
used to assay respiratory enzymes
Substrate Eo' (mV) Wavelength (nm) (mM-lcm-') Reference
MV - 446 603 13.7 101
BV -356 550 7.8 43
Morfamquat -374 600 9.0 43
Diquat -349 460 2.7 43
NAD+ -320 340 6.22 16
Phenosafranin -252 520 40 97
Lapachol -157 481 2.66 76
MQ - 80 263 11.3 50, 106
Qi - 70 277 106
DMN - 80 260 8.0 108
Plumbagin -40 419 3.95 76
Methylene blue + 11 578 17.1 16
PES + 55 366 5.1 16
Ascorbic acid + 58 106
PMS + 80 366 5.1 36
Ubiquinone + 90 292 13 20,31
DCIP +217 600 21.8 16
TMPD +260 606 11.6 71
Ferricyanide +408 420 1.09 16
Cytochrome c +270 550 19.7 16
"Extinction coefficient expressed at the indicated wavelength.

21.2. ENZYME LOCALIZATION tein) is an indication of an enzyme's purity. It is generally


Respiratory enzymes are not all equally positioned within the the highest in the compartment where the enzyme is lo-
cell. Some are membrane associated, while others are soluble. cated. However, activity levels may be lower if the cell frac-
If membrane associated, the enzyme may have the active site tionation method used leads to enzyme complex disinte-
for interaction with a substrate exposed to the cytoplasm or, gration or loss of essential cofactors (also see chapters 7 and
alternatively, facing the periplasmic space of gram-negative 19 of this volume).
bacteria (Fig. 1). Soluble enzymes may be located within the
cytoplasm or in the periplasmic space. A n understanding of 21.2.1. Intact Cells
the cellular location(s) for substrate utilization and product Enzyme assays are performed on intact cells to provide a
formation is useful in deciding what type of diagnostic en- starting point to define the overall level of enzyme activity.
zyme assay(s) to perform. Conversely, if the cellular location It also serves as a control for any leakage of enzyme from the
of the enzyme is unknown, it can be determined experimen- cellular preparation. Cells are harvested and washed (e.g.,
tally. Finally, knowledge of the enzyme location can be useful with 50 mM Tris-HC1 [pH 81-10 mM EDTA-0.3 M su-
in understanding the physiological role(s) of the enzyme in crose). After recentrifugation at 30,000 X g for 20 min at
cell metabolism and in energy conservation. 4"C, the cells are resuspended in the same buffer and stored
This section outlines experimental approaches to deter- on ice prior to enzyme assay. Intact cells, when assayed with
mine the cellular location of a redox enzyme following cell a membrane-permeative redox dye (Table 3), exhibit high
fractionation. While this may reveal the presence of isoen- activity (i.e., defined to be 100% activity). If little or no ac-
zymes located in distinct cellular fractions, it will, in gen- tivity is detected using an impermeative redox dye as co-
eral, not distinguish between two or more isoenzymes lo- substrate (Table 4),the enzyme is likely to be located inside
cated in the same cellular compartment. The latter issue the cell (e.g., soluble within the cytoplasm or membrane as-
may be resolved only after separating the isoenzymes chro- sociated with the active site facing the cytoplasm [Fig. 11).
matographically or by applying genetic methods to delete If high enzyme activity is detected, the enzyme is likely to
specific genes before analyzing their individual kinetic be localized on the outer surface of the cytoplasmic mem-
properties by biochemical methods. brane or located in the periplasmic space. Examples of the
To identify the cellular location of the enzyme, assays latter enzymes are the E. coli TorC TMAO reductase and
should be performed on intact cells and/or permeabilized DmsABC DMSO reductases, and an enzyme for ferrihy-
cells. This is followed by assays with cell spheroplasts, the droxide reduction (55).
periplasmic fraction, the cytoplasmic membrane fraction,
and the insoluble cytoplasmic fraction. Enzyme activity is 21.2.2. Permeabilized Cells
evaluated in total units for each cellular compartment to Cells are readily permeabilized by the addition of a deter-
identify the compartment with the highest percentage of gent such as 0.1% Triton X-100 or 0.1% dodecyl maltoside.
activity. The specific activity (units per milligram of pro- This provides an important control for enzymes that may
544 rn METABOLISM

active site soluble enzyme in periplasm


exposed to periplasm
S P
S P 1

periplasm

cytoplasmic membrane

cytoplasm

S P
active site S P
exposed to cytoplasm soluble enzyme in cytoplasm

FIGURE 1 Location of redox respiratory enzymes in the cell. The active site of a membrane-
bound oxidoreductase may face the cytoplasm (A) or the periplasm (B). A soluble-type oxidore-
ductase may be located either in the periplasmic space (C) or in the cytoplasm (D). The electron
acceptor substrate (S) is converted to the reduced product (P).

lose activity upon cell breakage. However, this activity mg/liter DNase, and stirred for 30 min under a stream of Nz
measurement may reveal limited information as to the en- at 4C. Spheroplast breakage can be facilitated by freeze-
zyme's cellular compartment. thawing the suspension.
21.2.3. Spheroplasts and the Periplasmic Fraction 21.2.5. Membrane and Cytoplasmic Protein
To separate the periplasmic proteins, gram-negative cells are Fractions
incubated in 50 mM Tris-HC1(pH 8.0)-I0 mM EDTA4.3 M To separate membranes from the cytoplasmic fraction the
sucrose for 10 min at room temperature. Lysozyme is then cell extract is centrifuged for 1 h at 100,000 X g a t 4C. The
added to a final concentration of 20 mg/liter, and the cell sus- supernatant fraction containing the cytoplasmic proteins
pension is gently stirred for 1 h at 4C. Following centrifuga- can be further concentrated as necessary and stored on ice.
tion for 5 min at 3,000 X g and 4"C, the supernatant con- The pellet containing the membranes is washed once in 50
taining the periplasmic protein complement is concentrated mM phosphate or other suitable buffer, pH 7.5, resuspended
by filter centrifugation or by other methods and stored on ice in the same buffer, and also stored on ice.
(4C). The pelleted spheroplasts are resuspended (50 mM
Tris-HC1 [pH 8.01, 10 mM EDTA, 0.3 M sucrose) and stored 21.2.6. Location(s) of Enzyme Activity
on ice. It is essential to measure the periplasmic fraction for in Cell Compartments
the presence of marker enzymes for the cytoplasmic mem- Enzyme assays are performed on the above cells and cell
brane and cell cytoplasm, e.g., succinate dehydrogenase fractions to (i) establish total activity and (ii) determine
(membrane bound) and malate dehydrogenase (soluble), to the cell compartment(s) that contains the activity. If signif-
monitor cross-contamination due to cell lysis. icant activity is detected in more than one compartment
(assuming minimal cross-contamination of fractions during
21.2.4. Cell Extracts
Pelleted spheroplasts are resuspended in 10 mM phosphate
or other suitable buffer, pH 7.5, with 2 mM MgC12, and 4
TABLE 4 Artificial redox dye substrates that are unable to
permeate to the cytoplasmic membrane
TABLE 3 Artificial redox dye substrates that are able to Dye Form Reference
uermeate to the cvtoulasmic membrane
BV Reduced and oxidized 43
Dye Form Reference
MV Reduced 43
DCIP Reduced and oxidized 51 Diquat Reduced and oxidized 43
MV Oxidized 43 PMS Oxidized 36
PES/PMS Reduced 51 Morfamquat Reduced and oxidized 43
PMS Reduced 36 Morfamquat Reduced and oxidized 16
Q, MQ, and analogs Reduced and oxidized 51 Dithionite 51
Methylene blue Reduced and oxidized 51 Ferricyanide/ferrocyanide 52
21. Bacterial Respiration 545

preparation), the cell may contain two or more distinct en- Physiological reaction: NADH + H+ + Q --+ NADf + QH2
zyme species. For example, some bacteria possess a cytoplas-
mic hydrogenase, a periplasmic hydrogenase, and/or a dis- where Q is ubiquinone and QH2 is ubiquinol. The NDH-1
tinct membrane-bound hydrogenase. Finally, detection of enzyme is energy conserving, is located on the inner face of
enzyme activity in a single compartment does not rule out the cytoplasmic membrane, and contains 13 to 14 subunits
the presence of several isoenzymes in that compartment with multiple prosthetic groups including flavin mononu-
(e.g., distinct hydrogenases). cleotide (FMN) and up to 9 iron-sulfur centers. I t is similar
to the much larger mitochondria1 complex I of eukaryrotes.
21.2.7. Determining the Orientation The NDH-1 enzymes in many bacteria and archaea are like
of the Enzymes Substrate Site that found in the well-studied microbe Escherichia coli and,
Knowing the orientation of the active site of a membrane- by inference, in Paracoccus denitrificans, Rhodobacter c a p -
bound enzyme with respect to the cytoplasmic membrane is latus, Thermus thermophilus, Klebsiella pneumoniae, and
useful in evaluating its potential bioenergetic role, and in Synechocystis (23). In contrast, the NDH-2 class of NADH
choosing appropriate enzyme assays for further investiga- dehydrogenase is composed of a single polypeptide with
tions. The active site may be facing the periplasm or facing flavin adenine dinucleotide (FAD) as the only redox group.
the cytoplasm. Active-site orientation is determined using It is not energy conserving (60). As both NDH-1 and
redox dyes as electron donors or acceptors that are not able NDH-2 utilize NADH as a substrate, specific assays have
to permeate the cytoplasmic membrane (Table 4). Whole been developed to discriminate between the two enzymes
cells exhibit little activity if the enzyme active site is lo- (60). NDH-1 utilizes both NADH and deamino (d)-
cated on the cytoplasmic side of the membrane or if it is sol- NADH as the electron donor, and enzyme turnover gener-
uble within the cytoplasm. Easily assayed cytoplasmic en- ates a membrane potential in the presence of KCN and
zymes, such as the tricarboxylic acid (TCA) cycle enzymes membrane-intrinsic ubiquinone (60).
malate and succinate dehydrogenase, are used as a control In contrast, the single-subunit NDH-2 enzyme does not
to evaluate cell intactness. When cells are permeabilized by generate a membrane potential when using NADH to re-
the addition of detergent, the redox dyes gain access to the duce ubiquinone. Thus, use of d-NADH and appropriate
active site in the cytoplasm and full activity is observed. electron acceptors can measure NDH-1 activity even in
Periplasmic active sites or enzymes are usually not affected membrane preparations containing NDH-2. I t should be
by detergent addition and exhibit full activity like that seen noted that both NADH and d-NADH are unable to per-
for intact cells. Even though most natural enzyme substrates meate the inner membrane (i.e., cytoplasmic membrane),
are charged, they can cross the cytoplasmic membrane via and thus it is essential that either inverted membrane vesi-
their cognate transporters and are thus rapidly delivered to cles or permeabilized cells be used in order to assay maximal
the enzymes active site. A number of artificial electron NDH-1 activity.
donors and acceptors unable to permeate to the cytoplasmic The d-NADH-ferricyanide reductase assay is useful for de-
membrane are listed in Table 3 (51). Note: for certain redox termining the content of functional NDH-1 in membrane
dyes, only the reduced or oxidized form is not membrane preparations. Various buffers can be used where a typical
permeative. For comparison, redox dyes that are membrane assay buffer contains 50 mM potassium phosphate (pH 7.5),
permeative are listed in Table 3. with 0.2 mM EDTA, 2 mM KCN, and 1 mM potassium fer-
A n example of the cellular orientation of fumarate re- ricyanide as the electron acceptor. Addition of the inhibitor
ductase from the rumen bacterium Wolinella succinogenes is KCN prevents the reoxidation of ferrocyanide by cyto-
described by Kroger and coworkers (51). The fumarate re- chrome oxidase. Membranes in this buffer are equilibrated
ductase assays are performed anaerobically as described in a temperature-controlled cuvette (37C). Reactions are
below on intact and lysed cells. Cell lysis is easily accom- initiated by the addition of 125 to 150 p M d-NADH, and
plished by either sonication or osmotic shock. Alter- the rapid reduction of ferricyanide is monitored in a spec-
natively, the cells can be permeabilized with a detergent as trophotometer at 420 nm. Activity is calculated by using
follows. After initiation of the fumarate reductase assay by the millimolar extinction coefficient of 1.0 for potassium
the addition of intact cells, the initial rate is recorded. A ferricyanide ( ~ 4 2 0= 1.09 mM- cm- [Table 21). The site
small volume of an aerobic detergent stock solution (e.g., 10 in the enzyme that donates electrons to ferricyanide is un-
pl of 10% Triton X-100 or 10% lauryl maltoside) is then known, although it is believed to involve an FMN molecule
added to the assay mixture and the rate is continuously at the enzyme active site since subcomplexes of NDH-1
monitored. A n increase in rate upon detergent addition with FMN still retain NADH-ferricyanide reductase activ-
suggests that the enzymes active site is oriented to the cy- ity (98).
toplasm. Note: the orientation of an enzyme active site may Physiologically, NDH- 1 reduces ubiquinone as the elec-
not be clearly resolved if the organism contains isoenzymes tron acceptor: the NADH-Q reductase assay is commonly
located in both the periplasmic and cytoplasmic compart- used to monitor the fully intact NDH-1 activity. The same
ments. buffer and incubation conditions as described above can be
used. However, ubiquinone analogs are used as the electron
acceptor in place of the natural substrate, ubiquinone-8.
21.3. AEROBIC RESPIRATORY ENZYMES One common analog is ubiquinone-1 or Q1, a ubiquinone
The following enzymes participate in aerobic electron analog with one isoprenoid unit at position 6 of the benzo-
transport pathways in many types of aerobic and facultative quinone ring, available from Eisai Co. Ltd. (Tokyo, Japan).
microorganisms. NADH:Q oxidoreductase is also present in Other quinone analogs are decylubiquinone and undecylu-
many anaerobic respiring microbes. biquinone (available from Sigma-Aldrich). However, the
quinone analogs give different rates of activity, so caution
21.3.1. NADH:Q Oxidoreductase should be exercised in comparing with NDH-1 activity
Many bacteria contain either one or two types of NADH- among different electron acceptors. Because quinone
ubiquinone oxidoreductases. For E. coli these have been analogs have limited solubility, a small amount of detergent
termed NDH- 1 and NDH-2. (0.03 to 0.1% dodecyl maltoside) is added to the assay
546 w METABOLISM

mixture prior to addition of Q1 in order to increase solubil- zyme. The activity is monitored spectrophotometrically by
ity. Following the addition of 20 to 70 FM Q1 (or another following the reduction of DCIP at 600 nm ( ~ = 621.8~ ~
quinone analog) to the assay mixture, the reaction is initi- mM- cm- [Table 21).
ated by adding 125 to 150 y M d-NADH and the reaction
rate is followed by monitoring the decrease of NADH ab- 2 1.3.2.3. Succinate Oxidation with Ferricyanide
sorbance at 340 nm ( ~ = 6.22 3 ~mM-
~ cm- [Table 21). It Ferricyanide is another electron acceptor commonly used to
is known that when NDH-1 from either bacterial or mito- assay succinate dehydrogenase activity.
chondrial preparations is solubilized with detergents, added
+
Nonphysiological reaction: succinate ferricyanide
lipids can stimulate NADH-quinone reductase activity.
Thus, for solubilized preparations of NDH-1, lipids such as
+
-+ fumarate 2H +
ferrocyanide
soybean asolectin or native E. coli lipids are added to the Succinate-ferricyanide oxidoreductase activity is determined
assay mixtures to stimulate activity (29, 80). There is a sig- in the buffer described above but containing 0.45 mM potas-
nificant (up to eightfold) stimulation of activity, so condi- sium ferricyanide ( E =~ 1.09 ~ mM-
~ cm- [Table 21) in
tions of solubilization and addition of lipids must be consid- place of PES and DCIP. The overall assay protocol is other-
ered to accurately measure complex I activity for an isolated wise the same as described above.
enzyme.
21.3.2.4. Succinate Oxidation
21.3.2. Succinate Dehydrogenase with Ubiquinone Analogs
Succinate dehydrogenase (SQR, succinate:ubiquinone Either soluble or membrane-bound forms of succinate dehy-
oxidoreductase) is the only membrane-bound enzyme of drogenase can by assayed by the PES-DCIP method (58).
the T C A cycle. It provides a dual role in generating the However, reduction of ubiquinone, or various ubiquinone
T C A cycle intermediate, fumarate, and in providing elec- analogs, can only be accomplished by membrane-bound
trons derived from succinate oxidation for the reduction forms of succinate dehydrogenase since the membrane an-
of ubiquinone to ubiquinol. Reducing equivalents are chor peptides (e.g., SdhC and SdhD) are required to form a
then passed along the electron transport chain of the cy- binding site for quinones. The latter assay is monitored by
toplasmic membrane to oxygen in bacteria and archaea the following coupled reaction.
(14, 41). + Q -+ fumarate + QH2
Reaction 1: succinate
Physiological reaction: succinate + Q -+ fumarate + QH2 Reaction 2: QH2 + DCIP,, -+ Q + DCIP,,d
The succinate:quinone oxidoreductase assay is per-
21.3.2.1. Activation of Succinate Dehydrogenase formed essentially as described above for the succinate:PES
Since succinate dehydrogenase may be partially deactivated assay. However, a ubiquinone analog is used in place of PES.
by tightly bound oxaloacetate at its active site, the enzyme Low solubility of the quinone limits its accessibility to
requires activation prior to enzyme assay by incubation with succinate dehydrogenase. Quinone reduction is therefore
either malate or succinate. To accomplish this, the succi- assayed by determination of the V,, using increasing
nate dehydrogenase preparation is diluted to 0.5 to 1.0 mg quinone concentrations. Quinones that are typically used
of protein per ml in buffer (30 mM potassium phosphate to assay succinate:quinone oxidoreductase activity include
[pH 7.41,O.Z mM EDTA, 0.1 % [wt/vol] Thesit [C12E9;poly- decylubiquinone or DB (2,3-dimethoxy-5-methyl-6-decyl-
oxyethylene dodecyl ether], and either 10 mM malonate or 1,4 benzoquinone), DPB (2,3-dimethoxy-5-methyl-6-
succinate) and incubated for 10 to 20 min at 37C . The pentyl-1,4-benzoquinone), or Q1 (Table 2). DB and DPB
activated enzyme solution can then be stored at 4C for can be obtained from Sigma-Aldrich, and Q1 may be ob-
subsequent assays. tained from Eisai Chemical Company. The hydrophobic na-
ture of quinones usually precludes using quinone analogs
21.3.2.2. Succinate Oxidation with PES or PMS that contain greater than two isoprenoid side chains in
length. The quinone analogs are dissolved in ethanol and
Succinate oxidation by succinate dehydrogenase is rou-
used in the assay at 1 to 40 pM concentrations in order to
tinely assayed spectrophotometrically using either phena-
zine ethosulfate (PES; N-ethylphenazonium ethyl sulfate
obtain V,,. DCIP reduction is stimulated by the addition
of elevated levels of the quinone analog. Usually there is a
salt) or phenazine methosulfate (PMS; N-methylphena-
short lag phase after the addition of quinone that precedes
zonium methyl sulfate) as the electron acceptor (Table 2).
the linear decrease in DCIP absorbance. The lag phase is
When using PES or PMS, the electrons are chemically me-
dependent upon the solubility of the quinone. Succinate-
diated to 2,6-dichlorophenol indophenol (DCIP) as the
quinone oxidoreductase activity is inhibited by quinone site
final electron acceptor. The coupled reactions using PES are
inhibitors, with pentachlorophenol usually effective for
shown below.
most bacterial enzymes at 20 pM. Detailed descriptions of
Reaction 1: succinate + PESO, -+ fumarate 4- PEs,d assays for both succinate oxidase and succinate-quinone ox-
idoreductase activities are provided in references 1 and 58
Reaction 2: PES,d + DCIP,, -+ PES,, + DCIP,d and references therein.
The succinate:PES (or succinate:PMS) reaction is measured
aerobically in 1-cm-path-length cuvettes at an assay tem- 21.3.3. Cytochrome Oxidase
perature of 37C. The cuvette contains 50 mM potassium Prokaryotes exhibit considerable enzymatic diversity with
phosphate (pH 7.6) (or another appropriate buffer), respect to the number and types of cytochrome oxidase en-
0.006% (wtlvol) Anapoe C12Eg (Anatrace, Maumee, OH) zymes present for reduction of molecular oxygen to water.
to maintain enzyme solubility, and 0.2 mM EDTA. Then 10 Some microbes contain only a single type of oxidase, while
mM succinate, 1.5 mM PES, and 50 yM DCIP are added to others are capable of synthesizing two, three, or more dis-
the cuvette and the reaction is initiated by addition of en- tinct enzyme types. They may differ in type of electron
21. Bacterial Respiration H 547

donor used (quinol or cytochrome c), in number and types convenient. Oxygen consumption is then monitored using
of cofactors present, and in efficiency for energy conserva- a Clark-type electrode (see above). Specific enzyme activ-
tion. This diversity may be explained in part by the ability ity is calculated in micromoles of oxygen consumed per
of bacteria to respond to environmental stress and to chang- minute per milligram of protein. If purified enzyme pre-
ing O2 conditions (70). For general reviews of cytochrome parations are used, activity may be calculated as electrons
oxidase enzymes, see references 4, 25, and 105. consumed per second per complex (i.e., the turnover
number).
General reaction: electron donor + O2
+ oxidized electron donor + 2H20 21.3.3.1.2. Oxygen Reduction with TMPD
Based on the immediate electron donor, cytochrome ox- As for the electron donor for oxygen reduction, Q1 can
idases are divided into two classes: (i) quinol oxidases that also be replaced with TMPD at a final concentration of 1
receive electrons from the membrane-intrinsic reduced mM (37, 109). Oxygen consumption is monitored using a
quinone pool (Q), i.e., cytochrome b03, ba,, and bd oxi. Clark-type electrode as described above, and enzyme activ-
dases, and (ii) cytochrome c oxidases that obtain electrons ity is calculated in micromoles of oxygen consumed per
from a small periplasmically located c-type cytochrome minute per milligram of protein.
(i.e., cytochrome aa3 and cbb3 oxidases). With the excep-
tion of the bd-type oxidase, all cytochrome oxidases belong 21.3.3.2. Cytochrome c as Electron Donor
to the heme-copper superfamily (12, 13). Both heme and The cytochrome c-type oxidases (e.g., cytochrome aa3 and
copper are important to transfer electrons to catalyze oxy- cbb3 oxidases) obtain electrons for oxygen reduction from
gen reduction within the enzyme complex. Various hemes a small soluble periplasmic c-type cytochrome in the gram-
(tetrapyrrole ring structures with a central iron atom) are negative bacteria, or from a membrane-bound c-type cy-
present in the oxidases such as a, b, d, and o heme. The tochrome located at the outer surface of the cytoplasmic
hemes differ by their tetrapyrrole ring substituents and sat- membrane in gram-positive bacteria and in archaea.
uration, which cause the heme-specific light absorbance
characteristics that can be determined by optical spec-
Physiological reaction: 4cyt c,,d + 4H+ + 0 2
troscopy. Of all oxidases, only cytochrome aa3 oxidase is
-+ 4 cyt c,, + 2H20
also present in eukaryotes, where it is located in the mito- where cyt c,d is the cytochrome c reduced form and cyt cox,
chondria (68). Several well-established assays are described is the cytochrome c oxidized form.
below as a starting point to evaluate cellular cytochrome ox- The following procedure is used to assay the cytochrome
idase levels. c oxidase of Rhodobacter sphaeroides and related organisms
(67). Membranes are prepared and resuspended by homog-
21.3.3.1. Oxygen Reduction with Ubiquinol enization in 50 mM phosphate (pH 7.4) and detergent (e.g.,
as Electron Donor 1% dodecyl-maltoside). Appropriately diluted membranes
are then added to the same buffer but containing 1.1 mg/ml
The quinol oxidases include the cytochrome bo3, ba3,and bd
asolectin (Sigma-Aldrich), and 2.8 mM ascorbate con-
oxidases that receive electrons from the membrane-intrin-
tained in the oxygen electrode chamber, and are equili-
sic reduced quinone pool. They are therefore assayed with
brated to the desired assay temperature. Cytochrome c is
the ubiquinol-1 analog, Q1,or the artificial electron donor
prereduced using ascorbate, and the reaction is initiated by
TMPD (tetramethylphenylene-diamine) as the electron addition of horse heart cytochrome c (Sigma-Aldrich) at a
donor. For both assays, oxygen consumption is monitored
final concentration of 40 pM. As described above for the
using a Clark-type electrode (Yellow Springs Instrument
ubiquinol-1 and TMPD assays, oxygen consumption is
Co. [http://www.ysi.com]). See also chapter 6 of Methods for
monitored using a Clark-type electrode where a value of
General and Molecular Bacteriology (MGMB) (8a). If the cell
237 p M 0 2 is used for the buffer, assuming air saturation at
material being evaluated contains more than one quinol-
type oxidase, the enzyme measurement will yield a combi-
37C (109). Assays of the cytochrome c-type oxidase en-
zymes (e.g., cytochrome oxidase c q ) in the gram-positive
nation of the activities. In this event, further analysis using
bacteria Paracoccus denitrificans and Bacillus subtilis are simi-
purified enzyme preparations is needed to establish the con-
lar (37, 73). Note: if a different assay temperature is used,
tributions of each complex.
the oxygen saturation value will differ accordingly (see
Physiological reaction: 2QH2 + O2+ 2Q + 2 H 2 0 chapter 19.3 in this volume).

21.3.3.1.1. Oxygen Reduction with Q1


The following procedure is used to assay cytochrome bd 21.4. ANAEROBIC RESPIRATORY ENZYMES
oxidase of Escherichia coli and related organisms (109). The following section describes a number of commonly
Membranes are prepared (section 21.2.5) and then resus- used assays for anaerobic respiratory enzymes that act on
pended by homogenizing in 30 mM Tris-HC1 (pH 7.5), 1 terminal electron acceptors, including nitrate, nitrite, nitric
mM EDTA. Appropriately diluted membranes are then in- oxide, nitrous oxide, fumarate, TMAO, DMSO, and metal
troduced into the same buffer but containing 2.5 mM oxides. Many of these respiratory enzymes are assayed by
dithiothreitol (DTT) or 4 mM ascorbate equilibrated at following the substrate-dependent oxidation of a reduced
37C. Once a stable oxygen electrode baseline signal is ob- nonphysiological electron donor such as BV, MV, or
tained, the reaction is initiated by addition of Q1 at a final phenosafranin (Table 2).
concentration of 250 FM (see section 21.3.2.4). A value of
For example: 2 BVred + 2H+ + natural substrate
237 pM O2 is calculated assuming 100% air saturation at
37C (109). Details of oxygen measurement by electrode
-+ ZBV,, + natural product
are addressed in reference 107 and chapter 17 of this vol- Alternatively, Q1,MQ, or lapachol (Table 2) can be used in
ume. The assay volume may vary depending on the sample place of the above nonphysiological dyes if the enzyme is
chamber size selected, where a 1.9-ml liquid volume is membrane bound and interacts with quinol.
548 rn METABOLISM

General reaction: MQHz + natural substrate Physiological reaction: fumarate + MQHz


-+MQ + product -+ succinate + MQ
where MQHz is menaquinol. The enzyme complex is usually present as a membrane-
The anaerobic assays described in detail below for fumarate bound menaquino1:fumarate oxidoreductase (QFR) com-
reductase can also be applied to assay many other anaerobic plex with significant structural and functional homology to
respiratory reductases (e.g., nitrate reductase, DMSO reduc- succinate:ubiquinone oxidoreductase (SQR).It should be
tase, TMAO reductase, and selenate reductase). Ferric re- noted that succinate dehydrogenases can be assayed in the
ductase and sulfur reductase activities are determined using reverse direction as fumarate reductase and vice versa.
alternative assays since reduced BV reacts directly with These enzymes have their catalytic sites inward-facing to-
Fe3+ or So and is therefore not a suitable electron donor. wards the cytoplasm (Fig. 1A).
21.4.1. Anaerobic Enzyme Assays Some bacteria that are versatile with regard to respira-
tion, such as Shewanella, contain a soluble fumarate reduc-
Since the reduced forms of many artificial electron donors
tase in the cell periplasm (Fig. 1C) (30).These are single-
have a low redox potential (Table 2) they are thus easily au-
subunit flavocytochromes that are homologous to the FrdA
toxidized by molecular oxygen. It is therefore necessary to
flavoprotein subunit of the membrane-bound fumarate re-
assay enzyme activities under strictly anaerobic conditions.
ductases. Both the soluble and membrane-bound fumarate
To ensure fully anaerobic conditions, the assay should be
reductase enzymes can be assayed using reduced BV as an
performed in anaerobic 1-ml glass cuvettes filled with an
electron donor as described below. Unlike succinate dehy-
anaerobic assay solution and sealed using butyl rubber or sil-
drogenase, fumarate reductase does not require preactiva-
icone stoppers (Fig. 2). The assay buffer solutions can be
tion to remove oxaloacetate since binding of this inhibitor
prepared in an anaerobic glove box and then introduced
is considerably weakened upon reduction of the enzyme by
into the cuvettes once they are oxygen free. The latter is ac-
the artificial electron donor.
complished by placing the cuvettes and stoppers into the
glove box for several hours before use to allow outgassing.
Alternatively, anaerobic buffer solutions can be prepared in 2 1.4.1.1.1. Fumarate Reduction by Reduced MV,
serum flasks using the Hungate technique (57, 63) and sub- BV, or Phenosafranin
sequently filled into stoppered cuvettes that have been pre- To measure the steady-state kinetics of soluble and mem-
flushed with oxygen-free nitrogen or argon gas using an N2- brane-bound fumarate reductase, the assay solution includes
flushed needle and syringe. A third approach involves the 30 mM bis-Tris-propane buffer (pH 7.0), 200 pM MV, 0.1
alternating evacuation and gassing of the stoppered cuvette mM EDTA, and 10 mM glucose in a 1-ml volume. Fumarate
containing the assay solution using an oxygen-free inert gas concentrations can be varied from 10 pM to 10 mM to ob-
(i.e., Nz or Ar). When using this approach it is important tain K,, and V,, values. After establishing anaerobic condi-
to gently tap the cuvette during the gas evacuation cycle to tions, glucose oxidase (40- to 100-pg/ml final concentra-
more efficiently remove residual Oz.To maintain anaero- tion) and 5 1.1 of catalase (diluted 50-fold from the original
bicity by scavenging trace oxygen, a solution of glucose/glu- crystalline suspension obtained from Sigma-Aldrich) are
cose oxidase/catalase can be added to the anaerobically pre- added. The cuvettes are then incubated for 7 to 10 min at
pared assay solution as an optional approach (see below for the temperature of the assay (usually 37C) to remove resid-
fumarate reduction). Small additions (up to 50 11.) to the ual oxygen. MV is reduced by the addition of sodium
stoppered anaerobic cuvettes are made from anaerobic stock dithionite from a stock solution (10 mg of Na2S204/ml,0.1
solutions using a gastight Hamilton syringe. M NaHC03, stored on ice) using a gastight Hamilton sy-
ringe until an absorbance reading at 603 nm of about 2.0 is
21.4.1 . I . Fumarate Reduction obtained. It is important to note that excess dithionite will
The ability to respire with fumarate as a terminal electron inhibit most reductase enzymes. The absorbance is moni-
acceptor is a widely distributed feature among the faculta- tored for 1 to 2 min to establish a stable baseline and to de-
tive anaerobic bacteria. Most fumarate reductase enzymes termine if all residual oxygen has been removed from the sys-
are membrane bound and have similar subunit composi- tem; no absorbance change should be observed if oxygen is
tions and structures (for reviews see references 14 and 41). absent. The enzymatic reaction is started by the addition of
a known amount of enzyme using a gastight Hamilton sy-
ringe (usually from a 0.1- to 1.0-mg protein/ml stock solu-
tion). The reaction is monitored by recording the decrease of
N, or Argon gas absorbance of MV (cbo3= 9.2 mM-' cm-' [Table 21). The
assay with reduced BV is identical to the MV assay except
that absorbance is monitored at 550 nm ( ~ 5 = 5 ~7.8 mM-'
cm-' [Table 21). As both reduced BV and MV are 1 e-
donors and fumarate is a 2 e- acceptor, by convention the
rate of the reaction is divided by 2 to express rates of fumarate
reduction. Phenosafranin, by contrast, is a 2 e- donor. The
assay with reduced phenosafranin as the electron donor is
performed as described above; however, the reaction is mon-
itored at 520 nm (qZ0 = 5.1 mM-' cm-' [Table 21).
Assays with artificial electron donors:
+ 2Hf + 2MVred -+ succinate + 2MV,,
cuvette
U fumarate
fumarate + 2H' + 2BVred -+ succinate + 2BV,,
FIGURE 2 Anaerobic cuvette gassing arrangement for per- fumarate + 2H+ + phenosafranin,,d
forming anaerobic dye-dependent enzyme assays. + succinate + phenosafranin,,
21. Bacterial Respiration w 549

21.4.1.1.2. Fumarate Reduction by Quinol trogen for amino acid, nucleoside, and cofactor biosynthe-
Various methods have been developed to measure the sis. In this case, the end product of the nitrate reduction
quino1:fumarate oxidoreductase activity of membrane-bound pathway is assimilated into the cell mass, and the pathway
fumarate reductase. As quinones have a maximum absor- is labeled assimilatory nitrate reduction (ANR). The assim-
bance at 277 nm, protein absorbance significantly interferes ilatory pathway is common to many microbes, fungi, and
with most attempts to directly monitor the formation of plants. The enzymes involved are soluble, localized in the
quinone from the reduced quinol. Thus, quinol oxidation can cytoplasm, and NAD(P)H dependent. For assays to measure
be measured effectively only with a dual-wavelength spec- the activities of the assimilatory pathway, please refer to the
trophotometer. However, alternative assays have been devel- corresponding literature (11, 21).
oped that require less sophisticated instrumentation. DNR is performed exclusively by certain bacteria, ar-
A recently developed assay system uses hydroxylated chaea, and fungi and occurs primarily under anaerobic or
naphthoquinones as electron donor substrates for anaerobic microaerophilic conditions (72, 90). Depending on the ge-
menaquino1:oxidant oxidoreductases (76). The naphtho- netic capability of the microorganism, nitrate reduction can
quinone analogs lapachol and plumbagin can be used as an proceed by the denitrification pathway to produce dinitro-
artificial electron donor (Table 2). Both compounds can be gen gas or by the ammonification pathway to generate am-
obtained from Sigma-Aldrich. The reduction of quinone is monium. Both types of DNR pathways release the end prod-
accomplished by exposing a 20 mM stock solution of either uct to the environment, in contrast to the assimilatory
compound (in ethanol containing 0.18 M HC1) to metallic pathway. Not all enzymes of the ammonification or the den-
zinc powder in a small glass vial sealed with a septum from itrification pathway participate in the generation of a pro-
which quinol can be removed using a gastight Hamilton sy- ton gradient to generate energy. They may rather serve to
ringe. Anaerobic assay conditions as described above are detoxify the harmful pathway intermediates. The following
used for the quino1:fumarate oxidoreductase assay. The cu- section provides a short summary of the denitrification and
vettes contain the glucose oxidaselcatalase O2salvaging sys- ammonification pathways as they occur in prokaryotes.
tem (section 21.4.1.1.1), EDTA, and fumarate. After prein- Denitrification proceeds via four enzymatic steps
cubation of the cuvettes at 37"C, reduced lapachol or whereby a total of five electrons are transferred from a suit-
plumbagin is added to approximately 0.3 mM. The reaction able electron donor to the substrates (nitrate and the re-
is initiated by addition of enzyme. For reduced plumbagin duced intermediates).
the absorbance is followed at 419 nm ( q 1 9 = 3.95 mM-' N03- +NO2- +N O +NzO -----+ Nz
cm-' [Table 2]), whereas for reduced lapachol the ab- nitrate nitrite nitric nitrous
sorbance is monitored at 481 nm (s4B1 = 2.66 mM-' cm-' reductase reductase oxide oxide
[Table 21). Note that the general quino1:fumarate oxidore- reductase reductase
ductase assay is useful for assay of nitrate reductase and
DMSO reductase activities (76) simply by supplying the ap- Ammonification involves only two enzymes that catalyze
propriate substrate as the electron acceptor in place of fu- the following reactions.
marate. NO3- +NO2- +NH4+
Hydroxylated naphthoquinone derivatives are highly nitrate nitrite
soluble; however, they generate high K,,, values. Thus, reductase reductase
some reductase enzymes may need to be assayed with the
more hydrophobic quinones, for which they exhibit a A number of well-characterized enzyme assays are described
higher affinity. Thus, a simple coupled enzyme assay has for the various reduction reactions. Several are provided
been developed using NADH, NAD( P)H-quinone oxi- below, and the reader is directed to more advanced descrip-
doreductase (DT-diaphorase) and MQ to measure the tive materials for additional assays and details.
quino1:fumarate oxidoreductase reaction (33,58). MQ-1 is 2 1.4.1.2.1. Nitrate Reductase
available from Eisai Co. Ltd. and is the preferred substrate
Two types of respiratory nitrate reductase enzymes have
for the reaction due to its high reactivity with fumarate re-
been described based on their location with respect to the cy-
ductase. The assay mixture contains 120 p,M NADH, fu- toplasmic membrane: these include a membrane-associated
marate reductase, and 20 to 80 p g of protein/ml of DT- NAR-type enzyme with the active site exposed in the cyto-
diaphorase (either rat or human enzyme obtained from
plasm in bacteria (Fig. 1A) and a periplasmic NAP-type en-
Sigma-Aldrich). The assay is carried out in anaerobic cue
zyme (Fig. 1C). For further details, see reference 72. Both
vettes and the reaction is initiated by the addition of 10
types of enzymes catalyze the quinol-dependent reduction of
mM fumarate. The decrease in NADH absorbance at 340 nitrate to nitrite and can be a component of either the deni-
nm ( E =~6.22 ~ mM-'
~ cm-' [Table 21) is monitored fol-
trification or ammonification pathway. The membrane-bound
lowing the addition of fumarate. This assay is very sensitive
and can be done with a variety of quinone analogs that in-
NAR complex interacts directly with the quinone pool, while
the periplasmic NAP receives its electrons via a membrane-
teract with fumarate reductase. The coupled reactions are
bound and/or a periplasmic, soluble cytochrome c.
indicated below.
NO3- + MQHz + NO2- + MQ + HzO
Reaction 1: NADH + Hf + MQ -+ NAD+ + MQHz Both NAR and NAP activities are measured essentially as
Reaction 2: fumarate + MQHz + succinate + MQ described for the fumarate reductase assay in section
2 1.4.1.1.1, with reduced BV (or MV) as the electron donor
21.4.1.2. Nitrate Reduction (Denitrification, and with 10 mM KN03 as the electron acceptor.
Ammonification, and Assimilation) NO3- + 2H+ + 2BV,d --+ NO2- + 2BV,, + H20
Nitrate reduction serves one of two general purposes (64):
(i) it is coupled to the generation of cellular energy, a 2 1.4.1.2.2. Nitrite Reductase
process that is termed dissimilatory nitrate reduction The ammonification and denitrification pathways em-
(DNR), or (ii) it functions to provide the cell with fixed ni- ploy distinct nitrite reductases. In the ammonification
550 H METABOLISM

pathway a pentaheme cytochrome c-type nitrite reductase mixture yielding approximately 100 p M dissolved N O at
reduces nitrite to ammonium. This nitrite reductase occurs room temperature. Following the addition of membranes or
in two types, a membrane-associated Nrf-type nitrite reduc- pure NOR, the reaction is initiated with ascorbate (10
tase that receives its electrons from the quinol pool and mM)-phenazine methosulfate (100 pM).
plays a respiratory role, and a soluble cytoplasmic, NADH- The activity of quinol-reactive NOR is measured with
dependent NirB-type nitrite reductase enzyme that plays a MQH2 (65 to 300 pM), Menaquinol is prepared by reduc-
role in nitrite detoxification. ing 100 pM M Q with 200 pM NADH using 0.2 U of di-
The membrane-associated Nrf nitrite reductase: aphorase (Sigma-Aldrich). It is important to choose
NADH and diaphorase concentrations that are not rate
NO2- + 3MQH2 + 2H+ + NH,+ + 3MQ + 2 H 2 0 limiting to N O reduction. This may have to be adjusted em-
pirically. Alternatively, menaquinol can be prepared by re-
The soluble NirB nitrite reductase: ducing MQ with a 10%molar excess of sodium borohydride
NO2- + 3NADH + 5H+ + NH,' + 3NAD+ + 2 H 2 0 under a nitrogen atmosphere. Cytochrome c-dependent
NORs can be assayed with 20 p M horse heart cytochrome
The denitrification pathway employs two distinctly differ- c as the electron donor (Table 2) in the presence or absence
ent types of nitrite reductases, a cytochrome cdl (NirS) and of electron mediators such as PES.
a copper-containing enzyme (NirK) (see references 100 and
110 for reviews). Both are located in the periplasmic space 21.4.1.2.4. Nitrous Oxide Reductase (NzO to N,)
and are rarely membrane bound. They receive their elec- Nitrous oxide reductases (NOS or N,OR) are copper-
trons from small periplasmic proteins, cytochrome c, or containing periplasmic soluble enzymes (Fig. 1C) that cat-
small blue copper proteins (azurin and pseudoazurin) and alyze the reduction of N 2 0 to N2 using periplasmic, soluble
reduce nitrite to nitric oxide (NO). c-type cytochromes, azurin or pseudoazurin, as their physio-
The NirS and NirK denitrification nitrite reductases: logical electron donor.
NO2- + cyt c,d + 2 H+ + N O + cyt c,, + H2O N2O + 2cyt c,,d + 2H' + N2 + H20 + 2cyt c,,
All nitrite reductases can be assayed using a BV or MV assay N2OR activity is assayed anaerobically in stoppered cu-
as described above for fumarate reductase (section vettes with reduced BV as the electron donor (see above for
21.4.1.1.1) The activity of NADH-dependent NirB-type fumarate reductase) (18, 49, 78). The assay mixture (1 ml)
enzymes is assayed by monitoring the oxidation of NADH. contains 50 mM Tris-HC1 buffer, pH 7, and 0.4 mM BV and
NirS and NirK enzyme activities can be measured using 10 is prereduced with 0.48 mM sodium dithionite. The enzyme
p M reduced cytochrome c as their physiological electron is then added to the assay and incubated at 25.0"C for 90
donor. For this type of assay, yeast or horse heart ferricy- min for complete activation of N20R. The enzyme reaction
tochrome c can be used as an electron donor (obtained from is initiated by the injection of a saturated nitrous oxide so-
Sigma-Aldrich). Samples are removed periodically and as- lution (final concentration, 1.5 mM). The oxidation of re-
sayed by colorimetric determination of the residual nitrite duced BV is monitored as the decrease in absorbance at 550
(74). Alternatively, oxidation of cytochrome c can be fol- nm (&550 = 7.8 mM-' cm-' [Table 21) as described above
lowed at 550 nm (8550 = 19.7 mM-' cm-' [Table 21). for fumarate reductase, section 21.4.1.1.1).

21.4.1.3. Anaerobic Reduction of Amine


21.4.1.2.3. Nitric Oxide Reductase (NO to NzO) N-Oxides and Sulfoxides
Three types of nitric oxide reductase (NOR) enzymes A variety of amine N-oxides and organic sulfoxides are re-
have been identified that participate in the denitrification duced as terminal electron acceptors during anaerobic cell
pathway (see references 100 and 110 for reviews). All growth by many enteric bacteria, by certain marine bacte-
NORs share a membrane localization and an outward- ria, and by nonsulfur purple bacteria (61). While some en-
facing orientation of their catalytic site (Fig. 1B). They in- zymes are specific for either DMSO or TMAO, others ex-
clude the cytochrome bc complex-type cNOR that uses hibit broad substrate specificity for these plus other related
membrane or soluble c-type cytochromes or small blue cop- amine N-oxides and organic sulfoxides. For example, the E.
per proteins (azurin and pseudoazurin) as physiological elec- coli enzyme can reduce methionine sulfoxide, nicotinic
tron donors. Another type of NOR, the cytochrome b- acid-hi-oxide, hydroxylamine, and 2-hydroxy pyridine-N-
containing qNOR, uses ubiquinol or menaquinol as the oxide (56, 104).
electron donor. The recently discovered copper-containing DMSO and TMAO reduction occurs by two types of re-
qCuANOR uses menaquinol and cytochrome c as electron ductase enzymes. The soluble, single-subunit DorA and
donors. Thus, two types of electron donors should be evalu- TorA enzymes are located in the periplasmic space of the
ated. cells (Fig. 1C) and receive electrons from a pentaheme c-
Donor type 1: 2 N 0 + 2 cyt c,d + 2H' type cytochrome (called DorC and TorC, respectively) via
+ N2O + HzO + 2cyt ,c the cytoplasmic membrane quinone pool (61 ). While DorA
catalyzes the reduction of both DMSO and TMAO, TorA
Donor type 2: 2 N 0 + MQH2 -+ N 2 0 + H2O + MQ exhibits high specificity only for TMAO. In contrast, the
NOR activity is usually assayed polarographically using a DmsABC-type enzyme is membrane bound by a hydropho-
modified Clark electrode (28, 89). Rates are determined bic anchor polypeptide, very similar to the membrane-
from the steepest slope of the curved activity trace. Briefly, bound nitrate reductase (NarGHI) enzyme, although the
DmsA catalytic site is exposed to the periplasm (Fig. 18).
the reaction is performed in a closed 1.4-ml chamber at a
temperature optimal for the enzyme. The assay chamber Physiological reactions:
contains an anaerobic buffer of 20 mM potassium phos- TMAO (or DMSO) + MQHz
phate, pH 7, which is saturated with a 5% NO-95% N2 gas + TMA (or DMS) + H Z O+ MQ
21. Bacterial Respiration 551

TMAO (or DMSO) + 2cyt cred + 2H+ respiratory (47, 66). In many microbes, the reduction is in-
-+ TMA (or DMS) + HZO+ 2cyt cox volved in detoxification (42, 75). Reduced BV may be used
as the electron donor as in the anaerobic fumarate reductase
Reduced BV ( BVrCd)is used as the electron donor in the assay (section 21.4.1.1.1) where 5 mM arsenate is used in
anaerobic assay of either the membrane-bound or the place of fumarate.
periplasmic form of the enzyme. For details, see the fu-
marate reductase assay (section 21.4.1.1.1) where 5 mM As0;- + 2H+ + 2BVIed-+ As03'- + 2BV,, + HzO
DMSO or 5 mM TMAO is used in place of fumarate.
TMAO (or DMSO) + 2BV,d + 2H' 21.4.1.4.4. Ferric Iron Reduction
+ TMA (or DMS) + 2BVOx+ HzO Ferric reductase enzymes (FER) constitute a heterogeneous
group of oxidoreductases that catalyze the reduction of ferric
Steady-state kinetics of the DMSO enzymes can be de- iron (Fe3') to ferrous iron (Fez') (55, 83). Dissimilatory fer-
termined using the dithionite-reduced MV:DMSO oxido- ric reductases function in the terminal step of the iron respi-
reductase activity. The PES-dependent dimethylsulfide ratory pathway in iron-reducing bacteria and archaea.
(DMS):DCIP oxidoreductase activity, as described by Assimilatory ferric reductases function to generate soluble fer-
McEwan and coworkers, is in principle similar to that de- rous iron that is incorporated in the prosthetic groups (hemes,
tailed above for fumarate reductase (35, 62). Fe-S centers, etc.) of metallo-redox enzymes. Both types of
ferric reductases can reduce ferric iron in the form of Fe3+-
21.4.1.4. Reduction of Metal Oxides (Selenate, citrate or Fe3+-EDTAas substrate. Alternatively, an entirely
Tellurate, Arsenate, and Iron) different class of ferric iron enzymes can interact with insolu-
A variety of metal oxides are reduced by various prokary- ble ferric iron, although the nature of these enzymes is not
otes. In some instances the reduction is coupled to an elec- well understood (for reviews see references 55 and 83).
tron transport chain, where energy is conserved. In other
instances, the reduction is driven within the cytoplasm, 21.4.1.4.4.1. Ferric reductase assay. Many ferric re-
where energy is expended. ductases are assayed using NADH or NADPH as the elec-
tron donor for iron reduction. Catalytic amounts of FMN
21.4.1.4.1. Selenate Reduction and FAD can be added to stimulate the activity.
The selenate reductase enzymes belong to the nitrate re-
ductase, formate dehydrogenase family of molybdopterin ox- NADH + H+ + 2Fe3+-EDTA -+
idoreductase enzymes. There are two distinct types that vary NAD' + 2FeZ+-EDTA+ 2H'
in cellular location (103). The Thaueru selenatis selenate re-
ductase enzyme is located in the periplasm and resembles the Ferric reductase is assayed spectrophotometrically by fol-
NAP-type nitrate reductase enzyme (46, 102). It contains lowing Fe3+-dependent oxidation of NADH or NADPH at
three subunits with molybdopterin, iron-sulfur, and b-type 340 nm (94). The assay is performed anaerobically in stop-
heme prosthetic groups. Assay of selenate reductase is per- pered quartz cuvettes to prevent oxidation of the reduced
formed in anaerobic cuvettes using reduced BV dye as the flavin by 02.The assay mixture contains 50 mM sodium
electron donor (82). The assay is performed essentially as de- phosphate (pH 7.0), 0.25 mM Fe3+-EDTA, 5 pM FMN or
scribed for fumarate reductase (section 21.4.1.1) except that FAD, and enzyme sample. The reaction is initiated by the
selenate (5 mM) is used in place of fumarate. addition of NADH or NADPH to a final concentration of
0.1 mM using a gastight Hamilton syringe, and the oxida-
Se0;- + 2H+ + 2BV,,d -+ Se03'- + ZBV,, + HzO tion of NADH or NADPH is monitored at 340 nm ( E~~~ =
The second type of selenate reductase is membrane bound 6.22 mM-' cm-' [Table 21).
and resembles the NAR-type nitrate reductase that inter- Ferric reductase activity can also be assayed anaerobi-
acts with the reduced menaquinol pool (103). Enzyme as- cally by photometric monitoring of the appearance of Fez+
says are performed as for the fumarate reductase enzyme de- by the method of Lascelles and Burke (24,54). Either 1 mM
scribed above (section 21.4.1.1.1) where Q1(Eisai Co. Ltd.) NADH or 4 mM dithionite-reduced horse heart cyto-
is used as the electron donor. Membrane preparations are chrome c is used as the electron donor, while 0.15 mM
used in place of soluble protein, and 5 mM selenate is used Fe3'-nitriloacetic acid (NTA), Fe3+-citrate, or Fe3+-EDTA
in place of fumarate. is used as the electron acceptor (purchased commercially).
The assay mixture contains Nz-saturated 50 mM HEPES
Se04'- + MQHz -+ Se03'- + MQ + HzO buffer (pH 7) with 0.5 mM ferrozine as the Fez+-chelating
agent. Horse heart cytochrome c is added from a stock solu-
21.4.1.4.2. Tellurate Reduction tion that is prepared by adding 50 ml of a 4.6 mM sodium
Tellurate is reduced by a variety of gram-negative and dithionite solution to 1 ml of 50 FM HEPES buffer, pH 7,
gram-positive bacteria. However, the enzymology of tellu- containing 2 mg of horse heart cytochrome c. The concen-
rite reduction is not as well understood as that of selenate, tration of reduced horse heart cytochrome c in the sample
fumarate, or nitrate (45,65,77).Some enzyme types are res- cuvette can be calculated by measuring the absorbance at
piratory, while others are involved in detoxification where 552 nm (&552 = 29.5 mM-' cm-'). The reaction is initiated
elemental tellurium is formed (92). Reduced BV may be by addition of whole cells or cell extracts (0.03 to 0.5 mg of
used as the electron donor as in the anaerobic fumarate re- protein) to both the sample and the reference cuvette. The
ductase assay (section 21.4.1.1.1) where 5 mM tellurate is formation of an Fez'-ferrozine complex is monitored by
used in place of fumarate. following the increase in absorbance at 562 nm ( ~ = 528.0~ ~
TeOt- + 2H+ + 2BVred-+ Te03'- + 2BVOx+ HzO mM-' cm-') (88). The reaction rate should be compared
to a reference cuvette where no electron donor was added.
2 1.4.1.4.3. Arsenate Reduction In the absence of ferrozine, the enzyme activity may also be
Arsenate reduction also occurs in a variety of gram- monitored at 552 nm, following the oxidation of the re-
negative and gram-positive bacteria. Some enzyme types are duced horse heart cytochrome c (39).
552 METABOLISM

21.4.1.5. Sulfur Oxide Reduction phorylate glucose. Glucose-6-phosphate is then consumed


Dissimilatory sulfate reduction leads to hydrogen sulfide by glucose-6-phosphate dehydrogenase to reduce NADP'
production in a variety of anaerobic habitats. Sulfate- (22, 48).
reducing bacteria include species of the gram-negative gen-
era Desulfowibno, Desulfuromonas, and Desulfobacterium; the
Reaction 1: APS + PPi + ATP + S042-
gram-positive genus Desulfotomaculum; and the archaeon Reaction 2: ATP + glucose + glucose-6-phosphate
Archaeoglobus fulgidus. Cell growth is supported by a variety Reaction 3: glucose-6-phosphate + NADP'
of organic compounds that serve as electron donors either + glucose + NADPH+
directly or indirectly. Typical substrates include lactate, ac-
etate, propionate, fatty acids, methanol, ethanol, formate, 21.4.1.5.2. APS Reductase
hydrogen, and a variety of aromatic compounds. The APS reductase enzyme assay is based on the AMP-
All enzymes involved in sulfate reduction are soluble cy- dependent reduction of the artificial electron acceptor, fer-
toplasmic enzymes, and the coupling of sulfate reduction to ricyanide ( ~ 4 2 0= 1.09 mM-' cm-' [Table 2]), using APS as
the generation of a proton motive force across the cytoplas- the electron donor (22, 48).
mic membrane is not well understood. Commonly assayed
reactions of the dissimilatory sulfate reduction pathway in- HS03- + AMP + 2ferricyanide +APS + 2ferrocyanide
clude the four enzymatic steps shown in Fig. 3. APS
Assays for the sulfate reduction enzymes are detailed reductase
elsewhere (22, 48) and described briefly here. Cell extracts
are prepared as outlined in section 21.2.4 above using an N2 21.4.1.5.3. Sulfite Reductase
gassed buffer containing the reducing agent dithiothreitol Sulfite reductase, a siroheme enzyme, reduces sulfite to
(DTT) (2 mM). Cell extracts can be assayed directly or stored sulfide and is present in most sulfate-reducing bacteria. The
under anaerobic conditions at -20 or -70C (22,48). Ifcells assay is based on the sulfite-dependent oxidation of reduced
are to be evaluated for hydrogenase and/or formate dehydro- MV dye under strict anaerobic conditions (19, 22), and is
genase activity, the investigator should also consider the cel- performed essentially as described for the fumarate reduc-
lular location(s) of these enzymes (e.g., membrane associated, tase assay (section 21.4.1.1.1). The MV dye is first reduced
cytoplasmic, or periplasmic). Please consult the section above with limiting amounts of dithionite, and the reaction is ini-
(21.2.6) for a discussion of enzyme localization. tiated by addition of cell extract where the decrease in ab-
sorption is followed at 600 nm ( ~ 6 = 0 ~13.0 mM-' cm-').
21.4.1.5.1. ATP Sulfurylase 6BVRd+ 6Hf + HS03- +6BV,,' + HS- + 3H20
ATP sulfurylase catalyzes the first step in the dissimila- Sulfite
tory pathway for sulfate reduction. It is an energy-consum- reductase
ing reaction that requires ATP to form adenosine-5-phos-
phosulfate (APS) and PPi. Due to the very negative redox 21.4.1.5.4. Pyrophosphatase
potential of the sulfate/sulfite couple (about -515 mV), The diphosphate produced in the formation of APS in
sulfate is not a suitable electron acceptor when NADH is the ATP sulfurylase reaction above is subsequently hy-
the reductant (-320 mM). Rather, sulfate must first be ac- drolyzed by the enzyme diphosphatase (pyrophosphatase).
tivated prior to its reduction. APS has a much higher redox Two moles of ATP is then required to regenerate ATP from
potential, about -60 mV, and is then easily reduced with
NADH-mediated APS reductase.
+
AMP 2 Pi. This initial reaction of dissimilatory sulfate re-
duction is identical to the assimilatory process that provides
ATP + SO4'- _ _ _ j APS + PPi the cell with sulfide for the biosynthesis of the sulfur con-
ATP sulfurylase taining amino acids (i.e., cysteine and methionine), and
coenzymes (lipoate and coenzyme A).
ATP sulfurylase activity is assayed in the reverse direc- Pyrophosphatase is assayed anaerobically by following
tion by following the pyrophosphate-dependent formation the formation of orthophosphate from pyrophosphate ( 15,
of ATP in a coupled assay. NADP' reduction is followed 48).
spectrophotometrically at 320 nm using glucokinase to phos-
PPi + HZO+ 2Pi
21.4.1.6. Formate Dehydrogenase
ATP 2e- 6e- Formate dehydrogenase (FDH) catalyzes the oxidation of
SO4- A+ APS __+ HS03- HS- formate to C 0 2and H'. Formate serves as a major electron
donor to a variety of respiratory terminal acceptors. The lo-
cation of the enzyme can vary depending on the physiolog-
PP ical role of the enzyme. For example, E. coli contains three
types of formate dehydrogenases. The one-subunit FDH-H
APS APS sulfite

-
is part of the anaerobic formate hydrogen-lyase complex
sulhrylase reductase reductase and is located in the cytoplasmic fraction of the cell (79).
In contrast, the three-subunit FDH-N is membrane bound,
with a periplasmic orientation of its active site (44). The
PP 2Pi latter enzyme serves as an electron donor to nitrate respira-
tion. A third enzyme, FDH-0, is similar to FDH-N but is
Pyrophosphatase produced in transition to anaerobic cell growth. Multimeric
periplasmic formate dehydrogenases have been isolated
FIGURE 3 Reactions of the dissimilatory sulfate reduction from Desulfovibrio species (17,85). In many bacteria the ex-
pathway. pression of formate dehydrogenase is influenced by envi-
21. Bacterial Respiration 553

ronmental conditions. Therefore, cell growth parameters duce protons and thus function to generate Hz. The en-
that are optimal for enzyme production should be consid- zymes also differ in subunit composition, electron carrier
ered prior to assay, and for determining cellular enzyme lo- specificity, cofactor content, and sensitivity to inactivation
cation(s). The reaction catalyzed by the respiratory-type by O2 (2,3). Depending on the nature of the enzyme's 0 2
FDH-N enzyme follows. sensitivity, either the protein must be maintained under
strict anoxic conditions or an enzyme activation step must
Physiological reaction: HC03- + H+ + MQ precede activity measurement.
-+ COZ + MQHz Suitable artificial electron acceptors that are commonly
A general assay capable of detecting each activity is pre- used to measure hydrogenase activity include MV, BV,
sented below. Formate dehydrogenase activity is measured methylene blue, or ferricyanide (Table 2) (81). As noted
under anaerobic conditions by following the reduction of previously, the investigator should consider conditions of
BV, methylene blue (52), or ferricyanide (Table 2). The cell growth optimal for enzyme production, and what cell
assay mixture contains 50 mM phosphate buffer (pH 7.0), compartment the enzyme is located in (i,e., active site on
0.5 mM BV, and 0.2 mM methylene blue or 1.0 mM ferri- either the inner or outer surface of the cytoplasmic mem-
cyanide. After the assay mixture has been equilibrated to brane, soluble within the cytoplasm, or within the periplas-
the enzyme's optimal temperature, enzyme is added with a mic space).
Hamilton syringe and the assay is started by the addition of
10 mM sodium formate. Certain formate dehydrogenases, 2 1.4.1.7.1. Assays and Activation Procedures
especially tungsten-containing enzymes, are sensitive to Hz oxidation activity with redox dyes as electron accep-
oxygen exposure and may require reduction for activation. tors is determined by following the reduction of MV or BV
In this case, enzyme addition is followed by approximately at 578 nm (86). The anaerobic assay mixture (e.g., 0.8 ml)
0.05 mM sodium dithionite. Dithionite will reduce some of containing 50 mM morpholinepropanesulfonic acid
the electron acceptor in the absence of formate. A lag phase (MOPS)/KOHbuffer (pH 7.0), 2 mM DTT, and either 0.5
of several minutes may precede the formate-dependent re- mM MV or BV is equilibrated with 100% Hz headspace.
duction of the electron acceptor caused by formate dehy- The assay temperature is adjusted to the optimum growth
drogenase. temperature of the microorganism. The reaction is then ini-
tiated by addition of enzyme, and the change in absorbance
21.4.1.7. Hydrogenase is followed at the appropriate wavelength spectrophotomet-
rically (Table 2).
Hydrogenase ( Hzase), the enzyme of hydrogen metabolism,
can function in bacteria for either the physiological uptake Hz + ZBV,, -+ 2BV,,d + 2H+
or the release of hydrogen gas (84). Hydrogen production
H2formation (reduction of protons) activity is measured
arises mainly as a result of fermentation of decomposing
by following the oxidation of reduced MV at 578 nm. The
organic matter by bacteria that reduce protons to Hz.
standard assay is performed essentially as described for fu-
Hydrogen consumption, on the other hand, occurs when
marate reductase (section 21.4.1.1.1) in anaerobic buffer
hydrogen gas serves as an electron donor during anaerobic
containing 50 mM MOPS/KOH (pH 7.0), 2 mM DTT, and
or aerobic respiration.
The number and location of hydrogenase enzymes vary
0.5 mM MV with an N2 gas phase. MV is prereduced with
sodium dithionite to an absorbance (A578)of approximately
in bacteria. For example, Ralstonia eutrogha contains three
2.0, and the reaction is initiated by addition of enzyme.
distinct hydrogenases involved in hydrogen uptake (10).
One, a membrane-bound enzyme with a periplasmically ori- 2H+ + 2MV,,d -+ Hz + 2MV,,
ented active site, serves as an electron donor to the respira-
NAD( P)-dependent hydrogenase activity is monitored
tory chain. A second is a soluble cytoplasmic enzyme that
by following the oxidation of NAD(P)H with K3Fe(CN)6
reduces NAD(P)+ with Hz as the electron donor to provide
as an electron acceptor, as described by Soboh et al., at 420
the cell with reducing power for other cellular reactions.
nm (86).This assay can be performed aerobically using a re-
The third is a low-activity Hz-sensing regulatory hydroge-
action mixture containing 50 mM Tris-HC1 (pH 8.0), 1.25
nase that mediates gene expression in response to Hz avail-
mM NAD(P)H, and enzyme. The reaction is started by the
ability. The facultative bacterium E. coli has four distinct
addition of 1 mM K3Fe(CN),. The assay is performed at
hydrogenases that are membrane bound and interact with
room temperature since K3Fe(CN), is chemically reduced
the quinone pool, where at least two are involved in hydro-
by NAD(P)H at a low rate at higher temperatures. Assays
gen production and one in hydrogen uptake. Several physi-
without added enzyme are used to correct for the rate of
ological reactions are as follows.
chemical K3Fe(CN)6reduction.
Hydrogen uptake reaction
Hz + MQ -+ MQHz NAD(P)H + H+ + ferricyanide
-+ NAD(P)+ + ferrocyanide
Hz + NAD(P)+ -+NADPH + HS Hydrogenase activity can also be assayed by directly fol-
Hydrogen release reaction lowing the decrease of H2 concentration amperometrically
in a closed, stirred 2.1-ml cell equipped with a Clark-type
2 H+ + 2Fd,,d + Hz + 2 Fd, electrode as described by van der Linden et al. (95). This
where Fd,d is reduced ferredoxin and Fd,, is oxidized ferre- assay can be performed aerobically (or anaerobically for
doxin. oxygen-sensitive enzyme) using 50 mM Tris-HC1, pH 8.0, to
Hydrogenases that catalyze H2reduction and/or Hz evo- which 1 p.M FMN is added as an electron mediator. For the
lution are divided into three classes based on the metal con- aerobic assay, Hz is added as Hz-saturated water to a final
tent at their active site: [NiFe] hydrogenase, [FeFe] hydro- concentration of 36 to 90 FM. Enzyme and the desired elec-
genase, and [Fe] hydrogenase (6, 96). [NiFe] hydrogenases tron acceptor, such as BV (2mM), MV (2 mM), or NAD+
generally oxidize Hz, while [FeFe] hydrogenases usually re- (5 mM), is added (10). To perform the hydrogenase assay
554 METABOLISM

anaerobically all solutions are flushed with argon, and glu- 9. Burgdorf, T., 0. Lenz, T. Buhrke, E. van der Linden,
cose (50 mM) and glucose oxidase (9 U/ml) (see section A. K. Jones, S. P. Albracht, and B. Friedrich. 2005.
21.4.1.1.1 ) are added to the assay solution to remove resid- [NiFeI-hydrogenases of Ralstonia eutrophu H16: modular
ual oxygen (95). The solution is then incubated for 3 min enzymes for oxygen-tolerant biological hydrogen oxida-
prior to the addition of NADH for enzyme activation. tion. J. Mol. Microbiol. Biotechnol. 10:181-196.
Before use, H2 must be passed over a palladium catalyst 10. Burgdorf, T., E. van der Linden, M. Bernhard, Q. Y. Yin,
(type E236P; Degussa, Dusseldorf, Germany), or alterna- J. W. Back, A. F. Hartog, A. 0. Muijsers, C. G.
tively, argon is passed through an Oxisorb cartridge (MG de Koster, S. P. Albracht, and B. Friedrich. 2005. The sol-
Industries) to remove trace amounts of O2(9). uble NAD+ ,reducing [NiFeI-hydrogenasefrom Ralstonia
eutrophu H16 consists of six subunits and can be specifically
As noted above, most hydrogenase enzymes are inhib- activated by NADPH. J. Bucteriol. 187:3122-3132.
ited by molecular oxygen. The 0 2 inhibition of [FeFe] hy- 11. Cabello, P., M. D. Rolan, and C. Moreno-Vivian. 2004.
drogenases is generally irreversible, while [NiFe] hydroge- Nitrate reduction and the nitrogen cycle in archaea.
nases can be reactivated. In [NiFe] hydrogenases Oz forms Microbiology 150:35 27-3546.
an 0 or O H bridge between the N i and the Fe atoms. This 12. Castresana, J., M. Lubben, and M. Saraste. 1995. New
bridging ligand can be removed upon reduction of the en- archaebacterial genes coding for redox proteins: implica-
zyme prior to the activity assay (38, 53). Hydrogenase acti- tions for the evolution of aerobic metabolism.J. Mol. Biol.
vation can be accomplished by incubating the enzyme with 250:202-210.
a small amount of a reduced electron donor under an H2 gas 13. Castresana, J., M. Lubben, M. Saraste, and D. G. Higgens.
phase prior to the addition of the oxidized electron accep- 1994. Evolution of cytochroine oxidase, an enzyme older
tor. For example, NAD-dependent hydrogenases are acti- than atmospheric oxygen. EMBOJ. 13:2516-2525.
vated with 5 IJ.MNADH followed by the addition of BV, 14. Cecchini, G., I. Schroder, R. P. Gunsalus, and E.
MV, or NAD+ as an electron acceptor (5 mM) to assay hy- Maklashina. 2002. Succinate dehydrogenase and fumarate
drogen oxidation. When hydrogenase activity is assayed reductase from Escherichia coli. Biochim. Biophys. Actu
with the amperometric method using methylene blue or an- 1553:140-157.
other electron acceptor, a small amount of dithionite is 15. Chen, P. S., T. Y. Toribara, and H. Warner. 1956.
added from a 2 mM stock solution until a slight reduction Microdetermination of phosphorus. Anal. Chem. 28:
of the methylene blue is observed. If desired, additional 1756-1758.
methylene blue can be added to restore the original level of 16. Clark, W. M. 1972. Oxidation-Reduction Potentials of
Organic Systems. Robert E. Krieger, Huntington, NY.
oxidized methylene blue. The reaction is initiated and the 17. Costa, C., M. Teixeira, J. LeGall, J. J. G. Moura, and I.
activity is followed spectrophotometrically at the desired Moura. 1997. Formate dehydrogenase from Desulfowibrio
wavelength (Table 2) by adding Hz-saturated buffer. desulfuricans ATCC 27774: isolation and spectroscopic
characterization of the active sites (heme, iron-sulfur cen-
2 1.5. REFERENCES ters and molybdenum).J. Biol. Inorg. Chem. 2:198-208.
1. Ackrell, B. A. C., E. B. Kearney, and T. P. Singer. 1978. 18. Coyle, C. L., W. G. Zumft, P. M. Kroeneck, H. Korner,
Mammalian succinate dehydrogenase. Methods Enzymol. and W. Jakob. 1985. Nitrous oxide reductase from denitri-
53:466-483. fying Pseudomonas perfectomarina. Purification and proper-
2. Adams, M. W. W. 1990. The structure and mechanism ties of a novel multicopper enzyme. Eur. J. Biochem. 153:
of iron hydrogenases. Biochim. Biophys. Actu 1020:115- 459-467.
145. 19. Dahl, C., N. Speich, and H. G. Truper. 1994. Enzy-
3. Adams, M. W. W., L. E. Mortenson, and J. S. Chen. mology and molecular biology of sulfate reduction in ex-
1981. Hydrogenases.Biochim. Biophys. Actu 594:105-176. tremely thermophilic archaeon Archueoglobus fulgidw.
4. Anraku, Y., and R. B. Gennis. 1987. The aerobic respi- Methods Enmmol. 243:331-349.
ratory chains of Escherichia coli. Trends Biochem. Sci. 12: 20. Ding, H., C. C. Moser, D. E. Robertson, M. K. Tokito,
262-266. F. Daldal, and P. L. Dutton. 1995. Ubiquinone pair in the
5. Antipov, A. N., D. Y. Sorkin, N. P. Lvov, and H. G. Q, site central to the primary energy conversion reactions
Kuenen. 2003. New enzyme belonging to the family of of cytochrome bcl complex. Biochemistry 34:15979-15996.
molybdenum-freenitrate reductases. Biochem. J. 369: 185- 21. Flores, E., J. E. Frias, L. M. Rubio, and A. Herrero.
189. 2005. Photosynthetic nitrate assimilation in cyanobacte-
6. Armstrong, F. A., and S. P. Albracht. 2005. [NiFeI- ria. Photosynth. Res. 83:117-133.
hydrogenases: spectroscopic and electrochemical definition 22. Frederiksen, T. M., and K. Finster. 2003. Sulfite-oxidoz
of reactions and intermediates. Phil. Trans. R. SOC.363: reductase is involved in the oxidation of sulfite in Desul-
937-954. focapsa sulfoexigens [sic] during disproportionation of thio-
7. Baker, S. C., S. J. Ferguson, B. Ludwig, M. D. Page, sulfate and elemental sulfur. Biodegradation 14: 189-198.
0. M. Richer, and R. J. van Spanning. 1998. Molecular 23. Friedrich, T. 1998. The NADH:ubiquinone oxidoreduc-
genetics of the genus Paracoccus: metabolically versatile tase (complex I) from Escherichia coli. Biochim. Biophys.
bacteria with bioenergetic flexibility. Microbiol. Mol. Biol. Actu 1364:134-146.
Rew. 62:1046-1078. 24. Gaspard, S., F. Vazquez, and C. Holliger. 1998.
8. Bock, A., and G. Sawers. 1996. Fermentation, p. Localization and solubilization of the iron(111) reductase of
262-282. In E C. Neidhardt, R. Curtiss 111, J. L. Ingraham, Geobacter sulfurreducens. Appl. Enwiron. Microbiol. 64:
E. C. C. Lin, K. B. Low, B. Magasanik, W. S. Reznikoff, M. 3188-3194.
Riley, M. Schaechter, and H. E. Umbarger (ed.), 25. Gennis, R. B. 1987. Cytochromes of Escherichia coli.
Escherichia coli and Salmonella: Cellular and Molecular FEMS Microbiol. Rev. 46:387-399.
Biology, 2nd ed. ASM Press, Washington, DC. 26. Gennis, R. B., and V. S. Stewart. 1996. Respiration,
8a.Breznak, J. A., and R. N. Costilow. 1994. Physico- p. 217-261. In F. C. Neidhardt et al. (ed.), Escherichia coli
chemical factors in growth, p. 135-154. In P. Gerhard, and Salmonella typhimurium: Cellular and Molecular Biology,
R. G. E. Murray, W. A. Wood, and N. R. Krieg (ed.), 2nd ed., vol. 1. American Society for Microbiology,
Methods for General and Molecular Bacteriology. American Washington, DC.
Society for Microbiology,Washington, DC. 27. [Reference deleted.]
21. Bacterial Respiration 555

28. Girsch, P., and S. de Vries. 1997. Purification and initial 46. Krafft, T., A. Bowen, F. Theis, and J. M. Macy. 2000.
kinetic and spectroscopic characterization of N O reduc- Cloning and sequencing of the genes encoding the
tase from Paracoccus dmitrificans. Biochim. Biophys. Actu periplasmic-cytochrome b-containing selenate reductase
1318:202-216. of Thauera selenutis. DNA Sequence 10:365-377.
29. Gong, X., T. Xie, L. Yu, M. Hesterberg, D. Scheide, T. 47. Krafft, T., and J. M. Macy. 1988. Purification and char-
Friedrich, and C.-A. Yu. 2003. The ubiquinone-binding acterization of the respiratory arsenate reductase of
site in NADH:ubiquinone oxidoreductase from Escherichia Chrysiogenes arsenatis. Eur. J. Biochem. 255:647-653.
coli. J. Biol. Chem. 278:25731-25737. 48. Kramer, M., and H. Cypionka. 1989. Sulfate formation
30. Gordon, E. H., S. L. Pealing, S. K. Chapman, F. B. via ATP-sulfurylase in thiosulfate-disproportionating and
Ward, and G. A. Reid. 1998. Physiological function and sulfide-disproportionating bacteria. Arch. Microbiol. 151:
regulation of flavocytochrome c3, the soluble fumarate 232-237.
reductase from Shewanella putrefaciens NCIMB 400. 49. Kristjansson, J.K., and T. C. Hollocher. 1980. First prac-
Microbiology 144:937-945. tical assay for soluble nitrous oxide reductase of denitrify-
31. Green, G. N., and R. B. Gennis. 1983. Isolation and ing bacteria and a partial kinetic characterization.J. Biol.
characterization of an Escherichia coli mutant lacking cy- Chem. 255 :704-707.
tochrome d terminal oxidase. J. Bacteriol. 154:1269-1275. 50. Kroeger, A. 1978. Determination of contents and redox
32. Greenberg, A. E., L. S. Clesceri, and A. D. Eaton. 1992. states of ubiquinone and menaquinone. Methods Enzymol.
Standard Methods for the Examination of Water and 53 :5 79-59 1.
Wastewater, 18th ed. American Public Health Association, 51. Kroger, A., E. Dorrer, and E. Winkler. 1980. The orien-
Washington, DC. tation of the substrate sites of formate dehydrogenase and
33. Grivennikova, V. G., E. V. Gavrikova, A. A. 'Iimoshin, fumarate reductase in the membrane of Vibrio succinogenes.
and A. D. Vinogradov. 1993. Fumarate reductase activity Biochim. Biophys. Actu 589:118-136.
of bovine heart succinate-ubiquinone reductase. New as- 52. Kroger, A., E. Winkler, A. Innerhofer, H. Hackenberg,
say system and overall properties of the reaction. Biochim. and H. Schagger. 1979. The formate dehydrogenase in-
Biophys. Actu 1140:282-292. volved in electron transport from formate to fumarate in
34. Gunsalus, R. P. 1992. Control of electron flow in Esche- Vibrio succinogenes. Eur. J. Biochem. 94:465475.
richia coli: coordinated transcription of respiratory pathway 53. Lamle, S. E., S. P. Albracht, and F. A. Armstrong. 2005.
genes. J. Bacteriol. 174:7069-7074. The mechanism of activation of a [NiFeI-hydrogenase by
35. Halnon, S. P., T. H. Toh, P. S. Solomon, R. A. Holt, and electrons, hydrogen, and carbon monoxide. J. Am. Chem.
A. G. McEwan. 1996. Dimethylsu1fide:acceptor oxidore- SOC.127:6595-6604.
ductase from Rhodobacter sulfidophilw. The purified enzyme 54. Lascelles, J., and K. A. Burke. 1978. Reduction of ferric
contains b-type haem and a pterin molybdenum cofactor. iron by L-lactate and D~-glycerol-3-phosphate in membrane
Eur. J. Biochem. 239:391-396. preparations from Stuphylococcus aureus and interactions
36. Hauska, G., A. Trebst, and W. Draber. 1973. Lipo- with the nitrate reductase system. J. Bacteriol. 134:585-559.
philicity and catalysis of photophosphorylation. 11. 55. Lovley, D. R., D. E. Holmes, and K. P. Nevin. 2004.
Quinoid compounds as artificial carriers in cyclic pho- Dissimilatory Fe(II1) and Mn(IV) reduction. Adv. Microb.
tophosphorylation and photoreductions by photosystem I. Physiol. 49:219-286.
Biochim. Biophys. Actu 5:222-232. 56. Lubitz, S. P., and J. H. Weiner. 2003. The Escherichia coli
37. Henning, W., L. Vo, V. Albanese, and B. C. Hill. 1995. ynfEFGHI operon encodes polypeptides which are par-
High-yield purification of cytochrome aa3 and cytochrome alogues of dimethyl sulfoxide reductase (DmsABC). Arch.
c a d oxidases from Bacillus subtilis plasma membranes. Biochem. Biophys. 418:205-216.
Biochem. J. 309:279-283. 57. Macy, J. M., J. E. Snellen, and R. E. Hungate. 1972. Use
38. Higuchi, Y., H. Ogata, K. Miki, N. Yasuoka, and T. of syringe methods for anaerobiosis.Am. J. Clin. Nutr. 25:
Yagi. 1999. Removal of the bridging ligand atom at the Ni- 1318-1323.
Fe active site of [NiFe] hydrogenase upon reduction with 58. Maklashina, E., and G. Cecchini. 1999.Comparison of ca-
H2, as revealed by X-ray structure analysis at 1.4 A resolu- talytic activity and inhibitors of quinone reactions of succi-
tion. Structure 7549-556. nate dehydrogenase (succinate-ubiquinoneoxidoreductase)
39. Hulse, C. L., J. M. Tiedje, and B. A. Averill. 1988. A and fumarate reductase (menaquinol-fumarateoxidoreduc-
spectrophotometric assay for dissimilatory nitrite reduc- tase) from Escherichia coli. Arch. Biochem. Biophys. 369:223-
tases. Anal. Biochem. 172:420426. 232.
40. Ingledew, W. J., and R. K. Poole. 1984. The respiratory 59. Manchenko, G. P. 1994. Handbook of Detection of Enzymes
chains of Escherichia coli. Microbiol. Rev. 48:222-271. on Electrophoretic Gels. CRC Press, Boca Raton, FL.
41. Iverson, T. M., C.. Luna-Chavez, I. Schroder, G. 60. Matsushita, K., T. Ohnishi, and H. R. Kaback. 1987.
Cecchini, and D. C. Rees. 2000. Analyzing your com- NADHubiquinone oxidoreductases of the Escherichia coli
plexes: structure of the quinol-fumarate reductase respira- aerobic respiratory chain. Biochemistry 26:7732-7737.
tory complex. Cum. Opin. Srruct. Biol. 10:448455. 61. McCrindle, S. L., U. Kappler, and A. G. McEwan. 2005.
42. Ji, G., and S. Silver. 1995. Bacterial resistance mecha- Microbial dimethylsulfoxide and trimethylamine-N-oxide
nisms for heavy metals of environmental concern. J. I d . respiration. Adu. Microb. Physiol. 50:147-198.
Microbiol. 14:6 1-75. 62. McEwan, A. G., S. J. Ferguson, and J. B. Jackson. 1991.
43. Jones, R. W., and P. B. Garland. 1977. Sites and speci- Purification and properties of dimethyl sulphoxide reduc-
ficity of the reaction of bipyridylium compounds with tase from Rhodobacter capsulatus. A periplasmic molyb-
anaerobic respiratory enzymes of Escherichia coli. Effects of doenzyme. Biochem. J. 274:305-307.
permeability barriers imposed by the cytoplasmic mem- 63. Miller, T. L., and M. J. Wolin. 1974. A serum bottle mod-
brane. Biochem. J. 164:199-211. ification of the Hungate technique for cultivating obligate
44. Jormakka, M., B. Bryne, and S. Iwata. 2003. Formate de- anaerobes. AppZ. Microbiol. 27:985-987.
hydrogenase-versatile enzyme in changing environ- 64. Moreno-Vivian, C., and S. J. Ferguson. 1998. Definition
ments. Cum. Opin. Stwct. Biol. 13:418423. and distinction between assimilatory, dissimilatory and res-
45. Klonowska, A., T. Heulin, and A. Vermeglio. 2005. piratory pathways. Mol. Microbiol. 29:664-666.
Selenite and tellurite reduction by Shewanella oneidensis. 65. MOSCOSO, H., C. Saavedra, C. Loyola, S. Pichuantes, and
Appl. Enuiron. Microbiol. 7 15607-5609. C. Vasquez. 1998. Biochemical characterization of the
556 H METABOLISM

tellurite-reducing activities of Bacillus stearothermophilusV. 83. Schroeder, I., E. Johnson, and S. DeVries. 2003.
Res. Microbiol. 149:389-397. Microbial ferric iron reductases. FEMS Microbiol. Reu.
66. Newman, D. K., E. K. Kenney, J. D. Coates, D. 27:427-447.
Ahmann, D. J. Ellis, D. R. Lovely, and F. M. M. Morel. 84. Schwartz, E., and B. Friedrich. 2006. The H2-metabo-
1977. Dissimilatory arsenate and sulfate reduction in lizing prokaryotes. In M. Dworkin, S. Falkow, E.
Desulfotomulum auripigmentum sp. nov. Arch. Microbiol. Rosenberg, K.-H. Schleifer, and E. Stackebrandt (ed.),
168:380-388. The Prohryotes, 3rd ed., vol. 3. Springer, New York, NY.
67. Pawate, A. S., J. Morgan, A. Namslauer, D. Mills, P. 85. Sebban, C., L. Blanchard, M. Bruschi, and F.
Brzezinski, S. Ferguson-Miller, and R. B. Gennis. 2002. Guerlesquin. 1995. Purification and characterization of
A mutation in subunit I of cytochrome oxidase from the formate dehydrogenase from Desulfovibrio vulgaris
Rhodobacter sphueroides results in an increase in steady- Hildenborough. FEMS Microbiol. Lett. 133: 143-149.
state activity but completely eliminates proton pumping. 86. Soboh, B., D. Linder, and R. Hedderich. 2004. A mul-
Biochemistry 41: 13417-13423. tisubunit membrane-bound [NiFe] hydrogenase and an
68. Pereira, M. M., M. Santana, and M. Teixeira. 2001. A NADH-dependent Fe-only hydrogenase in the ferment-
novel scenario for the evolution of haem-copper oxygen ing bacterium Thermoanmrobucter tengcongensis. Micro-
reductases. Biochim. Biophys. Actu 1505:185-208. biology 150:245 1-2463.
69. Phillips, A. T. 1994. Enzymatic activity, p. 555-586. In 87. Sprott, G. D., S. F. Koval, and C. A. Schnaitman. 1994.
P. Gerhardt, R. G. E. Murray, W. A. Wood, and N. R. Krieg Cell fractionation, p. 72-103. In P. Gerhardt, R. G. E.
(ed.), Methods for General and Molecular Bacteriology. Murray, W. A. Wood, and N. R. Krieg (ed.), Methods for
American Society for Microbiology, Washington, DC. General and Molecular Bacteriology. American Society for
70. Poole, R. K., and G. M. Cook. 2000. Redundancy of aer- Microbiology, Washington, DC.
obic respiratory chains in bacteria? Routes, reasons and 88. Stookey, L. L. 1970. Ferrozine: a new spectrophotomet-
regulation. Adv. Microb. Physiol. 43:165-224. ric reagent for iron. Anal. Chem. 42:779-781.
71. Prince, R. C., S. J. Linkletter, and P. L. Dutton. 1981. 89. Suharti, S., M. J. Strampraad, I. Schroder, and S. de
The thermodynamic properties of some commonly used Vries. 2001. A novel copper A containing menaquinol
oxidation-reduction mediators, inhibitors and dyes, as NO reductase from Bacillus arotoformans. Biochemistry 40:
determined by polarography. Biochim. Biophys. Actu 635: 2632-2639.
132-148. 90. Takaya, N. 2002. Dissimilatory nitrate reduction metab-
72. Richardson, D. J., B. C. Berks, D. A. Russell, S. Sprio, olisms and their control in fungi. J. Biosci. Bioeng. 94:
and C. J. Taylor. 2001. Functional, biochemical and ge- 506-510.
netic diversity of prokaryotic nitrate reductases. Cell. Mol. 91. Thauer, R. K., K. Jungermann, and K. Decker. 1977.
Life Sci. 58:165-178. Energy conservation in chemotrophic anaerobic bacteria.
73. Riistama, S., A. Puustinen, M. I. Verkhovsky, J. E. Bucteriol. Rev. 41:lOO-180.
Morgan, and M. Wikstrom. 2000. Binding of 0 2 and its 92. Toptchieva, A., G. Sisson, L. J. Bryden, D. E. Taylor,
reduction are both retarded by replacement of valine 279 and P. S. Hoffman. 2003. An inducible tellurite-resistance
by isoleucine in cytochrome c oxidase from Paracoccus den- operon in Proteus mirabilis. Microbiology 149:1285-1295.
itrificans. Biochemistry 39:6365-6372. 93. Unden, G., and J. Bongaerts. 1997. Alternative respira-
74. Robinson, M. K., K. Martinkus, P. J. Kennelly, and R. tory pathways of Escherichia coli: energetics and transcrip-
Xmkovich. 1979. Implications of the integrated rate law tional regulation in response to electron acceptors.
for the reactions of Paracoccus denitrificans nitrite reduc- Biochim. Biophys. Actu 1320:217-234.
tase. Biochemistry 18:3921-3926. 94. Vadas, A., H. G. Monboquette, E. Johnson, and I.
75. Rosen, B. P., H. Bhattacharjee, and W. Shi. 1995. Schroder. 1999. Identification and characterization of a
Mechanisms of metalloregulation of anion-translocating novel ferric reductase from the hyperthermophilic ar-
ATPase. J. Bioenerg. Biomembr. 27:85-91. chaeon Archaeoglobusfulgidus. J. Bid. Chem. 274:36715-
76. Rothery, R. A., I. Chatterjee, G. Kiema, M. T. 36721.
McDermott, and J. H. Weiner. 1998. Hydroxylated naph- 95. van der Linden, E., T. Burgdorf, A. L. de Lacey, T.
thoquinones as substrates for Escherichia coli anaerobic re- Buhrke, M. Scholte, V. M. Fernandez, B. Friedrich, and
ductases. Biochem. J. 332:35-41. S. P. J. Albracht. 2006. An improved purification proce-
77. Sabaty, M., C. Avazeri, D. Pignol, and A. Vermglio. dure for the soluble [NiFeI-hydrogenase of Ralstonia eu-
2001. Characterization of the reduction of selenate and trophu: new insights into its (in)stability and spectro-
tellurite by nitrate reductases. Appl. Enuiron. Microbiol. scopic properties. J. Biol. Inorg. Chem. 11:247-260.
6 7 5 122-5 126. 96. Vignais, P. M., and A. Colbeau. 2004. Molecular biology
78. Sato, K., A. Okubo, and S. Yamazaki. 1999. Anaerobic of microbial hydrogenases. Cum. Issues Mol. Biol. 6:159-
purification and characterization of nitrous oxide reduc- 188.
tase from Rhodobacter sphueroides f. sp. denitrificans IL106. 97. Vik, S. B., and Y. Hatefi. 1981. Possible occurrence and
J. Biochem. (Tokyo) 125:864-868. role of an essential histidyl residue in succinate dehydro-
79. Sawers, G. 1994. The hydrogenases and formate dehydro- genase. Proc. Natl. Acud. Sci. USA 78:6749-6753.
genases of Escherichiu coli. Antmie Leeuwenhoek 66:57-88. 98. Vinogradov, A. D. 1998. Catalytic properties of the mi-
80. Sazanov, L. A., J. Carroll, P. Holt, L. Toime, and I. M. tochondrial NADH-ubiquinone oxidoreductase (corn,
Fearnley. 2003. A role for native lipids in the stabilization plex I) and the pseudo-reversible active/inactive enzyme
and two-dimensional crystallization of the Escherichia coli transition. Biochim. Biophys. Actu 1364:169-185.
NADH-ubiquinone oxidoreductase (complex I). J. Biol. 99. Von Jagow, G., and H. Schager. 1994. A Practical Guide
Chem. 278:19483-19491. to Membrane Protein Purification, 2nd ed. Academic Press,
81. Schneider, K., H. G. Schlegel, and K. Jochim. 1984. San Diego, CA.
Effect of nickel on activity and subunit composition of pu- 100. Wasser, I. M., S. de Vries, P. Moenne-Loccoz, I.
rified hydrogenase from Nocardia opaca lb. Eur. J. Biochem. Schroder, and K. D. Karlin. 2002. Nitric oxide in biologi-
138533-541. cal denitrification: Fe/Cu metalloenzyme and metal com-
82. Schroder, I., S. Rech, T. Krafft, and J. M. Macy. 1997. plex NO(x) redox chemistry. Chem. Rew. 102:1201-1234.
Purification and characterization of the selenate reductase 101. Watanabe, T., and K. Honda. 1982. Measurement of ex-
from Thuuera selenatis. J. Biol. Chem. 272:23765-23768. tension coefficient of the methyl viologen cation radical
21. Bacterial Respiration 557

and the efficiency of its formation by semiconductor pho- 106. Wilson, G. S. 1978. Determination of oxidation-
tocatalysis. ]. Phys. Chem. 86:2617-2619. reduction potentials. Methods Enqmol. 54:396-410.
102. Watts, C. A., H. Ridley, K. L. Condie, 1. T. Leaver, 107. Wood, W. A., and J. R. Paterek. 1994. Physical analysis,
D. J. Richardson, and C. S. Butler. 2003. Selenate re- p. 465-51 1. In P. Gerhardt, R. G. E. Murray, W. A. Wood,
duction by Enterobacter cloacae SLDla-1 is catalysed by a and N. R. Krieg (ed.), Methods fur General and Molecular
molybdenum-dependent membrane-bound enzyme that Bacteriology. American Society for Microbiology,
is distinct from the membrane-bound nitrate reductase. Washington, DC.
FEMS Microbiol. Lett. 228:273-379. 108. Yamamoto, I., N. Okubo, and M. Ishimoto. 1986.
103. Watts, C. A., H. Ridley, E. J. Dridge, J. T. Leaver, Further characterization of trimethylamine N-oxide re-
A. J. Reilly, D. J. Richardson, and C. S. Butler. 2005. ductase from Escherichia coli, a molybdoprotein. ]. Bio-
Microbial reduction of selenate and nitrate: common chem. 99:1773-1 779.
themes and variations. Biochem. SOC.Trans. 33:173-175. 109. Zhang, J., P. Hellwig, J. P. Osborne, H. W. Huang, P.
104. Weiner, J. H., D. P. Macisaac, R. E. Bishop, and P. T. Moenne-Loccoz, A. A. Konstantinov, and R. B.
Bilous. 1988. Purification and properties of Escherichia Gennis. 2001. Site-directed mutation of the highly con-
coli dimethyl sulfoxide reductase, an iron-sulfur molyb- served region near the Q-loop of the cytochrome bd
doenzyme with broad substrate specificity. J. Bacteriol. quinol oxidase from Escherichia coli specifically perturbs
170~1505-1511. heme b595. Biochemisq 40:8548-8556.
105. Wikstrom, M. 2004. Cytochrome c oxidase: 25 years of the 110. Zumft, W. G. 1997. Cell biology and molecular basis of
elusive proton pump. Biochim. Biophys. Acta 1655241-247. denitrification. Microbiol. Mol. Biol. Rev. 61:533-616.
22
Carbohydrate Fermentations
R . MEGANATHAN. YAMINI RANGANATHAN. AND C. A . REDDY

22.1. FERMENTATION PRODUCTS FROM PYRUVATE .............. 559


22.2. FERMENTATION BALANCE ............................... 559
22.2.1. Construction of Fermentation Balance
.2. Carbon Recovery
.................... 560
...................................
559
.3. Oxidation/Reduction Balance .......................... 560
.4. Available Hydrogen Balance ........................... 560
22.3. ALCOHOLIC FERMENTATION ............................. 560
22.3.1. Embden-Meyerhof-Parnas Pathway ...................... 560
.2. Entner-Doudoroff Pathway ............................ 561
.3. Radiorespirometry .................................. 561
22.4. LACTIC ACID FERMENTATION ............................ 562
22.4.1. Homolactate Fermentation ............................ 562
.2. Heterolactate Fermentation ............................ 563
.3. Heterofermentative Bifidum Pathway .................... 563
22.5. BUTYRIC ACID-BUTANOL AND ACETONE-ISOPROPANOL
FERMENTATIONS ....................................... 563
22.6. ETHANOL-ACETATE FERMENTATION ...................... 564
22.7. MIXED ACID AND BUTANEDIOL FERMENTATION ............ 564
22.7.1. Pyruvate-Formate Lyase .............................. 565
.2. a-Acetolactate Synthase .............................. 566
.3. Formate Hydrogen Lyase ............................. 566
..4.5. Acetaldehyde Dehydrogenase and Alcohol Dehydrogenase ...... 566
Fumarase ......................................... 566
22.8. PROPIONATE AND SUCCINATE FERMENTATION ............ 567
22.8.1. Acrylate Pathway ................................... 567
.2. Succinate-Propionate Pathway .......................... 567
22.9. ACETATE FERMENTATION BY BACTERIA ................... 568
22.10. ACETATE FERMENTATION I N HYPERTHERMOPHILIC
ARCHAEA ............................................. 569
22.10.1. Kinases .......................................... 569
22.10.1.1. Phosphorylation of Glucose ................... 569
.2. Phosphorylation of Fructose-6-Phosphate .......... 569
.3. Bifunctional Kinase ......................... 570
22.10.2. Oxidation of Glyceraldehyde-3-Phosphate ................. 570
.3. Conversion of Acetyl Coenzyme A to Acetate .............. 570
22.11. ASSAYTABLES .......................................... 571
22.12. REFERENCES ........................................... 582

Fermentation was defined by Pasteur as la vie sans air part of their ATP for growth by anaerobic respiration.
(life without air) during the middle of the 19th century. which is defined as an energy-yielding metabolism in
Even though in common usage now. the term fermentation which either an organic compound (such as formate) or in-
is used for an aerobic process (as in antibiotic production) organic compound (such as H2) serves as an electron donor
or an anaerobic process (as in lactic acid production). and an organic compound (such as fumarate) or inorganic
strictly speaking. fermentation is defined as an energy- compound (such as nitrate. sulfate. or C02) serves as an
yielding metabolic process (in the dark) in which an or- electron acceptor.
ganic compound(s) serves as both an electron donor and an Historically. studies on ethanolic fermentation of glu-
electron acceptor. Fermentations are carried out by obligate cose via the glycolysis or the Embden-Meyerhof-Parnas
anaerobes or facultative anaerobes. and ATP is formed by (EMP) pathway were very important because these led to
substrate level phosphorylation.Facultative anaerobes such profound fundamental discoveries that laid the foundations
as enterobacteria grow by fermentative metabolism under for modern physiology and biochemistry as well as for the
anaerobic conditions. while under aerobic conditions they fermentation industry. More than one hundred years after
grow as aerobic heterotrophs . Obligate anaerobes. on the these initial investigations. important new discoveries con-
other hand. obtain energy for growth mostly by fermenta- tinue to be made in the fermentation field. Recently. some
tive metabolism. However. a few anaerobes obtain at least important discoveries were made in the study of glycolysis.

558
22. Carbohydrate Fermentations 559

These include for the first time the rather dramatic and star- a compound derived from pyruvate (Fig. 1). The type of
tling demonstration that ADP, rather than ATP, is the electron acceptor compound formed leading to the produc-
phosphoryl donor for the phosphorylation of glucose and tion of a given end product of fermentation is determined
fructose-6-phosphate in thermophilic archaea. Another by the substrate fermented, the organism responsible for the
surprising finding is the reported presence of a bifunctional fermentation, and various physiological factors.
kinase capable of phosphorylating both glucose and fruc- An examination of Fig. 1 also shows that formation of
tose-6-phosphate using ADP as the phosphoryl donor in the acetyl coenzyme A (acetyl-CoA) from pyruvate cleavage or
hyperthermophilic archaeon Methanococcus jannaschii. from acetate activation is a key reaction in the pathways
This chapter provides a brief review of some of the more presented. Acetyl-CoA is converted to acetate via acetyl-
common carbohydrate fermentation pathways, schematics of PO4 or undergoes condensation with another molecule of
the flow of carbon in these pathways, characteristic products acetyl-CoA to eventually form butyryl-CoA, or undergoes
produced, typical energy yields (in the form of ATP), and reduction to form ethanol. Furthermore, it is worthy of note
assay procedures for the key enzymes. The reader is referred that pyruvate undergoes two types of clastic reactions: one
to a wide array of books and reviews (18, 32,60, 68, 76, 103, yields acetyl-CoA and formate (coli-aerogenes type), while
118, 123) that give more details on the topics covered here. the other yields acetyl-CoA, CO2, and Hl (clostridial type).
Anaerobic respiratory processes such as sulfate reduction are Fermentative organisms are constrained to grow and survive
not covered in this chapter, but some aspects of bacterial res- mostly on substrate level phosphorylation that results in rel-
piration are covered in chapter 21 of this volume. atively poor energy yield per mole of substrate fermented, in
contrast to aerobic respiring organisms that produce large
amounts of ATP by oxidative phosphorylation (in addition
22.1. FERMENTATION PRODUCTS to substrate level phosphorylation). Thus, the inherent lim-
FROM PYRUVATE itations in energy yield in fermentation necessitate catabo-
Fermentations are classified according to the key fermenta- lism of large amounts of substrate, resulting in the accumu-
tion end products of each as exemplified by bacterial lation of large quantities of end products. Efficient and
ethanolic, homolactic, heterolactic, propionic, mixed-acid, economic production of fermentation products such as
butyrate-butanol, homoacetogenic, and other fermenta- ethanol is currently a multibillion-dollar industry and is ex-
tions (see below). Pyruvate is a key intermediate in many of pected to grow in the future.
these fermentations; i.e., substrate carbon flows via pyru-
vate to produce an end product(s) characteristic of a given
fermentation. The various fermentation products derived 22.2. FERMENTATION BALANCE
from pyruvate are shown in Fig. 1. Microorganisms catabo-
lize sugars to pyruvate and obtain energy for growth by sub- 22.2.1. Construction of Fermentation Balance
strate level phosphorylation. Since by definition, a fermen- The basic requirement for studying fermentation is to de-
tative organism is unable to use O2as an electron acceptor, termine the optimum conditions for the growth of the or-
it has to generate its own alternate electron acceptor. This ganism and determine the fermentation products quantita-
alternate electron acceptor to oxygen is usually pyruvate or tively. A variety of media used for growing of bacteria is

2H CO? 2H

Clostridia
Acetokctate
HO-FH-COOH
CH2-COOH
Malate CH3-COOH Acetyl-CoA
Acetate Ethanol H-C-OH
p 2 0 v I
c=o
CH2-CO- CHYCOOH
CH-COOH Acetoacitate I
I1 CH3
HOOC-CH Acetom
Fumarate A

CH3-CO-CH3 CH3-CH2-CH2-COOH
2HJ
CH2-COOH
Acetone Butyrate 2H/
#
\2H
I
I CH3 CH3
CH2-COOH I I
Succinate CH3-CHOH-CH3 CH3-CH2-CH2-CH20H
H-C-OH c=o
I I
Isopropanol Butanol H-C-OH c=o
bCO2 I I
CH3-CH2-COOH CH3 CH3
2.3-Buta~dbl Dlacetyl
Propionate

FIGURE 1 Fermentation products formed from pyruvate by various organisms.


560 rn METABOLISM

listed in chapter 10 in this volume. A complete description 22.2.3. Oxidation/Reduction Balance


of methods used for the determination of fermentation For the calculation of O/R balance, it is usually customary
products is beyond the scope of this chapter, but an excel- to assign an arbitrary O/Rvalue of 0 for formaldehyde. Each
lent review on the chemical methods for the analysis of fer- excess 2H is counted as - 1, and deficiency of 2H is counted
mentation products is that by Dawes et al. (24). Also see as +1 (32, 118). However, in a simpler alternative proce-
chapter 18 in this manual. With the recent advances in dure, hydrogen is assigned an arbitrary value of -0.5 and
technology, chemical methods have been superseded by
techniques such as high-performance liquid chromatogra-
+
oxygen is assigned a value of 1. The two methods yield
the same results.
phy and liquid chromatography/mass spectrometry. High-
performance liquid chromatography is now the most com- 22.2.4. Available Hydrogen Balance
mon method used for the analysis of organic acid and For the calculation of the available hydrogen balance, it is
solvents produced during fermentation (42, 108, 113). customary to determine the number of available hydrogens
Fermentatively produced gases can be determined with an in the substrate and the products of fermentation. During
acoustic gas monitor (17, 42). Also see chapters 17 and 18 fermentation, the products derive their hydrogens from the
in this volume for selected analytical procedures. substrate and water. Hence, the substrate and products are
Since fermentation often involves reactants, the sub- completely oxidized to COZ with water and the numbers of
strate, and water only, stoichiometry of the substrate con- available hydrogens are determined.
sumed and products formed in fermentation can be deter- From the nature and quantity of the fermentation prod-
mined accurately and this bookkeeping serves as an ucts formed, it is often possible to deduce the pathway. This
invaluable tool that will aid in deducing the presumed path- information can be verified by radiorespirometry (see sec-
way employed by the organism. In the construction of a fer- tion 22.3.3 below) and other techniques (see chapters 17
mentation balance, usually the carbon recovery, oxida- and 18), and the validity of the results can be further con-
tionlreduction (Om) balance, and a balance of available firmed by the assay of key enzymes of the pathways.
hydrogens are determined. Data on butanol-isopropanol
fermentation carried out by Closhidium beijerinckii (syn.
Closhidium butylicum) were presented in the classic review 22.3. ALCOHOLIC FERMENTATlON
by Wood (123). A typical fermentation balance is illus- Understanding of microorganisms as agents of fermentation
trated in Table 1. largely came from detailed studies on alcoholic fermenta-
tion in which breakdown of glucose to pyruvate occurs by
22.2.2. Carbon Recovery one of two main pathways: the EMP pathway and the
The carbon recovered in the fermentation shown in Table Entner-Duodoroff (ED) pathway. Other six-carbon sugars
1 is about 96%, which is in agreement with the expected are generally interconverted to glucose by the action of ac-
value. It is not uncommon in certain fermentations to re- cessory cellular enzymes.
cover significantly more carbon than expected. Such an in-
crease in recovered carbon indicates C 0 2 fixation (32, 22.3.1. Embden-Meyerhof-Parnas Pathway
123). In anticipation of this, it is customary to include bi- Much of our understanding of early fermentations came
carbonate in the fermentation media (see chapter 10). from alcoholic fermentation of sugar by the yeast Saccha-

TABLE 1 Glucose fermentation by Clostridium butylicum:calculation of carbon recovery, O/R balance, and available hydrogen
balance
O/Rbalance Available hydrogen balance
Substrate or mo1/100 mol mol of
product [I] of substrate" [2] carbonb[3] O/R O/R value mo1/100 ml' [5] Available Available He
value [4] Reduced Oxidized Hd [6] (mo1/100 mol) [7]
Substrate
Glucose 100 600 0 - 24 2,400
Products
Butyrate 17 68 -2 -34 20 340
Acetate 17 34 0 - 8 136
co2 204 204 f 2 +408 0 -
H2 78 - -1 -78 2 156
Butanol 59 236 -4 -236 24 1,416
Isopropanol 12 36 -3 -36 18 216

Total 578 -384 +408 2,264


"Data in this column are taken from reference 123.
bThis value is the product of the number of carbons X the number in column [2]. Carbon recovered = 578/600 X 100 = 96.3%; O/R balance = 408/384 = 1.06;
balance of available H = 2,400/2,264 = 1.06.
'This value is the product of column [2] X column [4].
dCalculation of available [HI is based on the following equations: C6H1206+ 6 H 2 0 + 24H + 6H2Q C4H802+ 6 H 2 0 + 20H + 4 c 0 2 ; C2H,02 + 2 H 2 0+ 8H
+ 2C02; C4HloO + 7H2O + 24H + 4 c o 2 ; C,H80 + 5H20 + 18H + 3 c o 2 .
T h i s value is the product of column [2] X column [6).
22. Carbohydrate Fermentations w 561

H-C=O H-C=O CH20H


I
H-C-OH
1 I
I ATp ADP H-c-oH
HO-C-H di% H0-C-HI
I
H-C-OH
I
H-C-OH
-2' I
H-C-OH
I I I I I
H-C-OH H-C-OH H-C-OH H-C-OH H-C-OH
I I I
CH20H CHzO@ CHzO@

COOH2A~mp$X30H 2 H20 COOH COOH coo@


I
2H-C-OH &2H-&-OH
I = 0 *2C-O@
2& II 2H--t-O@
CHzOH I
CHzO@ 7 I
CH3 CHZ CHzO@

2 c'H3 -CHO 2 CH3 -CHzOH


2NADH + H'
ZNAD'
FIGURE 2 Ethanolic fermentation of glucose by the EMP pathway. 1, hexokinase; 2, glucose-6-
phosphate isomerase; 3, PFK; 4, fructose bisphosphate aldolase; 5 , triose phosphate isomerase;
6, GAPDH; 7, 3-phosphoglycerate kinase; 8, phosphoglycerate mutase; 9, enolase; 10, pyruvate
kinase; 11, pyruvate decarboxylase; 12, ADH.

romyces cereuisiae. This is the classical fermentation em- pathway are 6-phosphogluconate dehydratase and 2-keto-3-
ployed in the manufacture of wine and other popular alco- deoxy-6-phosphogluconate(KDPG) aldolase. In contrast to
holic drinks. In this fermentation, glucose is degraded via the EMP pathway, which yields 2 mol of ATP per mol of
glycolysis or the EMP pathway to produce 2 mol of ethanol glucose fermented, the ED pathway yields only 1 mol of ATP/
for each mole of glucose metabolized (Fig. 2). Yeast cells mol of glucose. The low ATP yield might explain why the
produce three enzymes for the phosphorylation of glucose presence of the ED pathway in anaerobes is rare in spite of
and other sugars: hexokinases 1 and 2 and a glucokinase its widespread occurrence in aerobes.
(31, 37). Hexokinase 2 is thought to be the major enzyme For demonstrating the presence of the ED pathway in
during growth on glucose since it is the isozyme that pre- a n organism, extracts from glucose-grown cells are assayed
dominates. The other two isozymes present are individually for the key enzymes, dehydratase and KDPG aldolase. The
sufficient to support growth on glucose, but their expression presence of these enzymes and the absence of phospho-
is higher when the cells are grown on other carbon sources fructokinase (PFK) (the key enzyme of the EMP pathway)
(37, 61). Further, hexokinase 2 plays an important role in are clear indicators for the operation of the ED pathway
the regulation of glucose repression (29, 30). (Fig. 3). The two molecules of pyruvate formed are decar-
In alcoholic fermentation, 2 mol of pyruvate is produced boxylated to acetaldehyde and then reduced to ethanol by
for each mole of glucose degraded. The key enzyme that is alcohol dehydrogenase (ADH). It should be mentioned
unique to the EMP pathway in the formation of pyruvate is that there are two ADHs involved in the conversion of ac-
6-phosphofructokinase. All the other enzymes are involved etaldehyde to ethanol. These enzymes are designated as
in other pathways or reactions of intermediary metabolism. ADH I and ADH 11. These enzymes constitute approxi-
The unique enzyme that is involved in the conversion of mately 20 and 80% of the total ADH activity. ADH I con-
pyruvate to ethanol is pyruvate decarboxylase. The electron tains zinc, while ADH I1 contains ferrous iron as a cofac-
acceptor is the acetaldehyde. The 2 mol of NADH pro- tor. For further information, the reader should consult the
duced in the glyceraldehyde-3-phosphate dehydrogenase original publications (5, 22, 52, 62, 72). The assay meth-
(GAPDH) reaction are consumed in the reduction of ac- ods for the various enzymes of the pathway are shown in
etaldehyde to ethanol, resulting in an even hydrogen bal- Table 3 (see section 22.11).
ance. The net energy yield of yeast ethanolic fermentation
is 2 mol of ATP/mol of glucose consumed (Fig. 2). The assay 22.3.3. Radiorespirometry
methods for the various enzymes of the pathway are shown From the nature and quantity of the fermentation products
in Table 2. (See assay tables in section 22.1 1.) formed, it is possible to deduce the likely pathway. This in-
formation can be confirmed by radiorespirometry. For ex-
22.3.2. Entner-Doudoroff Pathway ample, radiorespirometry is a useful method for distin-
Zymomonas mobitis is the organism used in the fermentation guishing between the EMP pathway and the ED pathway.
of agave juice to Mexican pulque and tequila (111); it fer- The expected labeling patterns during fermentation of glu-
ments glucose via the ED pathway, resulting in the produc- cose by the EMP and ED pathways are shown in Fig. 4.
tion of 2 mol each of ethanol and COz from each mole of Each pathway produces 2 mol of pyruvate for each mole of
glucose (Fig. 3). 2. mobilis contains pyruvate decarboxylase, glucose consumed. However, the pyruvate originating from
which is unusual for a bacterium. The key enzymes of the the first three carbon atoms of glucose is different. In the
562 METABOLISM

H-C=O H-C=O
I I
H-C-OH H-C-OH
I
HO-C-H
I
H-C-OH
I I
H-C-OH H-C-OH
I
CH20H

HO-C-H
I
2 CH3 -CHO
H-C-OH
I 6 k 2 co2
H-C-OH
I
COOH
I I
c=o c=o
I
CH3 CH3
YOOH 4

H-C-OH
I
CH20@ H-C-OH
I
CH20@

FIGURE 3 Ethanolic fermentation of glucose by the ED pathway. 1, glucokinase; 2, glucose-


6-phosphate dehydrogenase;3, 6-phosphogluconate dehydratase; 4,KDPG aldolase; 5, GAPDH; 6,
3-phosphoglyceratekinase; 7, phosphoglycerate mutase; 8, enolase; 9, pyruvate kinase; 10,pyruvate
decarboxylase; 11, ADH.

ED pathway, the carbon one (aldehyde group) of glucose where hexokinase is used (76, 78). Glucose-6-phosphate is
becomes the carboxyl of pyruvate, while in the EMP path- fermented by the EMP pathway to pyruvate. The 2 mol of
way, the aldehyde group becomes the methyl group of pyru- pyruvate produced is reduced using the 2 mol of NADH de-
vate. The converse is true in the case of carbon 3 of glucose rived from the GAPDH reaction. This fermentation yields
in the EMP pathway. If glucose 14Clabeled in carbons 1 , 3 , 2 mol of ATP/mol of glucose fermented. The assay methods
and 4 is fermented, and if the organism uses the EMP path- for the various enzymes of the pathway are shown in Table
way, carbons 3 and 4 will appear as COz. O n the other 4 (section 22.11).
hand, if the ED pathway is used, then carbons 1 and 4 will
appear as C02(Fig. 4).
1 H-C=O 1 CH3 lCH3
I I I
22.4. LACTIC ACID FERMENTATION 2 H-C-OH
I 2y=O 2CHzOH

-
A group of bacteria commonly referred to as lactic acid bac-
teria ferment glucose and produce lactic acid. If a bacter-
I \ I
ium ferments each mole of glucose to 2 mol of lactate (lac- I
5 H-C-OH 5 k=O 5CH20H
tate is the sole product), then the pathway is referred to as I I I
homolactate fermentation. If glucose is fermented to lac- 6 CHzOH 6 CH3 6CH3
tate, ethanol, and COz, then it is referred to as heterolac-

-
'
tate fermentation. Depending on the organism, the lactic 1 H-C=O
I
1 COOH
I
p* I
acid bacteria produce either L-( +), D-(-) or, DL-lactic acid 2c=o 2CH20H
2 H-C-OH

-{ -
(123). The lactic acid produced by muscle, on the other I
hand, is L-( +). 3HO-C-H ED 3CH3 3CH3

22.4.1. Homolactate Fermentation


4 H-C-OH
I
4;OOH 1-
The homolactate fermentation pathway is shown in Fig. 5 . 5 H-C-OH 5C=O 5CH20H
I I
The lactic acid bacteria usually phosphorylate glucose 6 CH2OH 6 AH3 6CH3
by the phosphoenolpyruvate phosphotransferase system
(PEP-PTS). (For an excellent review on the genomic analy- FIGURE 4 Flow of carbon when ''[C]glucose is fermented
sis of PEP-PTS, see reference 6a). But there are instances to ethanol and COZ via the EMP and ED pathways.
22. Carbohydrate Fermentations m 563

Glucose& Glucose-6-@ 2Fructose-6- @ 22.4.2. Heterolactate Fermentation


The initial transport and phosphorylation of glucose in
heterolactate fermentation (Fig. 6) occur the same way as
in homolactate fermentation. The glucose-6-phosphate is
Fructose-I , 6-bisphosphate converted to gluconate 6-phosphate by the same reactions
as in the ED pathway. In the subsequent reactions, the
gluconate 6-phosphate is decarboxylated to ribulose 5 -
2 Lactate 2 Glyceraldehyde-3-@ phosphate and epimerized to xylulose 5-phosphate. A thi-
amine pyrophosphate-dependent phosphoketolase cleaves
the xylulose !%phosphate to G A P and acetylphosphate.
The GAP is converted to lactate by the same reactions as
in the homolactate pathway. The acetylphosphate is first
- - -~ N
7 k -- 2 A
NDm H+ +3H2+ p i converted to acetyl-CoA by a phosphotransacetylase and
I f reduced to acetaldehyde and, finally, to ethanol. The ATP
2 Pyruvate 2 1,3-bisphosphoglycerate yield is 1 mol/mol of glucose fermented. The assay meth-
ods for the various enzymes of the pathway are shown in
Table 5 (section 22.11).
FIGURE 5 Homolactate fermentation. 1, PEP-PTS; 2 to 6, 22.4.3. Heterofermentative Bifidum Pathway
conversion of glucose-6-phosphate to pyruvate, which is ac-
complished by the same enzymes as in Fig. 2; 7, lactate dehy- The transport and phosphorylation of glucose in the bifido-
drogenase. bacteria are similar to that described above for homo- and
heterolactic bacteria. In the bifidum pathway, Bifido-
bacterium bifidum ferments glucose to lactate and acetate
(Fig. 7). There are two mechanistically identical phospho-
clastic reactions that cleave fructose-6-phosphate and xylu-
lose 5-phosphate with inorganic phosphate. In human-
derived species like Bifidobacterium dentium, there is a sepa-
Glucose rate enzyme for each substrate, namely, a fructose-6-phos-
phate phosphoketolase and a xylose 5-phosphate phospho-
1
Glucose-6-@
ketolase. In contrast, in animal-derived species such as
Bifidobacterium globosum and Bifidobacterium lactis, a single
[Ethanol] enzyme cleaves both substrates (66, 77).

6@gluconate
2K:tz + H' There is no reduction or oxidation of NAD+ in this
pathway until GAP is formed. The net result is the forma-
Acetaldehyde tion of l mol of lactate and 1.5 mol of acetate per mol of
CO2+ :
EH + H+ glucose fermented. The ATP yield of the pathway is higher
H2-C-OH than that of other fermentative pathways: 2.5 mol ATP/mol
I of glucose fermented. The assay methods for the various en-
c=o zymes of the pathway are shown in Table 6 (section 22.1 1).
I
H-C-OH Acetyl -CoA
I
H-C-OH
I 22.5. BUTYRIC ACID-BUTANOL A N D
CH2OQ CoA
ACETONE-ISOPROPANOL FERMENTATIONS

-+
41

H2-
4
C-OH
CH3-CO-OP03H2 Butyrate as a major fermentation product of glucose fer-
mentation is primarily seen in anaerobes such as clostridia
I and in strains of genera Butyriuibno, Fusobacterium, and
I &Po4 Eubacterium. The clostridia employ PEP-PTS for the uptake
HO-C-H and phosphorylation of glucose to glucose-6-phosphate
I
H-C=O (Fig. 8). A full description of phosphotransferase is beyond
c=o
I
H-C-OH
CHzO@ I
H-?-OH the scope of this chapter, but recent reviews (28, 68) pro-
vide more details. The glucose-6-phosphate formed is con-
verted to 2 mol of pyruvate by reactions of the EMP path-
way (as shown in Fig. 2 ) . Pyruvate is then oxidized to
2A T P d acetyl-CoA, H2, and C02(Fig. 8) mediated by pyruvate-
ferredoxin oxidoreductase (reaction 1 below) and ferre-
doxin-linked hydrogenase (reaction 2 below).
Reaction 1: pyruvate + ferredoxin + CoA
-+ acetyl-CoA + ferredoxin * H2
Reaction 2: ferredoxin H2 + ferredoxin + H2
*
FIGURE 6 Heterolactate fermentation. 1, PEP-PTS; 2 , glu-
cose-6-phosphate dehydrogenase; 3, 6-phosphogluconate de- Evolution of H2 is an advantage to the organism, as it is an
hydrogenase; 4, ribulose 5-phosphate 3-epimerase; 5, phospho- effective way to dispose of the electrons and protons released
ketolase; 6, conversion of GAP to pyruvate, which is during pyruvate oxidation in that, even in an environment
accomplished by the same enzymes as in Fig. 2 and 3; 7, lactate containing high levels of hydrogen gas, reduced ferredoxin
dehydrogenase; 8, phosphotransacetylase; 9, acetaldehyde de- can transfer electrons to hydrogenase to produce H2. Some
hydrogenase; 10, ADH. of the clostridia also contain a pyridine nucleotide-linked
564 METABOLISM

2 Glucose CoA by another mole of NADH. The butyryl-CoA under-


goes a phosphorylytic cleavage resulting in the formation of
11 the high-energy phosphorylated intermediate butyryl phos-
phate, which in turn is converted to butyrate with the con-
CHzOH CHzOH comitant phosphorylation of ADP to ATP by substrate level
I I
c=o Acetyl-@-[=] c=o phosphorylation. The reactions involved in the conversion
I I
H-C- H H-C=O
I
f
H - L - o H ~ H-C-OH
HO-C-H
I
of a mole of glucose to 1 mo1,of butyrate result in the net
production of 3 ATPs/mol of glucose fermented (Fig. 8).
H-C-OH
I I During butyrate fermentation by Clostridium aceto-
H-C-OH H-L-OH H-C-OH
I butylicum and certain other clostridia, the production of
CHzO@ acids ceases when the pH of the medium drops below pH
5.0 and two neutral products, butanol and acetone, are pro-
CHzOH H-C=O duced with the consumption of butyrate and a concomitant
rise in pH. This observation was made very early during the
c=o H-C-OH
history of fermentation research (23,46). C. ucetobutylicum
I I
HO-C-H CW@ produces two butanol dehydrogenase isozymes that are
I
H-C-OH NADH dependent; however, the number of isozymes varies
I
H-C-OH depending on the strain. Similarly, Closmidium beijerinckii
I
H-C-OH contains three isozymes of NADH/N ADPH-dependent pri-
I mary ADHs. Again, the number of isozymes is dependent
CHzOQ
L J on the strain examined (16).
I4 Certain clostridia such as C. beijerinckii possess the abil-
ity to further reduce acetone to isopropanol. The regulation
of the ratio of acid and solvent products has been dealt with
in excellent reviews by Thauer et al. (109) and Jones and
Woods (46). The assay methods for the various enzymes of
the pathway are shown in Table 7 (section 22.1 1) .
~-6-0~ &-OH H-LoH ~-6-0~
I I I I
CHzO@ CHzO@ CHzO@ CH20@ 22.6. ETHANOL-ACETATE FERMENTATION

TjF One of the most interesting and remarkable of the fermen-


tations is that by Closcridium kluyveri. The organism requires
a mixture of ethanol and acetate for growth. However, the
acetate can be replaced by propionate. The primary prod-
2 AceQl-@ H-C=O ucts of this fermentation are butyrate and caproate in addi-
2 H-L-OH tion to hydrogen. The ratio of the two fatty acids is deter-
I mined by the concentration of ethanol in the medium.
CHzO@
Thus, an increase in ethanol concentration favors an in-
crease in the production of caproate and vice versa. The
4 ATP pathway for the formation of butyrate is presented in Fig. 9.
As seen from the figure, the oxidation of ethanol is in bal-
ance with the reduction of acetyl-CoA. Thus, the 2 mol of
NAD(P)H generated in the formation of acetyl-CoA is ox-
idized during the reduction of acetoacetyl-CoA to butyrate.
Evolution of hydrogen from the reduced NAD( P)H formed
FIGURE 7 Formation of acetate and lactate from glucose by in the first and second reactions of the pathway plays a cen-
the heterofennentative bifidum pathway. 1, PEP-PTS; 2, fructose- tral role in the generation of ATP. For each mole of H2
6-phosphate phosphoketolase; 3, transaldolase; 4, transketolase; evolved, half a mole of acetyl-CoA is not required for the
5, ribose-5-phosphate isomerase; 6, ribulose 5-phosphate 3- butyric acid cycle and the surplus can be used for the gen-
epimerase; 7, xylulose 5-phosphate phosphoketolase; 8, acetate eration of ATP. The stoichiometry between H2 evolution
kinase; 9, conversion of GAP to pyruvate, which is accomplished and ATP generation can be calculated (32, 103). For con-
by the same enzymes as in Fig. 2 and 3; 10, lactate dehydrogenase. venience, in Fig. 9, the concentrations of various interme-
diates produced during the production of 1 mol of ATP are
shown. The assay methods for the various enzymes of the
hydrogenase for the oxidation of NADH via ferredoxin to pathway are shown in Table 8.
yield H2 gas. When the levels of hydrogenase and ferredoxin
synthesis decrease due to iron limitation or when the hydro-
genase activity is inhibited by carbon monoxide, some 22.7. MIXED ACID A N D BUTANEDIOL
clostridia use pyruvate reduction to lactate as an alternate FER MENTATlON
sink for disposing of the electrons and protons (46). The enterobacteria carry out mixed acid and butanediol fer-
In butyrate production in clostridia, 2 mol of acetyl-CoA mentation, and this is the basis for the methyl redwoges-
(derived from pyruvate produced by the EMP pathway) is Proskauer test (see chapter 18) that is used to distinguish the
converted to 1 mol of acetoacetyl-CoA. The acetoacetyl- genera. Members of the genera Escherichiu, Salmonella, and
CoA is reduced using 1 mol of NADH derived from the Shigella ferment glucose to lactate, acetate, succinate, and
GAPDH reaction and then undergoes dehydration to yield formate (Fig. 10).They also form C02, H2, and ethanol. In
crotonyl-CoA. The crotonyl-CoA is reduced to butyryl- contrast, Enterobacter, Erwiniu, and Sewutia produce COZ,
22. Carbohydrate Fermentations 565

Glucose
2- E % ' NADH + H' NAD+
N A D s f2pi
2 NADH +2H'
2 CH3-CO-COOH d
lCH3-HCOH-COOH I

ADP A"
C H 3 - C O O a * w ]
CoA __--
__-*

CoA ...--._. __--__--


__--
..-. 16 __--- w CH3-CO-CH2-COOH
I- CH3-CO-CH2-CO-CoA
L
Acetoacetvl-CoA Acetoacetate
1
7bCO2

(CH3-CO-CH31
CH3-CH- CH2-CO-CoA / Acetone
I
OH
L (+)-n-hydroxybutyyl-CoA

H20 /
/
':
I:,
NADHtH'
NAD'

1-1
4
Isopropanol
CH~-CH=CH-CO-COA/
Crotonyl-CoA ;
NADH + H'
NAD'q i !
CH~-CH~-CH~-CO-COA CH3-CH2-CH2-COOH

Butanol

FIGURE 8 Butyrate-butanol-acetone-isopropanolfermentation in C. acetobutylicum. 1, PEP-PTS


and EMP pathway enzymes; 2, pyruvate-ferredoxin oxidoreductase; 3 , acetyl-CoA acetyltransferase
(thiolase); 4, L-( +)-P-hydroxybutyryl-CoA dehydrogenase; 5, crotonase; 6, butyryl-CoA dehydro-
genase; 7, butyraldehyde dehydrogenase; 8, butanol dehydrogenase; 9, NADH-ferredoxin oxido-
reductase and hydrogenase; 10, hydrogenase; 11, acetaldehyde dehydrogenase; 12, ethanol dehy-
drogenase; 13, lactate dehydrogenase; 14, phosphotransacetylase; 15, acetate kinase; 16,
acetoacetyl-CoA:acetate/butyrate:CoA transferase; 17, acetoacetate decarboxylase; 18, isopropanol
dehydrogenase; 19, phosphotransbutyrylase; 20, butyrate kinase.

ethanol, and large amounts of 2,3-butanediol and produce specific for the D and the other for the L isomer. These two
less acid. The enterobacteria employ PEP-PTS for the trans- unidirectional aerobic enzymes are not produced under fer-
port and phosphorylation of glucose. The glucose-6- mentative conditions and should properly be described as
phosphate is converted to pyruvate by the EMP pathway. oxidases rather than as dehydrogenases (18). In butanediol
The formation of the various fermentation products from fermentation, lactate formation is reduced and the forma-
PEP and pyruvate and the enzymes involved are shown in tion of other products of the pathway is determined by three
Fig. 10. A branch of the pathway leading from PEP forms enzyme activities: (i) pyruvate-formate lyase (PFL), (ii) a-
succinic acid. The rest of the products are formed from acetolactate synthase, and (iii) formate hydrogen lyase
pyruvate. Large amounts of lactate are formed in the mixed (FHL). Assay procedures for the enzymes involved in these
acid fermentation of Escherichia coli. There are three lactate fermentations are given in Table 9 (section 22.1 1).
dehydrogenases in this organism. One of these enzymes is a
soluble NAD-linked fermentative lactate dehydrogenase 22.7.1. Pyruvate-Forrnate Lyase
which produces D-lactate from pyruvate (12, 18). The other PFL plays a key role in the fermentation of glucose by a
two are membrane-bound flavoproteins, one of which is number of bacteria. The enzyme catalyzes the CoASH-
566 METABOLISM

iz.zzxl Glucose

H ~ ~ ~ ~ + ~ ~ +

6 Acetuldehyde
1 1 r

Malate

Furnarate

%OA

ltl 9g7
~utyvyI-~o~ L <+ )-J-hydroaybutylyl-CoA cetate
a /
Glucose
( 4 H ) F P Z

2 PEP
G2ADP
FIGURE 9 Ethanol-acetate fermentation by C. kluyueri. 1, 1

ADH; 2, acetaldehyde dehydrogenase; 3, Hz-evolving enzyme


system; 4, thiolase; 5, L-( +)-P-hydroxybutyryl-CoA dehydro-
genase; 6, crotonase; 7, butyryl-CoA dehydrogenase; 8, CoA (2W
transferase; 9, phosphotransacetylase; 10, acetate kinase. The
substrates, ethanol and acetate, are enclosed in shaded rectan-
gles, while the major product, butyrate, and the minor product, A aacetolactate
p
+Acetaldehyde

acetate, are enclosed in rectangles. Formation of acetate pri- 15kc02 rEZz4


marily yields the energy required for growth of the organism. acetoin

dependent cleavage of pyruvate, resulting in the formation of FIGURE 10 Mixed acid (top) and butanediol (bottom) fer-
formate and acetyl-CoA. The enzyme is induced only under mentation. 1, PEP-PTS; 2, pyruvate kinase; 3, lactate dehydro-
anaerobic conditions when the aerobic pyruvate dehydroge- genase; 4, PFL; 5, FHL; 6, acetaldehyde dehydrogenase; 7,
nase complex is completely repressed. The advantage of this ADH; 8, phosphotransacetylase; 9, acetate kinase; 10, PEP
switch is obvious in that the PFL forms acetyl-CoA anaero- carboxylase; 11, malate dehydrogenase; 12, fumarase; 13, fu-
bically without the requirement for the reduction of NAD. marate reductase; 14, a-acetolactate synthase; 15, a-acetola-
For the catalysis of the reaction, the enzyme has to be acti- cate decarboxylase; 16, acetoin reductase.
vated by PFLeactivating enzyme, which requires S-adenosyl-
methionine and dihydroflavodoxin as cosubstrates (see refer-
ence 54 for discussion of the activation process).
conditions when nitrate is present (also see chapter 21).
22.7.2. a-Acetolactate Synthase The second enzyme, formate dehydrogenase 0 (Fdh-0), is
As the name suggests, a-acetolactate synthase synthesizes synthesized at low levels independent of the availability of
acetolactate from pyruvate. The enzyme decarboxylates either oxygen or nitrate. The third enzyme, formate dehy-
pyruvate by a thiamine diphosphate (TPP)-dependent reac- drogenase H (Fdh-H), is the enzyme that in complex with
tion, with the formation of hydroxyethyl-TPP and COZ. hydrogenase is synthesized under anaerobiosis in the pres-
The enzyme-bound active acetaldehyde is subsequently ence of formate (6, 117).
transferred to a second molecule of pyruvate, resulting in
the formation of a-acetolactate. Consistent with its role in 22.7.4. Acetaldehyde Dehydrogenase
shifting the fermentation from the formation of acid end
and Alcohol Dehydrogenase
products to that of neutral products, the enzyme is synthe.
sized and active under acidic conditions (-pH 6). As shown in Fig. 10, acetaldehyde dehydrogenase converts
acetyl-CoA to acetaldehyde, which is further reduced to
22.7.3. Formate Hydrogen Lyase ethanol by ADH. In E. coli, these two reactions are carried
FHL is synthesized by E. coli and Enterobacter during fer- out by a single bifunctional protein encoded by the adhE
mentative growth. The enzyme cleaves formate to COz and gene (41, 59).
HZ. In contrast, Erwinia and Shigella species accumulate for-
mate due to the absence of this enzyme. 22.7.5. Fumarase
E. coli produces three formate dehydrogenases. Formate E. coli produces three distinct fumarases known as FumA,
dehydrogenase N (Fdh-N) is synthesized under anaerobic FumB, and FumC. The major enzyme produced during
22. Carbohydrate Fermentations 567

fermentative growth is FumB (124). The assay methods OH


for the various enzymes of the pathway are shown in I
(L)3CH3-C-COOH
Table 9 (section 22.11). I
H
22.8. PROPIONATE A N D SUCCINATE
FERMENTATION
Propionate is an end product of many fermentative bacte-
ria. These organisms ferment glucose to acetate, propionate,
11
H
I
@)3 CH3-C-COOH
and C02. The most common substrate for propionate-
producing bacteria is lactate, the end product of lactate fer- OH
OH
mentations (see above). Propionic acid-producing bacteria I
contain a lactate racemase because of which these organ- 2CH3-C-CO-CoA
I
isms are capable of fermenting D-, L-, or DL-lactate. There
are two different pathways for the formation of propionate.
Lactate is reduced to propionate in the acrylate pathway,
which is of limited distribution in bacteria. In contrast, in 2 CH~~CH-CO-COA
the propionate-succinate pathway (randomizing pathway),
the formation of propionate involves pyruvate and succi-
nate as intermediates and appears to be much more wide- CH3-CO-COOH
spread.
22.8.1. Acrylate Pathway
The acrylate pathway is present in C l o s d u m propionicum, CH3-C'O-CoA 2CH3-CH2 -CO-CoA
Bacteroides ruminicola, Megasphaera elsdenii, and a few other
organisms. These organisms contain lactate racemase for the
interconversion of L- and D-lactate. In the oxidative branch of
the pathway, D-lactate is converted to pyruvate by the enzyme
C O A T
211---
1

2CH3-CH2 -COOH
lactate dehydrogenase. There are two types of lactate dehy-
drogenases present in organisms that use the acrylate pathway.
For example, M. ekdmii has an NAD+-independent D-lactate
dehydrogenase (8), while C. propionicum has an NAD+-
dependent enzyme (93). In organisms containing the NAD+-
independent enzyme, an electron-transferring flavoprotein is
NET 3 Lactate - acetate + 2 propionate + C02
FIGURE 11 Fermentation of lactate to propionate by the
involved in the transfer of electrons. acrylate pathway. 1, lactate racemase; 2, propionyl-CoA trans-
The pyruvate formed by the lactate dehydrogenase is ferase; 3, lactyl+CoAdehydratase; 4, acrylyl-CoA reductase; 5,
converted to acetyl-CoA by pyruvate-ferredoxin oxidore- D-lactate dehydrogenase; 6, pyruvate-ferredoxin oxidoreduc-
ductase (see section 22.5). T h e enzymes phospho- tase; 7, phosphotransacetylase; 8, acetate kinase. ETFP, elec-
transacetylase and acetate kinase are responsible for the tron-transferring flavoprotein.
formation of acetate from acetyl-CoA. In this pathway, four
[HI released during the oxidation of one molecule of lactate
to acetate are used to reduce the two molecules of acrylyl-
CoA arising from dehydration of two molecules of lactate. The remaining two pyruvates are carboxylated to yield
This pathway yields 1 mol of ATP/3 mol of lactate metabo- oxaloacetate (OAA). It should be pointed out that in other
lized (Fig. 11). The assay methods for the various enzymes anaplerotic reactions employed by bacteria the carboxylation
of the pathway are shown in Table 10 (section 22.11). of pyruvate to OAA is an energy-consuming reaction requir-
ing ATP. However, Propionibacterium carboxylates pyruvate
22.8.2. Succinate-Propionate Pathway and forms OAA by methylmalonyl-CoA-pyruvatetranscar-
A majority of the propionate-producing organisms utilize boxylase, without consuming ATE The two OAAs are re-
the succinate-propionate pathway, and many of the classical duced to two molecules of malate, and four electrons are con-
early investigations that led to the characterization of this sumed for this reaction. The two malates are dehydrated to
pathway were done using Propionibacterium (Fig. 12). This two fumarates which are in turn reduced to two succinates,
pathway is commercially important in the manufacture of consuming four more electrons. Thus, the eight electrons
Swiss cheese. The characteristic flavor of Swiss cheese is generated are consumed and the pathway is balanced. The
due to the propionic acid, and the holes are due to COzpro- reduction of two fumarates to two succinates also yields two
duction. As seen from Fig. 12, succinate is an intermediate ATPs by substrate level phosphorylation. The succinates
in the pathway and in addition, depending on the organism, formed are esterified to succinyl-CoA by a CoA transferase.
it also accumulates in various amounts as an end product, Thus, the thioesterification of succinate also spares the con-
whereas succinate is not an end product in the acrylate sumption of energy. Isomerization of the two molecules of
pathway described above. succinylCoA results in the formation of two molecules
In this pathway, three molecules of lactate are oxidized of methylmalonyl-CoA. This is an unusual reaction where
to pyruvate by lactate dehydrogenase, resulting in the re- the CoASH molecule moves from the a-carbon to the p-
moval of six electrons. One of the pyruvates formed under- carbon of succinyl-CoA, resulting in the formation of (R)-
goes oxidative decarboxylation, resulting in the formation methylmalonyl-CoA in a vitamin B12-dependent reaction.
of acetyl-CoA and release of two additional electrons (not The (R)-methylmalonyl-CoA molecules are first racemized
shown). The acetyl-CoA is converted to acetate via to (S)+methylmalonylCoAbefore being transcarboxylated,
acetylphosphate, generating an ATP in the process. resulting in the formation of propionyl-CoA. The CoA from
568 METABOLISM

lates pyruvate, resulting in the formation of OAA. Instead,


3 CH3-CHOH-COOH B. fiagilis and S. ruminantium form O A A by carboxylating

3CH3-CO-COOH
p6H 2H
__fg_f,
cO2
CH3-CO-CoA
the glycolytic high-energy intermediate PEP by the enzyme
PEP-carboxykinase. The subsequent reactions of the path-
way from OAA to (S)-methylmalonyl-CoA are identical.
(S )-methylmalonyl-CoA is decarboxylated to propionyl-
CoA. The enzyme involved in this reaction, methyl-
malonyl-CoA decarboxylase, is membrane bound and acts
as a primary Naf pump. The Na+ ion gradient established
by the decarboxylase is used to drive ATP synthesis by an
2 HOOC-CH2-CO-COOH CH3-CO;OPO3H2 Na+- translocating ATP synthase (for a review on energy
2 NADH + H" conservation by decarboxylation of decarboxylic acids, see
2 NAD reference 25).
As the name of the pathway indicates, propionibacteria
ZHOOC-CH2-CHOH-COOH 1-1 can produce succinate in addition to propionate as the end

pl,o product. When the organism is growing on glucose or


other glycolytic substrates, it carboxylates the C3 interme-
diate PEP to OAA, which is then reduced to succinate.
2HOOC-CH=CH-COOH
T h e enzyme that carboxylates PEP is PEP carboxy-
transphosphorylase. It carries out the following unusual
2ADP +Pi anaplerotic reaction:
2 ATP + H20 PEP + C02 + Pi + OAA + PPi
2 HOOC-CH2-CH2-COOH In this reaction, the phosphate is transferred from PEP to
Biotin
I/------ Pi, resulting in the formation of PPi. In this connection, it is
Biotin-COz worth mentioning that propionibacteria use PPi to phospho-
rylate fructose-6-phosphate to fructose-1,6-diphosphate,
ZHOOC-CH2-CH2-CO-CoA originally discovered in Entamoeba histolytica (82). The assay
methods for the various enzymes of the pathway are shown

HI li
B12 -Em

2HOOC-C-CO-COA (R)
I
inTable 11 (section 22.11).

22.9. ACETATE FERMENTATION BY BACTERIA


CH3 A number of bacteria produce acetate as the sole product
during fermentation and are called homoacetogens. Many
2( 4 genera and species of bacteria are able to obtain energy and
grow solely on the reactions involving the formation of ac-
etate from C02and H2. However, in this chapter, only the
2HOOC-C-CO-CoA ($3) acetate fermentative pathway of hexoses is considered. The

Q-2
gram-positive, anaerobic, spore-forming bacterium Moorella
themoucetica (previously C. themaceticum) ferments hexose
to 3 mol of acetate (Fig. 13). The organism uses the EMP
pathway for the catabolism of glucose to two pyruvates. The
2CH3-CH2-CO-CoA two pyruvates are degraded by the action of pyruvate-
ferredoxin oxidoreductase, phosphotransacetylase, and ac-
etate kinase, resulting in the formation of acetate and C02.
Thus, from each molecule of glucose, two molecules each of
2 CH3-CH2-COOH acetate and C02are formed. The synthesis of the third mol-
ecule of acetate is initiated from the two molecules of COz.
FIGURE 12 Fermentation of lactate by the succinate- One of the C02molecules is first reduced to formate. In M.
propionate pathway. 1, lactate dehydrogenase (H acceptor is thermoacetica, the electron donor for the reaction is
flavoprotein); 2, (S)-methylmalonyl-CoA-pyruvatetranscar- NADPH. The formate combines with tetrahydrofolate
boxylase; 3 , rnalate dehydrogenase;4, fumarase; 5 , furnarate re- (THF) by an ATP-requiring reaction, resulting in the for-
ductase; 6, CoA transferase; 7, (R)-methylmalonyl-CoA mu- mation of formyl-THE It, in turn, undergoes a dehydration
tase; 8, methylmalonyl-CoA racemase; 9, pyruvate-ferredoxin reaction followed by two reductions resulting in the forma-
oxidoreductase; 10, phosphotransacetylase; 11, acetate kinase. tion of CH3-THF(Fig. 13). The methyl group is transferred
to a corrinoid iron-sulfur protein (CFeSP). The C02 from
the second molecule of pyruvate is not released but becomes
enzyme bound with carbon monoxide dehydrogenasel
the two molecules of propionyl-CoA are transferred to succi- acetyl-CoA synthetase (CODH/ACS). The C02 is now re-
nate, resulting in the formation of two propionate. duced to CO and finally becomes the carboxyl group of ac-
In this connection, it is worth mentioning that the etate. CODH/ACS accepts the methyl group from CFeSP.
anaerobes Bucteroides fraglis and Selenomonas ruminantium CODH also contains a binding site for CoASH and is re-
ferment glucose to propionate (63, 67). However, these or- sponsible for the synthesis of acetyl-CoA (60, 70, 79, 121).
ganisms lack methylmalonyl-CoA-pyruvatetranscarboxy- The typical enzymes acting on acetyl-CoA form the third
lase, a key enzyme of the pathway which normally carboxy- acetate and ATP. This pathway is unique in that all six car-
22. Carbohydrate Fermentations 569

Glucose
~ A D P+ 2Pi +NAD
2 ATP 2NADH + H'

I
Pyruvate Pyruvate

, , L g i d H2
Acetyl-CoA
CoA+"

5rADpH+H+
COZ
NADPH + H+
Acetyl- @ Acetyl- @
NADP+

? 'PT
AJ%
&
,
?!L
k? Formate
~
[COI
IAcetate
ATP
]
ADP + Pi methyl- CFeSP CoA
1 OZ~OKVJYI-H~F

H20 4 Acetvl-CoA

5 , 10-methenyl-H4F ,o II Acetyl- @
NADH + H+

NAD 4
5 , 10-methylene-H4F
Acetate

Fd
5 -methyl-H4F
FIGURE 13 Acetate fermentation of glucose by bacteria. 1, enzymes of the EMP pathway; 2,
pyruvate-ferredoxin oxidoreductase; 3, phosphotransacetylase; 4, acetate kinase; 5, formate dehy-
drogenase; 6, formyl-THF synthetase; 7, methenyl-THF cyclohydrolase; 8, 5,lO-methylene-THF
dehydrogenase; 9, 5,lO-methylene-THF reductase; 10, THF:B12 methyltransferase; 11, CO dehy-
drogenase/acetyl-CoA synthase.

bons of hexose are converted to three molecules of two car- 90). However, quantitative determination of substrate uti-
bon compounds. The assay methods for the various enzymes lization and product formation and construction of fermen-
of the pathway are shown in Table 12 (section 22.11). tation balances have only been described for a very few or-
ganisms.
Sugars are catabolized via the modified versions of the
22.10. ACETATE FERMENTATION IN EMP pathway (Fig. 14). The modifications usually occur
H Y PERTHERMOPHILIC ARCHAEA in (i) the phosphorylation of glucose and fructose-6-
Hyperthermophiles are those organisms that have the re- phosphate, (ii) the oxidation of GAP, and (iii) the conver-
markable ability to grow at, near, or above the boiling point sion of acetyl-CoA to acetate.
of water. By definition, according to Stetter, any organism
with an optimum growth temperature of between 80 and 22.10.1. Kinases
106C is a hyperthermophile (104). I t is worth noting that
the upper temperature limit of life (121C) is held by an Phosphorylation of Glucose
22.10.1 .l.
unidentified chemolithoautotrophic archaeon related to The phosphorylation of glucose is carried out by either glu-
Pyrodictium occultum and Pyrobaculum aerophilum (48). cokinase or hexokinase. It appears that among the archaea
Further, it has been demonstrated that vegetative cells of so far examined, the organisms that contain glucokinase ap-
Pyrolobus and Pyrodictium are able to survive autoclaving at pear to use ADP as the phosphoryl donor for the phospho-
121C for 1 h (105)! rylation of glucose. In contrast, the few archaea that have
Many of the organotrophic hyperthermophiles ferment been reported to contain the enzyme hexokinase appear to
sugars or peptides. Carbohydrates fermented include the use ATP as the phosphoryl donor (Table 13).
polysaccharides, such as starch and glycogen; the disaccha-
rides maltose, cellulose, and lactose; and the monosaccha- 22.1 0.1.2. Phosphorylation of Fructose-6-Phosphate
rides, such as glucose, galactose, pyruvate, and lactate. The The phosphorylation of fructose-6-phosphate is carried out
fermentation products formed include acetate, alanine, pro- by the enzyme PFK. The archaea contain three different
pionate, isovalerate, isobutyrate, and butanol (for a list of PFKs which use different phosphoryl donors to phosphorylate
substrates fermented and products formed, see reference fructose-6-phosphate. They are referred to as ATP-PFK,
570 rn METABOLISM

Glucose 22.10.1.3. Bifunctional Kinase

I1
Glucose 6-phosphate
Recently, an unusual kinase has been described for Meth-
umcoccus junnaschii. The enzyme is capable of phophorylat-
ing both glucose and fructose-6-phosphate.The phosphoryl
donor for the reaction is ADP. Hence, the enzyme is re-
12
Fructose 6-phosphate
ferred to as ADP-dependent glucokinase/PFK.
22.1 0.2. Oxidation of Glyceraldehyde-3-Phosphate

k
Fructose 1,bbisphosphate
In many of the anaerobic species of archaea examined so
far (Pyrococcus, Thermococcus, Themoproteus, and Desul-
furococcus), GAP is directly oxidized to 3-phosphoglyceric
acid. Depending on the organism, this oxidation is carried

Dihydroxyacetone phosphate
A &-+Glyceraldehyde 3 -phosphate
out by either glycera1dehyde:ferredoxin oxidoreductase or
the NAD+-dependent GAPDH. Both the enzymes carry
out irreversible conversion of GAP to 3-phosphoglyceric
2NAD++
2 NADH a H'
pL2 acid. In the conventional EMP pathway, the conversion
of GAP to 3-phosphoglycerate is carried out by two en-
2x 3-Phosphoglycerate zymes: a Pi-dependent, NADP-requiring GAPDH and a
phosphoglycerate kinase. In this reaction sequence, 1,3-
bisphosphoglycerate is an intermediate and ADP is phos-
2x 2-Phosphoglycerate phorylated to ATP during its conversion to 3-phospho-

l9
2x Phosphoenolpyruvate
glycerate. The anaerobic archaea, by eliminating the
formation of 1,3-bisphosphoglycerate as an intermediate,
also dispense with the formation of 2 mol of ATP/mol of

p2:;
2xpyrUvate
glucose oxidized.

22.10.3. Conversion of Acetyl Coenzyme A to


%
;*2I:;:; Acetate
During the fermentation of sugars or lactate, the acetate-

2 CoA +*=
2x Acewl-CoA
pi
forming archaea oxidize pyruvate by the enzyme pyruvate-
ferredoxin oxidoreductase (see discussion of clostridia
above) to acetyl-CoA and COz.The hyperthermophilic ac-
2x Aietate etate-forming archaea convert acetyl-CoA to acetate by the
action of a unique enzyme, acetyl-CoA synthetase (ADP
FIGURE 14 Acetate fermentation in archaea. 1, kinase
forming). This novel enzyme couples substrate level phos-
(glucokinase/hexokinase); 2, glucose-6-phosphate isomerase;
phorylation of ADP to the conversion of acetyl-CoA to ac-
3 , PFK; 4,fructose bisphosphate aldolase; 5 , triose phosphate
isomerase; 6, GAPDH; 7, GAP ferredoxin oxidoreductase; 8, etate. In this connection, it is worth mentioning that the
phosphoglycerate mutase; 9, enolase; 10, pyruvate kinase; 11, conversion of acetyl-CoA to acetate in bacteria requires
two enzymes, phosphate acetyltransferase and acetate ki-
pyruvate-ferredoxin oxidoreductase; 12, acetyl-CoA synthetase
(ADP forming). nase.
The conversion of acetyl-CoA to acetate and ATP by
acetyl-CoA synthetase is the most important energy-
which uses ATP as the phosphoryl donor; ADP-PFK, which conserving reaction during sugar fermentation in archaea.
uses ADP as the phosphoryl donor; and PP,-PEK, which uses The assay methods for the various enzymes of the pathway
pyrophosphate as the phosphoryl donor (Table 13). are shown in Table 14 (section 22.11).

TABLE 13 Phosphoryl donors for kinases of archaea"


Organism(s) ADP-GK ATP-HK ATP-PFK ADP-PFK PP,-PFK Reference(s)
Aeropyrum pernix + + 27,35
Archoglobus fulgidus + + 57, 125
Desulfurococcus umylolyticw + + 34,97
Methunococcus junnaschiib + + 87
Many methanogens + 116
Pyrococcus furiosus + + 27,51, 115
Thermococcus cekr + + 97
Thermococcus litoralis + + 54,97
Thermococcus zilligii + + 84, 125
Thermoproteus tenax + + 97,100
"ADP-GK, ADP-glucokinase;ATP-HK, ATP-hexokinase.
M. jannaschii, ADP-GK and ADP-PFK activities are carried out by a single bifunctional enzyme.
2 2 . Carbohydrate Fermentations 571

22.1 1 ASSAY TABLES

TABLE 2 Assay procedures for enzymes of the EMP pathway a

Enzyme Assay mixture AA (nm) E (M-lcm-)

1. Hexokinase (65) 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12, 5 mM 340 6.23 x 10


(EC 2.7.1.1) glucose, 1 mM ATP, 0.05 mM NADP+, 0.3 U of glucose-6-phosphate
dehydrogenase, and cell extract
2. Phosphoglucoisomerase 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12, 1 mM 340 6.23 x lo3
(65) (EC 5.3.1.9) fructose-6-phosphate-free glucose-6-phosphate, 0.05 mM NADP+, 0.3
U of glucose-6-phosphate dehydrogenase, and cell extract
3. PFK (65) (EC 2.7.1.11) 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12,5 mM 340 6.23 x 103
glucose-6-phosphate, 2 U of phosphoglucoisomerase, 0.03 mM
NADH, 1 mM ATP, 0.3 mM ADP, 1 U each of aldolase and a-
glyceraldehyde phosphate dehydrogenase, 10 U of triose-phosphate
isomerase, and cell extract. The reaction velocity should be divided
by 2.
4. Fructose bisphosphate 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12,0.03 340 6.23 x 10)
aldolase (65) mM NADH, 1 mM fructose 1,6-bisphosphate, 1 U a-glyceraldehyde-
(EC 4.1.2.13) phosphate dehydrogenase, 10 U triose-phosphate isomerase, and cell
extract. The reaction velocity should be divided by 2.
5. Triose phosphate iso- 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgCl2, 0.03 340 6.23 X lo3
merase (65) mM NADH, 0.4 mM GAP, 1 U of a-glyceraldehyde phosphate dehy-
(EC 5.3.1.1) drogenase, and cell extract
6. GAPDH (65) 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12, 0.03 340 6.23 x lo3
(EC 1.2.1.12) mM NADH, 5 mM cysteine, 1 mM 3-phosphoglycerate, 1 mM ATP,
1 U of phosphoglycerate kinase, and cell extract
7. 3-Phosphoglycerate 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgCI2,0.03 340 6.23 x lo3
kinase (65) (EC mM NADH, 5 mM cysteine, 1 mM 3-phosphoglycerate, 1 mM ATP,
2.7.2.3) 1 U of GAPDH, and cell extract
50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12, 0.03 340 6.23 x lo3
8. Phosphoglycerate mM NADH, 1 mM 3-phosphoglycerate, 0.5 U enolase, 1 mM ADP,
mutase (65) 1 U each of pyruvate kinase and muscle lactate dehydrogenase, and
(EC 5.4.2.1) cell extract
9. Enolase (65) 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12,0.03 340 6.23 x lo3
(EC4.2.1.11) mM NADH, 1 mM 3-phosphoglycerate, 0.5 U of phosphoglycerate
mutase, 1 mM ADP, 1 U each of pyruvate kinase and muscle lactate
dehydrogenase, and cell extract
10. Pyruvate kinase (65) 50 mM triethanolamine hydrochloride (pH 7.4); 10 mM MgCl2; a 1 mM 340 6.23 x 103
(EC 2.7.1.40) concn each of PEP, ADP, and fructose-1,6-diphosphate;0.03 mM
NADH; 1 U of muscle lactate dehydrogenase; and cell extract
11. Pyruvate decarboxylase 50 mM triethanolamine hydrochloride (pH 7.4), 10 mM MgC12, 50 mM 340 6.23 x lo3
(65) (EC 4.1.1.1) pyruvate, 5 mM cysteine, 0.03 mM NADH, 0.25 mM TPP, 1 U of
ADH, and cell extract
12. ADH (26) (EC 1.1.1.1) 21 mM glycine, 75 mM semicarbazide HCI, 75 mM sodium pyrophos- 340 6.23 x 10
phate buffer (pH 8.7), 150 mM ethyl alcohol, 1 mM NAD+, and cell
extract
Most reagents and enzymes described are readily available from various commercial sources, such as Sigma-Aldrich.
572 w METABOLISM

TABLE 3 Assay procedures for the enzymes of the ED pathway


Enzyme Assay mixture AA (nm) E (M-lcm-')
1. Glucokinase (96) 20 mM imidazole, 20 mM KHZPO, buffer (pH 6.8), 5 mM MgC12, 1 mM 340 6.23 x lo3
(EC 2.7.1.2) ATP, 10 mM glucose, 1 mM NAD+, 2 U of glucose-6-phosphatede-
hydrogenase, and cell extract
2. Glucose-6-phosphate 30 mM Tris-30 mM KC1-2 mM MgSO4 buffer (pH 6.8), 1 mM glucose- 340 6.23 x lo3
dehydrogenase (96) 6-phosphate, 1 mM NAD+, 0.2 mg of BSA," and cell extract
(EC 1.1.1.49)
3. 6-Phosphogluconate 20 mM K-MES buffer (pH 6.5) containing 2 mM MgC12 and 50 mM 340 6.23 x lo3
dehydratase (95) NaC1, 1 mM 6-phosphogluconate,0.15 mM NADH, 5 U each of lac-
(EC 4.2.1.12) tate dehydrogenase and KDPG aldolase, and cell extract
4. KDPG aldolase (94) 20 mM K-MES buffer (pH 6.5) containing 50 mM NaCl and 2 mM 340 6.23 x lo3
(EC 4.1.2.14) MgC12, 0.15 mM NADH, 5 U of rabbit muscle lactate dehydrogenase,
1 mM KDPG, and cell extract
5.GAPDH(75) 50 mM K-MES buffer (pH 6.5), 30 mM KC1,3 mM MgClz, 5 mM 3- 340 6.23 x lo3
(EC 1.2.1.12) phosphoglycerate, 1 mM ATP, 0.15 mM NADH, 10 mM P-mercap-
toethanol, 10 U of phosphoglycerate kinase, and cell extract
6. 3-Phosphoglycerate ki- Same as that described for GAPDH except for 10 U of GAPDH as the
nase (75) (EC 2.7.2.3) coupling enzyme
7. Phosphoglycerate 50 mM K-MES buffer (pH 6.5) containing 30 mM KCl and 3 mM 340 6.23 x lo3
mutase (75) MgCl,; 2 mM 3-phosphoglycerate;0.5 mM ADP; 0.15 mM NADH;
(EC 5.4.2.1) 10 mM P-mercaptoethanol; 10 U each of enolase, pyruvate kinase,
and lactate dehydrogenase; and cell extract
8. Enolase (75) (EC 30 mM triethanolamine buffer (pH 7.5) containing 0.5 mM ADP, 340 6.23 x lo3
4.2.1.1 1) 30 mM KC1, and 3 mM MgC12; 10 U each of pyruvate kinase and
lactate dehydrogenase; 0.15 mM NADH; 0.4 mM 2-phosphoglycerate;
and cell extract
9. Pyruvate kinase (75) 50 mM MES buffer (pH 6.5) containing 100 mM KC1 and 0.5 mM 340 6.23 x lo3
(EC 2.7.1.40) ADP, 0.15 mM NADH, 0.5 mM PEP, 10 U of lactate dehydrogenase,
and cell extract
10. Pyruvate decarboxylase 50 mM K-MES buffer (pH 6.5), 5 mM MgC12,O.l mM TPP, 0.1 mg of 340 6.23 x lo3
(71) (EC 4.1.1.1) BSA, 0.15 mM NADH, 10 U of yeast alcohol dehydrogenase, 5 mM
pyruvate, and cell extract
11. ADH (5, 72) 30 mM Tris-HC1buffer (pH 8.5), 1 M ethanol, 1 mM NADf, and cell 340 6.23 x lo3
(EC 1.1.1.1) extract
"BSA, bovine serum albumin; M E ,morpholineethanesulfonicacid.

TABLE 4 Assay procedures for enzymes of the homolactate fermentation pathway


Enzyme Assay mixture AA (nm) E (M-'cm-I)
1. PEP-PTS (15,56) 2.5, 5, and 10 pl of permeabilized cells" and 100 ~1 of 50 mM KP04 buffer
(EC 2.7.3.9) (pH 6.5) containing 12.5 mM NaF, 5 mM MgCl2, 2.5 mM DTT, 10 mM
PEP, and 10 mM I4C-labeledcarbohydrate (specific activity, 200 dpm .
nmol-I) are incubated for 15-30 min at 37C; the phosphorylated
carbohydrates are separatedbby columns and the radioactivity is
determined by liquid scintillation counting (see chapter 17)
2-6. Glucose-6-phosphate Assays as described in Table 2
to pyruvate
7. Lactate dehydrogenase 50 mM triethanolamine (pH 7.5), 1 mM fructose-l,6-bisphosphate,0.2 mM 340 6.23 x lo3
(4) (EC 1.1.1.27) NADH, and 10 mM sodium pyruvate are used to initiate the reaction with
permeabilized cells at 28C
"Preparation of permeabilized cells: Bacterial culture is washed twice with ice-cold 50 mM KPO, buffer (pH 6.5) containing 2 mM MgS04 (KPM buffer), resus-
pended in 1/100 of culture volume of KPM buffer containing 20% (wt/vol) glycerol, and rapidly frozen in liquid nitrogen and kept at -80C until use. After thawing,
cells are washed once with KPM buffer; resuspended in 50 mM KPO, buffer (pH 6.5) containing 12.5 mM NaE 5 mM MgC12, and 2.5 mM dithiothreitol (DTT); and
permeabilized. First, 2.5 pl of toluene/acetone (1:9, vol/vol) is added per 250 p1of cell suspension and vortexed for 5 min at 4C. The cells are then centrifuged (150
X g a t 4C for 2 min) and the cell pellet is resuspended in the same buffer (optical density at 600 nm of 50) and again treated with toluene/acetone as described above
(50). After 5 min of vortexing, permeabilized cells are kept on ice.
*Separationmethod: The product, ['4C]hexose-phosphate,is separated from excess 14C-substrateby ion-exchange chromatography. The incubation mixtures are di-
luted with 0.5 ml of water and transferred to columns, 0.8 by 9 cm, of analytical-grade,chloride form, ion-exchange resin (AG I-X2,50 to 100 mesh; Bio-Rad);the ex-
cess 14C-substrateis washed from each column with 15 ml of water; and the labeled product is eluted from the column with 6 ml of 1.0 M LCI. Each elute is collected
in a liquid scintillation spectrometer vial, and the I4C is counted after the addition of 15 rnl of a mixture containing 333 mi of Triton X-100 (Packard Instrument
Company), 666 ml of toluene, 5.5 g of 2,5-diphenyloxazole, and 125 mg of dimethyl-l,4-bis[2-(5-phenyloxazolyl)] benzene (dimethyl POPOP). The efficiency of the
counting system is determined with 14C-standards(see chapter 17).
2 2 . Carbohydrate Fermentations rn 573

TABLE 5 Assay procedures for enzymes of the heterolactate fermentation pathway


Enzyme Assay mixturea AA (nm) E (M-lcm-')
1. PEP-PTS (EC 2.7.3.9) Assay as described in Table 4
2. Glucose-6-phosphate Assay as described in Table 3
dehydrogenase (EC 1.1.1.49)
3. 6-Phosphogluconate Assay as described in Table 3
dehydrogenase (EC 1.1.1.44)
4. Ribulose-5-phosphate-3- 100 mM triethanolamine buffer (pH 7.4), 10 mM D-ribose- 290 72
epimerase (122) (EC 5.1.3.1) 5-phosphate, 2.2 U of heat-treated ribose-5-phosphate
isomerase, and cell extract
5 . Phosphoketolase (66) 33.3 mM KP04 (pH 6.5), 1.9 mM L-cysteine hydrochloride, 505
(EC 4.1.2.9) 23 mM sodium fluoride, 8 mM sodium iodoacetate,
27 mM ~-fructose-6-phosphateor D-xylulose-
5-phosphate, and cell extract. Incubate at 37C for
30 min, and then add 0.075 ml of 2 M hydroxylamine
hydrocholoride (pH 6.5) at room temp. After 10 min,
add 0.05 ml of 15% (wt/vol) trichloroacetic acid,
4 M HC1, and 5% (wtlvol) FeC13 . 6H20 in 0.1 M HCI.
Acetylphosphate is used as the standard.
6. Conversion of GAP to pyruvate Assays as described in Tables 2 and 3
7. Lactate dehydrogenase Assay as described in Table 4
8. Phosphotransacetylase(9) 100 mM Tris-HC1 buffer (pH 8), 5 mM MgC12,0.5 mM NAD', 340 6.23 x 103
(EC 2.3.1.8) 0.5 mM CoA, 5 mM L-malate, 12.5 pg of crystalline malate
dehydrogenase, 25 pg of crystalline citrate synthase, 10 mM
lithium acetyl phosphate, and cell extract
9. Acetaldehyde dehydrogenase (19) 50 mM CHES buffer (pH 9.5), 100 pM CoA, 1.0 mM DTT, 340 6.23 x 103
(EC 1.2.1.4) 75 pM NAD+, 10 mM acetaldehyde, and cell extract
10. ADH (19) (EC 1.1.1.1) 12 mM sodium pyrophosphate (pH 8.5), 75 pM NAD+, 20 pl 340 6.23 x lo3
of ethanol, and cell extract
"CHES, 2(N-cyclohexylamino) ethanesulfonate; DTF, dithiothreitol.

TABLE 6 Assay procedures for enzymes of the heterofermentative bifidum pathway


Enzyme Assay mixture AA (nm) E (M-lcm-')
1. PEP-PTS (EC 2.7.3.9) Assay as described in Table 4
2. Fructose-6-phosphate Assay as described in Table 5
phosphoketolase (EC 4.1.2.9)
3. Transaldolase (114) (EC 2.2.1.2) 40 mM triethanolamine buffer (pH 7.6), 100 mM 340 6.23 x lo3
EDTA, 3.2 mM fructose-6-phosphate,0.2 mM
erythrose-4-phosphate,0.1 mM NADH, 10 pg
a-glycerophosphate dehydrogenase-triose phosphate
isomerase mixture, and cell extract
4. Transketolase (101) (EC 2.2.1.1) 50 mM glycylglycine buffer (pH 8.5), 0.2 mM NADH, 340 6.23 x lo3
2 mM xylulose 5-phosphate, 5 mM MgC12, 1 mM TPP,
and cell extract
5. Ribose-5-phosphateisomerase 100 mM triethanolamine buffer (pH 7.4), 10 mM D-ribose- 290 72
(122) (EC 5.3.1.6) 5-phosphate, 2.2 U of ribose-5-phosphate epimerase, and
cell extract
6. Ribulose-5-phosphate- Same as above except 2.2 U of heat-treated ribose-5-phosphate
3-epimerase (EC 5.1.3.1) isomerase was added
7. Xylulose-5-phosphate GAP formed is measured by coupling it to the GAP assay as
phosphoketolase (EC 4.1.2.9) described in Table 2
8. Acetate kinase (102) 100 mM Tris-HC1buffer (pH 6.5), 3 mM MgC12, 2 mM 340 6.23 x 103
(EC 2.7.2.1) glucose, 0.5 mM NADP+, 1 U of hexokinase, 1 U of
glucose-6-phosphatedehydrogenase, 4 mM acetylphosphate,
1 mM ADP, and cell extract
9. Conversion of GAP to pyruvate Assays as described in Tables 2 and 3
10. Lactate dehydrogenase Assay as described in Table 4
574 rn METABOLISM

TABLE 7 Assay procedures for the enzymes of the butyrate-butanol-acetone-isopropanolfermentation in C. acetobutylicum


Enzvme Assav mixturea AA (nm) E (M-'cm-')
1. PEP-PTS and EMP
pathway enzymes
PEP-PTS (69) 10 mM Tris-HC1 (pH 7.5), 0.5 mM PEP, 2 mM DTT, 5 mM MgC12,
12 mM KF, and 0.1 mM radiolabeled sugar (1.05 Ci mol-'), with
25 mM potassium phosphate (pH 7.0) added to ensure adequate pre-
cipitation of sugar phosphate. At intervals, 0.15-ml samples were
withdrawn and added to 2 ml of 1% barium bromide in 80% ethanol.
Precipitates were removed by filtration on fiberglass disks (Whatman
GF/F), washed with 5 ml of 80% ethanol, and dried under a heat
lamp. The disks were added to 4 ml of scintillation cocktail and the
radioactivity was determined by scintillation counting.
EMP pathway enzymes Assays as described in Table 2
2. Pyruvate ferredoxin 50 mM K2HP04buffer (pH 7.0), 0.1 mM CoA, 20 mM DTT, 2 mM 578 8.65 x lo3
oxidoreductase (58, benzyl viologen, 5 mM pyruvate, and cell extract
102) (EC 1.2.7.1)
3. Acetyl-CoA acetyltrans- 100 mM Tris-HC1 (pH 8.0), 100 mM KCL, 0.6 mM CoA, 10 mM 233 4.44 x lo3
ferase (36) (EC 2.3.1.9) lithium acyl phosphate, and cell extract
4. L-( +)-P-Hydroxybutyryl- 50 mM 2-(N-morpholine)propanesulfonicacid-KOH buffer (pH 7.0), 1 340 6.23 x lo3
CoA-dehydrogenase mM DTT, 0.2 mM NADH, 75 pM acetoacetyl-CoA, and cell extract
(36) (EC 1.1.1.157)
5.Crotonase (36) (EC 100 mM Tris-HC1buffer (pH 7.6), 0.15 mM crotonyl-CoA, and cell 263 6.7 x lo3
4.2.1.17) extract
6. Butyryl-CoA dehy- 100 mM KP04 buffer (pH 7.0), 0.15% Triton X-100,0.2 mM meldola- 492 19.4 x lo3
drogenase (36) (EC blau (ZnCl salt of 8-dimethyl-amino-2,3-benzophenoxazine), 0.3 mM
1.3.99.2) iodonitrotetrazolium, and cell extract. Butyryl-CoA (0.1 mM) is
added to initiate the reaction after flushing with H2 gas for 2 min.
7. Butyraldehydedehydro- 80 mM Tris-maleate buffer (pH 6.0), 0.2 mM butyryl-CoA, 0.4 mM 340 6.23 x lo3
genase (3) (EC 1.2.1.57) NADH, 72 mM semicarbazidehydrochloride, and cell extract
8.Butanol dehydrogenase 70 mM Tris-HC1 buffer (pH 7.8), 20 mM butanol, 0.4 mM NAD, 340 6.23 x lo3
(3) 72 mM semicarbazide-HC1, and cell extract
9. NADH-ferredoxin 100 mM Tris-HC1 (pH 7.6), NADH-regenerating system (250pM 320 9.31 x lo3
oxidoreductase (7) NADH, 30 p1 of 96% ethanol, 45 U of ADH from yeast), and acetyl-
(EC 1.18.1.3) CoA-generating system [20 mM acetylphosphate, 2 U of phospho-
transacetylase, 1 mM CoA, 30 mM (N&)ZSO4, and 2 mM GSH to
activate phosphotransacetylase], 10 pM FAD, 0.1mM metronidazole,
0.5-8 pM ferredoxin, and cell extract. The reaction is carried out
under an atmosphere of CO in anaerobic cuvettes fitted with serum
stoppers.
10. Hydrogenase (102) 100 mM Tris-HC1 buffer (pH 6.5), 2 mM benzyl viologen, 2 mM DTT, 1 578 8.65 x lo3
(EC 1.12.7.2) atm of H2 gas, and cell extract
11. Acetaldehyde 100 mM Tris-HC1 (pH 6.5), 1 mM NAD(P), 1 mM DTT, 0.1 mM CoA, 340 6.23 X I d
dehydrogenase 7 mM sodium arsenate, 0.01 mM acetaldehyde, 0.5 U of phospho-
(58, 102) (EC 1.2.1.4) transacetylase, and cell extract
12. Ethanol dehydrogenase 100 mM Tris-HC1(pH 6.5), 10 mM DTT, 0.3mM NAD(P)H, 10 mM 340 6.23 x lo3
(102) (EC 1.1.1.1) acetaldehyde, and cell extract
13. Lactate dehydrogenase 20 mM MOPS buffer (pH 7.0), 30 mM sodium pyruvate (pH 7.5), 0.2 340 6.23 x lo3
(12) (EC 1.1.1.27) mM NADH, and cell extract
14. Phosphotransacetylase 100 mM Tris-HC1 buffer (pH 6.5), 1 mM CoA, 30 mM NH4Cl, 10 mM 233 4.44 x lo3
(67, 102) (EC 2.3.1.8) DTT, 2 mM acetylphosphate, and cell extract
15. Acetate kinase (102) 100 mM Tris-HC1buffer (pH 6.5), 3 mM MgC12, 2 mM glucose, 0.5 mM 340 6.23 x lo3
(EC 2.7.2.1) NADP+, 1 U of hexokinase, 1 U of glucose-6-phosphatedehydroge-
nase, 4 mM acetylphosphate, 1 mM ADP, and cell extract
16. Acetoacetyl- 100 mM Tris-HC1 (pH 7.5), 150 mM carboxylic acid (potassium salt of 310 8.0 x lo3
CoA:acetate/butyrate: acetate, butyrate, or propionate) (pH 7.5), 40 mM MgC12,0.1mM
CoA transferase (13) acetoacetyl-CoA, 5% (vol/vol) glycerol, and cell extract
(EC 2.8.3.9)
(Continued on next page)
22. Carbohydrate Fermentations 575

TABLE 7 (Continued)
Enzyme Assay mixturea AA (nm) E (M-lcm-')

17. Acetoacetate 40 mM sodium acetate buffer (pH 5.0), 80 mM lithium acetoacetate, 270 55.0
decarboxylase (3) and cell extract in a 3-mi total reaction volume. COZ is measured in a
(EC 4.1.1.4) Warburg manometer.
18. lsopropanol dehydroge- 50 mM Tris-HCI buffer (pH 7.5), 1 mM DTT, 0.2 mM NADPH, 6.7 340 6.23 X 10'
nase (44) (EC 1.1.1.80) mM acetone, and cell extract
19. Phosphotransbutyrylase 100 mM KPO, buffer (pH 7.0), 0.1 mM butyryl-CoA, and cell extract 233 4.44 x 10)
(36) (EC 2.3.1.19)
20. Butyrate kinase (36) 75 mM Tris-HCI (pH 8.0), 6 mM ADP, 10 mM MgClz, 2 mM glucose, 1 340 6.23 x lo3
(EC 2.7.2.7) U each of hexokinase and glucose-6-phosphate dehydrogenase, 0.2
mM NADP+, 7 mM butyryl phosphate, and cell extract
"DTT,dithiothreitol; KF, potassium fluoride; GSH, glutathione (reduced); FAD, flavin adenine dinucleotide; MOPS, morpholinepropanesulfonicacid

TABLE 8 Assay procedures for the enzymes involved in ethanol and acetate fermentation by C. kluyweri
Enzyme Assay mixture' AA (nm) E (M-lcm-')
1. ADH (EC 1.1.1.1) Assay as described in Table 7
2. Acetaldehyde dehydrogenase Assays as described in Table 7
(EC 1.2.1.4)
3. Hz-evolving enzymes (EC 1.12.2.1)
NADH ( 110) 75 mM Tris-HCI (pH 7.4), 2 mM glutathione red, 12pM FAD,
1 U of phosphotransacetylase, 25 mM acetylphosphate
(potassium-lithium salt), 0.75 mM CoA, 0.9 mg of C .
kluyweri ferredoxin (Fdkl), NADH-RS, and 5 mg of DEAE-
cellulose-treated extract. NADH-RS is 40 mM galactose,
2.5 mM NADH, and 0.5 U of galactose dehydrogenase.
Assays are carried out in a total volume of 1 ml in 17.5-ml
Thunberg tubes at 37C with shaking; gas phase, argon. HZ
is detected by gas chromatography!
NADPH (47) 70 mM Tris-HCI buffer (pH 8.0), 2 mM glutathione red, 12
p M FAD, 1 mg of ferredoxin (Fdkl), NADPHARS,NADf-
RS, and 5 mg of DEAE-cellulose-treated extract. NADPH-RS
is 16 mM glucose-6-phosphate, 3.5 U of glucose-6-phosphate
dehydrogenase, and 0.5 mM NADP+. NAD-RS is 2 mM
fructose-1,6-diphosphate, 0.5 U of aldolase, 2.5 U of triose
phosphate isomerase, 0.7 U of glycerol-I-phosphate dehydro-
genase, and 0.5 mM NAD. Assays are carried out in a total
volume of 2.5 ml in 17.5-1111 Thunberg tubes at 37C usin
an argon gas phase. Hz is detected by gas chromatography 8
4. Thiolase (120) (EC 2.3.1.9) 67 mM Tris-HC1 buffer (pH 8.1), 0.2 mM acetoacetyl-CoA, 25 233 4.4 x 10'
mM potassium arsenate (pH 8.1), 2 U of phosphotransacety-
lase, 0.2 mM CoA, and cell extract
5. L-( +)-P-Hydroxybutyryl-CoA Assay as described in Table 7
dehydrogenase (EC 1.1.1.157)
6. Crotonase (EC 4.2.1.17) Assay as described in Table 7
7. Butyryl-CoA dehydrogenase Assay as described in Table 7
(EC 1.3.99.2)
8. CoA transferase (119) (EC 2.8.3.8) 100 mM Tris-HCI buffer (pH 7.5), 100 mM potassium butyrate 310 8.0 x 10'
(pH 7.5), 20 mM MgC12,O.lO mM acetoacetyl-CoA, 5%
(vol/vol) glycerol, 0.05 U of CoA transferase, and cell
extract
9. Phosphotransacetylase (EC 2.3.1.8) Assay as described in Table 7
10. Acetate kinase (EC 2.7.2.1) Assay as desctibed in Table 7
"FAD,flavin adenine dinucleotide; RS, regenerating system.
"H, is quantified by gas chromatography. Sample column: Length, 4 m; inner diameter, 2 mm; material, steel; temperatures, 50C for both injection port and col-
limn; carrier gas. argon; detection, thermal conductivity detector; gas samples, 2 ml of 15-ml gas phase is injected with a gastight syringe.
576 METABOLISM

TABLE 9 Assav urocedures for t h e enzvmes of the mixed acid and butanediol fermentation"
Enzyme Assay mixture AA (nm) E (M-lcm-')

1. PEP-PTS (1) 50 mM KPO, buffer (pH 7.4),10 p M 14C-sugar,5 mM PEP, 12.5 mM


(EC 2.7.3.9) MgClZ, 25 mM potassium fluoride, 2.5 mM DTT, plus excess quanti-
ties of the soluble PTS enzymes for PEP-dependent reaction. For the
sugar-P-dependent reaction (transphosphorylation), PEP is replaced
by 10 mM sugar, the pH of the KPO, buffer is 6.0,and the soluble en-
zymes are omitted!
2.Pyruvate kinase Assay as described in Tables 2 and 3
3. Lactate dehydrogenase Assay as described in Table 7
(EC 1.1.1.27)
4.PFL (53)(EC 2.3.1.54)
Activation of PFL A two-armed test tube equipped with a ground-glass stopper with an
outlet for gas exchange is used for the anaerobic activation of PFL. To
one arm, enzyme I1 in MOPS buffer and 9 mM DTT are added to a
volume of 0.25 ml. To the second arm, 0.1 M MOPS buffer, 9 mM
DTT, 0.4 mM Fe(NH4)#304)2, 20 pM dichloro-phenol-indophenol,
10 pM 3(3,4-dichlorophenyl)-l,l-dimethylurea, 4 mM potassium
oxamate, 0.1 mM adenosylmethionine, 0.02 mg of flavodoxin, chloro-
plast fragments (with 10 pg of chlorophyll), and 0.2 mg of inactive
PFL are added. Volume, 0.25 ml; pH, 7.7;temp, 30C.'
Coupled assays for PFL
Forward reaction Tris-HC1 (0.1M) buffer (pH 8.1),10 mM DTT, 0.1 mM Fe(NH4)2(S04),, 340 6.24 x lo3
5 mM DL-malate, 1 mM NAD+, 10 mM sodium pyruvate, 0.055 mM
CoASH, 20 pg of citrate synthase, 25 pg of malate dehydrogenase,
0.1mg of BSA, and activated enzyme (in a total volume of 1 ml) are
added to the anaerobic cuvette.
Reverse reaction Tris-HCI (0.1M) buffer (pH 8.1),10 mM DTT, 0.1 mM Fe(NH4)2(S04)2, 340 6.24 x lo3
10 mM lithium acetylphosphate, 0.3mM NADH, 0.055 mM CoA, 53
mM potassium formate, 25 pg of lactate dehydrogenase, 5 pg of phos-
phate acetyltransferase, 0.1 mg of BSA, and activated enzyme are
added to an anaerobic cuvette in a final volume of 1 ml.
5. FHL (6)(EC 1.2.1.2) 50 mM Tris-HC1 buffer (pH 7.5),20 mM sodium formate, 2 mM benzyl 578 8.65 x 103
viologen (dichloride), and cell extract
6.Acetaldehyde dehydro- Assay as described in Table 5
genase (EC 1.2.1.4)
7.ADH (EC 1.1.1.1) Assay as described in Table 5
8. Phosphotransacetylase Assay as described in Table 5
(EC 2.3.1.8)
9.Acetate kinase Assay as described in Table 7
(EC 2.7.2.1)
10.PEP carboxylase 100 mM Tris-HC1 (pH 6.5),10 mM DTT, 0.15mM NADH, 10 mM 340 6.24 x lo3
(67,102) (EC 4.1.1.31) MgC12, 25 mM NaHC03, 1 U of malate dehydrogenase, 5 mM PEP,
and cell extract
1 1. Malate dehydrogenase 50 mM Tris (pH 7.6),50 mM L-malate (pH 7.0),2 mM NAD+, and cell 340 6.24 x lo3
(107)(EC 1.1.1.37) extract
12.Fumarase (39) 50 mM L-malate, 50 mM sodium phosphate buffer (pH 7.3),and cell 240 2.53 x lo3
(EC 4.2.1.2) extract
13. Fumarate reductase 50 mM Tris-SO4, 0.1mM EDTA buffer, 250 pM benzyl viologen, 20 mM 578 8.65 x lo3
(14)(EC 1.3.99.1) fumarate, and 20 mM glucose. The cuvette is evacuated and flushed
with 02-free argon before the addition of glucose oxidase and catalase
to ensure complete removal of residual oxygen. Sodium dithionite
(0.2mM) is then added to reduce the benzyl viologen and incubated
at 38C for 3 min prior to the addition of enzyme to start the reaction.
14.a-Acetolactate synthase 50 mM sodium acetate (pH 5.8), 40 mM sodium pyruvate, 0.87 mM TPP, 540
(106)(EC 2.2.1.6) 0.5 mM MnC12, and cell extract. After 25 min of incubation at 37"C,
0.9 ml of 2.5 N NaOH is added and 0.5 ml of this solution is mixed
with 2 ml of solution containing creatine and a-naphthol (1:l).This
reaction mixture is incubated for 20 min with intermittent shaking,
and the reaction is terminated by adding 2.5 ml of 2.5 N NaOH.

(Continued on next page)


22. Carbohydrate Fermentations 577

TABLE 9 (Continued)
Enzyme Assay mixture AA (nm) E (M-lcm-l)
15. a-Acetolactate 200 mM KPO, (pH 6.2), 10 mM DL-a-acetolactate, and cell extract. 540
decarboxylase (106) After 25 min of incubation at 37"C, 0.9 ml of 2.5 N NaOH is added
(EC 4.1.1.5) and 0.5 ml of this solution is mixed with 2 ml of solution containing
creatine and a-naphthol (1:l). This reaction mixture is incubated for
20 min with intermittent shaking. The reaction is terminated by
adding 2.5 ml of 2.5 N NaOH.
16. Acetoin reductase 340 6.23 x lo3
(106) (EC 1.1.1.4)
Reduction of acetoin 50 mM KP04 (pH 7.0), 5 mM acetoin, 0.1 mM NADH, and cell extract 340 6.23 x lo3
Oxidation of 2,3- 50 mM KP04 (pH 7.0), 100 mM 2,3-butanediol, 0.1 mM NAD, and cell
butanediol extract
"DTT,dithiothreitol; MOPS, morpholinepropanesulfonic acid; BSA, bovine serum albumin.
''The sugar phosphates used are glucose.6-phosphate with [L4Clglucose,fructose-1-phosphate with [L4C]fructose,and mannitol-1-phosphate with ['4C]mannitol. The
resin used for separating "C-sugar from ''C-sugar-phosphate is AG1-XZ, 50- to 100-mesh size, chloride form.
'The tube is evacuated and filled with 02-free argon, and 10 kl of 20 mM Fe(NH4)z(S04)zis then added to the first arm. The tube is preincubated under argon for
30 min. The contents of the two arms are mixed and the tube is illuminated by a daylight lamp. The enzyme is fully activated usually within 30 min. The larger chloro-
plast fragments are centrifuged and the mixture is stored at 0C. The low-molecular-weight components of the activated enzyme solution are removed by gel filtration
under argon with a Sephadex (3-25 column (1.6 by 20 cm) with an anaerobic buffer containing 50 mM MOPS (pH 7.6), 9 mM DTT,and 0.2 mM Fe(NH,),(SO,),.
Temperature, 4C; flow rate, 2 ml/min. The protein fraction is collected into argon-flushed tubes and stored at 0C.

TABLE 10 Assay procedures for the enzymes of the acrylate pathway for propionate production
Enzyme Assay mixture AA (nm) E (M-lcm-')
1. Lactate racemase (40) 100 mM KP04 (pH 7.2), 0.4 mM NAD+, 125 mM L-lactate, 2 U of 340 6.23 x lo3
(EC 5.1.2.1) D-lactate dehydrogenase, and cell extract
2. Propionyl-CoA 111 mM KPO, (pH 7.0), 11 mM sodium acetate, 0.44 mM 5,5'- 412 1.36 x 104
transferase (10, 93) dithiobis( 2-nitrobenzoate), 1.1 mM OAA, 0.2 mM propionyl-CoA,
(EC 2.8.3.1) 2.2 U of citrate synthase, 11 mM D-lactate, and cell extract
3. Lactyl-CoA dehydratase 50 mM KPO, (pH 7.5), 1 mM acrylyl-CoA, 10 mM sodium dithionite,
(10) (EC 4.2.1.54) 0.4 mM ATP, 10 mM MgS04, and cell extract. The reaction is termi-
nated after 5 min by adding 20 pl of concentrated perchlorate, the
product is centrifuged, and lactate in the supernatant is measured.
4. Acrylyl-CoA reductase 50 mM Tris-HCI (pH 7.0), 0.1 mM NADH, and cell extract. The reac- 340 6.23 x lo3
(38) tion is started by the addition of 0.03 mM acrylyl-CoA under anaero-
bic conditions.
5. D-Lactate dehydrogenase
(40, 63) (EC 1.1.1.28)
NAD+ independent 50 mM KPO, (pH 7.6), 0.15 mM 2,6+dichlorophenolindophenol, and 1 600 17.3 x 103
mM D-lactate or L-lactate, and cell extract or 100 mM KPO, (pH
7.0), 1 mM K3Fe(CN)6, 0.2 mg of BSA," 125 mM sodium-D-lactate, 420
and cell extract
NAD dependent 13.2 mM KPO, (pH 7.5), 6.6 mM pyruvate, 0.12 mM NADH, and cell 340 6.23 x 10'
extract
6. Pyruvate-ferredoxin oxi+ Assay as described in Table 7
doreductase (EC 1.2.7.1)
7. Phosphotransacetylase Assay as described in Table 7
(EC 2.3.1.8)
8. Acetate kinase Assay as described in Table 7
(EC 2.7.2.1)
"BSA, bovine serum albumin.
578 m METABOLISM

TABLE 11 Assay procedures for the enzymes of the succinate pathway for propionate production
Enzyme Assay mixture AA (nm) E (M-'cm-')
1. Lactate dehydrogenase (EC 1.1.1.28) Assay as described in Table 10
2. Methylmalonyl-CoA-pyruvate trans- 350 mM KP04 (pH 6.8), 10 mM sodium pyruvate, 0.4 mM 340 6.23 x lo3
carboxylase (112) (EC 2.1.3.1) methylmalonyl-CoA, 0.6 mM NADH, and cell extract
3. Malate dehydrogenase (EC 1.1.1.37) Assay as described in Table 9
4. Fumarase (EC 4.2.1.2) Assay as described in Table 9
5. Fumarate reductase (67) (EC 100 mM KPO, (pH 7.2), 5 mM fumarate, 0.15 mM NADH, 340 6.23 x lo3
1.3.1.6) and cell extract
6. CoA Transferase (119) (EC 2.8.3.8) 100 mM Tris-HCI (pH 7.5), 100 mM potassium butyrate 233 4.44
(pH 7.5), 20 mM MgC12, 0.10 mM acetoacetyl-CoA, 5%
(vol/vol) glycerol, and cell extract
7. (R)-Methylmalonyl-CoA mutase 12.5 mM KPO, (pH 7.5), 3 mM glutathione (reduced), 6.25 340 6.23 x 103
(49) (EC 5.4.99.2) mM sodium pyruvate, 6.25 mM sodium succinate, 0.05 U
of malate dehydrogenase (diluted in 1% bovine albumin),
0.1 U of OAA transcarboxylase, 0.1 U of methylmalonyl-
CoA racemase, 0.1 U of CoA transferase (enzymes are di-
luted in 50 mM phosphate buffer, pH 7.4), 0.125 mM
NADH, 1.5 mM acetyl-CoA, 0.003 mM DBC," and cell
extract. All the reagents except DBC are mixed by inver-
sion in the cuvette and incubated in a water bath with
same temp as the spectrophotometric cell chamber for
5 min. DBC is then added under dim light.
8. Methylmalonyl-CoA racemase (2) 12.5 mM Tris-HC1 (pH 7.8), 3 mM glutathione (reduced), 233 4.44
(EC 5.1.99.1) 6.25 mM sodium pyruvate, 6.25 mM sodium succinate,
0.05 U of malate dehydrogenase, 0.125 mM NADH, 0.1 U
of OAA transcarboxylase, 0.005 U of methylmalonyl-
CoA mutase, 1.5 mM acetyl-CoA, 0.1 U of CoA trans-
ferase, 0.01 pM DBC, and cell extract. The reaction con-
ditions are as described above for (R)-methylmalonyl-
CoA mutase.
9. Pyruvate-ferredoxin oxidoreductase Assay as described in Table 7
(EC 1.2.7.1)
10. Phosphotransacetylase (EC 2.3.1.8) Assay as described in Table 7
11. Acetate kinase (EC 2.7.2.1) Assay as described in Table 7
"DBC [(5,6-dimethyl benzimidazolyl) C0-5'-deoxyadenosine cobamide] should be stored in a light-proof container. Benzimidazolylcobamideor adenylcobamide can
also be used as coenzymes

TABLE 12 Assay procedures for the enzymes of the bacterial acetate fermentation pathway
Enzyme Assay mixture AA (nm) E (M-lcm-')
1. EMF' pathway enzymes Assay as described in Table 2
2. Pyruvate-ferredoxin oxi- Assay as described in Table 7
doreductase (EC 1.2.7.1)
3. Phosphotransacetylase Assay as described in Table 7
(EC 2.3.1.8)
4. Acetate kinase (EC Assay as described in Table 7
2.7.2.1)
5 . Formate dehydrogenase 100 mM triethanolamine hydrochloride (pH 7.5), 20 mM sodium 340 6.23 x lo3
(74) (EC 1.2.1.2) formate, 3 mM DTT," 1 mM NAD, or 1.4 rnM methyl viologen and
cell extract: 600 1.13 x 104
6. Formyl-THF synthetase 100 mM triethanolamine-hydrochloride(pH 8.0), 40 mM sodium formate, 350 2.49 x 104
(74) (EC 6.3.4.3) 5 mM (ATP), 10 mM magnesium chloride, 100 mM 2-mercaptoethanol,
40 mM Tris-HC1 (pH 7.4), 1 mM THF (stored in the Tris-HC1buffer
supplemented with mercaptoethanol), and cell extract. After 10 min of
incubation, 1 ml of 0.36 N HCI is added to stoD the reaction. The reac-
tion conditions are as described for formate dehydrogenase.
(Continued on next page)
22. Carbohydrate Fermentations 579

TABLE 12 (Continued)
Enzyme Assay mixture AA (nm) E (M-'cm-')
7. Methenyl-THF cyclo- 200 mM potassium maleate, (pH 7.0), 80 mM 2-mercaptoethanol, 356 24.9 x 103
hydrolase (74) (EC 40 mM KOH, 0.2 mM 5,lO-methenyl-THE and cell extract. The re-
3.5.4.9) action conditions are as described for formate dehydrogenase above.
8. 5,lO-Methylene-THF 200 mM potassium maleate (pH 7.0), 2.4 mM NAD', 0.96 mM 5,lO- 356 2.94 x 104
dehydrogenase (74) methylene-THF, 240 mM 2-mercaptoethanol, and cell extract. The
(EC 1.5.1.5) reaction measures the formation of NADH and 5,lO-methylenetetra-
hydrofolate. The reaction conditions are as described for formate de-
hydrogenase above.
9. 5,lO-Methylene-THF 100 mM KP04 (pH 7.2), 20 mM sodium ascorbate, 20 mM benzyl 578/555 8.65 X lo3
reductase (20) viologen, 0.13 mM methyl-THE and cell extract. The assay is per-
(EC 1.5.1.20) formed in argon-filled, serum-stoppered anaerobic cuvettes.
10. THF:B12 methyltrans- 0.066 mM methyl-B12,0.3 mM H4 folate, 5 mM DTT, and cell extract 525 8.6 mM
ferase (83) in a total volume of 0.8 ml
(EC 2.1.1.13)
11. CO dehydrogenase/
acetyl-CoA synthase
(21,80,81) (EC
1.2.99.2)
CO-MV reductase 100 mM KP04 buffer (pH 7.0), 30 mM methyl viologen, 3.2 mM DTT, 600 11.4 x 103
and cell extract. 02-freehydrogen gas or CO is bubbled into the assay
mixture in a serum-stoppered cuvette at 60C for 5 min. The nonspe-
cific reduction of the dye is corrected by using nitrogen as the gas
phase.
Exchange between 150 mM/5mM KPOdDTT (pH 6.0), 200 nmol/liter ''C-acetyl-CoA,
l-'4C-acetyl-CoA and 10 pg of ferredoxin 11. The amount of CO exchanged into the
and CO C-1 of acetyl-CoA is determined by bubbling the acidified reaction
mixture with nitrogen to remove I4COfrom the solution, and the I4C
in an aliquot of the reaction mixture is determined. The nanomoles of
[l-'4C]acetyl-CoA exchanged with CO are calculated by subtracting
the counts per minute left in the solution from the counts per minute
in the acetyl-CoA at time zero.
Exchange between 100 mM KP04 (pH 6), 0.266 mM acetyl-CoA, 4 mM ATP, 1.2 mM
I4CO and C-1 of Fe(NH4)z(S04)2,18.4 mM DTT, and cell extract. The assay mixture
acetyl-CoA is bubbled with CO for 5 min, and then 200 pl of CO is replaced
with 200 p1of I4CO (1.4 mM, 4.7 X lo6 cpm) and cell extract.
Incubation is at 55C for 3 min.'
"DTT, dithiothreitol.
bAllreagents are prepared anaerobically by boiling the water containing reagents and storing them under nitrogen. The assay is performed in nitrogen-filled,serum-
stoppered anaerobic cuvettes and the enzyme is added via a syringe. NAD+ reduction is monitored by measuring the absorbance at 340 nm, and methyl viologen re-
duction is measured at 600 nm.
T h e reaction is initiated with the addition of acetyl-CoA and is allowed to proceed for 15 min at 55C. The reaction is stopped by adding 0.04 ml of 4 M acetic
acid, which drops the pH to 3.5. The solution is then made alkaline with the addition of 0.2 ml of 2 N NaOH to hydrolyze acetyl-CoA over a period of 6 h at room
temperature. Carrier acetic acid (0.0032 mM) is added and the acetate is then isolated using chromatographyon Celite before assaying for I4C.
580 w METABOLISM

TABLE 14 Assay procedures for the enzymes of the acetate fermentation pathway in archaea
Enzyme Assay mixture
1. Kinases
Glucokinase (ADP) (50, 51) 100 mM Tris-HC1 (pH 7.8), 10 mM Mg Cl2,0.5 mM NADP+, 15 mM 340 6.23 x lo3
(EC 2.7.1.2) D-glucose, 2 mM ADF', 0.35 U of ~-glucose-6-phosphatedehydroge-
nase, and cell extract
Glucokinase (ATP) (86) 50 mM Tris-HC1 buffer (pH 8.0), 10 mM glucose, 2.5 mM ATP, 10 340 6.23 x lo3
(EC 2.7.1.2) mM MgCl2 . 6H20, 1 U of glucose-6-phosphatedehydrogenase, 1
mM NADP, and cell extract
Hexokinase (ATP) (27) 100 mM Tris-HC1 (pH 7.5), 4 mM ATF', 4 mM MgC12,l mM NADP+, 340 6.23 x lo3
(EC 2.7.1.1) 3 U of yeast glucose-6-phosphatedehydrogenase, 10 mM glucose,
and cell extract. The reaction is carried out at 50C.
2. Glucose-6-phosphate
isomerase
Formation of fructose-6- 100 mM Tris-HC1 (pH 7.0), 40 mM glucose-6-phosphate, 3 mM ATP, 340 6.23 x lo3
phosphate from glucose-6- 5 mM MgC12,0.5 mM NADH, 1 U of PFK, 1 U of fructose bisphos-
phosphate (33) phate aldolase, 50 U of triose phosphate isomerase, 9 U of glycerol-
(EC 2.7.1.11) 3-phosphate dehydrogenase, and cell extract. The reaction is car-
ried out at 50C.
Formation of glucose-6- 100 mM Tris-HC1 (pH 7.0), 10 mM fructose-6-phosphate,0.5 mM 3651340 6.23 X lo3
phosphate from fructose-6- NADP+, 0.3 U of glucose-6-phosphatedehydrogenase, and cell ex-
phosphate (33) tract. The reaction is carried out at 50C.
(EC 2.7.1.11)
3. PFK (ATP) (34)
(EC 2.7.1.11)
Forward reaction 100 mM Tris-HC1 (pH 6.0), 10 mM fructose-6-phosphate, 2.5 mM 365/340 6.23 X lo3
(discontinuous) ATF', and 12.5 mM MgC12. After preincubation at 85C the reaction
is started with an aliquot of cell extract (1 mg of enzyme), incubated
for 1-20 min, and stopped by rapid addition of EDTA to a final
concn of 50 mM. Fructose-l,6-bisphosphateformation is quantified
by adding 0.6 mM NADH, 0.2 U of aldolase, 1 U of triose phos-
phate isomerase, and 0.3 U of glycerol-3-phosphatedehydrogenase.
Reverse reaction 100 mM Tris-HC1 (pH 6.0,), 20 mM fructose-1,6-bisphosphate, 5 mM 340 6.23 x lo3
(continuous) ADP, and 25 mM MgC12. After preincubation at 85"C, the reaction
is started with an aliquot of cell extract (3 mg), incubated for 1-30
min, and stopped by rapid addition of EDTA to a final concn of
50 mM. Fructose-6-phosphate formation is quantified by adding
- - . isomerase. and 0.2 U
0.6 mM NADR 0.2 U of elucose-6-~hos~hate
of glucose-6-phosphatedehydrogenase.
PFK (ADP) (50, 115) (EC 100 mM MES buffer (pH 6.5), 10 mM MgC12, 10 mM fructose-6- 340 6.23 x lo3
2.7.1.146) phosphate, 0.2 mM NADH, 2.5 mM ADF', 3.9 U of glycerol-3-
phosphate dehydrogenase, 11 U of triose phosphate isomerase,
0.23 U of aldolase, and cell extract
PFK (PPi) (EC 2.7.1.90)
Fructose-6-phosphate as 100 mM Tris-HC1 (pH 7.0), 0.4 mM NADH, 1 mM fructose-6-phos- 340 6.23 x lo3
substrate (99) phate, 5 mM potassium pyrophosphate, 2 U of fructose bisphos-
phate aldolase, 5 U of glycerol-3-phosphatedehydrogenase, 5 U of
triose phosphate isomerase, and cell extract
Fructose-l,6-bisphosphate 100 mM Tris-HC1 (pH 7.0), 5 mM NADP+, 10 mM fructose-1,6- 340 6.23 x lo3
as substrate (99) bisphosphate, 5 mM potassium pyrophosphate, 3 U of glucose-6-
phosphate dehydrogenase, 2 U of glucose-6-phosphate isomerase,
and cell extract
4. Fructose bisphosphate al- 50 mM Tris-HC1 (pH 7.0), 0.2 mM NADH, 2.5 mM fructose bisphos- 340 6.23 x lo3
dolase (98) (EC 4.1.2.13) phate, 4 U glycerol-3-phosphatedehydrogenase, 11 U of triose
phosphate isomerase, and cell extract. The reaction is carried out at
50C.
5. Triose phosphate isomerase
(EC 5.3.1.1)
Dihydroxyacetone phos- 0.1 M Tris-HC1 (pH 7.3), 5 mM potassium arsenate, 8 mM NAD+, 340 6.23 x lo3
phate as substrate (91) 4 mM DHAF', 4 U of GAPDH, and cell extract
(Continued on next page)
22. Carbohydrate Fermentations rn 581

TABLE 14 (Continued)
Enzyme Assay mixture AA (nm) E (M-lcm-')
GAP as substrate (91) 100 mM HEPES (pH 7.3), 0.5 mM NADH, 4 mM GAP, 4 U of 340 6.23 x lo3
glycerol-3-phosphate dehydrogenase, and cell extract
6. GAPDH (NAD)
(EC 1.2.1.12)
Forward reaction (11) 100 mM HEPES-KOH buffer (pH 7), 200 mM KCL, 10 mM NAD+, 2 340 6.23 x lo3
mM ~~-glyceraldehyde-S-phosphate, and cell extract
Reverse reaction ( I 1) 100 mM HEPES-KOH buffer (pH 7), 200 mM KCL, 1 mM NADH, 340 6.23 x lo3
0.5-10 mM 3-phosphoglycerate, 100-500 pM 1,3-bisphosphoglyc-
erate, and cell extract
7. GAP:ferredoxin oxidore- 2 ml of 100 mM EPPS buffer (pH 8.4), 50 pl of 100 mM benzyl violo- 5 781600 7.4 x lo3
ductase (85) (EC 1.2.7.6) gen, a few microliters of 100 mM sodium dithionite, 250 pM GAP,
and cell extract
8. Phosphoglycerate mutase 100 mM Tris-HC1 (pH 8.0), 5 mM 2-phosphoglycerate, 5 mM PEP, 340 6.23 x lo3
(88) (EC 5.4.2.1) 0.5 mM ATP, 10 mM MgC12,0.4 mM NADH, 4.5 U of phospho-
glycerate kinase, 2.5 U of pyruvate kinase, 7.5 U of lactate dehydro-
genase, and cell extract
9. Enolase (89) (EC 4.2.1.11)
PEP formation 100 mM Tris-HCI (pH 8.0), 4 mM 2-phosphoglycerate, 3 mM 240 1.50?
MgS04, and cell extract (protein prepared in 50 mM glycyl-
glycine/NaOH buffer, pH 8.0)
PEP formation with 100 mM Tris-HC1 (pH 8.0), 2.5 mM 2-phosphoglycerate, 0.3 mM 340 6.23 x lo3
NADH oxidation NADH, 3 mM MgS04,5 mM ADP, 10 U of pyruvate kinase, 7.5 U
of lactate dehydrogenase, and cell extract. The reaction is carried
out at 60C.
10. Pyruvate kinase (45)
(EC 2.7.1.40)
Discontinuous assay 100 mM Tris-HCI, 1-5 mM PEP, 2 mM ADP, 5-10 mM MgC12, and 340 6.23 x 103
cell extract are incubated for 15-20 s, and the reaction is stopped by
rapid addition of 750 pl of ice-cold buffer (100 mM Tris-HC1, pH
7.0), 0.6 mM NADH, and 0.5 U of lactate dehydrogenase
Continuous assay 100 mM triethanolamine (pH 7.0), 1 mM PEP, 2 mM ADP, 5 mM 340 6.23 x lo3
MgC12,0.3 mM NADH, 1 U of lactate dehydrogenase, and cell ex-
tract. The reaction is carried out at 60C.
11. Pyruvate-ferredoxin oxi- 50 mM EPPS buffer (pH 8.4), 1 mM methyl viologen, 2 mM MgC12, 578 8.65 x lo3
doreductase (92) (EC 0.4 mM thiamine PPi, 0.1 mM CoA, 500 mM sodium pyruvate, and
1.2.7.1) cell extract
12. ACS (ADP forming) (43,
64) (EC 6.2.1.13): coupled
assay for acid production
from CoA derivative
ACS I 10 mM pyruvate, 5 mM MgCL2,0.4 mM thiamine PPi, 0.025 mM CoA, 600 9.78 x lo3
5 mM methyl viologen, 1 mM ADP, 10 mM KZHP04, and 40 mg
of pyruvate-ferredoxin oxidoreductase in 50 mM EPPS buffer (pH
8.4). Assays are carried out in serum-stoppered cuvettes under Ar.
ACS I1 Same as above except that the assay mixture contained 40 mg of 600 9.78 x lo3
indole pyruvate-ferredoxin oxidoreductase instead of pyruvate-ferre-
doxin oxidoreductase and 10 mM of indole pyruvate instead of
pyruvate
"DHAP, dihydroxyacetone phosphate; EPPS, N-(2-hydroxyethyl) piperazine-N9-3-propanesulfonicacid; ACS, acetyl-CoA synthetase
582 METABOLISM

22.12. REFERENCES precision, accuracy and dynamics of detection. Chem. Eng.


1. Aboulwafa, M., and M. H. Saier, Jr. 2002. Dependency Sci. 50:2601-2610.
of sugar transport and phosphorylation by the phospho- 18. Clark, D. P. 1989. The fermentation pathways of Esche-
enolpyruvate-dependent phosphotransferase system on richia coli. FEMS Microbiol. Rew. 5:223-234.
membranous phosphatidyl glycerol in Escherichiu coli: stud- 19. Clark, D. P., and J. E. Cronan, Jr. 1980. Acetaldehyde
ies with a pgsA mutant lacking phosphatidyl glycerophos- coenzyme A dehydrogenase of Escherichia coli. J. Bacteriol.
phate synthase. Res. Microbiol. 153:667-677. 144: 179-184.
20. Clark, J. E., and L. G. Ljungdahl. 1984. Purification and
2. Allen, S. H. G., R. W. Kellermeyer, and H. G. Wood. properties of 5,lO-methylenetetrahydrofolatereductase, an
1969. Methylmalonyl CoA racemase from Propioni-
bacterium shemnii. Methods Enxymol. 13:194-198. iron-sulfur flavoprotein from Clostridium formicoaceticum.
J. Biol. Chem. 259:10845-10849.
3. Andersch, W., H. Bahl, and G. Gottschalk. 1983. Level 21. Clark, J. E., S. W. Ragsdale, L. G. Ljungdahl, and J.
of enzymes involved in acetate, butyrate, acetone and bu-
tanol formation by Closnidium acetobutylicum. Eur. J. Appl. Wiegel. 1982. Levels of enzymes involved in the synthesis
of acetate from COz in Clostridium thennoautocrophicum.
Microbiol. Biotechnol . 18:3 2 7-33 2.
J. Bucteriol. 151:507-509.
4. Andersen, H. W., M. B. Pedersen, K. Hammer, and 22. Conway, T., G. W. Sewell, Y. A. Osman, and L. 0.
P. R. Jensen. 2001. Lactate dehydrogenase has no control Ingram. 1987. Cloning and sequencing of the alcohol de-
on lactate production but has a strong negative control on hydrogenase I1 gene from Zymomonas mobilis. J. Bacteriol.
formate production in Lactococcus lactis. Eur. J. Biochem. 169:2591-2597.
268~6379-6389. 23. Davies, R., and M. Stephenson. 1941. Studies on the ace-
5. Arfman, N., V. Worrell, and L. 0. Ingram. 1992. Use of tone butyl alcohol fermentation. 1. Nutritional and other
the tac promoter and &Iq for the controlled expression of factors involved in the preparation of active suspension
Zymomonas mobilis fermentative genes in Escherichiu coli of Cl. acetobutylicum (Wiezmann). Biochem. J. 35:1320-
and Zymomona mobilis. J. Bucteriol. 174:7370-7378. 1331.
6. Axley, M. J., D. A. Grahame, and T. C. Stadtman. 1990. 24. Dawes, E. A., D. J. McGill, and M. Midgley. 1971.
Escherichia coli formate-hydrogen lyase. Purification and Analysis of fermentation products. Methods Microbiol. 6A:
properties of the selenium-dependentformate dehydroge- 63-21 5.
nase component. J. Biol. Chem. 265:18213-18218. 25. Dimroth, P., and B. Schink. 1988. Energy conservation in
6a.Barabote, R. D., and M. H. Saier, Jr. 2005. Comparative the decarboxylation of dicarboxylic acids by fermenting
genomic analyses of the bacterial phosphotransferase sys- bacteria. Arch. Microbiol. 170:69-7 7.
tem. Microbiol. Mol. Biol. Rev. 69:608-634. 26. Dombek, K. M., and L. 0. Ingram. 1987. Ethanol pro-
7. Blusson, H., H. Petitdemange, and R. Gay. 1981. A new, duction during batch fermentation with Sacchuromyces
fast and sensitive assay for NADH-ferredoxin oxidoreduc- cereuisiue: changes in glycolytic enzymes and internal pH.
tase detection in Clostridia. Anal. Biochem. 110:176-181. Appl. Enuiron. Microbiol. 53:1286-1291.
8. Brockman, H. L., and W. A. Wood. 1975. D-Lactate de- 27. Dorr, C., M. Zaparty, B. Tjaden, H. Brinkmann, and B.
hydrogenase of Peptostreptococcus elsdenii. J. Bacteriol. Siebers. 2003. The hexokinase of the hyperthermophile
124: 1454-1461. Thermoproteus tenax. ATP-dependent hexokinases and
9. Brown, T. D., M. C. Jones-Mortimer, and H. L. ADP-dependent glucokinases, two alternatives for glucose
Kornberg. 1977. The enzymic interconversion of acetate phosphorylation in Archaea. J. Biol. Chem. 278: 18744-
and acetyl-coenzyme A in Escherichia coli. J. Gen. 18753.
Microbiol. 102:327-336. 28. Duree, P., and H. Bahl. 1996. Microbial production of
10. Brunelle, S. L., and R. H. Abeles. 1993. The stereo- acetone/butanol/isopropanol,p. 230-267. In H. G. Rehm
chemistry of hydration of acryly-CoA catalyzed by lactyl- and G. Reed (ed.), Biotechnobgy, 2nd ed, vol. 6. VCH,
CoA dehydratase. Bioorg. Chem. 21:118-126. New York, NY.
11. Brunner, N. A,, and R. Hensel. 2001. Nonphos- 29. Entian, K. D. 1980. Genetic and biochemical evidence for
phorylating glyceraldehyde-3-phosphatedehydrogenase hexokinase PI1 as a key enzyme involved in carbon catabo-
from Thennopoteus tenax. Methods EnxymoE. 331:117- lite repression in yeast. Mol. Gen. Genet. 178:633-637.
131. 30. Fernandez, R., P. Herrero, M. T. Fernandez, and F.
12. Bunch, P. K., F. Mat-Jan, N. Lee, and D. P. Clark. 1997. Moreno. 1986. Mechanism of inactivation of hexokinase
The ldhA gene encoding the fermentative lactate dehy- PI1 of Sacchuromyces cerewisiae by D-xylose. J. Gem
drogenase of Escherichia coli. Microbiology 143: 187- Microbiol. 132:3467-3472.
195. 31. Gancedo, J. M., D. Clifton, and D. G. Fraenkel. 1977.
13. Cary, J. W., D. J. Petersen, E. T. Papoutsakis, and G. N. Yeast hexokinase mutants. J. Biol. Chem. 252:4443-4444.
Bennett. 1990. Cloning and expression of Closnidium ace- 32. Gottschalk, G. 1985. Bacterial Metabolism. Springer-
tobutylicum ATCC 824 acetoacetyl-coenzyme Aacetate/ Verlag, New York, NY.
butyrate:coenzyme A-transferase in Escherichiu coli. Appl. 33. Hansen, T., M. Oehlmann, and P. Schonheit. 2001.
Enwiron. Microbiol. 56:1576-1583. Novel type of glucose-6-phosphate isomerase in the hyper-
14. Cecchini, G., B. A. Ackrell, J. 0. Deshler, and R. P. thermophilic archaeon Pyrococcus furiosus.J. Bacwriol. 183:
Gunsalus. 1986. Reconstitution of quinone reduction and 3428-3435.
characterization of Escherichia coli fumarate reductase ac- 34. Hansen, T., and P. Schonheit. 2000. Purification and
tivity. J. Biol. Chem. 261:1808-1814. properties of the first-identified archaeal, ATP-dependent
15. Chaillou, S., P. W. Postma, and P. H. Powels. 2001. 6-phosphofructokinase, an extremely thermophilic non-
Contribution of the phosphoeno1pyruvate:mannose phos- allosteric enzyme, from the hyperthermophile Desul-
photransferase system to carbon catabolite repression in ~UTOCOCCWamylolyticus. Arch. Microbiol. 173: 103-109.
Lactobacillus pentosus. Microbiology 147:671-679. 35. Hansen, T., and P. Schonheit. 2001. Sequence, expres-
16. Chen, J. S. 1995. Alcohol dehydrogenase: multiplicity sion, and characterization of the first archaeal ATP-
and relatedness in the solventeproducingclostridia. FEMS dependent 6-phosphofructokinase, a non-allosteric en-
Microbiol. Rev. 17:263-273. zyme related to the phosphofructokinase-B sugar kinase
17. Christensen, L. H., U. Schulze, J. Nielsen, and J. family, from the hyperthermophilic crenarchaeote
Villadsen. 1995. Acoustic off-gas analyser for bioreactors: Aeropyrum pemix. Arch. Microbiol. 177:62-69.
22. Carbohydrate Fermentations 583

36. Hartmanis, M. G., and S. Gatenback. 1984. Intermediary 54. Koga, S., I. Yoshioka, H. Sakuraba, M. Takahashi, S.
metabolism in Clostridium ucetobutylicum: levels of enzymes Sakasegawa, S. Shimizu, and T. Ohshima. 2000.
involved in the formation of acetate and butyrate. Appl. Biochemical characterization, cloning, and sequencing of
Environ . Microbiol . 4 7:12 77- 12 83. ADP-dependent (AMP-forming) glucokinase from two
37. Herrero, P., J. Galindez, N. Ruiz, C. Martinez-Campa, hyperthermophilic archaea, Pyrococcus furiosus and
and F. Moreno. 1995. Transcriptional regulation of the Thermococcus litoralis. J. Biochem. (Tokyo) 128:1079-
Succharomyces cerevisiue HXK1, HXK2 and GLKl genes. 1085.
Yeast 11:137-144. 55. Kundig, W., S. Ghosh, and S. Roseman. 1964. Phosphate
38. Hetzel, M., M. Brock, T. Selmer, A. J. Pierik, B. T. bound to histidine in a protein as an intermediate in a
Golding, and W. Buckel. 2003. Acryloyl-CoA reductase novel phosphotransferase system. Proc. Natl. Acud. Sci.
from Clostridium propionicum. An enzyme complex of pro- USA 52~1067-1074.
pionyl-CoA dehydrogenase and electron-transferring 56. Kundig, W., and S. Roseman. 1971. Sugar transport. 11.
flavoprotein. Eur. J. Biochem. 270:902-910. Characterization of constitutive membrane-bound en-
39. Hill, R. L., and R. H. Bradshaw. 1969. Fumarase. zymes I1 of the Escherichia coli phosphotransferase system.
Methods Enzymol. 13:91-99. J. Biol. Chem. 246:1407-1418.
40. Hino, T., and S. Kuroda. 1993. Presence of lactate dehy- 57. Labes, A., and P. Schonheit. 2003. ADP-dependent glu-
drogenase and lactate racemase in Megasphaera elsdenii cokinase from the hyperthermophilic sulfate-reducing ar-
grown on glucose or lactate. Appl. Enuiron. Microbiol. 59: chaeon Archaeoglobus fulgidus strain 7324. Arch. Microbiol.
255-259. 180:69-75.
41. Holland-Staley, C. A., K. Lee, D. P. Clark, and P. R. 58. Lamed, R., and J. G. Zeikus. 1980. Glucose fermentation
Cunningham. 2000. Aerobic activity of Escherichia coli al- pathway of Thermoanaerobacterium brockii. J. Bucteriol.
cohol dehydrogenase is determined by a single amino acid. 141: 1251-1257.
J. Bucteriol. 182:6049-6054. 59. Leonardo, M. R., P. R. Cunningham, and D. P. Clark.
42. Horvath, S. I., C. J. Franzen, M. J. Taherzadeh, C. 1993. Anaerobic regulation of the udhE gene, encoding
Niklasson, and G. Liden. 2003. Effects of furfural on the the fermentative alcohol dehydrogenase of Escherichia coli.
respiratory metabolism of Saccharomyces cerevisiue in J. Bucteriol. 175:870-878.
glucose-limited chemostats. Appl. Environ. Microbiol. 69: 60. Ljungdahl, L. G. 1986. The autotrophic pathway of ac-
407611086. etate synthesis in acetogenic bacteria. Annu. Rev.
43. Hutchins, A. M., X. Mai, and M. W. W. Adams. 2001. Microbiol. 40:415450.
Acetyl-CoA synthetases I and I1 from Pyrococcus furiosus. 61. Lobo, Z., and P. K. Maitra. 1977. Physiological role of
Me thods Enzymol. 331:158-1 67. glucose-phosphorylating enzymes in Saccharomyces cere-
44. Ismaiel, A. A., C. X. Zhu, G. D. Colby, and J. S. Chen. uisiue. Arch. Biochem. Biophys. 182:639-645.
1993. Purification and characterization of a primary sec- 62. Mackenzie, K. F., C. K. Eddy, and L. 0. Ingram. 1989.
ondary alcohol dehydrogenase from two strains of Modulation of alcohol dehydrogenase isoenzyme levels in
Clostridium beijerinckii. J. Bucteriol. 175:5097-5105. Zymomonas mobilis by iron and zinc. J. Bucteriol. 171:
45. Johnsen, U., T. Hansen, and P. Schonheit. 2003. 1063-1067.
Comparative analysis of pyruvate kinases from the hyper- 63. Macy, J. M., L. G. Ljungdahl, and G. Gottschalk. 1978.
thermophilic archaea Archglobus ful$w, Aeropyrum Pathway of succinate and propionate formation in
pernix, and Pyrobuculum uerophilum and the hyperther- Bucteroides fiagilis. J. Bucteriol. 134:84-91.
mophilic bacterium Thermotoga maritima: unusual regula- 64. Mai, X., and M. W. Adams. 1996. Purification and
tory properties in hyperthermophilic archaea. J. Biol. characterization of two reversible and ADP-dependent
Chem. 278:25417-25427. acetyl coenzyme A synthetases from the hyperthermo-
46. Jones, D. T., and D. R. Woods. 1986. Acetone-butanol philic archaeon Pyrococcus furiosus. J. Bacteriol. 178:
fermentation revisited. Microbiol. Rev. 50484-524. 5897-5903.
. .. ..
47. Jungermann, K., R. K. Thauer, E. Rupprecht, C. 65. Maitra, P. K., and Z. Lobo. 1971. A kinetic study of gly-
Ohrloff, and K. Decker. 1969. Ferredoxin mediated hy- colytic enzyme synthesis in yeast. J. Biol. Chem. 246475-
drogen formation from NADPH in a cell-free system of 488.
Clostridium kluyoeri. FEBS Lett. 3: 144-146. 66. Meile, L., L. M. Rohr, T. A. Geissmann, M. Heren-
48. Kashefi, K., and D. R. Lovley. 2003. Extending the upper sperger, and M. Teuber. 2001. Characterization of the D-
temperature limit of life. Science 301:934. xylulose 5-phosphate/~-fructose6-phosphate phosphoke-
49. Kellermeyer, R. W., and H. G. Wood. 1969. 2- tolase gene (xfp) from Bifidobucterium lactis. J. Bucteriol.
Methylmalonyl CoA mutase from Propionibucterium sher- 183:2929-2936.
manii. Methods Enzymol. 13:207-215. 67. Melville, S. B., T. A. Michel, and J. M. Macy. 1988.
50. Kengen, S. W., J. E. Tuininga, C. H. Verhees, J. van der Pathway and sites for energy conservation in the metabo-
Oost, A. J. Stams, and W. M. de Vos. 2001. ADP- lism of glucose by Selenomonas ruminantiurn. J. Bucteriol.
dependent glucokinase and phosphofructokinase from 170:5298-5304.
Pyrococcus furiosw. Methods Enqmol. 33141-53. 68. Mitchell, W. J. 1998. Physiology of carbohydrate to sol-
51. Kengen, S. W. M., J. E. Tuininga, F. A. M. de Bok, vent conversion by clostridia. Ado. Microb. Physiol. 39:
A. J. M. Stams, and W. M. de Vos. 1995. Purification and 31-130.
characterization of a novel ADP-dependent glucokinase 69. Mitchell, W. J., J. E. Shaw, and L. Andrews. 1991.
from the hyperthermophilic archaeon Pyrococcus furiosus. Properties of the glucose phosphotransferase system of
J. Biol. Chem. 270:30453-30457. Clostridium acetobutylicum NCIB 8052. Appl. Environ.
52. Keshav, K. F., L. P. Yomano, H. J. An, and L 0. Ingram. Microbiol. 5 7 :2534-2539.
1990. Cloning of the Zymomonas mobilis structural gene 70. Muller, V. 2003. Energy conservation in acetogenic bacte-
encoding alcohol dehydrogenase I (udhA):sequence com- ria. Appl. Enuiron. Microbiol. 69:6345-6353.
parison and expression in Escherichia coli. J. Bucteriol. 71. Neale, A. D., R. K. Scopes, J. M. Kelly, and R. E. H.
172:2491-2497. Wettenhall. 1986. The two alcohol dehydrogenases of
53. Knappe, J., H. P. Blaschkowski, P. Grobner, and T. Zymomonas mobilis. Eur. J. Biochem. 154:119-124.
Schmitt. 1974. Pyruvate formate lyase of Escherichiu coli: the 72. Neale, A. D., R. K. Scopes, R. E. H. Wettenhall, and
acetyl-enzyme intermediate. Eur. J. Biochem. 50:253-263. N. J. Hoogenraad. 1987. Pyruvate decarboxylase of
584 METABOLISM

Zymomonas mobilis: isolation, properties, and genetic ex- therophilic archaeon Pyrococcus furiosus: evidence for the
pression in Escherichia coli. J. Bacteriol. 169:1024-1028. operation of a novel sugar fermentation pathway. Arch.
73. OBrien, W. E., S. Bowien, and H. G. Wood. 1975. Microbiol. 158: 188-202.
Isolation and characterization of a pyrophosphate-depen- 89. Schafer, T., and P. Schonheit. 1993. Gluconeogenesis
dent phosphofructokinase from Propionibacterium sher- from pyruvate in the hyperthermophilic archaeon Pyro-
manii. J. Biol. Chem. 250:8690-8695. coccus furiosus: involvement of reactions of the Embden-
74. OBrien, W. E.,and L. G . Ljungdahl. 1972. Fermentation Meyerhof pathway. Arch. Microbiol. 159:354-363.
of fructose and synthesis of acetate from carbon dioxide by 90. Schonheit, P., and T. Schafer. 1995. Metabolism of hy-
Clostridium furmicoaceticum. J. Bacteriol. 109:626-632. perthermophiles. World J. Microbiol. Biotechnol. 11:26-
75. Pawluk, A., R. K. Scopes, and K. Griffiths-Smith. 1986. 57.
Isolation and properties of the glycolytic enzymes from 91. Schramm, A., M. Kohlhoff, and R. Hensel. 2001.
Zymomonas mobilis. Biochem. J. 238:275-281. Triose-phosphate isomerase from Pyrococcus woesei and
76. Poolman, B. 1993. Energy transduction in lactic acid bac- Methunothermus fervidus. Methods Enzymol. 331:62-77.
teria. FEMS Microbiol. Rew. 12:125-147. 92. Schut, G. J., A. L. Menon, and M. W. Adams. 2001.
77. Posthuma, C . C., R. Bader, R. Engelmann, P. W. 2-Keto acid oxidoreductases from Pyrococcus furiosus
Postma, W. Hengstenberg, and P. H. Pouwels. 2002. and Thennococcus litoralis. Methods Enzymol. 331:144-
Expression of the xylulose 5-phosphate phosphoketolase 158.
gene, @kA, from Lactobacillus pentosus MD363 is induced 93. Schweiger, G., and W. Buckel. 1984. On the dehydra-
by sugars that are fermented via the phosphoketolase path- tion of (R)-lactate in the fermentation of alanine to pro-
way and is repressed by glucose mediated by CcpA and the pionate by Clostridium propionicum. FEBS Lett. 171:79-
mannose phosphoenolpyruvate phosphotransferase sys- 84.
tem. Appl. Enuiron. Microbiol. 68:831-837. 94. Scopes, R. K. 1984. Use of differential dye-ligand chro-
78. Postma, P. W., J. W. Lengeler, and G. R. Jacobson. 1993. matography with affinity elution for enzyme purification:
Phosphoeno1pyruvate:carbohydrate phosphotransferase 2-keto-3-deoxy-6-phosphogluconate aldolase from Zymo-
system of bacteria. Microbiol. Rew. 57543-594. m o w mobilis. Anal. Biochem. 136525-529.
79. Ragsdale, S. W. 1992. Enzymology of the acetyl-CoA 95. Scopes, R. K., and K. Griffiths-Smith. 1984. Use of dif-
pathway of autotrophic COz fixation. Crit. Rew. Biochem. ferential dye-ligand chromatography with affinity elution
Mol. Biol. 26:261-300. for enzyme purification: 6-phosphogluconate dehydratase
80. Ragsdale, S. W., J. E. Clark, L. G. Ljungdahl, L. L. Lundie, from Zymomonas mobilis.Anal. Biochem. 136530-534.
and H. L. Drake. 1983. Properties of purified carbon monox- 96. Scopes, R. K., V. Testolin, A. Stoter, K. Griffiths-
ide dehydrogenase from Closnrdium thernwaceticum, a nickel, Smith, and E. M. Algar. 1985. Simultaneous purification
iron-sulfur protein. J. Biol. Chem. 258:236+2369. and characterization of glucokinase, fructokinase, and
81. Ragsdale, S. W., and H. G. Wood. 1985. Acetate biosyn- glucose-6-phosphatedehydrogenasefrom Zymomonas mo-
thesis by acetogenic bacteria. Evidence that carbon bilis. Biochem. J. 228:627-634.
monoxide dehydrogenase is the condensing enzyme that 97. Selig, M., K. B. Xavier, H. Santos, and P. Schonheit.
catalyzes the final steps of the synthesis. J. Biol. Chem. 1997. Comparative analysis of Embden-Meyerhof and
260:3970-3977. Entner-Doudoroff glycolytic pathways in hyperther-
82. Reeves, R. E., D. J. South, H. 1. Blytt, and L. G. mophilic archaea and the bacterium Thermotoga. Arch.
Warren. 1974. Pyrophosphate:D-fructose6-phosphate 1- Microbiol. 162217-232.
phosphotransferase. A new enzyme with the glycolytic 98. Siebers, B., H. Brinkmann, C. Dorr, B. Tjaden, H.
function of 6-phosphofructokinase. J. Biol. Chem. 249: Lilie, J. van der Oost, and C. H. Verhees. 2001.
7737-7741. Archaeal fructose-1,6-bisphosphatealdolases constitute a
83. Roberts, D. L., S. Zhao, T. Doukov, and S. W. Ragsdale. new family of archaeal type class I aldolase. J. Biol. Chem.
1994. The reductive acetyl coenzyme A pathway: se- 276:28710-28718.
quence and heterologous expression of active methyltet- 99. Siebers, B., and R. Hensel. 2001. Pyrophosphate-depen-
rahydrofolate:corrinoid/iron-sulfur protein methyltrans- dent phosphofructokinase from Thermoproteus tenax.
ferase from Clostridium thermoaceticum. J. Bacteriol. 176: Methods Enzymol. 33 154-62.
6 127-6130. 100. Siebers, B., H. P. Klenk, and R. Hensel. 1998. PPi-
84. Ronimus, R. S., E. de Heus, and H. W. Morgan. 2001. dependent phosphofructokinase from Thermoproteus
Sequencing, expression, characterisation and phylogeny of tenax, an archaeal descendant of an ancient line in phos-
the ADP-dependent phosphofructokinase from the hyper- phofructokinase evolution. J. Bacteriol. 180:2 137-2143.
thermophilic, euryarchaeal Thermococcus zilligii. Biochim. 101. Sprenger, G. A., U. Schorken, G. Sprenger, and H.
Biophys. Acta 1517:384-391. Sahm. 1995. Transketolase A of Escherichia coli K12. Eur.
85. Roy, R., A. L. Menon, and M. W. W. Adams. 2001. J. Biochem. 230525-532.
Aldehyde oxidoreductases from Pyrococcus furiosus. 102. Sridhar, J., M. A. Eiteman, and J. W. Wiegel. 2000.
Methods Enzymol. 331:132-1 43. Elucidation of enzymes in fermentation pathways used by
86. Sakuraba, H., Y. Mitani, S. Goda, Y. Kawarabayasi, and Clostridium thermosuccinogenes growing on inulin. Appl.
T. Ohshima. 2003. Cloning, expression, and characteriza- Enwiron. Microbiol. 66:246-251.
tion of the first archaeal ATP-dependent glucokinase from 103. Stanier, R. Y., J. L. Ingraham, M. L. Wheelis, and P. R.
aerobic hyperthermophilic archaeon Aeropyrum pernix. J. Painterm. 1986. The Microbial World. Prentice Hall, NJ.
Biochem. (Tokyo) 133:2 19-224. 104. Stetter, K. 0. 1988. Hyperthermophiles-physiology and
87. Sakuraba, H., I. Yoshioka, S. Koga, M. Takahashi, Y. enzymes. J. Chem. Technol. Biotechnol. 42:315-317.
Kitahama, T. Satomura, R. Kawakami, and T. Ohshima. 105. Stetter, K. 0. 1999. Extremophiles and their adaptation
2002. ADP-dependent glucokinase/phosphofructokinase,a to hot environments. FEBS Lett. 452:22-25.
novel bifunctional enzyme from the hyperthermophilic ar- 106. Stormer, F. C. 1975. 2,3-Butanediol biosynthetic system
chaeon Methanococcusjannaschii. J. Biol. Chem. 277: 12495- in Aerobacter aerogenes. Methods Enzymol. 41518-532.
12498. 107. Sutherland, P., and L. McAlister-Henn. 1985. Isolation
88. Schafer, T., and P. Schonheit. 1992. Maltose fermenta- and expression of the Escherichia coli gene encoding
tion to acetate, COZ and Hz in the anaerobic hyper- malate dehydrogenase.J. Bacteriol. 163:1074-1079.
22. Carbohydrate Fermentations 585

108. Taherzadeh, M. J., L. Gustafsson, C. Niklasson, and G. mophilic methanogenic archaea. J. Bacteriol. 183:7145-
Liden. 2000. Inhibition effects of furfural on aerobic 7153.
batch cultivation of Saccharomyces cereuisim growing on 117. Wang, H., and R. P. Gunsalus. 2003. Coordinate regu-
ethanol and/or acetic acid. J. Biosci. Bioeng. 90:374-380. lation of the Escherichia coli formate dehydrogenase
109. Thauer, R. K., K. Jungermann, and K. Decker. 1977. fdnGH1 and fdhF genes in response to nitrate, nitrite, and
Energy conservation in chemotrophic anaerobic bacteria. formate: roles for NarL and NarP. J. Bactaiol. 1855076-
Bacteriol. Rew. 41:lOO-180. 5085.
110. Thauer, R. K., K. Jungermann, E. Rupprecht, and K. 118. White, D. 2000. The Physiology and Biochemistry of Pro-
Decker. 1970. Hydrogen formation from NADH in cell- karyotes, 2nd ed. Oxford University Press, New York, NY.
free extracts of Closdium kluyweri. Acetyl coenzyme A 119. Wiesenborn, D. P., F. B. Rudolph, and E. T.
requirement and ferredoxin dependence. FEBS Lett. Papoutsakis. 1988. Thiolase from Clostridium aceto-
4: 108-1 12. butylicum ATCC 824 and its role in the synthesis of acids
111. Thimann, K. V. 1963. The Life of Bacteria. The and solvents. Appl. Enwiron. Microbiol. 54:2717-2722.
Macmillan Company, New York, NY. 120. Wiesenborn, D. P., F. B. Rudolph, and E. T.
112. Thompson, T. E.,and J. G. Zeikus. 1988. Regulation of Papoutsakis. 1989. Coenzyme A transferase from
carbon and electron flow in Propionispira arboris: relation- Closeridium acetobutylicum ATCC 824 and its role in the
ship of catabolic enzyme levels to carbon substrates fer- uptake of acids. Appl. Enwiron. Microbiol. 55323-329.
mented during propionate formation via the methyl- 121. Wood, H. G., S. W. Ragsdale, and E. Pezacka. 1986.
malonyl coenzyme A pathway. ]. Bacteriol. 170:5298- The acetyl-CoA pathway of autotrophic growth. FEMS
5304. Microbiol. Rew. 39:345-362.
113. Toh, H., and H. Doelle. 1997. Changes in the growth 122. Wood, T. 1970. Spectrophotometric assay for D-ri'bose-
and enzyme level of Zymomonas mobilis under oxygen- 5-phosphate ketol isomerase and for D-ribulose-5-P 3-
limited conditions at low glucose concentration. Arch. epimerase. Anal. Biochem. 33:297-306.
Microbiol. 168:46-52. 123. Wood, W. A. 1961. Fermentation of carbohydrates and
114. Tsolas, O., and B. L. Horecker. 1970. Isoenzymes of related compounds, p. 59-149. In I. C. Gunsalus and
transaldolase in Candido utilis. Arch. Biochem. Biophys. R. Y. Stanier (ed.), The Bacteria: a Treatise of S m t u r e and
136:287-302. Function, vol. 2. Academic Press, New York, NY.
115. Tuininga, J. E., C. H. Verhees, J. van der Oost, and 124. Woods, S. A., S. D. Schwartzbach, and J. R. Guest.
W. M. de Vos. 1999. Molecular and biochemical charac- 1988. Two biochemically distinct classes of fumarase in
terization of the ADP-dependent phosphofructokinase Escherichia coli. Biochim. Biophys. Acta 95414-26.
from the hyperthermophilic archaeon Pyrococcw furiosw . 125. Xavier, K. B., M. S. da Costa, and H. Santos. 2000.
J. Biol. Chem. 274:21023-21028. Demonstration of a novel glycolytic pathway in the hy-
116. Verhees, C. H., J. E. Tuininga, S. W. Kengen, A. J. perthermophilic archaeon Thermococcw &gii by ( 13)C-
Stams, J. van der Oost, and W. M. de Vos. 2001. ADP- labeling experiments and nuclear magnetic resonance
dependent phosphofructokinases in mesophilic and ther- analysis. J. Bacteriol. 182463211636.
Metabolism of Aromatic Compounds
JEROME J. KUKOR, BORIS WAWRIK, AND GERBEN J. ZYLSTRA

23.1. GROWTH OF BACTERIA WITH VOLATILE AROMATIC


SUBSTRATES ............................................ 586
23.2. APPROACHES TO THE STUDY OF AROMATIC HYDROCARBON
DEGRADATION .......................................... 588
23.3. EXTRACTION, DERIVATIZATION, AND ANALYSIS
OF METABOLITES ........................................ 589
23.4. ARENE MONOOXYGENASES VERSUS DIOXYGENASES ......... 591
23.5. OXYGEN CONSUMPTION ASSAYS ........................... 591
23.6. CELL-FREE ASSAYS: ARENE OXIDATION ..................... 592
23.7. CELL-FREE ASSAYS: RING CLEAVAGE AND SUBSEQUENT
REACTIONS ............................................. 593
23.8. REFERENCES ............................................ 593

There has been a long-standing fundamental and practical compound is somewhat dependent on the logP value (the
interest in microbial metabolism of aromatic compounds. logarithm of the partition coefficient in an n-octanol-water
The topic is of interest to ecologists and soil scientists who two-phase system) and the propensity of the substrate to dis-
are concerned with organic matter sequestration and rupt the cell membrane and cause lysis. In fact, many differ-
turnover as part of the global carbon cycle; to environmen- ent types of bacteria have evolved mechanisms to tolerate
tal engineers who want to design treatment systems to re- aromatic compounds and other solvents, mostly involving
mediate aquifers contaminated with aromatic hydrocar- metabolic changes resulting in membrane strengthening and
bon-containing fuels or solvents; to geneticists and efflux pumps to remove the compound from the membrane
evolutionary biologists who use genes encoding aromatic (25, 30, 46). Thus, it is not unexpected that direct addition
biodegradative pathways as models to understand tran- of liquid or solid aromatic compounds to growth media can
scriptional control, lateral gene transfer, and patchwork as- cause erratic or unpredictable bacterial growth due to toxicity
sembly of catabolic pathways; to biochemists who seek to of high concentrations of the substrate. The most obvious
probe the mechanisms by which the stable aromatic nu- way to overcome this problem is to initially add a small con-
cleus can be broken; and to biotechnologists in their quest centration of the substrate to the culture medium and then
to bioprospect f o r - o r construct via molecular rearrange- continually feed the substrate to the culture over time or to
ment-novel enzymatic catalysts that can be used to spike the culture at appropriate time intervals with new sub-
specifically modify chemical structures. strate. Continually feeding aromatic substrates to the culture
Given this long-standing interest in the metabolism of can be accomplished in a variety of ways, as described in the
aromatic compounds from such a diverse array of scientists, following paragraphs.
it is not surprising that the publications focused on the The most efficient method of providing an aromatic
topic are legion. A perusal of the Biocatalysis and Bio- compound as a carbon and energy substrate for growth is
degradation Database (http://umbbd.msi.umn.edu/) allows through the vapor phase, provided that the compound is
one to readily access several thousand publications in volatile. This methodology can be applied to cultures
PubMed that deal with specific enzymes or pathway inter- grown in either liquid or solid media as shown in Fig. 1
mediates related to aromatic compound biodegradation. A (49). In the case of a liquid culture in a flask, a glass bulb
single chapter that would cover all methods for all aromatic containing the liquid volatile aromatic substrate is sus-
compounds is an impossibility. Instead, the goal of this pended above the medium (Fig. 1A). As the substrate
chapter is to provide an overview of the generally used, evaporates, the atmosphere inside the flask becomes satu-
well-established methods that are the hallmark of aromatic rated and the substrate diffuses into the culture medium.
metabolism studies. To do this, we have chosen to focus on For most substrates it is important to vent the flask to the
toluene as a model aromatic substrate. In addition, we re- outside atmosphere (as shown in Fig. 1) in order to prevent
strict our focus to methods developed to study metabolism the substrate concentration from becoming too high. In the
of aromatics under aerobic conditions. case of highly toxic compounds it is sometimes advanta-
geous to insert a second glass tube through the stopper to
allow more outside air into the flask to keep the vapor con-
23.1. GROWTH OF BACTERIA WITH centration of the substrate low. The upper part of the glass
VOLATILE AROMATIC SUBSTRATES bulb should be stoppered with nonabsorbent cotton to
Bacteria variably tolerate the presence of aromatic com- maintain sterility. Since the stoppered flasks are open to
pounds in culture medium, where the toxicity of the aromatic the atmosphere, cultures should be grown in incubators

586
23. Metabolism of Aromatic Compounds H 587

a distance from each other in an appropriately vented incu-


A bator. Alternatively, petri dishes can be placed within
Nalgene screw-cap containers to contain the vapors. In
cases where a particularly volatile substrate is being used as
a growth substrate, several plates can be stacked up together
in one container, with only one or two tubes of substrate.
When using a less volatile substrate, it is important to in-
clude one tube of substrate per plate. A n alternative to this
approach is to place several petri plates in a glass desiccator
with a small beaker containing the growth substrate. It is es-
pecially important in these cases that the substrate concen-
perture tration not rise above the toxicity level for the organisms.
Since glass desiccators are completely sealed systems, it is
Volatile possible to add a substrate, let it evaporate completely, and
Hydrocarbon saturate the atmosphere at a target level. Thus, the calcu-
lated volume of the desiccator determines the amount of
liquid substrate added.
Mineral Although adding a volatile liquid substrate through the
Salts Medium vapor phase, as described above, works well, these methods
do not work very well with volatile solid substrates such as
naphthalene and biphenyl. These substrates can often be
added directly to liquid culture medium for growth, pro-
vided that the toxicity levels are low (see alternative meth-
ods below). Growth on solid medium can be accomplished
by placing crystals of the volatile solid substrate in the petri
dish lid. One disadvantage to this technique is that al-
though the entire atmosphere will become saturated with
vapors from the substrate, the area of the medium closest to
the crystals is exposed to a higher concentration. This can
often result in erratic growth or even no growth of the or-
ganism in areas on the petri dish where there are higher
Volatile Cotton Plug concentrations of the substrate. To evenly distribute the
Hydrocarbon substrate across the entire plate, a filter method can be em-
ployed. In this case, a sterile Whatman no. 1 filter is placed
FIGURE 1 Method for growing bacteria in the presence of in the petri dish lid. A solution (typically 100 to 300 1.1) of
volatile liquid aromatic hydrocarbons either in a shake flask substrate in methanol (or other appropriate volatile carrier)
(A) or on a solid medium (B). is applied to the filter. After the carrier liquid evaporates,
the filter is evenly saturated with the solid volatile com-
pound. The concentration of the solid substrate added to
with appropriate ventilation, for instance, with a shaker in the filter can be empirically adjusted based on the growth
a hood. In cases where the growth chamber is not ade- rate of the strain and tolerance for the hydrocarbon.
quately vented, an alternative is to place activated charcoal Less volatile and insoluble aromatic hydrocarbons, such
in the glass tubing between two cotton plugs to absorb the as phenanthrene, carbofuran, atrazine, etc., are more diffi-
substrate and prevent it from diffusing into the atmosphere cult to apply to solid culture medium. Three techniques are
of the incubator. As most rubber stoppers absorb hydrocar- commonly used in these cases: the use of top agar, spraying
bons, it is important to label each stopper with the substrate the surface of the plate, or deposition by sublimation. In the
chemical used and, after cleaning and resterilization, use case of top agar, an appropriate amount of substrate (finely
the stopper-bulb pair for the same substrate in subsequent ground) can be added to 5 ml of 0.5% Noble agar in media
experiments. A n alternative to the suspended bulb tech- salts. Even though the substrate does not dissolve, the solu-
nique is to utilize a flask with an enclosed side arm, such as tion can be mixed thoroughly and immediately poured on
a biometer flask or a Klett flask. The volatile hydrocarbon top of prepared medium in a petri dish. After the medium
can be placed in the side arm in a fashion similar to that of sets, cells can be applied to the surface and growth will
placing it in a suspended bulb. occur as the substrate diffuses slowly through the agar from
A similar approach can be used for growth of bacteria on the crystals. Alternatively, the substrate can be added to a
solid medium. A small (6- by 50-mm) tube containing the low-melting-point agar, the solution is cooled to just above
volatile liquid hydrocarbon can be placed in the lid of a gelling temperature, and then the cells are added directly to
petri dish (Fig. 1B). Evaporated liquid will saturate the at- the solution before it is poured onto base agar in a petri
mosphere, diffuse into the solid medium, and be available dish. This way, colonies will form within the top agar in
for growth of the bacteria. When stoppering these tubes close proximity to crystals of the growth substrate. The
with nonabsorbent cotton, it is important to make sure that spray plate technique can be utilized to apply a solid aro-
the cotton is fully inserted into the tube. If any part of the matic compound directly to the surface of the medium (32).
cotton extends out of the tube, it is possible that this will In this case the cells are commonly plated out on the agar
act as a wick and the liquid hydrocarbon substrate will es- first (and allowed to dry if cells were applied in solution). A
cape from the tube and perhaps melt the petri dish lid. 0.1% ether solution of the substrate is then carefully sprayed
When testing for growth on different volatile substrates, it on the plate in a chemical hood using a sprayer originally
is important to place petri dishes prepared in this fashion at designed for spraying thinelayer chromatography (TLC)
588 METABOLISM

plates. Care must be taken to cover the entire plate in an (dissolved in acetone) to an empty culture flask to give a
even fashion. Varying the thickness of the applied layer final effective concentration of 1 to 4 mM. The acetone was
controls the amount of substrate available for growth. evaporated under a stream of air, leaving a coat of fine dini-
More-controlled deposition can be achieved by the subli- trotoluene crystals on the bottom of the culture flask. An
mation technique (1). A visible layer of a water-insoluble appropriate amount of basal salts medium and XAD-7 resin
compound can be deposited onto plates by chilling and in- (3.5 g [dry weight] per liter; washed three times with
verting them onto a heated aluminum dish containing the methanol) were added to the dinitrotoluene-coated flask
substrate. As the bacteria grow and form colonies, clear prior to autoclaving. A final dissolved dinitrotoluene con-
zones should develop, indicating that the substrate is being centration of 20 to 200 mM was achieved in this manner
utilized (Fig. 2). Although ether is commonly used as a base after autoclaving and cooling. Following growth of a bacte-
to apply the substrate, any sufficiently volatile liquid can be rial culture in this medium, the culture was filtered through
utilized. Care must be taken that the solvent used is not glass wool to remove the XAD-7 resin prior to centrifuga-
harmful to the bacteria and does not itself serve as a growth tion to harvest the cells. A similar procedure was developed
substrate for the bacteria. by the same authors (42) to provide continuous but gradual
Another way to reduce the toxicity of volatile aromatic release of dinitrotoluene to bacteria in solidified media. For
substrates to cells in batch cultures is to provide the volatile this, the agar plates were prepared by grinding the XAD-7
substrate in a reservoir of a less-volatile organic solvent. For resin to a paste with a mortar and pestle. The ground resin
example, Hartmans et al. (24) were able to successfully ob- was added to media at a concentration of 7 g (hydrated
tain and grow styrene-degrading bacteria by providing 25 ~1 weight) per liter.
of styrene in a small tube containing 5 ml of dibutylphtha- Growing cells in large liquid volumes such as in a fer-
late. This tube was placed inside a larger flask in which the mentor or chemostat is a challenge as well. The high aera-
bacteria were to be grown. Using an organic solvent as a tion rate of these cultures strips volatile aromatic substrates
reservoir for potentially toxic volatile aromatic compounds out of the medium. This necessitates addition of the volatile
in enrichment and growth experiments allows for the slow growth substrate through the culture air feed. One way of
release of the substrate into the vapor phase. One can then doing this is to pass the air that is being used to sparge the
achieve high biomass concentrations at low aqueous sub- culture through a column or flask containing substrate crys-
strate concentrations in the growth medium without the tals or liquid. Although a single air feed set up in this way
need for continuous monitoring and addition of new sub- provides for efficient growth, two air feeds, one with air
strate. Care should be taken to ensure that a culture is not, only and one with air plus substrate, allow for more control
in fact, growing on the less volatile solvent. over the final substrate concentration in the growth
Amberlite XAD-7 resin can also be added to culture medium. Flowmeters should be used to control the influent
media as a way to provide continuous but gradual release of air stream(s) and hence the concentration of influent sub-
potentially toxic aromatic compounds. Nishino et al. (42) strate. Substrate concentrations can be measured in the in-
used this approach to grow dinitrotoluene degraders. The fluent and effluent air by gas chromatography (24,47,53).
protocol involved addition of an amount of dinitrotoluene Several methods for the enumeration of hydrocarbon-
degrading microorganisms are available. These methods are
generally based either on direct counting on agar plates or
on a type of most probable number (MPN) assay performed
in microtiter plates. Direct plate counting of hydrocarbon-
degrading bacteria can be performed by spraying agar plates
of serial dilutions of cells (26, 32) and counting the result-
ing clearing zones. Alternatively the substrate can be incor-
porated into a soft-agar overlay (7) to obtain clearing zones.
Many different variations of MPN methods are available.
Oil-degrading microorganisms, for example, can be enu-
merated by the Sheen Screen method (8). This procedure
estimates the number of oil-degrading bacteria by recording
their ability to emulsify when grown on oil as a sole carbon
source. Other methods consider the fact that complex en-
vironmental samples often contain both aliphatic and aro-
matic constituents. Aliphatic- and aromatic-hydrocarbon-
degrading bacteria can be enumerated separately by using
the MPN approach (see chapter 9) in microtiter plates con-
taining different substrates (62). In this method, alkane
degradation is indicated after 2 weeks of growth by the re-
duction of iodonitrotetrazolium violet, which was added to
the medium. Similarly, aromatic-hydrocarbon degradation
is indicated by the accumulation of yellow to greenish-
brown partially oxidized substrate.

FIGURE 2 Photograph of bacterial colonies on a plate that 23.2. APPROACHES TO THE STUDY OF
has phenanthrene applied to it by the spray plate technique. As AROMATIC HYDROCARBON DEGRADATION
the colonies grow, they form clear zones, indicating that the Aromatic hydrocarbon degradation is as complex as there
substrate is being utilized. This photograph was kindly provided are potential substrates. There are many ways by which mi-
by J. Philp, Napier University, Edinburgh, United Kingdom. croorganisms can metabolize aromatic hydrocarbons, but
23. Metabolism of Aromatic Compounds 589

while the enzymes and their substrates and products vary, on toluene, m- and p-xylene, and many other methyl-
the mechanisms utilized and the catabolic trends observed substituted aromatic compounds but not on benzene or eth-
are the same for all. Toluene and related aromatic com- ylbenzene. Organisms with the r i n g oxidizing dioxygenase
pounds have long been used as models for the analysis of pathway can grow on toluene, benzene, and ethylbenzene
aromatic hydrocarbon degradation due to the fact that but normally not on m- or p-xylene. Organisms with the
there are several catabolic pathways known for its degrada- ring-oxidizing monooxygenase pathway can often grow on
tion, illustrating the many possible ways by which aromatic toluene, benzene, phenol, and cresol. Of course, although
hydrocarbons can be metabolized (for reviews see references the substrate range for growth falls into distinct patterns,
67 and 68). As shown in Fig. 3 , toluene degradation under the exact substrate range for growth within each group is
aerobic conditions generally proceeds by oxidation to dihy- variable based on the ability of the initial oxygenase to ox-
droxylated intermediates which undergo ring cleavage and idize the substrate, the ability of subsequent enzymes in the
further metabolism to tricarboxylic acid (TCA) cycle inter- pathway to continue metabolism, and the ability of the
mediates. The initial oxidation reactions are catalyzed by strain to tolerate the toxic effects of a given aromatic com-
mono- or dioxygenases which add one atom or two atoms, pound and its metabolic products.
respectively, of molecular oxygen to the substrate. In the Although determining the substrate range for growth of
case of toluene, initial monooxygenases are known that can a particular strain points the investigator toward the partic-
oxidize the aromatic ring at any of the three possible ring ular class of enzyme and type of pathway involved in the
positions, resulting in phenolic compounds. Also, dioxyge- degradation of a particular hydrocarbon, further in-depth
nases that oxidize the aromatic ring adjacent to the methyl chemical and biochemical investigations must be performed
group (2,3-oxidation), resulting in cis-dihydrodiol com- to positively identify the enzymes and pathways involved.
pounds and monooxygenases that oxidize the methyl group Elucidation and confirmation of the catabolic pathway and
to a benzylic alcohol, have been described (see references the enzymes involved come from a combination of experi-
67 and 68). These initial catabolic enzymes act to channel ments involving analysis of wild-type and mutant culture
growth substrates into their respective catabolic pathways, filtrates for catabolic intermediates, I8O2experiments (to
and thus, their substrate range (and identity of the product distinguish mono- from dioxygenases), oxygen uptake assays
formed) determines the ability of the catabolic pathway to and biotransformation assays with whole cells, and enzyme
metabolize a particular substrate. For instance, in general, assays using crude cell extracts. The exact combination of
the different toluene catabolic pathways can be distin- approaches utilized depends on the goal(s) of the investiga-
guished based on the substrates upon which a micro- tor, the resources available, and the vagaries of the organism
organism can grow. Organisms with the methyl-oxidizing (ease of growth, stability and activity of the enzymes, etc.).
pathway, exemplified by the TOL plasmid (60), can grow Each of these approaches is outlined in depth in the sec-
tions below.

2 3.3. EXTRACTI0N, DERIVATlZ AT I 0N,


A N D ANALYSIS OF METABOLITES
Bacterial metabolism of aromatic hydrocarbons can result
in a variety of chemically different compounds, resulting
from the ring oxidation and the later ring cleavage steps.
These metabolites can be either neutral or charged and ei-
ther chemically stable or unstable, and thus, care must be
taken in the extraction process to stabilize and competently
extract the chemicals. These metabolites can be directly
identified by high-performance liquid chromatography
(HPLC) in culture supernatants if in high concentration or
identified by HPLC or gas chromatography (GC) following
1iquid:liquid extraction of the culture. Extraction of neutral

r r r s r
OH
metabolites is commonly performed with the addition of an
equal volume of ethyl acetate (or other solvent) to the cul-
ture broth or enzyme reaction mixture. After mixing and

I Dihydroxylated Intermediate
I phase separation, the organic layer is removed and the ex-
traction is repeated two more times with equal volumes of

r J. J. J.
solvent. The aqueous phase is then acidified to pH 1.8 with
hydrochloric or sulfuric acid. Acidic metabolites are then
extracted from the medium by three additional extractions

I Ring Cleavage
I with equal volumes of ethyl acetate. The ethyl acetate ex-
tracts from the neutral and acidic extractions are then sep-
arately dried over sodium sulfate, and the ethyl acetate is re-
moved by evaporation under vacuum with a rotary
evaporator. Because several of the metabolites are unstable
at elevated temperatures, care should be taken during rotary
evaporation not to heat the solvent above the temperature
at which the culture was grown. Following ethyl acetate re-
FIGURE 3 Representative known pathways for the degrada- moval, residues are taken up in a small volume of methanol
tion of toluene illustrating typical reactions catalyzed by en- for further chromatographic analysis. These methanolic ex-
zymes that act on aromatic compounds. tracts can be stored at -20C until analyzed.
590 METABOLISM

The extracted compounds can directly be examined by perature for several hours. Following derivatization, the re-
HPLC, and their retention times can be compared to au- action mixture is evaporated at room temperature with a
thentic compounds as standards. Typical HPLC conditions gentle stream of nitrogen and the dried derivatized metabo-
involve use of different solvent gradients under either neu- lites are suspended in methanol or another suitable solvent.
tral or acidic conditions. The exact solvent gradient utilized If any cis-dihydrodiols are present in the extract, then either
depends on the polarity and complexity of the suspected of these methodologies will result in a stable derivatized
metabolites. More-polar compounds elute early, whereas product with a corresponding identifiable peak on the GC.
compounds with multiple rings elute later. Methanol or ace- Although late-log-phase cultures actively growing on
tonitrile are often utilized as the mobile phase, with ace- the aromatic compound of interest or stationary-phase cul-
tonitrile commonly giving shorter retention times. Acidic tures are the preferred starting points for metabolite analy-
HPLC effluents (resulting from the use of 0.1% acetic acid sis, they often do not yield a large enough quantity of
or trifluoroacetic acid as the mobile phase) are commonly metabolites in the culture broth for full analysis and identi-
monitored based on absorbance at 254 or 280 nm for aro- fication. Mutant strains blocked in metabolism of the aro-
matic compounds and 190 or 210 nm for products resulting matic hydrocarbon (16, 17, 48) or modular units of genes
from ring cleavage. cloned and expressed in Escherichia coli (48, 69) provide for
Although neutral extracted compounds can often be di- the accumulation of large amounts of pathway intermedi-
rectly examined by GC-mass spectrometry (GC-MS), it is ates for chemical analysis. A concentrated suspension of
often advantageous to derivatize the compounds to produce bacterial cells that have been grown on a particular aro-
sharper GC peaks and to aid in MS identification. Common matic substrate can be used in a catalytic, resting-cell mode
methods involve acetylation or silylation of the hydroxyl to test the ability of these cells to transform the original sub-
groups and esterification of acidic groups. There are many strate as well as an array of other substrates (18). Resting-
different reagents and methods for acetylation and silyla- cell analyses involving highly concentrated induced bacte-
tion available commercially from companies such as rial cells can also be utilized to increase the possibility of
Alltech (Deerfield, IL) or Pierce (Rockford, IL). A common obtaining identifiable intermediates from the culture
method for acetylation is to evaporate the solvent from a medium due to differential rates of catalysis of the individ-
portion of the extracted metabolite with a gentle stream of ual enzymes in the catabolic pathway.
nitrogen in a test tube. Two milliliters of anhydrous pyridine Resting-cell assays can be performed on wild-type
and 1 ml of anhydrous acetic anhydride are added, and the strains, mutant strains, or induced clones in heterologous
solution is heated to 37C for 15 min. One milliliter of hosts. A typical resting-cell experiment consists of growing
water is then added, and the derivatized metabolite is ex- a wild-type bacterial culture in an appropriate basal salts
tracted from the reaction mixture with ether. The ether medium with an aromatic substrate as the sole carbon
layer is removed, dried with sodium sulfate, and evaporated source or growing a mutant bacterial strain on a suitable
under a gentle stream of nitrogen, and the derivatized non-catabolite-repressing substrate in the presence of the
metabolite is suspended in methanol or another suitable pathway-inducing compound to an A600of 0.6 to 1.0. In the
solvent for analysis. There is an amazing array of silylation case of heteroiogous expression of genes in E. coli, recombi-
reagents and procedures available for derivatization of nant cells are grown to an A6O0of 0.5 to 0.7, induced with
acidic and hydroxylation metabolites, many of which are the appropriate method (depending on the expression vec-
explained in detail elsewhere (6, 33). tor), and allowed to grow for one or two more hours. In all
Analysis of cis-dihydrodiol compounds, the products of cases, cells are harvested by centrifugation, washed at least
dioxygenase-catalyzed addition of molecular oxygen to the once in a suitable buffer (e.g., 50 mM sodium phosphate,
aromatic ring, can be problematic as these are not very sta- pH 7.5) to remove the growth medium, and resuspended in
ble and readily dehydrate to the corresponding phenols in a one-half to 1/20 of the original culture volume of the same
predictable fashion. For example, toluene cis-dihydrodiol buffer. This concentrated suspension of cells is no longer ca-
dehydrates under acidic and/or elevated temperature condi- pable of significant cell division since essential macronutri-
tions to o-cresol (16) while biphenyl cis-dihydrodiol dehy- ents such as nitrogen are lacking, hence the designation
drates to a 1:2 mixture of 2-hydroxy- and 3-hydroxy- resting cells. Resting cells can be used to test for the abil-
biphenyl (17). Thus, cis-dihydrodiols may dehydrate when ity of the strain to transform other carbon sources, to deter-
accumulating in culture medium (especially if the pH is less mine the substrate range of a particular catabolic enzyme
than neutral), during the extraction process, and even dur- (where cell-free assays are not possible), to determine po-
ing injection onto a GC or in the GC column. It is impor- tential pathway intermediates, and to obtain transforma-
tant therefore that these compounds be derivatized for ap- tion products that can be subsequently extracted and chem-
propriate analysis. Derivatization reactions have been ically characterized.
designed that not only derivatize the cis-dihydrodiols but Examples showing the utility of resting cells for the pro-
also, due to steric effects, demonstrate unequivocally that duction and analysis of cis-dihydrodiols from a wide range of
the hydroxyl groups are in the cis configuration. Two widely monocyclic and polycyclic compounds using various mu-
used methods are the synthesis of phenylboranate and ace- tant strains (16, 17, 48) and cloned genes (48, 69) have
tonide derivatives. Acetonide derivatives (16, 17) are syn- been elegantly described by Gibson and his coworkers.
thesized by drying the metabolite extract under a gentle Seeger et al. (51, 52) also used resting cells of E. coli carry-
stream of nitrogen and suspending the metabolites in 2 ml ing various cloned DNA fragments from Burkholderia sp.
of dimethoxypropane. p-Toluenesulfonic acid (100 pl of a strain LB400 or from Rhodococcus globerulus P6. The clones
10 mg ml- solution in dimethoxypropane) is added, and expressed enzymes in a pathway for degradation of substi-
the mixture is incubated for 1 h on ice. Phenylboronate de- tuted biphenyls. Use of individual recombinants allowed
rivatives (48) are prepared in a similar fashion, suspending the investigators to produce pathway intermediates that
the dried metabolite extract in 2 ml of ethyl acetate, adding were extracted, derivatized, and analyzed by GC-MS.
20 ~1 of a 10 mg ml- phenylboronic acid solution (in ethyl Similar approaches to obtain and identify products of aro-
acetate), and incubating at 75C for 15 minor at room tem- matic substrate transformation using cloned genes in resting
23. Metabolism of Aromatic Compounds rn 591

cells are given by Habe et al. (20) for transformation prod- pernatant can be extracted (see above) and analyzed by
ucts of dibenzofurans and dibenzo-p-dioxins, by Pollmann GC-MS.
et al. (45) for products of chlorotoluenes, and by Lessner et Caution should be exercised in the use of " 0 2 incorpo-
al. (36) for products of nitrobenzene. ration experiments to determine whether a dioxygenase or
a monooxygenase is involved in a new catabolic pathway in
an incompletely characterized isolate. Spain et al. (54) have
23.4. ARENE MONOOXYGENASES shown that toluene dioxygenase of P. putida F1 appears to
VERSUS DlOXYGENASES function as a monooxygenase in the transformation of 2,5-
As mentioned above, under oxic and suboxic conditions, dichlorophenol to 3,6-dichlorocatechol. Therefore, "0,
bacteria utilize a variety of oxygenases to initiate attack on incorporation studies alone are not always sufficient to un-
aromatic compounds. Oxygen incorporation into the aro- ambiguously determine the type of oxygenase that is in-
matic nucleus mediated by arene oxygenases results in the volved in an aromatic catabolic pathway.
production of dihydrodiols when the initial oxygenase is a
dioxygenase and in the production of phenolic intermedi-
ates when the initial oxygenase is a monooxygenase. When 23.5. OXYGEN CONSUMPTION ASSAYS
analyzing new bacterial strains with novel catabolic capa- Aerobic metabolism of aromatic compounds by bacteria in-
bilities, it is often necessary to demonstrate that an arene volves consumption of oxygen, either as a cosubstrate by
oxygenase is involved in a particular catabolic pathway and bacterial aromatic oxygenases or as a terminal respiratory
that the detected hydroxylated intermediates have arisen electron acceptor. Oxygen consumption can be used as a
from incorporation of molecular oxygen into their struc- measure of the ability of whole cells or cell extracts to uti-
tures. If one can identify a cis-dihydrodiol intermediate in lize an aromatic substrate. For whole cells, oxygen uptake
culture supernatants, then a priori the pathway proceeds via can be measured polarographically with a Clark-type oxy-
an initial dioxygenase attack of the aromatic nucleus. gen electrode (obtainable from Rank Brothers Ltd.,
However, if one detects only phenolic or dihydroxylated Cambridge, England, or Yellow Springs Instrument Co.,
products in the culture medium then it is uncertain whether Yellow Springs, OH). A typical reaction mixture (53, 56)
the catabolic pathway proceeds by an initial dioxygenase or would contain a cell suspension (typically containing 0.25
by two initial monooxygenases. Identifying which class of and 0.5 mg of protein) that has been washed free of growth
oxygenase initiates the catabolic pathway can be accom- medium by using aerated 20 to 50 mM sodium phosphate
plished through the use of an experimental system in which buffer (pH 7.0). After equilibration, background oxygen
the atmosphere is enriched in " 0 2 (15). For example, consumption is measured and substrate is added to a final
determination of whether a nonhydroxylated aromatic concentration of 500 pM (typically from a 10 mM stock).
compound is being converted to a dihydroxylated interme- Substrates can be added from aqueous stock solutions or, for
diate (e.g., a catecholic intermediate) via the action of an substrates that are poorly water soluble, from stocks made in
initial dioxygenase or via the sequential action. of two carriers such as dimethylformamide or dimethylsulfoxide. It
monooxygenations can be done in an atmosphere contain- is important to ascertain the effect of the substrate carrier
ing a 5050 mixture of l 6 0 2 and " 0 2 . From analysis of the on the basal oxygen consumption rate. The rate of sub-
dihydroxylated intermediates, one would expect a 5050 strate-dependent oxygen consumption must be corrected by
mixture of a dihydroxylated intermediate containing two subtracting the rate of endogenous oxygen consumption.
atoms of l60or two atoms of "0 from an initial dioxyge- The rates of oxygen consumption without a substrate (i.e.,
nase attack, whereas the product distribution of the dihy- endogenous consumption) and with an aromatic substrate
droxylated intermediate would be 25% containing two atoms are measured for at least 5. min.
of l60,25% containing two atoms of "0, and 50% con- It should be noted that oxygen uptake experiments mea-
taining one atom of l60and one atom of "0 from sequential sure all processes within the cell that consume oxygen.
monooxygenase attacks. The explanation for this type of Thus, addition of any substrate that enters the aromatic hy-
product distribution is that a dioxygenase incorporates both drocarbon catabolic pathway will result in whole-cell oxy-
atoms of molecular oxygen yielding a cis-dihydrodiol inter- gen consumption. provided that it is metabolized through
mediate which is subsequently dehydrogenated to yield a cat- one of the three routes requiring oxygen (initial oxygenase,
echol, whereas a monooxygenase incorporates only one atom ring-cleavage oxygenase, or tricarboxylic acid cycle). It is
of molecular oxygen at a time (the other atom being con- thus possible in the analysis of toluene degradation, for in-
verted to water). stance, to add cis-toluene dihydrodiol to toluene-induced
A typical experimental setup (23,54,56) consists of in- cells and observe oxygen uptake for strains metabolizing
duced cells harvested by centrifugation and resuspended in toluene by the cis-dihydrodiol pathway, even though the
a suitable buffer (e.g., 20 mM sodium phosphate at pH 7.0) nearest oxygen-consuming enzyme is the second metabolic
to a n A600 of 0.5 to 1.0. The suspension is transferred to a step downstream. Care should be taken in interpreting oxy-
flask that is sealed with a stopcock and placed on a mag- gen uptake experiments, as many aromatic hydrocarbon
netic stirrer. The air in the headspace of the flask is re- catabolic enzymes have broad substrate ranges. Thus, the
moved under vacuum and is replaced with nitrogen four ability of toluene-grown cells to oxidize cresols is not nec-
times. The headspace is then evacuated and refilled with air essarily due to a monooxygenase but could also be due to a
containing a 5050mixture of 1 6 0 2 and The gas in the dioxygenase (54). Certain bacterial strains have more than
headspace is analyzed by MS to determine the final compo- one catabolic pathway for a particular aromatic hydrocar-
sition of 1 6 0 2 and "0,. The experimental system is allowed bon (27), and this must also be taken into account when in-
to equilibrate for 15 min, and then the aromatic substrate to terpreting the data. One drawback to whole-cell oxygen up-
be tested is added, typically to a final concentration of 10 take experiments is the ability of the added substrate to
to 100 pM. The suspension is incubated with stirring for cross the cell membrane. While many aromatic hydrocar-
about 1 h and sparged with nitrogen for 5 min, and the cells bons partition into and diffuse across the membrane with-
are then removed by centrifugation. Metabolites in the su- out difficulty, many aromatic and aliphatic compounds,
592 METABOLISM

especially polar or charged compounds, have difficulty in scintillation counter. A negative control should be per-
diffusing across the membrane. This, however, is a problem formed to determine the background counts, especially as
only for testing compounds related to the growth substrate commercial radiolabeled toluene is often contaminated
or testing potential intermediates in the catabolic pathway, with minor amounts of radiolabeled polar compounds. A
since by definition the cells should be induced for transport similar assay has been developed for measuring the rate of
of the growth substrate (if a transport mechanism is oxidation of compounds less volatile than toluene. In the
needed). case of naphthalene (11, 12) or biphenyl (21, 22) dioxyge-
nase a similar reaction can be performed. In these cases,
however, the less volatile substrate must be removed under
23.6. CELL-FREE ASSAYS: ARENE OXIDATION a stream of warm air for 30 min in a chemical hood prior to
In order to get around the problem with transport of the en- scintillation counting of the products.
zyme substrate into the cell and to more precisely assay the Simple single-component aromatic oxygenases can eas-
enzymes for aromatic-hydrocarbon degradation, cell-free as- ily be assayed by measuring the disappearance of a specific
says should be performed. This is not an easy task, as many cosubstrate. For example, the activity of phenol hydroxylase
of the arene monooxygenases and dioxygenases consist of from the yeast Trichosporon curaneum (39) or chlorophenol
multiple components including an electron transport chain hydroxylase from Ralstonia eutrophu JMP134(pJP4) (37) can
that mediates transfer of electrons from NAD( P)H to the be measured by monitoring the disappearance at 340 nm of
oxygenase component, which actually performs the cat- the cosubstrates NADPH or NADH (as appropriate for the
alytic reaction. Due to this multicomponent nature of many individual enzyme). A typical assay mixture would contain,
oxygenases, cell-free enzymatic activity is quite low and in a 1-cm quartz cuvette, the following: 100 pmol of an ap-
does not show linearity with protein concentration. propriate buffer (e.g., potassium phosphate at pH 7.6), 0.5
Phthalate dioxygenase, one of the simplest multicompo- pmol of NAD(P)H, suitably diluted enzyme or cell extract
nent enzymes, consisting of a reductase and an oxygenase (diluted so that the reaction rate will be linear and propor-
component, can be assayed spectrophotometrically by fol- tional to the enzyme concentration), 0.5 pmol of phenolic
lowing the phthalate-dependent oxidation of NADH at substrate, and water to 3 ml. The reaction is initiated by ad-
340 nm (3). In a typical reaction, crude cell extract, ferrous dition of the substrate, and its rate is recorded for 1 to 4 min.
ammonium sulfate (10 p M final concentration from a freshly When using crude extracts, endogenous oxidation of
prepared stock solution), and NADH (100 p M final con- NAD(P)H must be taken into account. However, if this
centration) are added to 100 mM HEPES, pH 8.0. After the presents a problem, endogenous NAD( P)H oxidase activity
background rate of NADH oxidation is measured, phthalate can be removed by centrifugation of the extract at 100,000
is added at a final concentration of 2 mM. The rate of oxi- x g and using the supernatant for the assay (see, for exam-
dation of NADH is again monitored, and the difference in ple, Chapman and Ribbons [9]). One enzyme unit is defined
the rates before and after phthalate addition is the enzy- as the amount of enzyme which in the presence of a pheno-
matic activity of phthalate dioxygenase. lic substrate causes the oxidation of l pmol of NAD(P)H
Measuring the rate of oxygenases consisting of more per min.
than two components in crude cell extracts is problematic Some aromatic oxygenases require the addition of
due to the very low levels of measurable activity. Purified other cofactors besides NADH for optimal activity. For
electron transfer components, if available, can be added in instance, p-hydroxybenzoate hydroxylase, an enzyme in-
excess to the crude cell extract, and the rate of substrate- volved in the degradation of toluene via p-cresol (57, 58,
induced NAD( P)H oxidation, which is equivalent to the 63, 65, 66), requires the addition of flavin adenine dinu-
amount of oxygenase component present, is monitored. In cleotide (FAD) for maximal activity. p-Hydroxybenzoate
many cases, purified electron transfer components are not hydroxylase acts as a monooxygenase in the conversion of
readily available and enzyme assays must be performed using p-hydroxybenzoate to the dihydroxylated compound pro-
the native concentration of oxygenase. Given these condi- tocatechuate. p-Hydroxybenzoate hydroxylase activity can
tions, a radiometric assay is valuable due its extreme sensi- be measured spectrophotometrically by following the rate of
tivity. In general terms, the oxygenase reaction can be per- p-hydroxybenzoate-dependentoxidation of NADPH at 340
formed using a radiolabeled substrate, the volatile substrate nm. The assay mixture (13) contains, in a 1-cm cuvette in a
can be removed at a defined endpoint, and the polar non- final volume of 3 ml, the following: 33 mM Tris-sulfate (pH
volatile radioactive products can be measured in a scintilla- 8.0),0.33 mM sodium EDTA, 0.33 mM sodium p-hydroxy-
tion counter. In a typical 400-pl assay for toluene dioxyge- benzoate, 0.23 mM NADPH, and 3.3 p M FAD. The reac-
nase (55,64),crude cell extract is added to 50 mM Tris-HC1 tion is initiated by adding suitably diluted enzyme or cell
buffer, pH 7.5, along with 400 p M freshly prepared ferrous extract. When using cell extracts, endogenous oxidation of
sulfate. The mixture is incubated for 2 min to allow the fer- NADPH should first be determined. The FAD in this case
rous iron to reconstitute any enzyme that has lost its origi- is needed to reconstitute those enzymes which have lost
nal ferrous iron. NADH (1.2 mM final concentration) and their FAD.
14C-radiolabeled toluene (80 pM final concentration from Dihydrodiols are products of the action of dioxygenases
an N,N-dimethylformamide stock) are added, the reactants on aromatic substrates, as illustrated in Fig. 3. Dihydrodiol
are mixed, and the incubation is continued for 5 min. A 10- dehydrogenases are assayed using an NAD+/NADH-linked
~1 aliquot is then removed and spotted onto a 1- by 1-cm assay. Dihydrodiol dehydrogenase activity can be measured
plastic-backed silica gel square (Kodak chromagram sheet spectrophotometrically by following the increase in ab-
13181). The TLC square is placed in a chemical hood to sorbance at 340 nm concomitant with the conversion of
allow the volatile toluene to evaporate from the spotted re- NAD+ to NADH. A typical reaction mixture (53) would
action mixture. Nonvolatile polar products remain on the contain 0.5 pmol of NAD+, 0.1 pmol of the dihydrodiol
TLC square. Following an adequate drying period (30 min), substrate, 18 pmol of an appropriate buffer (e.g., Tris hy-
the TLC square is added to a scintillation vial along with drochloride at pH 8.0),and suitably diluted cell extract
scintillation cocktail and the radioactivity is measured in a (typically 0.02 to 0.06 mg of protein) in a final volume of
2 3 . Metabolism of Aromatic Compounds rn 593

1.O ml. When measuring extremely low rates of dihydrodiol substrate found in aromatic catabolic pathways.
dehydrogenase in crude cell extracts, it is important to use Protocatechuate 3,4-dioxygenase catalyzes intradiol cleav-
a sealable cuvette with gas ports so that the reaction mix- age of protocatechuate to yield P-carboxy-cis&-muconic
ture can be sparged with nitrogen. This prevents the oxida- acid. This enzyme can be assayed by monitoring the de-
tion of NADH by NADH oxidase in the cell extract. crease in A290due to the degradation of the substrate (14).
The assay mixture contains, in a final volume of 3 ml, 0.3
ml of 50 mM Tris-acetate (pH 7.5), 0.12 ml of 10 mM pro-
23.7 CELL-FREE ASSAYS: R I N G CLEAVAGE tocatechuate, and 0.1 ml of suitably diluted enzyme or cell
A N D SUBSEQUENT REACTIONS extract. A n alternative assay method is to polarographically
As illustrated in Fig. 3, metabolism of toluene yields dihy- monitor the consumption of oxygen during the reaction
droxylated intermediates such as catechol or methylcate- (59, 70). For this, the chamber of the oxygen polarograph is
chols, which are further converted into intermediates that filled with air-saturated MOPS (3-[N-morpholino]propane-
ultimately enter the T C A cycle. Metabolism of dihydroxy- sulfonic acid) buffer (pH 7.0). Once the temperature has
lated compounds proceeds by cleavage of the aromatic ring equilibrated, 1 pl of 100 mM protocatechuate is added and
either between (ortho-cleavage) or adjacent to (meta-cleav- a baseline is established on the recording device. A suitably
age) the hydroxyl groups. Aromatic ring cleavage in the lat- diluted aliquot of enzyme or cell extract is added, and oxy-
ter case is mediated by an extradiol catechol dioxygenase, gen consumption is monitored. As in the case of
commonly designated (methy1)catechol 2,3-dioxygenase (methyl)catechol, ring cleavage of protocatechuate can be
(C230) or metapyrocatechase. The activity of C 2 3 0 is de- via rneta-cleavage at either the 2,3- or the 45- position on
termined spectrophotometrically from the rate of product the aromatic ring. The activities of these two enzymes can
accumulation. A typical assay mixture (4,5,29,31,34,43) also be measured polarographically as for protocatechuate
contains in a quartz cuvette (l-cm light path and a final 3,4-dioxygenase (2, 61).
volume of 3 ml) the following: 0.3 ml of 50 mM potassium
phosphate buffer (pH 7.5), 0.1 ml of 10 mM catechol or We thank Kristen Foster for artwork. J.J.K. thanks NlEHS for their
methylcatechol dissolved in water (made fresh for each use support through Superfund basic research program grant P42-ES04911,
and G.J.Z. thanks NSF for their support through grants MCB-
as the material rapidly auto-oxidizes in the presence of air),
0078465 and CHE-9810248.
and 0.1 ml of suitably diluted enzyme or cell extract. The
rate of increase in absorbance is monitored at 375 nm for
catechol, 388 nm for 3-methylcatechol, and 382 nm for 4-
methylcatechol. When using cell extracts, heat treatment
23.8. REFERENCES
of the extract at 60C for 10 min prior to use allows the ring 1. Alley, J. F., and L. R. Brown. 2000. Use of sublimation to
fission product to accumulate by inactivating the enzymes prepare solid microbial media with water-insoluble sub-
involved in further metabolism of the muconate semialde- strates. Appl. Enuiron. Microbiol. 66:439-442.
hydes (53). 2. Arciero, D. M., A. M. Orville, and J. D. Lipscomb. 1990.
Metabolism of chlorobenzene yields chlorocatechol, Protocatechuate 4,5-dioxygenasefrom Pseudomonas testos-
which is further converted into intermediates that ulti- teroni. Methods Enzymol. 188:89-95.
3. Batie, C. J., E. LaHaie, and D. P. Ballou. 1987.
mately enter the T C A cycle by cleavage of the aromatic
Purification and characterization of phthalate oxygenase
ring between the hydroxylated carbons (i.e., intradiol and phthalate oxygenase reductase from Pseudomom cepa-
cleavage) to produce chloro-cis,cis-muconic acid. Further cia. J. Biol. Chem. 262:1510-1518.
metabolism of chloro-cis,cis-muconic acid proceeds by a 4. Bayly, R. C., S. Dagley, and D. T. Gibson. 1966. The me-
modified form of the p-ketoadipate pathway, with a diene tabolism of cresols by species of Pseudomonas. Biochern.
lactone (trans-4-carboxymethylene-but-2-en~4~olide) and J. 101:293-301.
maleylacetate as key intermediates (10, 28, 35,44, 47, 50). 5. Bayly, R. C., and G . J. Wigmore. 1973. Metabolism of
Chlorocatechol 1,2-dioxygenase (also known as catechol phenol and cresols by mutants of Pseudomom putida.
dioxygenase 11) has a broad substrate specificity (10, 38) J. Bacteriol. 113:1112-1120.
and relatively low k,,, values with catechol and chlorocate- 6. Blau, K., and J. Halket. 1993. Handbook of Deriwutiwes for
chols (41). The activity of chlorocatechol 1,2-dioxygenase Chromatography, 2nd ed. John Wiley & Sons, New York,
is determined spectrophotometrically from the rate of accu- NY.
mulation of the chloro-cis,cis-muconic acid product at 260 7. Bogardt, A. H., and B. B. Hemmingsen. 1992.
nm. A typical assay mixture (40) contains (final volume of Enumeration of phenanthrene-degrading bacteria by an
3 ml in a quartz cuvette with a l-cm light path) 0.1 mM cat- overlayer technique and its use in evaluation of petroleum-
echo1 or chlorocatechol, 1 mM EDTA, 33 mM Tris-HC1 contaminated sites. Appl. Enwiron. Microbiol. 58:2579-
buffer (pH 8.0),and suitably diluted enzyme or cell extract. 2582.
The enzyme that further transforms the cis,cis-muconic acid 8. Brown, E. J., and J. E Braddock. 1990. Sheen Screen, a
requires Mn2+for its activity, and unlike the chlorocatechol miniaturized most-probable-number method for enumera-
1,2-dioxygenase, this enzyme is inhibited by EDTA, allow- tion of oil-degrading microorganisms. Appl. Enwiron.
Microbiol. 56:3895-3896.
ing accumulation of the ring fission product. 9. Chapman, P. J., and D. W. Ribbons. 1976. Metabolism of
Many other aromatic substrates are also metabolized by resorcinylic compounds by bacteria: orcinol pathway in
way of catechol as the key ring fission intermediate (see, for Pseudomom putida. J. Bacteriol. 125:975-984.
example, p. 157-162 in Gottschalk [19]) and the catecholic 10. Dom, E., and H. J. Knackmuss. 1978. Chemical structure
substrate is cleaved by an intradiol catechol 1,2-dioxyge- and biodegradability of halogenated aromatic compounds.
nase (also known as catechol dioxygenase I, or pyrocate- Two catechol 1,2-dioxygenases from a 3-chlorobenzoate-
chase). Assay of this enzyme's activity is conducted in the grown pseudomonad. Biochem. J. 174:73-84.
same manner as for chlorocatechol 1,a-dioxygenase. 11. Ensley, B. D., and D. T. Gibson. 1983. Naphthalene
Besides catechol and methylcatechol, protocatechuate dioxygenase: purification and properties of a terminal oxy-
(the dihydroxylated substrate) is a common ring cleavage genase component. J. Bacten'ol. 155:505-511.
594 D METABOLISM

12. Ensley, B. D., D. T. Gibson, and A. L. Laborde. 1982. tion of an efflux pump involved in Pseudomom putidu S12
Oxidation of naphthalene by a multicomponent enzyme solvent tolerance. J. Biol. Chem. 273:85-91.
system from Pseudomonas sp. strain NCIB 9816. J. 31. Kim, E., and G. J. Zylstra. 1995. Molecular and bio-
Bacteriol. 149:948-954. chemical characterization of two meta-cleavage dioxyge-
13. Entsch, B. 1990. Hydroxybenzoate hydroxylase. Methods nases involved in biphenyl and m-xylene degradation by
Enzymol. 188:138-1 47. Beijhnckia sp. strain B1. 1. Bacteriol. 177:3095-3103.
14. Fujisawa, H. 1970. Protocatechuate-3,4-dioxygenase 32. Kiyohara, H., K. Nagao, and K. Yana. 1982. Rapid screen
(Pseudomom). Methods Enzymol. 17A:526-529. for bacteria degrading water-insoluble, solid hydrocarbons
15. Gibson, D. T., G. E. Cardini, F. C. Maseles, and R. E. on agar plates. Appl. Environ. Microbiol. 43:454-457.
Kallio. 1970. Incorporation of oxygen-18 into benzene by 33. Knapp, D. R. 1979. Handbook of Analytical Derivatization
Pseudomom putida. Biochemisq 9:1631-1635. Reactions. John Wiley & Sons, New York, NY.
16. Gibson, D. T., M. Hensley, H. Yoshioka, and T. J. 34. Kukor, J. J., and R. H. Olsen. 1991. Genetic organization
Mabry. 1970. Formation of (+)-cis-2,3-dihydroxy-l- and regulation of a meta cleavage pathway for catechols
methylcyclohexa-4,6-dienefrom toluene by Pseudomom produced from catabolism of toluene, benzene, phenol,
putida. Biochemisq 9: 1626-1630. and cresols by Pseudomom pickettii PKO1. J. Bacteriol.
17. Gibson, D. T., R. L. Roberts, M. C. Wells, and V. M. 173:4587-4594.
Kobal. 1973. Oxidation of biphenyl by a Beijerinckia 35. Kukor, J. J., R. H. Olsen, and J. S. Siak. 1989.
species. Biochem. Biophys. Res. Commun. 50:211-219. Recruitment of a chromosomally encoded maleylacetate
18. Gibson, D. T., G. J. Zylstra, and S. Chauhan. 1990. reductase for degradation of 2,4-dichlorophenoxyacetic
Biotransformations catalyzed by toluene dioxygenase from acid by plasmid pJP4. J. Bacteriol. 171:3385-3390.
Pseudomom putita F1, p. 121-132. In S. Silver, A. M. 36. Lessner, D. J., G. R. Johnson, R. E. Parales, J. C. Spain,
Chakrabarty, B. Iglewski, and S. Kaplan (ed.), Pseudo- and D. T. Gibson. 2002. Molecular characterization and
monas: Biotrunsformations, Pathogenesis, and Evolving Bio- substrate specificity of nitrobenzene dioxygenase from
technology. American Society for Microbiology, Wash- Commonas sp. strain JS765. Appl. Environ. Microbiol.
ington, DC. 68:634-641.
19. Gottschalk, G. 1986. Bacterial Metabolism, 2nd ed. 37. Liu, T., and P. J. Chapman. 1984. Purification and prop-
Springer-Verlag,New York, NY. erties of a plasmid-encoded 2,4-dichlorophenol hydroxy-
20. Habe, H., J. S. Chung, J. H. Lee, K. Kasuga, T. Yoshida, lase. FEBS Lett. 173:314-318.
H. Nojiri, and T. Omori. 2001. Degradation of chlori- 38. Nakagawa, H., H. Inoue, and Y. Takeda. 1963.
nated dibenzofurans and dibenzo-p-dioxinsby two types of Characteristics of catechol oxygenase from Brevibacterium
bacteria having angular dioxygenases with different fea- fuscum. J. Biochem. (Tokyo) 54:65-74.
tures. Appl . Environ. Microbiol. 67:36 10-36 17. 39. Neujahr, H. Y., and A. Gaal. 1973. Phenol hydroxylase
21. Haddock, J. D., and D. T. Gibson. 1995. Purification and from yeast. Purification and properties of the enzyme from
characterization of the oxygenase component of biphenyl Trichosporon cutaneum. Eur. J. Biochem. 35:386-400.
2,3-dioxygenase from Pseudomom sp. strain LB400. J. 40. Ngai, K. L., E. L. Neidle, and L. N. Ornston. 1990.
Bacteriol. 177:5834-5839. Catechol and chlorocatechol 1,2-dioxygenases. Methods
22. Haddock, J. D., L. M. Nadim, and D. T. Gibson. 1993. Enzymol. 188:122-126.
Oxidation of biphenyl by a multicomponent enzyme sys- 41. Ngai, K. L., and L. N. Ornston. 1988. Abundant expres-
tem from Pseudomom sp. strain LB400. J. Bacteriol. 175: sion of Pseudomom genes for chlorocatechol metabolism.
395-400. 1. Bacteriol. 170:24 12-24 13.
23. Haigler, B. E., and J. C. Spain. 1991. Biotransformation 42. Nishino, S. F., G. C. Paoli, and J. C. Spain. 2000.
of nitrobenzene by bacteria containing toluene degrada- Aerobic degradation of dinitrotoluenes and pathway for
tive pathways. Appl. Environ. Microbiol. 57:3156-3162. bacterial degradation of 2,6-dinitrotoluene. Appl. Environ.
244 Hartmans, S., M. J. van der Werf, and J. A. de Bont. Microbial. 6622139-2147.
1990. Bacterial degradation of styrene involving a novel 43. Nozaki, M. 1970. Metapyrocatechase. Methods Enzymol.
flavin adenine dinucleotide-dependent styrene monooxy- 17At522-525.
genase. Appl. Environ. Microbiol. 56:1347-1351. 44. Pieper, D. H., K..H. Engesser, and H.-J. Knackmuss.
25. Heipieper, J. H., F. J. Weber, J. Sikkema, H. Keweloh, 1989. Regulation of catabolic pathways of phenoxyacetic
and J. A. M. de Bont. 1994. Mechanisms of resistance of acids and phenol in Alcaligenes eutrophus JMP134. Arch.
whole cells to toxic organic solvents. Trends Biotechnol. Microbiol. 151:365-37 1.
12409-415. 45. Pollmann, K., S. Beil, and D. H. Pieper. 2001.
26. Heitkamp, M. A., and C. E. Cerniglia. 1987. Effects of Transformation of chlorinated benzenes and toluenes by
chemical structure and exposure on the microbial degrada- Ralstonia sp. strain PS12 tecA (tetrachlorobenzene dioxy-
tion of polycyclic aromatic hydrocarbons in freshwater and genase) and tecB (chlorobenzene dihydrodiol dehydroge-
estuarine ecosystems. Environ. Toxicol. Chem. 6:535-546. nase) gene products. Appl. Environ. Microbiol. 67:4057-
27. Kahng, H. Y., J. C. Malinverni, M. M. Majko, and J. J. 4063.
Kukor. 2001. Genetic and functional analysis of the tbc 46. Ramos, J. L., E. Duque, J. J. RodriguezeHerva, P.
operons for catabolism of alkyl- and chloroaromatic com- Godoy, A. Haidour, F. Reyes, and A. Fernandez-
pounds in Burkholderia sp. strain JSl50. Appl. Environ. Barrero. 1997. Mechanisms for solvent tolerance in bacte-
Microbiol . 67 :4805-48 16. ria. J. Biol. Chem. 272:3887-3890.
28. Kaphammer, B., J. J. Kukor, and R. H. Olsen. 1990. 47. Reineke, W., and H. J. Knackmuss. 1984. Microbial me-
Regulation of tfdCDEF by tfdR of the 2,4-dichlorophe- tabolism of haloaromatics: isolation and properties of a
noxyacetic acid degradation plasmid pJP4. J. Bacteriol. chlorobenzene-degrading bacterium. Appl. Environ.
172:2280-2286. Microbiol. 47:395-402.
29. Kataeva, I. A., and L. A. Golovleva. 1990. Catechol2,3- 48. Resnick, S. M., and D. T. Gibson. 1996. Regio- and
dioxygenases from Pseudomom aeruginosa 2x. Methods stereospecific oxidation of fluorene, dibenzofuran, and di-
Enzymol. 188:115-1 2 1. benzothiophene by naphthalene dioxygenase from
30. Kieboom, J., J. J. Dennis, J. A. de Bont, and G. J. Pseudomom sp. strain NCIB 9816-4. Appl. Environ.
Zylstra. 1998. Identification and molecular characteriza- Microbiol. 62:4073-4080.
23. Metabolism of Aromatic Compounds 595

49. Rosenberg, E. 1992. The hydrocarbon-oxidizing bacteria, 60. Williams, P. A., and K. Murray. 1974. Metabolism of
p. 446459. In A. Balows, H. G. Truper, M. Dworkin, W. benzoate and the methylbenzoates by Pseudomonas putida
Harder, and K.-H. Schleifer (ed.), The Prokmyotes, 2nd ed. (uruilla) mt-2: evidence for the existence of a TOL plas-
Springer-Verlag,New York, NY. mid. J. Bacteriol. 120:416-423.
50. Schmidt, E., and H. J. Knackmuss. 1980. Chemical 61. Wolgel, S. A., and J. D. Lipscomb. 1990. Protocatechuate
structure and biodegradability of halogenated aromatic 2,3-dioxygenase from Bucillw mucerans. Methods Enrymol.
compounds. Conversion of chlorinated muconic acids into 188:95-101.
maleoylacetic acid. Biochem. J. 192:339-347. 62. Wrenn, B. A., and A. D. Venosa. 1996. Selective enu-
51. Seeger, M., B. Camara, and B. Hofer. 2001. Dehalo- meration of aromatic and aliphatic hydrocarbon degrading
genation, denitration, dehydroxylation,and angular attack bacteria by a most-probable-number procedure. Can. J.
on substituted biphenyls and related compounds by a Microbiol. 42: 252-25 8.
biphenyl dioxygenase.J. Bucteriol. 183:3548-3555. 63. Wright, A., and R. H. Olsen. 1994. Self-mobilization and
52. Seeger, M., K. N. Timmis, and B. Hofer. 1995. organization of the genes encoding the toluene metabolic
Conversion of chlorobiphenyls into phenylhexadienoates pathway of Pseudomom mendocinu KR1. Appl. Enuiron.
and benzoates by the enzymes of the upper pathway for Microbiol. 60:235-242.
polychlorobiphenyl degradation encoded by the bph locus 64. Yeh, W. K., D. T. Gibson, and T. N. Liu. 1977. Toluene
of Pseudomom sp. strain LB400. Appl. Environ. Microbiol. dioxygenase: a multicomponent enzyme system. Biochem.
61:2654-2658. Biophys. Res. Commun. 78:401410.
53. Spain, J. C., and S. E Nishino. 1987. Degradation of 1,4- 65. Yen, K. M., and M. R. Karl. 1992. Identification of a new
dichlorobenzene by a Pseudomonas sp. Appl. Enuiron. gene, tmoF, in the Pseudomom mendocina KR1 gene clus-
Microbiol. 5 3 :1010-10 19. ter encoding toluene-4-monooxygenase.
.- J. Bacteriol.
54. Spain, J. C., G. J. Zylstra, C. K. Blake, and D. T. 1~:7253-7261.
Gibson. 1989. Monohydroxylation of phenol and 2,5- 66. Yen, K. M., M. R. Karl, L. M. Blatt, M. J. Simon, R. B.
dichlorophenol by toluene dioxygenase in Pseudomom Winter, P. R. Fausset, H. S. Lu, A. A. Harcourt, and
putidu F1. Appl. Environ. Microbiol. 55:2648-2652. K. K. Chen. 1991. Cloning and characterization of
55. Subramanian, V., T. N. Liu, W. K. Yeh, and D. T. a Pseudomom mendocinu KR1 gene cluster encoding
Gibson. 1979. Toluene dioxygenase: purification of an toluene-4-monooxygenase. J. Bucteriol. 1 7 3 5 315-
iron-sulfur protein by affinity chromatography. Biochem. 5327.
Biophys. Res. Commun. 91:1131-1139. 67. Zylstra, G. J. 1995. Molecular analysis of aromatic hydro-
56. Warhurst, A. M., K. F. Clarke, R. A. Hill, R. A. Holt, carbon degradation, p. 83-115. In S. J. Garte (ed.), Molec-
and C. A. Fewson. 1994. Metabolism of styrene by ular Environmental Biology. Lewis Publishers, Boca Raton,
Rhodococcus rhodochrous NCIMB 13259. Appl. Environ. FL.
Mimobiol. 60: 1137-1 145. 68. Zylstra, G. J., and D. T. Gibson. 1997. Aromatic hydro-
57. Whited, G. M., and D. T. Gibson. 1991. Separation and carbon degradation: a molecular approach, p. 183-203. In
partial characterization of the enzymes of the toluene-4- J. K. Setlow (ed.), Genetic Engineering: Principles and
monooxygenase catabolic pathway in Pseudomonas men- Methods. Plenum Press, New York, NY.
docina KR1. J. Bucteriol. 173:3017-3020. 69. Zylstra, G. J., and D. T. Gibson. 1989. Toluene degrada-
58. Whited, G. M., and D. T. Gibson. 1991. Toluene-4- tion by Pseudomonas putidu F1. Nucleotide sequence of the
monooxygenase, a three-component enzyme system that todCl CZBADE genes and their expression in Escherichia
catalyzes the oxidation of toluene to p-cresol in coli. J. Biol. Chem. 264:14940-14946.
Pseudomom mendocinu KR1. J. Bacteriol. 173:3010-3016. 70. Zylstra, G. J., R. H. Olsen, and D. P. Ballou. 1989.
59. Whittaker, J. W., A. M. Orville, and J. D. Lipscomb. Cloning, expression, and regulation of the Pseudomom
1990. Protocatechuate 3,4-dioxygenase from Brevibac- cepuciu protocatechuate 3,4-dioxygenase genes. J. Buc-
terium fuscum. Methods Enumol. 188:82-88. teriol. 1715907-59 14.
Cellulases, Hemice1lulases, and Pectinases
MICHAEL E. HIMMEL, JOHN 0. BAKER, WILLIAM S. ADNEY,
AND STEPHEN R. DECKER

24.1. INTRODUCTION ......................................... 596


24.1.1. Cellulases .......................................... 597
.2. Hemicellulases ....................................... 599
24.2. CELLULASE, HEMICELLULASE, AND PECTINASE ASSAYS-
OVERVIEW.............................................. 599
24.2.1. IUPAC General Methods for Cellulase and Hemicellulase
..................................... 599
Mixtures
24.2.1.1. Method Using Automated IUPAC Approach .......... 600
24.2.2. General Non-IUPAC Methods for Cellulase Mixtures .......... 600
24.2.2.1. Methods from Empirical Mathematical Models ......... 600
.2. Method Using a Membrane Dialysis Reactor .......... 600
.3. Use of Multiangle Laser Light Scattering to Estimate
MW Reduction of Cellulose ...................... 601
.............. 601
.4. Detection of Cellulase Activity in Gels
24.2.3. Assays for Specific Cellulase Enzyme Classes................. 601
24.2.3.1. Endoglucanase Analysis ......................... 601
.2. Exoglucanase Analysis .......................... 602
.3. p-D-Glucosidase Analysis ........................ 602
24.2.4. General Hemicellulase Assays ............................ 602
24.2.4.1. Hemicellulose-DebranchingEnzymes ............... 604
24.2.5. Hemicellulose Depolymerization Enzymes ................... 604
.6. Pectinases .......................................... 605
24.3. REFERENCES ............................................ 605

24.1. INTRODUCTION strength and flexibility to plants, but also the crystalline
Fossil fuels, which are finite and nonrenewable, supplied cores are quite resistant to chemical and biological degra-
86% of the energy consumed in the United States in 2002. dation. As a result, proposed renewable energy schemes
Biomass is renewable, offers an alternative to conven- that rely on fermentable sugars from plant polysaccharides
tional energy sources, and provides national energy secu- are impeded partly because of the recalcitrance of one of
rity, economic growth, and environmental benefits. The the major constituents, crystalline cellulose. The study of
potential impact of biofuels is great, but both mid- and cellulases is also important from the standpoint of micro-
long-term research strategies to obtain a better under- bial conversion of biomass to feeds and chemical feed
standing of plant cell wall biosynthesis and degradation stock.
are needed. The complex structure of hemicelluloses has dictated a
Plant cell walls are the planets dominant form of bio- correspondingly diverse array of hemicellulases. Generally,
mass. Plant cell walls and structural tissues are composed each structural feature in hemicellulose has an associated
of three primary structural polymers: cellulose, a ho- enzyme that can hydrolyze or chemically modify this fea-
mopolymer of p-( 1,4)-linked cellobiose residues; hemicel- ture; however, many enzymes needed to form and decon-
lulose, a branched cross-linking heteropolymer of varied struct cell wall structures remain a mystery today. The
composition; and lignins (see chapter 25 in this volume for definitive enzymatic degradation of cellulose to glucose,
a discussion of lignin and ligninolytic enzymes). Plant cell probably the most desirable fermentation feedstock, is gen-
wall is a complex of cellulose microfibrils embedded in a erally accomplished by the synergistic action of three dis-
matrix of hemicellulose and lignin. Studies consistently tinct classes of enzymes (Fig. 1): endoglucanases, exoglu-
show positive correlation between the enzymatic di- canases, and p-glucosidases (cellobiases) (115). The
gestibility of cellulose and the removal of hemicellulose enzymes involved in the deconstruction of various hemi-
(62, 81), supporting the notion of close spatial relation- celluloses have significantly more variability, as the bond
ships between these two polymers. Cellulose is the most specificities are much more complex than those in cellulose
abundant renewable organic polymer in the biosphere and (Fig. 2 and 3 ) . A recent review by Wyman et al. (163) cov-
is highly crystalline, water insoluble, and relatively resis- ers more details on these enzymes and their activity on
tant to depolymerization. Crystalline cellulose is of partic- plant biomass.
ular scientific interest because strands of cellulose make up Nature has evolved numerous enzymes that effect the
the core of the structural elements comprising plant cell depolymerization of cellulose to glucose. Bacterial cellu-
walls. Not only do these elementary microfibrils provide lases allow herbivores to digest cellulose into glucose, and

596
24. Cellulases, Hemicellulases, and Pectinases m 597

+ +-
- - - - - - - -
+ \$
- - - - - - - -
GIG GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC GIC

\
\
\ +
\
\
I
I

\
%.+\
- endoglucanase GIC- GIC
q*.........
+
GIG + GIC

- exoglucanase 4
4 - p-Glucosidase
FIGURE 1 Action of the major cellulase enzymes on cellulose. Endoglucanases cleave random
internal P-1-4 bonds. The major exoglucanases cleave cellobiose units from the reducing chain
ends in a processive manner, whereas minor exoglucanases may cleave glucose units from the non-
reducing end of the chain (not shown). P-Glucosidases cleave cellobiose to two glucose units.

fungal celluloses allow organisms to decompose woody ma- the natural deconstruction of plants lies the key to im-
terials. Of particular interest in the conversion of biomass to proved biomass conversion processes.
transportation fuels is the cellobiohydrolase I (Cel7A) en-
zyme isolated from Trichoderma reesei. In many ways, this 24.1 .l.Cellulases
highly active enzyme with specificity for crystalline cellu- 1. The endo-l,4-P-glucanases or 1,4-P-D-glucan 4-
lose is the key component in a complex cellulase system glucanohydrolases (EC 3.2.1.4) act randomly on soluble
containing many other (lesser) activities. Also of keen in- and insoluble 1,4-P-glucan substrates. These are commonly
terest are the pathways of degradation utilized by microbial measured by detecting the decrease in viscosity or reducing
plant decay enzyme systems, for hidden in the knowledge of groups released from carboxymethylcellulose (CMC) (137).

A.
Ph
01
MeGlcA
@I ESA?
Xyl - Xyl - Xyl - Xyl - Xyl - Xyl - Xyl - Xyl - Xyl
4 4
Xyl - Xyl
Endoxylanase @ a-Glucuronidase 0 Esterase
p-Xylosidase @ a-Arabinosidase

B.

Gal
I
Glc - Man - Man - Glc - Man - Man - Man
4 4
Glc - Man Man - Man
8
4 Endomannanase @ B-Glucosidase 0Esterase
a-Galactosidase -&b-Mannosidase

FIGURE 2 Action of major enzymes involved in the depolymerization of generic xylan (A) and
glucomannan (B) chains. Additional debranching enzymes are required for the numerous variants
found in the different hemicelluloses.
598 rn METABOLISM

6 y 3

FIGURE 3 More specific action of debranching enzymes involved in the deconstruction of ara-
binoxylan. The structure is a generalized diagram of arabinoglucuronoxylan. Enzyme activities: 1,
endoxylanase; 2, acetyl xylan esterase; 3, a-L-arabinofuranosidase; 4, a-D-glucuronidase; 5 , ferulic
acid esterase; 6, acetyl esterase; and 7, P-xylosidase.

2 . T h e exo- 1,4-@.D-ghcanasesRinclude both the 1,4- which liberates D-cellobiose from 1,4+-glucans. Differ-
@-D-glucan glucohydrolases (EC 3.2.1.74), which liberate entiation of these enzyme classes requires analytical tech-
D-glucose from 1,4-@-D-glucans and hydrolyze D-cellobiose niques to distinguish glucose and cellobiose and is usually
slowly, and 1,4-@-D-glucancellobiohydrolase (EC 3.2.1.91), carried out by high-performance liquid chromatography
24. Cellulases, Hemicellulases, and Pectinases 599

(HPLC) or gas chromatography (GC) (5, 22). These en- ined by microscopy, using traditional light microscopy,
zymes can be further distinguished by their ability to liber- electron microscopy, and more recently atomic force mi-
ate free sugars from either the reducing or the nonreducing croscopy (63, 66, 85, 89, 120, 126, 131, 166). This type of
end of the cellulose chain (59,90). Determination of which observation is critical when the properties of the cellulose
specificity a given enzyme has is usually carried out through fiber are in question, not when the goal is total hydrolysis.
synergistic studies with enzymes of known orientation (67, There has also been work using gel permeation chromatog-
70, 128). raphy to characterize changes to cellulose structure by ex-
3. The p-D-glucosidases or P-D-glucoside glucohydro- amining the products of cellulase action on wood fiber
lases (EC 3.2.1.21) release D-glucose units from cellobiose (121). This said, workers in the field are reminded that
and soluble cellodextrins, as well as an array of glycosides. only assays designed to measure the conversion of cellulose
Measurement of this activity is carried out on cellobiose or from the actual biomass substrates in question are ulti-
cello-oligomers, with product analysis by HPLC or G C mately valid performance measures. As with much of
or via direct spectrophotometric or fluorometric analysis of biotechnology today, high-throughput methods have also
various chromogenic and fluorogenic analogues of cel- been developed to increase the speed and accuracy of cel-
lobiose and cello-oligomers (17, 38, 102, 144). lulase assay (41).
For critical measurement of cellulase and hemicellulase
specific activity, precise and accurate measurements of pro-
24.1.2. Hemicellulases tein content are also necessary. A rounderobin assay of the
1. The endoacting hemicellulase enzymes attack poly- protein content in commercial cellulase preparations
saccharide chains internally, with very little activity on demonstrated that labs already working in the field were
short oligomers; i.e., the degree of polymerization (DP) is not able to agree within 1 or 2 standard deviations when
<3. Endoxylanases (EC 3.2.1.8) are specific for @-(1+4)- using the Bradford dye binding, Kjeldahl, and Biuret meth-
xylopyranose polymers (i.e., the backbone of xylan) while ods (4). Note that for these experiments the samples were
others are specific for other hemicellulose polymers, such as standardized using protein determination by amino acid
mannan [endo-(1+4)-P-mannosidases; EC 3.2.1.781 or p- analysis.
glucans [endo-(1+3)-P-~-glucosidase; EC 3.2.1.391. As
with endocellulases, these activities can be measured 24.2.1. IUPAC General Methods for Cellulase
through viscosimetry or by measuring the production of and Hemicellulase Mixtures
reducing sugar end groups from a given hemicellulosic As a result of significant effort by an international commit-
polymer. tee of cellulase researchers and the International Union of
2. The exoacting enzymes tend to act from either the re- Pure and Applied Chemists (IUPAC), a procedure was pub-
ducing or the nonreducing termini and are specific to the lished in 1987 describing the use of microcrystalline cellu-
type and length of the polymer. Some exoacting enzymes lose and measurement of reducing sugars by the dinitrosali-
act preferentially on short-chain substrates (DP, 2 to 4), and cylic acid method of Miller (109) in the context of a highly
some are more active on larger substrates (DP, >4). Xylan specific assay protocol (56). In fact, the text of this protocol
(1+4)-P-xylosidase (EC 3.2.1.37), glucan (1+3)-P- must be followed carefully to achieve comparable results.
glucosidase (EC 3.2.1.58), and mannan (1+4)-manno- The rationale developed in this IUPAC method is that to
biosidase (EC 3.2.1.100) are exoacting enzymes specific for be maximally useful, all assays for cellulase activity must be
xylan, p-(1+3)-glucan, and mannan, respectively. applied to an identical cellulosic substrate, i.e., Whatman
3. The so-called accessoryenzymes are also required for no. 1 filter paper, and that enzyme action on the substrate
hydrolysis of hemicellulose in native plant tissue. This cat- must be measured during the initial first-order reaction
egory includes a variety of acetyl xylan esterases (EC rates. The definitive level of hydrolysis has been set at 4%
3.1.1.72), acetyl esterases (EC 3.1.1.6), and esterases that (wtlwt) of the cellulose from a 1.0- by 6.0-cm (50-mg)
hydrolyze lignin glycoside bonds, such as ferulic acid es- Whatman no. 1 test coupon; i.e., 2 mg, converted to glu-
terase (EC 3.1.1.73) (146) as well as enzymes involved in cose after a 60-min incubation at 50C. After accounting
the cleavage of specific hemicellulose sidechains (such as for the addition of water to the glycosidic bond, the actual
a-L-arabinofuranose), glucuronic acid, and 4-0-methyl- level of hydrolysis measured is 3.6% of the substrate. The
glucuronic acid groups. concentration (or actually dilution) of enzyme preparation
required to effect this level of depolymerization is con-
verted, through a somewhat indirect procedure, to the cel-
lulase activity in filter paper units (FPU) per milliliter. For
24.2. CELLULASE, HEMICELLULASE, example, an undiluted cellulase preparation that yields ex-
AND PECTlNASE ASSAYS-OVERVIEW actly 2 mg of glucose during the IUPAC assay has 0.37
Literature describing the assay of general cellulase activity FPU/ml. This fractional unit is the lowest cellulase activity
(or of individual component enzymes) has broadened con- measurable with the IUPAC assay. Note that because the
siderably since the first reports by Mandels et al. (94) indi- IUPAC FPU assay is nonlinear due to hydrolysis of an in-
cating that reducing-sugar release and substrate weight loss soluble (and structurally heterogeneous) substrate, the use
could serve as suitable cellulase assay methods. To some ex- of traditional international units of enzyme activities, based
tent (for appropriate substrates), these methods are still on initial velocities, is invalid. Here, a single incubation
considered adequate. However, modern assays based on time (60 min), a single temperature (50C), and, crucially
molecular weight (MW) analysis detected by HPLC-size important, a single resulting extent of hydrolysis (2 mg of
exclusion chromatography (SEC), coupled enzymes, vis- glucose-equivalents released from 50-mg filter paper) are
cometry, hydrolysis of dyed or derivatized insoluble and sol- used for all samples. It should be stressed that this assay is
uble polymers, and hydrolysis of derivatized or labeled low difficult to reproduce consistently. Attention must be paid
MW substrates have greatly enhanced the understanding to all of the details, including assay tube diameter, rolling
of these complex systems. Cellulose structure and physical and positioning of the filter paper strip, and avoidance of
disruption of cellulose microfibrils have also been exam- large, single-step dilutions.
600 METABOLISM

The IUPAC cellulase assay has many significant limita- 24.2.2.1. Methods from Empirical
tions, and it merely serves as the best existing method. The Mathematical Models
IUPAC commission warns, for example, that extrapolation Contributions made in the assay of cellulase preparations by
of required glucose release from highly diluted or concen- Sattler et al. (133) describe a relationship between extent of
trated solutions of enzyme is not permitted. Indeed, the as- hydrolysis, reaction time, and enzyme concentration. This
says used to confirm the release of 2 mg of glucose must be procedure permits the effectiveness of different enzymes and
conducted with enzyme dilutions that closely bracket the of different pretreatment methods to be ranked. For this
actual value. The implication is that cellulase solutions too method, cellulose hydrolysis data collected from hyperbolic
dilute to release 2 mg of glucose must either be concen- functions of substrate concentration versus cellulase enzyme
trated to an appropriate level or pronounced not assayable concentration at various timed incubations are examined.
by the IUPAC method. The authors have also noted that A double-reciprocal plot based on the relationship
the nonlinearities inherent in the assay render erroneous
the estimation of new cellulase activities from simple dilu-
tion. For highest accuracy, every working solution made
from an enzyme stock must be reanalyzed for activity, a con- where Y/C, is the fraction of substrate hydrolyzed; [El is
dition that complicates most analytical procedures. given in filter paper units per gram of substrate initially
A similar approach was proposed for measuring hemicel- added; and Ym,,/C, is the fraction of substrate that could be
lulases in 1987 (58). This method relies on meeting a stan- maximally hydrolyzed at an infinite enzyme concentration.
dard level of conversion of the xylan fraction in oat spelt The y-axis intercept in the double-reciprocal plot,
xylan to xylose in a specified period of time under standard (Ymax/Co)-,may be used to quantify the quality of the en-
conditions. zyme preparation. Ideally, an enzyme should have a high
Y,, and a low value for KCJY,,,. Adney and coworkers
24.2.1 .I. Method Using Automated ( 3 ) used this general method successfully to model the ac-
IUPAC Approach tion of commercial Trichodem reesei cellulase preparations
From the above discussion, it is clear that traditional cellu- on Sigmacell 50, a cotton-linter-derived microparticulate
lase assays are tedious and time-consuming. Multiple en- cellulose preparation (Sigma-Aldrich Co.). Results from
double-reciprocal plots of enzyme activity, (percent conver-
zyme components, an insoluble substrate, and generally
slow reaction rates have plagued cellulase researchers inter- sion)-, versus loading, (FPU/g cellulose)-, enabled
ested in creating cellulase mixtures with increased activities extrapolation to infinite enzyme loading or maximal di-
gestibility.
and/or enhanced biochemical properties. Although the
IUPAC standard measure of cellulase activity, the filter
paper assay (FPA), can be reproduced in most laboratories 24.2.2.2. Method Using a Membrane
with some effort, this method has long been recognized for DiaIysi s Reactor
its complexity and susceptibility to operator error. The au- A new saccharification assay uses a continuous buffer flux
tomated FPA method reported by Decker and coworkers through a membrane reactor to remove the solubilized sac-
(41) is based on a Cyberlabs C400 robotics deck. This au- charification products, thus allowing high extents of sub-
tomated system is equipped with customized incubation, strate conversion without significant inhibitory effects from
reagent storage, and plate-reading capabilities which allow the buildup of either cellobiose or glucose (11). This diafil-
rapid evaluation of cellulases with a maximum throughput tration saccharification assay (DSA) was originally de-
of 84 enzyme samples per day when performing the auto- signed on the assumption that it could be used to obtain di-
mated FPA. Although the assay is not directly comparable rect measurements of the performance of combinations of
to the standard IUPAC FPA, the results are reproducible cellulase and substrate under simulated simultaneous sac-
and function well to compare cellulase mixtures and com- charification and fermentation (SSF) conditions, without
ponents assayed previously. the saccharification results being complicated by factors
that may influence the subsequent fermentation step (5).
24.2.2. General Non-IUPAC Methods Comparisons between actual SSF and DSA experiments
for Cellulase Mixtures using the same ranges of cellulase loadings (ratios of cellu-
Many cellulase enzyme preparations are simply not concen- lase protein to the cellulose content of biomass substrates)
trated enough to cause the required release of 2 mg of glu- have since made it plain, however, that there are special
cose from the 50-mg filter paper sample in 60 min as in the conditions existing in the SSF environment, as opposed to
IUPAC method described above. If these samples cannot be the much simpler DSA environment, that can affect the
concentrated accurately (which is often the case), tradi- saccharification step as well as the fermentation step of
tional FPU cannot be measured. In such cases, however, the the process. Rather than being an exact representation
IUPAC committee recommends that the reducing-sugar re- of the saccharification process that occurs in SSF, DSA data
lease per unit time be accepted as a provisional measure of were useful for comparison with SSF data in efforts to iden-
enzyme activity. This is similar to the pseudo-initial-rate ap- tify the influences of factors other than product inhibition
proach often used in the decade previous to the IUPAC re- on the performance of cellulases in SSE The principle value
port to measure cellulase activity from a wide variety of sub- of DSA, however, lies in its ability to measure cellulase sac-
strates. These substrates may include filter paper (95), charification performance substantially independent of end
Avicel(161), a plant-fiber-derived microcrystalline cellu- product inhibition over a wide range of substrate conver-
lose originated by FMC Corporation and presently supplied sion.
in 50+m particle size by Sigma-Aldrich Co. (St. Louis, An additional characteristic of DSA that must be taken
MO) under the brand name BioChemika, dewaxed cotton into account in data analysis is that the appearance of prod-
(57), or phosphoric-acid-swollen cellulose (136). Methods uct is measured in the effluent stream, rather than in the re-
based on the use of antibiotic disks (135) and turbidity de- action vessel itself. For all practical cases, which involve
velopment (72) also predated the IUPAC study. neither an infinitely small reactor volume nor an infinite
24. Cellulases, Hemicellulases, and Pectinases H 601

rate of flow of buffer solution through the membrane, there estimation (ccmpared to the reducing end group analysis)
is a lag between the generation of product in the reactor of values for (M,,), probably due to insufficient sensitivity of
vessel and the detection of the product in the effluent the light-scattering detector for the low-molecular-weight
stream. This lag is especially pronounced at high enzyme products of CMC degradation.
loadings, which result in rapid generation of soluble-sugar
products, at low buffer flow rates (low dilution rates), or 24.2.2.4. Detection of Cellulase Activity in Gels
with a combination of the two. This effect may be accom- Overall cellulose-depolymerizing activity can also be de-
modated by data-processing approaches that account for the tected by differential dye binding at low concentrations
amount of product held up in the reactor vessel at a given (0.002% [wt/vol]) of finely ground microcrystalline cellulose.
instant. After corrections are made for the lag in detection, According to the procedures of Sharrock (134) or Bartley et
the very detailed and precise progress curves yielded semi- al. (12), Congo red visualization of degraded insoluble cel-
automatically by this assay (Fig. 4) become intrinsic to the lulose can be used to estimate the presence of general cellu-
saccharification being studied and are transportable and lase activity in polyacrylamide gels. Bartley et al. (12) also
scalable to cases in which the reaction occurs in different utilized reduction of tetrazolium dyes by cello-oligomers to
reactor geometries. distinguish between endo- and exocellulase activity. Congo
red staining has also been used in in vitro assays for cellu-
24.2.2.3. Use of Multiangle Laser Light Scattering lase activity (27). Although useful for detecting cellulase
To Estimate MW Reduction of Cellulose components that solubilize microcrystalline cellulose, such
More recently, methods to use solvents to solubilize cellu- as endoglucanases and some exoglucanases, its usefulness is
lose that has been treated with purified enzymes and char- limited with components such as endodependent exoglu-
acterize the products using SEC have been developed canases and P-glucosidases, which do not efficiently solubi-
( 154). The high-performance size exclusion chromatogra- lize cellulose microfibrils. Cellulases may also be detected in
phy-multiangle laser light scattering (HPSEC-MALLS) slab gels using either Western blotting or enzyme-linked im-
technique was used to determine the number-average mo- munosorbent assay, as reported for enzymes from T. reesei
lecular weight (M,,) of CMC that has been treated with cel- (80, 116). Although useful for both quantification and de-
lulase plus solvents, thus allowing the quantification of the tection of cellulase, both methods are dependent on access
number of the bonds broken during degradation of CMC. to appropriate antibodies of known reactivity. One particu-
However, reproducibility of the method was low, especially lar problem noted by the authors is the difficulty of obtain-
for the high-molecular-weight fragments of CMC at the be- ing antibodies against the catalytic domain of T. reesei
ginning of hydrolysis. As hydrolysis proceeded to the more cel7A. With one exception reported in the literature (7),
advanced stages, the HPSEC-MALLS method gave over- anti-T. reesei cel7A antibodies are specific to either the
linker or cellulose-binding module, making detection of
catalytic domain degradation products difficult.

24.2.3. Assays for Specific Cellulase Enzyme Classes


..
I V

Process conversion target I

...........
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _,
_ _ _ _. L_-_m_
I
--- 24.2.3.1. Endoglucanase Analysis

t
.-0
80-
.. .. I
I
I
Endoglucanase activity is determined primarily by two
(widely different) procedures, both of which release reduc-
ing sugars (114, 140, 161) from CMC and reduce fluidity,
r - .r A+, of CMC measured by either capillary or rotary-type vis-
: ..i
I
I cometers (33, 43, 74, 97). In general, however, endoglu-
60- I canases have the widest range of hydrolytic potential of the
8 . . Time to target I specific cellulases, because it is possible to hydrolyze poly-
+
c
a,
240-
a,
i
jI
\: I
I
meric, substituted substrates, such as ostazin brilliant red-
hydroxyethylcellulose (OBR-HEC), azo-dyed cellulose (cellu-
lose azure) and azo-dyed and cross-linked HEC (AZCL-HEC),
as well as the low MW fluorogenic substrates, such as 4-
a . i I
methylumbelliferyl-cellobiose (MUC), 4-methylumbelliferyl-
I
lactopyranoside (MUL), or 4-methylumbelliferyl-cellotriose
20-
j II
(MU-G3) (6,17,165).

4%
- 1 f--
Conversion level of
filter paper assay
I
I
Reducing-Sugar Release from CMC
Reducing-sugar detection methods are traditionally based
on initial rate measurements introduced by Wood and
on I I I I I I I [ I , , i I ,
McCrae in 1977 (161). The reducing sugars are typically
measured by the Somogyi (140) and Nelson (114) proce-
dures, which measure reduction of Cu2+to Cuf in alkaline
solution, or by the DNS assay, whereby reducing sugars re-
FIGURE 4 DSA progress curve of commercial cellulase duce DNS to 3-amino,5-nitrosalicylic acid under alkaline
preparation (Spezyme from Genencor International) at a load- conditions. One unit of enzyme activity is defined as the
ing of 20 FPU/g of cellulose acting on Sigmacell 20 at 38C. amount of enzyme needed to liberate reducing sugars equiv-
After the actual experimental measurements, empirical fits to alent to 5 pg of glucose per h. A variation of the original
the data were done. The expression time-to-target kinetics is IUPAC method for filter paper activity uses CMC as a sub-
considered more descriptive of the approach than is the classi- strate (56). Here, the IUPAC CMC unit of activity is found
cal integrated-rate kinetics. from the dilution of enzyme necessary to produce 0.5 mg of
602 METABOLISM

glucose from 0.5 ml of a 2.0% CMC solution after 30 min specificities can be determined for proposed exoglucanases
at 122F (50C). The recommended substrate was CMC from analytical product evaluation by HPLC (13, 76). This
7L2 (Aqualon Division of Hercules, Inc., Wilmington, DE; is a much more definitive method of distinguishing endo-
degree of substitution [DS] = 0.7; and approximate DP, from exoacting cellulases. In general, exoglucanases such as
400) in 0.05 M sodium citrate buffer, pH 4.8. For compari- CBH I can be expected to hydrolyze the aryl substrates
son purposes, this method should be used to establish the MUC and MUL at the agluconic bond, and not the substi-
specific activity of a purified endoglucanase preparation. A tuted, soluble celluloses such as AZCL-HEC, OBR-HEC,
more commonly used method is that of Mandels et al. (96), and CMC (152). Phosphoric-acid-swollencellulose is also
where CMC 4M6F (Hercules, Inc.; DS, 0.38 to 0.48) is used used as a substrate for exoglucanases; however, some en-
as a substrate and units are expressed as micromoles of re- doglucanases hydrolyze this cellulose form as well (76).
ducing sugar released per minute of incubation time. Analysis of higher oligomeric derivatives has proven to be
Methods proposed by Shoemaker and Brown (136) and complex, especially with endoglucanases and CBH I (153).
HQkanssonet al. (65) also rely on initial-rate reducing-sugar Furthermore, as endoglucanases are highly synergistic with
release from various CMCs. In contrast, Rescigno et al. exoacting glucanases, the presence of endoglucanases sig-
(123) have used ruthidium red to stain CMC and use spec- nificantly complicates efforts to quantify exoglucanase
trophotometric absorption of the soluble fragments for de- activity and can be compensated for only by the separate
termination of endocellulase activity. purification and kinetic characterization of the endoglu-
The measurement of the initial rate of reducing end canase. Attempts persist in trying to link this synergy effect
group formation from the action of endoglucanases on to the cellulose binding module. Some evidence indicates
cellulose can also be accomplished using disodium 2,2'- that the cellulose binding module alone can have a syner-
bicinchoninate (BCA) (73). This reagent was found to be gistic effect on the activity of cellulases, both exo- and en-
the best choice in a recent comparison of methods for the doacting types (87,88,93).
determination of endoglucanase activity (154). The BCA
method was highly sensitive and simple to perform and di- 24.2.3.3. P-D-GI ucosidase Analysis
rectly gave the number of bonds broken, thus allowing for P-D-Glucosidase and cellobiase activities are usually deter-
expression of endoglucanase activity in international units mined according to the method of Wood (159) as aryl-P-
(micromoles of beta-1,4-glucosidic bonds hydrolyzed in 1 glucosidase activity by the hydrolysis of p-nitrophenyl-P-D-
min during the initial period of hydrolysis). glucopyranoside. The concentration of p-nitrophenol was
determined from the absorbance at under alkaline con-
Viscosimetric Assays ditions induced by the addition of 2 M Na2C03.One unit of
Viscometric approaches to cellulase measurement activities activity was defined as the amount of enzyme that catalyzes
are important because other methods measure only the the cleavage of 1.0 pmol of substrate per min at 37C. If
number of glycosidic bonds cleaved in a polymeric sub- necessary, P-D-glucosidases can be distinguished from cel-
strate, without providing any information about location in lobiases by the relative differences in the initial rates for
the substrate of the bonds cleaved. Viscometric methods aryl-P-D-glucosides and cellobiose. Furthermore, the unique
measure a substantial change in a physical property of the and acute sensitivity of P-D-glucosidase to inhibition by glu-
substrate polymer that is a very sensitive function of both conolactone provides a method to assess exoglucanase activ-
the number and the location of the bonds cleaved (33,43, ity in mixed systems of these two enzymes. This approach is
97). For this reason, even though the recommended inter- necessary because P-D-glucosidase also cleaves the aglu-
national units of carboxymethylcellulase are given in terms conic, as well as the holosidic bond of aryl-glucosides (44).
of glycosidic bonds cleaved, it is important to measure both A similar approach is often used to assay other aryl-
bond cleavage (most often by measurement of sugar-reduc- glycosidases (2, 17). This practice has been made possible
ing groups) and the change in solution viscosity as enzy- by the availability of many 0- and p-linked aryl-glycosides
matic hydrolysis proceeds. Vlasenko and coworkers (154) including (but not limited to) P-xylosides, p-mannosides,
found the viscometric method to be simple to perform and P-galactosides, and L-arabinofuranosides.
highly sensitive for the internal bonds cleaved. However,
this method does not account for the hydrolysis of CMC 24.2.4. General HemicellulaseAssays
near the chain end and thus only allows for expression of In 1991, 20 laboratories participated in a collaborative in-
endoglucanase activity in arbitrary viscometric units. vestigation of assays for endo-1,4-beta-xylanase activity
based on production of reducing sugars from polymeric 4-0-
24.2.3.2. ExoglucanaseAnalysis methyl glucuronoxylan (9). The standard deviation of the
The process of detecting and verifying exoglucanases (cel- results reported in this analysis was 108% of the mean when
lobiohydrolases [CBHs] in context of the fungal cellulase the laboratories used their own substrates and methods rou-
systems) has long been controversial. If purified proteins are tinely in each laboratory. Significant reduction in inter-
available, careful comparisons of reducing-sugar yields and laboratory variation was obtained when all the participants
fluidity values from CMC hydrolysis as a function of enzyme used the same substrate for activity determination, each
concentration can be used to judge whether an enzyme is with their own assay procedure. Not surprisingly, the level
more endoglucanase-like or CBH-like. Of course, purified of agreement was further improved (i.e., 17% of the mean)
enzymes can also be subjected to product analysis from the when both the substrate and the assay procedure were stan-
hydrolysis of a series of derivatized; i.e., radiolabeled, chro- dardized. A subset of these laboratories (9) also participated
mophoric, or fluorophoric cello-oligomers for further verifi- in preliminary testing of an assay based on the release of
cation (153, 155). One class of these derivatives, cellobio- dyed fragments from 4-0-methyl glucuronoxylan dyed with
syl fluorides, has been reported to distinguish between CBH Remazol Brilliant Blue (RBB) (16). The relative standard
I and CBH I1 from T. reesei based on cleavage activity on deviations of the results obtained by these laboratories were
the alpha and beta conformations of the cellobiosyl fluo- about 30% for an optimum range of xylanase activity in the
rides (82). Claeyssens et al. have also reported this type of reaction mixture. A major distinction in hemicellulase as-
rigorous analysis for fungal CBH I and CBH I1 (39). Further says arises from the heterogeneous structure of these poly-
24. Cellulases, Hemicellulases, and Pectinases 603

saccharides. Ignoring the actual chemical composition of include azurine (azo-), RBB, and OBR, among others (14,
hemicelluloses from various sources, the enzyme activities 19, 55, 106). For azo-, RBB- and OBR-linked substrates,
required are divided into those that debranch the polymeric clearing zones on petri plate or acrylamide gel agar overlays
backbone and those that depolymerize the polysaccharide indicate active colonies or protein bands (23,98, 138, 143,
chain(s). 157). The cross-linked version (AZCL-polysaccharides-s;
A general approach to detecting glycosyl hydrolase ac- Megazyme, Inc., Bray, Ireland) has also been used to screen
tivity is through the utilization of dyed polysaccharide sub- for activity of various glycosyl hydrolases (139, 165). In the
strates. Dyed polysaccharides have been utilized to deter- case of AZCL substrates, the result of activity is a blue halo
mine activities of cultures on various polysaccharides, both surrounding active colonies or dye release into microtiter
as activity screens and as quantitative measures. These sub- plate wells. The authors have used this technique exten-
strates include both soluble and insoluble forms (dependent sively to screen both environmental samples and recombi-
mainly on the properties of the native polysaccharides) and nant libraries for glycosyl hydrolase activities (Fig. 5).

FIGURE 5 AZCL-polysaccharide hydrolysis in a petri plate (top) and a microtiter plate. The
dark particulates are the AZCL-P-glucan (top) and AZCL-galactan (bottom). Soluble blue dye is
released upon hydrolysis.
604 rn METABOLISM

24.2.4.1 . HemicelIu lose-Debranching Enzymes birchwood glucuronoxylan. The same report also demon-
The debranching of the xylan backbone results in a wide strated that acetylation interferes with glucuronidase activ-
variety of soluble low-molecular-weight compounds as ity and that higher activity was observed on soluble soft-
products. Typically, these products are measured either by wood 4-O~methylglucuronoxylan ( 148). Such synergy has
HPLC or GC. The difficulty in assaying these products is been reported by other workers (35, 45, 148). Para-
not so much in the detection as in obtaining the correct nitrophenyl-aD-glucuronideis used as a substrate for a-
substrate for the enzyme. Most commercial xylan products glucuronidase (132), whereas xylan a-l,2-glucuronosidase
are extracted by alkaline treatment, essentially hydrolyzing is specific for an a(1-+2)-linked glucuronoside. Some glu-
any ester linkages by saponification; i.e., any acetyl-, curonidases, including membrane-bound enzymes, have
coumaroyl-, or feruloyl esters are destroyed. Glycosidic side been found more active on glucuronoxylo-oligomersas sub-
chains, such as arabinose or glucuronic acid, are left intact; strates (29, 35, 46, 111). One recent report has demon-
however, the polymer is typically insoluble. Enzyme studies strated the specific requirement for the 4-0-methyl group
using these substrates must be interpreted with caution, as for efficient binding and positioning of the side chain in the
the native esterified xylan is soluble. Extraction by dimethyl enzyme active site (112).
sulfoxide or steam has been used to prepare native xylan,
in which the esters are still intact and the polymer is solu-
24.2.5. Hemicellulose Depolymerization Enzymes
ble in water (37). As noted for cellulases, hemicellulose-depolymerizing en-
zymes are divided into three classes; the endoacting, exoact-
Arabinofuranosidases ing, and oligomer-hydrolyzing. Although research into the
a-L-Arabinofuranosidase(EC 3.2.1.55) cleaves a-L-arabino- mechanisms of hemicellulose hydrolysis has been steady
furanosides from the arabinoxylan xylose backbone. It has over the years, it has not received the attention given to
also been shown to enhance the release of ferulic and cellulose hydrolysis. Despite this history, a general pattern
coumaric acid from arabinoxylan, presumably through an of degradation is beginning to emerge.
affinity for hydrolyzing phenolic acid-substituted arabinose
side chains (160). Although in the context of hemicellulose X y lanases
Depolymerization of the xylan backbone is mediated by en-
hydrolysis the activity most often reported is hydrolysis of
the a-(1+2)-glycosidic linkage of the arabinofuranoside to doxylanases, and the oligomers are hydrolyzed by p-xylosi-
the xylan backbone, some of these enzymes have been dases. Based primarily on structural differences, the endoxy-
shown to cleave linear and/or branched a-(1+5)-linked lanases are divided into two major categories, the glycosyl
hydrolase families 10 and 11. Assays for endoxylanases fol-
arabinan side chains found in some pectins, resulting in
low the same general patterns as endocellulase assays.
some confusion regarding the specificity of this enzyme class
(99, 105, 129, 164). Although most assays are carried out Viscosity reduction, reducing sugar production, dye-release,
solubilization, zymogram analysis, and colorimetric/fluoro-
with extracted arabinoxylan, p-nitrophenyl-arabinofura-
metric analogues are all utilized in determining the endoxy-
noside has also been used as a substrate (42, 60, 86).
lanase activity (10, 25,49,61,91,92, 108, 125, 142). DNS
Esterases detection of reducing sugars from xylan is the most cited
Acetyl esterase (EC 3.1.1.6) removes acetyl esters from method. Endoxylanases tend to act preferentially on poly-
acetylated xylose and short-chain xylo-oligomers. Its poly- mers of a certain DP. Schizophyllum commune endoxylanase
mer-acting counterpart, acetyl xylan esterase (EC 3.1.1.72), preferred oligomers with seven subunits (24), while other
has a similar activity but is more active on polymeric xylan enzymes exhibited true endo-type activity, with decreasing
(37). In addition to acetate-specific enzyme detection kits, activity for lower DP oligomers (36,40, 117). There are nu-
HPLC, or G C analysis of acetate release from native ex- merous reports of P-xylosidases that cleave short-chain
tracted xylan and chemically acetylated xylan, colorimetric xylo-oligomers to xylose. In these cases, product detection
substrates such as p-nitrophenol acetate and P-napthyl ac- was carried out by direct HPLC analysis or hydrolysis of p-
etate or the fluorometric substrate 4-methylumbelliferylac- nitrophenyl-P-D-xylopyranoside(84, 122, 130, 141).
etate are also used (18,37). The third esterase, ferulic acid p-Clucanases
esterase (EC 3.1.1.73), hydrolyzes the ester bond between PeGlucan is a glucopyranose polymer containing either p-
ferulic acid or coumaric acid and the arabinose side chain of (1+3) or mixed p-( 1+3), p-( 1+4) linkages. The ratio of
arabinoxylan. Assays for this activity are usually carried out ( 1 4 4 ) to (1+3) linkages varies in different species and
using starch-free wheat bran or cellulase-treated gramina- gives specific properties to individual P-glucan polymers.
ceous biomass as a substrate and monitoring ferulic- or Because of the differences in the linkages, different enzymes
coumaric acid released by HPLC or thin-layer chromatog- are required to cleave the two forms of P-glucan (26,68,69,
raphy. When preparing enzyme-treated substrates, care 78, 83, 162). Notably, one report utilized an enzyme-linked
must be taken to employ phenolic acid esterase-free cellu- immunosorbent assay in microtiter plates coated with bio-
lases (37). Other substrates include methyl and ethyl esters tinylated P-glucan to determine activity (156).
of the phenolic acids and finely ground plant biomass (20,
30, 104). Mannanases, Clucomannanases, and
Calactomannanases
Clucuronidases While mannan is characteristically described as a linear p-
In hardwood xylans, xylan a-l,2-glucuronosidase (EC ( 1+4) mannopyranose polymer, galactomannan is com-
3.2.1.131) and a-glucuronidase (EC 3.2.1.139) are involved posed of a polymeric p-( 1+4) mannopyranosyl backbone
in debranching the xylan backbone through removal of a- highly substituted with p-(1+6)-linked galactopyranose
(1+2)-linked glucurono- and 4-0-methyl-glucuronosides residues (32, 107). The degree of substitution varies with
(71, 79, 112, 148). Although relatively little work on these the source. Glucomannan, found mainly in the root of the
enzymes has been carried out, Tenkanen and Siika-aho re- Konjac plant (Amurphophallus konjac), consists of a p-
ported synergy with endoxylanase utilizing deacetylated ( 1+4)-linked mannopyranose and glucopyranosebackbone
24. Cellulases, Hemicellulases, and Pectinases 605

in a ratio of 1.6:l (150). The enzymes involved in depoly- syl hydrolases, which cleave glycosidic bonds by an acid-
merization of the mannans consist of P-mannanase (EC base catalysis mechanism, and polysaccharide lyases, which
3.2.1.78), the endoacting enzyme, and p-mannosidase (EC hydrolyze the glycosidic bond through a p-elimination
3.2.1.25), which produces mannose from the nonreducing mechanism, resulting in a double bond between the C-4
end of the mannose chain (34, 118, 119). Debranching of and C-5 of the new nonreducing end (106). Recent reviews
galactomannan is primarily carried out by a-D-galactosidase by Kashyap et al. (77) and Naidu and Panda (1 13) outline
(EC 3.1.2.22) (1, 54). Tenkanen and coworkers have also the pectinase enzymes in detail. Assay techniques involve
reported an acetyl glucomannan esterase active on the the usual assortment of reducing-sugar production, viscosity
acetyl sidechains in glucomannan (145, 147, 149). There reduction, HPLC analysis, and dye release (8, 15,31,50,53,
are few other data on specific debranching enzymes in- 75, 106, 110). As with other polysaccharide degradation
volved in degradation of glucomannan. Assays for mannan studies, product structure has been determined with nuclear
hydrolysis have been carried out using extracted polysac- magnetic resonance spectroscopy (110).
charides as substrates, colorimetric analogues, and dyed Ruthenium red staining in plates and zymograms has
polysaccharides (21, 48, 52, 103, 127, 151). also been used for assay of pectinase enzymes (53). Because
of its solubility, pectin incorporated into plates can be de-
24.2.6. Pectinases tected by precipitation with ruthenium red (53) or hexade-
In addition to cellulose and the hemicelluloses, pectins are cyl-trimethyl-ammonium bromide, resulting in clear halos
a third class of polysaccharides found in the cell wall matrix of hydrolysis around active colonies (64). Polygalacturonase
of plant cells. Pectin polymers predominantly consist of 1,4- (EC 3.2.1.15) is often assayed by measuring the increase in
linked a-D-galacturonic acid residues, the carboxylic groups reducing groups resulting from the hydrolysis of polygalac-
of which are methoxylated to varying extent. Further infor- turonic acid to oligo-galacturonates. The latter are mea-
mation and good structural diagrams can be found in the re- sured using the Somogyi-Nelson assay (140). In doing this
cent review by Ridley et al. (124). Pectins are major con- procedure, citrate buffer should be avoided in the reaction
stituents of middle lamellae and primary cell walls of higher mixture because it interferes with the assay. Hydrolases such
plants. Pectins fall into three classes differentiated by their as polygalacturonases generally have pH optima at 56.0.
backbone structure and branching patterns (28, 47). Pectinlyase (EC 4.2.2.10) cleaves pectin by p-elimination
Homogalacturonan (xylogalacturonan) is composed of a- and generates products with 4,5-unsaturated residues which
( 1+4)-linked galacturonic acid chains containing xylose are measured by the increase in absorbance at 232 nm.
side chains and makes up the smooth region of pectin Lyases generally have a pH optimum around 8.5. Pectin es-
( 101). Homogalacturonan is methylated through ester-link- terase (EC 3.1.1.11) acts on pectin by liberating methanol
ages to the galacturonic acid residues. Once in place, pectin and pectic acid, and the assay for this enzyme is based on
methyl esterases (EC 3.1.1.1 1) remove these side chains the decrease in pH of the reaction mixture as a result of in-
and allow formation of the gel matrix (158). The rhamno- crease in acid production. For details of these and other as-
galacturonans make up the hairy region of pectin. In says, the reader is referred to the literature cited above.
rhamnogalacturonan I (RG I), the backbone chain is com-
posed of the disaccharide (-+4)-a-D-ga~acturonic acid- This work was funded by the U.S. Department of Energy Office of
a-(1+2)-a-~-rhamnopyranose-(l+). The rhamnose is typ- the Biomass Program.
ically substituted at the C-4 position with a branched chain
of sugars made up of either galactose or arabinose or a com- 24.3. REFERENCES
bination of both and other sugars. The arabinose residues 1. Ademark, P., R. P. de Vries, P. Hagglund, H. Stalbrand,
can be derivatized with ferulic acid. The galacturonic acid and J. Visser. 2001. Cloning and characterization of
residues in the backbone are usually 0 - 2 or 0-3 acetylated Aspergillus niger genes encoding an alpha-galactosidaseand
and 0 - 6 methylated. The structure and substitution pat- a beta-mannosidase involved in galactomannan degrada-
terns of RG I vary widely across plant species. Where the tion. Eur. J. Biochem. 268:2982-2990.
majority of the side chain is composed of arabinose, the side 2. Adney, W. S., J.0. Baker, T. B. Vinzant, S. R. Thomas,
chains are referred to as arabinans. These arabinans are pre- and M. E. Himmel. 1995. Kinetic comparison of beta-
dominantly a-(1+5)-linked arabinofuranosyl residues sub- D-glucosidases of industrial importance. Abstr. Pap. Am.
stituted at the 0 - 2 and/or 0 - 3 positions (28). Side chains Chem. SOC.209:119-BTEC.
composed of galactose residues are referred to as galactans. 3. Adney, W. S., C. I. Ehrman, J. 0. Baker, S. R. Thomas,
When these galactans are further substituted with arabinan and M. E. Himmel. 1994. Cellulase assays-methods from
chains, they are referred to as arabinogalactans (51). In empirical mathematical-models, p. 218-235. In J. N.
contrast to RG I, the structure of rhamnogalacturonan I1 Saddler and M. H. Penner (ed.), Enzymatic Conversion of
(RG 11) is highly conserved across the plant kingdom (100). Biomass for Fuels Production, vol. 566. American Chemical
It comprises 28 glycosyl residues, of which 7 are found in Society, Washington, DC.
the galacturonic acid backbone. The backbone is specifi- 4. Adney, W. S., A. Mohagheghi, S. R,Thomas, and M. E.
cally branched at four points (designated A through D Himmel. 1995. Comparison of protein contents of cellu-
lase preparations in a worldwide round-robin assay, p. 256-
chains) with some unusual sugars such as 2-O-methly-L- 271. In M. E. Himmel, J. 0. Baker, and R. P. Overend
fucose and 3-deoxy-D-manno-2-octu~osonic acid, aceric (ed.), Enzymatic Degradation of Insoluble Carbohydrates, vol.
acid, and apiose (100). The structures of each branch are 618. American Chemical Society, Washington, DC.
known, although the exact point of attachment of branch 5. Agblevor, F. A., A. Murden, and B. R. Hames. 2004.
D is still unclear. Improved method of analysis of biomass sugars using high-
As is apparent from its complex structure, there is a di- performance liquid chromatography. Biotechnol. Lett.
verse enzyme suite required to hydrolyze pectin. As in 26: 1207-1 2 10.
hemicellulase systems, there are depolymerizing and de- 6. Aho, S. 1991. Structural and functional analysis of
branching enzymes, mainly esterases, that act synergistically Trichoderma reesei endoglucanase I expressed in yeast
(53, 106). The depolymerizing enzymes include both glyco- Sacchuromyces cerewisiue. FEBS Lett. 291:45-49.
606 w METABOLISM

7. Aho, S., V. Olkkonen, T. Jalava, M. Paloheimo, R. 23. Braithwaite, K. L., G. W. Black, G. P. Hazlewood, B. R.
Buhler, M. Niku-Paavola, D. Bamford, and M. Korhola. Ali, and H. J. Gilbert. 1995. A non-modular endo-beta-
1991. Monoclonal antibodies against core and cellulose- 1,4-mannanase from Psewlomonas fluorescens subspecies
binding domains of Trichoderma reesei cellobiohydrolases cellulosa. Biochem. J. 305(Pt. 3):1005-1010.
I and I1 and endoglucanase I. Eur. J. Biochem. 200:643- 24. Bray, M. R., and A. J. Clarke. 1992. Action pattern of
649. xylo-oligosaccharidehydrolysis by Schizophyllum commune
8. Antov, M. G., D. M. Pericin, and G. R. Dimic. 2001. xylanase A. Eur. J. Biochem. 204:191-196.
Cultivation of Polyporus squamosus for pectinase produc- 25. Bronnenmeier, K., A. Kern, W. Liebl, and W. L.
tion in aqueous two-phase system containing sugar beet Staudenbauer. 1995. Purification of Thermotoga maritima
extraction waste. J. Biotechnol. 91:83-87. enzymes for the degradation of cellulosic materials. Appl.
9. Bailey, M. J., P. Biely, and K. Poutanen. 1992. Inter- Enuiron. Microbiol. 6 1:1399-1407.
laboratory testing of methods for assay of xylanase activity. 26. Brummell, D. A., C. Catala, C. C. Lashbrook, and A. B.
J. Biotechnol. 23 :257-2 70. Bennett. 1997. A membrane-anchored E-type endo-1,4-
10. Bailey, M. J., and K. Poutanen. 1989. Production of xy- beta-glucanase is localized on golgi and plasma membranes
lanolytic enzymes by strains of Aspergillus. Appl. Microbiol. of higher plants. Proc. Natl. Acud. Sci. USA 944794-
Biotechnol. 30:5-10. 4799.
11. Baker, J. O., T. B. Vinzant, C. I. Ehrman, W. S. Adney, 27. Carder, J. H. 1986. Detection and quantitation of cellu-
and M. E. Himmel. 1997. Use of a new membrane-reactor lase by congo red staining of substrates in a cup-plate dif-
saccharification assay to evaluate the performance of cel- fusion assay. Anal. Biochem. 153:75-79.
lulases under simulated SSF conditions-effect on enzyme 28. Cardoso, S. M., A. M. Silva, and M. A. Coimbra. 2002.
quality of growing Trichodem reesei in the presence of tar- Structural characterisation of the olive pomace pectic
geted lignocellulosic substrate. Appl. Biochem. Biotechnol. polysaccharide arabinan side chains. Carbohydr. Res.
63-65 :585-595. 337:9 17-924.
12. Bartley, T. D., K. Murphy-Holland, and D. E. Eveleigh. 29. Castanares, A., A. J. Hay, A. H. Gordon, S. I. McCrae,
1984. A method for the detection and differentiation of and T. M. Wood. 1995. D-xylan-degradingenzyme system
cellulase components in polyacrylamide gels. Anal. from the fungus Phanerochaete chrysosporium: isolation and
Biochem. 140: 157-16 1. partial characterization of an alpha-(4-O-methyl)-D-
13. Beldman, G., M. Searle-Van Leeuwen, F. Rombouts, and glucuronidase.J. Biotechnol. 43: 183-194.
F. Voragen. 1985. The cellulase of Trichoderma uiride; pu- 30. Castanares, A., and T. M. Wood. 1992. Purification and
rification, characterization and comparison of all de- characterization of a feruloyl/p-coumaroyl esterase from
tectable endoglucanases, exoglucanases and beta-glucosi- solid-state cultures of the aerobic fungus Penicillium
dases. Eur. J. Biochem. 146:301-308. pinophilum. Biochem. SOC.Trans. 20:275S.
14. Berens, S., H. Kaspari, and J. H. Klemme. 1996. 31. Castilho, L. R., T. L. M. Alves, and R. A. Medronho.
Purification and characterization of two different xylanases 1999. Recovery of pectolytic enzymes produced by solid
from the thermophilic actinomycete Microtewaspora flexu- state culture of Aspergillus niger. Process Biochem. 34: 181-
osa SIIX. Antonie Leeuwenhoek 69:235-241. 186.
15. Bhattacharya, S., and N. K. Rastogi. 1998. Rheological 32. Chaubey, M., and V. P. Kapoor. 2001. Structure of a
properties of enzyme-treated mango pulp. J. Food Eng. galactomannan from the seeds of Cassia angustifolia Vahl.
36:249-262. Carbohydr. Res. 332:439444.
16. Biely, P., D. Mislovicova, and R. Toman. 1988. Remazol 33. Chee, K. K. 1990. Kinetic study of random chain scission
brilliant blue xylan-a soluble chromogenic substrate for by viscometry. J. Appl. Polym. Sci. 41:985-994.
xylanases. Methods Enzymol. 160536-541. 34. Chhabra, S., K. N. Parker, D. Lam, W. Callen, M. A.
17. Biely, P., D. Mislovicova, and R. Toman. 1985. Soluble Snead, E. J. Mathur, J. M. Short, and R. M. Kelly. 2001.
chromogenic substrates for the assay of endo-1,+beta- Beta-mannanases from Thermotoga species. Methods
xylanases and endo-1,4-beta-glucanases.Anal. Biochem. Enzymol. 330:224-238.
144: 142-146. 35. Choi, I. D., H. Y. Kim, and Y. J. Choi. 2000. Gene
18. Blum, D. L., X. L. Li, H. Chen, and L. G. Ljungdahl. cloning and characterization of alpha-glucuronidase of
1999. Characterization of an acetyl xylan esterase from the Bacillus stearothermophilus no. 236. Biosci. Biotechnol.
anaerobic fungus Orpinomyces sp. strain PC-2. Appl. Biochem. 64:2530-2537.
Environ. Microbiol. 65:3 990-3 995. 36. Christakopoulos, P., W. Nerinckx, D. Kekos, B. Macris,
19. Bolam, D. N., N. Hughes, R. Virden, J. H. Lakey, G. P. and M. Claeyssens. 1997. The alkaline xylanase 111 from
Hazlewood, B. Henrissat, K. L. Braithwaite, and H. J. Fusarium oxysporum F3 belongs to family F/10. Carbohydr.
Gilbert. 1996. Mannanase A from Pseudomom fluorescens Res. 302:191-195.
ssp. cellulosa is a retaining glycosyl hydrolase in which 37. Christov, L. P., and B. A. Prior. 1993. Esterases of xylan-
E212 and E320 are the putative catalytic residues. degrading microorganisms: production, properties, and sig-
Biochemistry 35:16195-1 6204. nificance. Enzyme Microb. Technol. 15:460-475.
20. Borneman, W. S., L. G. Ljungdahl, R. D. Hartley, and 38. Claeyssens, M., and G. Aerts. 1992. Characterization of
D. E. Akin. 1991. Isolation and characterization of p- cellulolytic activities in commercial Trichoderma reesei
coumaroyl esterase from the anaerobic fungus Neocalli- preparations-an approach using small, chromogenic sub-
mastix strain MC-2. Appl. Environ. Microbiol. 57:2337- strates. Bioresour. Technol. 39: 143-146.
2344. 39. Claeyssens, M., H. Van Tilbeurgh, P. Tomme, T. M.
21. Bourgault, R., and J. D. Bewley. 2002. Gel diffusion as- Wood, and S. I. McRae. 1989. Fungal cellulase systems.
says for endo-beta-mannanase and pectin methylesterase Comparison of the specificities of the cellobiohydrolases
can underestimate enzyme activity due to proteolytic isolated from Penicillium pinophilum and Trichodem reesei.
degradation: a remedy. Anal. Biochem. 300:87-93. Biochem. J. 261:819-825.
22. Boussaid, A., J. Robinson, Y. J. Cai, D. J. Gregg, and 40. Debeire, P., B. Priem, G. Strecker, and M. Vignon. 1990.
J. R. Saddler. 1999. Fermentability of the hemicellulose- Purification and properties of an endo- 1,4-xylanase ex-
derived sugars from steam-exploded softwood (Douglas creted by a hydrolytic thermophilic anaerobe, Clostridium
fir). Biotechnol. Bioeng. 64:284-289. thermolacticum. A proposal for its action mechanism on
24. Cellulases, Hemicellulases, and Pectinases 607

larchwood 4-0-methylglucuronoxylan.Eur. J. Biochem. Linko (ed.), Proceedings of the Symposium on Enzymatic


187573-580. Hydrolysis of Cellulose. VTT, Aulanko, Finland.
41. Decker, S. R., W. S. Adney, E. Jennings, T. B. Vinzant, 58. Ghose, T. K., and V. S. Bisaria. 1987. Measurement of
and M. E. Himmel. 2003. Automated filter paper assay for hemicellulase activities. 1. Xylanases. Pure Appl. Chem.
determination of cellulase activity. Appl. Biochem. 59:1739-1751.
Biotechnol. 105-1 08:689-703. 59. Gilkes, N. R., E. Kwan, D. G. Kilburn, R. C. Miller, and
42. Degrassi, G., A. Vindigni, and V. Venturi. 2003. A ther- R. A. J. Warren. 1997. Attack of carboxymethylcellulose
mostable alpha-arabinofuranosidase from xylanolytic at opposite ends by two cellobiohydrolases from Cell-
Bacillus pumilus: purification and characterisation. J. Bio- ulomoms fimi. J. Biotechnol. 57:83-90.
technol. 101:69-79. 60. Gomes, J., I. I. Gomes, K. Terler, N. Gubala, G.
43. Demeester, J., M. Bracke, and A. Lauwers. 1979. Ditzelmuller, and W. Steiner. 2000. Optimisation of culture
Absolute viscometric method for the determination of en- medium and conditions for alpha-L-arabinofuranosidasepro-
docellulase (Cx) activities based upon light-scattering in- duction by the extreme thermophilic eubacterium
terpretations of gel chromatographic fractionation data. Rhodothemus murinus. Enyyme Microb. Technol. 27:414422.
Ado. Chem. Ser. 181:91-125. 61. Green, F., 111, C. A. Clausen, and T. L. Highley. 1989.
44. Deshpande, M. V., K.-E. Eriksson, and L. G. Pettersson. Adaptation of the Nelson-Somogyi reducing-sugarassay to
1984. An assay for selective determination of exo-1,q-p- a microassay using microtiter plates. Anal. Biochem.
glucanases in a mixture of cellulolytic enzymes. Anal. 182:197-199.
Biochem. 138:481-487. 62. Grohmann, K., R. Torget, and M. E. Himmel. 1985.
45. de Vries, R. P., H. C. Kester, C. H. Poulsen, J. A. Optimization of dilute acid pretreatment of biomass.
Benen, and J. Visser. 2000. Synergy between enzymes Biotechnol. Bioeng. Symp . 15:5 9-80.
from Aspergillus involved in the degradation of plant cell 63. Gustafsson, J., L. Ciovica, and J. Peltonen. 2003. The ul-
wall polysaccharides. Carbohydr. Res. 327:401410. trastructure of spruce kraft pulps studied by atomic force
46. de Vries, R. P., C. H. Poulsen, S. Madrid, and J. Visser. microscopy (AFM) and X-ray photoelectron spectroscopy
1998. aguA, the gene encoding an extracellular alpha- (XPS). Polymer 44:661-670.
glucuronidase from Aspergillus tubingensis, is specifically in- 64. Hadj-Taieb, N., M. Ayadi, S. Trigui, F. Bouabdallah, and
duced on xylose and not on glucuronic acid. J. Bacteriol. A. Gargouri. 2002. Hyperproduction of pectinase activi-
180:243-249. ties by a fully constitutive mutant (CT1) of Penicillium oc-
47. de Vries, R. P., and J. Visser. 2001. Aspergillus enzymes citanis. Enzyme Microb. Technol. 30:662-666.
involved in degradation of plant cell wall polysaccharides. 65. Hikansson, U., L. Fagerstam, G. Pettersson, and L.
Microbiol. Mol. Biol. Reo. 65:497-522. Anderson. 1978. Purification and characterization of a
48. Downie, B., H. W. M. Hilhorst, and J. D. Bewley. 1994. low molecular weight 1,4-beta-glucan glucanohydrolase
New assay for quantifying endo-beta-D-mannanase activ- from the cellulolytic fungus Trichoderma oiride QM 9414.
ity using congo red-dye. Phytochemistry 36:829-835. Biochim. Biophys. Actu 524:385-392.
49. Dupont, C., N. Daigneault, F. Shareck, R. Morosoli, and 66. Helbert, W., J. Sugiyama, M. Ishihara, and S. Yamanaka.
D. Kluepfel. 1996. Purification and characterization of an 1997. Characterization of native crystalline cellulose in
acetyl xylan esterase produced by Streptomyces liuidans. the cell walls of Oomycota. J. Biotechnol. 57:29-37.
Biochem. J. 319(Pt. 3):881-886. 67. Hoshino, E., M. Shiroishi, Y. Amano, M. Nomura, and
50. Fanta, N., A. Quaas, P. Zulueta, and L. M. Perez. 1992. T. Kanda. 1997. Synergistic actions of exo-type cellulases
Release of reducing sugars from citrus seedlings, leaves and in the hydrolysis of cellulose with different crystallinities.
fruits. Effect of treatment with pectinase and cellulase from J. Ferment. Bioeng. 84:300-306.
Alternaria and Trichoderma. Phytochemistry 3 1:3359-3364. 68. Hrmova, M., and G. B. Fincher. 2001. Structure-function
51. Fransen, C. T., S. R. Haseley, M. M. Huisman, H. A. relationships of beta-D-glucan endo- and exohydrolases
Schols, A. G. Voragen, J. P. Kamerling, and J. F. from higher plants. Plant Mol. Biol. 47:73-91.
Vliegenthart. 2000. Studies on the structure of a lithium- 69. Hu, G., and F. H. Rijkenberg. 1998. Subcellular localiza-
treated soybean pectin: characteristics of the fragments tion of beta-l,3-glucanase in Puccinia reconditu f.sp. tritici-
and determination of the carbohydrate substituents of infected wheat leaves. Plantu 204:324-334.
galacturonic acid. Carbohydr. Res. 328539-547. 70. Irwin, D. C., M. Spezio, L. P. Walker, and D. B. Wilson.
52. Fulop, L., and T. Ponyi. 1997. Rapid screening for endo- 1993. Activity studies of 8 purified cellulases-specificity,
beta-l,4-glucanase and endo-beta-1,4-mannanase activi- synergism, and binding domain effects. Biotechnol. Bioeng.
ties and specific measurement using soluble dye-labelled 42: 1002-1013.
substrates. J. Microbiol. Methods 29:15-21. 71. Jeffries, T. W. 1996. Biochemistry and genetics of micro-
53. Gainvors, A., N. Nedjaoum, S. Gognies, M. Muzart, M. bial xylanases. Curr. Opin. Biotechnol. 7:337-342.
Nedjma, and A. Belarbi. 2000. Purification and charac- 72. Johnson, E. A., M. Sakajoh, G. Halliwell, A. Madia, and
terization of acidic endo-polygalacturonase encoded by the A. L. Demain. 1982. Saccharification of complex cellu-
PGL1-1 gene from Saccharomyces cereoisiae. FEMS losic substrates by the cellulase system from Clostridium
Microbiol. Lett. 183:131-135. thermocellum. Appl. Enoiron. Microbiol. 43:1125-1132.
54. Ganter, J. L., J. C. Sabbi, and W. F. Reed. 2001. Real- 73. Johnston, D. B., S. P. Shoemaker, G. M. Smith, and
time monitoring of enzymatic hydrolysis of galactoman- J. R. Whitaker. 1998. Kinetic measurements of cellulase
nans. Biopolymers 59:226-242. activity on insoluble substrates using disodium 2,2' bicin-
55. Ghangas, G. S., Y. J. Hu, and D. B. Wilson. 1989. choninate. J. Food Biochem. 22:301-319.
Cloning of a Thermomonospora fusca xylanase gene and its 74. Joos, P., W. Sierens, and R. Ruyssen. 1969. The deter-
expression in Escherichia coli and Streptomyces liuidans. J. mination of cellulase activity by viscometry. J. Pharm.
Bacteriol. 171:2963-2969. Pharmucol. 21:848-853.
56. Ghose, T. K. 1987. Measurement of cellulase activities. 75. Kapoor, M., Q. K. Beg, B. Bhushan, K. S. Dadhich, and
Pure Appl. Chem. 59:257-268. G. S. Hoondal. 2000. Production and partial purification
57. Ghose, T. K., A. N. Pathak, and V. S. Bisaria. 1975. and characterization of a thermo-alkali stable polygalac-
Kinetic and dynamic studies of Trichoderma oiride cellulase turonase from Bacillus sp. MG-cp-2. Process Biochem.
production, p. 111. In M. Bailey, T.-M. Enari, and M. 36:467-473.
608 METABOLISM

76. Karlsson, J., M. Siika-aho, M. Tenkanen, and E 93. Linder, M., and T. T. Teeri. 1997. The roles and func-
Tjerneld. 2002. Enzymatic properties of the low molecular tion of cellulose-binding domains. J. Biotechnol. 57: 15-
mass endoglucanases Cell2A (EG 111) and Ce145A (EG 28.
V) of Trichoderma reesei. J. Biotechnol. 99:63-78. 94. Mandels, M., G. L. Miller, and R. W. Slater. 1961.
77. Kashyap, D. R., P. K. Vohra, S. Chopra, and R. Tewari. Separation of fungal carbohydrases by starch block zone
2001. Applications of pectinases in the commercial sector: electrophoresis. Arch. Biochem. Biophys. 93:115-121.
a review. Bioresour. Technol. 77:215-227. 95. Mandels, M., R. Andreotti, and C. Roche. 1976. Mea-
78. Keitel, T., K. K. Thomsen, and U. Heinemann. 1993. surement of saccharifying cellulase. Biotechnol. Bioeng.
Crystallization of barley (1-3,1-4)-beta-glucanase,isoen- Symp. 6:21-33.
zyme 11.J. Mol. Biol. 232:1003-1004. 96. Mandels, M., D. Sternberg, and R. E. Andreotti. 1975.
79. Khandke, K. M., P. J. Vithayathil, and S. K. Murthy. Growth and cellulase production by Trichoderma, p. 81. In
1989. Purification and characterization of an alpha-D- M. Bailey, T.-M. Enari, and M. Linko (ed.), Proceedings of
glucuronidase from a thermophilic fungus, Themoascus the Symposium on Enzymatic Hydrolysis of Cellulose. VTT,
aurantiucus. Arch. Biochem. Biophys. 274511-517. Aulanko, Finland.
80. Kolbe, J., and C. P. Kubicek. 1990. Quantification and 97. Manning, K. 1981. Improved viscometric assay for cellu-
identification of the main components of the Trichodema lase methods. J. Biochem. Biotechnol. 5:189-202.
cellulase complex with monoclonal antibodies using an 98. Markovic, O., D. Mislovicovi, P. Biely, and K.
enzyme-linked immunosorbent assay (ELISA). Appl. Heinrichovs. 1992. Chromogenic substrate for endo-
Microbiol. Biotechnol. 34:26-30. polygalacturonase detection in gels. J. Chromatogr. A
81. Kong, F., C. R. Engler, and E. J. Soltes. 1992. Effects of 603:243-246.
cell-wall acetate, xylan backbone, and lignin on enzymatic 99. Matsuo, N., S. Kaneko, A. Kuno, H. Kobayashi, and I.
hydrolysis of aspen wood. Appl. Biochem. Biotechnol. Kusakabe. 2000. Purification, characterization and gene
34/35 :23-35. cloning of two alpha-L-arabinofuranosidasesfrom Strep-
82. Konstantinidis, A. K., I. Marsden, and M. L. Sinnott. tomyces chartreusis GS901. Biochem. J. 346(Pt. 1):9-15.
1993. Hydrolyses of alpha- and beta-cellobiosyl fluorides 100. Mazeau, K., and S. Perez. 1998. The preferred confor-
by cellobiohydrolases of Trichoderma reesei. Biochem. J. mations of the four oligomericfragments of rharnnogalac-
291(Pt. 3):883-888. turonan 11. Carbohydr. Res. 311:203-217.
83. Kotake, T., N. Nakagawa, K. Takeda, and N. Sakurai. 101. McCartney, L., A. P. Ormerod, M. J. Gidley, and J. P.
1997. Purification and characterizationof wall-bound exo- Knox. 2000. Temporal and spatial regulation of pectic
1,3-beta-D-glucanase from barley (Hordeum wulgare L.) (l+g)-beta-D-galactan in cell walls of developing pea
seedlings. Plant Cell Physiol. 38:194-200. cotyledons: implications for mechanical properties. Plant
84. La Grange, D. C., I. S. Pretorius, M. Claeyssens, and J. 22:105-113.
W. H. van Zyl. 2001. Degradation of xylan to D-xylose by 102. McCleary, B. V. 1980. New chromogenic substrates for
recombinant Succhuromyces cereoisiae coexpressing the the assay of alpha-amylase and (1 leads to 4)-beta-D-
Aspergillus niger beta-xylosidase (xlnD) and the glucanase. Carbohydr. Res. 86:97-104.
Trichodem reesei xylanase I1 (xyn2) genes. Appl. Enuiron. 103. McCleary, B. V. 1978. Simple assay procedure for beta-
Microbiol. 6 7 5 5 12-5519. D-mannanase. Carbohydr. Res. 67:213-221.
85. Lee, I., B. R. Evans, and J. Woodward. 2000. The mech- 104. McDermid, K. P., C. W. Forsberg, and C. R.
anism of cellulase action on cotton fibers: evidence from MacKenzie. 1990. Purification and properties of an
atomic force microscopy. Ultramicroscopy 82:213-221. acetylxylan esterase from Fibrobacter succinogenes S85.
86. Lee, R. C., R. A. Burton, M. Hrmova, and G. B. Appl . Enuiron. Microbiol . 56:3 805-3 8 10.
Fincher. 2001. Barley arabinoxylan arabinofuranohydro- 105. McKie, V. A., G. W. Black, S. J. Millward-Sadler, G. P.
lases: purification, characterization and determination of Hazlewood, J. I. Laurie, and H. J. Gilbert. 1997.
primary structures from cDNA clones. Biochem. J. 356: Arabinanase A from Pseudomom fluorescens subsp. cellu-
181-189. losa exhibits both an endo- and an exo- mode of action.
87. Lemos, M. A., J. A. Teixeira, M. R. M. Domingues, M. Biochem. J. 323(Pt. 2):547-555.
Mota, and F. M. Gama. 2003. The enhancement of the 106. McKie, V. A., J. P. Vincken, A. G. Voragen, L. A. van
cellulolytic activity of cellobiohydrolase I and endoglu- den Broek, E. Stimson, and H. J. Gilbert. 2001. A new
canase by the addition of cellulose binding domains de- family of rhamnogalacturonan lyases contains an enzyme
rived from Trichoderma reesei. Enzyme Microb. Technol. that binds to cellulose. Biochem. J. 355:167-177.
32:35-40. 107. Mestechkina, N. M., 0. V. Anulov, N. I. Smirnova,
88. Levy, I., and 0. Shoseyov. 2002. Cellulose-binding do- and V. D. Shcherbukhin. 2000. Composition and struc-
mains: biotechnological applications. Biotechnol. Adw. ture of a galactomannan macromolecule from seeds of
20: 191-2 13. Astragalus lehmannianus Bunge. Appl. Biochem. Microbiol.
89. Li, H., M. Rief, F. Oesterhelt, H. E. Gaub, X. Zhang, 36~502-506.
and J. Shen. 1999. Single-molecule force spectroscopy on 108. Milagres, A. M. F., and R. M. Sales. 2001. Evaluating
polysaccharides by AFM-nanomechanical fingerprint of the basidiomycetes Poria medula-panis and WolfiporiaCOCOS
alpha-( 1,4)-linked polysaccharides.
. . Chem. Phys. Lett. for xylanase production. Enzyme Microb. Technol.
305: 197-201. 28:522-526.
90. Limam, F., S. E. Chaabouni, R. Ghrir, and N. Marzouki. 109. Miller, G. L. 1959. Dinitrosalicylic acid reagent for de-
1995. Two cellobiohydrolases of Penicillium occitunis mu- termination of reducing sugar. Anal. Chem. 3 1426428.
tant Pol 6: purification and properties. Enzyme Microb. 110. Mutter, M., C. M. Renard, G. Beldman, H. A. Schols,
Technol. 17:34&346. and A. G. Voragen. 1998. Mode of action of RG-
91. Lin, J., L. M. Ndlovu, S. Singh, and B. Pillay. 1999. hydrolase and RG-lyase toward rhamnogalacturonan
Purification and biochemical characteristics of beta-D- oligomers: characterization of degradation products using
xylanase from a thermophilic fungus, Themomyces hnugi- RG-rhamnohydrolase and RG-galacturonohydrolase.
nosw-SSBP. Biotechnol. Appl. Biochem. 30:73-79. Carbohydr. Res. 3 11:155-164.
92. Lin, L. L., and J. A. Thomson. 1991. An analysis of the 111. Nagy, T., K. Emami, C. M. Fontes, L. M. Ferreira,
extracellularxylanases and cellulases of Butyriwibriofibrisol- D. R. Humphry, and H. J. Gilbert. 2002. The mem-
wens H17c. FEMS Microbiol. Lett. 68:197-203. brane-bound alpha-glucuronidase from Pseudomonas
24. Cellulases, Hemicellulases, and Pectinases 609

cellulosa hydrolyzes 4-O-methy~-D-g~ucuronoxy~oo~igosac- tion of crystalline cellulose. Carbohydr. Res. 140:lll-


J. Bac-
charides but not 4-0-methyl-D-glucuronoxylan. 120.
teriol. 184:49254929. 129. Sakamoto, T., and T. Sakai. 1995. Analysis of structure
112. Nagy, T., D. Nurizzo, G. J. Davies, P. Biely, J. H. of sugar-beet pectin by enzymatic methods. Phytochemistry
Lakey, D. N. Bolam, and H. J. Gilbert. 2003. The 39:821-823.
alpha-glucuronidase,GlcA67A, of Cellvibrio japonicus uti- 130. Saluzzi, L., H. J. Flint, and C. S. Stewart. 2001.
lizes the carboxylate and methyl groups of aldobiouronic Adaptation of Ruminococcus flavefaciens resulting in in-
acid as important substrate recognition determinants. creased degradation of ryegrass cell walls. FEMS
J. Biol. Chem. 278:20286-20292. Microbiol. Ecol. 36:131-137.
113. Naidu, G. S. N., and T. Panda. 1998. Production of pec- 131. Samejima, M., J. Sugiyama, K. Igarashi, and K.-E. L.
tolytic enzymes-a review. Biopocess Eng. 19:355-361. Eriksson. 1997. Enzymatic hydrolysis of bacterial cellu-
114. Nelson, N. 1944. A photometric adaptation of the lose. Carbohydr. Res. 305:281-288.
Somogyi method for the determination of glucose. 1.Biol. 132. Saraswat, V., and V. S. Bisaria. 1997. Biosynthesisof xy-
Chem . 153:3 75-380. lanolytic and xylan-debranching enzymes in Melano-
115. Nidetzky, B., W. Steiner, M. Hayn, and M. Claeyssens. carpus albomyces IIS 68.1. Ferment. Bioeng. 83:352-357.
1994. Cellulose hydrolysis by the cellulases from 133. Sattler, W., H. Esterbauer, 0. Glatter, and W. Steiner.
Trichodema reesei: a new model for synergistic interac- 1989. The effect of enzyme concentration on the rate of
tion. Biochem. J. 298:705-710. the hydrolysis of cellulose. Biotechnol. Bioeng. Symp.
116. Nieves, R. A., Y. C. Chou, M. E. Himmel, and S. R. 33:1221-1234.
Thomas. 1995.Quantitation of Acidothemus cellulolyticus 134. Sharrock, K. R. 1988. Cellulase assay methods: a review.
E l endoglucanase and Themomonospora fusca E(3) exo- J. Biochem. Biophys. Methods 17:81-106.
glucanase using enzyme-linked-immunosorbent-assay 135. Sheir+Neiss, G., and B. S. Montenecourt. 1984.
(ELISA). Appl. Biochem. Biotechnol. 5 1-2:211-223. Characterization of the secreted cellulases of Trichoderma
117. Nishitani, K., and D. J. Nevins. 1991. Glucuronoxylan reesei wild type and mutants during controlled fermenta-
xylanohydrolase. A unique xylanase with the require- tions. Appl. Microbiol. Biotechnol. 20:46-53.
ment for appendant glucuronosyl units. J. Biol. Chem. 136. Shoemaker, S. P., and J. R. D. Brown. 1978. Char-
266:6539-6543. acterization of endo-1,4-beta-D-glucanasespurified from
118. Parker, K. N., S. Chhabra, D. Lam, M. A. Snead, E. J. Trichodema viride. Biochim. Biophys. Actu 523:147-161.
Mathur, and R. M. Kelly. 2001. beta-Mannosidase from 137. Sieben, A. 1975. Cellulase and other hydrolytic enzyme
Thennotoga species. Methods Enzymol. 330:238-246. assays using an oscillating tube viscometer. Anal.
119. Parker, K. N., S. R. Chhabra, D. Lam, W. Callen, Biochem. 63:2 14-2 19.
G. D. Duffaud, M. A. Snead, J. M. Short, E. J. Mathur, 138. Sipat, A., K. A. Taylor, R. Y. Lo, C. W. Forsberg, and
and R. M. Kelly. 2001. Galactomannanases man2 and P. J. Krell. 1987. Molecular cloning of a xylanase gene
man5 from Themotoga species: growth physiology on from Bacteroides succinogenes and its expression in Esche-
galactomannans, gene sequence analysis, and biochemi- richia coli. Appl. Enuiron. Microbiol. 53477-481.
cal properties of recombinant enzymes. Biotechnol. 139. Skjot, M., S. Kauppinen, L. V. Kofod, C. Fuglsang, M.
Bioeng. 75:322-333. Pauly, H. Dalboge, and L. N. Andersen. 2001.
120. Pere, J., A. Puolakka, P. Nousiainen, and J. Buchert. Functional cloning of an endo-arabinanase from
2001. Action of purified Trichodem reesei cellulases on Aspergillus aculeatus and its heterologous expression in A.
cotton fibers and yam. J. Biotechnol. 89:247-255. oryzae and tobacco. Mol. Genet. Genomics 265:913-921.
121. Ramos, L. P., A. Zandona Filho, F. C. Deschamps, and 140. Somogyi, M. 1952.Notes on sugar determination. J. Biol.
J. N. Saddler. 1999. The effect of Trichoderma cellulases Chem. 195:19-23.
on the fine structure of a bleached softwood kraft pulp. 141. Suzuki, T., E. Kitagawa, F. Sakakibara, K. Ibata, K.
Enzyme Microb. Technol. 24:371-380. Usui, and K. Kawai. 2001. Cloning, expression, and
122. Ratanakhanokchai, K., K. L. Kyu, and M. Tan- characterization of a family 52 beta-xylosidase gene
ticharoen. 1999. Purification and properties of a xylan- (xysB)of a multiple-xylanase-producingbacterium, Aero-
binding endoxylanase from alkaliphilic Bacillus sp. strain m o m caviae ME-1. Biosci. Biotechnol. Biochem. 65:487-
K-1. Appl. Environ. Microbiol. 65:694-697. 494.
123. Rescigno, A., A. C. Rinaldi, N. Curreli, A. Olianas, 142. Taguchi, H., T. Hamasaki, T. Akamatsu, and H.
and E. Sanjust. 1994. A dyed substrate for the assay of Okada. 1996. A simple assay for xylanase using o-nitro-
endo-1,q-beta-glucanases. J. Biochem. Biophys. Methods phenyl-beta-D-xylobioside.Biosci. Biotechnol. Biochem.
28: 123-129. 60:983-985.
124. Ridley, B. L., M. A. ONeill, and D. Mohnen. 2001. 143. Takahashi, R., K. Mizumoto, K. Tajika, and R. Takano.
Pectins: structure, biosynthesis, and oligogalacturonide- 1992. Production of oligosaccharides from hemicellulose
related signaling. Phytochemistry 5 7:929-967. of woody biomass by enzymatic-hydrolysis. 1. A simple
125. Ruiz-Arribas, A., J. M. Fernandez-Abalos, P. Sanchez, method for isolating beta-D-mannanase-producing
A. L. Garda, and R. I. Santamaria. 1995. Over- microorganisms.Mokuzai Gakkaishi 38:1126-1135.
production, purification, and biochemical characteriza- 144. Ten, L. N., W. T. Im, M. K. Kim, M. S. Kang, and
tion of a xylanase (Xysl) from Streptomyces hulstedii JM8. S. T. Lee. 2004. Development of a plate technique for
Appl. Environ. Microbiol. 6 1:2414-2419. screening of polysaccharide-degrading microorganismsby
126. Rutland, M. W., A. Carambassis, G. A. Willing, and using a mixture of insoluble chromogenic substrates.
R. D. Neuman. 1997. Surface force measurements be- J. Microbiol. Methods 56:375-382.
tween cellulose surfaces using scanning probe microscopy. 145. Tenkanen, M. 1998. Action of Trichoderma reesei and
Colloids Surf. A 123-124:369-374. Aspergillus oryzae esterases in the deacetylation of hemi-
127. Sachslehner, A., G. Foidl, N. Foidl, G. Gubitz, and D. celluloses. Biotechnol. Appl. Biochem. 27:19-24.
Haltrich. 2000. Hydrolysis of isolated coffee mannan and 146. Tenkanen, M., and K. Poutanen. 1992. Significance of
coffee extract by mannanases of Sclerotium rolfsii. J. esterases in the degradation of xylans, p. 203-212. In 1. G.
Biotechnol. 80:127-134. Visser, M. A. Beldman, K.-V. Someren, and A. G. J.
128. Sadana, J. C., and R. V. Patil. 1985. Synergism between Voragen (ed.), X y h s and Xylames. Elsevier, New York,
enzymes of Sclerotium rolfsii involved in the solubiliza- NY.
610 METABOLISM

147. Tenkanen, M., J. Puls, M. Ratto, and L. Viikari. 1993. 157. Whitehead, T. R., and R. B. Hespell. 1989. Cloning and
Enzymatic deacetylation of galactoglucomannans. Appl. expression in Escherichia coli of a xylanase gene from
Microbiol. Biotechnol. 39:159-165. Bacteroides ruminicola 23. Appl. Environ. Microbiol.
148. Tenkanen, M., and M. Siika-aho. 2000. An alpha- 55:893-896.
glucuronidase of Schizophyllum commune acting on poly- 158. Willats, W. G., C. Orfila, G. Limberg, H. C. Buchholt,
meric xylan. J. Biotechnol. 78:149-161. G. J. van Alebeek, A. G. Voragen, S. E. Marcus, T. M.
149. Tenkanen, M., J. Thornton, and L. Viikari. 1995. An Christensen, J. D. Mikkelsen, B. S. Murray, and J.P.
acetylglucomannan esterase of Aspergillus oryzm-purifi- Knox. 2001. Modulation of the degree and pattern of
cation, characterization and role in the hydrolysis of 0- methyl-esterification of pectic homogalacturonan in
acetyl-galactoglucomannan. J. Biotechnol. 42: 197-206. plant cell walls. Implications for pectin methyl esterase
150. Teramoto, A., and M. Fuchigami. 2000. Changes in action, matrix properties, and cell adhesion. J. Biol.
temperature, texture, and structure of konnyaku (konjac Chem. 276:19404-19413.
glucomannan gel) during high-pressure-freezing.J. Food 159. Wood, T. M. 1971. The cellulase of Fusarium solani: pu-
Sci. 65:491497. rification and specificity of the beta-( 1,4)-glucanaseand
151. Torto, N., T. Buttler, L. Gorton, G. Markovarga, H. the beta-D-glucosidase components. Biochem. J. 121:
Stalbrand, and F. Tjerneld. 1995. Monitoring of enzy- 353-362.
matic-hydrolysis of ivory nut mannan using online mi- 160. Wood, T. M., and S. I. McCrae. 1996. Arabinoxylan-
crodialysis sampling and anion-exchange chromatogra- degrading enzyme system of the fungus Aspergillus
phy with integrated pulsed electrochemical detection. awamori: purification and properties of an alpha-l-arabi-
Anal. Chim. Acta 313:15-24. nofuranosidase. Appl. Microbiol. Biotechnol. 45 :53 8-545.
152. Tuohy, M. G., D. J. Walsh, P. G. Murray, M. 161. Wood, T. M., and S. I. McCrae. 1977. Cellulase from
Claeyssens, M. M. Cuffe, A. V. Savage, and M. P. Fusarium solani purification and properties of the C-1
Coughlan. 2002. Kinetic parameters and mode of action component. Carbohydr. Res. 57:117-133.
of the cellobiohydrolases produced by Talaromyces emer- 162. Wu, C. T., G. Leubner.Metzger, F. Meins, Jr., and K. J.
sonii. Biochim. Biophys. Actu/Prot. Struct. Mol. Enzymol. Bradford. 2001. Class I beta-l,3-glucanase and chitinase
1596:366-380. are expressed in the micropylar endosperm of tomato
153. van Tilbeurgh, H., M. Claeyssens, and C. K. seeds prior to radicle emergence. Plant Physiol.
DeBruyne. 1982. The use of 4-methylumbelliferyl and 126: 1299-13 13.
other chromophoric glycosides in the study of cellulolytic 163. Wyman, C. E., S. R. Decker, M. E. Himmel, J.W.
enzymes. FEBS Lett. 149:152-156. Brady, C. E. Skopec, and L. Viikari. 2005. Hydrolysis of
154. Vlasenko, E. Y., A. I. Ryan, C. F. Shoemaker, and S. P. cellulose and hemicellulose, p. 995-1034. In S. Dimitriu
Shoemaker. 1998. The use of capillary viscometry, reduc- (ed.), Polysucchurides: Structural Diversity and Functional
ing end-group analysis, and size exclusion chromatogra- Versatility. Marcel Dekker, New York, NY.
phy combined with multi-angle laser light scattering to 164. Yanai, T., and M. Sato. 2000. Purification and charac-
characterize endo-1 ,4-beta-D-glucanases on carboxy- terization of a novel alpha-L-arabinofuranosidase from
methylcellulose: a comparative evaluation of the three Pichia capsulata X91. Biosci. Biotechnol. Biochem.
methods. Enzyme Microb. Technol. 23:350-359. 64:1181-1188.
155. Vrsanska, M., and P. Biely. 1992. The cellobiohydrolase 165. Zantinge, J. L., H. C. Huang, and K. J. Cheng. 2002.
I from Trichodem reesei QM 9414: action on cello- Microplate diffusion assay for screening of beta-
oligosaccharides. Carbohydr. Res. 227:19-27. glucanase-producing microorganisms. BioTechniques 33:
156. Wang, G., R. R. Marquardt, H. Xiao, and Z. Zhang. 798, 800, 802.
1999. Development of a 96-well enzyme-linked solid- 166. Zauscher, S., and D. J. Klingenberg. 2000. Normal
phase assay for beta-glucanase and xylanase. J. @c. forces between cellulose surfaces measured with colloidal
Food Chem. 47:1262-1267. probe microscopy. J. Colloid Interface Sci. 229:497-5 10.
25
Lignin and Lignin-Modifying Enzymes
CARLOS G. DOSORETZ AND C. A. REDDY

25.1. INTRODUCTION ......................................... 61 1


25.2. FUNGAL LIGNIN DEGRADATION ........................... 61 1
25.2.1. Primary Degraders of Lignin ............................. 61 1
.2. White Rot Fungi ..................................... 612
.3. Initial Studies on Lignin Degradation by White Rot Fungi ....... 612
25.3. LIGNIN-MODIFYING ENZYMES ............................. 612
25.3.1. Lignin Peroxidase (Lip) ................................ 613
25.3.1.1. Catalytic Cycle ............................... 613
.2. Role of Redox Mediators........................ 614
.3. Lip Activity Measurement....................... 614
25.3.2. Manganese Peroxidase (MnP) ............................ 614
25.3.2.1. Catalytic Cycle ............................... 614
.................
.2. Role of MnP in Lignin Degradation 615
.3. MnP Activity Measurement ...................... 615
25.3.3. LAC .............................................. 615
25.3.3.1. Role of LAC in Lignin Degradation ................ 615
.2. LAC Activity Measurement ...................... 616
25.3.4. Versatile Peroxidase ................................... 616
.5. Role of Reactive Oxygen Species in Lignin Degradation......... 616
25.4. REFERENCES ............................................ 6 17

25.1. INTRODUCTION gal enzymes, designated lignin-modifying enzymes (LMEs),


Lignin, next to cellulose, is the most abundant renewable consist of lignin peroxidases, manganese peroxidases, and
organic polymer in the biosphere. Lignin occurs in plant laccases, and these play a key role in lignin biotransforma-
cell walls in intimate association with the carbohydrate tion. There is intense worldwide interest in studying LMEs
polymers cellulose and hemicellulose, which are, respec- because of their potential biotechnological applications.
tively, the first and second most abundant polysaccharide These include transformation of lignocellulosic biomass
polymers in nature. These three polymers are the major into feeds, fuels, and chemicals; for biopulping and
constituents of plant cell walls. Lignin, which is struc- biobleaching in pulp and paper production; for decolorizing
turally complex and recalcitrant to microbial degradation, and detoxifying Kraft bleach plant effluents; and for de-
and hemicellulose form an amorphous matrix in which lie grading toxic environmental pollutants such as dioxins,
the cellulose fibrils (23, 49, 51). The sheath-like covering polychlorinated biphenyls, dye effluents, and polyaromatic
of lignin-hemicellulose makes the cellulose component rel- hydrocarbons (4, 11, 20,32,51,64,65,82, 107). A briefre-
atively less accessible to microbial degradation. Thus, view of lignin degradation and the basic methodologies as-
lignin biodegradation plays a central role in global carbon sociated with the study of LMEs are presented here. For
cycling, which is of considerable ecological interest. greater details, the reader is referred to selected books (20,
Interest in lignin biodegradation also lies in the fact that 29, 51, 104) and numerous reviews (3,4, 9, 11, 13, 14,47,
lignin removal enhances the efficiency of conversion l i g 49,50,52,58,59,62,69, 77) that have appeared regarding
nocellulosic biomass as a feed for ruminants, as a source of the physiology and biochemistry of lignin degradation and
feedstock chemicals for industry, and for production of fuels LME. For information on cellulases and hemicellulases, the
such as ethanol and that lignin is itself a potential source of reader is referred to chapter 24 of this volume.
useful industrial chemical feedstock (11, 60, 64, 65, 73,
107). Moreover, there is increasing worldwide interest in 25.2. FUNGAL LlGNlN DEGRADATION
the use of ligninolytic fungi for bioremediation purposes
and for biopulping applications (11, 12,32, 53, 78, 87). 25.2.1. Primary Degraders of Lignin
Free-radical condensation of lignin precursors, Basidiomycete fungi that cause white rot decay of wood are
coniferyl, synapyl, and p-coumaryl alcohols by plant cell the most efficient lignin degraders in nature and show ex-
wall peroxidases results in the formation of a heteroge- tensive and complete mineralization of lignin to carbon
neous, amorphous, optically inactive, and highly branched dioxide and water (13, 20, 29,51,59). Two other groups of
aromatic polymer with many different types of inter- fungi, designated soft rot fungi and brown rot fungi, are also
monomer linkages. It is these unique structural features of capable of causing wood decay (8, 35, 29). Brown rot ba-
lignin that impose unusual restrictions on its degradation sidiomycetes extensively degrade cellulose and hemicellu-
by microbes (11, 13, 20, 51, 59, 60). Three families of fun- lose in plant cell walls but are limited in their ability to

61 1
612 METABOLISM

mineralize lignin (8, 10, 20, 29, 51). Demethoxylation is and for isolating lignin-peroxidase-negative and nitrogen-
the most obvious consequence of attack on lignin by these deregulated mutants (see references 11 and 104); however,
fungi. Soft rot fungi such as Chaetomium, Ceratocystis, and poly R dye decolorization by a fungus does not necessarily
Phialophora degrade cellulose and hemicellulose but cause correlate in all cases with its ability to degrade 14C-DHP
only limited modification of lignin. Some bacteria such as lignin to l4Co2.A simple screening procedure that distin-
streptomyces have been shown to partially depolymerize guishes between fungi that cause decay by selectively remov-
lignin to yield low-molecular-weight products but are not ing lignin and those that degrade both cellulose and lignin si-
believed to be significant in causing mineralization of lignin multaneously has been developed (91). This involves
to C02 (20, 29, 51). staining of lignin with Astra-blue, which stains cellulose blue
only in the absence of lignin. Safranin, on the other hand,
25.2.2. White Rot Fungi - stains lignin regardless of the presence of cellulose.
Although white rot fungi are capable of extensive mineral- Dimeric model compounds that represent the principal
ization of lignin to C02 and H 2 0 , lignin is not a significant substructures of lignin have been used widely to character-
source of cell carbon or energy for these organisms (13, 51, ize the ligninolytic systems of white rot fungi (20, 51).
59). It is widely believed that the ability to degrade lignin en- Dimeric models also played a large role in revealing the type
ables white rot fungi to gain access in wood to cellulose and of lignin substructures that are degraded by fungal lignin
hemicellulose, which serve as the main carbon and energy peroxidase (Lip) (42, 49-51). For example, oxidation of
sources for these fungi. White rot fungi cause more extensive p-0-4 linkages (which represent 50 to 60% of the bonds in
degradation of hardwood from deciduous trees (angiosperms) lignin molecule) by Lip indicated that these linkages are
than of softwood trees such as conifers. In a survey of a total cleaved via the cation radical intermediates formed by one-
of 65 central European wood-decaying basidiomycetes, 34 electron oxidation of the aromatic ring of the substrate.
were reported to attack angiosperms exclusively and 27 at- Thus, Lip does cleave the predominant linkages in lignin
tacked both hardwood and coniferous wood, while only four (49, 50,58) . Dimeric models have also been used to detect
species exclusively attacked coniferous wood, (8, 9, 86). Lip activity in situ in fungus-colonized wood, where extrac-
White rot fungi typically colonize the cell lumen and cause tion and conventional assay of the enzyme are technically
cell wall erosion. Degradation is usually localized to cells col- difficult (90). Degradation of a (P-0-4)-(5-5')-type lignin
onized by fungal hyphae. Progressive erosion of the cell wall trimer compound, arylglycerol-P-(dehydrodivanillyl alco-
occurs when components are degraded simultaneously during hol) ether (substrate I) by Lip showed formation of
nonselective delignification and eroded zones coalesce as the Ccu-Cp cleavage, p-0-4 bond cleavage, and opening of p-
decay progresses, forming large voids filled with mycelium. etherified aromatic ring (B-ring) (reviewed in reference
During selective delignification, a diffuse attack of lignin oc- 50). The results also showed that the B-ring of substrate (I),
curs and a white pocket or white-mottled type of rot results which is more difficult to degrade than arylglycerol-p-
(8, 10). Electron microscopic studies have revealed that guaiacyl and +-( 2,6-dimethoxyphenyl) ethers, was oxi-
lignin is degraded at some distance from the hyphae and is re- dized by Lip (50). The disadvantage in the use of dimeric
moved progressively from the lumen towards the middle lignin model compounds, however, is the fact that, unlike
lamella, which is also degraded (8,9). the lignin polymer, they can be taken up and metabolized
intracellularly by microorganisms, which can make it diffi-
25.2.3. Initial Studies on Lignin Degradation cult to determine whether the degradation products ob-
by White Rot Fungi - served really reflect actual ligninolytic activity. Therefore,
The main mechanism of lignin degradation by white rot ideally, lignin model compounds should be sufficiently
fungi involves one-electron oxidation reactions catalyzed by macromolecular but at the same time facilitate efficient
extracellular oxidases and peroxidases (38, 50, 52, 58, 77). product analysis. To this end, dimeric lignin compounds at-
Lignin-degrading ability is commonly studied by growing the tached to a polymer backbone such as polystyrene and poly-
organism in media containing relatively low levels of nitro- ethylene glycol have been used (57).
gen (2.4 mM), because higher levels of nitrogen (24 mM) re-
sult in cessation of lignin degradation (13,59,77).Similarly,
low levels of carbon promote lignin degradation, while 25.3. LICNIN-MODIFYING ENZYMES
higher levels inhibit lignin degradation. Lignin degradation The three major families of LMEs produced by white rot
in such media is measured by determining the amount of fungi that are involved in the oxidative degradation of
I4CO2produced from I4C-labeled lignin preparations, such lignin are Lip (96), manganese-dependent peroxidases or
as ''C-r~ng-labeled dehydrogenative polymer (DHP) of lig- manganese peroxidases (MnP) (36), and laccases (LAC)
nin (20,29). To date, measurement of 14C02evolution from (4) (Table 1). Both Lip and MnP are heme-containing en-
14C-DHP lignin is reported to be the most reliable method zymes that require hydrogen peroxide (H202) for activity.
for testing ligninolytic activity of a given organism. In addition to Lip and MnP, a versatile peroxidase (VP),
Therefore, this method has been extensively used for deter- which shares some of the properties of both Lip and MnP,
mining ligninolytic activity of many white rot basidiomycete has also been described (16, 76, 98). LACs are a group of
fungi as well as for elucidating the role of different fungal en- blue multicopper oxidases that couple substrate oxidation
zymes and other constituents in lignin degradation (20, 29). to four electron reduction of molecular oxygen to water.
Other methods such as nuclear magnetic resonance spec- LACs appear to be present in most white rot fungi studied
troscopy have also been used to study the degradation of to date (4, 62, 70). Two other groups of enzymes are indi-
polymeric lignin (33), but these methods are not easily rectly involved in lignin degradation. These include (i) en-
amenable for detailed physiological and biochemical studies zymes involved in the reduction of aromatic radicals pre-
on white rot fungi and their enzymes. Decolorization of the venting repolymerization such as cellobiose dehydrogenase,
polymeric dye poly R has also been used by a number of cel1obiose:quinone oxidoreductase, and pyranose 2-oxidase;
investigators as a quick screen for ligninolytic organisms and (ii) enzymes involved in the generation of hydrogen
25. Lignin and Lignin-Modifying Enzymes 613

TABLE 1 Key features of LMEs


Enzyme Electron acceptorlmetal Metal cofactor Mediator Bonds cleaved
Lip H202/Fe None Veratryl alcohol, veratrole, Ca-Cp, p-0-4; aryl-Ca bonds,
dimethoxycinnamic acid, P-ring oxidation, demethoxylation;
1,2-dimethoxybenzene nonphenolic moieties in the
presence of mediators (54)
Ca-Cp cleavage; nonphenolic
moieties in the presence of
mediators (48, 54)
VP H202/Fe MnZf Veratryl alcohol Presumably similar to MnP (69)
LAC Oz/CU None 3-Hydroxyanthranillic Ca-Cp;nonphenolic moieties in
acid, ABTS, the presence of mediators (54)
hydroxybenzotriazole

peroxide such as glyoxal oxidase, glucose oxidase, and aryl fectively extending the substrate range. Reactions catalyzed
alcohol oxidase (1, 11, 22, 29, 58, 77, 98). by Lip include benzyl alcohol oxidations, side chain cleav-
ages, ring-opening reactions, demethoxylations, and oxida-
25.3.1. Lignin Peroxidase (Lip) tive dechlorinations (46, 50, 68, 81).
Lip (EC 1.11.1.14) of Phanerochaete chrysosporium, the most
extensively studied ligninolytic fungus, is secreted as a series 25.3.1 . I . Catalytic Cycle
of glycosylated isoenzymes with PIS ranging from 3.2 to 4.0 T h e catalytic cycle of Lip is similar to that of other perox-
and molecular masses ranging from 38 to 43 kDa; each idases (14, 50, 59, 79). As shown in Fig. 1, reaction of na-
isozyme contains 1 mol of heme per mol of protein (14, 59, tive ferric enzyme (Fe-Lip; Fe3+-P) and H202 yields LiP-
77, 83, 105). U p to 15 different isoenzymes have been re- compound I (LiPI), a complex of high valent 0x0-iron and
ported in cell extracts of P. chrysosporium which are struc- porphyrin cation radical (Fe4+=O-P'+). One-electron ox-
turally similar but show some variation in stability, quantity, idation of a phenolic reducing substrate (SH) by Lip1 yields
and catalytic properties (14, 37, 74, 77). Lip possesses a a phenoxy radical (So) and the one-electron-oxidized en-
higher redox potential and a lower p H optimum than that of zyme intermediate, Lip-compound I1 (LiPII; Fe4+=O-P).
most other isolated peroxidases or oxidases (15, 42, 58). LiP, A single one-electron oxidation of a second substrate mol-
similar to other peroxidases, is capable of oxidizing most phe- ecule returns the enzyme to Fe-LiP completing the cat-
nolic compounds through the generation of phenoxy radi- alytic cycle. Lip is uniquely different from other peroxi-
cals; however, due to its exceptionally high redox potential dases in that it exhibits a n unusually high reactivity
and low pH optimum, it is also able to oxidize nonphenolic between LiPII and H 2 0 2(14, 102). In the absence of suit-
aromatic substrates, typically not oxidized by other peroxi- able reducing substrate or at high HzOz concentrations,
dases, including the nonphenolic phenylpropanoid units of LiPII is further oxidized by H z 0 2 to Lip-compound I11
lignin (22,34,45-47,50,59). Stable cation centered radicals (LiPIII; F e 3 + = 0 i - - P ) , a species with limited catalytic
formed during the oxidation of nonphenolic aromatic nuclei activity. LiPIII is inactivated rapidly in the presence of ex-
may serve as redox mediators for Lip-catalyzed oxidations, ef- cess H202.

LIP LIP
(Fe3+-P) (Fe3+-P)

LiPII arc\(,e Lip1 LiPII dTFeP


!I
(Fe4+=O-P) 4+-0-p'+) (Fe4+=O-P) 4+-0-P.+)
VAD VA

x
S- SH

S. SH

FIGURE 1 Catalytic cycle of lignin peroxidase (Lip) in the presence (left panel) and absence
(right panel) of a mediator, as exemplified by the veratryl alcohol in this figure. Symbols: Lip, na-
tive lignin peroxidase; Lip I, compound I; Lip 11, compound 11; VA, veratryl alcohol; VAD, vera-
traldehyde; SH, reducing substrate; S.,substrate cation radical.
614 w METABOLISM

25.3.1.2. Role of Redox Mediators This assay mixture consists of VA, 2mM; H202,0.4 mM;
LiPIII (see above) has been shown to readily return to the d-tartaric acid, 50 mM; and the reaction is started by adding
native ferric state in the presence of H202 and veratryl al- a Lip preparation. Optimal conditions for Lip activity are
cohol (3,4-dimethoxybenzyl alcohol) (VA) (6, 14, 49). pH 2.5 and 30C. Care should be taken to minimize prein-
Ligninolytic cultures of P. chrysosporium normally produce cubation of Lip preparation with buffer or with H 2 0 2 (in
VA, one of the physiological roles of which is believed to be the absence of VA) because the enzyme is likely to get in-
to protect LiP from H202-dependent inactivation by re- activated and/or show lower activity otherwise.
verting LiPIII to the native state. This is of particular sig- The oxidation of azure B, involving absorbance mea-
nificance since during oxidation of certain chemicals such surements in the visible range, was suggested as an altema-
as phenols, LiPIII has been shown to accumulate, indicating tive method for measuring the activity of Lip. This method
that either they are poor substrates for LiPII or they lack the is claimed to be less susceptible to interferences due to ab-
ability to revert LiPIII to the native state (14, 17, 19, 58). sorbance of organic material in the near UV range when
Thus, oxidation of such chemicals is inefficient at high measuring activity in crude extracellular culture fluid (2);
H202concentrations. The catalytic cycle of LiP in the pres- however, this method has not gained much popularity.
ence of VA as mediator is shown in Fig. 1. VA has also been
shown to act as a charge-transfer mediator in Lip-catalyzed 25.3.2. Manganese Peroxidase (MnP)
reactions (41). During the catalytic cycle of Lip, VA is oxi- MnP (EC 1.11.1.13) is an extracellular enzyme that is pro-
dized to VA cation radical (VA+'), which in the presence of duced as a part of the lignin-degrading enzyme system in
a suitable reducing substrate is reduced back to VA and some white rot fungi. MnP exists as a series of glycosylated
then becomes ready for another LiP-catalyzed charge- isozymes with PIS ranging from 4.2 to 4.9 and molecular
transfer reaction. masses ranging from 45 to 47 kDa. Similar to Lip, each
Low-molecular-weight redox mediators such as VA are isozyme contains 1 mol of iron per mol of protein (22, 39,
believed to be important for lignin degradation, since elec- 48, 51, 52, 58, 75). To date, five major isozymes have been
tron microscopic studies have indicated that enzymes as detected in P. chrysosporium. MnP catalyzes the following
large as peroxidases and LAC cannot have direct contact reaction:
with lignin, since they appear to be too large for the pene-
tration of the cell wall or the middle lamella (15). Pine
2Mn(II) + H202 + 2H+ + ZMn(II1) + 2 H 2 0
decayed by wood-rotting fungi was infiltrated with a con- The product of MnP reaction is Mn(III), which nonspecif-
centrated culture filtrate of the white rot fungus P. ically oxidizes a variety of organic compounds including the
chrysosporium and labeled for Lip by postembedding immu- key substructures in lignin.
noelectron microscopy. This method demonstrated that en-
zymatic attack on pine by LiP and presumably also other en- 25.3.2.1. Catalytic Cycle
zymes of the same size is restricted to the surface of the wood MnP, similar to LiP and several other peroxidases, has a cat-
cell wall (92). alytic cycle involving a two-electron oxidation of the heme
Measurements of the stability of VAf' have led to the sug- by H202, followed by two subsequent one-electron reduc-
gestion that it would be unable to act as a diffusible oxidant tions to the native ferric enzyme. The primary reducing sub-
without some form of stabilization (17, 22, 50). It was sug- strate in the MnP catalytic cycle is MnZ+,which efficiently
gested that a Lip-VA+' complex acts as the redox partner, reduces MnP I and MnP I1 (Fig. 3) generating Mn3+,which
with the cation radical being stabilized by a protein microen- then serves to oxidize phenols to phenoxy radicals (46-48).
vironment of acidic character (see references 22 and 50). Just as cation radicals of aromatic substrates (such as that of
Nonphenolic aromatic compounds other than VA such VA) maintain the active form of Lip by oxidatively con-
as 3,4-dimethoxycinnamic acid, 1,2-dimethoxybenzene, verting compound 111 to the native enzyme and prevent
and 2-chloro- 1,4-dimethoxybenzene have also been shown HzOz-dependent inactivation ( 6 ) ,Mn3" has similarly been
to be capable of mediating oxidation (94, 100). The medi- shown to convert MnP I11 to native enzyme (50).
ation phenomenon appears to be driven by the difference in Additionally, Mn2+ also reactivates compound I11 (al-
the oxidation potential (OP) and site-binding affinity of though this reaction is slower) and this reaction is also
the mediators (possessing higher OP values and higher
affinity) and the target substrates (possessing lower OP val-

y \s..
ues and lower affinity) (101). MnP
25.3.1.3. Lip Activity Measurement (Fe3+-P)
Lip activity is assayed by measuring the increase in ab-
sorbance at 3 10 nm resulting from the oxidation of VA to
veratraldehyde (2,3-dimethoxybenzaldehyde)in the pres-
ence of HzO2 (14, 96, 104), as shown in Fig. 2. SH

+ HZOZ - ($,
Lignin
peroxidase
+ 2HZO
(Fe4+=O-P)
Mnf3 Mn+2
MnPI
MnPrr 7 F ( ~ e ? + = o - p . + )

6Me
OMe
6Me
OMe
x
s* SH

FIGURE 2 Oxidation of VA to veratraldehyde by LIP in the FIGURE 3 Catalytic cycle of manganese-dependent peroxi-
presence of HZ02. dase (MnP). See the text for details.
25. Lignin and Lignin-Modifying Enzymes 615

known to prevent compound I11 accumulation when excess This assay works best with purified enzyme because con-
Mn2+ is present. taminating metals such as copper and iron in crude enzyme
preparations interfere with the activity.
25.3.2.2. Role of MnP in Lignin Degradation Paszczynski et al. (75) described a slightly different MnP
In many fungi, MnP is thought to play a crucial role in the assay procedure in that the reaction mixture contained (in
primary attack on lignin, because it generates Mn3+, a a total volume of 1 ml) 100 mM sodium tartrate (pH 5.0)
strong diffusible oxidant that is able to penetrate the small and 100 p M each MnS04 and H 2 0 z .In this procedure, the
"molecular pores" between cellulose microfibrils, which reaction is initiated by the addition of H202 (rather than
precludes the action of Lip because of steric hindrances the enzyme preparation) and increase in absorbance is mea-
(30). Organic acids such as oxalate, fumarate, and malate sured at 238 nm during the first 5 to 30 s of reaction.
(52, 67, 84), which are produced by cultures of white rot Sodium malonate buffer (pH 4.5) can be used instead of tar-
fungi, chelate Mn3+. These stable complexes are believed trate, and Mn(I1) conversion to Mn(II1) is determined by
to be responsible for the oxidation of the substrate. measuring the change in absorbance at 270 nm. Even
Although MnP does not oxidize nonphenolic lignin struc- though the principle is the same, different investigators
tures during normal turnover, these structures have been make some minor changes (to suit their convenience) to
shown to be slowly co-oxidized when MnP peroxidatively the basic procedure.
oxidizes unsaturated fatty acids (54, 55). Bao et al. (5) de-
scribed the oxidation of a nonphenolic lignin model by a Assay with ABTS as the Substrate
lipid peroxidation system that consisted of P. chrysospon'um The reaction mixture (in a total volume of 1 ml) consists of
MnP, MnZ+,and unsaturated fatty acid esters. Substrate ox- 50 mM each of sodium succinate and sodium lactate buffer
idation occurred via benzylic hydrogen abstraction, and it (pH 4.5), 100 pM MnS04, ABTS (40 pg), and 50 pm
was suggested that this process might enable the white rot H202.The assay is done at room temperature. The reaction
fungi to accomplish the initial delignification of wood. The is initiated by adding the enzyme preparation. The initial
importance of MnZ+and lipid peroxidation in depolymer- rate of ABTS oxidation is determined spectrophotometri-
ization and mineralization of 14C-labeled, polyethylene cally by measuring the increase in absorbance at 415 nm.
glycol-linked, p-0-4 lignin model compound by Ceripor-
iopsis subuemispora has been demonstrated in wood block Other Substrates for MnP Assays
cultures as well as in defined medium (54). Thus, lipid per- Oxidative coupling of 3-methyl-2-benzothiazolinonehy-
oxidation has been suggested to be the mechanism involved drazone and 3 4dimethylamino) benzoic acid in the pres-
in the oxidation of the nonphenolic lignin structures by ence of H20zand Mn2+ was also reported to be specific for
white rot fungi that do not produce Lip (54, 72). assaying MnP activity (18). This reaction gives a deep pur-
ple-blue color with a broad absorption band that peaks at
25.3.2.3. MnP Activity Measurement 590 nm. The extinction coefficient is high (53,000 M-'
MnP activity assay is based on the oxidation of a substrate cm-l), and therefore, relatively low MnP activities can be
(S) such as ABTS [2,2'-azinobis(3-ethylbenzothiazoline- detected. When DMP is used as the substrate, it is oxidized
6-sulfonic acid)] or vanillylacetone [4-(4-hydroxy-3- to the dimer 3,3' ,5'5-tetramethoxy-p,p'diphenylquinone (E
= 49,600 M-' . cm-'). The oxidation of NADH in the
methoxyphenyl)-3-buten-Z~one] in the presence of H202
and Mn2+,according to the scheme shown in Fig. 4. The presence of H z 0 2and Mn2+ is also employed by some for
use of a buffer having chelating activity such as citrate, lac- determining the MnP activity.
tate, succinate, tartrate, or malonate has been reported to
improve the reaction (36, 75). Commonly used substrates 25.3.3. LAC
for MnP assay are Mn(II), ABTS, 2,6-dimethoxyphenol LACs (EC 1.10.3.2) are blue multicopper oxidases that ox-
(DMP), vanillylacetone, phenol red, syringaldazine, and idize polyphenols, methoxy-substituted phenols, aromatic
several others (38,39,44, 48, 52). Two commonly used as- diamines, and a range of other aromatic compounds but do
says are described here. not oxidize tyrosine (which is a substrate for tyrosinase) (4,
62). Laccases are found in all the three domains of life:
Assay with Mn(ll) as the Substrate Eukarya, Prokarya, and Archuea. Biochemically, LACs are
This assay measures oxidation of Mn(I1) to Mn(II1). The blue multicopper oxidases that couple four-electron reduc-
reaction mixture (in a total volume of 1 ml) contains 50 tion of molecular oxygen to water concomitant with the ox-
p M MnS04, 50 p M H202, and 50 mM sodium lactate idation of an aromatic substrate (15, 62, 89, 95). LACs are
buffer (pH 4.5). The reaction, performed at room tempera- glycosylated enzymes that are generally larger than Lip and
ture, is initiated by adding the appropriate amount of MnP, MnP, having molecular masses of approximately 60 kDa and
and the initial rate of Mn(II1)-lactate formation is deter- above (4, 80). In white rot fungi, they are produced as mul-
mined by measuring the increase in absorbance at 240 nm. tiple isozymes (4, 24, 62) and are encoded by gene families
One unit of MnP is usually defined as the amount of enzyme (4, 106). A number of excellent reviews on the physiology,
needed to oxidize 1 pmol of Mn(I1) to Mn(II1) in 1 min. biochemistry, and applications of LACs have appeared (4,
25, 50, 62, 70, 89, 92, 95).
In comparison to classical LACs, which are blue, a blue
LAC as well as a novel yellow LAC were isolated from cul-
tures of Pannus tigrinus (63). The yellow LAC lacks the ab-

Mn3+ jf- Mn2+


sorption maxima typically associated with the blue color in
classic LACs.

sred sox
25.3.3.1. Role of LAC in Lignin Degradation
LAC oxidizes phenolic lignin model compounds, but it was
FIGURE 4 MnP activity assay scheme. widely believed (even a few years ago) that the redox
616 w METABOLISM

potential of LAC is too low to directly oxidize the nonphe- 25.3.4. Versatile Peroxidase
nolic components of lignin (4, 50, 60). However, Eggert et A heme peroxidase different from other microbial, plant,
al. (26, 27) showed that the white rot fungus Pycnopo.rms and animal peroxidases, termed versatile peroxidase (VP),
cinnabarinus, which does not produce either MnP or Lip but was discovered in Pleurotus and Bjerkanderu species (16, 40,
produces LAC, degrades lignin efficiently, suggesting the im- 69, 71, 85,97). VP shares catalytic properties of both MnP
portance of LAC in lignin degradation (27). Furthermore, and Lip. The enzyme exhibits high affinity for Mn2+,hy-
P. cinnubarinw was shown to produce 3-hydroxyanthranilate droquinones and dyes, and also oxidizes VA, dimethoxy-
(HAA) that apparently mediates the oxidation of nonphe- benzene, and lignin dimers (50, 61, 69, 76, 85). Molecular
nolic substrates by LAC. Based on several lines of evidence, models and crystal analysis of native and recombinant VP
it is now clear that LAC is able to oxidize lignin and a wide show an Mn2+-bindingsite formed by three acidic residues
range of lignin-related aromatic compounds in the presence near the heme internal pro ionate accounting for the abil-
of appropriate low-molecular-weight mediators such as ity of VP to oxidize Mn2P (69, 76, 85). VP can oxidize
ABTS, HAA, and l-hydroxybenzotriazole (4, 15,21,26,50, Mn2+ as well as phenolic and nonphenolic aromatic com-
62). The LAX-HAA cou le was shown to be more effective pounds. With regard to oxidation of aromatic substrates, VP
in degrading synthetic '%-lignin than is the LAC-ABTS shows a putative long-range electron transfer pathway from
couple (26). It is widely believed now that low-molecular- an exposed tryptophan to heme, similar to that postulated
weight mediators such as HAA act as diffusible lignin- in Lip (69). Mutagenesis and chemical modification of this
oxidizing agents and that LACs play a far more important tryptophan and the acidic residues forming the Mn2+-bind-
role in lignin degradation than was previously thought (60, ing site confirmed their role in catalysis.
62). The data to date clearly indicate that LACs and MnPs
can degrade both phenolic and nonphenolic aromatic moi- Determination of VP Activity
eties of lignin in the presence of selected low-molecular- Since VP shares many catalytic properties of both MnP and
weight mediator compounds. A LAC-mediated system Lip, determination of VP activity is essentially similar to the
(LignozymeRProcess) was reported to be effective in delig above-mentioned assays for Lip and MnP. In crude prepara-
nifying wood in making paper pulp (15, 25). tions, VP is determined as Mn-independent peroxidase by
using 0.1 mM DMP in 0.1 M sodium tartrate (pH 4 to 5) in
25.3.3.2. LAC Activity Measurement the presence of 0.1 mM H 2 0 z (16) or as Lip in reaction
LAC activity assay is based on the oxidation of a phenolic mixtures containing 4 mM VA in 50 mM succinic acid
reducing substrate in the presence of 0 2 . LAC activities are buffer, pH 3.5 (99).
often assayed using ABTS as the substrate (24, 62). Activity of purified fractions of VP is determined by
Oxidation of ABTS is monitored by determining the in- measuring both the Mn+'-dependent and Mn+2-indepen-
crease in A420( E = 36,000 M-'cm-'). LAC activity is also dent activities (as described above) using purified VP frac-
monitored by measuring the oxidation of catechol as evi- tion (85, 99).
denced by increase in absorbance at 440 nm or by the oxi-
25.3.5. Role of Reactive Oxygen Species
dation of syringaldazine with the concomitant increase in
absorbance at 560 nm (44, 61). Determining LAC activity in Lignin Degradation
spectrophotometrically by measuring the conversion of 2 Reactive oxygen species have been reported to play a direct
mM DMP to 3,5,3,9,5,9-tetramethoxydiphenoquinone at role in lignin degradation by a number of investigators. The
468 nm ( E = 549.6 mM-'cm-l) has also been described (4, production of OH' radicals by white rot fungi using a
62, 66). Furthermore, the use of an oxygen electrode for Fenton-type reaction is well documented (7, 31, 93, 103).
measuring LAC activity has been reported (66). The opti- OH' radicals are very reactive and can attack the subunits
mal conditions for LAC activity are pH 5 and 25C. of lignin both by abstracting aliphatic Ca-hydrogens and by
adding to aromatic rings (43). Typical reactions of OH' rad-
LAC Assay with ABTS as the Substrate ical with the major arylglycerol-P-aryl ether structure of
The reaction mixture for this assay (24) contains, in a total lignin can result in demethoxylation, p-0-4 cleavage, hy-
volume of 1 ml, 14 kmol of ABTS, the enzyme preparation, droxylation, or Ca-oxidation (43). The oxidation of lignin
and 50 mM glycine-HC1 buffer, pH 3.0. The reaction is by OH' radicals therefore results in diverse reactions, some
monitored by determining the increase in absorbance at of which are expected to degrade the polymer. Hydroxy-
420 nm ( E = 36,000 M-lcm-') for 3 to 5 min. The LAC lation of both phenolic and nonphenolic lignin resulting in
activity is usually expressed as nanokatals (nanomoles/sec- new phenolic substructures on the lignin polymer may
ond) per unit amount of enzyme preparation. make it susceptible to attack by LAC or MnP (93).
When DMP is used as the substrate for LAC, the reac- In considering the role of OH' radicals in lignin degra-
tion mixture may contain 1 mM DMP, McIlvane's citrate- dation, it is also necessary to consider the possible role of
phosphate buffer adjusted to pH 5.0, and the MnP enzyme peroxyl (ROO') and hydroperoxyl (HOO') radicals on
preparation. Oxidation of DMP is monitored by measuring lignin degradation, since both of these radicals are expected
the increase in absorbance at 477 nm ( E = 14,800 as secondary radicals when OH' radicals oxidize wood poly-
M-'cm-'). mers (43, 56). MnP of white rot fungi cause peroxidation of
When syringaldazine is used as the substrate (44,61), the unsaturated fatty acids, which results in the formation of
reaction mixture contains 0.5mM syringaldazine (dissolved ROO' (72). ROO' radical-generating systems which appear
in ethanol) and 50 mM phosphate buffer. Syringaldazine to oxidize a nonphenolic P-O-+linked lignin model dimer
oxidation is followed by measuring the increase in ab- to products indicative of hydrogen abstraction (56). Since
sorbance at 525 nm ( E = 65,000 M-' cm-'). white rot fungi do produce extracellular lipids (28), the for-
Several other substrates such as guaiacol and 6-hydroxy- mation of ROO' radicals by an MnP-dependent mechanism
dopamine (2,4,5-trihydroxyphenethylamine) can also be and their involvement in lignin degradation seems reason-
used as substrates for LAC activity measurements. able.
25. Lignin and Lignin-Modifying Enzymes 617

Both Lip and MnP have been reported to decompose ox- oxidase substrate interaction sites. J. Biol. Chem. 274:
alate in the presence of VA and Mn2+, respectively (88), 10324-10330.
and indeed other organic acids (see references 52 and 98). 17. Candeias, L. P., and P. J. Harvey. 1995. Lifetime and re-
These reactions account for the observed oxidation of phe- activity of the veratryl alcohol radical-cation-implica-
nol red and kojic acid by MnP in the presence of Mn2+, tions for lignin peroxidase catalysis. J. Biol. Chem.
without exogenous addition of HI01 (see references 14 and 270:16745-16748.
50). The reduction of VA+' or Mn3+ by oxalate suggests 18. Castillo, M. D., P. J. Stenstrom, and P. Ander. 1994.
that as long as oxalate coexists with Lip and MnP, it would Determination of manganese peroxidase activity with 3-
inhibit lignin degradation. Indeed, oxalate has been shown methyl-2,benzothiazolinonehydrazone and 3-(dimethyl-
amino) benzoic acid. Anal. Biochem. 218:399-404.
to strongly reduce the rate of lignin mineralization in ligni-
19. Chung, N., and S. D. Aust. 1995. Inactivation of lignin
nolytic cultures of white rot fungi (88). peroxidase by hydrogen-peroxide during the oxidation of
C.A.R. acknowledges financial support from U.S. Department of phenols. Arch. Biochem. Biophys. 316:851-855.
Energy, Biological Sciences Diwision. 20. Crawford, R. L. 1981. Lignin Biodegradation and
Transformation. John Wiley & Sons, Inc., New York, NY.
21. Crestini, C., L. Jurasek, and D. S. Argyropoulos. 2003.
25.4. REFERENCES On the mechanism of laccase-mediator system in the oxi-
1. Ander, P., C. Mishra, R. Farrel, and K.-E. Eriksson. dation of lignin. Chem. Eur. J. 95371-5378.
1990. Redox interactions in lignin degradation: interac- 22. Cullen, D., and P. J. Kersten. 2004. Enzymology and mo-
tion between laccase, different peroxidases and cel- lecular biology of lignin degradation, p. 249-273. In R.
1obiose:quinone oxidoreductase. J. Biotechnol. 13:189- Brambl and G. A. Marzluf (ed.), The Mycota III.
198. Biochemistry and Molecular Biology, 2nd ed. Springer-
2. Archibald, F. S. 1992. A new assay for lignin-type peroxi- Verlag, Berlin, Germany.
dases employing the dye azure B. Appl. Enwiron. Microbiol. 23. Davin, L. B., and N. G. Lewis. 2006. Lignin primary
58:3110-3116. structures and dirigent sites. Curr. Opin. Biotechnol.
3. Aro, N., T. Pakula, and M. Pentila. 2005. Transcriptional 16:407415.
regulation of plant cell wall degradation by filamentous 24. D'Souza, T. M., C. S. Merritt, and C. A. Reddy. 1999.
fungi. FEMS. Microbiol. Rew. 29:719-739. Lignin-modifying enzymes of the white rot basidiomycete
4. Baldrian, P. 2006. Fungal laccases-occurrence and prop- Gunoderma lucidum. Appl. Enwiron. Microbiol. 655307-
erties. FEMS. Microbiol. Rew. 30:215-242. 5313.
5 . Bao, W. L., Y. Fukushima, K. A. Jensen, M. A Moen, 25. DurAn, N., M. A. Rosa, A. Dannibale, and L. Gianfreda.
and K. E. Hammel. 1994. Oxidative-degradation of non- 2002. Applications of laccases and tyrosinases (phenol ox-
phenolic lignin during lipid-peroxidation by fungal man- idases) immobilized on different supports: a review.
ganese peroxidase. FEBS. Lett. 354:297-300. Enzyme Mimob. Technol. 31:907-931.
6. Barr, D. P., and S. D. Aust. 1994. Conversion of lignin 26. Eggert, C., U. Temp, J. F. Dean, and K.-E. Eriksson.
peroxidase compound I11 to active enzyme by cation radi- 1997. A fungal metabolite mediates degradation of non-
cals. Arch. Biochem. Biophys. 312611-515. phenolic lignin structures and synthetic lignin by laccase.
7. Barr, D. P., M. M. Shah, T. A. Grover, and S. D. Aust. FEBS Lett. 391:144-148.
1992. Production of hydroxyl radical by lignin peroxidase 27. Eggert, C., U. Temp, and K.-E. Eriksson. 1997. Laccase
from Phanerochaete chrysosporium. Arch. Biochem. Biophys. is essential for lignin degradation by the white-rot fungus
298480485. Pycnoporus cinnabarinus. FEBS Lett. 407:89-92.
8. Blanchette, R. A. 1991. Delignification by wood-decay 28. Enoki, M., T. Watanabe, S. Nakagame, K. Koller, K.
fungi. Annu. Rew. Phytopathol. 29:381-398. Messner, Y. Honda, and M. Kuwahara. 1999. Extra-
9. Blanchette, R. A. 1995. Degradation of the lignocellulose cellular lipid peroxidation of selective white-rot fungus,
complex in wood. Can. J. Bot. 7351:999-1010. Ceriporiopsis subwermispora. FEMS Microbiol. Lett. 180:
10. Blanchette, R. A. 2000. A review of microbial deteriora- 205-2 11.
tion found in archaeological wood from different environ- 29. Eriksson, K.-E., R. A. Blanchette, and P. Ander. 1990.
ments. Int. Biodeterior. Biodegrud. 46:189-204. Microbial and Enzymatic Degradation of Wood and Wood
11. Boominathan, K., and C. A. Reddy. 1992. Lignin degra- Components. Springer, New York, NY.
dation by fungi: biotechnological applications,p. 763-822. 30. Flournoy, D. S., J. A. Paul, T. K. Kirk, and T. L.
In D. K. Arora, K. G. Mukerji, and R. P. Elander (ed.), Highley. 1993. Changes in the size and volume of pores in
Handbook of Applied Mycology, ~01.4.Biotechnology. Marcel sweetgum wood during simultaneous rot by Phanerochaete
Dekker Inc, New York, NY. chrysosporium Burds. Holzforschung47:297-301.
12. Breen, A., and F. L. Singleton. 1999. Fungi in lignocellu- 31. Forney, L. J., C. A. Reddy, M. Tien, and S. D. Aust.
lose breakdown and biopulping. Cum. Opin. Biotechnol. 1982. The involvement of hydroxyl radical derived from
10:25 2-25 8. hydrogen peroxide in lignin degradation by the white rot
13. Buswell, J. A., and 0. Odier. 1987. Lignin biodegrada- fungus Phanerochaete chrysosporium. J. Biol. Chern. 257:
tion. CRC Crit. Rew. Biotechnol. 6:l-60. 11455-1 1462.
14. Cai, D., and M. Tien. 1993. Lignin-degrading peroxidases 32. Gadd, G. M. (ed.). 2001. Fungi in Bioremediation.
of Phnerochaete chrysosporium. J. Biotechnol. 30:79-90. Cambridge University Press, Cambridge, United Kingdom.
15. Call, H. P., and I. Mucke. 1997. History, overview and 33. Gamble, G. R., A. Sethuraman, D. E. Akin, and K.-E.
applications of mediated lignolytic systems, especially Eriksson. 1994. Biodegradation of lignocellulose in
laccase-mediator-systems [Lignozym(R)- process]. J. Bio- Bermuda grass by white-rot fungi analyzed by solid-stateC-
technol. 53: 163-202. 13 nuclear magnetic resonance. Appl. Enwiron. Microbiol.
16. Camarero, S., S. Sarkar, F. J. Ruiz-Duenas, M. J. 60:3 138-3 144.
Martinez, and A. T. Martinez. 1999. Description of a ver- 34. Geng, X., and K. Li. 2002. Degradation of non-phenolic
satile peroxidase involved in the natural degradation of lignin by the white-rot fungus Pycnoporus cinnabarinus.
lignin that has both manganese peroxidase and lignin per- Appl. Microbiol. Biotechnol. 60:342-346.
618 METABOLISM

35. Gilbertson, R. L. 1980. Wood-rotting fungi of North nonphenolic lignin structures by Ceriporiopsis subvermis-
America. Mycologia 72:l-49. pora in the absence of lignin peroxidase. Appl. Environ.
36. Glenn, J. K., and M. H. Gold. 1985. Purification and Microbiol. 62:3679-3686.
characterization of an extracellular Mn( 11)-dependentper- 55. Kapich, A., M. Hofrichter, T. Vares, and A. Hatakka.
oxidase from the lignin-degrading basidiomycete, Phan- 1999. Coupling of manganese peroxidase-mediated lipid
erochaete chrysosporium. Arch. Biochem. Biophys. 242:329- peroxidation with destruction of nonphenolic lignin
341. model compounds and C-14-labeled lignins. Biochem.
37. Glumoff, T., P. J. Harvey, S. Molinari, M. Goble, G. Biophys. Res. Commun. 259:212-219.
Frank, J. M. Palmer, D. G. Smit, and M. S. A. Leisola. 56. Kapich, A. N., K. A. Jensen, and K. E. Hammel. 1999.
1990. Lignin peroxidase from Phanerochaete chrysosporium. Peroxyl radicals are potential agents of lignin biodegrada-
Molecular and kinetic characterization of isozymes. Eur. tion. FEBS Lett. 461:115-119.
J. Biochem. 187615520, 57. Kawai, S., K. A. Jensen, W. Bao, and K. E. Hammel.
38. Gold, M. H., and M. Alic. 1993. Molecular biology of the 1995. New polymeric model substrates for the study of mi-
lignin-degrading basidiomycete Phanerochaete chrysospo- crobial ligninolysis. Appl. Environ. Microbiol. 61:3407-
rium. Microbiol. Rew. 57:605-622. 3414.
39. Gold, M. H., H. Wariishi, and K. Valli. 1989. 58. Kirk, T. K., and D. Cullen. 1998. Enzymology and molec-
Extracellular peroxidases involved in lignin degradation by ular genetics of wood degradation by white-rot fungi,
the white rot basidiomycete Phanerochaete chrysosporium. p. 273-307. In A. R. Young and M. Akhtar (ed.), Envi-
ACS Symp. Ser. 389:127-140. ronmentally Friendly Technologies for the Pulp and Paper
40. Gomez-Toribio, V., A. T. Martinez, M. J. Martinez, and Industry. John Wiley & Sons, Inc., New York, NY.
F. Guillen. 2001. Oxidation of hydroquinones by the ver- 59. Kirk, T. K., and R. L. Farrell. 1987. Enzymatic combus-
satile ligninolytic peroxidase from Pleurotus eryngii. H 2 0 2 tion: the microbial degradation of lignin. Annu. Rev.
generation and the influence of Mn2+. Eur. J. Biochem. Microbiol. 41:465-505.
26847874793. 60. Krause, D. O., S. E. Denman, R. I. Mackie, M.
41. Goodwin, D. C., S. D. Aust, and T. A. Grover. 1995. Morrison, A. L. Rae, G. T. Attwood, and C. S.
Evidence for veratryl alcohol as a redox mediator in lignin McSweeney. 2003. Opportunities to improve fiber degra-
peroxidase-catalyzed oxidation. Biochemistry 34:5060-5065. dation in the rumen: microbiology, ecology and genomics.
42. Hammel, K. E., K. A. Jensen, M. D. Mozuch, L. L. FEMS Microbiol. Rev. 27:663-693.
Landucci, M. Zen, and E. A. Pease. 1993. Ligninolysis 61. Leonowicz, A., and K. Grzywnowicz. 1981. Quantitative
by a purified lignin peroxidase.J. Biol. Chem. 268:12274- estimation of laccase forms in some white-rot fungi using
12281. syringaldazine as a substrate. Enzyme Microb. Technol.
43. Hammel, K. E., A. N. Kapich, K. A. Jensen, Jr., and 35-58.
Z. C. Ryan. 2002. Reactive oxygen species as agents of 62. Leonowicz, A., N. S. Cho, J. Luterek, A. Wilkolazka,
wood decay by fungi. Enzyme Microb. Technol. 30:445-453. M. Wojtaswasilewska, A. Matuszewska, M. Hofrichter,
44. Harkin, M., and J. R. Obst. 1973. Syringaldazine, an ef- D. Wesenberg, and J. Rogalski. 2001. Fungal laccases:
fective reagent for detecting laccase and peroxidase in properties and activity on lignin. J. Basic Microbiol. 41:
fungi. Experientia 2 9: 38 1-3 87. 185-227.
45. Harvey, P. J., and J. M. Palmer. 1990. Oxidation of phe- 63. Leontievsky, A., N. Myasoedova, N. Pozdnyakova, and
nolic compounds by ligninase. J. Biotechnol. 13:169-180. L. Golovleva. 1997. Yellowlaccase of Pannus tigrinus ox-
46. Hatakka, A. 1994. Lignin-modifying enzymes from se- idizes non-phenolic substrates without electron-transfer
lected white-rot fungi-production and role in lignin mediators. FEBS. Lett. 413:446-448.
degradation. FEMS Microbiol. Rev. 13:125-135. 64. Lin, Y., and S. Tanaka. 2006. Ethanol fermentation from
47. Hatakka, A. 2001. Biodegradation of lignin, p. 129-184. biomass resources. Appl. Microbiol. Biotechnol. 69:627-
In M. Hofrichter and A. Steinbuchel (ed.), Biopolymers, 642.
vol. 1. Lignin, Humic Substances and Coal. Wiley-VCH, 65. Lynd, H. R., W. H. VanZyl, J. E. Mcbride, and M. Laser.
Weinheim, Germany. 2005. Consolidated bioprocessing of cellulosic biomass: an
48. Hatakka, A., T. Lundell, M. Hofrichter, and P. Maijala. update. Cum. Opin. Biotechnol. 16577-583.
2003. Manganese peroxidase and its role in the degrada- 66. Mai, C., W. Schorman, 0. Milstein, and A. Hutterman.
tion of wood lignin, p. 230-243. In S. D. Mansfield and 2000. Enhanced stability of laccase in the presence of
J. N. Saddler (ed.), Applications of Enzymes to Ligno- phenolic compounds. Appl. Microbiol. Biotechnol. 54510-
cellulosics. ACS Symposium Series 855. Oxford University 514.
Press, Washington, DC. 67. Makela, M., S. Galkin, A. Hatakka, and T. Lundell.
49. Higuchi, T. 1990. Lignin biochemistry: biosynthesis and 2002. Production of organic acid and oxalate decarboxy-
biodegradation. Wood Sci. Technol. 24:23-63. lase in lignin-degrading white rot fungi. Enzyme Microb.
50. Higuchi, T. 2004. Microbial degradation of lignin: role of Technol. 30542-549.
lignin peroxidase, manganese peroxidase, and laccase. Jpn. 68. Martinez, A. T. 2002. Molecular biology and stmcture-
Acad. Ser. B 80:204-214. function of lignin-degrading heme peroxidases. Enmrne
51. Higuchi, T., H.-M. Chang, and T. K. Kirk (ed.). 1983. Microb. Technol. 30:425-444.
Recent Advances in Lignin Biodegradation Research. Uni- 69. Martinez, A. T., M. Speranza, F. J. Riz-Duenas, P.
publishers Co. LTD., Tokyo, Japan. Ferreira, S. Camarero, F. Guillen, M. J. Martinez, A.
52. Hofrichter, M. 2002. Review: lignin conversion by man- Gutierrez, and J. C. del Rio. 2005. Biodegradation of lig-
ganese peroxidase (MnP). Enzyme Microb. Technol. 30: nocellulosics: microbial, chemical, and enzymatic aspects
454-466. of the fungal attack of lignin. Int. Microbiol. 8:195-204.
53. Ikehata, K., I. D. Buchanan, and D. W. Smith. 2004. 70. Mayer, A. M., and R. C. Staples. 2002. Laccase: new
Recent developments in the production of extracellular functions for an old enzyme. Phytochemistry 6 0 5 5 1-565.
fungal peroxidases and laccases for waste treatment. J. 71. Mester, T., and J. A. Field. 1998. Characterization of a
Enwiron. Eng. Sci. 3:l-19. novel manganese peroxidase-lignin peroxidase hybrid iso-
54. Jensen, K. A., W. L. Bao, S. Kawai, E. Srebotnik, and zyme produced by Bjerkandera species strain BOS55 in the
K. E. Hammel. 1996. Manganese-dependent cleavage of absence of manganese. J. Biol. Chem. 273:15412-15417.
25. Lignin and Lignin-Modifying Enzymes 619

72. Moen, M. A., and K. E. Hammel. 1994. Lipid-peroxida- 90. Srebotnik, E., K. A. Jensen, Jr., and K. E. Hammel.
tion by the manganese peroxidase of Phunerochuete 1994. Fungal degradation of recalcitrant nonphenolic
chrysosporium is the basis for phenanthrene oxidation by lignin structures without lignin peroxidase. Proc. Natl.
the intact fungus. Appl. Environ. Microbiol. 60:1956-1961. Acud. Sci. USA 91:12794-12797.
73. Mosier, N., C. Wyman, B. Dale, R. Elander, Y. Y. Lee, 91. Srebotnik, E., and K. Messner. 1994. A simple method
M. Holtzapple, and M. Ladisch. 2005. Features of promis- that uses differential staining and light microscopy to as-
ing technologies for treatment of lignocellulosic biomass. sess the selectivity of wood delignification by white-rot
Bioresour. Technol. 96:673-686. fungi. Appl. Enwiron. Microbiol. 60:1383-1386.
74. Ollikka, P., V. M. Leppanen, T. Anttila, and I. 92. Srebotnik, E., and K. Messner. 2001. Oxidative mecha-
Suominen. 1995. Purification of major lignin peroxidase nisms involved in lignin degradation by white-rot fungi.
isoenzymes from Phunerochuete chrysosporium by chromato- Chem. Rev. 101:3397-3413.
focusing. Protein Expr. Purif. 6:337-342. 93. Tanaka, H., S. Itakura, and A. Enoki. 1999. Hydroxyl
75. Paszczynski, A., R. Crawford, and V.-B.Huynh. 1986. radical generation by an extracellular low-molecular-
Manganese peroxidase of Phunerochuete chrysosporium: pu- weight substance and phenol oxidase activity during
rification. Methods Enzymol. 161:264-270. wood degradation by the white-rot basidiomycete
76. PCrez-Boada, M., F. J. Ruiz-Dvenas, R. Pogni, R. Basosi, Trametes wersicolor. J. Biotechnol. 7557-70.
T. Choinowski, M. J. Martinez, K. Piontek, and A. T. 94. Teunissen, P. J., and J. A. Field. 1998. 2-Chloro-1,4-
Martinez. 2005. Versatile peroxidase oxidation of high dimethoxybenzene as a mediator of lignin peroxidase cat-
redox potential aromatic compounds: site-directed muta- alyzed oxidations. FEBS Lett. 439:219-223.
genesis, spectroscopic and crystallographic investigations 95. Thurston, C. F. 1994. The structure and function of fun-
of three long-range electron transfer pathways. J. Mol. gal laccases. Microbiology 140:19-26.
Biol. 354:385-402. 96. Tien, M., and K. Kirk. 1984. Lignin degrading enzyme
77. Reddy, C. A., and T. M. DSouza. 1994. Physiology and from Phunerochaete chrysosporium: purification, character-
molecular biology of the lignin peroxidases of Phanero- ization, and catalytic properties of a unique H202-requiring
chuete chrysosporium. FEMS Microbiol. Rew. 13:137-152. oxygenase. Proc. Natl. Acad. Sci. USA 81:2280-2284.
78. Reddy, C. A., and Z. Mathew. 2001. Bioremediation po- 97. Tsukihara, T., Y. Honda, T. Watanabe, and T.
tential of white-rot fungi, p 52-78. In G. M. Gadd (ed.), Watanabe. 2006. Molecular breeding of white rot fungus
Fungi in Bioremediation. Cambridge University Press, Pleurocus ostreatw by homologous expression of its versatile
Cambridge, United Kingdom. peroxidase MnP2. Appl. Microbiol. Biotechnol. 7 1:114-120.
79. Renganathan, V., and M. H. Gold. 1986. Spectral char- 98. Urzua, U., P. J. Kersten, and R. Vicuna. 1998.
acterization of the oxidized states of lignin peroxidase, an Manganese peroxidase-dependent oxidation of glyoxylic
extracellular heme enzyme from the white rot basidiomy- and oxalic acids synthesizedby Ceriporiopsis subvermisporu
cete Phanerochuete chrysosporium. Biochemistry 25:1626-1631. produces extracellular hydrogen peroxide. Appl. Environ.
80. Rheinhammar, B. 1984. Laccase, p. 4-10. In R. Lontie Microbiol. 64:68-73.
(ed.), Copper Proteins and Copper Enzymes. CRC Press, 99. Verdin, J., R. Pogni, A. Baeza, M. C. Baratto, R.
Inc., Boca Raton, FL. Basosi, and R. Vazquez-Duhalt. 2006. Mechanism of
81. Rheinhammar, B., and T. K. Kirk. 1990. Comparison of versatile peroxidase inactivation by Ca(*+) depletion.
lignin peroxidase,horseradish-peroxidase and laccase in the Biophys. Chem. 121:163-170.
oxidation of methoxybenzenes. Biochem. J. 268:475480. 100. Ward, G., P. Belinky, Y. Hadar, I. Bilkis, and C. G.
82. Rodriguez, C. S., and H. J. L. Toca. 2006. Industrial and Dosoretz. 2002. The influence of non-phenolic media-
biotechnological applications of laccases: a review. tors and phenolic co-substrateson the oxidation of 4-bro-
Biotechnol. Adv. 24500-513. mophenol by lignin peroxidase. Enzyme Microb. Technol.
83. Rothschild, N., C. Novotny, S. Sasek, and C. G. 30:490498.
Dosoretz. 2002. Ligninolytic enzymes of the fungus Irpex 101. Ward, G., Y. Hadar, I. Bilkis, and C. G. Dosoretz. 2003.
lacteus (Polyporus tulipifera): isolation and characteriza- Mechanistic features of lignin peroxidase catalyzed oxida-
tion of lignin peroxidase. Enzyme Microb. Technol. 31: tion of substituted phenols and 1,2-dimethoxyarenes.
627-633. J. Biol. Chem. 278:39726-39734.
84. Roy, B. P., M. G. Paice, F. S. Archibald, S. K. Misra, and 102. Wariishi, H., K. Valli, and M. H. Gold. 1992.
L. E. Misiak. 1994. Creation of metal-complexing agents, Manganese I1 oxidation by manganese peroxidase from
reduction of manganese-dioxide, and promotion of man- the basidiomycete Phanerochuete chrysosporium. Kinetic
ganese peroxidase-mediated Mn(111) production by cel- mechanism and role of chelators. J. Biol. Chem. 267:
lobiose-quinone oxidoreductase from Trametes wersicolor. 23688-23695.
J. Biol. Chem. 269:19745-19750. 03. Wood, P. M. 1994. Pathways of production of Fenton
85. Ruiz-Duenas, F. J., S. Camarero, M. Perez-Boada, M. J. reagent by wood-rotting fungi. FEMS Microbiol. Rew.
Martinez, and A. T. Martinez. 2001. A new versatile per- 13:3 13-320.
oxidase from Pleurotus. Biochem. SOC.Trans. 29:116-122. 04. Wood, W. A., and S. T. Kellogg. 1988. Lignin, Pectin,
86. Rypacek, V. 1977. Chemical composition of hemicellu- and Chitin. Methods in Enzymology, vol. 161. Academic
loses as a factor participating in the substrate specificity of Press, Inc., San Diego, CA.
wood-destroying fungi. Wood Sci. Technol. 1159-67. 05. Wymelenberg, A. V., P. Minges, G. Sabat, D. Martinez,
87. Scott, G. M., M. Akhtar, M. J. Lentz, T. K. Kirk, and R. A. Aerts, A. Salamov, I. Grigoriev, H. Shapiro, N.
Swaney. 1998. New technology for papermaking: com- Putnam, P. Belinky, C. Dosoretz, J. Gaskell, P. Kersten,
mercializing biopulping. Tappi J. 81:220-225. and D. Cullen. 2006. Computational analysis of the
88. Shimada, M., D. B. Ma, Y. Akamatsu, and T. Hattori. Phanerochuete chrysosporium v2.0 genome database and
1994.A proposed role of oxalic acid in wood decay systems mass spectrometry identification of peptides in lignin-
of wood-rotting basidiomycetes. FEMS Microbiol. Rev. 13: olytic cultures reveal complex mixtures of secreted pro-
285-296. teins. Fungal Genet. Biol. 43:343-356.
89. Solomon, E. I., U. M. Sundaram, and T. E. Machonkin. 106. Yaver, D. S., F. Xu, E. J. Golightly, K. M. Brown, S. H.
1996. Multicopper oxidases and oxygenases. Chem. Rev. Brown, M. W. Rey, P. Schneider, T. Halkier, K. Mondorf,
96:2563-2605. and H. DalbQge. 1996. Purification, characterization,
620 w METABOLISM

molecular cloning, and expression of two laccase genes of Bjerkundera udusta with synthetic lignin in acetone so-
from the white-rot basidiomycete Trumetes villosa. Appl. lution. J . Wood Sci. 44:486490.
Environ. Microbiol. 62:834-841. 108. Zadrazil, F., and P. Reinger (ed.). 1988. Treatment of
107. Yoshida, S., A. Chatani, Y. Honda, T. Watanabe, and Lignocellulosics with White-Rot Fungi. Elsevier Applied
M. Kuwahara. 1998. Reaction of manganese peroxidase Science, London, United Kingdom.
MOLECULAR GENETICS

Introduction to Molecular 31. Gene Transfer in Gram-Negative


Genetics 621 Bacteria 735
C. A. REDDY, Editor JOSEPH E. PETERS
L. R. SNYDER, Co-Editor
32. Genetic Exchange in Gram-Positive
26. Similarity Analysis of DNAs 624 Bacteria 756
JOHN L. JOHNSON AND CHRISTOPHER J. KRISTICH,
WILLIAM B. WHITMAN CHRISTINE E. SALOMON, AND
GARY M. DUNNY
27. Nucleic Acid Analysis 653
WILLIAM HENDRICKSON AND 33. Genetics of Archaea 800
DON WALTHERS KEVIN R. SOWERS, PAUL H. BLUM, AND
SHILADITYA DAsSARMA
28. Measuring Spontaneous Mutation
Rates 676 34. Genetic Manipulations Using Phages
PATRICIA L. FOSTER 825
GRAHAM F. HATFULL,
29. Transposon Mutagenesis 684 DEBORAH JACOBS-SERA,
SILVIA ROSSBACH AND FRANS J. DE BRUIJN MICHELLE H. LARSEN, AND
WILLIAM R. JACOBS, JR.
30. Plasmids 709
MARCEL0 E. TOLMASKY, LUIS A. ACTIS,
TIMOTHY J. WELCH, AND JORGE H. CROSA
Introduction to Molecular Genetics
C. A. REDDY

Philipp Gerhardt wrote in his Introduction to Molecular a table. This is another useful chapter of basic methodology
Genetics section in Methods for General and Molecular in molecular biology.
Bacteriology (MGMB), The growth of knowledge about Measuring Spontaneous Mutation Rates (chapter 28),
the molecular genetics of bacteria has accelerated like a authored by Foster, is new to this edition. Spontaneous mu-
chain reaction and that molecular genetics has become a tations occur in the absence of exogenous causes and can
premier part of research in microbiology. These state- be due to a variety of factors such as errors made by DNA
ments are as true today as they were then. The explosive polymerases during replication (or repair), errors made dur-
growth in molecular genetics in the past few decades has ing recombination, and several others. Measuring sponta-
been so extensive that it has permeated all the subdisci- neous mutation rates is quite challenging, and this chapter
plines of microbiology and in fact practically all areas of should be very useful for researchers in this area.
modern biology. Many of the major topics in molecular The chapters Transposon Mutagenesis (chapter 29) by
genetics have grown so big that one or more books have Rossbach and de Bruijn, Plasmids(chapter 30) by Tolmasky
been published on the individual topics. Therefore, any- et al., Gene Transfer in Gram-Negative Bacteria (chapter
thing beyond a basic coverage of the methodology in this 31) by Peters, and Genetic Exchange in Gram-Positive
field is not practical in a book of this kind. However, the Bacteria (chapter 32) by Kristich et al. are retained from the
methods covered in this section constitute a good start for previous edition. Each of these chapters has been revised and
most of the basic techniques in molecular genetics, and updated for this edition and each continues to be a key com-
the reader may then seek more detailed and specialized ponent in this section. To accommodate the large body of re-
information on advanced procedures from other pub- search information that has become available on the molec-
lished sources. ular biology of Archaea since the last edition of this book,
Chapter 26, Similarity Analysis of DNAs by Johnson chapter 33, Genetics of Archaea, authored by Sowers et al.,
and Whitman, is a new inclusion in this section and is an is a welcome new addition to this edition. Another excellent
update of a chapter by the same name from the previous edi- chapter that is being presented for the first time in this edi-
tion (but in a different section) and authored by Johnson tion is chapter 34, Genetic Manipulations Using Phages,
(now deceased). This is a comprehensive chapter of basic authored by Hatfull et al. In this chapter, the authors focus on
methods such as DNA isolation and measurement, DNA la- the use of bacteriophages to manipulate host bacteria geneti-
beling, and DNA-DNA reassociation. Chapter 27, Nucleic cally and as sources for the development of genetic tools with
Acid Analysis, authored by Hendrickson and Walthers, is wide applications in microbiology.
a revised and updated version of a chapter by the same name I thank my coeditor, Loren Snyder, for his assistance in
in the previous edition and includes several new figures and editing the chapters in this section.

623
26
Similarity Analysis of DNAs
JOHN L . JOHNSON AND WILLIAM B. WHITMAN

26.1. CELL GROWTH AND DISRUPTION .......................... 625


26.1.1. Growth Conditions ................................... 625
.2. Cell Disruption...................................... 626
.........................
26.1.2.1. Gram-Negative Bacteria 626
. .........................
2. Gram-Positive Bacteria 626
. ...........................
3. Recalcitrant Bacteria 626
26.2. DNA ISOLATION ......................................... 627
26.2.1. Marmur Procedure .................................... 627
.2. CetyltrimethylammoniumBromide Lysis .................... 629
.3. Hydroxylapatite Adsorption............................. 630
26.3. DNA MEASUREMENT ..................................... 631
26.3.1. UV Spectrophotometry ................................ 631
.2. Hyperchromic Shift ................................... 631
.3. Colorimetric Method (Diphenylamine Reaction)............... 631
. ........................
4. Fluorimetric Dye-Binding Methods 632
26.3.4.1. Mithramycin................................. 632
. .............
2. 4,6.Diamidin o.2+Phenylindole . 2HC1 632
. ....................
3. Bisbenzimide (Hoechst 33258) 632
. .............................
4. Ethidium Bromide 633
26.4. GUANINE*PLUS+CYTOSINECONTENT OF DNA ............... 633
26.4.1. Thermal Melting Method ............................... 633
.2. High-Performance Liquid Chromatography .................. 634
.3. Fluorimetric Dye-Binding Methods ........................ 636
26.4.3.1. Purified DNA ................................ 636
. ..............................
2. Intact-Cell DNA 636
..........
26.4.4. Differential Scanning Calorimetry of Intact-Cell DNA 636
26.5. DNA LABELING ......................................... 636
26.5.1. Iodination .......................................... 637
.2. Photobiotination ..................................... 638
.3. Glutaraldehyde Procedure .............................. 638
.4. Nick Translation Procedure ............................. 638
.5. Random Primer Procedure .............................. 638
26.6. DNA-DNA REASSOCIATION ............................... 638
26.6.1. Special Apparatus .................................... 638
.2. Free-Solution Methods ................................. 639
26.6.2.1. Preliminary Procedures......................... 639
. ..............
2. Reassociation Measured by Photometry 641
. ............
3. Reassociation Measured with S1 Nuclease 642
. 4. Reassociation Measured with HA.................. 642
. ...................
5. Thermal Stability Measurement 643
. ......................
6. Determination of CotValues 644
26.6.3. Membrane Methods ................................... 645
26.6.3.1. DNA Immobilization on Membranes ................ 646
. ........
2. Preincubation of DNA-Containing Membranes 646
. ............
3. Reassociation Measured by Direct Binding 647
. ...................
4. Thermal Stability Measurement 648
26.6.4. Fluorometric Plate Method .............................. 648
26.7. RELIABILITY AND COMPARISON OF RESULTS ................ 649
26.7.1. DNA Reassociation Values .............................. 649
.2. DNA Thermal Stability Values........................... 649
26.8. REFERENCES ............................................ 649
26.8.1. General References ................................... 649
.2. Specific References................................... 650

624
26. Similarity Analysis of DNAs 1 625

Elucidation of the structure and the physical properties of is directly related to the sequence similarity. In fact, it is the
DNA and RNAs has enabled investigators to determine change in the melting temperature of the hybrid DNA
phylogenetic and taxonomic relationships among bacteria complexes that is directly related to sequence similarity
by comparing their genomes. A preliminary comparison be- (18), while the percent reassociation is the fraction of DNA
tween two organisms can be made with respect to the over- with a sequence similarity sufficient to form hybrids under
all nucleotide base composition of DNA. This is usually ex- the experimental conditions chosen. Thus, the criteria that
pressed as the moles percent guanine plus cytosine, which is are used to delineate prokaryotic species, reassociation val-
considered a constant feature of that organism. A large dif- ues below 70% and a difference in the melting temperature
ference in the moles percent G + C values from DNAs of of the DNA hybrids of greater than 5C, suggest that less
two organisms indicates the lack of a close genetic related- than 70% of the DNA forms hybrids which have 91 to 92%
ness; however, a similarity in the moles percent G+C does or greater sequence similarity. Even though the interpreta-
not necessarily mean that the two organisms are closely re- tion of DNA reassociation values is complex, these values
lated with regard to the sequence of nucleotides in their are still useful to express relatedness between prokaryotes,
DNAs. In such cases, methods more precise than DNA base especially closely related species, subspecies, and strains of a
composition are required for comparing the genomes, species.
namely DNA-DNA reassociation (the formation of du- In theory and in practice, DNA reassociation experi-
plexes between single-stranded DNA fragments by comple- ments are relatively easy to do. However, there are many
mentary base pairing), rRNA-DNA hybridization (the for- technical details or problems associated with the experi-
mation of duplexes between single-stranded DNA and ments. These include the initial isolation of high-purity
rRNA), and the nucleotide sequencing of specific genes and DNA, experimental conditions, and the type of experi-
rRNAs. Methods involving DNA reassociation are covered ment. Some problems may be unique for a given group of
in this chapter, and those involving rRNA hybridization bacteria, and there are no universal solutions. The methods
and nucleotide sequencing are dealt with in a following described in this chapter are, for the most part, those that
chapter. have been successfully employed in one of the authors lab-
DNA hybridization is especially important because it re- oratories, and familiarity has contributed to the selection of
mains one of the major criteria for the definition of a the specific methods. In some cases specialized instruments
prokaryotic species (109). In this proposal by an Ad Hoc are required, such as those found in core facilities (e.g., de-
Committee of the International Committee of Systematic termination of moles percent G + C in intact cells by flow
Bacteriology, the criteria for placement of strains in separate cytometry or differential scanning colorimetry).
species include DNA reassociation values below 70% and a Additional background information on DNA reassocia-
difference in the melting temperature of the DNA hybrids tion methods, as well as on DNA base composition meth-
of >5C. These numerical values for the species boundary ods, can be found in the general references at the end of this
represent the values below which phenotypic similarity is chapter (1-6).
difficult to demonstrate (for examples, see references 23 and
54). Thus, when the DNA reassociation is above 70%, the
strains possess significant phenotypic similarity. Below 70% 26.1. CELL GROWTH AND DISRUPTION
DNA reassociation, the phenotypic similarities of strains The rapid growth of many bacteria, relative to other organ-
are often too low to establish taxonomic relationships. isms, is advantageous for the quick generation of cells from
Although sequencing of 16s rRNA genes has proven which to isolate DNA. The growth rates and the extent of
enormously successful in elucidating the deeper taxonomic growth, however, vary greatly and dictate how long cultures
relationships at the genus level and below, it has failed to must be grown and what volumes are produced.
provide reliable information for making species level dis-
tinctions (58, 98). Thus, DNA reassociation remains the 26.1 . I , Growth Conditions (also see chapters
major method for bridging the modern molecular criteria 9 to 16)
based on sequencing with the phenotypic descriptions The group of organisms being investigated determines the
based on numerical taxonomy (97). culture medium requirements. A peptone-yeast extract-
The reassociation of denatured DNA fragments or the based medium supplemented with a carbohydrate energy
hybridization between denatured DNA and rRNA have source works well for cultivating many chemoheterotrophs.
commonly been referred to as homologies, with the results If high levels of acidic by-products are produced, inclusion
reported as relative percent homology values. In retro- of a buffer (e.g., 50 mM potassium phosphate [pH 7.01) may
spect, the word homology was not the best choice to de- result in larger cell numbers.
scribe these experiments, and it is not used in this chapter The consideration of oxygen requirements is very im-
because of its long use in a different sense among classical portant. Although reducing agents are usually added to
evolutionalists: homology (or homologous) refers to phylo- anaerobic media, many anaerobic bacteria grow well in a
genetic development, in which, for example, the arm and freshly prepared medium with a nitrogen-filled headspace
the wing are viewed as homologous appendages, with de- (26, 43) and a 5% (vol/vol) inoculum of an actively grow-
grees of similarity between the two. Thus, homology refers ing culture. For anaerobic bacteria requiring COz, sterile
to a common ancestry and is not used quantitatively. Also, sodium bicarbonate can be added at the time of inoculation.
in the literature on DNA reassociation, the relative Gentle agitation (stirring bar or shaking) may also be im-
amounts of DNA from one organism forming duplexes with portant for organisms that tend to settle under static growth
DNA from the same organism or from another organism conditions. Facultatively anaerobic organisms are best cul-
have been referred to in the literature as percent similarity, tured under aerobic conditions, because smaller amounts of
percent identity, or percent reassociation. For consistency, acidic end products are produced. Actively growing aerobic
only the term percent reassociation is used in this chap- cultures rapidly deplete oxygen in the medium; therefore,
ter. This terminology is chosen to avoid the misconception the rate at which oxygen is dissolved from the atmosphere
that the extent of reassociation between two types of DNA limits the growth rate. Aeration is optimized by using small
626 w MOLECULAR GENETICS

volumes of medium per flask (e.g., 250 ml per 2-liter flask), other staphylococci; and achromopeptidase, a peptidase
flasks with fluted walls, and rapid shaking (250 to 450 rpm). that is isolated from Achromobacter lyticus and is also active
The oxygen requirements of microaerophilic organisms are on the cross-linking peptides.
the most difficult to fulfill. The medium is first equilibrated
by sparging with a mixture of air and COZ, and the sparging Lysozyme
is continued during growth (104). Use the same suspending buffer described previously for
The preferred time for harvesting a culture is at the early gram-negative organisms. Add 1 to 3 mg of lysozyme per ml,
stationary phase, when the amount of growth is near maxi- and incubate at 37C until the cells are susceptible to lysis
mum and the death rate is still minimal. The amount of at- by a detergent.
tention that must be given to the growth curve depends on N-Acetylmuramidase
the organism. Many organisms have a long stationary phase First isolated by Yokogawa et al. (1 15),N-acetylmuramidase
with little cell death, so that the exact harvest time is not has the same substrate specificity as lysozyme, but the range
very critical. Others may start dying quite soon after reaching of organisms that can be lysed by it differs (114). The rec-
maximal growth. With sporulating organisms, vegetative-cell ommended buffer for this enzyme is 20 mM Tris-HC1 (pH
DNA can be converted into sporal DNA during the 1- to 7.0). Add approximately 6 kg of the enzyme per ml to the
3-h sporulation period of the stationary phase. cell suspension and incubate at 50C.
26.1.2. Cell Disruption (also see chapter 7) Lysostaphin
The first step in DNA isolation is the disruption of the bac- Lysostaphin is specific for the cross-linking peptides in the
terial cells. Depending on the nature of the cell wall, bacte- cell walls of staphylococci (88). The recommended buffer
rial cells can be disrupted (i) with a detergent alone, (ii) for the enzyme is 50 mM Tris-HC1-0.145 M NaCl (pH 7.5).
with a combination of detergents and hydrolytic enzymes, Add the enzyme to a concentration of 25 kg/ml and incu-
or (iii) by physical methods such as sonic oscillation, shear- bate at 37C.
ing release from a pressure cell, or shaking in the presence
of glass beads. Achromopeptidase
The general protocols are for culture volumes of 250 to The Achromobacter lyticus strain that produces achromopep-
350 ml, and after centrifugation, the cell pellets are sus- tidase was first isolated at Takeda Chemical Industries, Ltd.,
pended in 25 ml of suspending buffer (see below). These Osaka, Japan. The enzyme has been found to be useful for
volumes can be scaled up or down depending on the plasmid isolation (44) and is also useful for routine DNA
amount of growth or the amount of DNA needed. For many isolations. The recommended buffer is 10 mM Tris-HC1
organisms, using cell pellets that have been stored frozen (pH 8.2). Add the enzyme to a concentration of 50 yg/ml
greatly reduces the yield of DNA, and these procedures and incubate at 50C until the cells become sensitive to
work best with fresh cell material. lysis.

26.1.2.1. Gram-Negative Bacteria 26.1.2.3. Recalcitrant Bacteria


Detergents alone can cause many gram-negative bacteria to There are many bacteria whose cell walls are not suscep-
lyse, but some species may lyse only partially or not at all. tible to enzymic digestion and detergent lysis. Although
A n initial incubation of the cells in the presence of hen egg this group may include some gram-negative bacteria, most
white lysozyme often provides a more uniform lysis on are typical gram-positive bacteria (such as Actinomyces,
subsequent treatment with the detergent. (Lysozyme is a Streptococcus, Peptococcus, and Propionibacterium species)
muramidase, a hydrolytic enzyme that acts on the peptido- and archaea (such as Methanosarcina and Methanobacterium
glycan of the cell wall. It is inhibited by high salt concen- species). There are two approaches available for disrupting
trations and low pH.) these recalcitrant bacteria. The first involves making the
Suspend the cells in a buffer consisting of 10 mM Tris- cells susceptible to one or more lytic enzymes by growing
HC1 (pH 8.0),1 mM sodium EDTA, and 0.35 M sucrose. them in the presence of wall-component analogs or antibi-
The sucrose stabilizes the resulting spheroplasts until the otics, and the second involves the use of any of several
addition of the lysing solution. The EDTA binds the diva- physical methods for cell disruption.
lent cations that stabilize the outer membrane, and Tris One can make cells more susceptible to lysozyme by
buffer also disrupts the outer membrane. Add lysozyme (0.5 growing them in the presence of glycine, threonine, or ly-
to 1.0 mglml), and incubate at room temperature for 5 min; sine (22, 113). However, a problem with using high con-
then, add the lysing solution (see section 26.2.1). If the centrations of these amino acids is that they may inhibit
cells are not readily lysed by the lysing solution, use a tem- growth. Another way of making cells susceptible to ly-
perature of 37C for the initial lysozyme digestion. It is also sozyme is by adding penicillin at late log phase to inhibit
useful to microscopically follow spheroplast formation to wall synthesis. Here one has to be concernea thatthicells
ensure successful lysis. do not lyse before or during harvesting. Required penicillin
levels may vary from strain to strain. Lysozyme-induced lysis
26.1.2.2. Gram-Positive Bacteria of some gram-negative bacteria can be improved by washing
In contrast to gram-negative bacteria, nearly all gram- the cells with a mild detergent (0.1% Sarkosyl) and then
positive bacteria must be digested with a lytic enzyme be- subjecting them to a mild osmotic shock (89).
fore they can be lysed by a detergent. In addition to Many physical methods have been used to disrupt bacte-
lysozyme, which is the enzyme most commonly used, several rial cells. The following are some methods that have been
other enzymes are available. These include N-acetylmur- routinely used for DNA isolation.
amidase, which is isolated from Streptomycesglobisporus and
also cleaves the muramic acid backbone; lysostaphin, an Sonication
endopeptidase that is isolated from Staphylococcus sp. strain The major problems with sonication are that it may not dis-
K-6-WI and is specific for the cross-linking peptides of rupt cells having thick cell walls or small sizes and that it
26. Similarity Analysis of DNAs 627

causes substantial fragmentation of the DNA. The amount sonably large fragments such that, in addition to all of the
of fragmentation increases with time of sonication. DNA standard DNA reassociation and RNA hybridization exper-
preparations from cells disrupted in this manner are not use- iments, it can be used in endonuclease restriction digestions
ful for restriction endonuclease digestion and cloning, but if and partial digestions for fragment size estimations, fragment
the sonication times can be standardized the DNA prepara- size polymorphism studies, and construction of genomic li-
tions can be used in reassociation experiments. braries. The major problem with the procedure, depending
on the organism, is the coisolation of high-molecular-weight
French Pressure Cell polysaccharides, which have ethanol precipitation proper-
Disruption by passage through a French pressure cell has the ties very much like those of DNA. The current variation of
same limitations as sonication. The advantage of the the Marmur procedure that is used in the authors laboratory
French pressure cell is that one obtains more evenly sized includes the addition of the reducing agent 2-mercap-
DNA fragments. toethanol (which was initially used to inactivate nucleases
Glass Bead Disruption in eucaryotic tissue but also seems to work with bacterial
When suspensions of organisms are shaken with glass beads cells) and proteinase K to degrade proteins. The procedure
at high speed (4,000 oscillations per min), the cells rupture is described for culture volumes of 250 to 500 ml but is
by being crushed between the colliding beads. The bead size amenable to scaling up or down. The protocol below is based
is very important; for bacteria, small beads of about 100 k m on the use of lysozyme to render the cells susceptible to SDS
in diameter are used. There are several instruments avail- disruption, but the suspending buffer can be modified for use
able commercially for shaking cells with glass beads. One with other lytic enzymes.
that works well is the Braun Cell Homogenizer (Braun Reagents
Instruments, Melsungen, Germany). The colliding beads
produce so much kinetic energy that the sample vessel must Cell-suspending buffer: 10 mM Tris-HC1 (pH 8.0),1 mM
be cooled with liquid COz.Although it can disrupt all cell sodium EDTA, and 0.35 M sucrose.
types, the homogenizer does not fragment DNA to the same Na-EDTA, 0.5 M solution (pH 8.0).
extent as does sonication or passage through a French pres- SDS, 20% (wtlvol) solution in water.
sure cell. Proteinase K, 20 mg/ml in Tris-EDTA (TE)buffer. The lat-
Alkaline Hydrolysis ter consists of 10 mM Tris-HC1 (pH 8.0) containing
Sodium hydroxide at a concentration of 0.03 N has been 1 mM sodium EDTA (pH 8.0).
used for the mild disruption of gram-negative bacilli and Lysing solution. The lysing solution consists of 100 mM
subsequent isolation of native DNA (9) (see section Tris-HC1 buffer (pH 8.0),0.3 M NaC1, 20 mM EDTA,
26.2.1). It has also been used at a concentration of 0.4 N for 2% (wtlvol) SDS, 2% (vol/vol) 2-mercaptoethanol, and
not only disrupting bacteria but also obtaining denatured 100 pg/ml proteinase K. Add the proteinase K and 2-
(single-stranded) DNA, as in the following procedure (77). mercaptoethanol the day of use; do not store the com-
Suspend the bacterial cells in water, and add NaOH plete solution longer than 1 day. Also, just before use,
from a 10 N stock solution to give a final concentration of add 0.5 vol of 5 M sodium perchlorate to the volume of
0.4 N. (CAUTION: Add the stock solution slowly, and be lysing solution needed. A precipitate will form but will
sure to wear safety goggles.) Incubate in a water bath at quickly dissolve if the solution is warmed in a 50 to 60C
80C for 30 min. Under these conditions the RNA, pro- water bath. Add 1.5 vol of this mixture to each volume
teins, and many of the polysaccharides are hydrolyzed, but of cell suspension.
the denatured DNA remains intact except for some depuri- Sodium acetate, 3 M (pH 6.0).
nation and strand scission (105). The alkaline lysate can be Standard saline-citrate (SSC): 0.15 M NaC1, 0.015 M
used directly to immobilize DNA on nylon membranes; al- trisodium citrate (pH 7.0). Other concentrations of SSC
ternatively, after neutralization, the denatured DNA can be are indicated by multipliers; for example, 20X SSC =
isolated by using hydroxylapatite (HA) (see section 26.2.3). 20-fold the standard concentration, 0.1X SSC = 1/10
the standard concentration.
26.2. DNA ISOLATION Chloroform-isopentanol: chloroform containing 3%
The pioneering work of Kirby et al. (59, 60) and Marmur (vol/vol) isopentanol.
(74) has provided the basis for many nucleic acid isolation Phenol-chloroform: water-saturated phenol mixed with an
protocols. Procedures involving cetyltrimethylammonium equal volume of chloroform that contains 3% (vol/vol)
bromide (CTAB) precipitation have evolved from the work isopentanol. Add 0.1% (wtlvol) hydroxyquinoline to
of Jones (53). Britten et al. (15) were the first to use H A ad- the mixture.
sorption for DNA purification. Ethanol, 80% (vol/vol): 80 parts 100% ethanol plus 20
parts distilled water. Store and use at -20C.
26.2.1. Marmur Procedure RNase A: bovine pancreatic RNase A, 1 mg/ml, dissolved
The Marmur procedure (74) is widely used for isolation of in 0.15 M NaCl (pH 5.0). Heat the solution at 80C for
large amounts of DNA. The bacterial cells are lysed with 10 min to inactivate any traces of DNase. Store in tubes
sodium dodecyl sulfate (SDS) in the presence of sodium containing small portions of the solution at -20C.
chloride, Na-EDTA, and sodium perchlorate. Sodium per- RNase T dissolve RNase TI in 0.02 M Tris-HC1 buffer (pH
chlorate is a chaotropic salt that helps dissociate proteins 6) to a concentration of 200 U/ml.
and polysaccharides from nucleic acids. The proteins are
then extracted with chloroform-isopentanol and discarded,
and the DNA is precipitated with ethanol. After treatment Procedure
with RNase and more chloroform-isopentanol extractions, 1. Harvest the cells by centrifugation. After decanting
the DNA is again precipitated. The isolated DNA has rea- the spent medium, remove any residual medium with a
628 MOLECULAR GENETICS

Pasteur pipette, and dry the inside of the tube with a tissue. mm) by slowly stirring and spinning the rod in the mixture
Suspend the cells in 20 ml of suspending buffer, and place (Fig. 1). Continue the spooling until the two phases are to-
them into a ground-glass-stoppered 250-ml Erlenmeyer tally mixed. Gently press and turn the rod on the wall of the
flask. beaker to squeeze out the remaining ethanol. Wash the rod
2. Add dry lysozyme to the 20-ml cell suspension with with 10 to 20 ml of cold 80% ethanol, invert, and stand the
a measuring spoon (one-eighth teaspoon gives about 2.5 rod in a test tube rack to air dry. Dissolve the DNA by plac-
mglml). Incubate the mixture at room temperature or at ing the rod in screw-cap test tube containing 3 to 5 ml of
37C until the cells become susceptible to lysis by the addi- 0.1X SSC or TE buffer. Usually after about 30 min the
tion of the next set of reagents. For gram-negative organ- DNA can be slipped off the glass rod by gently shaking the
isms, only about 5 min may be needed; gram-positive or- rod up and down (be careful not to hit the bottom of
ganisms may also become susceptible in a few minutes but the test tube at this point; the rod can easily punch through
sometimes require several hours. Remove small samples (0.1 the bottom). Alternatively, place the tube and rod over-
to 0.2 ml) to a small test tube and mix with 1.5 vol of lysing night in a refrigerator to dissolve the DNA.
solution to test for lysis. O n lysis, the cell suspension changes 11. Finally, dissolve the DNA in 3 to 5 ml of 0.1 X SSC
from turbid to opalescent and becomes very viscous. or TE and store in a freezer.
3. Lyse the cells by adding 30 ml of the warm lysing so-
lution. Quickly swirl to uniformly mix the components and Comments
obtain a uniform lysis of the cells. Incubate at 50 to 60C There are many DNA isolation procedures, but the Marmur
for 1 to 4 h. procedure is the best method for most applications. The fol-
4. Add 12 ml of the phenol-chloroform mixture, and lowing are some important variations from the procedure
swirl and shake to obtain a homogeneous mixture. (Shaking originally described by Marmur. (i) The lower-ionic-
may have to be vigorous for a highly viscous lysate.) Shake strength suspension buffer optimizes the efficiency of cell
on a wrist-action shaker for 20 min at a setting that will pro- wall lysis by lysozyme. (ii) The single lysing solution allows
vide vigorous mixing; however, if the phenol-chloroform is for an even distribution of all of the components in what
not first mixed in well by hand, the extraction will be in- will become a very viscous solution. (iii) The proteinase K
complete. digestion helps promotes purification of protein-free DNA.
5. Place the extracted lysate into centrifuge tubes (di- For some organisms, the binding of protein to the DNA can
viding the lysate between two 50-ml straight-wall poly- cause the DNA to be extracted with the proteins. (iv)
propylene tubes works well), and centrifuge for 10 min at Initial protein extraction with phenol-chloroform seems to
17,000 X g. Carefully decant and/or pipette the upper, be more effective than the use of chloroform-isopentanol
aqueous layer from the centrifuge tubes back into a clean alone. (v) The use of 0.6volume of isopropanol for the ini-
Erlenmeyer flask. A n inverted 5-ml pipette placed in a tial DNA precipitation promotes less coprecipitation of
pipetting device or pump works well for removing the vis- polysaccharides with the DNA. In the original procedure,
cous lysate. Discard the phenol-chloroform, bottom layer this selective precipitation step was near the end. (vi)
into an organic waste container. Letting the DNA form a clot permits efficient washing with
6.Add 10 ml of phenol-chloroform mixture to the
lysate, and again shake for 20 min on a wrist-action shaker.
Centrifuge, and collect the upper layer as in step 5. Repeat
this step until there is no protein layer at the aqueous-
chloroform interface. Each time discard the phenol-
chloroform, bottom layer into an organic-waste container.
7. Place the aqueous phase into a 125-ml ground-glass-
stoppered Erlenmeyer flask, and add 0.6 volume of iso-
propanol. Swirl the flask so that the precipitated DNA will
form a clot. Decant the lysate, leaving the DNA clot in the
flask. Add about 20 ml of cold 80% ethanol, swirl, and de-
cant after about 5 min. Repeat this ethanol wash step twice.
After the final ethanol wash, allow the clot to stick to the
bottom of the flask (this can usually be done by slowly tilt-
ing the flask) and then invert the flask on a paper towel to
drain. Air dry for 15 to 30 min in a 37C incubator.
8. Dissolve the DNA by adding 20 ml of 0.1 X SSC to
the flask. After the DNA is completely dissolved, add 0.25
ml of RNase A solution and 2.5 ml of RNase TI, solution.
Incubate at 37C for 30 min.
9. Add 5 ml of chloroform-isopentanol to the DNA so-
lution, and shake on a wrist-action shaker for 20 min.
Centrifuge at 17,000 X g for 10 min. Remove the aqueous
phase with a pipette and save it. If there is a substantial pro-
tein layer, repeat the chloroform-isopentanol step one more
time. Finally, transfer the aqueous phase to an 80- or 100-
ml beaker.
10. Again precipitate the DNA, as follows. Add 2.0 ml
of 3 M sodium acetate to the DNA solution, swirl to mix,
and then overlay the solution with 2 volumes of 95% FIGURE 1 Spooling DNA onto a glass rod during precipita-
ethanol. (Spool the DNA onto a glass rod (20 cm by 7 tion by ethanol. Photo by D.Arbour.
26. Similarity Analysis of DNAs 629

80% ethanol and rapid redissolution in buffer. (vii) In many Chloroform-isopentanol: see section 26.2.1.
cases, the use of a single RNase (RNase A) leaves RNA CTAB dilution-precipitation buffer: 50 mM Tris-HC1, 10
fragments that are still large enough to coprecipitate with mM EDTA, 1% CTAB (wtlvol).
the DNA. Using both RNase A and RNase T reduces this CTAB wash solution: 0.4 M NaC1.
problem.
There have been other variations of the Marmur pro- CTAB-DNA salt dissolving solution: 2.5 M ammonium
cedure reported in the literature, such as dialyzing DNA acetate.
preparations to remove small RNA fragments, replacing SSC: see section 26.2.1.
sodium perchlorate with NaCl(35), and performing an ini- RNase A: see section 26.2.1.
tial alkaline lysis of the cells (8). The major features of the RNase TI: see section 26.2.1.
procedure of Beji et al. (9) are as follows. Phenol-chloroform: see section 26.2.1.
Harvest late-log-phase cells (500 to 600 ml of culture)
and resuspend them in 50 ml of saline-EDTA (150 mM Sodium acetate, 3 M (pH 6.0).
NaC1, 100 mM EDTA [free acid] [pH 8.01). Centrifuge 80% ethanol: see section 26.2.1.
again and determine the wet weight of the cells. Lyse the
cells with 0.03 M NaOH (10 ml/g [wet weight] of cells);
Procedure
leave the cells in contact with NaOH for a maximum of 2
min at room temperature. Add (per gram [wet weight] of 1. Harvest the cells by centrifugation. After decanting
cells) 1.5 ml of 25% (wtlvol) SDS and 35 ml of saline- the spent medium, remove any residual medium with a
EDTA (pH 7). Mix well, and centrifuge at 17,000 X g for 5 Pasteur pipette. Transfer the cell pellet to a tared Braun
min at 4C. After treatment with 2.5 mg of RNase at 60C Disintegrator shaking bottle, and make up total mass to 10
for 30 min and 0.6 mg of proteinase K at 37C for 15 min, g with TE buffer. Add 10 ml of 2X CTAB lysing buffer, 0.2
add 5 M NaCl to increase the NaCl concentration to 1 M. ml of 2-mercaptoethanol, and 20 ml of glass beads (diame-
Precipitate the DNA with 1 volume of 95% ethanol and re- ter range, ca. 100 pm). Place the bottle in the Braun
dissolve in 20 ml of 0.1 X SSC. Add 0.45 M NaC1, and Disintegrator, and shake for 5 min with C02 cooling. The
again precipitate the DNA by adding an equal volume of exact shaking time required depends on the particular or-
isopropanol. Dissolve the final product in 0.1X SSC, and ganism; confirm cell breakage by microscopic observation
dialyze it against 0.1 x SSC. (phase contrast or Gram staining).
2. Separate the lysate from the glass beads by filtration
26.2.2. CetyltrimethylammoniumBromide Lysis through a sintered-glass filter (coarse pore). Filtration is fa-
The CTAB procedure has been used extensively for fungal cilitated by applying a vacuum (a water aspirator can pro-
materials, which contain polysaccharide-like contaminants duce a sufficient vacuum) to a vacuum flask. Use an addi-
that have plagued DNA isolation procedures (80,116). The tional 20 ml of 1X lysing buffer to wash the remaining lysate
procedure described here also works well for actinomycetes away from the glass beads. Add small amounts of buffer at a
and propionibacteria. time, and use a rubber-tipped stirring rod to mix it with the
The cetyltrimethylammonium ion is a cationic deter- glass beads. Avoid using metal or glass stirring rods, as they
gent and cannot be used in conjunction with SDS or phe- will damage the surface of the sintered glass filter.
nol, because these mixtures form very insoluble complexes. 3. To the pooled lysate, add proteinase K to a final con-
DNA is soluble in the presence of CTAB if there is also a centration of 50 pg/ml, and incubate the mixture for 1 to 2
high concentration of monovalent cations such as Naf or h at 60C.
NH,+. 4. Add 15 ml of chloroform-isopentanol to the lysate,
The protocol given here (52) is for recalcitrant organ- shake on a wrist-action shaker for 20 min, and centrifuge at
isms that must be physically disrupted. This disruption re- 17,000 X g for 10 min.
sults in fragmentation of the DNA, and the DNA works 5. Transfer the lysate (upper layer) into a 250-ml cen-
well for reassociation experiments involving only frag- trifuge bottle and add an equal volume of CTAB dilution-
mented DNA, i.e., the optical, HA, and S1 nuclease proce- precipitation buffer. Mix well, and pellet the CTAB-nucleic
dures. However, it may not work well if membrane-bound acid salts by centrifuging at 8,000 x g for 10 min. Wash the
DNA is required for DNA reassociation experiments or precipitate two or three times with 40-ml volumes of cold
rRNA hybridization, and it would not be suitable for re- 0.4 M NaCl wash solution to remove free CTAB. A glass
striction endonuclease digestions. The procedure can be homogenizer works well to disperse the pellet for more effi-
used for the isolation of high-molecular-weight DNA if cient washing.
lysozyme-digested cells (or cells digested by some other lytic 6. Dissolve the CTAB-nucleic acid pellet in 8 ml of 2.5
enzyme) are lysed by the CTAB. The procedure may also be M ammonium acetate. Use a 50-ml screw-cap polypropy-
useful when polysaccharide contamination of DNA prepa- lene centrifuge tube, and warm the material at 50C to fa-
rations is a problem. cilitate dissolution. After the nucleic acids are in solution,
place the tube on ice for 15 min to precipitate the large
Reagents RNAs. Remove the RNA by centrifugation at 17,000 X g
at 4C for 10 min. Transfer the supernatant to another cen-
Cell-suspending buffer (TE buffer): 10 mM Tris-HC1 buffer trifuge tube, and precipitate the remaining nucleic acids
(pH 8.0) containing 1 mM Na-EDTA (pH 8.0). with 2 volumes of 95% ethanol. Collect the precipitate by
CTAB lysing solution: 50 mM Tris-HC1 buffer (pH 8.0),0.7 centrifugation at 17,000 X g for 10 min.
M NaC1, 10 mM EDTA, 1% CTAB (wtlvol), 1% (vol/ 7. Dissolve the pellet in 10 ml of 0.1x SSC, and add
vol) 2-mercaptoethanol. 2 X and 5 X CTAB lysing solu- 0.25 ml of RNase A and 2.5 ml of RNase TI. Incubate at
tions can be prepared by using two and five times the 37C for 1 h.
concentrations of all of the components, respectively. 8. Extract the digest once with 5 ml of phenol-chloro-
Proteinase K: see section 26.2.1. form. After centrifugation, add 1 ml of 3 M sodium acetate
630 MOLECULAR GENETICS

to the extracted digest, and precipitate by adding 2 volumes Carefully draw off the upper (aqueous) layer (see step 5 of
of 95% ethanol. Collect the precipitate by centrifugation, section 26.2.1) and return it to the flask.
and wash the pellet with 10 ml of 80% ethanol. 6. Repeat steps 4 and 5.
9. Dissolve the DNA in 3 to 5 ml of 0.1 X SSC or TE and 7. After again returning the aqueous phase to the flask,
store in a freezer. add 2.0 ml of 1.0 M PB. Then, add 2 g (1 measuring tea-
spoon is convenient and sufficiently accurate) of dry HA.
Comments Suspend well, and gently shake on a rotary or reciprocal
The procedure described above can be used for organisms shaker for 1 h at a speed sufficient to keep the H A from set-
that are difficult to lyse and have high levels of nuclease ac- tling out.
tivity. A technical difficulty is that the detergent causes 8. Transfer the suspension to a 50-ml polypropylene
foaming and tends to reduce the velocity of the beads, centrifuge tube, and centrifuge for 2 to 3 min at 5,000 X g
thereby reducing their kinetic energy. Depending on the or- at room temperature. Return the supernatant layer (lysate)
ganism, this may result in inefficient cell breakage. If nu- to the flask (this can be used for a second DNA adsorption
clease activity is not a problem, suspend and disrupt the cycle), and use the sedimented H A (to which DNA is now
cells in TE buffer (it may be helpful to increase the EDTA adsorbed) in step 9.
concentration to 10 mM) and add the RNase at this time. 9. To the sedimented HA, add 8 ml of 0.10 M PB.
Then, remove the glass beads by filtration and add 0.25 vol- Suspend the HA with the aid of a Vortex mixer.
ume of 5 X CTAB lysing solution. Then proceed to step 3 of Immediately add an additional 24 ml of the PB (an auto-
the procedure. matic pipettor works well for these additions). The PB
If cell lysis is not difficult and the main reason for using should be added with some force, so that the HA will be-
this procedure is to eliminate contaminating polysaccha- come evenly mixed for maximum dilution of nucleotides
rides, one can start as if doing the Marmur procedure but and phenol. Allow the H A to settle for 1 to 2 min, and then
then lyse cells with 2X CTAB lysing solution. centrifuge for 2 to 3 min at 5,000 X g. Discard the super-
natant.
26.2.3. Hydroxylapatite Adsorption 10. Repeat step 9 six or seven times or until the ab-
Britten et al. (15) were the first to isolate DNA by adsorb- sorbance at 270 nm (or A270)of the supernatant is less than
ing it to HA. Since then, several modifications of the 0.05(the wavelength of maximum absorption by phenol).
method have been described (2, 49, 73) including the fol- 11. Suspend the H A in 5.0 ml of 0.5 M PB to desorb the
lowing adaptation. DNA. Centrifuge as before, but this time save the DNA-
containing supernatant.
Reagents 12. If most of the DNA has not been removed from the
Cell-suspending buffer (TE buffer): see section 26.2.2. lysate in step 8, wash the H A from step 11 once with dis-
SDS: see section 26.2.1. tilled water, and then add it to the lysate for a second ad-
sorption cycle. Alternatively, fresh HA may be used. Add
RNase A: see section 26.2.1. the DNA obtained from a second adsorption cycle to that
RNase TI:see section 26.2.1. from the first cycle.
Phenol: water-saturated phenol containing 0.1 % (wtlvol) 13. Filter the DNA preparation through a fiberglass fil-
8-hydroxyquinoline. ter (2.4-cm diameter) in a syringe-type filter holder to re-
HA, DNA grade Bio-Gel HTP (Bio-Rad Laboratories, move any remaining HA particles.
Richmond, CA) . 14. Dialyze the DNA preparation against TE buffer (pH
Phosphate buffer (PB), 1.0 M (pH 6.8) (prepare by mixing 8.0). Cut 13- to 15-cm lengths of dialysis tubing and wash
equal volumes of 1 M Na2HP04 and 1 M NaH2P04). out UV-absorbing materials by boiling the tubing in a 2 to
5% solution of sodium carbonate for a few minutes, thor-
This is used for preparing the lower concentrations as in-
dicated in the text. oughly rinsing the lengths of tubing under running tap
water, and then rinsing them under distilled water. The tub-
ing can be stored in a refrigerator for 2 to 3 days in distilled
Procedure
water. Cellulolytic organisms may hydrolyze the tubing if it
1. Suspend the centrifuged cells in 25 ml of TE buffer, is stored for a longer time. Seal one end of the tubing length
add lysozyme or other lytic enzyme, and incubate until the by tying a knot or using a dialysis tubing clamp, pour in the
cells are susceptible to detergent lysis (see section 26.2.1, DNA solution, and seal the other end of the tubing. Attach
step 2). Add 1.5 ml of the SDS solution. a string to one end, and wrap a piece of tape around the
2. Add 0.25 ml of RNase A and 2.5 ml of RNase TI. string for a convenient label. Dialyze in a 400- to 500-
Swirl the flask and warm in a 50 to 60C water bath until volume excess of the buffer for about 3 h, change the buffer,
lysis is complete. For bacteria not disrupted by detergent and continue dialyzing overnight. Store the DNA prepara-
alone, see Comments, section 26.2.2, for alternate methods. tion in a freezer, or add a few drops of chloroform and store
3. Reduce the viscosity of the lysate by briefly subject- in a refrigerator.
ing it to sonic oscillation or by use of a mechanical tissue
homogenizer. Proteinase K may be added at this time (50 Comments
Fg/ml). For some organisms proteinase digestion is not If the lysate is digested with proteinase K, a single phenol
needed, but for others it is essential. If the proteinase is extraction will usually be sufficient.
added, incubate it with the lysate at 50C for 1 h. A n important feature of the H A procedure is that RNA
4. Add 7 ml of water-saturated phenol, shake by hand is degraded to such an extent that it will not compete with
to get the two phases well mixed, and then shake the flask DNA for adsorption sites on the HA. For some groups of or-
on a wrist-action shaker for 20 min. ganisms, the RNase is inhibited when added in step 2. In
5. Centrifuge at 17,000 X g in a polypropylene cen- such cases, dialyzing the lysate against saline-EDTA buffer
trifuge tube, using a refrigerated centrifuge at 0 to 4C. for several hours removes the inhibitory effect.
26. Similarity Analysis of DNAs 631

The major contaminant in the DNA preparations, other DNA. A preparation of pure native DNA increases in A260
than RNA, is polysaccharide. Many polysaccharides do not by approximately 40% (Fig. 2) when denatured. If the sec-
adsorb to HA and therefore are easily separated from DNA ondary structure of the contaminating RNA has been de-
by this procedure. Others, however, can bind to HA and in- stroyed by RNase, the RNA will have no hyperchromic
hibit DNA adsorption. shift. If the secondary structure is intact, the RNA will have
In some cases a more dilute cell suspension of some or- a hyperchromic shift of approximately 30%. The hyper-
ganisms may be required to improve the efficiency of this chromic shift for RNA occurs over a lower and wider tem-
method. Dilution appears to promote more complete lysis perature range than for DNA, so the profiles for RNA and
and/or RNA degradation. Scale up the volumes four or five DNA can be easily distinguished. Therefore, to estimate the
times, and follow the above steps until the HA adsorption. A260 of a nucleic acid preparation that is due only to the
After adding the HA to part of the lysate, pour the HA sus- DNA, divide the hyperchromic shift due to DNA by 0.4.
pension into a 60-ml sintered-glass filter. The HA will For example, assume that the A260 of a sample (or a dilution
quickly settle, and the lysate will flow through the HA as of it) is 1.25 and that the change in A260 as a result of de-
through a column, resulting in more efficient adsorption. naturation is 0.4. The calculated DNA A260 would be
Transfer the HA to a centrifuge tube, and complete the pro- 0.4/0.4, or 1.00 (i.e., 1/1.25 or 80% of the A260 due to
cedure as described above. DNA).
26.3.3. Colorimetric Method
26.3. D N A MEASUREMENT (also see chapter (Diphenylamine Reaction)
18.6) Many colorimetric assays have been developed and modi-
fied over the years for the measurement of DNA (8, 16, 17,
26.3.1. UV Spectrophotometry 39, 93, 112). Although they can be used for measuring pu-
The molar extinction coefficient of native DNA is in the rified DNA, these procedures were often designed for mea-
range of 6,650 to 6,700 (51, 68). Conversion of the DNA suring DNA in tissue material. They have the disadvantage
concentration from molarity to milligrams per milliliter of using high concentrations of mineral acids and of requir-
yields the following formula, for cuvettes with a 1-cm light ing long periods for color development.
path. For native (double-stranded) DNA, the formula is The diphenylamine reaction is specific for the deoxyri-
Concentration (mglml) = A260/20 (1) bose in DNA. Although the Burton procedure (16) has
For denatured (single-stranded) DNA and for RNA, the
formula is
Concentration (mglml) = A260/23 (2)
In practice, most stock DNA preparations range from 0.5
to 2 mg of DNA per ml. It is convenient to dilute them 20-
fold (0.1 ml of DNA solution added to 1.9 ml of buffer) be-
cause the resulting A260 can be read directly as milligrams of
DNA per milliliter of the stock preparation (see equation 1).
UV spectrophotometry has also been used for detecting
contaminating materials (5 1). (i) Proteins absorb UV light
more strongly than nucleic acid polymers at 280 nm. The
A260/A280 ratio of a nucleic acid solution is used as an indi-
cator of protein contamination. The extinction coefficient
for protein, however, is very low compared with that for nu-
cleic acids; consequently, the A260/A280 ratio is not a very
sensitive indicator. (ii) The absorption spectrum for nucleic
acids has maxima at approximately 208 and 260 nm and a
minimum at 234 nm. The peak at 208 nm is not specific for
nucleic acids, because most compounds absorb light of that
wavelength. If contaminating material is present in a nu-
cleic acid preparation and contributes to a large peak at 208
nm, this peak will overlap the 234-nm minimum for the nu-
cleic acid. Therefore, the A260/A234 ratio is a more sensitive
indicator of contaminating material in a DNA preparation
than the A260/A280 ratio. (iii) The absorption maximum of
phenol is 270 nm; consequently, the &60/A270 ratio is quite
sensitive for detecting phenol contamination in a nucleic
acid preparation.

26.3.2. Hyperchromic Shift FIGURE 2 Hyperchromic shift tracings for two samples of
The major contaminant of DNA preparations is usually DNA in an automatic recording spectrophotometer. The
RNA, and unfortunately, the two nucleic acids cannot be points at which any vertical line crosses the absorbance and
differentiated directly by their UV absorbance. However, temperature curves give values for these two parameters at a
the hyperchromic shift (absorbance change) that occurs given time. The inflection points are estimated at one-half of
when DNA is thermally denatured (see section 26.4.1) can the hyperchromatic shift, and the temperature at that time is
be used to estimate the fraction of the preparation that is the melting temperature (T,).
632 w MOLECULAR GENETICS

often been used, the following variation of the more sensi- 2. Dilute the test DNA preparations so that they are
tive Giles and Meyers modification (37) is preferable. within the same range.
3. Add 0.1 ml of the mithramycin stock solution to the
1. Place 1.0 ml of the DNA sample (5 to 50 pg) in a 16-
standard and test DNA dilutions, mix, and determine the
by 125-mm screw-cap tube and add 1.0 ml of 20% (wtlvol) fluorescence (emission) at 540 nm with an excitation wave-
perchloric acid.
length of 440 nm.
2. Add 2.0 ml of glacial acetic acid containing 4%
(wtlvol) diphenylamine.
4. Plot a standard curve from the values obtained for the
DNA standards, and use it to estimate the concentrations of
3. Add 0.2 ml of 0.16% (wtlvol) solution of acetalde-
the test DNA samples.
hyde.
4. Mix and incubate overnight at 30C. 2 6.3.4.2. 4',6'-Diamidino-2-Phenyl indole.2 HCI
5. Read the A595 and A700, and subtract the A700 from
the A595. Construct a standard curve, using known amounts DAPI (4',6'-diamidino-2-phenylindole* 2HC1) forms a
(5 to 50 pg) of a pure standard DNA (calf thymus or salmon specific fluorescent complex with DNA, but the manner by
sperm DNA; Sigma Chemical Co.), and plot the microgram which it binds (i.e., by intercalation, or by a nonintercalat-
amount of DNA against the difference (A595 - A700). ing mode) is uncertain (67). DAPI does exhibit a prefer-
Determine the concentration of DNA in the sample from ence for several continuous A-T base pairs (56). The dye is
the standard curve. very stable and is specific for DNA. I t has been used to mea-
sure DNA directly in fixed cells of Escherichia coli and might
The diphenylamine procedure can also be used for quan- be similarly useful with other bacteria.
tifying the DNA bound to nitrocellulose or nylon mem-
branes in hybridization studies. It is particularly useful when Reagents
one cannot readily measure the amount of DNA being DAPI stock solution: 3 pg/ml in distilled water. This stock
bound to a filter (for an example, see section 26.6.3). After solution is stable at 4C for months.
the amounts of probe (radioactive or nonradioactive)
bound to the membranes have been measured (use a non-
Dilution buffer: 10 mM NaC1. 6.6 mM Na7SOd. * 5 mM
, I

aqueous counting solution so that the membranes will not N'-2-hydroxyethylpiperazine-N'-2-ethanesulfonicacid


be dissolved or the DNA eluted), dry the membranes and (HEPES; pH 7.0).
place each membrane in 2.0 ml of 10% perchloric acid (du- Toluene-fixed cells. Mix samples of bacteria with toluene
plicate membranes can be placed together in the same (l%,vol/vol), and shake vigorously. Store at 4C.
tube). Heat in a boiling-water bath for 5 min. After cooling
to room temperature, proceed with step 2 above. Procedure

26.3.4. Fluorimetric Dye-Binding Methods 1. Dilute a sample of a DNA standard in the dilution
buffer to give concentrations ranging from 5 ng/ml to 1.0
Fluorescent compounds, usually dyes, have been used for pg/ml in a final volume of 3.0 ml.
characterizing and quantitating DNA. After interacting 2. Similarly, dilute the fixed bacterial cells in the dilu-
with the DNA, the resulting complex causes an enhance- tion buffer to a final turbidity of 0.008 to 0.08 at 600 nm
ment of the fluorescence. The complexes are very specific, (turbidity values may differ from one spectrophotometer to
in that molecules other than DNA (such as RNA, proteins, another; the only important requirement is that the fluores-
and carbohydrates) seldom interfere with assays. A major cence values for DAPI [see step 31 be linear within the tur-
use of these procedures is the measurement of DNA in tis- bidity range used).
sue homogenates. Certain dyes may complex specifically 3. Add 100 p l of DAPI stock solution to each tube, mix,
with high-A+T or high-G+C regions of the DNA, and measure the fluorescence intensity with excitation at
whereas the binding of others is independent of the base 350 nm and emission at 450 nm. (The mixtures are stable
composition. As a result, the moles percent G + C content at 4C for several weeks.) Plot a standard curve from the
of the DNA may have to be taken into account when con- values obtained for the DNA standard and use it to estimate
structing standard curves, and in some cases differential the concentrations of the test DNA samples.
binding of two dyes can be used for estimating this value.
The following are some of the more commonly used fluori- 26.3.4.3. Bisbenzimide (Hoechst 33258)
metric methods for measuring DNA.
The Hoechst 33258 dye is probably the most widely used of
26.3.4.1. Mithramycin DNA-binding dyes (20, 29, 62, 82, 83, 99, 100). The dye
binds specifically to regions rich in A-T base pairs in the
The antibiotic mithramycin binds to double-stranded
large groove of the DNA helix (99). Consequently, the
DNA, and the amount of fluorescence is directly propor-
DNA used for the standard curve should have a moles per-
tional to the amount of DNA (42). The antibiotic binds to
cent G + C content similar to that of the test DNAs.
a lesser extent to denatured DNA and does not bind to
DNase-treated DNA or to RNA. Reagents
Reagent 20X Phosphate-buffered SSC (PB-SSC): 20X SSC (see
Prepare a stock solution containing 200 pg of mithramycin section 26.2.1) prepared in 200 mM sodium phosphate
per ml in 300 mM MgC12. buffer (pH 7.0).
Hoechst 33258 stock solution: 2 pg/ml in 1 X PB-%SC.
Procedure
(This is equivalent to a concentration of 3.7 X 10- M;
1. Dilute a sample of a DNA standard (a preparation the molecular weight of the dye is 533.89.) The dye con-
containing no RNA, with the concentration determined by centration can be measured with a spectrophotometer at
UV absorbance) with distilled water to give amounts rang- 338 nm; the molar extinction coefficient is 4.2 X lo4
ing from 0.4 to 32 pg in a final volume of 1.9 ml. M-' (103).
26. Similarity Analysis of DNAs 633

Procedure DNA preparations by Marmur and Doty (75) (Fig. 2). The
method has been reexamined by several investigators, and
1. Dilute samples of a DNA standard (a preparation con-
several equations correlating the T, with the base composi-
taining no RNA, with the concentration determined by
tion have been proposed. With the exception of the equa-
UV absorbance) in l x PB-SSC to give amounts ranging
tion proposed by Mandel et al. (72), which appears to have
from 0.01 to 10 p,g in final volumes of 2.0 ml.
a different slope (Z), all are similar to the one proposed by
2. Dilute the test DNA preparations so that they are Marmur and Doty. The T, values are greatly affected by the
within the same range.
ionic strength of the buffer used, and this effect is the same on
3. Add 2 ml of dye solution to each tube of DNA, mix, all DNA preparations. Consequently, one must either know
and measure the fluorescence intensity with excitation at
the ionic strength of the buffer that is used or include a ref-
360 nm and emission at 450 nm. Plot a standard curve from
erence DNA preparation at the same ionic strength as that
the values obtained for the DNA standard and use it to es-
used for the unknown samples. The latter is easy to do: one
timate the concentrations of the test DNA samples.
merely dialyzes all of the DNA preparations (including the
reference DNA) together in a single batch of buffer. Al-
26.3.4.4. Ethidium Bromide ternatively, if the DNA preparations are all dissolved in a
The intercalating dye ethidium bromide binds to double- common buffer, they can be diluted with the same buffer
stranded DNA with little dependence on the base composi- and tested directly.
tion. Although it binds to a lesser extent to intact RNA,
error from contaminating RNA can be eliminated by treat- Procedure
ing the DNA preparation with RNase (57,67, 82, 83). The following procedure is reliable and may be adapted to
other buffers or other buffer concentrations than those
Reagents given.
20X PB-SSC: see section 26.3.4.3.
1. Prepare 2 to 4 liters of 0.5X SSC (section 26.2.1) at
Ethidium bromide. Prepare a working solution of 2 p,g/ml in pH 7.0. Save some of the buffer for diluting the DNA prepa-
2X PB-SSC. rations and for rinsing out cuvettes just prior to putting the
DNA samples into them. Divide the rest into two equal
Procedure parts so that the buffer can be changed once during dialysis
1. Dilute samples of a DNA standard (a preparation con- (see step 3).
taining no RNA and the concentration determined by UV 2. Prepare 2- to 5-ml samples (depending on the size of
absorbance) in 2x PB-SSC to give amounts ranging from the cuvettes to be used) of DNA at 50 p,g/ml, using 0.5X
0.01 to 10 pg in final volumes of 2.0 ml. SSC to dilute the stock DNA preparations. Prepare a 5- to
2. Dilute the test DNA preparations so that they are 10-ml volume of the reference DNA (i.e., the DNA of E.
within the same range. coli b has a G + C content of 51 mol% and a T, of 90.5"C
3. Add 2 ml of dye solution to each tube, mix, and mea- in 1x SSC),because it will have to be included in each in-
sure the fluorescence intensity with excitation at 305 nm strument run.
and emission at 615 nm. Plot a standard curve from the val- 3. Dialyze all of the preparations together overnight in
ues obtained for the DNA standard, and use it to estimate the 0.5X SSC. Change the buffer once after the first few
the concentrations of the test DNA samples. hours of dialysis. After dialysis, return the DNA prepara-
tions to screw-cap test tubes.
4. Determine the melting profile with an automatic
26.4. G UANlNE-PLUS-CYTOSINE recording spectrophotometer having a cuvette holder that
CONTENT OF DNA is heated electronically. Start at 60C, and increase the
The moles percent G+C content of DNA can be estimated temperature linearly at 0.5 or 1.OC per min over the entire
by several different methods; see reference 1 for a review. range.
The methods described in detail here use thermal denatura- 5. Determine the T, values as in Fig. 2. Calculate the
tion, high-performance liquid chromatography (HPLC), or moles percent G + C of the sample DNA by the following
dye-binding fluorimetry. Although buoyant-density cen- equation:
trifugation has also been used for many moles percent G + C mol% G + C = (T, + [90.5 - Tm(ref)]
- 69.3)/0.41
estimations reported in the literature, there are few if any
analytical ultracentrifuges still functioning. All of these This is the Marmur and Doty equation, modified to nor-
methods require the prior isolation and purification of malize the T, values to those in SSC.
DNA.
Estimation of the moles percent G+C directly in intact Comments
whole cells is often desirable, especially in characterizing The thermal stability of double-stranded DNA is highly de-
newly isolated bacteria in ecological and systematics studies or pendent on the ionic strength of the buffer in which it is
bacteria whose DNA is difficult to extract or purify. One of dissolved. If the DNA preparations have been spooled out
the dye-binding fluorimetric methods described below (sec- of a sodium acetate-ethanol mixture and the spooled DNA
tion 26.4.3.2) can be used with formalin-fixed whole cells. A has been rinsed with 80% ethanol (i.e., there is little or no
calorimetric method involving native intact cells is also de- residual salt in the DNA preparations), and if the DNA
scribed (section 26.4.4); this method requires expensive spe- preparations have been dissolved in 0.1 x SSC, they can be
cialized instrumentation but can be performed quickly. diluted in a single batch of 0.1x SSC and not dialyzed.
Rinsing the cuvettes can be a cause of error if residual water
26.4.1. Thermal Melting Method is allowed to dilute the buffer. If rinse water remains in the
The midpoint temperatures of the thermal melting profiles cuvettes, give them a final rinse with the T, buffer prior to
(T,) were first correlated with the base composition of use.
634 MOLECULAR GENETICS

26.4.2. High-Performance Liquid Chromatography dC dG dA


(also see chapter 17.3.6)
The only direct approach for measuring the base composi-
tion of DNA is to hydrolyze it and quantify the individual
nucleosides or nucleotides. Originally this was done by
using paper chromatography to separate the nucleosides or
nucleotides, which were then eluted and measured with a
spectrophotometer. Many other methods have been based
on correlations with these early results. However, because of
the difficulty in quantifying these chromatographic analy-
ses, few have been done since the early 1960s. Recent ad-
vances in HPLC have enabled investigators to determine
the base composition of DNA more rapidly and accurately
(see reference 2 for a review). The procedure described here
is that of Mesbah et al. (78, 79), although similar proce-
dures have been described by others (61, 102). This proce- w
dure includes degrading denatured DNA with the single- 0
strand-specific nuclease PI from Penicillium citn'num. The z
resulting 5' nucleotides are then dephosphorylated with al-
kaline phosphatase, and the resulting nucleosides are sepa-
8
[r
rated on a reverse-phase C18 column. This method has a 0
cn I I
number of advantages. One, because of the high resolution m 0 10
of modern columns, ribo- and deoxyribonucleosides can be a
easily separated, and contamination with up to 50% RNA C
does not interfere (Fig. 3). Two, by limiting the analyses to
deoxyguanosine (dGuo) and thymidine (dThd), the effects
of base methylations and other modifications (which are
common for deoxycytidine and deoxyadenosine) are negli-
gible. Three, if the moles percent G+C is estimated from
the ratio of dGuo and dThd, it is also possible to obtain a
very high precision of 0.1 mol% with standard equipment.
Lastly, much of the analysis can be performed automatically
with an autosampler. L I

0 10
Instrumentation Requirements
The HPLC setup includes a pump that will produce pulse- TIME (minutes)
free isocratic separations of the nucleosides, an injector, a FIGURE 3 Separation of deoxynucleosides from the ribonu,
fixed-wavelength (254-nm) UV detector, and an integrator- cleosides by HPLC for determination of the moles percent
recorder. The column is a CISreverse-phase column, with +
G C. (A) Separation of ribonucleosides from deoxyribonucle-
5-pm particle size and column dimensions of 250 by 4.6 osides at 24C in 12% methanol on a CI8 reverse-phase col-
mm, or any comparable CIBcolumn. Because of the sensi- umn. The nucleosides were obtained from enzymatic degrada-
tivity of peak shape to temperature, the column tempera- tion of Methanococcus voltae nucleic acid. (B) Separation of
ture should be carefully maintained with a column oven or nucleosides under the optimal conditions for resolution of de-
water jacket. oxyguanosine (dG) and thymidine (dT), which were 38C and
Reagents 12% methanol for this particular column. Under these condi-
tions, guanosine (G) and 5-methyldeoxycytidine (mC) elute as
Stock triethylamine PB, 0.5 M. Prepare by adjusting the pH minor peaks just prior to dG and deoxycytidine (dC) and uri-
of a solution of 0.5 M triethylamine to 5.1 with 85% dine (U) are not resolved. The nucleoside mixture was pre-
phosphoric acid. For preparing this buffer, the triethyl- pared from standards. (C) Separation of nucleosides from the
amine should be nearly colorless. If yellow, it should be nucleotide monophosphates. The chromatography was per-
redistilled before use. formed as for panel B. The nucleoside mixture contained 2% of
Chromatographic buffer: 20 mM triethylamine phosphate the nucleotide monophosphates. Under these conditions, the
buffer (pH 5.1) containing about 8 to 12% (vol/vol) nucleotides appear as minor peaks near the nucleosides.
HPLC grade methanol. Before using, filter the buffer Possible products of incomplete degradation, the nucleotides
through a cellulose triacetate membrane filter with a can interfer with the determination of dG and dT.
pore size of 0.45 pm. The actual methanol concentra- Abbreviations: A, adenosine; dA, deoxyadenosine; dAMP, de-
tion of the buffer depends greatly on the individual col- oxyadenosine monophosphate; C, cytidine; dC, deoxycytidine;
umn and the analysis temperature, and it is determined mC, 5-methyldeoxycytidine; G, guanosine; dG, deoxyguano-
empirically. sine; dGMP, deoxyguanosine monophosphate; dT, thymidine;
P1 nuclease: the commercial enzyme is diluted to 340 U/ml U, uridine; X, unidentified compound. From reference 79.
in 30 mM sodium acetate (pH 5.3). Portions of 50 pl are
then stored in microfuge tubes at -20C for up to 4
months. (pH 7.6) or any comparable commercial preparation.
Calf intestinal alkaline phosphatase: 20 to 30 U/pl dis- Before using, dilute to 0.1 U/pl with alkaline phos-
solved in a solution consisting of 30 mM trieth- phatase buffer (see below).
anolamine, 3 M NaC1, 1 mM MgC1, and 0.1 mM ZnClz Sodium acetate buffer: 300 mM, pH adjusted to pH 5.3.
Next Page

26. Similarity Analysis of DNAs w 635

ZnS04 solution, 20 mM. rate of 1 ml/min at the beginning of the week. A t night or
Alkaline phosphatase buffer: 0.1 M glycine-HC1 (pH 10.4). during breaks in the analyses, the flow rate is decreased to
TE buffer: see section 26.2.2. 0.1 to 0.2 ml/min. When analyses are complete, the column
is washed sequentially with water and storage buffer (such
DNA standard: calf thymus DNA, 1 mg/ml in TE buffer, is as 70% methanol in water) prior to storage. Because of the
an inexpensive standard. Using the method described salts in the buffer, the column requires a continuous flow of
below, its moles percent G + C has been found to be buffer between uses. Moreover, the buffer is always slowly
44.03% t 0.05%,and the 5-methyldeoxycytosine con- mixed on a magnetic stir plate during the chromatography
tent is 1.0 rf: 0.1 mol%. Unmethylated lambda phage to prevent the formation of precipitates.
DNA can also be used. Because it has been sequenced, When the chromatographic conditions are known, in-
the exact moles percent G + C is known to be 49.858 ject a control sample prepared with the enzymes and buffers
mol%. but without DNA. Look for minor peaks that elute at the
same times as deoxyguanosine and thymidine. This control
Degradation Procedure tests for interference by contaminants that could adversely
1. Add standard and unknown DNAs, 2 to 25 pg, to 1.5- affect the analyses. Also inject a representative DNA sam-
ml microfuge tubes in a final volume of 70 pl of water or di- ple and allow the chromatography to run for 60 min. Check
lute TE buffer. For highly purified DNA, use 10 pg. Increase the chromatogram to ensure that no minor peaks are elut-
the amount of nucleic acid for samples believed to contain ing after the major deoxynucleosides. Program the auto-
large amounts of RNA. Denature the DNA by heating in a injector to load samples after the last peak in the chro-
boiling-water bath for 2 min, and then rapidly cool it by matogram, usually every 15 to 30 min.
placing it in an ice bath. Typically, the column is equilibrated for several hours at
2. Add 13 p1 of the P1 mix. This mix is composed of 50 a flow rate of 1 ml/min and the running temperature prior
p1 of 0.3 M sodium acetate buffer (pH 5.3), 50 p l of 20 mM to the analysis. During this time, 10 p l of each sample is an-
ZnS04 solution, and 30 pl of diluted P1 enzyme. Shake gen- alyzed once to establish the best injection volume. The in-
tly to mix, centrifuge for 5 s to collect the liquid at the bot- jection volume is then adjusted so that nucleosides from
tom of the tube. Incubate the mixture at 37C for 2 h. about 1 pg of DNA are injected. If the DNA sample is
3. Add 10 pl of alkaline phosphatase diluted in buffer to below about 0.2 pg, the integration will not be accurate. If
0.1 U/pl. Incubate at 37C for 6 h or overnight. the DNA sample is too large, the nucleosides will not be
4. Centrifuge the sample in a microcentrifuge for 5 min, well resolved on the column. The injection volume must be
transfer to a clean tube, and store at -20C until chro- between 5 and 40 pl. When the column is equilibrated,
matographed. Samples should be stored for no more than 1 samples are run in duplicate or triplicate, with a standard
week after the degradation and before the analyses. DNA run after every four or five samples.

Calculation
Chromatographic Procedure The moles percent G + C is calculated from the ratios of de-
Because the determination of the moles percent G + C is oxyguanosine and thymidine of the unknown and the stan-
based upon the ratio of deoxyguanosine and thymidine, dard DNAs:
only these two nucleosides need to be well resolved during
the chromatography. Ordinarily, all the major deoxynucle-
mol% G+C = [l + YT,,,/G,,,] -'
osides are well separated, but coelution of guanosine, 5- where Tap,is the integrator response for thymidine and Gap,
methyldeoxycytidine, and deoxyguanosine is frequently a is the integrator response for deoxyguanosine of the un-
problem, and the chromatography buffer needs to be opti- known DNA. Y = (Gapp/Tapp)x (T,,JG,,,) for the stan-
mized for the separation of these compounds. In general, dard DNA, where T,,, is the actual thymidine content and
these compounds migrate more closely as the methanol G,,, is the actual deoxyguanosine content. For calf thymus
concentration and temperature increase (79). However, the DNA, TaCt/G,,, is 1.2712.
effect of temperature is offset somewhat by the smaller peak
width at higher temperatures. Comments
In any case, it is necessary to establish the precise chro- A common error in this procedure is incomplete degradation
matographic conditions empirically when using a new type of the DNA caused by contaminating salts. Salts change the
of column. Since calf thymus DNA contains about 1 mol% pH optimum of the P1 nuclease and greatly reduce the ac-
of 5-methyldeoxycytidine and small amounts of RNA con- tivity at pH 5.3 (78). Moreover, contaminating buffers can
taminate the commercial preparations, this standard is suit- maintain the pH near neutrality when the alkaline phos-
able for testing the chromatography conditions. After the phatase buffer is added and inhibit that reaction as well.
degradation of the calf thymus DNA, perform the chro- These problems can be avoided by dialyzing the DNA
matography at 35C and 10% methanol. Look for the ap- against water, 0.01X SSC or TE buffer prior to the degrada-
pearance of two small peaks eluting just before the de- tion reaction. For this reason, it is also important to perform
oxyguanosine peak (Fig. 3 ) . These peaks can also be replicate degradation reactions and analyze each digestion in
identified by coelution with authentic standands. If both duplicate or triplicate. If the digestion reactions have gone
peaks are not observed, decrease the methanol concentra- to completion, they will not be significantly different.
tion by dilution with the aqueous components of the buffer, The precision of this method is generally better than 0.1
increase the temperature, or both. Be sure to allow suffi- mol%. Most of this precision comes from calculating the
cient time for the column to reequilibrate under the new moles percent from the deoxyguanosine/thymidine ratio be-
conditions (at least 60 min) prior to the chromatography. cause it allows one to neglect differences in injection vol-
Because of the sensitivity of the chromatography to umes and preparation of standards. However, the method is
small changes in the methanol concentration, large sensitive to systematic errors because of incomplete degra-
amounts of buffer sufficient to complete the entire analysis dations, contaminants that coelute with the deoxynucleo-
are prepared. Typically, the column is equilibrated at a flow sides, and changes in the chromatography. Even then, the
Nucleic Acid Analysis
WILLIAM HENDRICKSON AND DON WALTHERS

27.1. DNA SEQUENCING ....................................... 653


27.1.1. Manual DNA Sequencing ............................... 654
27.1.1.1. DNA Polymerases ............................ 654
.
2. DNA Templates .............................. 655
.
3. Manual DNA Sequencing Reactions ...............656
. 4. Gel Electrophoresis ...........................
27.1.2. Automated DNA Sequencing ............................
656
658
27.1.2.1. Overview .................................. 658
.2. Interpreting Chromatograms ..................... 658
.3. Troubleshooting .............................. 659
27.2. DNA-PROTEIN BINDING .................................. 659
27.2.1. Gel Mobility Shift Assay ............................... 659
27.2.1.1. DNA Fragments ............................. 660
.2. Gels and Buffers ............................. 660
.3. DNA Labeling ............................... 660
.4. Gel Preparation .............................. 661
.5. Binding Reactions and Electrophoresis .............. 661
.6. Protein Concentration ......................... 662
.7. Equilibrium Binding Constant .................... 663
.8. Kinetics .................................... 664
..19.0. Stoichiometry ............................... 664
Assay with Crude Protein ...................... 665
27.2.2. DNA Protection (Footprinting) Assays ..................... 665
27.2.2.1. DNase I Protection Assay ...................... 666
.2. DMS Protection Assay ......................... 667
.3. FeBABE Footprinting .......................... 668
27.3.
.4. ChIP Assay ................................. 668
RNA TRANSCRIPT MAPPING .............................. 670
27.3.1. S1 Nuclease Protection Assay ............................ 670
27.3.1.1. Crude-RNA Isolation .......................... 670
.2. Pure-RNA Isolation ........................... 670
.3. Assay Procedure ............................. 671
27.3.2. Primer Extension Assay ................................ 672
.3. 27.3.3.1.
Northern Blot Hybridization Assay ........................
Cell Lysate RNA Preparation ....................
673
673
.2. Assay Procedure ............................. 673
27.4. REFERENCES ............................................ 674

This chapter describes several of the most common nucleic a Northern blot hybridization. also described in section
acid analyses performed in vitro to characterize a cloned 27.3. Both the S1 protection and Northern (RNA) blot
DNA segment carrying a gene or genes. However. Southern hybridization methods provide information on the abun-
hybridization. which is done to locate a cloned DNA seg- dance of mRNA for studies of gene regulation.
ment within a physical map. is described in chapter
29.2.3.2 and will not be discussed here. DNA sequencing.
described in section 27.1, is essential for many other meth- 27.1. DNA SEQUENCING
ods and is often one of the first procedures done with a The most common method used to determine the sequence
cloned DNA segment. Section 27.2 describes the gel mo- of nucleotides in a DNA fragment (50) is based on the gen-
bility shift assay. DNase protection and chemical protec- eration of sequencing ladders by using a DNA oligomer as
tion (footprinting) assays. and an immunoprecipitation a primer. a double-stranded target DNA segment as the
assay used to locate DNA-binding sites for regulatory pro- template. various types of DNA polymerase. deoxynucleo-
teins at promoters and in control regions of genes. The pre- side triphosphates (dNTPs). and 2.3 .dideoxynucleoside
cise 5 end of the mRNA can be located by S1 nuclease triphosphates (ddNTPs). The polymerase extends the
protection or by primer extension methods. as described in primer from the 5 to the 3 direction. The ddNTPs are
section 27.3. The size of an mRNA is determined by doing analogs of dNTPs. but they lack a hydroxyl residue at the

653
654 m MOLECULAR GENETICS

3 position of deoxyribose. When a ddNTP residue is incor- versity has such a facility, and new, low-cost commercial fa-
porated into the growing chain of the newly synthesized cilities are opening as automation techniques are improved.
DNA strand, it cannot form a phosphodiester bond with Nevertheless, simple manual procedures of nucleotide se-
another dNTP and, consequently, the DNA chain termi- quencing are still used in the laboratory for routine molec-
nates. For manual sequencing, one specific ddNTP is added ular biology, for example, promoter mapping by primer ex-
to each of four reaction mixtures and the DNA chain is tension and footprinting studies.
terminated at specific base positions, generating sequence
(oligonucleotide) ladders of random sizes with a fixed 5 ter- 27.1 .l.Manual DNA Sequencing
minus. Separate reactions are run for each of the four nu-
cleotides. The DNA oligomers are separated by denaturing 27.1.1 . l . DNA Polymerases
polyacrylamide gel electrophoresis. A maximum of 400 to The large fragment of the Escherichia coli DNA polymerase
600 bases can be read accurately from one gel (Fig. 1).The (the Klenow fragment), originally used by Sanger et al. (50)
longer reads are achieved by loading additional aliquots of for DNA sequencing, has a low level of 3-to-5 exonucle-
the reaction mixes during the electrophoresis run. ase activity and an intermediate level of 5-to-3 poly-
Automated techniques of DNA sequencing that allow merase activity. The exonuclease can hydrolyze 3 -terminal
for large-scale sequencing projects have been improved over deoxy or dideoxy monophosphates, interfering with the
the last decade and make it possible to obtain a first draft of analysis of polymerase reactions, and therefore, the enzyme
an entire bacterial genome in as little as 1 day in a high- is no longer used for sequencing. Newer polymerases that
throughput sequencing facility. The average run length is are used for DNA sequencing include modified T7 phage-
now 700 to 1,000 bases, depending on the quality of the derived DNA polymerase (60, 61) and a variety of ther-
template, and the cost has been reduced to a few dollars per mostable DNA polymerases such as that from the thermo-
run. For high-throughput projects using templates submit- philic bacterium Themus aquaticus (11). A genetically
ted in 96-well plates, the cost is often reduced an additional engineered form of the enzyme is generally used for auto-
50%. Therefore, it is most cost-effective to have all se- mated sequencing (AmpliTaq; Perkin-Elmer, Emeryville,
quencing projects, whether large or small, performed at a CA). The modified T7 phage-derived polymerase is popular
DNA-sequencing facility. Almost every major research uni- for manual sequencing and is marketed under the trade
name Sequenase 2.0 (U.S. Biochemicals, Cleveland, OH;
http://www.usbweb.com).
The Klenow fragment polymerase has low processivity
(dissociates from and reassociates with the template before
10 nucleoside triphosphates are incorporated) and incorpo-
rates ddNTPs at a 1,000-fold-lower rate than dNTPs. These
properties result in differential band intensities and a low
net level of incorporation of the labeled substrate used and
limit the extension of the primer chain to approximately 350
nucleotides in a typical reaction. Sequenase 2.0 lacks ex-
onuclease activity and is highly processive (for several thou-
sand nucleotides), and the rate of the polymerization reac-
tion is very high. T i DNA polymerase lacks 3-to-5
exonuclease activity, has an intermediate level of processiv-
ity, and yields an intermediate-rate polymerization reaction,
but it has 5-to-3 exonuclease activity and works best at 70
to 80C (33). This enzyme is useful for the sequencing of
double-stranded DNA (dsDNA), especially when plasmid
dsDNA from mini preparations, phage dsDNA, or PCR
products are used as templates. There is a high degree of se-
lectivity of priming since the sequencing reaction is carried
out at a high temperature. Genetically modified versions of
the T i DNA polymerase (Thermo Sequenase for manual
sequencing, from US. Biochemicals, and AmpliTaq for au-
tomated sequencing, from Applied Biosystems Inc.) lack any
exonuclease activity and can survive repeated incubations at
90 to 95C. DNA polymerases from other thermophilic mi-
croorganisms are also now used for DNA sequencing. These
polymerases have specific characteristics that are suitable for
special needs. For a comprehensive update on these enzymes
and their use in DNA sequencing, see reference 2.
High-quality commercial sequencing kits are relatively
inexpensive, provide quality control, and come with de-
tailed descriptions of protocols and free access to a host of
technical tips. For example, kits include the dNTP-ddNTP
mixes that have been tested for quality and proper run
FIGURE 1 Autoradiogram of a sequencing gel. A central length for the reaction products in the gel. We have suc-
portion of the gel approximately 100 bases from the primer is cessfully used the kit marketed by US. Biochemicals for
shown. Lanes are identified at the top, and a portion of the se- many years; however, other products may be just as effec-
quence is indicated at the side. tive. It is strongly recommended that laboratories use kits
27. Nucleic Acid Analysis 655

from U.S. Biochemicals or competing manufacturers for ap- the sample. Additional washes with binding solution can
plications in which they need to do manual sequencing and help remove cellular contaminants, additional wash buffer
gel electrophoresis. Since the kits include detailed descrip- can reduce salts, and additional 70% ethanol washes can
tion of protocols, these descriptions will not be reproduced reduce salts and contaminants. Resuspend the DNA in
here. water or a 1/5 dilution of Tris-EDTA (TE) buffer (Table
1). TE can inhibit Tq polymerase and cause shortened
27.1.1.2. DNATemplates runs. Store the DNA samples frozen at 5 -20C for fu-
ture use. The average yield of DNA should be 10 to 20 Fg
The most critical aspects of both manual and automated
for a miniprep.
sequencing are the quality and quantity of the template.
dsDNA obtained from plasmids is the most common tem-
plate. Methods for the preparation of plasmid DNA are de-
scribed in chapter 30. Plasmid DNA preparations should be
free from RNA. Plasmid DNA prepared by using mini TABLE 1 Preparation of buffers and solutions
preparation kits can work very well; however, care must be Buffer or solution Ineredient or steu Amt
taken to ensure that RNA is completely digested and that
contaminants are minimized. NaTMM buffer 2 M Tris-HC1 100 pl
Several factors should be considered in preparing tem- (10x1 (PH 8.0)
plate DNA. 2 M NaCl 50 p1
1. The bacterial strain used to propagate the plasmid can 1 M MgClz 200 pl
affect the template quality. A number of strains that lack a 14.2 M 14 pl
functional endAl gene (encodes DNA endonuclease), in- 2-mercaptoethan01
cluding E. coli DHSa, DHlOB, TOP-10, XL1-Blue, and Water 1,636 pl
XL10-Gold, have been shown to produce good results.
Other commercially available E. coli strains, for example, Luria-Bertani medium Bacto tryptone 10 g
JM101, HB101, and TB1(54), should be avoided since they Bacto yeast 5g
release endonuclease and large amounts of carbohydrates extract
that can greatly reduce the template quality or recovery. NaCl 5g
2. Cells should be cultured properly. Growth for 16 to 18 Water 1 liter
h at 35C in Luria-Bertani or YT medium works well, and Adjust pH
for most plasmids and strains, the yield of DNA from a to 7.5
miniprep is more than sufficient for DNA sequencing. If
DNA yields are low, the amount of culture used in the Polyethylene glycol Polyethylene glycol 15 g
miniprep may be doubled or a midiprep can be done. solution 6000 or 8000
Alternatively, a super-rich medium such as superbroth or (Carbowax)
2X YT may be used, but growth should be monitored so NaCl 14.6 g
that cultures do not overgrow and begin to lyse. When more Water To 100 ml
cells are used, the prep columns may become overloaded or S1 buffer 3 M potassium 1.1 ml
the amount of contaminants may increase and interfere (lox) acetate (pH 4.6)
with the sequencing reactions. Resuspending the cells in a 5 M NaCl 5 ml
salt solution such as SSC ( l x SSC is 0.15 NaCl plus 0.015
M sodium citrate) and pelleting a second time will reduce Glycerol 5 ml
the levels of some contaminants. Low yields may also occur ZnS04 30 mg
if the proper antibiotic concentration is not maintained. T i sequencing 1 M Tris-HCl 1.1 ml
Ampicillin will degrade during storage and after exposure to buffer ( 5 x ) (PH 4.6)
moisture (such as that from repeated opening when the bot- 1 M MgClz 0.1 ml
tle is cold). In the absence of selection, cells will begin to Water 1.4 ml
lose the plasmid.
3. Template preparation methods must produce suffi- TBE buffer (5X ) Tris base 60.55 g
cient DNA with minimal contaminants. Although you Boric acid 25.8 g
may use very inexpensive methods such as boiling preps, Disodium- 1.85 g
to obtain the most consistent, long-sequence reads, it is EDTA
recommended that proven plasmid kits be used. QIAGEN TE buffer 5 M NaCl 0.2 ml
(http://wwwl .qiagen.com) and Invitrogen (http://www 2 M Tris HCl 0.5 ml
.invitrogen.com) market miniprep kits that work well for
(PH 8.0)
both manual and automated sequencing. For QIAGEN
kits, we find that an additional wash step before the elu- 0.2 M disodium- 50 p1
tion of the DNA can improve results. Modest amounts of EDTA (pH 8.0)
RNA contaminating the sample can be tolerated; how- Water To 100 ml
ever, determining the concentration of DNA in the sam- TES buffer 5 M NaCl 6 ml
ple by using a spectrophotometer will be impossible. Salts 2 M Tris-HC1 5 ml
or ethanol left over from the elution step will inhibit the (PH 8.0)
polymerase, especially in performing cycle sequencing. 0.2 M disodium- 0.5 ml
The presence of contaminants may be due to the improper EDTA (pH 8.0)
removal of supernatants, the precipitation of salt by
ethanol, the overloading of columns, or failure to fully dry Water To 100 ml
656 H MOLECULAR GENETICS

4. PCR products also may be sequenced directly; how- The radiolabeled dNTP (usually a n ~ t - ~ ~ S - l a b e l e d
ever, care must be taken to purify the products away from dNTP) should be obtained fresh. 35S labeling is preferred
contaminating deoxynucleotides, primers, and other inter- because it produces sharp bands upon exposure of the poly-
fering molecules. The contaminating primers can especially acrylamide gel to an X-ray film, and 35S-labeled dNTPs
interfere with cycle sequencing reactions. Also, the PCR have long half-lives (the 35Shalf-life is 87.4 days), thus re-
should produce a single product. These problems can be ducing waste. Overnight exposure of the dried gel to a phos-
avoided by gel purifying a single band from the PCR. The phor screen (used with laser digital imaging systems) or to
gel slice containing the band is purified by using a gel ex- sensitive X-ray film (Kodak XAR) is sufficient to observe
traction kit (QIAGEN and Invitrogen kits work well). the sequencing bands.
5. Determine the approximate amount of DNA by run- For standard sequencing reactions with T7 DNA poly-
ning samples and standard DNA solutions (DNA quantifi- merase, the following materials and reagents are needed:
cation standards may be obtained commercially) on an template DNA (2 to 3 bg for a 3-to-6-kb plasmid), a DNA
agarose gel and comparing intensities of ethidium bromide- oligonucleotide primer (an oligomer of at least 18 deoxynu-
stained bands in the gel (for agarose gel electrophoresis and cleotides; 10 ng/p,l), and the appropriate sequencing kit
staining procedures, see chapters 15 and 30). The concen- components. The SequenaseTMversion 2.0 DNA sequenc-
trations of PCR products should also be checked after gel ing kits from U.S. Biochemicals include Sequenase version
purification. Although spectrophotometer readings can be 2.0 DNA polymerase. It is highly processive and able to ef-
used for plasmid preps, any RNA or carbohydrates in the ficiently incorporate nucleotide analogs (ddNTPs, dITP, 7-
sample will make the readings inaccurate. Once the con- deaza-dGTP) and is not easily impeded by template sec-
centration has been determined by using gel electrophore- ondary structures. Reactions are done in two basic steps.
sis, readings from the spectrophotometer can be compared First, the polymerase, primer, and nucleotides are added in
and calculations can be adjusted if necessary. If the a short labeling reaction. Then the ddNTP mixes are added
minipreps are done consistently by using the same plasmids, (for each template, four reactions each with a mix includ-
strains, and media, spectrophotometer readings can then be ing one of the four ddNTPs) to terminate the chains.
used. The spectrophotometer also provides an estimate of Occasionally, weak bands may occur with prolonged reac-
protein contamination. A n A2,j0/A280ratio of less than 1.7 tion times or with the use of dITP in the sequencing reaction.
indicates contamination. Finally, accurate, sensitive mea- The addition of pyrophosphatase can prevent weak band
surements of DNA concentrations are made with a fluo- intensities brought on by sequence-specific pyrophospho-
rometer by using dyes such as Picogreen (Invitrogen) and rolysis catalyzed by the polymerase (U.S. Biochemicals
Hoechst 33258 (Molecular Probes or Invitrogen). Sequenase protocol). Specialized kits can be used for diffi-
cult templates with significant secondary structures. In one
27.1 .I .3. Manual DNA Sequencing Reactions kit, the substitution of 7-deaza-dGTP for standard dGTP
DNA polymerase uses dNTPs as substrates for the extension helps to resolve gel compressions that may arise from sec-
of the primer that hybridizes with the single-stranded DNA ondary structures of the DNA in the gel.
template. ddNTPs are substrate analogs, the incorporation Manual sequencing protocols using Tq DNA poly-
of which results in the termination of the polymerization merase (cycle sequencing) require much less of the template
reaction. If dGTP is used in the sequencing reaction mix- (0.1 Fg). The reactions are done in two basic steps: thermal
ture, upon electrophoresis anomalous separations of cycle labeling-amplification and thermal cycle termination
oligonucleotides occasionally result from the compression by the ddNTPs. Primers of 18- to 30-nucleotide oligomers
of G-C stretches present in the DNA, which forms a local- are used. Thirty to 35 thermal cycles are performed with the
ized secondary structure. This problem can be alleviated by inclusion of the labeled deoxynucleotide. In each cycle, the
replacing dGTP with dITP or with 7'-deaza-2'-dGTP in the temperature is first raised to 95C to denature the template
polymerization reaction mixture. The concentrations of and then it is reduced to 55C to anneal the primer, fol-
dNTPs and ddNTPs are critical for the sequencing reaction; lowed by an extension step at 70C. Depending on the
hence, they must be handled properly. The manufacturer's melting temperature of the primer and the empirical results
recommendations must be followed carefully, reagents must of the sequencing run, the annealing step may be adjusted
be kept on ice during use, and reagents that have been to obtain better annealing or to reduce nonspecific priming.
stored for a long period of time or that produce gradually Then the four termination mixes are added individually to
poorer results should be discarded. For manual sequencing, four tubes per template-primer pair, and an additional 30 to
four reactions are set up for each template and primer pair. 35 cycles are performed. These cycles produce a set of
Then a short labeling reaction is done to produce labeled single-stranded DNA fragments that are terminated at each
DNA fragments from the template and primer, dNTPs, and respective ddNTP, either A, C, G, or T, in a particular re-
a labeled dNTP. This step is followed by a termination re- action tube. The reaction mixtures are cleaned up and ap-
action that is started by adding a mix containing all four plied to a sequencing gel as described for standard sequenc-
dNTPs and only one ddNTP. Each of the four tubes gets ei- ing, producing the same type of gel image (Fig. 1). Cycle
ther the A, C, G, or T ddNTP. Since the elongation of the sequencing has the advantage that the higher temperature
DNA chain terminates with the incorporation of one mol- of the reaction reduces the effects of template secondary
ecule of ddNTP, the oligonucleotides in the sequencing lad- structure.
der will end with the specific ddNTP used in the reaction.
By using four different ddNTPs in separate reactions, and 27.1 .I .4. Gel Electrophoresis
then separating the oligonucleotides extended from the Gel apparatuses of various lengths and widths are commer-
primer in parallel on a polyacrylamide gel, the sequence of cially available. Larger and wider gels are difficult to handle.
bases is read (Fig. 1). The fragments that terminate near the Apparatuses from Bio-Rad work well. Gels of 40 cm
primer appear at the bottom of the gel. A readable sequence (length) by 20 cm (width) by 0.3 to 0.4 cm (thickness) can
adjacent to the primer is usually not obtained, so primers be conveniently handled. Plastic spacers placed between
should be designed at least 20 bases away from the sequence glass plates determine the thickness of the gel. In order to
to be read. read the maximum number of nucleotides from a gel,
27. Nucleic Acid Analysis 657

wedge-shaped spacers (tapered from 0.3 to 0.8 cm) may be sealed by using tape (3M yellow electrical tape or its equiv-
used (1, 34); however, they are more difficult to dry. alent) instead of the bottom spacer. The bottom can also be
Polyacrylamide gradient gels (4 to 18%) or buffer gradient sealed by inserting a piece of filter paper in between the
gels have also been used (4, 24, 43). Buffer gradient gels plates and soaking the filter paper with a mixture of 12 to
with different ionic concentrations of the electrode buffer 18% acrylamide solution and high concentrations of cata-
are the easiest to handle among all different kinds of gradi- lysts (51).
ent gels. Two different kinds of combs are used for forming 3. For casting two 7% gels, mix the following in a
wells in gels: those producing conventional square or rec- beaker: urea, 33 g; 40% acrylamide solution, 13.1 ml; 10%
tangular wells that are spaced a few millimeters apart from ammonium persulfate, 0.38 ml; 5 X TBE, 15 ml; and water to
one another and sharks tooth combs forming wells sepa- 75 ml. The addition of warm water (about 50C) will help
rated from one another by minimum spacing. In order to in dissolving urea. After the solutions are thoroughly mixed,
form conventional wells, the comb is inserted into the gel filter the solution through a fluted Whatman no. 1 filter
mold before the gel solution polymerizes. In contrast, the paper if you see any suspended particles (some batches of
flat end of the sharks tooth comb is inserted into the gel urea contain insoluble particles). Cool the solution on ice,
mold prior to the polymerization of the gel solution. The add 38 pl of TEMED, and mix by swirling. Cold solution
comb is removed after the gel is polymerized, the comb is will polymerize slowly in the presence of TEMED, allowing
rotated 180,and wells are formed by slightly inserting the sufficient time to pour the gel solution into the gel mold.
sharp teeth of the comb into the gel. The loading of samples 4. Draw the above-described solution into a 25-ml
onto the sharks tooth gel is more difficult, and leaking of pipette or a hypodermic syringe. Hold the glass plates at an
samples from one well to another well is a common prob- angle (45),and pour gel solution through one comer of the
lem. O n the other hand, the reading of sequences from such gel mold at a uniform rate. If any air bubbles are trapped,
a gel is improved since the lanes run very close together. the plates are not clean. However, air bubbles usually can be
For the preparation of sequencing gels, use a solution removed by tapping on the upper plate (the wooden handle
containing 38% (wtlvol) acrylamide and 2% (wtlvol) of a large spatula works well for tapping). When the mold is
N,N-methylenebisacrylamideas a stock solution. Acry- filled, lay the plates on the top of a rubber stopper in a near-
lamide is a neurotoxin, so wear gloves while handling it, horizontal position. Insert the comb. Place clamps holding
and use a mask while handling powdered acrylamide. Make the two glass plates together in the area where the comb is
the solution in warm water (40 to 50C),deionize by stir- inserted. Allow the gel to polymerize over a period of 1 to 2
ring with 5 g of Amberlite MB-1 resin (from Mallinckrodt hours at room temperature before electrophoresis. Efficient
or Sigma Chemical Co.) or an equivalent monobed polymerization can be achieved in 30 min at 37C.
resin/liter for a period of 30 min or longer, and then filter 5. When ready to use the gel for electrophoresis, re-
through Whatman no. 1 filter paper (preferably fluted). move the clamps, the bottom spacer (or the piece of sealing
The stock solution can be stored in the dark at 4C for tape), and the comb. Wash the top part of the gel with 1X
months (12 months). Ammonium persulfate and TEMED TBE. Remove any polymerized gel particles trapped in the
(N,N,N,N-tetramethylethylenediamine) are used as cata- wells by forcing l x TBE through a Pasteur pipette (with
lysts for the polymerization reaction. A stock solution of one end drawn to a capillary shape). If a sharks tooth comb
ammonium persulfate (10% [wt/vol] in water) can be stored is used, insert the comb with the teeth just penetrating the
for up to a week at 4C. acrylamide. Make sure all the teeth have penetrated, but do
The volumes of the solutions required for a gel depend not insert so far that the surface of the well is bowed.
on the sizes of the glass plates and the thickness of the spac- 6. Pour electrode buffer (1X TBE or a different buffer
ers used. For casting gels, follow the recommendation of the for buffer gradient gel) into the lower buffer reservoir. Place
supplier of the gel apparatus. Polymerized gels can be stored the plates vertically (unnotched plate facing outside), and
clamped with large binder clamps and wrapped with plastic secure them to the gel apparatus by using clamps or screws.
wrap at room temperature to prevent dehydration from the Air bubbles between the lower edge of the gel and the lower
area where the comb is inserted. Gels should not be stored electrode buffer should be avoided (forcing of buffer
for more than 24 h under the above-described conditions. through a Pasteur pipette may help remove any trapped air
Use clean glass plates to prevent the trapping of bubbles in bubbles). Fill the upper buffer reservoir with 1X TBE, mak-
the gel sandwiched between the glass plates. The glass ing connection between the upper part of the gel and the
plates are best cleaned with warm water without detergents upper electrode buffer.
or acids immediately following electrophoresis. Spray 7. Prerun the gel for 30 min to warm up to 40 to 45C
ethanol on the washed glass plates, and wipe with a few Kim (set the power as recommended by the supplier of the gel
wipes. Applying a layer of silicone to one side of one of the apparatus).
plates (with Sigmacoat [Sigma Chemical Co.] or an equiv- 8. Disconnect the power, and wash urea from the wells
alent chemical) facilitates the separation of the gel that is by squirting 1X TBE with a Pasteur pipette or syringe.
sandwiched between the glass plates. A protocol for the 9. Load 1 to 3 p l of samples into each well by using a
preparation of a 7% gel (20 by 40 by 0.3 cm) is given below. pipette and capillary tips (commercially available as flat
tips or gel4oading tips). Samples should be heated to
1. Materials and solutions required are as follows: glass 70C before loading. Load in the order G, A, T, and C for
plates, spacers, combs, large binder clamps, 40% acry- each template. Rinse the pipette tip by pipetting buffer from
lamide-bisacrylamide solution (38:2), 10% ammonium per- the lower reservoir in and out between the loading of each
sulfate, TEMED, ultrapure urea, and 5 X Tris-borate-EDTA sample, or use a separate tip for each sample. When loading
(TBE) (Table 1). samples with reactions from many templates, load samples
2. Two glass plates (one notched) are put together with for one to three templates and then electrophorese for 3
spacers in between the plates on the longitudinal edges and min before loading another set of samples. Remove urea
at the bottom (optional). The spacers must be placed on the from the wells, as described above, between each loading.
siliconized surface of one of the glass plates. Use clamps to 10. Turn on the power, and run the gel for about 20 min
hold the plates and spacers together. The bottom can be after the bromophenol blue dye runs out of the bottom of
658 MOLECULAR GENETICS

the gel. Previously loaded samples can be loaded again in ing, all four labeled dideoxynucleotides will be detected in
adjacent wells at this time, and electrophoresis is continued a single tube. The different bases are labeled with fluores-
for an equal amount of time to obtain the maximum se- cent probes that each emit light at a unique wavelength. As
quence information for each template from a single gel. The the DNA fragments are synthesized, termination and label-
choice of using multiple gels with different run times or ing occur when a labeled ddNTP is encountered. The ratio
staggered loading of one gel depends on the number of tem- of ddNTPs to dNTPs is set so that these termination reac-
plates and the width of the gel (number of wells) used. tions occur evenly across all possible positions. All reactions
11. After electrophoresis is complete, disassemble the are done in 96-well plates, greatly increasing the through-
glass plates from the electrophoresis apparatus, wash the put. The reaction mixtures are cleaned up to remove unin-
plates with tap water, and lay the plates flat on a sheet of ab- corporated ddNTPs and other contaminants by using 96-
sorbent paper (for safe handling of radioactive materials). well clean-up plates and then put into an automated DNA
Pry the plates apart by starting at one corner. Observe care- sequencer.
fully and lay down the plate to which the gel remains at- The most popular present capillary sequencing instru-
tached. If desired, cut about the top 3 cm of the gel with a ments are the ABI 3730 and 3730x1, which are capable of
pizza cutter (works best) or a scalpel and discard the top por- processing hundreds of samples per day with unattended op-
tion of the gel. This part of the gel contains the maximum eration. The sequencers are equipped with an autoloader, a
amount of radioactivity, and sequences cannot be read in sample injection system, and an array of 48 or 96 very thin
this area. Note that the buffer in the lower reservoir is ra- capillaries. A separation matrix (the latest is POP7) is au-
dioactive and should be disposed of properly. tomatically injected into the capillaries before each sample
12. Place the gel with the support of the attached glass run. Then an electrokinetic injection system draws samples
plate in a liter (or more) of 5% acetic acid bath (a plastic from the 96-well plate and the DNA is carried by an elec-
tray can be used for making the acetic acid bath). Soak the tric current into the capillary matrix.
gel in acetic acid for a period of 30 min at room temperature Although this remarkable system has greatly reduced
without shaking. Soaking with acetic acid helps fix the labor, it does have certain limitations. Templates must be
oligonucleotide bands in the gel and removes urea. cleaner than with previous gel-based systems. Any nega-
13. Carefully remove the gel with the support of the glass tively charged contaminants compete with the DNA injec-
plate from the acetic acid. Soak excess acetic acid from the tion, reducing the signal and possibly broadening bands. TE
top of the gel by placing two to three layers of tissue papers and salts eluted with the DNA from miniprep kits are com-
(Kim wipes) over the gel and slowly rolling the tissue papers mon culprits. It is highly recommended to elute with water
off the gel from one comer. Place a piece of a Whatman or a 1/10 dilution of TE. Any silica from miniprep kits car-
3MM paper on the gel, and press the paper slowly to adhere ried over with DNA can be especially damaging to the cap-
to the gel surface. illaries. Since the entire block of capillaries must be re-
14. Invert the plate with hand support onto the placed when even a few are blocked, contaminated DNA
Whatman paper, and slowly peel the plate away from the preps can greatly increase the cost of sequencing for all who
gel. Note the orientation of the gel to ascertain the proper use a facility. Even moderate levels of contaminants will re-
order of the samples loaded. Place a piece of plastic wrap on duce the lifespan of the capillaries.
the gel, and dry the gel under a vacuum at 70 to 80C. Gel- As the fluorescently labeled DNA fragments reach a de-
drying apparatuses are commercially available. tection cell at the end of the capillaries, a laser beam excites
15. Remove the plastic wrap, and expose the dried gel the fluors. The subsequent emissions are captured by a
overnight to an X-ray film (Kodak XAR-5 or its equivalent) charge-coupled device camera, and the software interprets
for autoradiography or expose it for at least 4 h to a phos- which color of dye is passing through at any time. The soft-
phor screen (be sure the screen is sensitive to for ware evaluates the size, spacing, and other properties of the
laser digital imaging. peaks and identifies the base sequence.
16. After the image is developed, sequences can be read
from the film or computer. Mark the lanes in the appropri- 27.1.2.2. Interpreting Chromatograms
ate order of loading. Start reading from the bottom of the The output provided to the user for each run consists of a
film in ascending order of bands present in the lanes (G, A, simple text file with the sequence identified by the software
T, and C). Note that the sequence read is complementary to and a much larger file with the chromatogram. ABI sup-
the sequence of the template DNA strand. plied the free viewing and editing program Editview for the
Macintosh, and later Chromas for personal computers be-
27.1.2. Automated DNA Sequencing came available. Editview is no longer supported and is not
compatible with the Mac OSX system. A new program
27.1 -2.1. Overview called FinchTV from Geospiza (http://www.geospiza.com/
Automated fluorescence sequencing utilizes the same general finchtv) is an excellent program for viewing chromatogram
Sanger chain termination methods as manual sequencing; traces on Linux, Mac OSX, Windows, and Solaris systems.
however, the methods are substantially modified to greatly Bases are shown in colors: black, G; red, T; blue, C; and
increase speed and reduce the cost per sample. As in man- green, A. The first things to notice are the peak heights and
ual cycle sequencing, a primer is hybridized to the DNA and spacing and the background. An example of a reasonably
the template is amplified by using 35 reaction cycles (note good sequence is shown in Color Plate 12A. The part of the
that this is not PCR since only one primer is used). Most se- sequence close to the primer is often not readable, so
quencing facilities use AmpliTaq and reagents marketed by primers should be designed 20 to 40 bp away from the re-
Applied Biosystems. The enzyme and reagents are premixed gion of interest. For 500 to 700 bases, the sequence should
in the present ABI BigDye terminator version 3.1 reagent be more than 99% accurate, and then as the peaks spread
kit. A set of four fluorescently labeled ddNTPs representing and lose intensity, the error rate increases. Even with 900 to
each of the four bases, AGCT, is present in each reaction 1,100 bases, a sequence with occasional errors is obtained if
mixture, along with all dNTPs. Unlike in manual sequenc- the template is of good quality. Due to the higher error rate,
27. Nucleic Acid Analysis 659

it is unadvisable to design primers beyond 700 bp from the cycle sequencing reactions, or sequencing from the op-
single-run data. With the use of a pure template, appropriate posite strand.
primers, and good sequencing procedures, the peak heights
are relatively even and the background signal is almost non-
existent. When the computer program is unable to make an 27.2. DNA-PROTEIN BINDING
identification, an N is placed at the position in question. A variety of techniques are available to study the factors
Experienced users can often detect patterns in the data and that influence the regulation of a gene. If sequence-specific
accurately extend the base identification beyond the limits DNA-binding proteins are thought to be involved, it is of
of the software, but as the base-identifying algorithms have interest to determine the locations of the binding sites and
improved, the benefit from peering over chromatograms to the biochemical properties of the protein-DNA interac-
determine a few additional bases has dwindled. tions. In bacteria, these sites can be close to the transcrip-
tion start site or they may be located more than a kilobase
27.1.2.3. Troubleshooting away. An analysis in vivo of variants with deletion and
Color Plate 12B illustrates a common problem that has point mutations can be used to obtain information on the
nothing to do with the quality of the sequencing reactions. locations of the sites, but ultimately biochemical assays
Throughout the entire sequence, two bands at each position must be used. To study in vitro the factors that influence
are observed. This is a clear indication that the template is transcription, several DNA-binding assays can be em-
a mixture of two plasmids. Excess DNA is often used for ployed. If the location of the suspected binding site is
bacterial transformations, and this practice often results in known within a few hundred base pairs, nuclease or chemi-
multiple plasmids entering the same cell. The remedy is to cal protection (footprinting) assays can be used (see section
simply plate the strain and isolate single colonies. Similar 27.2.2). If the location of the site is not suspected, or if the
results (although usually with more bands observed) may protein is of low abundance and purity, use gel mobility shift
also appear if there are multiple priming events. Either de- or chromatin immunoprecipitation (ChIP) assays. The re-
sign a new primer or ask if the sequencing reaction can be cently developed ChIP assay is capable of simultaneously
run with a higher annealing temperature. Most facilities isolating for one protein all protein-binding sites within a
perform all standard cycle sequencing at one temperature, genome. The basic aspects of the gel mobility shift assay
about 55C; however, they may do runs with problem sam- using purified protein are described first, and then the cell
ples at other temperatures. In Color Plate 12C, there is also lysate assay is discussed. Next, protection assays are consid-
a site with multiple peaks, near position 145. In this case, ered, followed by a review of the ChIP assay. Further dis-
since two bands appear only in two positions, it is likely that cussion of all the assays may be found in reviews (10,27,46)
the template is derived from a heterozygote. This scenario is and method manuals (2, 7).
most common in the sequencing of PCR products.
A total failure to obtain a sequence is shown in Color 27.2.1. Gel Mobility Shift Assay
Plate 12D. This result may be caused by improper design or The gel mobility shift assay (also called the gel retardation
degradation of the primer, loss of the primer binding site on assay) is based on the differences in the degrees of elec-
the plasmid, a greatly reduced concentration of intact trophoretic mobility between nucleic acid fragments and
DNA, or the inhibition of the reaction by contaminants. nucleic acid-protein complexes (14, 19). Although the
The result in Color Plate 12E is an example of inhibition by assay has been used successfully to study binding to RNA
a high salt content. Bands are broadened, and base intensi- (52), this discussion will be limited to the most common
ties are quite variable. The high level of background signal use, the study of binding to dsDNA. A solution of protein is
in Color Plate 12G was not sufficient to interfere with most mixed with DNA fragments in an appropriate binding
base identifications in the early part of the run; however, buffer. After allowing the protein to bind, the mixture is ap-
the readable sequence is very short. High levels of back- plied to a nondenaturing gel, and following a period of elec-
ground signals occur with multiple priming or insufficient trophoresis, the positions of the DNA fragments are deter-
purification of PCR products, poor-quality plasmid tem- mined. If the protein binds the DNA stably, the increased
plates, or primers containing shortened contaminants from mass and the alteration in the charge of the complex will
the oligonucleotide synthesis. Very large bands in the 60- to generally cause it to migrate more slowly than the DNA
80-base region are dye blobs resulting from contamination alone. The relative amounts of free DNA and complex
with unincorporated labeled ddNTPs. If the sequence is bands can be analyzed quantitatively to obtain information
weak due to a low template concentration or contamina- on the concentrations of the reactants, equilibrium and ki-
tion, the excess unincorporated nucleotides overwhelm the netic binding constants, stoichiometry, and some aspects of
purification procedure. If the dye blobs persist even with a the structure of the complex (12, 14-17, 19,41,52,59,67).
good sequence signal, the facility should consider alterna- The greatest advantage of the gel shift assay over other
tive purification procedures. methods is its versatility. Specific protein-DNA interac-
Truncated sequences such as that shown in Color Plate tions can be detected by using a mixture of DNA fragments,
12F also may result from a number of problems. G-C-rich such as a digest of a whole plasmid. This is valuable for
regions produce a high frequency of hairpin structures quickly locating binding sites within a large segment of
when single stranded. During the sequencing reaction, the DNA. By using a single DNA fragment, interactions with
hairpin forms and may block polymerase, causing an multiple proteins can usually be resolved since different pro-
abrupt drop in the intensity of the sequence signal. A teins or different stoichiometries of binding yield character-
gradual loss of the signal may also occur if the blockage is istic shift positions. Thus, the binding of multiple activator
only partial. The ABI BigDye 3.1 formulation has greatly or repressor proteins can be distinguished from the binding
reduced the effects of hairpins, but they are still seen reg- of one species, which in turn can be distinguished from the
ularly. There are several common remedies, including binding of a large molecule such as RNA polymerase (32,
adding denaturants such as dimethyl sulfoxide to the se- 59). This property allows for functional assays in studies of
quencing reaction mixtures, increasing the temperature of protein interactions at promoters. Even in a crude cell
660 MOLECULAR GENETICS

lysate, the specific binding of one protein can be detected if buffers such as TE may allow some denaturation of the
appropriate controls are performed to demonstrate speci- DNA, resulting in the appearance of a band migrating more
ficity. This is perhaps the most powerful aspect of the assay, slowly than the intact, dsDNA exactly where a protein-
since it provides a rapid, convenient method to quantify DNA complex is anticipated.
protein from new constructs or to assay fractions from pu-
rification schemes. The method can also be used prepara- 27.2.1.2. Gels and Buffers
tively to obtain the bound and free fractions of the DNA, Polyacrylamide and agarose gels have both been used suc-
to isolate bound proteins from a crude mixture, or to obtain cessfully for mobility shift assays. Polyacrylamide gels may
the protein-DNA complexes after treatments such as UV be 4 to 10% with 0.33 to 0.7% bisacrylamide, depending on
cross-linking and chemical modification (27,46). the sizes of the DNA fragment and the protein. For most
All these applications rely on the basic finding that regulatory proteins and DNA fragments of fewer than 600
protein-DNA complexes are more stable in the gel than in bp, 4 to 6% acrylamide is a good concentration for pilot ex-
solution. This observation has been explained on the basis periments. NuSieve agarose (Cambrex Corp.) at 3 to 4%
of the caging effect of the gel. The complexes are not may be substituted in most cases since it has sieving proper-
more stable; dissociated protein reassociates at a greatly in- ties similar to those of polyacrylamide. Regular agarose is
creased rate so that the reactants do not have a chance to generally useful only when the protein is large or multipro-
migrate apart. Since the electrostatic component of the tein complexes are bound.
binding interaction is stronger at lower salt concentrations, Many different gel buffers have been used. In the first gel
an additional stability of complexes may be obtained by mobility shift experiments, very-low-salt-content TE (10
using low-salt-content gels. mM Tris, 1 mM EDTA, pH 7.5) buffer was used to maximize
protein binding during electrophoresis (11, 16). The low
2 7.2.1 .l . DNA Fragments buffering capacity requires that a circulator be used during
The most common DNA templates used in mobility shift electrophoresis; in as little as 30 min without circulation,
assays are restriction fragments, synthetic oligodeoxynu- the pH of the gel can drop to 5 5 . Usually, l x or 2X TBE
cleotides, and PCR products. Best results, easily observable (45 mM Tris-borate, 1 mM EDTA, pH 8.3) and TAE (40
shifts, are obtained with fragments of less than 1 kb p, al- mM Tris-acetate, 15 mM Na acetate, 1 mM EDTA, pH 8.0)
though assays with larger fragments and even whole plas- may be substituted; these buffers have the advantage that
mids have been successful. It should be kept in mind that no circulation is necessary. Care should be taken with TBE,
the larger the DNA fragment, the smaller the shift in the however, since borate may significantly affect the DNA-
mobilities of the complexes and the greater the amount of binding properties of protein. The gel buffer need not be the
nonspecific protein binding. If the fragment of interest can same as the buffer used for the binding reaction. Some cofac-
be cut by restriction enzymes, leaving only one or two large tors required for the stable binding of protein to DNA may
vector fragments, the mixture may be used in the assay; oth- also be necessary in the gel, such as metal ions like Mg2+ or
erwise, the fragment should be isolated by gel electrophore- allosteric effectors, for example, cyclic AMP (CAMP) for
sis. Synthetic oligodeoxynucleotides and fragments pro- the E. coli catabolite gene activator protein.
duced by PCR should also be purified to eliminate any
contaminants that may interfere with binding. Mixtures 27.2.1.3. DNA Labeling
containing fragments that may comigrate with the protein-
The following protocol is for filling in the ends of restric-
DNA complexes should be avoided. Unlabeled DNA can tion fragments with 5 overhangs. End labeling with T4
be detected by soaking the gel in 1 pg of ethidium bro-
polynucleotide kinase (section 27.3.1.3) may be used for 5
mide/ml after electrophoresis. Labeling the fragments with
overhangs or blunt-end fragments. For fragments with 3
32P greatly reduces the necessary amounts of reactants and overhangs, T4 polymerase can be used to digest the ends
is the method of choice. One of the standard end-labeling
back and then to fill in with labeled nucleotides (49). The
or internal labeling protocols may be used; however, proto-
incorporation of radioactivity may be measured by placing a
cols such as nick translation and random primer labeling
portion of the sample in the scintillation counter without
should be used with caution as they may leave partially
using scintillation fluid and using the discriminator setting
filled-in DNA that will significantly alter quantitative mea-
for tritium or an open setting. This method of counting by
surements and produce smeared bands. For most applica-
using the Cerenkov effect is about 50% as efficient as
tions, labeling to the extent of producing high specific ac-
counting by using 32P with a scintillation fluid, but the
tivity is not necessary since as little as 200 cpm is sufficient
method is quick and it does not destroy the counted sample.
for the detection of bands upon overnight exposure to a
phosphor screen and imaging using a laser digital imager or 1. Use an appropriate restriction enzyme in a volume of
overnight exposure to X-ray film with an intensifying 40 pl to digest 5 to 10 kg of plasmid DNA containing the
screen. After labeling, the unincorporated nucleotides may region of interest (see chapter 30 for plasmid purification
be removed by chromatography on a 1-ml Sephadex G-50 and handling). Inactivate the enzyme by heating to 70C
column or by similar methods. The purification or removal for 10 min. In the case of a heat-resistant enzyme, remove
of unincorporated nucleotides may be done by using DNA by phenol extraction and ethanol precipitation or with a
clean-up kits (QIAGEN, Invitrogen, or Promega), but do clean-up kit. Tables for the heat inactivation of restriction
not use kits that employ loose silica beads. Frequently, a enzymes may be found in the catalogs of manufacturers such
small portion of the DNA remains bound with silica and is as New England Biolabs.
observed as a band that fails to migrate into the gel. Choose 2. If a low- or moderate-salt-content buffer or a 1x con-
kits that use a membrane-bound matrix. Gel purification centration of a universal buffer (50 mM potassium acetate or
can be used to isolate the fragment from vector fragments as glutamate) is used for the restriction digestion, proceed to step
well as free nucleotides. The DNA should be stored in a 3. If a high-salt-content buffer is used or if the sample was
buffer such as 10 mM Tris (pH 7.5)-50 mM NaC1-1 mM treated with phenol, remove the buffer by ethanol precipita-
EDTA. Storage of small fragments in low-salt-content tion with a 1/10 volume of 3 M Na acetate, pH 5, and 2.5 vol-
27. Nucleic Acid Analysis rn 661

umes of ethanol at -20C overnight or - 70C for 30 min. mm wide. Use 6% polyacrylamide-O.l% bisacrylamide in
Alternatively, isolate the DNA by using a silica method. If a TAE buffer (40 mM Tris base, 15 mM Na acetate, 1 mM
silica method is used, follow the manufacturers instructions EDTA; adjust to pH 8.0 with acetic acid), which is filtered
but repeat the final elution step and carefully draw off the su- and degassed. A 20% polyacrylamide-O.33% bisacrylamide
pernatant to reduce the amount of silica carried over into the stock can be stored in the dark at 4C for up to several
sample. Resuspend the DNA in 40 p1 of TE (Table 1). months; store TAE as a lox stock.
3. Add 50 pCi of an a-32P-labeled dNTP and 1 p1 of a 2. Polymerize with 20 pl of TEMED and 200 p1 of fresh
10 mM stock of each of the other dNTPs as necessary to fill 10% ammonium persulfate for 50 ml of gel.
in the fragment ends. The nucleotides used will depend on 3. If available, set up a pump to circulate the gel buffer.
the restriction enzyme, but it is best that the labeled nu- The gel should be run in the cold for pilot experiments until
cleotide not be the last to be incorporated. If the DNA is in the stability of the complex is known.
TE, add 5 p1 of lox polymerase buffer (500 mM Tris-HC1 4. Soak the gel in running buffer (the same as the gel
[pH 8.01, 100 mM MgC12, 10 mM dithiothreitol [ D m , 500 buffer) for several hours (preferably overnight) to allow the
mg of bovine serum albumin [BSA]/ml). Add 2 U of TEMED and persulfate to diffuse out. Add ligands necessary
Klenow fragment DNA polymerase, and incubate at room for the protein (such as cAMP for the cAMP receptor pro-
temperature for 15 min. tein) at this time. For horizontal gels, soak for 1 h.
4.Add 2 p l of a solution of all four nucleotides (5 mM 5. Prerun the gel at 5 to 10 V/cm for 1/2 to 1 h. If the
each), and continue the incubation for 5 min. buffer is not circulated, mix the buffer portions from the
5. Ethanol precipitate as described in step 2, and dry. upper and lower tanks or replace the buffer before loading
Centrifuge 10 min in a microcentrifuge, remove the super- the samples.
natant, add 70% ethanol to the pellet (without disturbing
it), centrifuge 2 min, remove the supernatant, and dry the 27.2.1.5. Binding Reactions and Electrophoresis
pellet in air or in a vacuum concentrator. Resuspend the 1. Make an appropriate binding buffer. Usually, some salt
DNA in 20 pl of TE, and add 2 pl of 1OX loading buffer is added to reduce nonspecific binding, components are
(50% glycerol, 0.05% bromophenol blue). added to stabilize the protein, and glycerol is added to allow
6. Purify the fragment of interest by electrophoresis on layering on the gel. A typical buffer is as follows: 10 mM Tris,
agarose or NuSieve agarose, depending on size. NuSieve 50 mM KC1, 1 mM EDTA, 5% glycerol, 50 pg of BSA/ml,
agarose (3% NuSieve-1% agarose mixture) will separate and 1 mM DTT. NP-40 (0.05%) may be added if the protein
small fragments in the range of 50 to 500 bp well. After is unstable and/or aggregates. An easy method is to make a
electrophoresis in the presence of 0.6 pg of ethidium bro- 5 X concentrated stock with the first four items (store them
midelml, visualize the band with UV light and make an in- frozen) and add the BSA, DTT, water, any additional factors,
cision in the gel just ahead of the band. Place a small piece and DNA to a single tube just before use. Make up 10%
of NA45 membrane (Schleicher & Schuell, Keene, N H ) more mix than required to allow for pipetting error.
into the incision down to the gel tray. Electrophorese the 2. Add enough DNA for 500 cpm (Cerenkov) for each
DNA onto the membrane at two times the voltage used for sample (assuming a single labeled DNA fragment). Pipette
the gel run; take care not to run additional bands onto the 20 p1 of the complete binding mix into 1.5-ml microcen-
membrane. Remove the membrane, wash in 1 ml of low- trifuge tubes for each binding reaction.
salt-content buffer (10 mM Tris [pH 7.51, 100 mM NaCl), 3. Make up the binding mix without DNA for diluting
and elute for 45 min in 300 p1 of high-salt-content buffer the protein. Make 10-fold serial dilutions of the protein,
(10 mM Tris [pH 7.51, 1 M NaC1) at 70C according to the and maintain on ice for a limited time. Never vortex solutions
manufacturers directions. Remove the membrane and pre- containing protein; mix gently by using the pipette tip or by
cipitate the DNA by adding 1.5 p1 of 1 M MgClz and 2.5 flicking the tube with a finger. For a pilot experiment, al-
volumes of ethanol, hold at -70C for 30 min, centrifuge ternate adding l and 3 p l of each dilution to reaction tubes.
for 10 min, and wash the pellet with 70% ethanol. For a purified protein, the lowest target concentration can
Resuspend in 100 p1 of TES (Table 1) and store frozen. This be estimated from the amount of DNA in each sample
method is simple and produces quite clean DNA with a spe- (generally less than 1 ng) by assuming 100% active protein.
cific activity of >5 x lo6 cpm per pg of fragment. A p;otein at 1 mg/ml should require a dilution of to
Clean-up kits (Qiaex or Invitrogen gel extraction kits) 10- . One tube should have no protein added. Thus, the
also work well to isolate the DNA from NuSieve or stan- full range of concentrations can be assayed in 12 reactions.
dard agarose gels. For fragments of fewer than 100 bp, such 4.After adding protein, incubate the reaction mixtures
as synthetic oligonucleotides, be sure that the clean-up kit for 30 min at room temperature. Many proteins require only
is rated for oligonucleotides. The gel purification may be 5 to 10 min.
done before labeling, especially if several fragments are pro- 5. Gently add 1 pl of 0.05% bromophenol blue, pH 7.5
duced by the restriction digestion. In this case, remove the (the dye should be neutralized with Tris or other buffer be-
fore use). Turn off the power (and circulator if used), and
free nucleotides by column chromatography after labeling.
load the samples carefully but quickly onto the gel. A con-
27.2.1.4. Gel Preparation sideration important for this assay is that the binding char-
acteristics map be changed when the sample is mixed with
The binding assay can be done most easily on horizontal,
submarine gels (28); however, since horizontal acrylamide gel-running buffer. To minimize this effect, samples are
gel apparatuses are not used widely, the following procedure loaded with a minimum of mixing and frequently the sam-
is for a standard vertical system. It is best if the vertical gel ples are loaded with the current turned on. (Caution: take
apparatus has fittings for buffer circulation, but this is not great care to avoid electric shock.) It takes about 1 to 3 min
essential if the running time is limited. for the complexes to enter the gel. After the dye has entered
the gel, the circulator may be turned on.
1. Cast a 1.5-mm-thick gel with standard size plates (usu- 6. Electrophorese at 5 to 10 V/cm (usually 100 to 150 V)
ally 14 to 16 cm long) and a comb to produce wells 5 to 10 until the dye is one-third of the way to halfway down the
662 MOLECULAR GENETICS

gel, 1.5 to 2 h). The dye should run with a DNA fragment in the gel, nonspecific binding is readily detected. Once one
of about 50 bp. The voltage should be adjusted so that min- molecule of protein is bound at its specific site, additional
imal heating occurs. protein added to the reaction mixture results in the moder-
7. Gently pry the plates apart by using a spatula, and pull ately stable binding of multiple proteins. Even if no specific
the top plate away, leaving the gel on one plate. Gently binding occurs, nonspecifically bound protein-DNA bands
press a piece of filter paper onto the gel, and pull it off the may be observed. Therefore, it is important to carefully
plate. Cover with a piece of plastic wrap, and dry in a vac- titrate the DNA and to characterize the binding with
uum dryer (1 h at 80C). proper controls. One simple control is to add equal amounts
8. Expose the dried gel to X-ray film. Expose overnight of a labeled, irrelevant DNA fragment and a specific DNA
with an enhancing screen (such as DuPont Cronex) for 300 fragment to the binding mix. The specific fragment should
to 1,000 cpm (Cerenkov) per band. Place the screen in the shift in position while the control fragment remains un-
film holder, then add the film, and finally add the gel (fat- changed. This result is easiest to observe if the control frag-
ing down). For shorter exposures, or if intensifying screens ment is somewhat smaller than the specific fragment.
are not available, use an amount of DNA corresponding to The gel mobility shift assay can be used to obtain rea-
2,000 to 5,000 cpm per band. Exposure to a phosphor screen sonably accurate quantitative information about the bind-
for 4 h to overnight will also provide a sufficient signal for ing reaction. Several methods can be employed to deter-
the laser digital imager. mine the equilibrium constant for binding. For any of these
methods, the concentration of the protein active in specific
An example of protein titration using the gel mobility
DNA binding must be determined accurately. The simplest
shift assay is given in Fig. 2. The relative mobility of the
method to accomplish this is to accurately determine the
shifted band can vary widely with different combinations of concentration of the DNA fragment containing a single,
protein and DNA. If only a slight shift is seen, the gel con-
specific binding site and then to titrate the protein against
centration can be increased. Running conditions may also
the DNA.
greatly affect the mobility. For trp repressor-operator com-
plexes, reducing the pH is necessary to noticeably lessen the 27.2.1.6. Protein Concentration
mobility of the complexes (9). Thus, running a sample of
1. Carefully measure the concentration of plasmid DNA
protein alone (detected by silver staining of the gel) may
containing the binding site of interest by absorbance at 260
show whether the protein runs far into the gel at the pH se-
nm against a blank with the same buffer. An A260 of 1 is
lected, and conditions can be altered to maximize the dif-
equivalent to 50 pg of DNA/ml. The A260/A2&) ratio should
ference in the degrees of mobility of the protein and the free
be approximately 1.8. It is important that the DNA be free
DNA. If the shifted band is near the top of the gel, repeat
of contaminating RNA. RNase treatment is not sufficient
the assay with an agarose gel. Sometimes the free-DNA
since mono- and oligonucleotides will still be present even
band disappears with increasing amounts of protein, but no
after CsCl purification of the DNA. Use DNA that has
complex band appears. In this case, the protein is probably
been purified two times by a CsC1-ethidium bromide gradi-
dissociating during the run. A lower-salt-content gel, differ-
ent method or some other method that ensures the removal
ent binding conditions, or different running buffer may
of RNA.
allow for more-stable complexes.
2. Digest 20 pg of the plasmid DNA with the restriction
A common observation is that a single shifted band is enzyme(s).
observed at one protein concentration and multiple, higher
3. Label a small portion (1 pg) of the DNA by filling in
bands are seen at higher protein concentrations, until all
the ends with the Klenow fragment polymerase as described
the DNA is near the gel origin at the highest concentra-
in section 27.2.1.3.
tions. Since weak protein-DNA interactions are stabilized
4.Inactivate the enzymes in the labeled sample by heat-
ing to 70C for 20 min or by phenol extraction. Remove the
unincorporated label by purification on a 1-ml Sephadex
G-50 column equilibrated with TE buffer or by a silica
method.
5. Inactivate the restriction enzyme in the unlabeled
sample by heating to 70C. Do not use procedures such as
ethanol precipitation that may cause a loss of DNA during
Protein-DNA - handling.
FREE DNA - 6. Mix the labeled and unlabeled samples together. The
molar concentration of the binding site is calculated from
the molecular weight of the plasmid. Since the labeled sam-
ple is only 5% of the total, even a significant loss during
handling will not affect the calculation. If the affinity of the
-0 protein for the specific site is thought to be weak or the
nonspecific binding is strong, protein binding to the vector
Increasing Protein sequences will alter the results. In such cases, at least 1 pg
of the fragment must be purified after restriction cutting
FIGURE 2 Gel mobility shift assay. Shown is an autoradi- and the fragment concentration must be determined spec-
ogram from an assay with a 200-bp DNA fragment correspon- trophotometrically. Do not add carriers such as tRNA or
ding to 500 cpm per sample run on a 6% polyacrylamide gel for BSA since these will interfere with the readings.
1 h at 8 V/cm. Lanes 2 through 5 contain samples with three- 7. Perform the binding assay as described in section
fold increases in levels of protein (purified E. coli AraC pro- 27.2.1.3 by using protein at a level increased by twofold or
tein). Film was exposed overnight at -70C with an intensify- more for each sample. The DNA concentration should be
ing screen. high so that the measurements are above the dissociation
Next Page

27. Nucleic Acid Analysis 663

binding constant of the protein. For most regulatory pro- the concentration and the type of salt in the buffer. Many
teins, a concentration above 10 nM is sufficient, and 100 proteins bind with much higher affinity in a lower salt con-
nM would be sufficient for even very-low-affinity specific centration and with anions such as acetate and glutamate
binding reactions. The binding can be quantitatively mea- than with chloride. All sequence-specific DNA-binding
sured by densitometry of the gel as long as data are within proteins also bind nonspecifically. The nonspecific binding
the linear range for the film. Generally, the film is pre- constant is mostly electrostatic and, therefore, more salt
flashed (cover a camera flash attachment with two layers of sensitive. To maximize specific over nonspecific binding, an
3MM paper and flash from a distance of about 2 ft) so that optimum salt concentration and type can be determined. A
the absorbance at 450 nm is about 0.1. The film is then good starting point is to use 50 mM KC1 and test increasing
tested by exposing a range of labeled standards. Alter- concentrations. A buffer with potassium acetate may also be
natively, the bands may be cut from the gel and measured beneficial.
directly in a scintillation counter. As long as the excised gel
fragments are nearly equal in size, quenching of the radia- 1. Use DNA labeled to an extent corresponding to high
tion for "'-labeled samples is not a problem. The best re- specific activity. Since only 100 cpm per reaction is neces-
sults are obtained by using a phosphorimager system. The sary, DNA concentrations of around lo-" M are easily
linear range for this system is as much as 5 orders of magni- obtained in a 20-pl reaction mixture. For lower concentra-
tude, or 100-fold, greater than that for film. tions, alternative labeling schemes are needed. T4 poly-
8. For the most accurate calculations, plot the following: merase can be used to digest and then fill in one strand of
(1 - fraction of DNA not bound) versus the level of pro- DNA with labeled nucleosides (49), or nick translation may
tein. The fraction of free DNA in each sample should be be used if the procedure is optimized so that intact frag-
calculated by dividing the counts for the sample band by ments are obtained.
the counts for the band from a control lane with DNA but 2. Perform a gel mobility shift assay as described in sec-
with no protein. Since some of the protein-DNA complex tion 27.2.1.5. Allow plenty of time for the reaction to come
may be lost during the gel run, the fraction of DNA in the to equilibrium. The time required for the approach to equi-
complex sometimes is not an accurate reflection of the librium is proportional to the product of the concentrations
amount of DNA bound in solution. The plot should be lin- of the reactants and the association and dissociation rate
ear to a point well beyond that where 50% of the DNA is constants, so at very low concentrations the time needed is
bound. At the point that 50% of the DNA is bound, the greatly extended. Nevertheless, most systems will be at
amount of protein is equal to one-half of the amount of equilibrium within 30 min. Use a wide range of protein con-
DNA, and the active protein concentration can be calcu- centrations, starting with one equal to the estimated con-
lated. It is quite common for highly purified proteins to be centration of the DNA. An easy method is to use 1 and 3
10 to 50% active in specific binding. This method is also pl of each of a series of 10-fold serial dilutions. In this way,
useful for estimating the amount of active protein from par- a range in protein concentrations of 104M can be tested
tially or unpurified samples (see below) as long as the im- with just eight samples. Very carefully load the gel while it
purities do not strongly interfere with the binding reaction. is running to minimize the mixing of the sample with the
running buffer. Since protein binding is salt sensitive, the
27.2.1.7. Equilibrium Binding Constant effects of exposure to a low-salt-content running buffer are
The simplest method to estimate the equilibrium binding a major concern; however, experiments with a number of
constant is to perform a titration step as described in section well-characterized proteins suggest that data from carefully
27.2.1.6; however, in this assay the DNA concentration is performed gel mobility shift assays with low-salt-content
held at a level lower than the equilibrium binding concen- running buffer closely match binding data from other meth-
tration. If the midpoint for binding is obtained under con- ods such as DNase footprinting. At 8 V/cm, a sample in 50
ditions in which the protein is in vast excess relative to the mM KC1 will fully enter the gel in about 1 min. Run the gel
DNA concentration, the equilibrium dissociation constant for sufficient time to resolve the complexes, dry it, and ex-
(kD) is identical to the protein concentration. This princi- pose it to X-ray film. At the lowest levels of the label,
ple follows from the simple binding relationship expressed exposures of several days may be necessary without an en-
+
here: Pf Df = [P * D], where Pf is the free-protein con- hancing screen.
centration, Df is the free-DNA concentration, and [P * D] 3. Determine the fraction of free DNA as described
is the concentration of the complex. The K D can be ex- above, and plot the fraction of free DNA (or the fraction of
-
pressed as follows: K D = PfD/[P D]. At the point where bound DNA) against the active protein concentration. If
50% of the DNA is bound, [b D] equals DF Substituting necessary, correct for any DNA that is not bound even at
D for [P * D] in the equation gives the following: K D = very high protein concentrations. This is sometimes a fac-
56%(Pf). If P, >> D,, where P, and D, are the amounts of tor when using hybridized, synthetic oligonucleotides. If-a
total protein and total DNA, respectively, then the amount densitometry analysis of autoradiograms is performed, the
of protein in the complex is negligible and P, = Pp Thus, linearity of the response must be checked under the condi-
K D = [PJ. If the DNA concentration is 10-fold lower than tions of the binding assay and film exposure. Accurate
that of the protein, Pf is only about 5% lower than the total quantification may also be achieved by assessing the gel
protein concentration and the K D obtained will be well with a beta-scanning device for direct counting or by excis-
within the experimental variation of the assay. The equilib- ing the bands and placing them in a scintillation counter.
rium constant may also be expressed as the equilibrium as- Mincing the gel slices and placing them in an aqueous
sociation constant, K,, which is simply the inverse of KD counting cocktail is sufficient for good counting efficiency
KDs of lop7 M to M for regulatory proteins in E. for 32P-labeledsamples. An alternative, simple method to
coli, have been obtained, with most falling in the range of obtain a reasonable estimate ( ? a factor of two) of the bind-
to (7-10,16,17). The constant may be affected ing affinity is to visually estimate the binding half point. For
strongly by the binding conditions. The electrostatic con- the initial characterization of a protein, such accuracy is
tribution to the binding free energy is highly dependent on usually sufficient.
28
Measuring Spontaneous Mutation Rates
PATRICIA L. FOSTER

28.1. INTRODUCTION ......................................... 676


28.2. TERMINOLOGY .......................................... 677
28.3. ASSUMPTIONS ABOUT MUTATIONAL PROCESSES ............ 677
28.4. MUTANT ACCUMULATION ................................ 677
28.5. FLUCTUATION ANALYSIS ................................. 678
28.5.1. Experimental Design .................................. 678
.2. Analyzing Results of Fluctuation Assays .................... 679
.3. Calculating the Mutation Rate ........................... 681
.4. Statistical Methods To Evaluate Mutation Rates ............... 682
.5. Departures from the Luria-Delbriick and Lea-Coulson Model ..... 682
28.6. REFERENCES ............................................ 682

28.1. INTRODUCTION mutation rates are very low, this number is best understood
Spontaneous mutations are mutations that occur in the ab- as a probability, usually of the order of 1 per lo9 to 10 cell
sence of exogenous causes. They can be due to errors made lifetimes for a given mutation. The mutant fraction or fre-
by DNA polymerases during replication or repair, errors quency is the proportion of cells that are mutant and is typ-
made during recombination, the movement of genetic ele- ically of the order of 1 per lo5 to lo7 cells for a given mu-
ments, spontaneous events such as DNA base loss, and tant phenotype. Although mutant frequencies are adequate
DNA damage from reactive compounds produced by nor- to assess rates of induced mutations, they are very inaccu-
mal cellular metabolism. The rate at which spontaneous rate measures of rates of spontaneous mutations. And, un-
mutations occur can yield useful information about cellular like most biological phenomena, the accuracy of the mea-
processes. For example, the occurrence of specific classes of surement does not improve with the number of replicates.
mutations in various mutant backgrounds has been used to This is the fundamental property of spontaneous mutation
deduce the prevalence and importance of specific DNA re- that was exploited in the famous fluctuation test of Luria
pair pathways (23). Of particular interest are the sponta- and Delbruck (21). The theory is as follows.
neous mutation rates of organisms that live in environ- During the growth of a population, every cell has an
ments so extreme that the coding properties of their DNA equal and constant chance of sustaining a mutation during
should be destroyed. Do these organisms have extremely ef- its lifetime. This probability, which is the mutation rate, is
ficient mechanisms to protect or repair their DNA, or do low, and the number of cells is large, so the numbers of mu-
they simply tolerate a high rate of genetic change (10, 12)? tations sustained among replicate cultures follow the
Accurately determining the rates of mutations due to Poisson distribution. However, once a mutation occurs, the
spontaneous events is more challenging than determining cell sustaining it will produce a clone of mutant progeny, all
the rates of mutations due to exogenously applied DNA- of which will contribute to the number of mutants in that
damaging agents. If a population of cells is subjected to culture. The size of a given mutant clone will depend on
DNA damage at a specific time point, and if the level of when during the growth of the population the mutation
induced mutation is well above the rate of spontaneous occurred; mutations occurring early will produce large
mutation, the frequency of mutants among the survivors re- clones, whereas mutations occurring late will produce small
flects only the number of mutations induced. (It is impor- clones. Thus, the distribution of the numbers of mutants
tant to distinguish a mutation-a heritable change in the among replicate cultures is far from Poisson and is usually
genetic material-from a mutant-an individual that car- referred to as the Luria-Delbriick distribution. Because of
ries a mutation.) Importantly, because spontaneous muta- this basic biology, the frequency of mutants in a culture has
tions are rare relative to induced mutations, the mutant no simple relationship to the mutation rate, but it can be
faction remains a constant as the cells proliferate. In con- used to estimate the mutation rate by various mathematical
trast, the events that give rise to spontaneous mutations manipulations.
occur continuously and so the fraction of cells that are mu- The Luria-Delbruck distribution, which has no closed-
tant due to spontaneous events increases with every divi- form solution, has stimulated the interest of rnathemati-
sion. In addition, as a population proliferates, the time at cians since the original paper describing it was published in
which a spontaneous mutation occurs has a dramatic effect 1943. Unfortunately, methods to calculate mutation rates
on the ultimate mutant fraction; mutations that occur early from this distribution remain opaque to most investigators.
will give rise to jackpots, which are cultures with abnor- This chapter will provide a guide to these methods in the
mally large numbers of mutant individuals. hope that it will be useful to scientists and students who
The mutation rate is the expected number of mutations wish to use mutation rates in their research. In addition,
that an average cell will sustain during its lifetime. Since the Luria-Delbruck distribution applies to other cases in

676
28. Measuring Spontaneous Mutation Rates 677

which a rare initiating event is amplified in a population. 28.3. ASSUMPTIONS ABOUT MUTATIONAL
For example, disease genes can be mapped by using the PROCESSES
Luria-Delbruck distribution to estimate the rate at which All methods to estimate spontaneous mutation rates are
linkage disequilibrium is lost after a founder mutation is in- based on theoretical models of mutational processes and
troduced into a population (13). cell growth. The most common model originated with Luria
There are two basic methods to determine mutation and Delbruck (21) and was further defined by Lea and
rates: mutant accumulation and fluctuation analysis. These Coulson (19); it is usually called the Lea-Coulson model. It
two methods are described below, with an emphasis on fluc- has the following assumptions.
tuation analysis.
The population increases exponentially
The initial number of cells is negligible compared to the
28.2. TERMINOLOGY final number of cells
It is unfortunate that many different symbols have been The probability of mutation is constant per cell lifetime
used for the terms that enter into the equations for calcu-
The probability of mutation per cell lifetime does not vary
lating mutation rates. Most of the terms used here date
during the growth of the culture
from the report by Luria and Delbruck (21) and are given
in Table 1. I t is particularly important to distinguish be- The growth rates of mutants and nonmutants are the same
tween m, the mean number of mutations (mutational The proportion of mutants is always small
events) that occurred during the growth of a culture, and p, The number of reverse mutations is negligible
the mutation rate, which is the probability that a cell will The death rate is negligible
sustain a mutation during its lifetime. Almost all methods
to calculate the mutation rate start by determining m and
All mutants are detected
then obtain p by dividing m by some measure of the num- No mutants arise after selection is imposed
ber of cells at risk. For example, Luria and Delbruck (21) A n additional implicit assumption is that the probability
divided m by the total number of cells, N,, to give the mu- that a mutation occurs is not influenced by previous muta-
tation rate. tional events, which is the randomness required for the
It is also important to distinguish between the number of Poisson distribution. Various theoretical papers have ad-
mutations per culture, m, and the number of mutants per dressed how departures from the Lea-Coulson model affect
culture, r. The number of mutants derives from both the the distribution of mutant numbers. In general, most of the
number of new mutants that arose during the growth of a models requirements can be met with proper experimental
culture and the clonal expansion of the progeny of each protocols, but some departures reflect real biological phe-
founder mutant. T is what we can actually observe and use nomena.
to calculate m. r divided by N,is the mutant fraction or mu-
tant frequency, f.
28.4. MUTANT ACCUMULATION
After an exponentially growing population reaches a suffi-
cient size so that m is >> 1, the accumulation of mutants
TABLE 1 Definition of terms becomes a simple function of the cell number (20). As il-
lustrated in Fig. l, the combination of new mutants and the
Term Definition growth of preexisting mutants results in a constant increase
in the mutant fraction; the mutation rate is equal to this
m . . . . . . . Number of mutations per culture increase divided by the number of individuals present.
p . . . . . . . Mutation rate; probability of mutation per cell per Therefore, the conceptually simplest of all ways to deter-
division or generation mine a mutation rate is to measure the change in the mu-
N o . . . . . . Initial number of cells in a culture; the inoculum tant fraction in a growing population. Considering the pop-
N, . . . . . . Final number of cells in a culture ulation growth to be continuous, p can be calculated by
using equation 1 (9):
r . . . . . . . Number of mutants in a culture
5. . . . . . . . Mean number of mutants per culture
i: . . . . . . . Median number of mutants per culture
f . . . . . . . Mutant fraction or frequency; r/N = ( h- A ) + ( l n N 2 - l n N l )
V . . . . . . . Volume of a culture
There are important limitations to the use of this method.
C . . . . . . . Number of cultures in an experiment
By the time the population reaches a size large enough to
po . . . . . Proportion of cultures without mutants
I
allow the mutant fraction to be measured, mutations have
p7 . . . . . . . Proportion of cultures with r mutants already occurred and their numbers will be subject to the
P, . . . . . . Proportion of cultures with r or more mutants fluctuation effect. In many cases, these fluctuations will
z . . . . . . . Dilution factor or fraction of a culture plated make it impossible to accurately measure the accumulation
of new mutants. However, the mutant accumulation method
Q,. . . . . . Value of r at 25% of the ranked series of rs can be used successfully when the number of mutations
Q,. . . . . . Value of r at 50% of the ranked series of rs; the median within a large population can be kept low. For example, sta-
Q3 . . . . . . Value of r at 75% of the ranked series of rs ble stocks containing large numbers of bacteriophages or
c, . . . . . . . Number of cultures with r mutants viruses can be generated and tested for mutants; the stocks
u . . . . . . . Standard deviation with low mutant frequencies can then be used repeatedly
for experiments. Mutant accumulation can also be used if a
CL . . . . . Confidence limit
population can be purged of preexisting mutants by using
678 MOLECULAR GENETICS

Generation Cells New Growth of Pre-existing Total Mutant Change per

=k =N Mutants Mutants Mutants Fraction Generation

=r = r/N =P

4 Nff16 1 16lNt 16lNt

3 NU8 2
I \ 4 32lNt 16lNt

2 NU4 \ :\4 12 48lNt 16lNt

1 NU2 8 \\\
4 32 64lNt 16lNt

0 Nt 16 \ 1:\1:\1h16 80 80/Nt 16lNt

FIGURE 1 Illustration of the constant increase in the mutant fraction after a population reaches
a size sufficiently large that the accumulation of mutants is simply a function of population size.
Lurias conventions are followed (20): k, the generation numbered backwards from 0; N, the num-
ber of cells present at each generation; N,, the final number of cells in the population; p, the
mutation rate per cell (assuminga synchronous population). At each generation, there are NJ2k in-
dividuals that produce ~ N t / new
2 ~ mutations, which will produce a total of pN, mutant progeny by
the last generation. Adapted from reference 10a with permission of the publisher.

a mutational target that can be selected both for and against a measure of reproducibility, not accuracy. Accuracy is
(i.e., a counterselectable marker). For example, if a popula- how well the resulting estimate reflects the actual muta-
tion of Lac- cells carrying a temperature-sensitive galE mu- tion rate, and that will depend on the applicability of the
tation is incubated in lactose medium at the nonpermissive underlying assumptions. The parameters that are impor-
temperature, Lac+ revertants will die (because one product tant in designing a fluctuation assay are as follows: m, the
of P-galactosidase is galactose and the accumulation of number of mutations per culture; T , the number of mutants
UDP-galactose is toxic in the absence of the epimerase en- per culture; No, the inoculum; N,, the final number of
coded by galE); if the population is then shifted to the per- cells; V, the culture volume; and C, the number of parallel
missive temperature, the accumulation of new Laci mu- cultures. Preliminary experiments should be done to de-
tants can be monitored (24). Another possibility is to use a termine the range of rs obtained when a given number of
mutational target that allows mutants to be eliminated by cells is plated. These experiments consist simply of grow-
cell sorting. For example, mutants that allow green fluores- ing three to five cultures in the desired medium, plating
cent protein to be produced can be eliminated by fluores- aliquots of various sizes onto the selective medium, and
cence-activated cell sorting; new mutants can then be de- determining the number of mutants per culture obtained.
tected by flow cytometry (4). A provisional m can then be estimated from the median of
these experiments (by using method 3 or 4;see below),
and this value can be used to adjust the other parameters.
2 8.5. FLUCT UATlON A NA LYS IS In general, m should be between 0.3 and 15; at low values
of m, the error is large, but if m is above 15, some of the
28.5.1. Experimental Design more convenient methods to calculate the mutation rate
A normal fluctuation assay begins by inoculating parallel are not valid (11, 2 5 ) . Obviously, m also has to be within
cultures with a population of cells that is small enough to be a range so that the number of mutants per plate is count-
unlikely to contain any preexisting mutants. The cultures able; however, if the number of mutants per plate
are allowed to grow, usually to saturation, and the total is in the typical range of 0 to 50, there is little loss in pre-
number of cells is determined by plating appropriate dilu- cision if occasionally high colony counts are truncated at
tions onto nonselective medium. The number of mutants a value of about 150 (3, 15).
is determined by plating each culture onto a selective The desired m can be achieved by adjusting N,, the final
medium, where each mutant produces a colony. The muta- number of cells. When defined medium is used, this adjust-
tion rate is calculated from the distribution of the numbers ment can be done by limiting the carbon source. However,
of mutants among the parallel cultures. This basic design it is not recommended to limit the cells with other growth
can be used for single-celled microorganisms, cultured cells, factors, such as vitamins or amino acids, because the cell
bacteriophage, and viruses. Selection usually takes place on physiology may change and mutants that do not require the
solid medium so that individual mutants can be enumer- growth factor may arise. The alternative to limiting cell
ated; however, the method utilizing the proportion of cul- growth is to adjust the volume of the culture to obtain the
tures without mutants (PO; see below) can be carried out desired N,. It has long been considered the best practice to
with liquid medium as well. subject the entirety of each culture to selection because
Proper design of a fluctuation assay will maximize the sampling introduces errors. Sampling or low plating effi-
precision of the estimate of the mutation rate. Precision is ciency (which is the same thing) narrows the distribution of
28. Measuring Spontaneous Mutation Rates 679

the number of mutants, increasing the influence of small 28.5.2. Analyzing Results of Fluctuation Assays
numbers and decreasing the influence of large numbers (7, A fluctuation assay yields a distribution of the number of
28). However, a recent analysis has concluded that it is bet. mutants per culture, T , which is used to calculated m, the
ter to plate a small aliquot from a large culture than all of a mean or most likely number of mutations per culture. It is
small culture because the large culture contains more infor- helpful to remember that although the distribution of mu-
mation than the small culture (15). In addition, if several tants is not Poisson the distribution of mutations is, so m is
mutant phenotypes are to be assayed simultaneously, sam- a Poisson parameter. All the methods to calculate m depend
pling may be unavoidable. Simple corrections for sampling on the theoretically described distribution of mutant clone
can be applied when using some, but not all, of the various sizes, but the methods differ in the mathematics used to
methods to calculate mutation rates. obtain m from the experimentally obtained distributions.
It is extremely important that when selection is applied, The equations and parameters used to calculate m are usu-
N,be the same for each culture. Usually this condition can ally called estimators. Over the last 50 years, the methods
be ensured by growing cells to saturation, although care have improved but the complexity of the calculations has
should be taken when the strain used has growth defects. increased, and access to a computer is required for some of
Also, in some cases it may be desirable to use exponentially the more complex calculations. The MSS maximum-likeli-
growing cells. Cell numbers can be monitored before mu- hood method (method 8) is the gold standard because it
tant selection by measuring the optical density or by count- utilizes all of the results of an experiment, it can be used
ing the cells microscopically (e.g., by using a Petroff- over the entire range of mutation rates, and it yields results
Hausser chamber); however, these measurements should be that can be statistically evaluated. Of the less complicated
confirmed by plating to determine the numbers of viable methods, the Lea-Coulson method of the median (method
cells. Presently there is no valid method to correct mutant 3 ) and the Jones median estimator (method 4) are reliable
numbers for different N,s, so deviant cultures must be elim- if mutation rates are low to moderate; if mutation rates are
inated from the analysis (11). very low (m 5 l ) ,only the po method (method 1) is appli-
To easily calculate m, the initial inoculum, No, must be cable (11, 25).
negligible compared to N,. A ratio of 1/1,000 is usually suf-
ficient if m is 5 1 0 (26), but a smaller inoculum may be
needed to ensure that there are no preexisting mutants. Method 1:the p o Method
Obviously, with a small inoculum, the cultures will take Since the distribution of the numbers of mutations per cul-
longer to saturate and viability may be a problem, which ture is Poisson, the mean can be calculated from po, the pro-
may increase the variation of N,s. A reasonable rule of portion of cultures with no mutants (because if there are no
thumb is that Noshould be approximately equal to ( N J m ) mutants, there were no mutations) (21).
x (11). Since
It is a common practice to begin a fluctuation analysis po = eCm (2)
by inoculating each culture with a single colony of the
strain being tested. There are two problems with this pro- Then
cedure. First, Nos will vary among the cultures. Second, a m = -lnpo (3)
colony has undergone the same process of mutant accumu-
lation as a culture, and each colony will contain a different The great advantage of the po method is its simplicity; N,is
number of mutants. Thus, some of the cultures in the fluc- adjusted so that about a third of the cultures have no mu-
tuation test may be inoculated with preexisting mutants. A tants, and m is calculated from equation 3. Although sim-
better practice is to dilute one culture or colony into an ap- ple, the method is limited. Obviously, it can be used only
propriate volume of fresh medium and then distribute when the number of mutants is small, and the error of small
aliquots into the individual cultures for nonselective numbers is large. The range of usefulness of the po method
growth. is: 0.3 5 m 5 2.3 (0.7 2 po 2 0.1). No method is reliable
Because the precision of the estimate of m depends on when m is <0.3 unless hundreds of cultures are used, and if
the number of parallel cultures, C, the larger C is, the bet- m is 22.3, there are too few cultures with no mutants to use
ter the estimate is. Most experiments have 10 to 100 paral- the po method. In addition, the po method is inefficient
lel cultures, with numbers around 40 being most common. (i.e., it requires more cultures for the same precision) com-
There is little gain in precision if C is increased above this pared to other methods (11, 18, 25).
level unless m is less than about 0.3 (11, 15, 25). The po estimator is also sensitive to factors other than
In experiments with nonlethal selections, for example, the mutation rate that increase po. For example, if the phe-
the reversion of an auxotrophy or the utilization of a carbon notype being selected is not immediately expressed (i.e., it
source, mutants can arise after selection has been applied. shows phenotypic lag), mutations that occur in the last few
These mutants can be the result of mutations occurring as generations will not produce clones and, thus, the po will be
the cells proliferate on the selective medium, or the result inflated. Likewise, if mutants die or have a poor plating ef-
of mutations occurring in the nongrowing cells (adaptive ficiency, po will be increased. However, if only a fraction of
mutations). In either case, the m estimated for preplating all cells (not just mutants) is detected and this fraction is
mutations will be inflated by these postplating mutations. If known, a correction factor can be applied to m. For exam-
nonmutant cells can grow on the selective medium because ple, if only a fraction, z,of a culture is plated, the po will be
of contaminating nutrients, the problem can be solved by increased, but the m calculated from this observed po (mobs)
adding an excess of scavenger cells that cannot mutate (be- can be corrected to give the actual m (matt) by using equa-
cause, for example, they have a deletion of the relevant tion 4 (14, 28).
gene) (5). If the cells proliferate because the selection is
not stringent, the time it takes for a mutant colony to form (4)
on the selective medium can be determined and then
colonies can be counted at the earliest possible time after Note that when z is 1, (7 - l)/[z In (731 equals 1 from
plating. lH6pitals rule and macrequals mobs.
680 MOLECULAR GENETICS

Method 2: the Method of the Mean tribution with its median equal to 0.5 (which also means
The method of the mean was first formulated by Luria and that po equals 0.5). The two advantages of the Jones esti-
Delbruck (21). Rearranging their equation gives the follow- mator are that it is explicit and that it accommodates dilu-
ing: tions. The basic equation is as follows:
T = m ln(mC) (5) r" - ln(2) - ? - 0.693
m= (9)
where f is simply the mean number of mutants per culture In(?) - ln[ln(2)] - ln(?) + 0.3665
and C is the number of cultures. This transcendental equa- If2: is the dilution factor (i.e., the fraction of the culture
tion can easily be solved by iteration by using the following that is plated) or the plating efficiency, then the observed
rearrangement: median, fobs, can be used in the following equation:
m ln(mC) - F = 0 (6)
This method is very easy to apply but, like all methods re-
lying on the mean, has severe limitations. Because the dis-
tribution of mutant numbers is inherently skewed even
without obvious jackpots, the mean will be dominated by
the few cultures with large numbers of mutants. Luria and
Delbruck attempted to eliminate this problem by consider- The Jones median estimator method was verified with sim-
ing only mutations that occur after the population reaches ulated data for the range of m from 1.5 to 10 (1.5 5 m 5 10;
a size sufficiently large that the accumulation of mutants is
3 5 r" 5 40) and proved to be as reliable as and more effi-
cient than the Lea and Coulson median estimator (16). I t
deterministic (see above) (Fig. 1).They took this to be the
point when NC, the whole population size in the experi- also performs well with real data (11, 25).
ment (i.e., all the cells in all the cultures), equals l/p.
Method 5: Drake's Formula
Alternatively, Armitage (2) considered the deterministic
Drake's formula (8) provides an easy way to calculate muta-
period to begin when the population of each culture reaches
tion rates from mutant frequencies and is especially useful
l/p, in which case the equation is as follows:
in comparing data from different sources. Starting from
mln(m)-f=O (7) equation 1 above, Drake sets N1 to be not the initial inocu-
lum, but l/p, which is the population size when the proba-
Obviously, mean estimators should not be used to calcu-
bility of mutation approaches unity. Setting fi as zero and fi
late mutation rates when N,is ~ l / p i.e.,
, when m is 51. In
as the final mutant frequency gives the following:
addition, the method assumes that no mutations occur be-
fore the deterministic period, which is unlikely to be true. u = f + ln(uN,) (11)
As a result, methods using the mean will frequently overes-
which can be solved for p by iteration. Drake's formula is
timate mutation rates. If jackpots could be eliminated, then
based on the same assumption as the Luria-Delbruck
the mean might be useful, but there is no way to nonarbi-
method of the mean, i.e., that mutations occur only during
trarily decide what is or is not a jackpot. The use of the me-
the deterministic period of mutant accumulation. To mini-
dian instead of the mean improves the method somewhat
(11, 25). mize the influence of jackpots, the median frequency is used
instead of the mean (8). By setting p equal to m/N,, Drake's
Method 3: Lea-Coulson Method of the Median formula can be rearranged into a form similar to that of Lea
Lea and Coulson (19) observed that for m over the range and Coulson's (see equation 8) as follows (25):
from 4 to 15, a plot of the probability that a culture contains ?
r or fewer mutants versus [r/m - ln(m)] gives a smooth -- ln(m) = 0
curve with a median of 1.24. Rearranging gives the follow- m
ing transcendental equation: Because of its derivation, Drake's formula is best used only
r
- when mutation rates are high. When m is <4, the estimates
- - ln(m) - 1.24 = 0 (8) of m from equation 12 are significantly higher than those
m from equation 8. As m becomes larger, values calculated from
where r" is the median number of mutants per culture. This equation 12 asymptotically approach those calculated from
equation can easily be solved by iteration. The Lea and equation 8; the differences are trivial when m is 230 (11,25).
Coulson method of the median is easy to apply and remark-
ably accurate as judged by its performance in computer sim- Method 6: Accumulation of Clones
ulations (3, 27). It performs well within the range 1.5 5 m Luria (20) pointed out that P,, the proportion of cultures
5 15 (2.5 5 f 5 60) (11, 25). Because it uses the median, with r or more mutants, approaches 2m/r during the deter-
it is relatively insensitive to deviations that affect either ministic portion of growth. Formally,
end of the distribution. For example, experiments can be i=N,
designed so the f is large enough to be unaffected by phe- 2m
notypic lag. However, the method of the median compares
-
p,=cp;=-
I
i=r
unfavorably to the maximum-likelihood method (method
8) because little of the information obtained from the fluc- Taking logarithms gives the following:
tuation test is used, making the method relatively ineffi-
cient (11, 25).
ln(P,) = -ln(r) + ln(2m) (14)
Thus, a plot of ln(P,) versus ln(r) will yield a straight line
Method 4: the Jones Median Estimator with a slope of - 1 and an intercept [where h ( r ) = 01 equal
Jones et al. (16) derived an estimator based on the hypo- to ln(2m) (Fig. 2). Dividing P,. at the intercept by 2 gives m
thetical dilution that would be necessary to produce a dis- (11, 25).
28. Measuring Spontaneous Mutation Rates 681

1
I are so rare that they contribute little to the distribution and
can be lumped into one category (3). The likelihood func-
0-. * - tion, equation 19, is the product of these calculated pr)~for
each culture. The easiest way to do this calculation is to
arrange the mutant counts from an experiment in order and
count the number of cultures with each r number of mu-
tants. Then the product of the pr)s is as follows:

where each p,. is from equation 18 and c, is the number of


cultures that had r mutants (note that because equation 18
is recursive, r's that were not obtained enter into the calcu-
lation of the pr)s, but because these give a value of 1 in equa-
0 1 2 3 4
tion 19, they do not have to appear in equation 20).
Alternatively, the values of c, times ln(p,) for each r can be
Ln (r) added. Several m's over a small range are evaluated to find
FIGURE 2 ln(P,) versus ln(r) for the Luria-Delbruck distri- the m that maximizes the likelihood function.
bution. The distribution (solid line) was calculated for m of 1 As mentioned above, the MSS maximum-likelihood
by using equation 18. A curve (dotted line) has been fitted by method is the best method presently available to estimate
the least-squaresmethod to the upper part of the distribution m. Unlike the other methods discussed above, except
[ln(r) 2 21; it has a slope of - 1.2 and an intercept of 0.6, giv- methods 6 and 7, the maximum-likelihood method uses all
ing an m of 0.9. the results from a fluctuation experiment. Because of its der-
ivation, it is valid over the entire range of mutation rates
(although the calculations become tedious at high values of
r). In addition, simulations have shown it to perform in a
Method 7: the Quartiles Method manner regular enough to allow for statistical evaluation
The median is the central (50%) quartile of a distribution (27).
and is the parameter used in most of the methods described
above. More of the data from a fluctuation assay are incor- 28.5.3. Calculating the Mutation Rate
porated into the calculation of m if the upper (75%) and The mutation rate, F, is calculated by dividing m, which is
lower (25%) quartiles are also used. By regressing m versus an estimate of the number of mutations that occurred dur-
the theoretical values of r at the quartiles, Koch (17) de- ing the growth of a culture, by some measure of the number
rived the following empirical equations: of cells at risk for mutation during that growth. There are
mi = 1.7335 + 0.4474Q1 - 0.002755(Q1)' (15) three such measures, each of which reflects different as-
sumptions about the underlying mutational process.
m2 = 1.1580 + 0.2730Q2 - 0.000761(Q2)2 (16) Luria and Delbruck (21) and most theorists assume that
7713 = 0.6658 + 0.1497Q3 - 0.0001387(Q3)' (17) the probability of mutation is constant over the cell divi-
sion cycle. Since the final number of cells in a culture, N,,
where Q1,Qz, and Q3 are the values of r at 25,50, and 75% arose from (N, - 1) divisions, the mutation rate is as fol-
of the ranked series of observed 7's. For a perfect Luria- lows:
Delbruck distribution, the three m's should be equal. These
equations are valid over the range of m from 2 to 14; Koch
also gives graphs that can be used up to values of m of 120
(17).
The same calculation applies if mutations are assumed to
Method 8: the MSS Maximum-likelihood Method occur at the end of the division cycle (i.e., at or during di-
Sarkar et al. (26) derived the following recursive algorithm vision), so that the number of "opportunities" for mutation
that efficiently computes the Luria-Delbriick distribution is equal to the number of divisions (1).
for a given value of m If mutations are assumed to occur at the beginning of the
division cycle (i.e., shortly after division), then the number
of cells at risk is equal to the total number of cells that ever
existed in the culture, which is 2N, (because N,cells had
where pr is the proportion of cultures with r mutants. This NJ2 parents and NJ4 grandparents, etc., and the sum of the
algorithm can be used to estimate m from experimental re- series is ZN,) . Thus, the mutation rate (1) is as follows:
sults by using the MMS maximum-likelihood method (22),
the formula for which is as follows: (22)

Finally, microorganisms rarely grow synchronously.


During one generation period for an asynchronously expo-
nentially growing population, there is an average of N/ln( 2)
where f(ri I m) equals p, from equation 18. The procedure is cells. Thus, the total number of divisions during the growth
to start with a trial m (obtained from equation 8, for exam- of a culture is NJln(2) and the mutation rate (1) is as fol-
ple) and use equation 18 to calculate the probability, pr, of lows:
obtaining each possible r from 0 to the maximum value ob-
tained (even if a given r was not obtained in the experi-
ment). For most experiments, values of r greater than 150
682 w MOLECULAR GENETICS

For the same m, these three methods will give mutation bility on the model of expansion of mutant clones originally
rates that differ by 1, 0.5, and 0.693. described by Luria and Delbruck (21) and extended by Lea
and Coulson (19). If the assumptions of the model are not
28.5.4. Statistical Methods To Evaluate true for a particular application, then the calculated muta-
Mutation Rates tion rate will be wrong. Several meaningful biological de-
The estimates of m or p. obtained from fluctuation tests are partures have been considered by theorists and can, with
neither normally distributed nor unbiased; therefore, nor- some care, be accounted for to derive a meaningful muta-
mal statistics cannot be used to evaluate the results (3, 16, tion rate.
27). No matter how many times a fluctuation experiment is A plating efficiency for all cells (not just mutants) of less
repeated, it is simply not valid to take the mean and stan- than 100% has the same effect as plating only samples of
dard deviation of the results. There are two general ap- the cultures: all clones will be reduced in size by the same
proaches that can allow reasonable confidence limits to be relative amount. The actual m can be calculated by using
placed around mutation rate estimates. equation 4 or 10, where 9 is either the fraction of the cul-
If the parameter that was used to calculate m has a de- ture that is plated or the fraction of the cells that can grow
fined distribution, that distribution can be used to evaluate after plating.
m. For example, because a culture either has mutants or Phenotypic lag means that the expression of a mutant
does not, po (but not m) can be considered a binomial pa- phenotype is delayed for several generations. As a result,
rameter (19). The confidence limits for a binomial parame- clones that arise in the last few generations of growth will
ter can be found by using standard statistical methods, and contribute few mutants, but clones that arise early during
the new po at each limit can be used to calculate new m's. growth will contribute a normal number of mutants. Thus,
These new m's are estimates of the confidence limits for m the lower end of the distribution will be affected, but de-
as determined by the po method. Similarly, when the me- pending on the length of the lag, the upper end will not, re-
dian is used to estimate m, standard methods can be used to sulting in an inflated m. The actual m can be estimated
calculate confidence limits for the median and then m can graphically with method 6 by using only the upper part of
be recalculated using these median limits to estimate the the curve and eliminating any obvious jackpots (11, 25)
confidence limits for m (11, 25). (Fig. 2). If the length of the phenotypic lag can be esti-
The second approach to statistically evaluating m is to mated, Koch (17) provides a method for estimating m from
find a transforming function that gives m a normal distribu- the quartiles (method 7). First, the length of the phenotypic
tion. Using simulated fluctuation tests, Stewart (27) com- lag is estimated in number of doublings, n. Then the three
pared the distributions of m's obtained by several methods. experimental r values that are the points 2"-fold less than
These results indicated that the logarithms of m's obtained the actual quartiles are used to derive provisional m's from
by using the MSS maximum-likelihood method (equation equations 15 through 17 or from the graphs given in refer-
19) are approximately normally distributed. From this, ence 17. Different values of n can be tried until the three
Stewart calculated the standard deviation, u, of the loga- m's have similar values. Then, the provisional m is multi-
rithm of In(m) to be as follows: plied by (2" - 1) to give the actual m.
If during nonselective growth mutants grow more slowly
that nonmutants, mutant clones arising early during the
growth of a culture will be reduced in size, whereas clones
arising late will be more normal. This occurrence shifts the
where C is the number of cultures. Since In(m) is normally
distribution of mutant numbers from the Luria-Delbruck to-
distributed, the 95% confidence limits for ln(m) should be
ward the Poisson (17, 28). Koch (17) gives graphs that can
ln(m) t 1.96u1,,,) be used to estimate m from the quartiles when the relative
growth of mutants ranges from 0.6 to 0.9 of that of the non-
While this is a reasonable approximation, the true confi-
mutants. If there is more than one type of mutant and each
dence limits must be calculated from the actual m and u of
type has a different growth rate, the distribution can be ap-
the population, not the experimentally determined m and
proximated by assuming that there is only one type whose
u. Methods to calculate or estimate the correct confidence
growth rate is the average (28).
limits are given by Stewart (27). A close approximation can
Deviations from the Luria-Delbruck distribution can be
be obtained from the following equations (11, 25)
detected as distortions of the curve defined by equation 14
(Fig. 2). For example, if mutations occur both during non-
selective growth and after selection is applied, a plot of
CLP95%= In(m) - 1.96u(e1.96")f0"'5 (27) ln(P,.) versus ln(r) gives a curve that is a combination of the
Once the upper and lower limits for ln(m) are obtained, the Luria-Delbruck and Poisson distributions (6). The effects
upper and lower limits form are simply the antilogarithms. that several other deviations from the Luria-Delbruck and
Once confidence limits are obtained for m, these can Lea-Coulson assumptions have on the shape of this curve
then be divided by N,(or 2N, or NJln 2) to estimate the have been modeled previously (28). Experimental data can
confidence limits for p., the mutation rate. Of course, this be fit to these curves to test whether a given factor, such as
procedure ignores the variance associated with the determi- selection against mutants, is operative. However, all of the
nation of N,, which is approximately equal to N,.Although deviant curves are rather similar, so any conclusion that a
this variance is nontrivial, most theorists consider it to be given factor is distorting the distribution would have to be
negligible, and it is probably justifiable to ignore it as long tested experimentally.
as N,is determined accurately.
28.6. REFERENCES
28.5.5. Departures from the Luria-Delbriick 1. Armitage, P. 1952. The statistical theory of bacterial pop-
and Lea-Coulson Model ulations subject to mutation. J. R. Stat. SOC.B 14:1-40.
All the methods for calculating mutation rates from fluctu- 2. Armitage, P. 1953. Statistical concepts in the theory of
ation tests discussed above are dependent for their applica- bacterial mutation. J. Hyg. 51:162-184.
28. Measuring Spontaneous Mutation Rates 683

3. Asteris, G., and S. Sarkar. 1996. Bayesian procedures for 16. Jones, M. E., S. M. Thomas, and A. Rogers. 1994. Luria-
the estimation of mutation rates from fluctuation experi- Delbruck fluctuation experiments: design and analysis.
ments. Genetics 142:313-326. Genetics 136:1209-12 16.
4. Bachl, J., M. Dessing, C. Olsson, R. C. von Borstel, and 17. Koch, A. L. 1982. Mutation and growth rates from Luria-
C. Steinberg. 1999. An experimental solution for the Delbriick fluctuation tests. Mutat. Res. 95:129-143.
Luria Delbruck fluctuation problem in measuring hyper- 18. Koziol, J. A. 1991. A note of efficient estimation of muta-
mutation rates. Proc. Natl. Acad. Sci. USA 96:6847-6849. tion rates using Luria-Delbruck fluctuation analysis.
5. Cairns, J., and P. L. Foster. 1991. Adaptive reversion of a Mutat. Res. 249:275-280.
frameshift mutation in Escherichiu coli. Genetics 128:695- 19. Lea, D. E., and C. A. Coulson. 1949. The distribution of
701. the numbers of mutants in bacterial populations.J. Genet.
6. Cairns, J., J. Overbaugh, and S. Miller. 1988. The origin 49: 264-285.
of mutants. Nature 335:142-145. 20. Luria, S. E. 1951. The frequency distribution of sponta-
7. Crane, B. J., S. M. Thomas, and M. E. Jones. 1996. A neous bacteriophage mutants as evidence for the exponen-
modified Luria-Delbruck fluctuation assay for estimating tial rate of phage reproduction. Cold Spring Harbor Symp.
and comparing mutation rates. Mutat. Res. 354:171-182. Quunt. Biol. 16:463-470.
8. Drake, J. W. 1991. A constant rate of spontaneous muta- 21. Luria, S. E., and M. Delbriick. 1943. Mutations of bacte-
tion in DNA-based microbes. Proc. Natl. Acud. Sci. USA ria from virus sensitivity to virus resistance. Genetics 28:
88:7 160-7 164. 49 1-5 11.
9. Drake, J. W. 1970. The Molecular Basis of Mutation. 22. Ma, W. T., G. v. H. Sandri, and S. Sarkar. 1992.Analysis
Holden-Day, Inc., San Francisco, CA. of the Luria-Delbruck distribution using discrete convolu-
10. Fitz-Gibbon, S. T., H. Ladner, U. J. Kim, K. 0. Stetter, tion powers. J. Appl. Prob. 29:255-267.
M. I. Simon, and J. H. Miller. 2002. Genome sequence of 23. Miller, J. H. 1996. Spontaneous mutators in bacteria: in-
the hyperthermophilic crenarchaeon Pyrobaculum aero- sights into pathways of mutagenesis and repair. Annu. Rev.
philum. Proc. Natl. Acud. Sci. USA 99:984-989. Microbiol. 50:625-643.
lOa.Foster, P. L. 1999. Sorting out mutation rates. Proc. Natl. 24. Reddy, M., and J. Gowrishankar. 1997. A genetic strat-
Acud. Sci. USA 96:6862-6867. egy to demonstrate the occurrence of spontaneous muta-
11. Foster, P. L. 2006. Methods for determining spontaneous tions in non-dividing cells within colonies of Escherichia
mutation rates. Methods E n ~ m o l409:195-213.
. coli. Genetics 147:991-1001.
12. Grogan, D. W. 2000. The question of DNA repair in hy- 25. Rosche, W. A., and P. L. Foster. 2000. Determining mu-
perthermophilic archaea. Trends Microbiol. 8:180-185. tation rates in bacterial populations. Methods 20:4-17.
13. Hastbacka, J., A. de la Chapelle, I. Kaitila, P. Sistonen, 26. Sarkar, S., W. T. Ma, and G. v. H. Sandri. 1992. On fluc-
A. Weaver, and E. Lander. 1992. Linkage disequilibrium tuation analysis: a new, simple and efficient method for
mapping in isolated founder populations: diastrophic dys- computing the expected number of mutants. Genetica
plasia in Finland. Nut. Genet. 2:204-211. 85:173-179.
14. Jones, M. E. 1993. Accounting for plating efficiency when 27. Stewart, F. M. 1994. Fluctuation tests: how reliable are the
estimating spontaneous mutation rates. Mutat. Res. 292: estimates of mutation rates? Genetics 137:1139-1146.
187-189. 28. Stewart, F. M., D. M. Gordon, and B. R. Levin. 1990.
15. Jones, M. E., S. M. Thomas, and K. Clarke. 1999. The Fluctuation analysis: the probability distribution of the
application of a linear algebra to the analysis of mutation number of mutants under different conditions. Genetics
rates. J. Theor. Biol. 199:ll-23. 124: 175-185.
Transposon Mutagenesis
SILVIA ROSSBACH AND FRANS J . DE BRUIJN

29.1. Tn5 AS A MODEL TRANSPOSON ............................ 686


.2. RANDOM Tn5 MUTAGENESIS .............................. 687
29.2.1. Phage X Suicide Vectors ............................... 687
29.2.1.1. Phage Lysate Preparation ....................... 688
.
2. Infection and Mutant Selection ................... 688
29.2.2. Plasmid Suicide Vectors ............................... 688
29.2.2.1. Diparental Conjugation and Mutant Selection ........ 689
.
2. Triparental Conjugation and Mutant Selection ........ 689
.
3. Mutagenesis Protocols and Mutant Characterization .... 690
29.2.3. Physical Mapping of Insertions .......................... 690
29.2.3.1. Isolation of Genomic DNA ..................... 690
.
2. Southern Blotting and Hybridization ............... 690
29.2.4. Genetic Mapping of Insertions and Tn5-Induced Mutations ...... 691
.5 . Cloning of TnS-Mutated Genes .......................... 691
29.3. REGION-DIRECTED Tn5 MUTAGENESIS ..................... 692
29.3.1. Vectors ........................................... 692
29.3.1.1. Phage X::TnS ............................... 692
.2. Mutagenesis Protocol and Mutant Characterization .... 693
29.3.2. Physical Mapping of Plasmid-Borne Insertions ............... 693
.3. Correlated Physical and Genetic Maps ..................... 693
.4. Gene Replacement Vectors and Methods ................... 693
29.3.4.1. Broad+Host+Rangeand Incompatible Plasmids ........ 694
.2. Narrow-Host-Range Plasmids .................... 694
.3. Screening for Double Crossovers ................. 694
29.4. USES OF Tn5 DERIVATIVES ................................ 695
29.4.1. Reporter Gene Fusions ................................ 695
.
2. Secondary Transposon Mutagenesis for the Identification
of Regulatory Genes .................................. 696
.
3. Portable Promoters ................................... 696
.
4. Transfer and Replication Origins ......................... 698
.
5 . Alternative Marker Genes ............................. 698
.
6. Primers for DNA Sequencing and PCR .................... 699
.
7. Genome Mapping .................................... 700
.
8. In Vitro Transposition ................................ 700
.
9. Protein Structure-Function Analysis Using Transposons ........ 701
. l 0. Signature-Tagged Mutagenesis........................... 701
29.5. RECIPES FOR MEDIA ..................................... 702
.6. REFERENCES ............................................ 702

Transposable elements are distinct DNA segments that general reviews. see the books edited by Berg and Howe
have the unique capacity to move (transpose) to new sites (21) and Craig et a1. (47).
.
within the genomes of their host organisms The transposi- Prokaryotic transposable elements can be divided into
tion process is independent of the classical homologous different groups. One group consists of simple elements
recombination (rec) system of the organism. Moreover. the such as insertion sequences (IS elements). which are ap-
insertion of a transposable element into a new genomic proximately 800 to 1.500 bp in length . IS elements nor-
site does not require extensive DNA homology between mally consist of a gene encoding an enzyme required for
the ends of the element and its target site. Transposable el- transposition (transposase). flanked by terminally repeated
ements have been found in a wide variety of prokaryotic DNA sequences which serve as substrate for the trans-
and eukaryotic organisms. where they can cause null muta- posase . IS elements were initially identified in the lactose
tions. chromosome rearrangements. and novel patterns of (lac) and galactose (gal) utilization operons of enteric bac-
gene expression upon insertion into the coding regions or teria. where they were found to cause often unstable. polar
regulatory sequences of resident genes and operons. For mutations upon insertion (198).

684
29. Transposon Mutagenesis 685

Traditionally, structurally more complex transposable el- ences 35, 150, and 206 for reviews and reference 141 for the
ements (transposons, or T n elements) have been assigned complete DNA sequence of phage Mu.
to three different classes. Class I transposons in prokaryotes Recently, other mobile elements have been discovered
have been defined as a class of composite transposons that that do not fit into these three categories. They include in-
contain IS elements (or parts thereof) as direct or inverted tegrons, conjugative transposons (CTns), mobilizable trans-
repeats at their termini. Tn5 and TnlO are members of this posons (MTns), and retrotransposons (213). Integrons usu-
class of transposons. Class I transposons behave formally ally consist of a gene encoding an integrase enzyme, a strong
like IS elements but carry additional genes unrelated to promoter, and a recombination site (159). The integrase is
transposition functions, such as antibiotic resistance, a site-specific recombinase, which integrates incoming cir-
heavy-metal resistance, or pathogenicity determinant genes cular gene cassettes (often carrying antibiotic resistance
(33, 34). Transposons carrying antibiotic resistance genes genes) at the recombination site. The expression of the in-
were first identified in the mid-1970s after transposition coming genes is driven by the promoter of the integron. For
from drug resistance transfer plasmids into other replicons example, the transposon Tn7 contains an integron with tri-
in the cell, such as bacteriophage A (20, 80, 92, 107); see methoprim, streptothricin, and streptomycin/spectinomycin
references 21 and 47 for general reviews. A simple italic resistance genes. CTns have the capability to move them-
number is assigned to each independent isolate from nature, selves from one cell to another (41, 42, 213). First discov-
e.g., Tn5 (33, 34). A n abbreviated list of the most exten- ered in gram-positive bacteria, they have also been found to
sively studied transposons is shown in Table 1; for more de- be prevalent among gram-negative Bacteroides strains.
tailed lists, see references 14 and 15. The insertion of a Tn916 is one of the best-described examples of a CTn. In
transposon into a particular genetic locus or replicon addition to genes responsible for the integration and exci-
(phage) is designated by using a double colon, e.g. hc2::TnS sion of the transposon and antibiotic (often tetracycline)
or A::Tn5 (33, 34). These designations will also be used in resistance genes, the CTns contain so-called tra genes re-
this chapter. sponsible for their conjugal transfer. After excision, CTns
Class II transposons (noncomposite transposons) are de- form a circular intermediate; however, they are not called
scribed as transposons that do not contain IS elements and plasmids, since they do not form self-replicating structures.
are related to Tn3. Usually, they contain genes encoding Nevertheless, comparable to conjugative plasmids, they do
antibiotic (ampicillin) resistance, a transposase, and an en- contain an origin of transfer (oriT), a site at which a nick
zyme involved in the resolution of the cointegrate transpo- occurs to generate single-stranded DNA, which is then
sition intermediate, the resolvase. transferred through the conjugation apparatus into another
Class III transposons include transposable bacterio- cell. MTns also contain an origin of transfer, but they do
phages, such as Mu and its relatives. Phage Mu is in fact not contain tra genes. For their transfer, they rely on the tra
both a virus and a transposon and has been extremely use- gene products provided by either CTns or plasmids. MTns
ful in elucidating the mechanism of transposition of trans- have been identified mainly in Bacteroides species (41,213).
posable elements (35, 150). It was first discovered in the Group I1 introns have been identified first in eukaryotes,
early 1960s as a novel phage that upon lysogenization could but also recently in prokaryotes (129, 223). They are also
integrate at multiple sites in the host chromosome, thereby called retrotransposons, since they have the ability to move
frequently causing mutations (mutator phage); see refer- to new, heterologous sites via an RNA intermediate. One

TABLE 1 Common bacterial transposable elements and their characteristics


Antibiotic Transposition
Designation resistance Length (kb) frequency (per Insertion specificity Reference(s)
phenotype(s)a cell generation)
Tn3 AP' 4.957 10-~-10-~ Prefers A+T-rich sequences; 82, 186
attracted to sites having some
similarity to transposon ends
Tn5 Km' Nm'; Bm'; 5.820 10-~-10-~ Nearly none; hot spot in 18, 163
Sm' pBR322
Tn7 Tp' Sm' Sp' 14 High frequency if Very high for target site attTn7 44, 46
target site attTn7
is present; low
frequencies at
secondary sites
Tn9 Cm' 2.6 2 x 10-~ Many sites 7, 80
TnlO Tc' 9.147 Insertion hot spots (symmetrical 86, 106
6-bp sequences); prefers
nontranscribed regions
Phage Mu 36.717 High frequency for Essentially random 35, 150
chromosome
"Abhreviations used for resistance phenotypes are as follows: Ap, ampicillin; Km, kanamycin; Nm, neomycin; Bm, bleomycin; Sm, streptomycin; Tp, trimethoprim;
Sp, spectinomycin; Cm, chloramphenicol; Tc, tetracycline. Bm' and Smr phenotypes associated with Tn5 are expressed only in certain nonenteric bacteria.
686 MOLECULAR GENETICS

example is the retrotransposon L1.LtrB of Lactococcus lactis. genetics, the reader is referred to the article by Kleckner et
The intron encodes a catalytic RNA and a multifunctional al. (108). For an in-depth overview of the use of different
protein, which possesses a maturase activity to stabilize the transposable elements in the genetic engineering of bacte-
RNA structure and to promote splicing, a reverse transcrip- ria, as well as comprehensive lists of transposable elements
tase activity to synthesize cDNA, and a DNA nuclease ac- and the map positions of known insertions in Escherichia coli
tivity for the cleavage of the recipient DNA (12, 43, 213). and Salmonella, the reader is referred to articles by Berg and
It was recognized soon after the discovery of transposons Berg (14, 15), Berg et al. (16), Altman et al. (6), Sherratt
that these elements could be used as extremely efficient (186), Craig ( 4 4 4 6 ) , Kleckner (106), Haniford (86), and
tools in bacterial genetics (30, 108) because of the follow- Hayes (91).
ing characteristics. Because of the great diversity of bacterial strains, trans-
posons, delivery vehicles, and gene transfer methods avail-
1. The transposition or insertion of a transposon into a
able today, it is not possible to present a complete set of
gene generally leads to the genes inactivation, and the re-
transposon mutagenesis protocols in this chapter. Never-
sulting null mutations are relatively stable since the fre-
theless, the methods, examples, and references included
quency of precise excision of the transposon is very low.
here should provide some basic tools.
2. If a transposon integrates into an operon, it usually
exerts a strong polar effect on genes located downstream of
its insertion site, allowing transposon mutagenesis to be 29.1. Tn5 AS A MODEL TRANSPOSON
used to determine operon structure.
Transposon Tn5 is a 5,820-bp composite transposon con-
3. Upon integration, transposons introduce new genetic sisting of two inverted repeats of 1,534 bp (IS50L and
and physical markers into the target locus, such as antibi-
otic resistance genes, new restriction endonuclease cleavage
IS5OR) flanking a unique region which carries three genes
that confer on certain bacterial hosts resistance to the an-
sites, and unique DNA sequences which can be identified
tibiotics kanamycin or neomycin (the kanamycin or
by DNA-DNA hybridization, electron microscopic het-
neomycin phosphotransferase [nptII]gene), bleomycin (the
eroduplex analysis, or PCR-based methods. The genetic
ble gene), and/or streptomycin (the s t r gene) (18) (Fig. 1).
markers can be used to rapidly map and transduce the mu-
The nptII, ble, and sty genes are organized in an operon and
tated loci.
are transcribed from a promoter located at the inside end of
4. Transposons can generate a variety of genomic re- ISSOL (18) (Fig. 1).ISSOR genes encode the transposase re-
arrangements, such as deletions, inversions, translocations,
sponsible for Tn5 transposition (the tnp gene), as well as a
and duplications, and can be used to introduce specific
transposition inhibitor (the inh gene) (18) (Fig. 1). The first
genes into the genome of a target bacterium.
19 bp of both ISSOL and ISSOR have been found to be es-
These characteristics have made transposon mutagenesis sential for the efficient transposition of Tn5 (18).
a valuable addition to the more classical techniques such as Tn5 is capable of transposing at a very high frequency
chemical mutagenesis or mutageensis induced by UV irra- ( loe2 to transpositions/cell generation) in E. coli by
diation. Transposon mutagenesis has been used to clone using a conservative cut-and-paste mechanism, and it
genes, to construct reporter gene fusions, to construct cor- generates a 9-bp target site duplication upon insertion (18).
related physical and genetic maps of cloned DNA segments, The three-dimensional structure of the transposition inter-
to map entire bacterial genomes by pulsed-field gel elec- mediate has been determined (49). Tn5 has a low inser-
trophoresis, to construct conditional mutations with tional specificity and therefore can insert itself into a large
portable promoters, to introduce desired origins of conjugal number of locations in bacterial genomes, as well as into
transfer or replication into chromosomes and plasmids of multiple positions within single genes, although occasional
interest, and to determine the sequence of large DNA re- hot spots for insertion have been reported (18, 19, 50, 52).
gions without the need of sub- or deletion cloning. For ad- Tn5-induced insertion mutations are very stable, and mu-
ditional information on the use of transposons and bacte- tants only occasionally revert to the wild type via a process
riophages for gene manipulations in bacteria, see chapters known as precise excision, although the reversion fre-
31, 32, and 34. quency is somewhat dependent on the precise location of
Even in the time of the availability of complete bacter- Tn5 in the genetic locus, the nature of the replicon, and the
ial genome sequences, transposon mutagenesis continues to host genotype (18). These parameters are important for the
play a role in analyses of the functions of putative genes. experiments described below and should be examined when
Often, when complete bacterial genomes are characterized, Tn5 mutagenesis experiments are performed with new or-
no function can be assigned to around 40% of the putative ganisms or when problems are encountered with the proto-
open reading frames. Thus, the technique of transposon cols presented here. For in-depth discussions of these pa-
mutagenesis can be combined with genomics to analyze the rameters, see reviews by Berg and colleagues (16, 18, 19)
phenotypes of mutants with transposon insertions in spe- and de Bruijn and Lupski (52).
cific genes. Tn5 mutagenesis experiments can be divided into two
In this chapter, a subset of these applications and the distinct categories: random mutagenesis and region-
corresponding experimental protocols are described. The directed mutagenesis (50). The terms site-directed and site-
focus is on one of the transposons, namely, Tn5, which has specific transposon mutagenesis have also been used in the
been extensively used in bacterial molecular genetics, but literature to describe the latter category (50). The term
the principles and most of the experimental strategies de- region-directed mutagenesis is used here to distinguish this
scribed are applicable to a variety of other transposons. For category of transposon mutagenesis from base pair-specific
a treatise on the biology of transposons and their transposi- oligonucleotide mutagenesis methods, which are usually re-
tion mechanisms, the interested reader is referred to the ferred to as site specific. Random mutagenesis involves the
publication by Berg and Howe (21) and the volume edited introduction of Tn5 into the bacterial species of interest via
by Craig et al. (47). For a detailed description of the appli- transformation, transduction, conjugation, or electropora-
cation of transposon mutagenesis to more classical bacterial tion by using suicide plasmid or phage vectors, followed
29. Transposon Mutagenesis w 687

Q som c9 >m OD> m>


r x za I o m nn mp.

1 1000 2000 3000 4000 5000 5818

FIGURE 1 Structure of the transposable element Tn5. Stippled bars represent the insertion se-
quences ISSOL (left) and ISSOR (right), present as terminal inverted repeats in Tn5. Gene symbols:
tnp, coding region for the transposase; inh, coding region for the inhibitor; UAA, stop codon re-
sponsible for the truncated versions of the transposase and the inhibitor in ISSOL; p, promoter for
the transcript of the operon coding for resistance to three antibiotics (nptII, neomycin or kanamycin
resistance; ble, bleomycin resistance; s t r , streptomycin resistance). Abbreviations for restriction en-
zymes: Ba, BamHI; Bg, BglII; H3, HindIII; Hp, HpaI; No, NotI; Ps, PstI; Pv, PvuII; Sa, SalI; Sm,
SmaI; Xh, XhoI.

by selection for the antibiotic resistance marker(s) carried cation for stable maintenance. Phage P1 has also been used
by Tn5. The transposition and insertion of Tn5 into the to introduce transposons such as Tn5 into different bacter-
genome of the recipient bacterium are detected after the ial species, and examples of the use of this vector system
vector has been lost by segregation, and the phenotype as- have been described previously (11, 114).
sociated with the Tn5-induced mutations can be analyzed The choice of the suicide vector and transposon element
subsequently. Region-directed mutagenesis usually involves to be used depends on the purpose of the random mutagen-
the isolation of Tn5 insertion mutations in genes cloned esis experiment and a number of criteria, including the
into (multicopy) plasmids in E. coli, followed by the char- phage sensitivity and the intrinsic antibiotic resistance of
acterization of the mutant phenotype of this (heterologous) the bacterium to be mutagenized, as well as the availability
host or the organism of origin of the cloned genes after the of a conjugal transfer or direct transformation system. A
reintroduction of the Tn5-mutated loci by gene replace- short list of transposons and their relevant properties
ment (SO). Experimental strategies for these two types of is shown in Table 1, but for a discussion of the above-
mutagenesis are presented in the following sections and mentioned parameters and an extensive list of the trans-
have been reviewed previously (16, SO, 52). posons available for specific purposes, the reader is referred
to previous reviews (14-16, 52).
This chapter focuses on the use of bacteriophage X and
29.2. RANDOM Tn5 MUTAGENESIS conjugable narrow-host-range or conditionally replicating
To generate transposon insertion mutations in the genomes plasmid vectors to deliver Tn5 and its derivatives and de-
of bacteria, the transposon must be introduced into its new scribes experimental protocols that are commonly used to
host and cells in which transposition has occurred must be carry out the random mutagenesis of (predominantly gram-
identified. These objectives are achieved by the use of so- negative) organisms. For a discussion of chemical transfor-
called suicide vectors, which can be of phage or plasmid ori- mation and high-voltage electroporation techniques (62) to
gin. In both cases, after the introduction of the suicide vec- introduce plasmids carrying Tn5 into cells, the reader is re-
tor carrying the Tn5 DNA molecule into the cell, the ferred to chapter 3 1.4. For a discussion of transposons in
vector must be incapable of stable replication or integration gram-positive organisms and their use for mutagenesis ex-
so that selection for one or more of the antibiotic resistance periments, the reader is referred to articles by Murphy (142)
markers on the transposon element leads to the identifica. and Youngman (222).
tion of cells in which the element has transposed from the
vector into the genome. A commonly used phage vector for 29.2.1. Phage X Suicide Vectors
the introduction of transposons into bacterial cells is a mod- For X::TnS-mediated random mutagenesis experiments, a
ified derivative of bacteriophage X, which can be used for derivative of phage X (A467) is routinely used; it has the fol-
the mutagenesis of all bacteria that are naturally susceptible lowing genotype: X b221 rex::Tn5 cI8S7 Oam29 Pam80 (52,
to A adsorption and infection or have been genetically mod- 108). The b221 mutation is a deletion in the X genome that
ified to be X sensitive. The protocol for generating the late removes the phage attachment (att) site. This deletion pre-
ter category of engineered organisms has been described vents the lysogenization of the target strain. The rex gene is
previously (121, 149) and is not presented here. For bacte- a nonessential A gene and carries the Tn5 insertion. Both
ria that are not X sensitive or cannot be engineered to be- the 0 and P genes are involved in phage replication, and
come X sensitive, the transposon can be introduced into the therefore, amber mutations in these genes will prevent
cells by using plasmids, which can be conjugally mobilized phage replication in a suppressor-negative (Su-) back-
or electroporated (62) into recipient cells (see chapter 3 1.4) ground. The X467-type phage can be propagated in a Su'
but lack the appropriate (wide-host-range) origins of repli- E. coli strain (see below), will adsorb to a A-sensitive strain,
688 w MOLECULAR GENETICS

and will inject its DNA; however, this DNA will be unable 5. Mix the phage and the cells gently, and allow the
to replicate and will be lost by segregation. Selection for phage to adsorb for 2 h at room temperature without agitat-
Km' Nm' (and, in selected cases, Bm' or Smr) survivors re, ing the tubes.
sults in the identification of cells in which Tn5 has trans- 6. Plate 200-pl aliquots of the infected cells on com-
posed and integrated into the genome. This type of Tn5 plete-medium plates containing kanamycin (or neomycin,
mutagenesis has been successfully used to isolate mutations bleomycin, and streptomycin) at the desired concentration,
in many genes of E. coli (15, 123, 184). and incubate the plates at the appropriate temperature until
clearly defined colonies appear on the selection plates (but
29.2.1 . I . Phage Lysate Preparation not on the control plates).
To prepare A::Tn5 lysates, the Su+ E. coli LE392 strain (F- 7. Purify the resistant colonies on complete-medium
hsdR514 supE44 supF58 lacy1 galK2 galT22 metBl trpR.55 plates with the desired antibiotics, and analyze them as de-
A-) and the following protocol (modified from reference scribed in sections 29.2.3 and 29.2.4.
52) are used.
Controls and Comments
1. Start an overnight culture of E. coli LE392 at 37C No Km' colonies should be observed on the plates onto
in 10 ml of Luria-Bertani (LB) medium (see section 29.5) which the uninfected cells were plated.
plus 0.2% maltose.
2. Pellet the cells by centrifugation, wash once with 5 ml 29.2.2. Plasmid Suicide Vectors
of 10 mM MgSO,, and pellet again. The first class of suicide plasmids to be used for the random
3. Resuspend the cells in 4 ml of 10 mM MgSO,. Tn5 mutagenesis of A-insensitive bacterial strains was based
4. Prepare five sterile glass tubes with 0.1 ml of washed on an IncP-type plasmid carrying a copy of Mu in addition
LE392 cells, and add 10 p l of A::Tn5 lysate or the phages to Tn5 (pJB4JI) (24). The presence of Mu in this replicon
from a single plaque to four of the tubes. Leave one tube un- was found to prevent its stable maintenance in different
infected as a control. gram-negative bacteria, and Tn5 transpositions could be
5. Mix gently, and let the phages adsorb for 2 h at room identified after antibiotic resistance selection (24). How-
temperature without agitating the tubes. ever, for several bacterial species, it was found that a large
6. Mix the infected cells with 3 ml of soft agar (45C) percentage of the insertion mutants carried Mu sequences
and pour the mixture evenly onto A agar plates (see section at the insertion site, in addition to Tn5 (130), and pJB4JI
29.5). was observed to be relatively stable in other species (191),
7. Incubate the plates for 6 to 8 h (or overnight) at leading to a reduced use of this class of vectors.
37"C, and scrape off the clear soft agar into a fresh tube. A second class of Tn.5-containing suicide plasmids is
8. Mix the agar-phage cell suspension well but gently based on replicons carrying a temperature-sensitive origin
with 2.5 ml of SM buffer (see section 29.5) and 0.5 ml of of replication, allowing the selection for Tn.5 transposition
chloroform in a sterile (Corex) centrifuge tube. Incubate at elevated temperatures (115). In addition, derivatives
the mixture for 5 min on ice, and centrifuge it for 15 min at of the broad-host-range conjugative plasmids R388
1,000 X g (e.g., at 3,000 rpm in an SS34 fixed-angle rotor) (pCHR7 1) and pRK290 (pRK340), carrying temperature-
at 4C. sensitive mutations in plasmid replication functions, have
9. Transfer the supernatant solution into a small glass been described and their utility in the Tn5 mutagenesis of,
tube, add 20 drops of chloroform, and store at 4C. for example, Shigella and Legionella species has been demon-
10. Determine the titer of the lysate by infecting LE392 strated previously (104, 179, 180).
cells (prepared as described in steps 1 to 3) with several di- A third class of suicide vectors for Tn5 mutagenesis,
lutions of the lysate (from lo-,, to and count- based on narrow-host-range IncW- and IncN-type plasmids,
ing the resulting single plaques on the indicator plates. has been described previously and used for the mutagenesis
of different bacterial species, such as Rhizobium meliloti
Controls and Comments (pGS9) ( 181) , Bradyrhizobium japonicum ( 171), Azorhizo-
The plaques on the titer plates should be small and well de- bium caulinohm (pGS9) (96), Pseudomonas solanacearum
fined (pinpoint). No plaques should be observed on the (pW1281) (140), and Azotobacter vinelandii (105), to men-
plates with bacterial cell suspensions to which no phage was tion a few examples.
added. The titer of the lysate should be in excess of 5 x 10" A fourth class of suicide vectors is derived from the R6K
for successful use in subsequent Tn5 mutagenesis experi- replicon and is based on the principle of transcomplemen-
ments. tation-dependent maintenance of the R6K replication ori-
gin (109). In this system, a plasmid carrying (a fragment of)
the R6K replication origin (e.g., pRK703) can be stably
29.2.1.2. infection and Mutant Selection maintained only if a trans-acting factor (the n protein en-
1. Start an overnight culture of the A-sensitive Su- coded by the pir gene) is provided in the bacterial cell, for
bacterial strain to be mutagenized in 5 ml of complete example, by a Apir prophage (109). Miller and Mekalanos
medium for the respective strain, supplemented with 0.2% (139) inserted into plasmid pRK703 an IncP-type origin
maltose. of conjugal transfer (mob, oriT of RP4) to allow high-
2. Inoculate 100 p l (1:lOO) of the saturated culture into frequency transfer to a variety of gram-negative bacteria
10 ml of complete medium + 0.2% maltose, and incubate with the help of RP4 mobilization functions provided in
the culture for 2 to 4 h. tram (see below), as well as a polylinker containing several
3. Pellet the cells, wash them with a sterile solution of 10 cloning sites (pGP704). Transposon Tn5 derivatives (mini-
mM MgS04, and pellet them again. Resuspend the pelleted TnS's), consisting of the 19 bp of the Tn5 inverted repeats
bacteria in 4 ml of 10 mM MgSO,. required for transposition coupled to antibiotic resistance or
4. Infect 1-ml aliquots of the washed cells with 100 p1of other selectable marker genes (see section 29.4.4), have
A::TnS lysate ( lo8 to lo9 CFU/ml), and keep 1 ml of unin- been inserted into this vector, in addition to the Tn5 trans-
fected cells as a control. posase (tnp) gene, to construct an elegant suicide transpo-
29. Transposon Mutagenesis 689

son mutagenesis system. The suicide plasmid carrying a 5. Spot 100 p l of the donor and recipient cell cultures
mini-Tn5 construct is conjugally mobilized (see sections separately and 100 pl of a 1:l mixture of both onto conju-
29.2.2.1 and 29.2.2.2) from an E. coli strain harboring Apir gation plates containing the complete medium suitable for
to the gram-negative bacterium to be mutagenized, and the recipient strain, in the absence of antibiotics. Allow the
selection for one of the mini-Tn5-borne marker genes is ap- spots to dry, and incubate the plates at the appropriate tem-
plied. This procedure results in the selection of transconju- perature for the recipient strain for 12 to 36 h.
gants in which the mini-Tn5 construct has transposed from 6. Scrape the mating mixtures and the single spots of
the vector, which itself is no longer capable of replicating in donor and recipient cells off the plates with a sterile spatula,
the absence of the pir gene, into the resident genome. Once resuspend the bacterial cells in 1 ml of 0.9% NaCl (or H20
inserted into the genome, the mini-Tn5 element is inca- for osmotically stable recipient bacteria, such as rhizobia),
pable of further transposition, since it is lacking the suicide and plate out the suspensions (or appropriate dilutions
vector-borne tnp gene, resulting in a very stable integration thereof) onto selective plates containing antibiotics to se-
event (95). The stable integration is especially relevant lect for the recipient strain (if the recipient strain harbors a
when Tn5-carried genes must be stably maintained in the gene conferring resistance to an antibiotic to which the
absence of antibiotic selection in natural environments donor strain is sensitive) and for the presence of Tn5
(95). For a detailed description of the use of these vectors, (kanamycin, neomycin, bleomycin, or streptomycin).
the reader is referred to the articles by Herrero et al. (95) 7. Purify the single colonies appearing on the selective
and de Lorenzo et al. (55,56). plates once onto the same selective medium to avoid con-
Another commonly employed class of suicide vectors for tamination with donor bacteria, or (when screening large
Tn5 mutagenesis in gram-negative bacteria other than E. numbers of colonies for mutant phenotypes) characterize
coli is based on replicons derived from plasmid RP4 carrying them directly.
the same broad-host-range conjugal transfer or mobilization
Km' derivatives of the recipient strain should be the re-
functions and sites as those used for the above-described
sult of the introduction of the suicide vector, the subsequent
vectors but a narrow-host-range origin of replication (190,
(random) transposition of Tn5 into the resident genome,
191). These vectors can be mobilized at a high frequency
and the loss of the vector. These colonies can now be sub-
from E. coli to other gram-negative bacteria but cannot be
jected to further studies (see sections 29.2.3 and 29.2.4).
stably maintained in the recipient species.
These vectors contain, in addition to Tn5, the IncP-type Controls and Comments
mobilization (mob; oriT) site and are based on commonly No colonies should appear on the selection plates onto
used E. coli cloning vectors, such as pACYC184 (37) and which the donor and recipient cells were plated separately.
pBR325 (28), which cannot replicate in nonenteric bacte- If colonies do appear repeatedly on the control plates, alter-
ria (pSUP series) (189, 191). pSUP-type plasmids can be native counterselection protocols should be considered (see
mobilized in diparental mating experiments (see section below). Care should be taken to treat the cells gently dur-
29.2.2.1) by providing the transfer functions in mans from a ing pelleting and washing and to keep them at the growth
chromosomally integrated copy of the IncP plasmid RP4 in temperature of the recipient strain. Instead of or in addition
the donor strain itself, e.g., with strain S17-1 (RP4-2- to the antibiotic selection, a specific minimal medium suit-
Tc::Mu Km::Tn7 Tp' Sm' thi pro hsdR) (190), or in tri- able for the recipient bacteria but lacking the appropriate
parental mating experiments (see section 29.2.2.2.) by pro- carbon source for E. coli (e.g., one containing sucrose) or
viding the transfer functions from plasmid pRK2013, lacking the amino acid or vitamin requirements of the E. coli
harbored by a nondonor, nonrecipient helper strain of E. donor strain (e.g., thiamine and proline for strain S17-1)
coli (E. coli/pRK2013) (59, 60). The use of this vector sys- (190) can be used to counterselect the E. coli donor bacte-
tem will be described here; for a detailed review of the con- ria. The latter method can of course be used only if one is
jugal transfer of pSUP vectors, the reader is referred to other not trying to isolate Tn5-induced auxotrophic mutations in
reviews (189, 191). As an alternative to conjugation meth- the target bacterial strain (see section 29.2.2.3).
ods to transfer suicide vectors, high-voltage electroporation
can be used (62) (chapter 31.4). 29.2.2.2. Triparental Conjugation and Mutant
Selection
29.2.2.1. Diparental Conjugation and Mutant
Selection 1. Prepare a culture of logarithmically growing recipient
bacteria as described above. Similarly, prepare a logarithmi-
1. Start a 10-ml culture of the recipient bacterial strain cally growing culture of a donor E. coli strain harboring the
in complete medium, and grow it until it has reached satu- RP4-derived Tn5 mutagenesis vector. In this case, the donor
ration at the appropriate temperature. Reinoculate the cells strain does not contain a chromosomally integrated copy of
1:lO in fresh medium, and incubate the cultures for another RP4 to provide transfer functions but can be any E. coli
2 to 5 h (depending on the doubling time of the bacterial strain, preferably a strain carrying one or more auxotrophic
strain used). markers for counterselection (see above). In addition, pre-
2. Start a 10-ml overnight culture of the donor strain E. pare a logarithmically growing culture of a third E. coli
coli S17-1 harboring the Tn5 delivery plasmid (189, 190) in (helper) strain harboring a plasmid (pRK2013) (59,60), pro-
LB medium supplemented with the appropriate antibiotics viding the RP4 transfer functions in trans, and allowing the
(e.g., usually kanamycin for Tn5 and tetracycline, ampi- conjugal transfer of the Tn5 mutagenesis vector from the
cillin, or chloramphenicol for the pSUP vector), and grow donor to the recipient bacteria in this type of triparental mat-
the culture until saturation at 37C. Reinoculate the cells ing experiment. Since the pRK2013 plasmid carries a Km' or
1:lO in fresh medium, and grow for 1 to 2 h at 37C. Nm' gene, kanamycin or neomycin should be added to the
3. Pellet the logarithmically growing recipient and donor growth medium to select for the presence of the plasmid.
cultures (1,000 X g for 10 min), wash the bacteria with 2. Pellet the cultures (1,000 x g for 10 min), wash the
0.9% NaCl to remove the antibiotics, and pellet again. bacteria with 0.9% NaC1, and pellet them again. Resuspend
4. Resuspend the pelleted bacteria in 1 ml of 0.9% NaC1. the pelleted bacteria in 1 ml of 0.9% NaC1.
690 MOLECULAR GENETICS

3. Spot 100 pl of the donor, helper, and recipient cell volving the suicide vector. Cointegration is of particular
cultures separately, in painvise combinations (l:l), and all importance since, in some bacterial species, the predomi-
three together (1:l:l) onto conjugation plates containing nantly Tn.5-mediated formation of cointegrates resulting in
the complete medium suitable for the recipient strain in the the integration of the entire Tn5-carrying suicide vector
absence of antibiotics. Incubate the plates at the appropri- into the genome is the cause of the observed mutant phe-
ate temperature for the recipient strain for 12 to 36 h. notype (for examples, see references 61 and 96). For a de-
4. Follow steps 6 and 7 of the diparental mating protocol. tailed description of this phenomenon, the reader is referred
to reference 18.
Controls and Comments The cointegration of the entire suicide vector can usu-
No colonies should appear on the selection plates onto ally be ruled out conveniently by examining the Km'
which the donor, helper, or recipient strain was plated sep- colonies resulting from a random Tn5 mutagenesis experi-
arately or in a pairwise combination. The use of an unsup- ment for the absence of one or more of the antibiotic re-
plemented minimal medium and auxotrophic donor and sistance genes carried by the non-Tn5 part of the vector,
helper E. coli strains should help reduce background on the e.g., the Tc', Ap', or Cm' gene for the pSUP vectors (189,
selection plates. If colonies do form consistently on the con- 190). Single versus multiple Tn5 insertions and the cointe-
trol plates, the individual strains should be checked or other gration of parts of the suicide vector should also be exam-
(or modified) counterselection conditions should be ex- ined by DNA-DNA hybridization (Southern blotting; see
plored (e.g., using a different minimal medium, using an al- the following sections).
ternative antibiotic, or raising the concentration of the an-
tibiotic). 29.2.3. Physical Mapping of Insertions
29.2.2.3. Mutagenesis Protocols and Mutant To physically characterize Km' colonies resulting from the
random Tn5 mutagenesis protocols outlined in sections
Characterization 29.2.1 and 29.2.2, total genomic DNA is isolated from cul-
The protocols outlined above normally result in approxi- tures derived from the respective colonies, digested with
mately lop3 to Tn5-containing colonies per recipient restriction enzymes, separated by agarose gel electrophoresis,
bacterial cell. If the total number of Tn5-containing strains transferred onto nitrocellulose or nylon membranes, and hy-
is too small for the planned mutant-screening program, the bridized with Tn5 and suicide vector-derived DNA probes.
mutagenesis protocol should be optimized. For the conjuga- The method for isolating total genomic DNA from bac-
tion protocols, the ratios of donor and recipient cells should terial cultures depends strictly on the genus and species
be varied (e.g., 2- to 20-fold excess of donor cells compared being analyzed, and therefore, no universal protocol is
to recipient cells) and the conjugation time should be ex- available. However, the following method modified from
tended. In addition, instead of spotting the conjugation references 50 and 130 is applicable to a variety of gram-
mixtures directly, the cell mixtures should be combined negative enteric and soil bacteria.
onto 0.2-pm-pore-size nitrocellulose filters placed on (pre-
warmed) conjugation plates, incubated for the desired time, 29.2.3.1. Isolation of Genomic DNA
and removed from the filters by vortexing in 0.9% NaCl (or
HzO) before plating onto selection plates. The concentra- 1. Generate a 1-ml culture of logarithmically growing cells.
tion of the antibiotic(s) used for (counter)selection should 2. Pellet the cells (e.g., in a 1.5-ml microcentrifuge tube),
also be varied. and wash them twice with 1 M NaCl to remove (most) ex-
When first carrying out random Tn5 mutagenesis exper- tracellular polysaccharides. Resuspend the pelleted cells in 1
iments with a particular bacterial strain, it is useful to ex- ml of TE (10 mM Tris, 1 mM EDTA, pH 8.0).
amine the frequency of Tn5-mediated gene inactivation. 3. Incubate the cells for 20 min at 37C in the presence
This is most conveniently done by determining the per- of 0.1 ml of a freshly prepared 2-mg/ml lysozyme solution in
centage of auxotrophic mutants resulting from a given Tn5 TE, with gentle mixing.
mutagenesis experiment. This determination requires the 4. Add 0.125 ml of a 10% Sarkosyl-5-mg/ml pronase so-
availability of a defined minimal medium for the strain lution in TE (which has been predigested for l h at 37"C),
under study. Five hundred to several thousand Km' colonies and incubate the mixture for 1 h at 37C.
are picked and streaked or replica plated (117) onto com- 5. Extract the viscous mixture of lysed cells with TE (pH
plete and minimal medium, and the frequency of colonies 8.0)-saturated phenol two or three times, and extract the
unable to grow on minimal medium is examined. Approx- final aqueous phase with chloroform.
imately 0.1 to 3.0% of the colonies should fall into the lat- 6. Add ammonium acetate to a final concentration of
ter category. The occurrence of different auxotrophic mu- 0.3 M, and precipitate the nucleic acids by the addition of
tants can be examined by using the Holliday test (100). The 2.5 volumes of ice-cold ethanol. "Spool out" the DNA
actual percentage of different auxotrophic mutants observed strands with a glass rod, or pellet the nucleic acids by cen-
is an important piece of data that can be used to determine trifugation. Resuspend the spooled-out DNA or the pellet
how many random Tn5-containing colonies must be in 0.1 to 0.5ml of TE by gentle mixing. Leave the DNA so-
screened to find an insertion in a particular gene of interest, lution at 4C overnight, and mix gently to achieve optimal
since it reflects the insertional specificity of Tn5 in the bac- resuspension.
terial strain under study. 7. Determine the concentration of the DNA prepara-
Once it has been established that Tn5 mutagenesis can tions, and load 0.1 to 0.5 pg of undigested DNA onto an
indeed lead to the identification of different mutations in agarose gel to verify its high molecular weight and to check
the bacterial strain of interest, it should be determined for the presence of nucleases in the preparations.
whether the Km' (mutant) colonies isolated carry a simple
insertion of a single Tn5 transposon into a particular ge- 29.2.3.2. Southern Blotting and Hybridization
nomic locus. These events should be distinguished from The total genomic DNA from the Km' colonies (1 to 2 pg)
multiple Tn5 insertions or aberrant cointegration events in- can now be digested with different restriction endonucle-
29. Transposon Mutagenesis 691

ases. For the first analysis, an enzyme which does not cut Tn5 derivatives carrying an origin of conjugal transfer (see
Tn5 should be used (e.g., EcoRI, KpnI, or ClaI). For modi- section 29.4.4).
fied Tn5 derivatives carrying additional sequences, the re- Detailed strategies for carrying out mapping experiments
striction maps should be examined to verify that these en- with transposon insertions are described by Kleckner et al.
zymes do not cut; if necessary, other enzymes should be (108), and such descriptions exceed the scope of this chap-
selected. The insertion of Tn5 into a specific EcoRI or ClaI ter. Examples of the mapping of specific Tn5-induced inser-
fragment (X) should give rise to a new, larger EcoRI or ClaI tion mutations have been described previously (89,184),and
+
fragment (X Tn5; Tn5 = 5.8 kb). In the second level of a list of mapped Tn5-induced insertion mutations in E. coli
analysis, an enzyme should be used which cuts the transpo- and Salmonella has been compiled elsewhere (15). Examples
son twice (in the inverted repeats; e.g., Hind111 or BglII) of the use of Tn5 mutagenesis to create a genetic map have
(Fig. l ) , thereby generating distinct Tn5 target junction also been described previously (9, 155). For an example of a
fragments, as well as constant internal Tn5 fragments (50). fine-structure analysis of Tn5 insertions within a single
To visualize the Tn5-containing (junction) fragments, the operon, see Miller et al. (138).
digested DNA should be separated by agarose gel elec- Once the genetic map of a region (operon or regulon)
trophoresis and transferred onto membranes by Southern has been established with the help of Tn5-induced inser-
blotting (for details, see chapter 30.4.4 and references 174 tion mutations and a recombinant plasmid corresponding to
and 175). To determine the orientation of the Tn5 inser- the wild-type operon (regulon) has been obtained, a corre-
tion relative to known restriction sites in the mutated locus, lated physical and genetic map can be prepared by using re-
enzymes which cut the transposon once (asymmetrically; striction mapping and Southern blotting. For a description
e.g., SalI and SmaI) (Fig. 1) should be used. The mem- of the strategy used and an example of this type of analysis,
branes should be hybridized with labeled Tn5 sequences see Riedel et al. (167).
(e.g., the HpaI internal fragment of Tn5) (Fig. l ) , and the
hybridizing fragments should be visualized by autoradiogra- 29.2.5. Cloning of Tn5-Mutated Genes
phy or another appropriate visualization technique (see ref-
O n the basis of hybridization data (see section 29.2.3), a
erences 174 and 175).
fragment generated by a restriction enzyme that does not
For the enzymes not cutting Tn5, single hybridizing
cut within the Tn5 sequence is usually chosen. This choice
fragments of different sizes (all larger than 5.8 kb) should
enables the cloning of regions upstream as well as down-
be observed in the DNA of different Km' colonies, sug-
stream of the Tn5 insertion. The fragment should be large
gesting that the fragments are the result of single Tn5
enough to carry, besides Tn5, the coding sequences of the
transpositions into the genome. For the enzymes cutting
gene(s) of interest, at least with a high probability. Any
Tn5 once, two distinct hybridizing fragments should be ob-
suitable vector can be used, but of course it will facilitate
served in the DNA of each Km' colony, representing de-
the cloning procedure if the antibiotic resistance marker of
fined transposon-host junction sequences. For the enzymes
the vector is not kanamycin resistance.
cutting Tn5 twice within the inverted repeats, two distinct
A great advantage at this step in the random mutagene-
junctions and one constant internal Tn5 fragment should
sis procedure is the use of a Tn5 construct that carries an
hybridize.
origin of replication (see section 29.4.4) in addition to the
The Southern blots carrying the digested genomic DNA
Tn5 antibiotic resistance genes. In this case, the chromo-
of the Km' colonies should also be probed with labeled sui-
somal DNA can be isolated, digested with a restriction
cide vector sequences (without Tn5). If simple Tn5 trans-
enzyme that does not cut within the Tn5 derivative, and
positions have occurred, fragments hybridizing with vector
self-ligated, and E. coli cells can be transformed with the
sequences should not be observed.
construct. Only fragments containing the Tn5 derivative
The Km' colonies have now been shown to be the result
with the origin will replicate and propagate in E. coli.
of the simple insertion of a single copy of Tn5 and can be
As soon as the mutated gene region is cloned, it can be
characterized further, both genetically (see section 29.2.4)
used to conduct a gene replacement experiment (see sec-
and physically, by more extensive restriction mapping,
tion 29.3.4) to verify that the Tn5 insertion indeed caused
cloning, and DNA sequencing (see sections 27.1, 29.2.5,
the observed phenotype.
29.4.6, and 29.4.7).
Finally, the mutated gene region can be used as a probe
29.2.4. Genetic Mapping of Insertions and to clone the corresponding wild-type region from the origi-
Tn.5-Induced Mutations nal bacterial strain. Different strategies are available for this
purpose; they are summarized in the following paragraphs
Once a Tn5 insertion, a Tn5-induced mutation, or a col-
(for detailed protocols, see molecular biology manuals [174,
lection thereof has been isolated and characterized, it can
be used for genetic mapping experiments to determine its 1751).
relative position with reference to previously identified mu- 1. After the plasmid carrying the Tn5-mutated region is
tations or to create a genetic map of an organism. This pro- mapped with restriction enzymes, prepare a DNA probe
cedure is facilitated by the presence of the antibiotic resis- from the cloned and mutagenized bacterial locus. This
tance marker on the transposon, which permits selection for probe may consist of a restriction fragment carrying DNA
the inheritance of the integrated transposon and selection sequences immediately adjacent to the Tn5 insertion or a
or screening for the mutations caused by the transposon's fragment carrying Tn5 plus target sequences. Alternatively,
insertion (108). Transposon-induced insertion mutations if the DNA sequence of the mutagenized locus has been de-
behave essentially as standard genetic markers and there- termined (see also section 29.4.6), a DNA oligonucleotide
fore can be used in two-point crosses to determine genetic probe can be synthesized or a specific PCR-generated probe
distances and in three-point crosses to establish gene order can be prepared for hybridization experiments (see chapter
(108). By using standard methods (see chapter 31.1), these 30.4.3 and reference 175).
crosses can be carried out and genetic map positions can be 2. Hybridize a Southern blot, containing genomic DNA
determined. This process is further facilitated by the use of from both the Tn5-mutated and the wild-type strains
692 H MOLECULAR GENETICS

digested with enzymes that do not cut the transposon (see (18, 52), the longer the vector sequence, the more inser-
section 29.2.3.2), with the DNA probe prepared in step 1. tions will be made in the vector and not in the cloned locus
This analysis should lead to the identification of the same of interest. In addition, the copy number of the cloning vec-
single hybridizing fragments found to hybridize with the tor is important. High-copy-number vectors will be easier to
Tn5 probe (see section 29.2.3.2) in the Tn5-containing mutagenize, and it will be more convenient to isolate DNA
mutant strains and corresponding smaller single fragments from strains harboring them. In addition, high-copy-number
(minus the length of Tn5) in the wild-type strain. plasmids can be used for Tn5 mutagenesis experiments with
3. Create a complete clone bank of DNA from the wild- a chromosomal copy of Tn5 as the donor (22, 188). This
type bacterial strain used to generate the Tn5 mutants in a protocol will not be presented here but is detailed in refer-
plasmid or phage cloning vector (see reference 175). ence 50.
Alternatively, a partial clone bank can be constructed by Some commonly used cloning vectors, such as pBR322
purifying DNA fragments of the expected size (see step 2) (29), carry hot regions for Tn5 insertion (18) and may
from a gel and cloning them into a plasmid vector. therefore be less useful. However, the Tn5 mutagenesis of
4. Screen the library (step 3) with the probe generated cloned genes has been performed successfully,not only with
in step 1, and purify hybridizing plaques or colonies (see ref- small, higher-copy-number vectors such as pACYC184 (37)
erence 175). but also with low-copy-number, broad-host-range vectors
5. Hybridize a Southern blot containing genomic DNA such as pRK290 and pLAFRl (60,66).
from the strain and DNA from the recombinant phages or The most commonly used Tn5 delivery (mutagenesis)
plasmids, digested with the same enzymes as those used in vector for region-directed mutagenesis is A::Tn5 (see sec-
step 2, with the probe (step 1) to verify that the fragment of tion 29.2.1), and a protocol modified from references 50
interest has been cloned. and 52 for this type of experiment follows.
6. Further analyze the cloned wild-type locus by restric-
tion mapping, complementation studies, DNA sequencing, 29.3.1 . I . Phage X::Tn5
and expression studies (175). In addition, the cloned region
can be subjected to region-directed Tn5 mutagenesis to 1. Transform any A-sensitive E. coli host with the plas-
create a precise, correlated physical and genetic map (see mid to be mutagenized (see chapter 31.1.2 and reference
section 29.3.3) or subjected to transposon mutagenesis to 175). The plasmid-selectable marker should not be
create gene fusions for regulation studies (see section kanamycin or neomycin resistance.
29.4.1). 2. Purify a single colony, make a small-scale plasmid
DNA preparation (see chapter 30.1.2 and reference 175),
In selected cases, the wild-type counterpart of a Tn5-
and verify the monomeric structure of the recombinant
mutated locus can also be cloned by direct complementa-
plasmid to be mutagenized. (Determine the size of the undi-
tion. A readily screenable or selectable phenotype of the
gested plasmid by agarose gel electrophoresis.)
complemented strain would have to be available. In this 3. Start an overnight culture of the E. coli strain with
case, the clone bank of the wild-type strain (step 3) would
the target plasmid in 5 ml of LB supplemented with the ap-
be introduced into the mutant strain via transformation,
propriate antibiotic (corresponding to the plasmid vector
transduction, or conjugation and individual transformants,
marker gene) and 0.2% maltose at 37C.
tranductants, or transconjugants would be examined for the
restoration of the wild-type phenotype.
4. Inoculate 100 pl(1:lOO) of the saturated culture into
10 ml of LB-antibiotic-0.2% maltose, and let it grow for 2
h at 37C.
5. Spin down the cells, wash them with 10 mM MgS04,
29.3. REGION-DIRECTEDTn5 MUTAGENESIS and pellet them again. Resuspend the pelleted bacteria in 4
When a wild-type locus has been cloned, region-directed ml of 10 mM MgSO,.
(Tn5) mutagenesis can be used to delimit the genes and 6. Infect 1-ml aliquots of cells with 100 p 1 of X::Tn5
operons carried by the cloned region. The whole procedure lysate, prepared as described in section 29.2.1.1 (lo8 to lo9
can be carried out most efficiently if the presence of the CFU/ml), and keep 1 ml of uninfected cells as a control.
cloned locus in E. coli results in an observable phenotype. 7. Mix gently, and let the phage adsorb for 2 h at room
Possible scenarios include those in which the cloned locus temperature without agitating.
of interest is able to complement an auxotrophic mutation 8. Plate 2OO-kl aliquots of infected cells onto LB plates
or a growth defect of a special E. coli strain, endows strains containing kanamycin (25 pl/ml) and the antibiotic select-
with the ability to catabolize or grow in the presence of ing for the presence of the plasmid. Incubate the plates for
novel compounds, or produces protein products which can 2 days at 37C.
be detected enzymatically or immunologically. If the cloned 9. Wash the colonies off the plates with 1 to 2 ml of LB
locus does not carry genes whose presence results in an ob- medium by using a sterile spreading rod, and pellet the cells
servable phenotype, region-directed Tn5 mutagenesis can by centrifugation. Resuspend the pellet in 2 to 5 ml of LB
still be performed as described below. The insertions are medium. Isolate plasmid DNA from the cell suspension by
mapped, and a subset is chosen for gene replacement exper- using a (scaled-up) miniprep plasmid DNA preparation pro-
iments (see section 29.3.4) to analyze the effects of the dif- tocol (see chapter 30.1.2).
ferent Tn5 insertions when introduced into the genomic 10. Transform competent E. coli cells (see chapter
locus of the original bacterial strain. 31.1.3) with the plasmid DNA isolated in step 9, and select
for kanamycin- and plasmid marker-resistant transformants.
29.3.1. Vectors Store the desired number of transformants on master plates
In principle, every plasmid cloning vector carrying a cloned for further analysis. These colonies should harbor plasmids
region but not a kanamycin resistance gene can be used as carrying (generally) a single copy of Tn5 inserted into a dis-
a target for region-directed Tn5 mutagenesis. The length of tinct plasmid location (pXX::Tn5, where pXX is any plas-
the vector relative to the length of the insert should be con- mid vector used), which can be determined by restriction
sidered. Assuming a random insertional specificity for Tn5 mapping (see section 29.3.2).
29. Transposon Mutagenesis 693

29.3.1.2. Mutagenesis Protocol and Mutant which do not cut Tn5 (e.g., EcoRI, KpnI, and ClaI) or cut
Characterization Tn5 once (e.g., SalI, SmaI, and BamHI) or twice (e.g.,
Generally, step 8 of the above-described protocol should HindIII, BglII, NotI, and HpaI) (Fig. 1). Depending on the
yield plates with at least 500 to 1,000 Km' colonies, from restriction sites available in the pXX plasmid, different sin-
which plasmid DNA is to be isolated. If fewer than 500 gle and double digestions can be carried out to map the po-
colonies appear, the titer of the h::Tn5 lysate should be sition and relative orientation of the Tn5 insertion. As dis-
checked. Steps 9 and 10 will select colonies in which Tn5 cussed above for the mapping of chromosomal T n 5
has inserted into the plasmid and not into the chromosome insertions (section 29.2.3.2), first use an enzyme which does
of the original E. coli/pXX strain infected with h::Tn5. The not cut Tn5 and subsequently use an enzyme which cuts
effect of the Tn5 insertion on the expression of the gene(s) (twice) within the inverted repeats in order to generate spe-
or operon(s) carried on the cloned DNA of plasmid pXX cific Tn5-pXX junction fragments. By measuring the
can now be directly examined if there is a measurable phe- lengths of the junction fragments and subtracting the num-
notype. ber of base pairs contributed by Tn5 (Fig. l ) , the approxi-
mate distance of the Tn5 insertion from the nearest cleav-
1. If the cloned DNA is capable of complementing an age site for the enzyme used can be deduced. To help
auxotrophy or growth requirement of the E. coli strain used determine the relative orientation of the transposon within
in step 10, plate the E. coli/pXX::Tn5 transformants onto the cloned DNA, an enzyme cutting Tn5 asymmetrically
the appropriate minimal medium and select pXX::Tn5 de- (e.g., SmaI) can be used. For a detailed discussion of map-
rivatives no longer capable of complementing. These plas- ping strategies, see references 50 and 52.
mids are expected to carry Tn5 insertions in the locus of
interest, either in the structural gene(s) or in the corre- 29.3.3. Correlated Physical and Genetic Maps
sponding control region(s). For examples of this type of The physical mapping data (section 29.3.2) can now be
analysis, see references 53, 54, and 169. A similar strategy combined with the data obtained by examining the pheno-
would apply in analyzing a cloned DNA region conferring types corresponding to the different insertions (section
on the host E. coli strain the ability to catabolize certain 29.3.1.2) to create a correlated physical and genetic map.
compounds. For an example of this type of analysis, see ref- The Tn5 insertions that disturb the expression of the
erence 70. cloned locus under analysis would be expected to cluster
2. If the detectable phenotype associated with the and to be flanked by Tn5 insertions that do not affect the
cloned region is solely the production of a polypeptide of a expression of the locus. Thus, the extent of the locus can be
known molecular weight, introduce the pXX::Tn5 plasmids mapped to an accuracy of approximately 100 bp, depending
and the parental plasmid into minicells (160) or maxicells on the extent of the saturation mutagenesis experiment and
(176) via transformation and analyze the plasmid-encoded the lack of insertional specificity of Tn5 in the particular
polypeptides by sodium dodecyl sulfate-polyacrylamide gel cloned region under investigation. For examples of corre-
electrophoresis (see chapter 17.5.1 and reference 175). The lated maps thus created, see references 53,54, and 169, and
insertion of Tn5 into the locus would result in the disap- for discussions of the insertional specificity of Tn5, see ref-
pearance of the polypeptide of interest. In many cases, be- erences 18, 19, and 52.
cause of the presence of transcriptional as well as transla- The information thus obtained on the precise location
tional stop signals on Tn5, truncated polypeptides will of a gene of interest in a large fragment of cloned DNA can
appear. The sizes of these truncated polypeptides are indica- now be used to characterize this gene further, for example,
tive of the position of the Tn5 in the gene (or the operon) by directed DNA sequencing with Tn5-derived primers (see
relative to the start codon, and this information can be used section 29.4.6), characterization of the gene's regulatory re-
to deduce the direction of the transcription of the gene gions and expression (see section 29.4.1), and the construc-
(operon) once it is combined with the physical mapping tion, by directed gene replacement, of well-characterized
data (section 29.3.2). Examples of this type of analysis have Tn5 insertion mutants of the bacterial species from which
been described previously (53, 54, 169). the gene was cloned (section 29.3.4).
3. If the cloned region encodes an enzymatic activity Since Tn5 insertion mutations are generally polar on
detectable in E. coli or produces a protein for which an an- genes located downstream of the mutated gene in the same
tibody has been isolated, screen E. coli/pXX::Tn5 transfor- operon (18), transposon mutagenesis can also be used to de-
mants appropriately for a loss of function. For antibody- termine the structure of an operon carried on a cloned seg-
screening methods, see reference 175, and for an example of ment of DNA, especially when the method is combined
the latter type of Tn5 mutant screening, see reference 122. with an analysis of the polypeptides encoded by the (muta-
Clearly, there are many variations not listed here on the genized) recombinant plasmids (see section 29.3.1.2).
theme of how to screen for the desired phenotype. For an However, Tn5-mediated polarity is not always observed
extensive list of transposon-induced insertion mutations in (23), especially in analyzing Tn5-mutated operons carried
distinct genes of E. coli and Salmonella (and screening pro- by multicopy plasmids (54), and therefore one should exer-
cedures used to identify them), the reader is referred to ref- cise caution when interpreting the results obtained with
erence 15. For further discussion, see references 16 and 52. region-directed Tn5 mutagenesis of cloned DNA segments;
for a discussion, see references 18 and 52.
29.3.2. Physical Mapping of Plasmid-Borne 29.3.4. Gene ReplacementVectors and Methods
Insertions
A powerful extension of the Tn5 mutagenesis protocol is
1. Isolate plasmid DNA from the desired E. coli/ the use of gene replacement techniques to replace the wild-
pXX::Tn5 strains by using a rapid, miniprep isolation proto- type gene in the original bacterial strain with its well-
col (see chapter 16.1.2). characterized Tn5-mutated analog carried by a plasmid in
2. Digest the DNA with several restriction enzymes. E. coli. This technique allows the determination of the phe-
Several convenient enzymes are available for the physical notypes associated with specific Tn5-induced insertion mu-
mapping of Tn5 insertions in pXX::Tn5 plasmids, some of tations in the original organism. The general principle is to
Next Page

694 MOLECULAR GENETICS

introduce the Tn5-mutated gene region back into the orig- region was derived), an E. coli strain harboring the Tn5-
inal organism and force a double-crossover event between mutated gene region cloned into pRK290, and an E. coli
the wild-type gene sequences in the bacterial genome and strain harboring the helper plasmid pRK2013. Select for the
the corresponding sequences flanking the Tn5 insertion, recipient strain, Tn5 (Km'), and pRK290 (Tc').
followed by the loss of the wild-type gene. 3. Purify the obtained exconjugant once on the same se-
Two methods widely applicable to gram-negative bacte- lective plates.
ria other than E. coli have been described previously (50). 4. Perform a diparental mating (section 29.2.2.1) with
The first method was developed by Ruvkun and Ausubel the recipient strain harboring the Tn5-mutated gene on the
(172). In this method, the broad-host-range vector pRK290 pRK290 replicon and an E. coli strain harboring pPH1JI.
(60) is used to introduce the Tn5-mutated gene into the Select for the recipient strain, Tn5 (Km'), and pPH1JI
original bacterial strain by triparental conjugation (see sec- (Gm') .
tion 29.2.2.2) with the helper plasmid pRK2013 (60). This 5. Pick 100 to 1,000 exconjugants.
step is followed by the introduction of a plasmid incompat- 6. Screen for the loss of the pRK290 replicon by streak-
ible with pRK290, e.g., pPH1JI (97), into the same strain. ing the exconjugants onto master plates with and without
Selection for the antibiotic resistance marker of Tn5 tetracycline.
(kanamycin resistance) and that of the newly introduced 7. Purify about 10 different tetracycline-sensitive excon-
plasmid pPHl JI (gentamicin resistance) forces a double-ho- jugants once or twice on selective plates.
mologous-crossover event and the subsequent loss of the 8. Isolate the total chromosomal DNA from these tran-
pRK290 replicon carrying the wild-type sequences. conjugants, digest it with one or two restriction enzymes,
The second method uses narrow-host-range plasmids separate it by agarose gel electrophoresis, blot it onto a
carrying the ColEl mobilization (bom) site (such as filter, and hybridize it with the cloned wild-type gene re-
pBR322). These plasmids are able to replicate only in E. gion. The wild-type region should be replaced by the Tn5-
coli, but they are transferable to a wide range of other gram- mutated region. (In a digest with a restriction enzyme that
negative bacteria if the ColEl Mob and Tra functions are does not cut Tn5, such as EcoRI, the wild-type fragment
provided in trans (see section 29.2.2). Van Haute et al. should be replaced by a fragment 5.8 kb larger [see section
(209) described the use of E. coli GJ23 harboring the plas- 29.2.3.2 and reference 501.)
mids pGJ28 (MobC) and R64drdll (Tra+) to transfer
pBR322 derivatives carrying a Tn5-mutated gene effec- 29.3.4.2. Narrow-Host-Range Plasmids
tively into Agrobacterium tumefuciens. The ColEl replicon
cannot be stably maintained, and therefore, upon selection 1. Clone the Tn5-mutated region into a narrow-host-
for the resistance marker of Tn5, the mutated region will in- range vector, such as pBR322 or pSUP202.
tegrate into the genome via homologous recombination be- 2. Transform an E. coli strain providing the Mob and Tra
tween cloned plasmid-borne and resident sequences. In functions with the clone obtained in step 1. (Use strain
most cases, a cointegration of the vector will occur via a sin- GJ23 for pBR322 derivatives and strain S17-1 for pSUP de-
gle crossover, but at a certain frequency (depending on the rivatives.)
host and the mutated gene region), a double crossover will 3. Perform a diparental mating (section 29.2.2.1) with
occur, leading to a bona fide gene exchange. Alternatively, the recipient strain (from which the region was cloned) and
the above-described vectors of the pSUP series (see section the E. coli strain harboring the narrow-host-range vector
29.2.2) can be used for the same purpose, since they carry with the cloned Tn5-mutated region. Select for the recipi-
the mob region of RP4 cloned into narrow-host-range plas- ent strain and Tn5 (Km').
mids such as pACYC184 and pBR325 (190). Therefore, 4. Pick 100 to 1,000 transconjugants.
they can be mobilized into other gram-negative bacteria by 5. Screen for loss of the vector sequences by streaking
providing the Tra functions in trans. This step is normally the exconjugants onto master plates with and without the
achieved by using strain S17-1, which carries a chromoso- antibiotic resistance marker of the vector used but contain-
mally integrated copy of RP4 (Tra+). ing kanamycin for the selection of Tn5.
Gene replacement techniques can also be used to re- 6. Purify, once or twice on selective plates, about 10
place already existing Tn5 insertions with other Tn5 deriv- colonies that do not exhibit the antibiotic resistance phe-
atives carrying alternative selectable marker genes, reporter notype conferred by the vector.
gene fusions, portable promoters, DNA primers for restric- 7. Isolate the total chromosomal DNA from these
tion sites, and origins of transfer or replication (see section transconjugants, digest it with one or two restriction en-
29.4 and references 23, 58, and 154). This replacement ale zymes, separate it by agarose gel electrophoresis, blot it onto
lows a large degree of versatility. The only limitation in a filter, and hybridize it with the wild-type gene region. The
these types of replacement experiments is the length of the wild-type region should have been replaced by the Tn5-
homologous sequences needed for the double-recombination mutated region (see section 29.3.4.1, step 8).
event. This consideration is particularly relevant for trans-
posons used for the construction of gene fusions and those 29.3.4.3. Screening for Double Crossovers
carrying portable promoters, in which very limited Tn5 se- In some bacterial systems, depending on the host or the spe-
quences may be present between the reporter gene or pro- cial gene region mutagenized, it may be difficult to find
moter and the end of the transposon. double-crossover events. Sometimes more than 1,000
colonies must be screened to find exconjugants that have
29.3.4.1. Broad-Host-Range and Incompatible undergone the double homologous crossover ( 15 1).
Alternatively, a conditionally lethal gene has been used to
Plasmids
improve the gene replacement protocol. This gene allows
1. Clone the Tn5-mutated region into the broad-host- selection and screening for the bona fide gene exchange
range vector pRK290. event in several bacterial species. The sacB gene of Bacillus
2. Perform a triparental mating, as outlined in section subtilis (coding for levansucrase) confers sensitivity to 5%
29.2.2.2, with the recipient strain (from which the cloned sucrose; i.e., gram-negative organisms expressing the sacB
Plasmids
MARCEL0 E. TOLMASKY. LUIS A . ACTIS. TIMOTHY J . WELCH. AND
JORGE H. CROSA

30.1. ISOLATION ............................................. 710


30.1.1. Ordinary Plasmids by Large-Scale Methods.................. 710
............................. 711
30.1.1.1. Cold Triton X.100
. ............................. 711
2. Hot Triton X-100
. ................................. 712
3. Alkaline pH
. ......................... 712
4. Sodium Dodecyl Sulfate
. ..................................... 712
5. Boiling
. .................................. 712
6. Lysostaphin
. ....................... 713
7. Lysostaphin and Lysozyme
. ..................................... 713
8. Sucrose
. ................................... 713
9. Penicillin
30.1.2. Ordinary Plasmids by Small-Scale Methods.................. 713
............................. 713
30.1.2.1. Cold Alkaline pH
. .............................. 714
2. Hot Alkaline pH
. ................................. 714
3. Acid Phenol
. ......................... 714
4. Phenol and Chloroform
. ..................................... 714
5. Boiling
.......................................
30.1.3. Large Plasmids 714
.............................. 715
30.1.3.1. Lysis in Solution
. ........................... 715
2. Lysis in Agarose Gel
......................................
30.1.4. Linear Plasmids 716
. ............................... 716
5. Single-Stranded Plasmids
. .............................. 716
6. Single-Stranded Phagemids
30.2. PURIFICATION .......................................... 717
......................... 717
30.2.1. Extraction with Organic Solvents
. ....................... 717
2. Equilibrium Gradient Centrifugation
. ..................... 718
3. Precipitation with Polyethylene Glycol
.4. Chromatography..................................... 718
.5. Commercial Methods.................................. 718
30.3. QUANTIFICATION ....................................... 718
.................... 718
30.3.1. Quantification of Plasmid Concentration
..................... 718
30.3.1.1. Spectrophotometric Methods
. .......................... 719
2. Fluorometric Methods
.......................... 720
30.3.2. Quantification of Plasmid Forms
...................... 720
30.3.2.1. Agarose Gel Electrophoresis
. ..................... 720
2. Capillary Gel Electrophoresis
30.4. CHARACTERIZATION .................................... 720
30.4.1. Size............................................... 721
.......... 721
30.4.1.1. Gel Electrophoresis of Native Plasmid DNA
. ......... 721
2. Gel Electrophoresis of Digested Plasmid DNA
. ................... 721
3. Pulsed-Field Gel Electrophoresis
30.4.2. Copy Number ....................................... 723
....... 723
30.4.2.1. CsCLEthidium Bromide Gradient Centrifugation
. ...................... 724
2. Agarose Gel Electrophoresis
30.4.3. Base Composition.................................... 724
.4. DNA Homology .................................... 724
................ 724
30.4.4.1. Single-Strand-Specific Endonuclease
. ............. 725
2. DNA Immobilization and Hybridization
..................... 727
30.4.5. Replication and Maintenance Functions
. ........................................ 727
6. Visualization
...................................... 728
30.4.6.1. FISH
. .............................. 728
2. GFP-Lac1 Fusion
...................................... 728
30.4.7. Incompatibility
.8 .Curing............................................ 729
.................. 729
30.4.8.1. Elevated Incubation Temperature
. ............................. 729
2. Intercalating Dyes

709
710 w MOLECULAR GENETICS

.3. Sodium Dodecyl Sulfate ......................... 729


.4. Electroporation ............................... 729
.5. Kick-Out ................................... 729
30.5. NOVEL USE OF PLASMID D N A ............................. 729
..............
30.5.1. Plasmid D N A from a Pharmaceutical Perspective 729
30.6. REFERENCES ............................................ 730

Plasmids range in size from 1 to more than 200 kbp (31), DNA have been recently developed and are currently being
and even larger megaplasmids were detected in Rhizobium used in many laboratories.
spp. (8,48,79). Although they replicate separately from the The initial characterization of a bacterial plasmid usually
host genome, plasmids do rely on host-encoded factors for is at the genetic level. If a bacterial trait is suspected of
their replication. Although not essential for the survival of being plasmid mediated, gene transfer experiments often
bacteria, plasmids may encode a wide variety of genetic de- document transmissibility of plasmid determinants inde-
terminants, which permit their bacterial hosts to survive pendently of chromosomal determinants. Moreover, the
better in an adverse environment or to compete better with elimination (curing) of a genetic trait by treating the bac-
other microorganisms occupying the same ecological niche. terial population with chemical or physical curing agents
Plasmids are found in a wide variety of bacteria, and it is as such as acridine dyes, ethidium bromide, sodium dodecyl
difficult to generalize about plasmids as it is to generalize sulfate (SDS), antibiotics, high temperature, or electropora-
about the bacteria that harbor them. The medical impor- tion (63, 95) indicates that the expression of that genetic
tance of plasmids that encode antibiotic resistance and spe- trait is linked to the presence of a plasmid. Most curing
cific virulence traits has been well documented (60, 132, agents are also mutagenic agents. In most cases, however, it
157). Plasmids have also been shown to influence signifi- is essential to document that a plasmid is present and un-
cantly several properties contributing to the usefulness of equivocally associated with the genetic trait in question.
bacteria in agriculture and industry (21, 32, 42, 61). The Clinically, the plasmid content of a cell can also be a useful
types of genes that encode these properties are frequently epidemiological marker (62, 103). If possible, it is best to
located in transposable elements, producing great variation transfer a plasmid by physical or genetic means into a bac-
and flexibility in the constitution of plasmids. These extra- terial host that is known to be devoid of plasmids. The ad-
chromosomal elements are of equal importance, however, vantage, of course, is that any single plasmid transferred to
for the study of the structure and function of DNA. and subsequently isolated from such a host can be analyzed
Plasmids have taken on paramount importance in recombi- without fear of contamination by a preexisting plasmid. In
nant DNA technology, particularly as cloning vehicles for some cases, genetic methods are not available, and so one
foreign genes. Most bacterial plasmids are usually covalently must directly examine a bacterium to determine its plasmid
closed circular (CCC) DNA; however, linear DNA plas- content. Such an analysis usually can be performed to de-
mids, as well as single-stranded plasmids which are interme- termine simply whether a plasmid is or not present.
diates in replication, have also been described (2, 24, 38, Subsequently, one can examine a strain cured of a trait to
39,50,64,75,79, 135, 137, 158). Plasmid-encoded mainte- determine whether a plasmid is lost concomitantly with a
nance functions have also been shown to interact with the particular host cell function. The compositions of the com-
host cell (72). Plasmids have also been found in eukaryotic monly used reagents mentioned throughout this chapter are
microorganisms. Recently, plasmid transfer from bacteria to indicated in Table 1. In this chapter we give a representa-
eukaryotic cell lines maintained in culture has also been tive rather than comprehensive account of techniques and
demonstrated (30, 55, 148). With the advent of the ge- references concerning the biology of plasmids and their use
nomics era, websites that include complete sequences of as tools in molecular biology and pharmacology. Additional
plasmids as well as a data base for plasmids replicons have information on bacterial plasmids can be found in refer-
been created (http://www.ncbi,nlm.nih.gov/genomes/static/ences 11,60, 114, and 132.
o.htm1; http://www.essex.ac.uk/bs/staff/osborn/DPR_home
.htm [in construction at the time of writing of this chapter];
http://www.genomics.ceh.ac.uk/plasmiddb/ [currently up- 30.1. ISOLATION
dated]).
In this section are described methods of isolating plasmid
This chapter deals with the isolation, purification, and
DNA that do not require the use of commercially available
characterization of bacterial plasmids and also provides
materials such as prepared chromatography columns or
some information concerning the novel application of plas-
reagents. Some characteristics are explained before each
mid biology in gene therapy. Many of the techniques cur-
procedure. When dealing with a new isolated bacterium, a
rently used for the isolation of plasmid DNA from bacteria
trial-and-error strategy in selecting the appropriate method
are based on its supercoiled CCC configuration. All of the
is recommended.
techniques require some means of gently lysing bacterial
cells so that the plasmid DNA is preserved intact and can
be physically separated from the more massive chromo- 30.1 .I. Ordinary Plasmids by large-Scale Methods
somal DNA. This separation is more easily achieved with Large-scale methods permit the preparation of ordinary
smaller plasmids, and the degree of difficulty increases as plasmid DNA in amounts such that repetitive analyses (re-
the size of the plasmid increases, particularly with some striction endonuclease digestions, preparative isolation of
megaplasmids. This complication is often compounded by specific DNA fragments, cloning and subcloning, etc.) can
the fact that very large plasmids are normally present in low be carried out. Generally, these methods are used in con-
copy numbers. Even more specialized techniques are needed junction with a subsequent purification step, such as equi-
to isolate linear plasmids. Kits for the isolation of plasmid librium density gradient centrifugation in CsC1-ethidium
30. Plasmids 711

TABLE 1 Composition of commonly used reagents


Reagent Composition
Acetate solution 60 ml of 5 M potassium acetate, 11.5 ml of glacial acetic acid, 28.5 ml of water
Boiled RNase ............................. 5 mg of RNase per ml in 0.15 M NaC1, heated at 100C for 20 min to destroy
DNase
50X Denhardt's solution ..................... 1% Ficoll, 1% polyvinylpyrrolidone, 1% bovine serum albumin
Gel-loading buffer 0.25% bromophenol blue, 0.25% xylene cyanol FF, 30% glycerol
Phenol .................................. Equilibrated with 0.1 M Tris-HC1 (pH 8) and 0.2% P-mercaptoethanol. Oxidation
is prevented by the addition of 0.1% 8-hydroxyquinoline
20X salt-sodium citrate (SSC) . . . . . . . . . . . . . . . . 175.3 g of NaCl and 88.2 g of sodium citrate per liter adjusted to pH 7.8
20X salt-sodium phosphate-EDTA (SSPE) . . . . . . . 3.6 M NaCI, 0.2 M sodium phosphate (pH 7.7), 0.02 M EDTA
Sucrose-Triton-EDTA-Tris (STET) . . . . . . . . . . . . . 8% Sucrose, 0.5% Triton X-100, 0.05 M EDTA, 0.05 M Tris-HC1 (pH 8.0)
TE buffer. ................................ 0.01 M Tris-HC1,O.OOl M EDTA (pH 8.0)
TES buffer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0.05 M Tris-HCI, 0.005 M EDTA, 0.05 M NaCl (pH 8.0)
Tris-acetate (electrophoresisbuffer) . . . . . . . 0.04 M Tris, 0.001 M EDTA (pH 8.0), 0.02 M glacial acetic acid
Tris-borate (electrophoresisbuffer) . . . . . . . . . . . . . 0.089 M Tris, 0.0025 M EDTA (pH 8.0), 0.089 M boric acid
Triton X-100 lytic mixture . . . . . . . . . . . . . . . . . . .1 ml of Triton X-100 in 0.01 M Tris-HC1 (pH 8.0), 25 ml of 0.25 M EDTA
(pH8.0), 5 ml of 1 m Tris-HC1 (pH 8.0), and 69 ml of water

bromide gradients or precipitation with polyethylene gly- can be used for further analysis. The plasmid-enriched su-
col. Most of the methods described below are generally used pernatant fraction (3.2 ml) can also be purified and con-
for isolating ordinary plasmid DNA with molecular size no centrated by centrifugation in a CsC1-ethidium bromide
greater than 160 kbp. density gradient as described in section 30.2.
30.1 . I . I . Cold Triton X-1 00 30.1 . I .2. Hot Triton X-1 00
The cold Triton X-100 procedure (28, 81) works well for
The hot Triton X-100 method is a modification of the fore-
isolating plasmid DNA from Escherichia coli K-12 strains,
going procedure, which improves the sharpness of the DNA
with the cells lysed by the nonionic detergent Triton X-100.
bands in agarose gels for lysates obtained from certain bacte-
1. Grow the cells at 37C either in a rich medium or in ria, e.g., Psewlomonas aeruginosa, Serratia marcescens, Proteus
a suitable minimal medium (under selection, if possible), rettgeri, and Klebsiella pneumoniae. This procedure was suc-
with gentle aeration achieved by shaking the flask at a rate cessfully used in the analysis of plasmid profiles of clinical iso-
just sufficient to keep the surface of the medium in motion. lates of Acinetobacter baumannii (3, 62). This method is use-
The following details are for a 100-ml culture in a 250-ml ful for isolation of plasmids of sizes ranging from 2 to 100 kbp.
Erlenmeyer flask, but the method can be scaled up or down
1. Grow the culture in 40 ml of brain heart infusion
proportionally. To ensure optimum lysis, harvest the cells in
broth or any other appropriate rich medium in a 100-ml flask
the mid-logarithmic phase of growth.
on a shaker, as in step 1 of the method described above.
2. Harvest the cells by centrifugation in a 250-ml bottle
2. Harvest the cells by centrifugation at 3,000 X g for
(e.g., for 10 min at 12,000 X g a t 5C). Resuspend the pel-
10 min.
let with 15 ml of Tris-EDTA-salt (TES) buffer (Table l),
transfer to a 40-ml tube, and centrifuge for 10 min at 12,000
3. Suspend the cell pellet in 5 ml of TES buffer as in
step 2 of the method described above.
X g at 5C. Resuspend the pellet with 1 ml of ice-cold 25%
sucrose solution (in 0.05 M Tris-0.001 M EDTA [pH 8.0]),
4. Centrifuge the cells as in step 2.
and place the tube in crushed ice for 30 min.
5. Suspend the cell pellet in 2 ml of a 25% sucrose so-
lution (in 0.001 M EDTA-0.05 M Tris-HC1 [pH 8.01). Place
3. Add 0.2 ml of a freshly prepared lysozyme solution (5
the tube in ice for 20 min.
mg/ml in 0.25 M Tris-HC1 [pH 8.01). Mix the contents by
swirling the tube several times, and then place the tube in
6. Add 0.4 ml of a lysozyme solution (10 mg/ml in 0.25
ice for 10 min. This step can be skipped when working with
M Tris HC1 [pH 8.01) to the suspension. Place the tube in
ice for 20 min.
some bacterial species.
4. Add 0.4 ml of 0.25 M EDTA (pH 8.0). Swirl the tube, 7. Add 0.8 ml of 0.5M EDTA (pH 8.0) to the cell sus-
pension.
and then place i.t in ice for another 10 min.
5. Add 1.6 ml of Triton X-100 lytic mixture (1 ml of 10% 8. Lyse the cells with 4.4 ml of Triton lytic mixture as
in step 5'of the method described above. Mix gently.
Triton X-100 in 0.01 M Tris-HCl [pH 8.01, 25 ml of 0.25 M
EDTA [pH 8.01, 5 ml of 1 M Tris-HC1 [pH 8.01, 69 ml of
9. Heat the tube at 65C for 20 min.
10. Sediment the cellular debris by centrifugation at
water). After very gentle swirling to mix the contents thor-
27,200 X g for 40 min.
oughly, place the tube in ice for 20 min.
11. Decant, and then adjust the supernatant solution to
6. Centrifuge at 35,000 X g at 5C for 20 min, decant
the supernatant, and discard the pellet.
0.5 M NaCl and 10% polyethylene glycol by use of stock so-
lutions of 5 M NaCl and 40% polyethylene glycol (molec-
About 95% of the plasmid is separated from the pellet ular weight, 1,000 to 6,000).
containing the chromosomal DNA and cellular debris and 12. Store the tube at 4C overnight.
712 MOLECULAR GENETICS

13. Sediment the resulting precipitate by centrifugation 5. Precipitate chromosomal DNA by adding 0.9 ml of
at 3,000 X g for 10 min, and resuspend the pellet in 1 to 2 3 M NaCl (to bring the final concentration to 1 M NaC1).
ml of 0.25 M NaCl containing 0.001 M EDTA and 0.01 M Place the tube in ice for at least 2 h to allow complete pre-
Tris-HC1 (pH 8.0) at 4C. cipitation.
14. Precipitate the DNA by adding 2 volumes of 95% 6. Centrifuge the precipitated chromosomal DNA and
ethanol at -20C and let the tube stand overnight at any remaining cell debris at 35,000 X g for 30 min at 4"C,
-20C. Alternatively, the DNA can be precipitated by let- and decant the supernatant (enriched for plasmid DNA)
ting the tube stand for 30 min at - 70C. Depending on the into a 15-ml Corex tube.
bacteria and the nature of the plasmid, the ideal length and 7. Remove the RNA by adding 1 volume of distilled
temperature of incubation may vary. water and 4 pl of a 5 mg/ml boiled RNase solution in 0.15
15. Collect the plasmid DNA pellet by centrifugation at M NaCl (the enzyme must be heated at 100C for 20 min
12,000 X g at 4C. Resuspend it in TES. before use to destroy DNases). Incubate the tube at 37C for
an additional 1 h.
30.1 .I .3. Alkaline pH 8. Add an equal volume of Tris-saturated phenol to de-
The alkaline pH method described below is a shorter ver- proteinize the mixture (as described in section 30.2.1).
sion of the one described by Birnboim and Doly (13). I t is Shake the mixture vigorously, and then centrifuge at 3,000
used routinely to prepare plasmid DNA from E. coli, Vibrio x g for 20 min at 2C. Remove the aqueous (upper) phase
anguillarum, K. pneumoniae, and Enterobacter cloacae and has containing the plasmid DNA. Repeat this step once. Ex-
been used by others for many other genera. The pH should tract the aqueous phase once with 1 volume of chloroform-
not be allowed to go above 12.5, to avoid plasmid DNA al- isoamyl alcohol (24:1, vol/vol) to remove residual phenol.
teration. Warning: phenol is poisonous and caustic.
9. Transfer the aqueous phase to a 30-ml Corex tube,
1. Harvest cells from a 50- to 200-ml (depending on the and add 0.6 ml of 3 M sodium acetate to make the final con-
plasmid copy number) culture by centrifugation at 12,000 centration 0.3 M.
X g at 4C. Resuspend the pellet with 5 ml of a solution 10. Add 2 volumes of cold (-20C) 95% ethanol. Mix
containing 0.05 M Tris-HC1,O.OlO M EDTA, and 0.050 M the solution well, and place at -20C overnight.
glucose (pH 8.0). 11. Sediment the precipitated DNA at 12,000 X g for 20
2. Add 10 ml of a solution containing 0.2 N NaOH and min at - lO"C, decant the ethanol thoroughly, and resuspend
1% SDS. Mix gently. The solution should clear and turn the plasmid DNA in 0.2 ml of 0.006 M Tris-HC1 (pH 7.5).
viscous.
3. Add 7.5 ml of acetate solution (make the solution by 30.1 .I .5. Boiling
adding 60 ml of 5 M potassium acetate, 11.5 ml of glacial
The boiling method described below is a modification by
acetic acid, and 28.5 ml of water). Mix thoroughly by shak-
Reddy et al. (110) of the original method of Holmes and
ing. A white precipitate should form.
Quigley (70), which is commonly used as a small-scale
4. Centrifuge at 30,000 X g for 20 min at 4C. method. A related modification was also reported by Lev
5. Discard the pellet. If the supernatant contains pellet (85) and Lev and Seveg (86). The following method has
particles or is milky, filter it to obtain a clear solution. Add
been used with E. coli cells, but it is possible that it could be
12.5 ml of isopropanol, and let stand 15 min at room tem-
used with other bacteria.
perature.
6. Sediment the plasmid DNA by centrifugation at 1. Collect cells from a 1-liter culture by centrifugation at
12,000 X g at room temperature. 16,000 X g for 10 min.
7. Resuspend the DNA in Tris-EDTA (TE) buffer (Table 2. Resuspend the cells in 10 ml of a solution containing
1). 0.050M Tris-HC1 (pH 7.5), 0.062 M EDTA, 0.4% Triton
X-100, and 2.5 M LiC1. Add 0.5 ml of 20% lysozyme in
The original technique recommends that, after addition
water. Mix gently, and incubate at 25C for 10 min.
of the alkali and acetate solutions in steps 2 and 3, the sam-
3. Transfer the solution to a plastic bag (8 by 8 cm), and
ple stand in ice for a certain time. However, skipping these
seal. Immerse the bag in a boiling-water bath for 1 min.
waiting steps results in the same yield and quality of plasmid
DNA.
4. Transfer the contents to a centrifuge tube, let it stand in
ice for 5 min, and centrifuge at 27,200 X gat 4C for 20 min.
5. Transfer the supernatant to a fresh tube, add 2 volumes
30.1 .I .4. Sodium Dodecyl Sulfate of ethanol, mix, and keep the tube in dry ice for 10 min.
The SDS method below is a modification (57) of that de- 6. Sediment the DNA by centrifugation as in step 4,
scribed by Hirt (69). wash with 70% ethanol, and let dry.
1. Sediment the cells by centrifugation at 12,000 x g 7. Resuspend the DNA in 0.5 ml of TE buffer (Table l),
for 10 min at 4"C, and then resuspend them in 1.5 ml of and add 10 pl of a boiled RNase solution (10 mg/ml dis-
25% sucrose containing 0.05M Tris-HC1 and 0.001 M solved in 0.15 M NaC1). Incubate at 25C for 20 min.
EDTA (pH 8.0). 8. Extract the plasmid DNA-containing solution with
2. Add 0.2 ml of lysozyme solution (10 mg of lysozyme phenol as described in section 30.2.1, and precipitate with
per ml in 0.25 M Tris-HC1 [pH 8.01) to the cell suspension, 1/9 volume of 5 M potassium acetate (pH 4.8) and 2.5 vol-
and mix gently. Place the tube in ice for 15 min. umes of ethanol.
3. Add 0.1 ml of 0.25 M EDTA at pH 8.0, mix gently 9. Collect the plasmid DNA by centrifugation, and re-
by slow inversion of the tube, and replace the tube in ice for suspend in TE buffer.
10 min.
4. Add 0.1 ml of 20% SDS solution. Mix the suspen- 30.1 .I .6. Lysostaphin
sion gently, and keep in ice for 10 min. During this time, the The following procedure has been used successfully to ob-
cells lyse and the solution becomes viscous. tain plasmid DNA from gram-positive bacteria, as well as
30. Plasmids 713

from gram-negative bacteria that are resistant to lysis by 6. Centrifuge at 10,000 x g at 4"C, and resuspend the
Triton X-100 or SDS. pellet in 0.7 ml of TE.
1. Pellet the cells from a 30-ml overnight culture by cen-
7. This preparation can be phenol extracted and ethanol
precipitated.
trifugation at 16,000 X g for 10 min and suspend them in
1.5 ml of 0.0075 M NaC1-0.050 M EDTA (pH 7.0). 30.1 .I .9. Penicillin
2. Add lysostaphin to a final concentration of 15 pg/ml. The following procedure can be used for obtaining plasmid
(Double the enzyme concentration for Staphylococcus epi-
DNA from gram-positive and gram-negative organisms that
dermidis.) Incubate the suspension at 37C for 15 min with are resistant to Triton and SDS but sensitive to penicillin.
gentle agitation, and then place the flask in ice.
If the bacterium under study produces a p-lactamase, how-
3. Add 1.5 volumes of a mixture containing 0.4% de- ever, a penicillinase-resistant antibiotic (e.g., a cephal-
oxycholate, 1% Brij 58, and 0.3 M EDTA (pH 8.0) to
osporin) may be effective. It is not certain whether other
achieve cell lysis. Mix the viscous contents of the tube gen-
antimicrobial agents active against cell wall biosynthesis
tly, and put the tube in ice for 15 min. would prove successful, although it may be useful to inves-
4. Pellet the cellular debris by centrifugation at 23,000 X tigate this possibility.
g for 20 min at 4"C, and decant the supernatant fluid into a
15-ml Corex tube. 1. Grow bacteria to the mid-logarithmic phase, add 1 mg
5. Add 1 volume of distilled water and 4 pl of boiled of penicillin G per ml, and incubate the culture at the opti-
RNase solution (1 mg/ml) to the supernatant fluid. mum growth temperature for an additional 2 h. (Note: It is
Incubate the tube at 37C for 1 h. Proceed with plasmid convenient to perform a preliminary experiment to deter-
DNA purification as described for SDS lysis, step 8. mine the time and antibiotic concentration that give opti-
/
/ m u p protoplast formation; the goal is to achieve this maxi-
30.1 .I .7. Lysostaphin and Lysozyme mum without massive lysis. The addition of 1 M sucrose to
The lysostaphin and lysozyme method was adapted from the growth medium may be beneficial in achieving this goal.)
that of Lyon et al. (88). 2. Harvest the penicillin-treated cells by centrifugation,
and wash them twice with an equal volume of 0.01 M Tris-
1. Collect the cells from a 20-ml culture, wash them, and HC1 (pH 8.2) of the original culture.
resuspend them with 1 ml of TES buffer (Table 1). Add 3. Suspend the cell pellet in 0.25 volume of 0.02 M Tris-
lysostaphin and lysozyme to a final concentration of 75 HC1 (pH 8.2)-0.5 volume of 1 M sucrose-0.25 volume of
pg/ml for each enzyme. Incubate for 1 h at 37C. lysozyme (4 mg/ml) in 0.02 M Tris-HC1 (pH 8.2).
2. Add 1 ml of a solution containing 2.5% Sarkosyl, 0.4 4. Incubate the cell suspension at 3C for 1 h with shak-
M EDTA (pH 8), and 100 pg proteinase K. Mix by inver- ing. Centrifuge at 27,000 X g for 15 min.
sion, and incubate at 65C for 30 min. 5. Gently suspend the cells in 0.02 M Tris-HC1 (pH 8.2)
3. Add 2 ml of a solution containing 3 M sodium acetate and 0.01 M EDTA, taking care to ensure that the cells are
and 0.005 M EDTA (pH 7.0), and keep in ice for 30 min. homogeneously dispersed.
4. Centrifuge at 40,000 X g at 4C for 30 min. Transfer 6. Add 0.1 volume of 10% SDS to the cells. Lysis should
the supernatant to a fresh tube, and add 1 volume of iso- be complete within 10 min.
propanol. Let stand at room temperature for 10 min.
5. Collect the DNA by centrifugation at 27,000 X g for Proceed from here as described for the SDS lysis method
10 min. (section 30.1.1.4, step 8).
6. Dissolve the DNA pellet in 0.04 ml of water. Add Among gram-positive bacteria such as streptococci, lac-
boiled RNase to a final concentration of 50 pg/ml. Incubate tococci, and lactobacilli, addition of 0.01 M L-threonine to
for 30 min at 37C. This DNA can be phenol extracted and the growth medium weakens the cell wall and leads to eas-
ethanol precipitated. ier lysis after lysozyme treatment followed by detergent lysis
(23,90). Glycine added at the early to mid-log phase can be
30.1 .I .8. Sucrose just as effective as penicillin but, like the latter and unlike
The sucrose method can be used with gram-negative as well threonine, will stop growth of the culture (90).
as gram-positive bacteria. Bacterial lysis is performed by ap-
plying an osmotic shock and then treating with detergent 30.1.2. Ordinary Plasmids by Small-Scale Methods
(112). Small-scale methods permit the rapid preparation of ordi-
nary plasmid DNA in small amounts, normally enough for
1. Harvest cells from 50 ml of culture, and wash them a few assays such as restriction endonuclease digestion for
with 0.7 ml of 0.05 M Tris-HC1 (pH 8.0) containing 25% screening or preparation of probes. Some of them produce a
sucrose. DNA of high enough quality to perform a few sequencing
2. Resuspend the cells in 0.7 ml of 0.05 M Tris-HC1 (pH reactions, and some produce a DNA of inferior quality.
8.0) containing 100% sucrose. Mix by pipetting, which These methods do not require a subsequent purification
takes time since 100% sucrose solution is very thick. step for the stated purposes.
Incubate for 3 h at 37C.
3. Add 3 ml of a solution made by mixing 0.5 parts of 30.1.2.1. Cold Alkaline pH
0.25 M EDTA (pH 8.1) with 2.3 parts of 1% Brij 58-0.4% The cold alkaline pH method is a scaled-down version of
sodium deoxycholate-0.05 M Tris-HC1 (pH 8.0)-0.06 M the one described in large scale (section 30.1.1.3) that has
EDTA-2 M NaC1. Mix gently, and incubate at room tem- been used to prepare E. coli and V. unguilhrum plasmids.
perature for 1 h.
4. Pellet the cellular debris for 30 min at 10,000 X g at 1. Harvest cells from 1.5 ml of culture by centrifugation
4C. in a microcentrifuge (16,000 X g) for 10 s. Resuspend the
5. Add 0.6 volume of isopropanol to the supernatant, pellet with 100 pl of a solution containing 0.05 M Tris-
mix, and keep for 15 min. HC1, 0.010 M EDTA, and 0.050 M glucose (pH 8.0).
714 MOLECULAR GENETICS

2. Add 200 pl of a solution containing 0.2 N NaOH and 1. Proceed through step 4 as described for the alkaline
1% SDS. Mix gently. The solution should clear and turn lysis method (section 30.1.2.1). Transfer 400 pl of the su-
viscous. pernatant to a fresh microcentrifuge tube, and add 400 pl of
3. Add 150 pl of acetate solution (make the solution acid phenol (made by equilibration of distilled phenol with
with 60 ml of 5 M potassium acetate, 11.5 ml of glacial 0.05 M sodium acetate pH 4.0). Vortex and separate the
acetic acid, and 28.5 ml of water). Mix thoroughly by shak- layers in a microcentrifuge (16,000 X g).
ing. A white precipitate should form. 2. Transfer the upper phase to another microcentrifuge
4. Centrifuge in a microcentrifuge (16,000 X g) for 5 tube, and extract with 1 volume of chloroform-isoamyl al-
min. cohol (24:1, vol/vol).
5. Transfer 400 pl of the supernatant to a fresh micro- 3. Transfer the upper phase to another microcentrifuge
centrifuge tube, and add 1 mi of cold ethanol. tube, and add 2.5 volumes of cold ethanol.
6. Recover the plasmid DNA by centrifugation for 5 4. Recover the plasmid DNA by centrifugation for 5
min, pour off the supernatant, wash once with 70% cold min, pour off the supernatant, wash once with 70% cold
ethanol, and dry. ethanol, and dry. Resuspend the pellet in 50 pl of TE buffer
7. Resuspend the DNA in 20 to 50 pl of TE buffer. (Table 1) containing 50 pg of boiled RNase per ml.
This DNA is suitable for restriction endonuclease analy- 30.1.2.4. Phenol and Chloroform
sis and Southern blot hybridization (see section 30.4.4).
The phenol-chloroform method involves the lysis of bac-
After RNase treatment and either phenol extraction or
terial cells by phenol-chloroform, which renders a DNA
purification with silica powder (e.g., GENECLEAN
preparation suitable for restriction endonuclease treatment
[QeBIOgene, Carlsbad, CAI), the DNA can be used for
as well as transformation (4, 110, 120). Broth cultures or
probe preparation and cloning experiments. CAUTION:
pooled bacterial growth from solid surfaces can be used for
(i) If the plasmid DNA is to be used as a probe against ge-
this method.
nomic DNA from an organism that is homologous to the
host from which the plasmid is isolated, hybridization might 1. Transfer 1.5 ml of culture or suspended scraped cells
occur between the blotted DNA and the residual chromo- from a plate to a microcentrifuge tube, and centrifuge for 3
somal DNA from the host present in the plasmid prepara- min.
tion. This situation can be avoided either by purifying the 2. Resuspend the pellet in 0.05 ml of a solution contain-
plasmid DNA by dye buoyant-density gradient centrifuga- ing 0.01 M Tris-HC1 (pH 8.0),0.1 M NaCl, and 0.001 M
tion or by preparing plasmid DNA by the acid-phenol EDTA. Add 0.05 ml of phenol-chloroform solution (made
method described below. (ii) Sometimes the plasmid DNA by mixing phenol, chloroform, and isoamyl alcohol
is not clean enough at this point for restriction endonucle- [25:24:1, vol/vol/vol]), and vortex.
ase analysis. In this case, a phenol extraction usually renders 3. Centrifuge for 5 min to yield a clear supernatant.
the preparation suitable for this purpose. Transfer the upper phase to another tube, add ammonium
acetate to a final concentration of 2 M, and add 2 volumes
30.1.2.2. Hot Alkaline pH of cold ethanol. Let stand in ice for 15 min.
The hot alkaline pH method is a modification (73) of the 4. Centrifuge for 10 min to collect the DNA. Wash the
alkaline lysis method described above (section 30.1.2.1). pellet with 70% cold ethanol, dry it, and resuspend it in
This rapid miniscreen method has been used to analyze 0.02 ml of TE buffer.
large and small plasmids in many types of gram-positive as
well as gram-negative bacteria. 30.1.2.5. Boiling
The boiling method described here is the original method
1. Harvest cells from 2 ml of culture by centrifugation,
of Holmes and Quigley (70),used effectively for screening
and resuspend the cell pellet in 1 ml of 0.002 M EDTA-
of high-copy-number plasmids.
0.04 M Tris-acetate (pH 8.0).
2. Add 2 ml of a solution containing 0.05 M Tris and 3% 1. Transfer 1.5 ml of culture to a microcentrifuge tube,
SDS (pH 12.6) (adjust the pH with 2 N NaOH). Mix uni- and pellet the cells.
formly, and incubate at 60C for 30 min. 2. Resuspend in 85 p1 of STET buffer (8% sucrose, 0.5%
3. Add 6 ml of phenol-chloroform (l:l),and mix until Triton X-100, 0.05 M EDTA, 0.05 M Tris-HC1 [pH 8.01).
complete emulsification occurs. Separate the aqueous phase Add 10 pl of 10 mg/ml lysozyme dissolved in water and let
by centrifugation at 10,000 x g for 15 min at 4C. stand on ice for 5 min.
4. Transfer the aqueous phase to a fresh tube containing 3. Place in a boiling-water bath for 90 s. The optimal
1 volume of chloroform. Mix, and again separate the phases. time should be determined for different strains, since effec-
5. Recover the aqueous phase containing the plasmid tiveness seems to be strain dependent.
DNA. 4. Immediately centrifuge for 10 min in a microcen-
trifuge at full speed.
30.1.2.3. Acid Phenol 5. Remove the viscous pellet with a pipette tip, and add
The acid phenol method (151) is based on the addition of 85 pl of cold isopropanol. Let stand at -20C for 20 min.
an acid phenol extraction to the cold alkaline lysis prepara- 6. Collect the DNA by centrifugation in a microcen-
tion of DNA (section 30.1.2.1) and has been used very suc- trifuge (16,000 X g), wash the pellet once with 70% cold
cessfully. The additional use of phenol produces a prepara- ethanol, and dry it. Resuspend the pellet in 20 pl of TE
tion of quality similar to that obtained after separation in a buffer. Another method involving lysis by boiling is re-
CsC1-ethidium bromide gradient. This DNA can be used ported in reference 161.
for sequencing and other procedures such as exonuclease
111-generated nested deletions. Acid phenol removes nicked 30.1.3. Large Plasmids
and linear DNA, leaving only the supercoiled molecules, by Large plasmids have been found in several bacterial systems,
a mechanism still not well understood (159). including the tumor-inducing and root-inducing plasmids
30. Plasrnids 715

of Agrobacterium tumefaciens and A. rhizogenes (145, 156), 7. Centrifuge in a microcentrifuge (16,000 X g) for 10
the Sym plasmids of Rhizobium spp. (6), the H incompatibil- min to sediment debris, and then pour the supernatant fluid
ity group of antibiotic resistance plasmids (3l ) ,the camphor- into another microcentrifuge tube (do not attempt to re-
degradative plasmid of Pseudomonas putidu (22, 102), and cover the small amount of fluid remaining in the bottom of
the various F factors (91, 122, 127). Several methods have the tube).
been developed which permit the isolation of small as well 8. Add 0.55 ml of isopropanol to the supernatant fluid
as large plasmids (5,36, 58, 149). In addition, several tech- to precipitate the DNA. After mixing, place the tube at
niques have been developed which permit the use of very -20C for 30 min.
small working volumes or even material from a single col- 9. Centrifuge for 3 min in a microcentrifuge (16,000 X
ony (43). A method similar to those mentioned above has g), pour off the supernatant fluid, and invert the tube on a
been applied to lactic acid bacteria and several other groups paper towel. Then, dry the tube under vacuum.
of gram-positive organisms (5). 10. Resuspend the precipitate in 30 pl of TES. Allow the
DNA to dissolve overnight at 4C. Ordinarily, 10 pl of this
30.1.3.1. Lysis in Solution solution shows readily visible DNA bands after gel elec-
trophoresis. Note that plasmid DNA obtained by this
The following method has the advantage of using small
method should be further purified for restriction endonu-
working volumes, shares the best features of the previously
clease analysis by the following additional steps.
published methods, and permits the isolation of small as
11. Resuspend the pellet obtained in step 9 in 100 p l of
well as large plasmids. It takes advantage of the efficiency of
0.010 M Tris (pH 8.0). Add 100 pl of phenol equilibrated
plasmid DNA molecules to renature after treatment with
with 0.010 M Tris-HC1 (pH 8.0). Mix well. Add 100 pl of
alkali, which may account for the improved yields in plas-
chloroform.
mid DNA of very high molecular mass (up to 500 kbp).
12. Centrifuge for 30 s in a microcentrifuge (16,000 X g)
This method has been used for the isolation of plasmids
to separate the aqueous phase from the phenol-chloroform
from gram-negative bacteria (e.g., A. tumefaciens, Yersiniu
phase. Remove the upper, aqueous phase.
enterocolitica, Salmonella enterica serovar Typhi, Klebsiella
13. Precipitate the plasmid DNA with 2 volumes of ice-
spp., and E. coli) and also gram-positive bacteria (e.g.,
cold 95% ethanol, and proceed as in step 9. Resuspend the
Staphylococcus and Streptococcus spp.). Consequently, it is
precipitate in a suitable buffer for the restriction endonu-
recommended for the routine screening of all types of bac-
clease reactions.
teria for all types of plasmids.
Plasmid DNA obtained by this method from even 2 ml For lactic streptococci, another lysis protocol is de-
of culture is relatively free from chromosomal DNA and, scribed in reference 5.
after a few additional steps, can be used directly for restric-
tion endonuclease analyses. The general procedure is as fol-
lows. 30.1.3.2. Lysis i n Agarose Gel
The following method is based on bacterial cell lysis di-
1. Grow 2 ml of culture overnight in an appropriate rectly in the wells of a vertical agarose gel (43). Since most
medium (e.g., brain heart infusion broth) on a shaker. The of the chromosomal DNA remains intact after the lysis, it
cells may be used directly or, better, can be used to obtain remains in the well, and therefore less than 0.5% of the
logarithmically growing cells by diluting the overnight cul- total chromosomal DNA appears in the gel as a band of lin-
ture 1:20 into 2 ml of fresh medium and incubating the sub- ear DNA. Megaplasmids have been successfully detected by
culture for 2 to 3 h. Harvest the cells by centrifugation at this method.
about 2,500 X g for 10 min.
2. Resuspend the cells in 2 ml of TE buffer (Table l ) , 1. Pick cells from a colony with a flat toothpick, and re-
recentrifuge, and resuspend the cell pellet thoroughly in 40 suspend them in the well of a gel containing 15 p l of a so-
pl of TE. For marine vibrios and other halophilic bacteria, lution consisting of 7,500 U of lysozyme per ml, 0.3 U of
it is necessary to carry out this and subsequent washing steps boiled RNase per ml, 20% Ficoll 400,000, and 0.05% bro-
in the presence of high salt concentrations (up to 1 M mophenol blue in electrophoresis buffer (0.09 M Tris base,
NaC1). 0.012 M EDTA, and 0.09 M boric acid) (for gram-negative
3. Add 0.6 ml of the lysis buffer (4% SDS in TE [pH bacteria) or 75,000 U of lysozyme per ml, 0.3 U of boiled
12.41) to a 1.5-ml microcentrifuge tube, and, with a Pasteur RNase per ml, 20% Ficoll400,000,0.05 M EDTA (pH 8.0),
pipette, transfer 40 p l of the cell suspension into the lysis 0.1 M NaCl, and 0.05% bromophenol blue in electrophore-
buffer. Mix the suspension well, but avoid vigorous agita- sis buffer (for gram-positive bacteria). The gel dimensions
tion or the use of mechanical mixing devices. It is impor- recommended are 110 by 140 by 2.5 mm, and the well sizes
tant to determine the pH accurately (use a high-pH elec- should be 6.5 by 15 by 2.5 mm. Gel concentrations may
trode), since values higher than 12.5 will irreversibly vary from 0.75 to 1.2% agarose.
denature the plasmid DNA. 2. O n top of the bacterium-lysozyme mixture add 30 p l
4. Incubate the suspension at 37C for 20 min to of a solution containing 0.2% SDS and 10% Ficoll400,OOO
achieve full lysis of the cells. in electrophoresis buffer for gram-negative bacteria or 30 pl
5. Neutralize the solution by adding 30 p1 of 2.0 M Tris- of the same solution but also containing 2% SDS for gram-
HC1 (pH 7.0). Slowly invert the tube until a change in vis- positive bacteria.
cosity is noted. 3. Gently mix the two layers with a toothpick by mov-
6. Precipitate chromosomal DNA by adding 0.24 ml of ing it from side to side (not more than twice for gram-
5 M NaC1. This step should be done immediately after step negative bacteria). Complete mixing should be avoided.
5. For complete removal of the chromosomal DNA, put the The two layers must still be distinguishable.
tube in ice for 4 h. If a large number of cultures are being 4. On top add 0.1 ml of a solution containing 0.2% SDS
screened and maximum plasmid purity is not important, the and 5% Ficoll400,OOO in electrophoresis buffer without dis-
time in ice can be shortened to 1 h. turbing the viscous DNA lysate.
716 MOLECULAR GENETICS

5. Seal the wells with hot agarose, fill both electro- impede the binding of the double-stranded DNA to the
phoretic chambers with electrophoresis buffer, and proceed membrane.
with electrophoresis for 60 min at 2 mA and then for 60 to Further purification can be achieved by hydroxyapatite
150 min at 40 mA. column chromatography. Dialyze the extract obtained in
step 6 against 0.075 M sodium phosphate buffer (pH 6.8).
30.1.4. Linear Plasmids Apply this sample to a hydroxyapatite column equilibrated
Although most plasmids are composed of circular DNA, with the same buffer. After washing, elute the single-
linear plasmids have also been described. They were found stranded DNA with a linear gradient consisting of 0.075 to
in lankamicin- and lankacidin-producing Streptomyces 0.250 M phosphate buffer.
rochei as well as in Borrelia burgdorferi and B . coriaceae, the
etiological agents of Lyme disease and epizootic bovine 30.1.6. Single-Stranded Phagemids
abortion, respectively (7, 67, 106, 113). Although several techniques that used to require that the
The protocols to isolate linear plasmids involve cell lysis, cloned DNA fragment be in the form of a single-stranded
in some cases followed by CsC1-ethidium chloride gradient template have been now replaced by other, newer tech-
purification, and the separation of the linear plasmid from niques, for some purposes it may still be desirable to purify
the chromosomal DNA by either sucrose gradient or aga- single-stranded plasmid DNA. Dotto et al. (41) demon-
rose gel electrophoresis (7, 67, 106). A procedure designed strated that a plasmid carrying the major intergenic region
to isolate linear plasmids of B. coriaceae is as follows. of f l , in the presence of a helper phage, enters the f l repli-
cation pathway by generating a single-stranded DNA that
1. Pellet cells from a 20-ml culture, and wash them with
is packaged and exported in phage-like particles. The
phosphate-buffered saline containing 0.005 M MgC12.
major intergenic region of f l contains all of the sequences
2. Resuspend the pellet in 0.4 ml of a solution contain- required in cis for initiation and termination of viral DNA
ing 0.05 M Tris-HC1 (pH 8.0) and 0.05 M EDTA.
synthesis and for morphogenesis of bacteriophage particles.
3. Add 0.1 ml of 10% SDS and 25 p l of 0.02 M pro-
Phagemids are plasmid vectors that have the ColEl origin
teinase K. Incubate at 37C for 1 h.
of replication, an antibiotic resistance gene for selection, a
4. Load this mixture onto a 10 to 40% sucrose gradient multiple cloning site, and the origin of replication of one
prepared in 0.05 M Tris-HC1 (pH 8.0)-0.05 M EDTA. Run
filamentous phage. Most of these phagemids also have the
overnight at 100,000 x g in an SW41 rotor (Beckman-
T3 and/or T7 promoter to allow the synthesis of specific
Coulter Instruments, Palo Alto, CA).
RNA by using the clone as a template. The cloned DNA
5. Collect 3-drop fractions from the bottom of the gradi- can be propagated as a regular recombinant plasmid.
ent.
Single-stranded DNA is obtained in the culture super-
6. Run the fractions in 0.3% agarose gel electrophoresis, natant after the E. coli cells harboring the phagemid are
and use UV transillumination to locate those containing
coinfected with a helper phage. Different phagemids are
the linear plasmid DNA.
available from several commercial sources (Stratagene,
7. Pool these fractions, and precipitate the DNA by ad-
Bio-Rad, and others). In the following paragraphs a general
dition of 2.5 volumes of cold ethanol and centrifugation.
protocol is described for the generation of single-stranded
Wash with 70% cold ethanol and dry. Resuspend the DNA
phagemid DNA.
in TE buffer.
1. Inoculate 5 ml of 2 X YT broth with a single colony
30.1.5. Single-Stranded Plasmids of E. coli harboring a phagemid, and add 2 X lo7 PFU of
Several plasmids of gram-positive bacteria may be present as helper phage suspension (e.g., phage M13K07 [Bio-Rad,
both single- and double-stranded DNA. All these plasmids Richmond, CAI, which has a kanamycin resistance gene)
replicate via a rolling-circle mechanism, and so the single- per ml.
stranded DNA is a replicative intermediate (56). Single- 2. Incubate at 37C for 30 min with strong agitation
stranded DNA plasmids have been isolated from gram- (250 to 300 cycles/min), and then add kanamycin to a final
positive bacteria: lysozyme or lysostaphin in combination concentration of 70 pg/ml. This addition prevents growth
with Sarkosyl were used with Bacillus subtilis and Staphylo- of cells not infected with the helper phage.
coccus aureus (134), and electroporation was recently applied 3. Incubate in a rotary incubator (250 to 300
to Clostridium acetobutylicum for the same purposes (76). The cycles/min) at 37C for 18 h.
method developed by te Riele et al. (134) is as follows. 4. Transfer 1.5 ml of the culture to a microcentrifuge
tube, and pellet the cells in a microcentrifuge (16,000 X g)
1. Collect cells from a culture of optical density at 650
for 5 min at 4C.
nm (OD650) of 1, and wash with a solution containing 0.1
M EDTA (pH 8) and 0.15 M NaCl. 5. Transfer 1.2 ml of the supernatant to a new tube, add
2. Resuspend the cells in a solution containing 0.01 M 7 pg of boiled RNase, and incubate for 30 min at room tem-
perature.
EDTA (pH 8.0) and 0.15 M NaC1.
3. Add 10 mg of lysozyme per ml (for B. subtilis) or 50 pg 6. Add 240 p1 of a solution containing 20% polyethyl-
ene glycol 8000 and 15% NaCl (a solution containing 3.5
of lysostaphin per ml (for S. aureus), and incubate at 37C.
4. Add Sarkosyl to a final concentration of 1%, and in- M sodium acetate instead of NaCl may also be used) to pre-
cipitate the phage particles. Let the mixture stand in ice for
cubate at 65C for 20 min.
30 min.
5. Extract the cell lysate with phenol and chloroform.
6. Add boiled RNase to 50 pg/ml, and incubate at 37C
7. Collect the phage by centrifugation in a microcen-
for 20 min. trifuge (16,000 X g) for 1.5 min. Remove the supernatant.
Resuspend the phage pellet in 200 pl of a solution contain-
The single-stranded plasmid can be recognized by per- ing 0.3 M NaC1, 0.1 M Tris-HC1 (pH 8.0),and 0.001 M
forming Southern blot hybridization (as described in sec- EDTA. Transfer to another tube, and let stand in ice for 30
tion 30.4.4) but skipping the alkali treatment of the gel to min. Centrifuge again, and transfer the supernatant to a
30. Plasmids 717

fresh tube (this step removes insoluble products). This To obtain a higher degree of purification, perform an ex-
phage suspension can be stored at 4C for 1 week. traction with phenol followed by another extraction with
8. To isolate the phagemid DNA, extract the phage sus- phenol-chloroform-isoamylalcohol (2524: 1).
pension with 1 volume of phenol (prepared as described in
section 30.2.1). Transfer the aqueous phase to a fresh tube. 30.2.2. Equilibrium Gradient Centrifugation
9. Extract once with 1 volume of phenol-chloroform- Equilibrium gradient centrifugation is a classical method to
isoamyl alcohol (25:24:1), recover the aqueous phase, and purify plasmid DNA from large preparations. Ethidium bro-
reextract several times with chloroform-isoamyl alcohol mide is one in a series of phenathridinium dyes that bind to
(24:l) until no interphase is visible. DNA and RNA and inhibit nucleic acid functions (29,65).
10. To the aqueous phase add 1/10 volume of 7.8 M am- The DNA is sedimented to equilibrium by high-speed cen-
monium acetate and 2.5 volumes of cold ethanol. Keep at trifugation. Ethidium bromide intercalates into the DNA,
- 70C for 30 min. thus reducing its density (27, 108). A linear DNA molecule
11. Recover the phagemid DNA by centrifugation, wash or an open-circular (OC) (nicked) molecule of plasmid
the pellet with 70% cold ethanol, dry, and resuspend in 20 DNA does not have the physical constraints that are im-
pl of TE buffer. posed on a CCC molecule of plasmid DNA. Consequently,
the former types of DNA can bind significantly more ethid-
ium bromide molecules and are rendered less dense than the
30.2. PURIFICATION latter type of DNA. This difference in binding permits the
The subsequent analysis of plasmid DNA determines the separation of the different forms of plasmid DNA. OC and
degree of purity required. For example, to analyze and com- linear plasmid DNA that may form as a result of handling
pare plasmid profiles for epidemiological purposes, further during the preparation, as well as linear chromosomal DNA
purification is normally not required. Conversely, other (also formed during handling), will all appear in the same
purposes such as restriction endonuclease analysis, cloning, band unless the G + C contents of plasmid and chromosome
and probe preparation may require further purification. are drastically different. Only CCC plasmid DNA and some
Furthermore, a higher degree of purification is required for replicative intermediates will be separated from the linear
nucleotide sequencing in which all linear or nicked DNA and OC DNA molecules. In gradients of cesium chloride,
molecules as well as RNA must be eliminated. After purifi- these forms can be visualized as discrete bands when the
cation, the plasmid DNA concentration can be determined gradient is illuminated with UV light, as a result of the flu-
by either classical methods such as spectrophotometric de- orescence of the ethidium bromide intercalated in the
termination or ethidium bromide fluorescence or commer- DNA.
cial methods such as colorimetric quantitation with
1. To a polyallomer ultracentrifuge tube (5/8 by 3 in. [1.6
Dipstick (Invitrogen, Carlsbad, CA). This is a rapid
by 7.6 cm]), add about 5 g of cesium chloride, 2 ml of TES
method that consists of spotting a small amount of the nu-
buffer, and 3.2 ml of the DNA solution.
cleic acid solution on the Dipstick, dipping the stick in a so-
2. Mix the contents of the tube until the cesium chloride
lution provided with the kit, and determining the concen-
is in solution, and add 0.2 ml of ethidium bromide solution
tration by matching the color intensity of the sample to a
(10 mg/ml in TES buffer). Add the ethidium bromide after
standard chart.
the CsC1, since the relaxation protein present in some plas-
30.2.1. Extraction with Organic Solvents mids (which leads to nicking of the DNA) can be activated
by ethidium bromide (82). The high CsCl concentration (7
Extraction with organic solvents is used to remove proteins
that are normally present after plasmid DNA isolation or
M) inactivates the relaxation complex of most plasmids
(82), and consequently higher yields of DNA can be ob-
after boiled RNase or restriction endonuclease treatment.
tained. Even at a high salt concentration, visible light (in
1. Mix the plasmid DNA solution with 1 volume of phe- the presence of ethidium bromide) can induce nicking of
nol (Table 1) or phenol-chloroform-isoamylalcohol (2524: plasmid DNA. Therefore, all operations must be conducted
1, vol/vol/vol) until emulsified. under indirect illumination.
2. Separate the phases by centrifugation in a microcen- 3. Adjust the refractive index of the solution to 1.3925
trifuge (16,000 X g) for 10 s. &0.001 g/ml by adding TES buffer or solid cesium chloride.
3. Transfer the aqueous upper phase to a fresh tube, add The refractive index is a suggested value and may vary for
1 volume of either chloroform-isoamyl alcohol (24:l) or each instrument; it must be standardized by doing trial ex-
water-saturated ether, and mix until the aqueous phase be- periments. If necessary, top the tubes off with mineral oil.
comes translucent. Warning: ether is very flammable and Rule of thumb: To the dried DNA pellet resuspended in 4.0
tends to form explosive peroxides under the influence of air ml of TE buffer, add 4.5 g of CsCl and 0.1 ml of 10 mg/ml
and light. ethidium bromide. Under these conditions, good separation
4. Separate the phases by centrifugation as in step 2, and of the bands is usually obtained.
recover the aqueous phase containing the plasmid DNA. 4. Prepare the tubes for ultracentrifugation as specified
With chloroform extraction, the aqueous phase is on top; by the centrifuge manufacturer, and place them in the ap-
with ether extraction, the aqueous phase is below. propriate rotor. Fixed-angle, vertical, swinging bucket or
5. Add 1/9 volume of 5 M potassium acetate (pH 4.8) near-vertical rotors can be used. However, in vertical and
and 2.5 volumes of cold ethanol. Precipitation can also be near-vertical rotors the equilibrium is reached faster.
achieved by addition of 0.5volume of 7.5 M ammonium ac- 5. Centrifuge the tubes at 180,000 X g at 15C for 4 to
etate and 3 volumes of ethanol. 8 h in a vertical or near-vertical rotor or 12 to 16 h in a
6. Pellet the DNA by centrifugation in a microcentrifuge fixed+angle rotor. After centrifugation, examine the gradi-
(16,000 x g) for 5 min, wash the pellet with 70% cold ents by illuminating the tubes with UV light and looking
ethanol, and dry. for the presence of fluorescent bands due to the DNA-
7. Resuspend the DNA in TE buffer. ethidium bromide complex. Two bands should be observed,
718 MOLECULAR GENETICS

the upper one corresponding to chromosomal and linear or ity for proteins but low affinity for nucleic acid at neutral
nicked plasmid DNA, and the lower band corresponding to pH, such as Strataclean Resin (Stratagene, La Jolla, CA).
CCC plasmid DNA. In some cases only the lower band is Quick minicolumn chromatography products are based on
observed. Collect the plasmid DNA by puncturing the tube ion exchange or adsorption. Some of these columns are pre-
at the bottom or just below the plasmid DNA band with a packed in small devices, such as pipette tips or small columns
needle. that can be centrifuged for easier elution, and are compo-
6. Free the DNA of ethidium bromide by extracting the nents of DNA purification kits. Commercial kits for plasmid
preparation at least three times with cesium chloride- DNA purification are available from several companies.
saturated isopropanol. Recently, plasmid DNA isolation and purification has
7. Dialyze the DNA against TE buffer for 2 to 3 h to become almost routine in most clinical and research labo-
eliminate the cesium chloride. Alternatively, the cesium ratories. Therefore, several companies have developed
chloride can be eliminated by twofold dilution of the DNA products to make nucleic acid isolation and purification
solution with TE buffer and precipitation with 2.5 volumes easier and faster. Most of these products are basically com-
of ethanol. binations of alkaline or boiling lysis methods followed by a
purification step such as those described above. Some ex-
30.2.3. Precipitation with Polyethylene Glycol amples of these kits are Prep-A-Gene (Bio-Rad), Wizard
DNA purification system (Promega), PlasmidQuick
1. Add 1 volume of 5 M LiCl (to precipitate high- (Stratagene), Miniprep Kit Plus (Pharmacia), and QIAGEN
molecular-weight RNA) to the DNA solution. Mix, and plasmid purification systems, including kits for the isolation
centrifuge at 12,000 X g at 4C for 10 min. of plasmids containing large genomic inserts such as those
2. Transfer the supernatant to a fresh tube, and add 1 generated using BAC, PAC, and P1 cloning vectors. In ad-
volume of isopropanol. Mix and centrifuge as in step 1 but dition, most of these systems have been adapted for the iso-
at room temperature. lation of plasmid DNA from a large number of samples such
3. Discard the supernatant, wash the pellet with cold as those processed by high-throughput systems involved in
70% ethanol, and allow it to dry. genome sequencing projects. The Concert96 lysis and pu-
4. Dissolve the DNA pellet in 0.5 ml of TE buffer con- rification system was developed recently by Invitrogen, in
taining boiled RNase (20 pglml). Let stand for 30 min at which the bacterial cells of up to 96 samples are collected
room temperature. and lysed directly onto the matrix used for the subsequent
5. Add 0.5 ml of a solution containing 1.6 M NaCl and purification of plasmid DNA. Some of these kits work with
13% polyethylene glycol 6000. Mix. both gram-positive and gram-negative organisms.
6. Collect the plasmid DNA by centrifugation in a mi-
crocentrifuge (16,000 x g) for 5 min, and discard the su-
pernatant.
7. Dissolve the DNA pellet in 20 p l of TE buffer, extract 30.3. QUANTl F ICATl ON
with phenol, and precipitate the DNA as described above. 30.3.1. Quantification of Plasmid Concentration
30.2.4. Chromatography 30.3.1 . I . Spectrophotometric Methods
Several authors have described the application of high- Absorbance at 260 nm is a widely used method to deter-
pressure liquid chromatography and fast protein liquid chro- mine the concentration of nucleic acids in aqueous solu-
matography to the purification of plasmid DNA (92, 155). tions ( I 14). Although very practical and nondestructive,
These methods can be fast and convenient, although they the drawbacks of this method include the requirement of
require special equipment. pure nucleic acid preparations, the relatively narrow range
at which reliable concentrations can be determined, its rel-
30.2.5. Commercial Methods atively low sensitivity, and the effect of pH and ionic
Many companies have introduced products for nucleic acid strength on the absorbance of UV light by the purines and
purification that are rapid and nontoxic alternatives to pyrimidines. A general practice when using this quantita-
organic-solvent extraction. These products can be used to tive method is the determination of the absorbance of the
purify and concentrate DNA preparations, to extract-purify sample at different wavelengths to determine the purity of
DNA fragments from agarose gels, and to separate labeled the sample being tested. For example, contamination with
DNA from unincorporated nucleotides. The methods con- phenolate and thiocyanate compounds, which are generally
sist basically of the adsorption of the DNA to glass or silica used for the isolation of nucleic acids, increases significantly
powder or of minicolumns for adsorption or ion-exchange the absorption at 230 nm, while absorption at 330 nm and
chromatography. higher wavelengths indicates contamination with particu-
The principle of glass or silica matrix methods is the se- late matter. The contamination of DNA samples with pro-
lective binding of DNA to these matrixes in the presence of teins can be tested by determining the OD260:OD280 ratio,
sodium iodide. The contaminants (RNA, proteins, salts, although it should be kept in mind that this ratio is a better
agarose, and polysaccharides normally present in agarose) indication of contamination of protein samples with nu-
are eliminated by washing steps. Finally the DNA is recov- cleic acids rather than the reverse. In general, the initial
ered from the matrix by elution in low-ionic-strength buf- data are obtained as the A260 or OD260 units of appropriate
fer. A few examples of these commercial products are dilutions of DNA or RNA samples. This information is
GENECLEAN kits (QOBIOgene), which are used to purify then transformed into relative concentration of nucleic
DNA fragments of 200 bp or larger; MERmaid kits acids by considering that 1 OD260unit is equal to 50 pg/ml
(Q*BIOgene), which are used to purify oligonucleotides of of double-stranded DNA or 38 pg/ml of single-stranded
10 to 200 nucleotides; or QIAquick DNA cleanup system DNA or RNA. The newer spectrophotometers, e.g.,
(QIAGEN, Valencia, CA). A n alternative method is based NanoDrop, obtain concentration and quality data by direct
on the use of a hydroxylated silica resin that has high affin- measurement of samples as small as 1 ~ 1 .
30. Plasmids 719

30.3.1.2. Fluorometric Methods 3. Add to each standard and sample spots 1 volume of
TE buffer containing 2 pg/ml ethidium bromide and mix
30.3.1.2.1. Nonintercalating Dyes either by pipetting up and down with a micropipette or
The concentration of DNA can be determined using mixing with a pipette tip.
some other approaches that are as simple as direct spec- 4. Turn on the UV transilluminator and determine the
trophotometry but more sensitive. One of these methods is fluorescence of the samples and standard dilutions immedi-
based on the binding of nonintercalating dyes such as ately either by direct visual inspection or by taking a pho-
Hoechst 33258 by double-stranded DNA larger than 1 kbp, tograph, using an appropriate gel documentation system
which results in a 60-fold increase in fluorescence at 458 equipped for DNA detection.
nm when the sample is excited at 365 nm. It is important to 5. Estimate the plasmid DNA concentration of the sam-
consider that this assay is also affected by the pH of the sam- ples by comparing their fluorescence intensity with that of
ple, the presence of detergents and salts above 3 M, and the the standard dilutions.
quenching of the fluorescence of the sample by excess of
dye in the final reaction mixture. The Agarose Slab Method
1. Dilute 50 p l of Hoechst 33258 stock solution (0.2 1. Prepare the samples and standard dilutions as de-
mg/ml in H20) in 100 ml of fluorometry buffer (2M NaC1, scribed for the Saran Wrap method.
50 mM NaP04, pH 7.4). Solutions should be filtered 2. Prepare a 1% agarose slab by pipetting 2 to 3 ml of 1%
through a 0.45-pm-pore-size filter, and the Hoechst 33258 agarose in TE buffer containing 0.5 pg/ml ethidium bro-
stock solution should be kept protected from the light. mide on a flat surface such as that of a microscope slide.
2. Transfer 3 ml of diluted dye to an appropriate number 3 . Spot the samples and standard dilutions on the surface
of glass tubes. of the agarose after it has solidified and incubate at room
3. Construct a standard curve using DNA of known con- temperature from 1 to 2 h.
centration (100 to 500 ng). 4. Transfer the gel slab onto the surface of a UV transil-
4. Mix and read the fluorescence of the standards imme- luminator carefully, turn it on, and photograph the gel im-
diately in a prewarmed fluorometer set to 365 nm and 458 mediately using an appropriate gel documentation system
nm excitation and emission wavelengths, respectively. equipped for DNA detection.
5. Add 0.1, 1.0, and 10 p l of the DNA sample being 5. Estimate the concentration of the samples by compar-
tested to individual tubes containing the diluted Hoechst ing their fluorescence intensity with that of the standard
33258 dye solution. Mix and read immediately as described dilutions.
for the standard samples.
6. Determine the concentration of DNA in the sample The Agarose Minigel Method
by using a curve constructed by plotting the fluorescence of This method is as rapid as those described in the two previ-
each standard point against its corresponding DNA con- ous sections with the advantage of providing information
centration. regarding the presence of different plasmid topoisomers as
well as the potential contamination with RNA.
30.3.1.2.2. Intercalating Dyes 1. Mix 2 to 5 p l of plasmid DNA sample with 2 pl of gel-
The UV-induced fluorescence emitted by ethidium bro- loading buffer on the surface of a piece of Parafilm or Saran
mide when intercalated with DNA is a practical and widely Wrap.
used alternative to determine as little as 1 ng of double- 2. Mix 2 to 5 p l of each of a twofold dilution series of
stranded DNA. In general, the most popular methods are plasmid DNA standard, covering a concentration range
only semiquantitative because they are based on the visual from 0 to 50 pg/ml, with 2 p1 of gel-loading buffer on the
comparison of the fluorescence produced by the test sam- surface of a piece of Parafilm or Saran Wrap. Purified plas-
ples with that produced by dilutions of standard DNA, mids similar in size to those being tested should be used for
which are all spotted on the surface of UV-transparent ma- this purpose.
terial (Saran Wrap). Alternatively, the samples and stan- 3 . Prepare a 0.8% mini-agarose gel (20 to 30 ml) con-
dard dilutions can be either spotted onto an agarose slab or taining 0.5 pg/ml ethidium bromide.
electrophoretically fractionated through agarose contain- 4. Load the plasmid samples and the standards and run
ing 0.5pg/ml ethidium bromide. These approaches avoid the electrophoresis using standard conditions until the bro-
interference due to contaminants. New dyes such as Pic0 mophenol blue present in the gel-loading buffer reaches
Green and SYBR Green I (Molecular Probes, Eugene, OR) one-third to one-half of the gel length.
are now being used to quantify DNA because of their low in- 5. Destain the gel by immersing it in 50 to 100 ml of dis-
trinsic fluorescence and other properties that make them tilled water or 0.01 M MgClz for 5 min at room tempera-
more suitable for DNA quantification (16, 126). Affordable ture. Skip this step if the gel background is low and does not
fluorometers are now on the market, e.g., Qubit (Invitrogen). interfere with the quantification of plasmid DNA.
Recently, a sensitive method (the microplate method) that 6. Photograph the gel immediately using an appropriate
allows the rapid and practical quantification of a large num- gel documentation system equipped for DNA detection.
ber of samples, such as those produced in high-throughput 7. Estimate the concentration of the samples by compar-
processes, was developed using SYBR Green I (146). ing their fluorescence intensity with that of the standard
dilutions.
The Saran Wrap Method
The Microplate Method
1. Stretch a piece of Saran Wrap over a UV transillumi-
nator and spot 5 p l of standard DNA serially diluted cover- 1. Dilute the dye SYBR Green I 6,250-fold in 10 mM
ing a concentration range from 0.1 to 20 pg/ml. Tris-HC1 (pH 7.5)-1 mM EDTA.
2. Spot 1 to 5 p1 of sample DNA close to the standard 2. Pipette up to 250 p1 of diluted dye into each well of a
spots. standard microtiter plate.
Next Page

720 MOLECULAR GENETICS

3. Prepare a serial dilution of standard DNA covering a 3. Irradiate an aliquot of the purified plasmid for 30 min
final concentration range from 2 ng/ml to 2 pg/ml of DNA. at room temperature with UV light (254 nm, 15 W) to cre-
4. Add 1 to 5 p1 of standard dilutions and plasmid sam- ate the OC (nicked) forms of plasmid monomers and mul-
ples to each well containing diluted SYBR Green I. timers.
5. Mix by tapping on the side of the microplate gently, 4. Mix each of the samples (up to 500 ng of DNA) with
and incubate at room temperature no longer than 15 min. gel-loading buffer.
6. Illuminate the samples by using a 365-nm UV transil- 5. Prepare a 0.8% medium-size agarose gel using the Tris-
luminator and capture the image with a CCD camera acetate buffer system.
(BioPrint gel documentation system; LTF-Labortechnik, 6. Load the plasmid samples and run the electrophoresis
Wasserburg, Germany) with a standard yellow filter (485 at 5 V/cm (50 V) until the bromophenol blue present in the
nm) using an aperture of 1.8. The plate should be located gel-loading buffer reaches the bottom of the gel.
with minimal proximity to the camera objective, and the 7. Stain the DNA samples with 1 pg/ml ethidium bro-
exposure time should be adjusted to obtain maximal sensi- mide for 30 min and record the gel image using an appro-
tivity. priate gel documentation system equipped for DNA detec-
7. Calculate the fluorescence intensity of each sample by tion.
determining their pixel density with a software such as the 8. Comparative analysis of the electrophoretic mobility
Image Tool 2.00 (University of Texas Health Science of the DNA bands detected in each sample provides infor-
Center, San Antonio, TX). mation regarding the plasmid forms present in each sample.
8. Estimate the concentration of the plasmid samples by Under the conditions described, the monomeric CCC form
comparing their fluorescence intensity with that of the is the fastest-migrating plasmid species, which is followed by
standard dilutions. the plasmid monomers in their nicked and linear forms, re-
spectively. The CCC, nicked, and linear forms of plasmid
30.3.2. Quantification of Plasmid Forms dimers migrate immediately above the monomers, with the
Plasmids can be isolated in different forms such as circular CCC dimer being the fastest species of this second group of
elements with different degrees of coiling, partially cleaved, plasmid forms.
as well as linear molecules. In addition, the extrachromoso- 9. Comparative analysis of the relative fluorescence of
ma1 elements can exist as monomers and multimers that each plasmid species present in the untreated sample pro-
can be present either as concatemer or catenete complexes. vides information regarding the relative proportion of each
Recently, a fluorescence-based method was developed to species in the plasmid preparation.
determine the presence of supercoiled DNA in plasmid 30.3.2.2. Capillary Gel Electrophoresis
preparations (87).This method is based on the fluorescence
enhancement of PicoGreen bound specifically to double- 1. Equip a capillary electrophoresis system such as a
stranded DNA and the fact that supercoiled DNA regains its Beckman P/ACE 2050 CE (Beckman) instrument with an
original double-stranded structure after denaturation with argon laser and a 37-cm-long coated capillary (100-pm
chemical and physical agents such as high temperature. In inner diameter, 0.1-pm coating thickness) (DB-17; J. and
contrast, OC and linear DNA molecules are irreversibly de- W. Scientific, Folsom, CA). Use 488 and 520 nm as the ex-
natured and remain as single-stranded DNA after these citation and detection wavelengths, respectively.
treatments, resulting in the minimal fluorescence of these 2. Prepare the running buffer, consisting of 89 mM Tris,
DNA forms because of the very limited binding of 89 mM boric acid, 2 mM EDTA, pH 8.4, and 0.1% (wtlwt)
PicoGreen to single-stranded DNA. Although sensitive hydroxypropylmethylcellulose (H-7509; Sigma Chemical
and practical for the rapid analysis of a large number of sam- Co., St. Louis, MO).
ples, this method does not provide information regarding 3. Add 1 ~1 of YOYO stain (Molecular Probes) to 15 ml
the presence of the different topoisomers and their relative of running buffer immediately before sample analysis.
concentration in a particular plasmid DNA preparation. 4. Stain the samples prepared as described for agarose gel
Most of these forms can be detected and quantified by sim- electrophoresis by incubating them at room temperature for
ple methods such as agarose gel electrophoresis and ethid- 2 min with YOYO at a DNA base pair-to-dye molar ratio
ium bromide staining. A similar analysis can also be con- of 5 1 .
ducted using capillary gel electrophoresis, which provides 5. Apply samples to the capillary column by pressure in-
more reliable and quantitative data and can be automated jection for 1 s and perform electrophoresis at 100 V/cm and
to examine a large number of samples. A comparative 30C.
analysis of these two methods was reported by Schmidt et 6. Collect the data as suggested by the instrument man-
al. (116), who concluded that capillary gel electrophoresis ufacturer and analyze them using a package such as the
is an appropriate alternative for the detection and quantifi- System Gold Software (Beckman), recording migration
cation of plasmid forms. time and peak area for each peak detected in the analyzed
samples.
30.3.2.1. Agarose Gel Electrophoresis 7. Quantify each plasmid form present in a particular
sample by determining the peak area:migration time ratio
1. Isolate plasmid DNA by a standard method. In gen- for each peak detected during the electrophoresis of a plas-
eral the preparation is composed mostly of monomers and mid sample.
dimers in their supercoiled or CCC forms.
2. Incubate an aliquot of the purified plasmid either with
excess or limiting amounts of a restriction endonuclease, 30.4. CHARACTERIZATION
which has a single recognition site within the plasmid mol- Physical characterization of plasmid DNA is covered in sec-
ecule, to produce monomers and multimers in their cognate tions 30.4.1 through 30.4.4. For details of genetic and func-
linear forms. tional characterization, see sections 30.4.5 through 30.4.8.
Gene Transfer in Gram-Negative Bacteriat
JOSEPH E. PETERS

31.1. INTEGRATING DNA INTO THE CHROMOSOME ............... 736


3 1.1.1. Homology-Based RecA Systems .......................... 736
3 1.1.1.1. Single-Crossover Systems........................ 736
. ..... 737
2. Double-Crossover Systems with Circular Substrates
. ...... 739
3. Double-Crossover Systems with Linear Substrates
3 1.1.2. Site-Specific Recombination Systems Can Remove Selectable
Markers ........................................... 739
.
3. Recombination Systems from Bacteriophages ................. 739
3 1.1.3.1. Other Applications with Bacteriophage Recombination
Systems .................................... 742
3 1.1.4. Introducing DNA at Specific Sites in the Chromosome .......... 742
3 1.2. INTRODUCING DNA ON PLASMIDS ......................... 743
.3. GENERALIZED TRANSDUCTION ........................... 744
.
31.3.1. Generalized Transduction in E coli with Bacteriophage P1 ....... 744
31.3.1.1. Protocol for Determining Plaque-Forming Units of P1 ... 744
.
2. Procedure for P1 Growth from Plaques .............. 744
.
3. Procedure for P1 Growth on Donor Strains ........... 745
.
4. Procedure for P1 Transduction .................... 745
31.3.3. Generalized Transduction in Other Systems .................. 745
.
4. Applications for Generalized Transduction ................... 745
3 1.4. TRANSFORMATION ...................................... 746
31.4.1. Chemical Transformation ............................... 746
31.4.1.1. Procedure for Calcium Chloride-Competent E coli . ..... 746
. .
2. Procedure for Competent E coli with the Inoue
Method ..................................... 746
.
3. Procedure for Rubidium Chloride-Competent E coli . .... 747
31.4.2. Electroporation ...................................... 747
3 1.4.2.1. Procedure for Electroporation ..................... 747
.
.2 Adaptations for Electroporation ...................748
3 1.4.3. Natural Competency .................................. 748
31.4.3.1. Procedure for Chromosomal DNA Isolation .......... 748
......... 748
31.4.4. Controls and Precautions for Transformation Strategies
31.5. CONJUGATION .......................................... 749
.6. BARRIERS T O DNA TRANSFER ............................. 749
. .....................
3 1.6.1. Restriction of Foreign DNA by E coli 749
. .
2. Barriers That Inhibit Moving DNA from E coli into Other
............................................ 749
Bacteria
3 1.7. BACTERIAL STRAINS ..................................... 750
. ......................... 750
3 1.7.1. Basics of E coli Strain Genotypes
. ............................ 750
2. How To Get Bacterial Strains
31.8. MEDIA ................................................. 750
31.8.1. LB Medium ......................................... 750
.
2. SOB Medium ........................................ 750
.
3. SOC Medium ....................................... 750
.
4. Psi Medium ......................................... 750
.
5. M9 Medium ........................................ 750
.
6. M63 Medium ........................................ 751
.
7. Supplements ........................................ 751
.
8. Preparation of Agar Media .............................. 751
. .............
9. Tetracycline.Sensitive.Selective (TSS) Agar Plates 751
31.9. BUFFERS ................................................ 751
3 1.9.1. Inoue Transformation Buffer ............................ 751

t A chapter by the same title was authored by D. L. Provence and Roy Curtis 111 for Methods for General and Molecular
Bacteriology . This chapter has been completely revised and reorganized by the current author.

735
736 MOLECULAR GENETICS

.2.TfbI ............................................ 751


.3.TfbII ........................................... 751
3 1.10. FINAL CONSIDERATIONS ................................ 752
.11. REFERENCES ........................................... 752
3 1.11.1. General References ................................. 752
.2. Specific References ................................. 752

One realization that has come from comparing multiple ification of chromosomes. Most of these strategies can also
bacterial genome sequences, including multiple isolates be applied to large low-copy-number plasmids, such as the
from the same species, is that gene transfer is an important Fertility or F plasmid that has been adapted to make bacte-
force in bacterial genome evolution. In the laboratory gene rial artificial chromosomes (BACs). Early recombination
transfer is essential for the study of bacteria and for learning methods used the host-encoded recombination system as a
more about all living organisms. tool for engineering the chromosome. These strategies can
Three processes in bacteria can broadly define the trans- still be useful in many bacteria for generating recombinants
fer of DNA: transformation, transduction, and conjugation. in wild-type bacteria via single-crossover and double-
In each transfer, genetic information from one bacterium, crossover events from circular and linear DNA molecules. In
the donor, is acquired by another bacterium, the recipient. E . coli and a number of other bacteria, mutations in various
Generally an organism which has had its genetic content host recombination functions are required to allow chromo-
modified is called a recombinant organism, but specific some modification with linear substrates. Site-specific re-
terms are also used depending on how DNA is introduced. combination systems can be utilized for introducing DNA
Transformation is a gene transfer process in which DNA into the chromosome via circular DNA molecules. The
from the donor is taken up directly by the recipient and greatest recent advance in bacterial genetics is the use of
incorporated into its genome. A recipient cell that now bacteriophage-encoded recombination systems like X Red-
expresses a donor genetic trait is called a transformant. mediated recombination that have been adapted as impor-
Transduction is a gene transfer process in which a bacterial tant laboratory tools.
virus propagating in the donor picks up some of its genetic
information and, on infection of the recipient, causes a her- 31.1 .l.Homology-Based RecA Systems
itable change in the recipient. A recipient cell that acquires The major bacterial recombination protein RecA is con-
a donor trait by this process is called a transductant. served in bacteria and plays an important role in DNA re-
Conjugation is a gene transfer process in which the so- pair (72, 73). RecA-mediated recombination was the origi-
called donor bacterium makes physical contact with, and nal source for manipulating bacterial genomes and provided
transfers genetic material to, the recipient. A recipient that the original evidence for sexual recombination in bacteria
acquires donor genetic information is called a transconju- (77). Work in E. coli indicates that RecA cannot act on its
gant. These three processes have been central in allowing own in recombination. A variety of accessory proteins are
bacterial geneticists to analyze the genetic and biochemical required for RecA to function by tailoring DNA substrates
basis of bacterial functions. In turn they have led to the es- and promoting the binding of RecA to a single-stranded
tablishment of principles of gene structure, function, and DNA in competition with the single-stranded DNA bind-
regulation as well as the dissection of more complex ing protein. The RecBCD machine allows RecA-mediated
processes of macromolecular synthesis, growth, and cell recombination to initiate at broken double-strand ends by
division. generating a 3' single-strand end and loading RecA (6).
This chapter focuses on the many genetic tools available The RecBCD machine is a potent nuclease, and recombi-
to manipulate the genetic content of Eschen'chiu coli. E . coli nation with linear DNA substrates in E. coli and many
remains important as a model system and as a workhorse for other bacteria is essentially impossible without inactivation
manipulating DNA prior to introduction into other organ- of its nuclease activity. Another RecA-dependent system,
isms. In addition to E . coli, references are made to a variety called the RecF system, requires a larger set of recombina-
of other proteobacteria (Table 1). It would be impossible to tion proteins and is predominantly involved in promoting
definitively cover gene transfer in all gram-negative organ- recombination at single-strand interruptions. Once RecA is
isms. However, an attempt is made to cover the basic ideas loaded onto a single-strand DNA substrate, it mediates re-
in gene transfer in organisms other than E. coli where there combination by searching for homology and carrying out
have been successes in a variety of proteobacteria and in strand invasion and exchange. Other recombination pro-
cyanobacteria. Plasmids are the topic of another chapter in teins are required for processing recombination intermedi-
this book and are mentioned only briefly in this chapter. ates, as well as DNA replication proteins. Bacteriophage re-
combination proteins can generate recombinant molecules
in the absence of RecA and are dealt with later.
31 .l.INTEGRATING DNA INTO THE
CHROMOSOME 31 .I .I . I . Single-Crossover Systems
After genetic information is introduced into a cell, it must Circular DNA substrates can be integrated into the bacter-
be replicated to create a recombinant bacterium. A DNA ial chromosome in a RecA-dependent process as a tool for
.molecule that does not have its own origin of replication manipulating the bacterial chromosome (104). This strat-
must integrate into either the host chromosome or another egy is based on selecting for a gene, such as antibiotic re-
autonomously replicating element such as an endogenous sistance, that is carried on a circular DNA substrate in a
plasmid. Therefore, one cannot really talk about gene trans- strain background where the DNA element cannot repli-
fer without describing how the genetic information will be cate autonomously (54,95) (Fig. 1). These experiments uti-
maintained after it is introduced. lize a conditional plasmid that cannot replicate in the given
DNA recombination systems encoded by bacteria and host naturally or because it lacks an essential muns-acting
bacteriophages provide important tools for the in vivo mod- protein (70, 89) or because it is temperature sensitive for
31. Gene Transfer in Gram-Negative Bacteria 737

TABLE 1 Methods for transferring DNA in selected species within the phylum proteobacteria
Method of DNA Narrow-host- Broad-host- Moving markers
Organism Subdivision introductiona range vectorsb range vectors' between strains Reference(s)
Agrobacterium Alpha No Yes None 24
tumefaciens
Caulobacter Alpha No Yes Transduction 13,46
crescentus
Rhodobac ter Alpha No Yes None 45
sphaeroides
Sinorhizobium Alpha No Yes Transduction 47,50, 115
meliloti
Neisseria Beta No Yes Natural transformation 51, 111, 123
gonorrhoeae
Helicobacter Epsilon No No Natural transformation
pylori
Escherichia coli Gamma Yes Yes Transduction 90
Salmonella Gamma Yes Yes Transduction
enterica
Vibrio cholerae Gamma Yes Yes Transduction 57, 116
Vibrio harveyi Gamma Yes Yes None 116
Pseudomonas Gamma No Yes Transduction 20, 106
aeruginosa
Pseudomonas Gamma No Yes None 106
syringae
Legionella Gamma No Yes None
pneumophila
Yersinia Gamma Yes Yes None 33
enterocolitica
Haemophilus Gamma Yes Yes Natural transformation 11
influenzae
Geobacter Delta No Yes None 35
sulfurreducens
'Mechanisms of introduction are abbreviated as follows (in order of preference): E, electroporation; M, mating via conjugation; N, natural competency; and C,
chemical competency.
'Representatives from the narrow-host-range plasmids are reported to replicate in this species (Table 3 ) .
'Representatives from the broad-host-range plasmids are reported to replicate in this species (Table 3).

DNA replication (54). This strategy can be used to inacti- vided in the vector. A marker in the vector backbone can be
vate genes as well as to introduce gene fusion to promoters screened to determine if the plasmid is lost (for example, by
and proteins (120). Promoter fusions can be very useful to screening for loss of a marker at positions 3 to 5 in Fig. 2). It
assay gene regulation. Despite the ease of this type of con- may sometimes be sufficient to introduce a selective marker
struction, it also has several limitations; these constructs that replaces a large portion of the gene (for example, by se-
may not be genetically stable, and the integrated DNA can lecting a marker cloned at position 1 or 2 in Fig. 2). However,
be polar on downstream genes. In addition, promoters in- this is not always an option because it can alter the expres-
cluded in the integrated DNA segment can activate down- sion of downstream genes in an operon. Therefore, integra-
stream genes. tion of a mutant allele with an in-frame deletion is often
more appropriate to eliminate the possibility of a polar effect
31 .I .I .2. Double-Crossover Systems with Circular on the transcription of downstream genes. Additionally, a
Substrates very specific mutation may be required (e.g., a point muta-
Circular DNA substrates can be used to isolate constructs tion conferring a specific amino acid change or a conditional
where the allele replacement has taken place via a double- mutation such as an amber suppressible mutation). In cases
crossover event (Fig. 2). In theory this procedure is similar to such as these one must have an alternative strategy to iden-
the single-crossover event, but only the new allele is left be- tify the recombinant organisms, for example, a phenotype or
hind in the chromosome and not the entire plasmid. These through the use of DNA sequencing.
experiments utilize conditional or temperature-sensitive rep- One potent limitation in procedures where circular sub-
licons as described above. Presumably a single-crossover strates are used to isolate recombinants by double-crossover
event is an intermediate in all double-crossover events. The events is that single-crossover events that allow integration
second crossover event that allows the plasmid to be lost can of the entire circular substrate often must be sorted away
have one of two outcomes; the plasmid is excised from the from the desired replacement event. Therefore, a counter-
replicon and leaves either the original allele or the allele pro- selection or negative selection may be required to select for
738 w MOLECULAR GENETICS

2 4
3Ql 35-

b. P
b. JkL
......................
-+I
c. i)
1 2 3
FIGURE 1 Genetic information can be integrated into the
i , ____-______________
* 1
chromosome of a bacterium using a single crossover from a cir-
C.
cular DNA construct that cannot replicate in the host. (a) A
1 2
single open reading frame (thick line) to be inactivated is FIGURE 2 Genetic information can be integrated into the
shown with its promoter (solid arrow) and RNA transcript chromosome of a bacterium using a double-crossover event
(dashed line with arrow). (b) A plasmid (circle) that cannot from a circular DNA construct that cannot replicate in the
replicate in the strain can be maintained only by integrating host. (a) A single open reading frame (thick line) to be inacti-
into the chromosome using homology provided on the plasmid vated is shown with its promoter (solid arrow) and RNA tran-
indicated with an X. Three arbitrary positions are indicated script (dashed line with arrow). (b) A plasmid (oval) that can-
with numbers to show the orientation. (c) Integration of the not replicate in the strain can be maintained only by
circular DNA substrate disrupts and fuses the plasmid-borne integrating into the chromosome using the homology provided
genes behind the target gene promoter. Depending on the con- on the plasmid indicated with Xs (thick lines). Two
struct, the transcript may be terminated within the integrated crossover events ensure that only a portion of the circular
DNA segment. Promoters within the plasmid could also acti- DNA is integrated into the gene. Five arbitrary positions are
vate adjacent genes in the chromosome. indicated with numbers to show the DNA that is integrated
and the orientation. A single crossover event likely occurs at a
mid-step in the reaction and is not shown (Fig. 1). (c) Inte-
gration of the circular DNA substrate removes all or a portion
loss of the integrating DNA element if the frequency of loss of the gene and fuses the encoded genes behind the target gene
is very low (79). One tool for negative selection capitalizes promoter. Depending on the construct, the transcript may
on the hypersensitivity of elements encoding the tetAR be terminated within the integrated DNA segment. Promo-
genes, like TnlO, to lipophilic chelating agents such as ters within the plasmid could activate adjacent genes in the
fusaric acid (17). Tetracycline-sensitive clones can be se- chromosome.
lected from an otherwise tetracycline-resistant population
by streaking the strain on tetracycline-sensitive-selective
(TSS) plates and incubating 1 to 2 days at 37C. The sacB the chromosome by inducing double-strand breaks at a spe-
gene from Bacillus subtilis, which imparts sensitivity to su- cific location in the chromosome (101). In this procedure,
crose in proteobacteria and cyanobacteria, can also be used the IeSceI meganuclease from yeast, which recognizes a
for negative selection by selecting for colonies capable of large sequence that is not found in E. coli and other bacte-
growth on rich media containing 5% sucrose (sucrose is fil- ria, is used. The desired mutant copy of the gene is intro-
ter sterilized and added after the medium cools) (16, 22,49, duced on a suicide plasmid. By selecting a plasmid-encoded
68). A n additional strategy involves the use of the rpsL gene drug resistance encoded on the plasmid backbone, the sui-
encoding the S12 subunit of the ribosome. Certain muta- cide plasmid will integrate into the chromosome by homol-
tions in the rpsL gene confer resistance to streptomycin. ogous recombination. The plasmid also contains the I-SceI
However, streptomycin resistance is recessive to the wild- recognition site in its backbone such that upon integration
type allele when placed in truns in the cell allowing nega- the I-SceI recognition site resides between the mutant and
tive selection for loss of the wild-type allele. This procedure wildetype alleles (for example at positions 1 to 3 in Fig. 1).
was initiallv used in E. coli and subseauentlv in other less In a second step, expression of the I-SceI endonuclease in
tractable bacteria like Bordetella pertussis andViVibno cholerue crc~nsallows for selection of recombinants that lost the plas-
(107, 119, 125). mid. Following loss of the integrated plasmid a second strat-
Negative selection can be used as a mechanism to facil- egy must be used to identify recombinants that have main-
itate and select RecA-mediated recombination events in tained the altered allele and not the wild-type allele. The
31. Gene Transfer in Gram-Negative Bacteria 739

double-strand break acts both as a negative selection and some with large linear DNAs (Table 2). Following intro-
also as a means to facilitate recombination. Variations on duction of a desired mutation in such a genetic background,
the use of I-SceI as a mechanism for negative selection have a second mechanism is needed to move the recombinant al-
also been used with bacteriophage-encoded recombination lele into a reef background. Some of the limitations of
systems (see below) (69). these recombination strategies that abolish RecBCD func-
tion are the need for long stretches of homology to the tar-
31 .I .I .3. Double-Crossover Systems with Linear get substrate (-300 to 1,000 bp) and a low frequency of
Substrates recombinants requiring direct selection for the recombina-
When linear DNA substrates are used to modify the chro- tion event. One strategy that appears to circumvent the re-
mosome, a double-crossover event must take place because combination barrier imposed by the exonuclease activity of
a single-crossover event would result in a broken chromo- RecBCD with linear DNA is through the inclusion of very
some and cell death (Fig. 3). However, as mentioned above, large stretches of homology flanking the DNA substrate.
the potent nuclease activity of RecBCD in wild-type E. coli This allowed large fragments (-20 kb) in linearized BACs
cells prevents linear DNAs from residing in the bacterium to be crossed into RecBCD+ E. coli strains when the frag-
long enough for recombination to occur. Mutant strain ment was flanked by -6-kb segments of DNA that were ho-
backgrounds have been used to introduce linear DNA, mologous to the E. coli chromosome (105).
which inactivate the nuclease activity of RecBCD but also
allow RecA to load onto the DNA substrate. Mutations 31 .I .2. Site-Specific Recombination Systems Can
abolishing recB and/or recC functions in a strain back- Remove Selectable Markers
ground (like JC7623) that also contains a sbcBC suppressor Another strategy for the removal of a selectable marker that
allele allow recombination with linear DNAs (63, 133). was introduced via a double crossover is the use of various
Mutations in the recD gene abolish the RecBCD exonucle- site-specific recombination systems like the Cre and Flp re-
ase activity without inactivating its ability to load RecA, combinases. The Cre recombinase functions at its cognate
thereby preserving its recombination function (108). A lox sites, and the Flp recombinase acts at frt sites. When the
strain with a TnlO insertion inactivating the recD gene, recombinase is expressed in trans, a selectable marker situ-
such as CAG12135, can be used for modifying the chromo- ated between the cognate recombination sites is efficiently
removed. The Cre system originally came from bacterio-
phage P1, but this system has been utilized in a number of
eukaryotic organisms as well (110). The Cre system has
been adapted for use in a variety of hosts as a tool to remove
antibiotic resistance determinants (8, 85). The Flp recom-
binase from the yeast 2km plasmid can be used in a similar
fashion and is described below.
31 .I .3. Recombination Systems from Bacteriophages
One of the greatest recent advances in tools for the genetic
modification of E. coli has been the widespread establish-
ment of the recombination system from bacteriophage A,
called the X Red recombinase, as well as a system from a
related phage (39, 136, 139). These systems now allow re-
b. i\ combination efficiencies that were previously possible only
in yeast (97, 126). Linear DNA substrates have been used
for some time in yeast, capitalizing on its highly efficient
recombination-based double-strand break repair system that
is stimulated by linear DNA. The segments of homology
that are required for recombination in yeast can be as short
as 50 bp (12, 76).
Bacteriophage-encoded recombination functions are well
i ) _+_ _ _ _i_ _ _ _ _ _ _ _ _ _ _ _ _ _ suited for manipulating bacterial chromosomes and large
C. low-copy-number plasmids such as BACs. However, there
1 2 are some limitations for manipulating small high-copy-
number cloning vectors (136). Red-mediated recombina-
FIGURE 3 Genetic information can be integrated into the tion is much simpler than the host-encoded RecA-mediated
chromosome of a bacterium using a double-crossover event systems. The A Red recombinase functions reside in three
from a linear DNA construct. (a) A single open reading frame genes, exo ( r e d a ) ,bet ( r e d p ) , andgum (37,74). The Beta and
(thick line) to be inactivated is shown with its promoter (solid Exo proteins have functions analogous to those of RecA
arrow) and RNA transcript (dashed line with arrow). (b) Two and the RecBCD proteins, respectively. The Beta protein
crossover events must occur for the linear DNA to be inte- binds stably to single-stranded DNA greater than 35 nu-
grated into the gene when selecting for gene products indicated cleotides and promotes pairing or annealing between com-
by 1 or 2. Homology provided on the fragment is indicated plementary single-stranded DNAs. The exonuclease func-
with Xs. (c) Integration of the DNA substrate removes all or tion of Ex0 can tailor double-stranded DNA substrates to
a portion of the gene and fuses the genes carried behind the tar- make them competent for recombination with Beta. The
get gene promoter. Depending on the construct, the transcript Exo function is dispensable when single-stranded DNA sub-
may be terminated within the integrated DNA segment. strates are used for recombination (136, 137). Homologs of
Promoters within the integrated DNA segment could activate the Exo and Beta proteins, called RecE and RecT, are en-
adjacent genes in the chromosome. coded on a cryptic prophage called Rac that is found in the
740 MOLECULAR GENETICS

TABLE 2 E. coli strains


Strain or plasmid Genotypea Commentsb
E. coli strains thr-1 aruC14 kuB6 A(gpt-proA)62 lacy1 tsx-33 A common laboratory strain (43); CGSC 1157
AB1157 qsr'-0 glnV44 gulK2 Rm-0 hisG4 rjbD1 mgl-5 1
rpoS396 rpsL31 kdgK51 xylA.5 mtl-1 urgE3 thi-1

BW20767 A 3 ( l a ~ ) ~RP4-2-tet:Mu-lkun:
~,, :Tn7 integrant leu- Strain for mobilizing plasmids with RP4 origins of transfer
63::ISlO recA1 creC510 hsdR17 endA1 sbf-5 via an integrated RP4 fragment (RP4-2-tet:Mu-
uidA(AMluI)::pir+ thi 1kan::Tn7) which imparts resistance to spectinomycin,
streptomycin, and trimethoprim. pir gene allows replica-
tion of conditional R6K origin plasmids (88, 89);
ATCC 47084.

BW23473 A(argF-lac) 169' robAl creC510 hsdR5 14 endA9 Chromosome has the wild-type pir gene which encodes
recA1 AuidA::pir+ the 7~ protein required for the R6K origin replication at
a copy number of -15 (88, 89); CGSC 7837.

BW23474 A(urgF-lac) 169' robAl creC510 hsdR5 14 endA9 Chromosome has the mutant pir-116 gene which encodes
recA1 AuidA::pir-116 a 7~ protein required for the R6K origin replication at a
copy number of -250 (88,89); CGSC 7838.

CAGl2135 MG1655 recD1901::TnlO MG1655 strain with a TnlO insertion in recD. Allows lin-
ear DNA to persist long enough for recombination into
the chromosome (1 18); CGSC 7429.

DH5a endA1 hsdRl7 glnV44 thi-1 recAl gyrA96 A(urgF- Cloning strain that allows a complementation. Available
lac) 169' deoR (+80dlacA(lacZ)M15) from Invitrogen (134).

ER1793 jhuA2 A(lacZ)rl glnV44 e14- up31 his-1 rpsL104 Cloning strain that lacks the native E. coli K-12 restriction
xyl-7 mtl-2 metB1 A(mcrC-mmr)114::1S10 systems. Should be used when cloning DNA directly
from non-E. coli strains. Available from New England
Biolabs.

ER2925 ara-14 kuB6 jhuA3 1 lacy1tsx78 glnV44 galK2 gulT22 Cloning strain that is both dam- and dcm-negative. Useful
mcrA dcm-6 hisG4 rfbD1 R(gb210::TnlO)TetS for propagating DNA that will be introduced into strains
endA1 rpsL136 daml3::Tn9 xylA-5 mtl-1 thi-1 that restrict methylated DNA. Available from New
mcrB1 hsdR2 England Biolabs.

HBlOl thi-1 hsdS20 glnV44 recA13 aru-14 leuB6 proA2 A common laboratory strain (75). ATCC 33694.
lacy1 gdK2 q ~ L 2 ~yl.5
0 mtl-1

HME5 inversim(nnD-nnE) 1 rph-1 A(argF-lac)169' Xc1857 Temperature-sensitive strain that allows for the induction
A(cro-bioA) of the A Red (Ex0 Bet Gam) proteins with a short period
of growth at 42C (136).

JC7623 AB1157 sbcC2Ol sbcB15 recB21 recC22 Mutations in recBC sbcBC allow linear DNA to persist
long enough for recombination into the chromosome
(63). CGSC 5188.

JM109 endA1 recAl gyrA96 thi hsdR17 relA1 glnV44 A(lac- Reference 135; ATCC 53323.
proAB) (F' truD36 proAB lacIqZAM15

CAG18642 MG1655 srlD3131::TnlO Can be used to move mutations in recA with P1 transduc-
tion; CGSC 7423.

MC4 100 uraD139 A(argF-lac)169'rpsL150 relA1 flhD5301 A common laboratory strain. The A(fruK-yeiU)725 allele
deoCl ptsF25 rbsR22 e14- A(fimB-fimE)632::1Sl was formally referred to as fruA25 (25, 99); CGSC
A(fruK-yeiU)725 6152.

MG1655 rph- 1 A common laboratory strain. The genome has been se-
quenced (15); CGSC 6300.
(Continued on next page)
31. Gene Transfer in Gram-Negative Bacteria w 741

TABLE 2 Continued
Strain or plasmid Genotype" Commentsb
S17-1 thi pro hsdR recA (RP4-2-tet::MuaphA::Tn7) Used for mobilizing plasmids with RP4 origins of transfer.
The strain is sensitive to kanamycin and tetracycline
but resistant to spectinomycin, streptomycin, and
trimethoprim. A derivative with Apir is also available
for maintenance of R6K origin plasmids (117); ATCC
47055.

SMlO C600 (RP44-2-tet:Mu-1kan) Used for mobilizing plasmids with RP4 origins of transfer.
The strain is sensitive to tetracycline but resistant to
kanamycin. A derivative with Xpir is also available for
maintenance of R6K origin plasmids (117).

W3110 inversion (rrnD-mE) 1 rph-1 W3110 is a common laboratory strain. The genome has
been sequenced (9); CGSC 4474.

XL1-Blue F'::TnlO proAfB+ lacI, A(lacZ)Ml5/recAI endAl Cloning strain that allows a complementation. Available
gyrA96 thi hsdRl7 glnV44 relAl lac from Stratagene.

Plasmids
pCP20 Temperature-sensitive plasmid with thermal induction of
Flp recombinase (27). CGSC 7629.

pKD46 Temperature-sensitive plasmid with the X Red proteins


under arabinose control (39); CGSC 7669.

pKD3 Plasmid encoding ampicillin resistance that allows PCR


amplification of a gene cassette encoding chloram-
phenicol resistance flanked by frt sites recognized by the
Flp recombinase (39); CGSC 7631.

pKD4 Plasmid encoding ampicillin resistance that allows PCR


amplification of a gene cassette encoding kanamycin re-
sistance flanked by frt sites recognized by the Flp recom,
binase (39); CGSC 7632.

pACBSR Chloramphenicol-resistantplasmid with X Red proteins and


the I-SceI meganuclease under arabinose control (58).
'Unless indicated, all of the strains do not contain bacteriophage X or the F plasmid.
bThe catalog numbers for the American Type Culture Collection (ATCC) and the E. coli Genetic Stock Center (CGSC) are given when available.
'The A(argF-h)169 deletion allele actually deletes ykfD-mhpD (99).

common laboratory E. coli strain K-12 and most other non- nated expression (3 7). Constructs made in this background
laboratory strains. RecE and RecT are normally not ex- will likely be moved to a new strain background via trans-
pressed except in the presence of activating mutations duction or conjugation. Other strategies utilize plasmid
called sbcA mutations (10). T h e Gam protein is a modifier constructs for expression of the Exo, Beta, and Gam pro-
of two exonucleases in E. coli, RecBCD and SbcCD. As teins utilizing a n arabinose-inducible promoter (39, 58,69,
mentioned above, RecBCD is a potent nuclease responsible 139). O n e advantage of the plasmid-based systems is the
for degrading linear DNAs in E. coli. The Gam protein is ability to move the system into the strain of choice. Indeed,
capable of binding RecBCD, thereby inhibiting many, but these plasmid constructs have even allowed the system to
not all, activities of RecBCD, and functionally protecting be adapted to a few other species including Yersinia sp.,
linear DNA. Shigella flexneri, and Salmonella enterica serovar Typhi-
There are a number of different systems that allow ex- murium (42, 87, 102, 113, 127, 128). Anecdotal accounts
pression of the A Red recombination functions. In one indicate that the system from bacteriophage lambda has
system using strain HME5 and its derivatives, the phage met with little success in nonenterics. T h e best strategy for
proteins are expressed at high temperature from the chro- organisms unrelated to E. coli may be to adapt the recombi-
mosome by a defective A phage (Table 2) (136). Expression nation proteins from bacteriophages that are endogenous to
of the Exo, Beta, and Gam proteins is induced to high lev- the target bacterium.
els by a 15-min heat shock at 42C just prior to making the One of the greatest advantages of using the A Red re-
cells electrocompetent (78). An advantage of the chromo- combination proteins to recombine substrates into the
somally encoded system is its tight regulation and coordi- chromosome is that small regions of homology (30 to 70 bp)
742 rn MOLECULAR GENETICS

are sufficient to allow recombination (Fig. 3). Recom-


bination that is catalyzed with RecA typically requires
much larger regions of homology (see above). PCR can be a. Antibiotic resistance
used to quickly synthesize recombination substrates for in
vivo recombination with the A Red recombinase, which FRT FRT
works efficiently with such small regions of homology (39,
136); primers can be designed to amplify a selectable marker
where a 5 tail can be included on the primer with homol-
ogy that flanks the region to be deleted (12, 76). This pro-
cedure provides a convenient way to introduce antibiotic-
resistance-conferring genes (cassettes) to replace genes in
the E. coli chromosome. However, it is often desirable to
remove the selectable marker since leaving a selectable
h
U.
marker in an operon could produce polar effects on down- Target gene
stream genes or the selectable marker may simply be needed
for other purposes. Template plasmids can be used to make
recombination substrates with fit recombination sites flank-
ing the antibiotic resistance determinant that are recog-
nized by the Flp recombinase (Fig. 4a) (39). When the tem-
plate plasmids pKD3, pKD4, and pKD13 are used as a
source for the antibiotic resistance cassette, the f i t sites
C.
FRT FRT
flanking the resistance gene can be used to remove the gene
and leave behind a deletion that allows translation of the
remaining portion of the gene intact; in-frame deletions
will not cause a polar effect on downstream genes. The Flp
recombinase that is used to catalyze site-specific recombi- ____________ F
nation between the frt sites is thermally induced from the
plasmid pCP20 (Table 2 and Fig. 4c). The plasmid also has
d.
FRT
a temperature-sensitive replication system that is therefore
lost (or cured) at 42C. FIGURE 4 Recombination substrates can be made in vitro
for use with A Red recombination. The antibiotic resistance
31 . I .3.1. Other Applications with Bacteriophage used to select for recombinants can subsequently be removed
Recombination Systems by expressing the Flp recombinase that will excise the region
There are numerous applications for the A Red recombina- between the two fit sites (FRT). (a) Primers are designed to
tion system for manipulating chromosomes and plasmids. amplify from a universal antibiotic-resistant cassette withfrt re-
One consideration when carrying out these procedures is combination sites using primer binding sites P1 and P2. The
the hazard of unwanted recombination events. Given that primers contain 5 tailsthat are complementary to the target
the frequency of unselected recombination events is re- gene at regions H1 and H2 for the subsequent crossover event.
ported to exceed 1% (36, 96, 136), it stands to reason that (b) The PCR-generated substrate is transformed into a cell ex-
a number of unintended recombination events will likely pressing the Exo, Beta, and Gam proteins (substrate shown
occur in the bacterial genome besides the one that was in- miniaturized). (c) Selecting for the antibiotic resistance marker
tended. Therefore, it is important that any time A Red crosses in the cassette with its flanking frt recombination sites
recombination is used, the recombinant allele should be using the homology at regions H1 and H2. (d) The Flp recom-
moved into a clean genetic background by using transduc- binase is transiently expressed and subsequently lost because
tion or natural transformation. As with RecA-mediated replication from the pCP20 vector is temperature sensitive.
recombination (see above), I-SceI-mediated double-strand The Flp recombinase efficiently catalyzes recombination be-
breaks can be used as a form of negative selection that also tween the fit recombination sites removing the antibiotic
enhances recombination. In such a procedure the I-SceI resistance-conferringcassette. See the text for details.
recognition site is first introduced into the gene of interest
by recombination or transposition. In a second step, a linear
recombination substrate is introduced into the cells at the
same time as a vector expressing the I-SceI meganuclease Other novel strategies for the Red proteins include using
and the A Red recombination proteins. In another efficient them to manipulate large DNA substrates encoded on bac-
procedure called gene-gorging, the DNA substrate that is to terial artificial chromosome (e.g., for building substrates for
be recombined into the genome is linearized in vivo from a mouse genetics), for cloning genes via homology, and as
plasmid through the use of I-SceI sites that flank the DNA a tool to recombine DNA substrates subjected to in vitro
segment (58). The ability to linearize the substrate in vivo transposition into the chromosome (34,37, 38).
addresses the issue of transformation efficiency, the greatest
limiting factor in the A Red system, because all cells will 31 .I .4. Introducing DNA at Specific Sites in the
have the linearized substrate. Single-stranded DNA oligo- Chromosome
nucleotides have also been reported to efficiently recom- There are multiple strategies for introducing DNA at spe-
bine into the chromosome in a process that obviates the cific locations in the chromosome. One mechanism in-
need for the Gam and Exo proteins (96, 137). As expected, volves using site-specific recombination systems endoge-
the process of A Red-mediated recombination is enhanced nous to E. coli and its bacteriophage. An advantage of
in strains that lack the methyl-directed repair system (36). integrating DNA directly into the chromosome is that it
31. Gene Transfer in Gram-Negative Bacteria 743

allows genes to be expressed at one copy per chromosome, for H. pylori, and an alternative system was therefore
thereby avoiding artifacts caused by expressing genes on needed to express genes in this organism (59). Null muta-
multiple-copy plasmids. One clever system for introducing tions in the rdxA gene of H. pylori impart resistance to
DNA into the chromosome makes use of five insertion sites metronidazole; therefore, inactivation of this gene could be
for bacteriophages in E. coli and their cognate recombinases used as a selection for gene cassettes into this site via
that can be supplied on plasmids (53). In this system the double-crossover events ( 121). Similar strategies can likely
genes of interest are cloned into plasmid vectors that are be developed for other organisms.
subsequently introduced into an E. coli host that allows in-
tegration into the chromosomal bacteriophage attachment
sites, but not replication of the plasmid. The availability of 31.2. INTRODUCING DNA ON PLASMIDS
multiple attachment sites for integration with constructs A n important strategy for manipulating the genetic content
containing various expression systems and different antibi- of bacteria is the use of plasmids. Plasmids can be described
otic resistances allows many genes to be expressed in a sin- in two broad families of replicons, narrow- and broad-host-
gle strain. Integrated vectors can be removed using plasmid range vectors. Narrow-host-range plasmids are stably main-
constructs encoding excision genes, or the construct can be tained in E. coli and its close relatives (Table 1). Common
rescued as a plasmid when transduced into the appropriate examples of narrow-host-range replicons are the ColEI/
host. Presumably, similar systems could be established in pMB1, p15A, and pSClOl replicons (Tables 2 and 3). The
other less tractable bacteria using bacteriophage integration ColEI/pMBl, p15A, and pSClOl replicons all fall into dif-
sites or integrative and conjugative elements (conjugative ferent incompatibility groups and can therefore reside in
transposons) (21). Another system that utilizes bacterio- the same host at the same time. Broad-host-range vectors
phage attachment sites is based on a A bacteriophage deriv- are capable of replicating in E. coli and a large number of
ative, called A Inch, encoding two regions that are homol- different proteobacteria (Table 3). Some of the broad-host-
ogous to common pBR322-based plasmids encoding range vectors are also capable of replicating in various
ampicillin resistance (19). A n advantage of this system is cyanobacteria (68). All of the vectors in Table 3 are in dif-
that genes that are already cloned in a pBR322-based vec- ferent incompatibility groups and could in theory be main-
tor can be constructed into the chromosome with minimal tained in the same permissive host organism. There are
additional effort. A strain containing the gene of interest multiple considerations when choosing which type of plas-
cloned into a pBR322-based plasmid is used as a host to mid to use to replicate any given substrate. Many vectors
grow the A I n c h bacteriophage. Recombination between contain controllable promoters to limit the expression of
the plasmid and a truncated copy of the bla gene encoded cloned genes that are derived from sugar catabolism systems
on the bacteriophage results in resistance to ampicillin; how- such as those from lactose or arabinose (52, 65).
ever, the same recombination event removes a bacteriophage- Plasmids often contain a region of sequence called a
encoded kanamycin resistance gene. This procedure allows multiple cloning site (MCS) with recognition sites for
the isolation of recombinant bacteriophages by selecting many different restriction endonucleases to facilitate
strains that have the bacteriophages integrated in the chro- cloning. A useful MCS often encodes a deletion derivative
mosome (lysogens), which are resistant to ampicillin but are of the LacZ protein called LacZa as a tool to identify clones.
kanamycin sensitive. Lysogens can be prevented from excis- When the A(LacZ)MI5 allele is expressed in a strain with
ing and locked into the final host by selecting for a second the LacZa mutant protein, it allows LacZ activity to be re-
recombination event that removes a large portion of the stored in a process called a-complementation (see Bac-
bacteriophage. terial Strains below).
Transposons can be used to introduce DNA into the A n additional consideration when selecting plasmids is
chromosomes of proteobacteria and cyanobacteria (31, the copy number, the steady-state number of copies of the
41). However, one disadvantage of using random transpo- plasmid that will reside in the cell. Copy number can vary
sition to insert cloned genes into the chromosome is that from very low (for example, pSClOl and F plasmid deriva-
the insertion events can cause problems associated with tives have a copy number of one or a few per genome) to
gene inactivation. A n additional consideration is that the very high (for example, ColEI/pMBl derivatives can have a
transcription of each transposition event will be affected to copy number approaching 1,000) (109).
different degrees depending on where the DNA inserted. A Conditional replicons, in which replication occurs only
solution for this problem came from the use of the transpo- in certain hosts or at certain permissive temperatures, are
son Tn7, which inserts at a single position, called its at- important tools for genetics. Multiple temperature-sensitive
tachment site (attTn7), and only in one orientation (98). derivatives of the pSClOl replicon have been adapted for
The attTn7 site is found in the transcriptional terminator cloning with the addition of a variety of antibiotic resis-
of the glmS gene in proteobacteria, and insertion into this tance selection systems and a useful MCS (100). A n ex-
site does not negatively impact the host. A transposition tremely useful conditional replication system has been as-
system has been developed whereby the DNA of interest is sembled using the R6K plasmid replicon (122). Plasmids
cloned into a T n 7 derivative that can then be introduced that contain the R6K-y origin are maintained only in cells
into the attTn7 site of proteobacteria following conjuga- where the T protein (encoded by the pir gene) is expressed
tion into the host (67). (70,89). E. coli normally does not encode the T protein and
Another option for introducing DNA directly into the will not replicate plasmids with the RGK-y origin. However,
chromosome involves direct selection for insertions by gene R6K-y origin plasmids will replicate in E. coli strains with
inactivation. For instance, examples exist where inactivat- the pir gene on a A phage or otherwise integrated into the
ing a single gene product results in resistance to an anti- chromosome, such as in the uidA gene in strains BW23473
biotic or other substance (89). This strategy has proved and BW23474 (Table 2) (88, 89). A mutant pir-116 gene
successful at introducing genes into the chromosome of also exists that allows R6K-y origin plasmids to be main-
Helicobacter pylori. There is only one shuttle plasmid system tained at a higher copy number in the host cell (Table 2).
744 MOLECULAR GENETICS

TABLE 3 General classes of plasmids commonly used in gram-negative bacteria


Founding replicon Common examples Host range Comments

pMBl/ColEI pBR322 (18), pUC vectors (135), Narrow pBR322 is a low-copy-numbervector (-20 copies/
pGEM vectors, pBluescript vectors cell) which has been adapted as a very-high-
copy number vector (>300).
p15A pACYC177, pACYC184 (26) Narrow The pACYC vectors are low-copy-numbervectors
(-15/cell). The p15A is similar to, but compati-
ble with, the pMBl/ColEI replicon.
pSCl0l pSCl0l Narrow Low-copy-number vector (-5) good for toxic genes.
Temperature-sensitivederivatives exist ( 100).
F plasmid pBeloBACl1 (114) Narrow The original Fertility (F) plasmid, Replication origin
utilized in BACs
RK2 (RP4) pSP329 (24),pCM62, pCM66 (86) Broad IncP incompatibility group
RSFl 010 pJRD215 (40), pSUP104, pSuP204 Broad IncQ incompatibility group
(103)
PSa pUCD2 (30) Broad IncW incompatibility group
R6K R6K Broad IncX incompatibility group
pBBRlMCS pBBR1MCS-2, pBBR1MCS-3, etc. (71) Broad Undefined incompatibility group

31.3. GENERALIZED TRANSDUCTION protocols are given here for use in transduction experi-
In generalized transduction, alleles from a donor strain can ments.
be moved to a recipient in a bacteriophage-mediated process
31.3.1 . I . Protocol for Determining Plaque-Forming
when host DNA is mistakenly packaged into the bacterio-
phage. This is contrasted by another form of bacteriophage-
Units of P I
mediated transduction called specialized transduction. In 1. Prepare an overnight culture with a recA+ E. coli
specialized transduction, genes adjacent to a bacteriophage strain in 5 ml of LB media with 5 mM CaC12.
integrated in the chromosome are packaged into the phage 2. Seriallv dilute the P1 stock DreDaration. called a
particle replacing a portion of the phage genome. The bacteriophagk lysate, with 100-fold diluiions (lo-, lop4,
process of specialized transduction was of important histor- and
ical significance and is still of great importance for making 3. Combine 100 r~.lof each phage dilution with 200 p1
libraries of large DNA fragments but has previously been of the overnight culture in a glass test tube.
covered in detail (124). 4. Incubate at 37C without shaking for 15 min to allow
the bacteriophage to adsorb to the cells.
31.3.1. Generalized Transduction in E. coli with 5. Add 3 ml of molten top agar to one test tube contain-
Bacteriophage P I ing the mixture of cells and diluted bacteriophage. The top
P1 is a well-studied bacteriophage that has been adapted for agar should have been prepared in advance and set out of
generalized transduction (124). P1 is normally a lysogenic the dry or wet bath so that it is warm to touch, but still
bacteriophage that replicates as a plasmid in E. coli. molten.
However, virulent derivatives of P1 have been isolated for 6. Gently mix the agar preparation in the test tube with-
use in generalized transduction. When P1 is grown on an E. out producing bubbles, pour onto a fresh LB plate, and
coli host, 0.3 to 6.0% of the phage particles contain host allow the solution to solidify. Repeat for each bacteriophage
DNA that can subsequently be moved or transduced into dilution and incubate inverted overnight at 37C.
a new host where lop4 to transductants per infected 7. The following day count the plaques from plates bear-
donor cell can be obtained. P1 packages 110 to 115 kbp of ing between 30 and 300 plaques to calculate the number of
DNA, 2.3% of the genome of standard laboratory strains. plaque-forming units of bacteriophage in the stock lysate.
Therefore, the longest possible distance where a selected ge-
netic marker can bring along a second marker, in a process 31.3.1.2. Procedure for P I Growth from Plaques
called cotransduction, is approximately 2 min according to
the genetic map of E. coli K-12 originally derived by conju- 1. Prepare an overnight culture of a recAf E. coli strain
gal mapping (14). in 5 ml of LB media with 5 mM CaC12.
Virtually all strains of E. coli can act as donors for P1 2. Use a sterile Pasteur pipette to collect bacteriophage
transduction. E. coli strains that are recA+ produce more P1 plaques from an LB plate as plugs. Assemble six test tubes
phage particles than recA-negative strains (25 to 150 versus with 100 pl of overnight cells. To each tube with cells add
5, respectively) per donor cell. The lower titers of P1 phage 0, 1, 2,3,4, or 5 plaques, mix gently, and incubate at 37C
found with recA-negative cells contribute to lower levels of for 5 min without shaking.
transducing phage particles. Host recombination is required 3. Add 5 ml of LB media containing 5 mM CaC12and
to recombine the DNA packaged into P1 particles into the continue incubating at 37C with vigorous aeration until
recipient chromosome. Therefore, recipient cells must be lysis is visible in one of the tubes (usually 3 to 5 h). If mul-
recA+. General principles for handling bacteriophages are tiple tubes show good lysis, use the tube that was produced
explained in depth in another chapter in this text. Brief with the fewest plaques.
31. Gene Transfer in Gram-Negative Bacteria 745

4. Add several drops of chloroform and vortex briefly. as much DNA as P1 and also is less sensitive to capsules
Centrifuge at 2,500 X g to remove the cell debris. found on many pathogenic strains of E. coli. Special T4 de-
5. Store phage lysates at 4C protected from light with rivatives must be used for transduction that do not degrade
additional chloroform. The chloroform will kill the residual and modify host DNA. P1 can also be used in some other
donor bacteria overnight at 4C or within a couple of hours bacteria, such as Myxococcus xanthus (64, 94). The bacte-
at room temperature. Phage titers with this procedure should riophage P22 was first used to demonstrate the process of
be in the range of lo9 to 10" PFU per ml. generalized transduction in the bacterium S. enterica serovar
Typhimurium (140). Bacteriophages that are capable of
31.3.1.3. Procedure for P I Growth on Donor Strains generalized transduction have since been reported in a
number of bacteria, including V. cholerae (CP-T1) (57),
1. Prepare an overnight culture of a donor strain of E. Vibrio puruhaemolyticus and Vibrio alginolyticus (phi VP253
coli in 5 ml of LB media with 5 mM CaClz (for example, and phi VP143) (91), Pseudomom aeruginosa (DMS3)
strain CAG12135 containing a Tnf 0 encoding tetracycline (20), Cuulobacter crescentus (Cr30T) (13), Xunthomona
resistance inserted in the recD gene) (Table 2). campestris pv. campestris (XTP1) (13 l ) , Sinorhizobium
2. Add 100 p1 of overnight cells to each of four test meliloti (phage 11 M12) (47, 115), and Myxococcus xunthus
tubes. (phage MX4) (23).
3. To each tube add 0,50,100, or 200 pl of stock P1 bac-
teriophage lysate and incubate at 37C with vigorous aera- 31.3.4. Applications for Generalized Transduction
tion until lysis is visible in one of the tubes (usually 3 to 5 There are many applications for generalized transduction in
h). Use the tubes that showed the best clearing with the genetics for proteobacteria. For example, generalized trans-
fewest phage as a donor lysate (see below). Use the tube duction is an important tool for moving mutations that
with 0 pl of stock for comparison. might be in different backgrounds into a single background;
4. Add several drops of chloroform and vortex briefly. an important consideration when determining the effect of
Centrifuge at 2,500 x g to remove the cell debris. various mutant alleles is the effect of strain background. If a
5. Store phage lysates at 4C protected from light with mutant allele has an associated selectable marker, such as a
additional chloroform. The chloroform will kill the residual transposon insertion in a given gene, transductants can be
donor bacteria overnight at 4C or within a couple of hours selected directly. However, it is important to confirm with a
at room temperature. separate assay that the allele has been recombined correctly
into the new background by using a phenotype associated
with the mutation or by physical means like PCR or
31.3.1.4. Procedure for P I Transduction Southern hybridization. Plasmids can also be moved by gen-
1. Prepare an overnight culture of a recipient strain of E. eralized transduction in a mechanism that must still accom-
coli in 5 ml of LB media with 5 mM CaClz (for example, the modate the headfull packaging mechanism (81,83).A very
tetracycline-sensitive strain MG 1655) (Table 2). clean way to move a given mutation is to utilize linked al-
2. Add 300 ~1 of overnight cells to each of four test leles that can be selected for and selected against. One ex-
tubes. ample involves producing bacteria that are recA-negative
3. To each test tube with recipient cells add 0,50,100, by using a linked T n f 0 insertion in the sorbitol genes,
or 200 pl of P1 bacteriophage grown on the donor cells in srlD3 131 ::Tn10. The tetracycline resistance encoded by
a test tube (i.e., the best donor phage lysate above). Tn10 can be used to introduce the srlD3131::Tnf 0 allele
Incubate without shaking at 37C for 25 min (do not allow into a target strain which in turn results in a strain inca-
to go over 30 min). pable of growth on sorbitol as a sole carbon source. A recA-
4. Add 5 ml of LB medium with 10 mM sodium citrate negative srlD+ strain can then be used as donor for trans-
to each tube and centrifuge at 2,500 x g to collect the cells. duction. By selecting for the ability to grow on sorbitol as
5. Remove the supernatant and resuspend the cells in 5 the sole carbon source with the recA-negative srlD+ donor
ml of LB medium with 10 mM sodium citrate and incubate lysate, the recA-negative allele can be cotransduced. The
at 37C with aeration for 1 h. transductants that have replaced the srlD3131::Tnl@allele
6. Centrifuge at 2,500 X g to collect the cells and plate with the srlD+ allele can now grow on sorbitol minimal
on selective medium. Also plate 200 pl of the donor phage plates. The transductants that can grow on sorbital are
lysate as a control. I t is also a good idea to spot all of the screened for the recA-negative allele by using sensitivity to
reagents on a selection plate. UV light or mitomycin C. Because the strain has lost the
srlD3131::Tnf 0 allele, the organisms are now tetracycline
This procedure should result in hundreds of transduc- sensitive.
tants. It is important to streak purify transductants at least Generalized transduction can also be used in mapping
once on selective media to ensure that the bacteria are free mutations and in a variety of mutagenesis strategies. For ex-
from residual bacteriophage (124). Very fresh plates or ample, a strain containing a mutation of interest can be
plates that are wet with condensation can limit the pro- used to make a pool of bacteria with random transposition
cedure because they contribute to the killing of potential events. The transposons are then transduced to a new ge-
recombinants by residual bacteriophage on the selection netic background where one can screen for the mutant al-
plate. Minimal growth media can be used instead of LB lele. Strains that contain both the transposon insertion and
where required. the mutation from the donor strain must have the two alle-
les linked. Transposon insertions that are very tightly linked
31.3.3. Generalized Transduction in Other Systems to the mutation can then be mapped using arbitrary PCR to
There are many bacteriophages that are suitable for gener- determine the region where the original mutation occurred
alized transduction in E. coli and in other bacteria (83). In (28).
E. coli a modified derivative of the bacteriophage T4 offers Generalized transduction can also be used for localized
some advantages for transduction in that it packages twice mutagenesis (60). A strain containing a selectable marker
746 w MOLECULAR GENETICS

in the region of interest is heavily mutagenized and then 7. Aliquot the cell suspension in 0.5-ml amounts into
used to grow bacteriophage. The region of the chromosome prechilled microcentrifuge tubes, freeze in a dry ice/ethanol
that is linked to the marker is then moved to a clean genetic bath or in liquid nitrogen and store at -70C. When
background, virtually ensuring that only a small region of needed for transformation, the frozen cells can be thawed
the chromosome is mutagenized and not the entire genome. on ice and used as described below.
A very useful set of transposon insertions at every minute 8. For transformation, thaw the cells on ice and trans-
position (about every 46 kb) in E. coli is available for map- fer 200 p l of cell suspension to a prechilled sterile 15-ml
ping and cotransducing mutations (92, 118). The set of (17- by 119-mm) plastic disposable centrifuge tube. Prepare
transposon insertions have isolates with antibiotic resis- one tube for each transformation plus tubes for the positive
tances to tetracycline or kanamycin at each position and are and negative controls (see below). Add the DNA in a vol-
available from the E. coli genetic stock center (see below). ume of 5 pl or less, swirl to mix, and incubate on ice for 30
Multiple projects are under way to produce null mutations min.
in every nonessential gene in E. coli. 9. Transfer the tubes in a plastic rack to a circulating-
water bath equilibrated at 42C. Allow the tube to stand
without shaking for 60 s.
31.4. TRANSFORMATION 10. Rapidly transfer the tubes back into ice for 2 min.
Transformation of bacteria by use of either naturally com- 11. Add 800 pl of SOC medium (see Media below)
petent organisms, the process of electroporation, or chemi- and incubate at 37C with aeration for 1 h to allow recov-
cal competency relies on the direct uptake of DNA by bac- ery of the transformants and expression of the genes encod-
teria. A number of commonly used protocols that are ing antibiotic resistance.
optimized for E. coli are given below. The chemical compe- 12. Plate out the cells on selective media in a volume of
tency procedures have limited success for organisms other 100 to 200 pl. It is also possible to collect the cells with
than E. coli; however, the electroporation procedure is centrifugation at 2,500 X g and plate the entire amount in
widely applicable to a variety of proteobacteria, cyanobac- a volume of 100 to 200 pl.
teria, gram-positive bacteria, and eukaryotic cells, often
with some modifications (Table 1) (56). 31.4.1.2. Procedure for Competent E. coli with the
lnoue Method
31.4.1. Chemical Transformation The Inoue method is not as convenient as making calcium
Chemical transformation was discovered over 30 years ago chloride-competent cells but allows for higher-competency
and provides a convenient strategy for introducing DNA cells (10 CFU of bacteria per pg of DNA) (61). The pro-
into E. coli and some other bacteria (Table 1) (82). While cedure requires growing cells at or below standard room
the parameters that affect the process are well understood, temperature (18 to 22C).
the exact mechanism that allows for the uptake of DNA re-
mains obscure (56). 1. Prepare and freeze Inoue transformation buffer in ad-
vance.
31.4.1 .I. Procedure for Calcium Chloride- 2. Grow a 25-ml bacterial culture in LB at 37C for 6
Competent . coli to 8 h from a plate that was streaked out the prior day.
3. In the evening subculture the starter culture into
This simple procedure should allow transformation efficien-
three 1-liter flasks with 250 ml of SOB medium (see
cies of lo6 to lo7 CFU of bacteria per pg of DNA, which is
Media below), adding 10, 4, or 2 ml individually to each
sufficient for many applications (32).
flask. Allow the flasks to incubate overnight at 18 to 22C
1. Inoculate 1 liter of LB broth with 20 ml of an with moderate shaking (this may need to be adjusted ac-
overnight culture of E. coli at 37C. cording to the strain used).
2. Grow the cells with shaking at 300 rpm at 37C to 4. The following morning, check the OD600 for all
an overnight density at 600 nm (OD600) of approximately three flasks to find if any is at the appropriate OD600of 0.55.
0.4 to 0.6. Cells should be collected in early to mid-log If none of the flasks is at the appropriate OD600, continue to
phase, and therefore, the appropriate OD600 and time will read the OD every 45 min until one reaches 0.55. Having
vary by strain and growth conditions. three cultures helps ensure that one reaches the appropriate
3. Chill cells as quickly as possible by swirling the flask OD in the correct time frame. Continue with this flask and
by hand in an ice bath for 15 min. From this point on, keep discard the other two.
cells at 0C. Transfer the cells to sterile prechilled cen- 5.Chill cells as quickly as possible by swirling the flask
trifuge bottles and centrifuge in a cold rotor at 4,000 x g for by hand in an ice bath for 15 min. From this point on, keep
15 min at 4C. Discard as much supernatant as possible in- cells at 0C. Transfer the cells to prechilled centrifuge bot-
cluding residual drops on the sides of the tubes, even if some tles and centrifuge in a cold rotor at 4,000 X g for 15 min at
cells are removed in the process. 4C. Discard as much supernatant as possible including
4. Gently resuspend the pellets in a total of 300 ml of residual drops on the sides of the tubes, even if some cells
ice-cold sterile MgC12-CaC12solution (80 mM MgC12, 29 are removed in the process.
mM CaC12). For example, resuspend by swirling instead of 6. Gently resuspend the cells in a total volume of 80 ml
pipetting or vortexing the suspension. Centrifuge as above. of ice-cold Inoue transformation buffer (for example by
5. Gently resuspend the pellets in a total of 100 ml swirling instead of pipetting or vortexing) and transfer the
0.1M CaC12 10% glycerol and transfer to prechilled sterile suspension to prechilled sterile 50-ml (28- by 115-mm)
50 ml (28- by 115-mm) plastic disposable centrifuge tubes. plastic disposable centrifuge tubes.
Centrifuge at 2,500 X g at 4C . 7. Centrifuge at 2,500 X g for 10 min at 4C. Discard
6. Gently resuspend in 40 ml of ice-cold sterile 0.1 M as much supernatant as possible including residual drops on
CaC12-10% glycerol and centrifuge as above. the sides of the tubes.
31. Gene Transfer in Gram-Negative Bacteria 747

8. Gently resuspend the cells in a total volume of 20 ml 9. Rapidly transfer the tubes back into ice and allow to
of ice-cold Inoue transformation buffer. Add 1.5 ml of di- chill for 2 min.
methyl sulfoxide (DMSO) with gentle swirling and store on 10. Add 800 pl of SOC medium and incubate at 37C
ice for 10 min. Quickly aliquot into microcentrifuge tubes, with aeration for 1 h to allow recovery of the transformants
freeze in liquid nitrogen, and store at -70C. and expression of the genes encoding antibiotic resistance.
9. For transformation, thaw an aliquot of cells on ice. 11. Plate out the cells on selective LB medium in a vol-
Add up to 2.5 p1 of DNA solution to 50 p1 of competent ume of 100 to 200 pl. It is also possible to collect the cells
cells in a prechilled sterile 15-ml (17- by 119-mm) plastic with centrifugation at 2,500 X g and plate the entire
disposable centrifuge tube. Prepare one tube for each trans- amount in a volume of 100 to 200 ~ 1 ,
formation plus tubes for the positive and negative controls
(see below). Gently mix the cell-DNA suspension with 31.4.2. Electroporation
swirling, and incubate on ice for 30 min.
Electroporation is a broadly successful strategy for introduc-
10. Transfer the tubes in a plastic rack to a circulating-
ing DNA into proteobacteria and cyanobacteria (56, 68,
water bath equilibrated at 42C. Allow the tubes to sit
93). In this technique, bacteria are subjected to a high-
without shaking for 60 s.
strength electrical field to form pores in the cells. While the
11. Rapidly transfer the tubes back into ice for 2 min.
pores are present, DNA in solution can diffuse into the
12. Add 800 pl of SOC medium and incubate at 37C
cells. Electroporation has proven to be useful in a variety of
with aeration for 1 h to allow recovery of the transformants
and expression of the genes encoding antibiotic resistance.
E. coli strains and, under optimal conditions, results in fre-
quencies of lo9 to 10 transformants per pg of plasmid
13. Plate out the cells on selective SOB medium with 20
DNA. The identification and optimization of electropora-
mM MgS04 in a volume of 100 to 200 pl. It is also possible
tion conditions are different for different genera, and often
to collect the cells with centrifugation at 2,500 X g and
for different species, and require systematic testing of sev-
plate the entire amount in a volume of 100 to 200 pl.
eral variables. These variables include, but are not limited
to, growth phase, cell density, DNA concentration, field
31.4.1.3. Procedure for Rubidium Chloride- strength, pulse duration, and temperature. Electroporation
Competent . coli is the mechanism of choice for making DNA libraries, in-
This procedure is reported to allow transformation efficien- troducing DNA substrates for X Red recombination, and
cies of 10 CFU of bacteria per pg of DNA or greater (55). difficult ligation reactions. Because of the high frequency of
transformation one must also consider the possibility of
1. Inoculate 100 ml of Psi medium with 1 ml of an cotransformation of multiple plasmids.
Overnight culture of E. coli at 37C.
2. Grow the cells with shaking at 300 rpm at 37C to 31.4.2.1. Procedure for Electroporation
an OD600 of approximately 0.4 to 0.6. Cells should be col-
The following procedure is a modification of the procedure
lected in early to mid-log phase, and therefore, the appro-
recommended by Bio-Rad Laboratories (Richmond, CA). It
priate OD600 and growth time will vary by strain and growth
is routinely used to transform E. coli and S. enterica serovar
conditions.
Typhimurium. The apparatus is a Bio-Rad Gene Pulser with
3. Chill cells as quickly as possible by swirling the a Pulse Control Unit or equivalent.
flask by hand in an ice bath for 15 min. From this point
on, keep cells at 0C. Transfer the cell suspension to two 1. Inoculate 1 liter of LB broth with 20 ml of an
prechilled sterile 50-ml (28- by 115-mm) plastic dispos- overnight culture of E. coli at 37C.
able centrifuge tubes. Centrifuge at 2,500 X g at 4C. 2. Grow the cells with shaking at 300 rpm at 37C to an
Discard as much supernatant as possible including residual OD600 of approximately 0.4 to 0.6. Cells should be collected
drops on the sides of the tubes, even if some cells are re- in early to mid-log phase, and therefore, the appropriate
moved in the process. OD,jOOand growth time will vary by strain and growth con-
4. Gently resuspend in a total of 40 ml of ice-cold TfbI ditions.
(see Buffers below) and leave on ice for 15 min. Centrifuge 3. Chill cells as quickly as possible by swirling the flask
at 2,500 X g at 4C. Discard as much supernatant as possi- by hand in an ice bath for 15 min. From this point on, keep
ble including residual drops on the sides of the tubes. cells at 0C. Transfer the cells to prechilled centrifuge bot-
5. Gently resuspend in a total of 4 ml of TfbII (see tles and centrifuge in a cold rotor at 4,000 x g for 15 min at
Buffersbelow) and leave on ice for 15 min. 4C. Discard as much supernatant as possible including
6. Aliquot the cell suspension in 0.5-ml amounts into residual drops on the sides of the tubes, even if some cells
prechilled microcentrifuge tubes, freeze in a dry ice/ethanol are removed in the process.
bath or on liquid nitrogen and store at -70C. When 4. Resuspend the pellets in a total of 1 liter of ice-cold
needed for transformation, the frozen cells can be thawed sterile H20(use very pure water, ideally Milli-Q water or
on ice and used as described below. higher). Centrifuge as above. Resuspend in 0.5 liter of ice-
7. For transformation, thaw an aliquot of cells on ice. cold sterile H2O and centrifuge as above.
Add up to 2.5 pl of DNA solution to 50 p1 of competent 5. Resuspend in a total of 20 ml of ice-cold sterile 10%
cells in a prechilled sterile 15-ml (17- by 119-mm) plastic glycerol in H20and transfer to prechilled sterile 50 ml (28-
disposable centrifuge tube. Prepare one tube for each trans- by 115-mm) plastic disposable centrifuge tubes. Centrifuge
formation plus tubes for the positive and negative controls at 2,500 X g for 10 min at 4C. Resuspend the cells in 3 ml
(see below). Gently mix the cell-DNA suspension with of ice-cold sterile 10% glycerol in H20. The cell concen-
swirling, and incubate on ice for 30 min. tration should be about 3 X 10 CFU of bacteria per ml.
8. Transfer the tubes in a plastic rack to a circulating- 6. Aliquot the cell suspension in 0.1- to 0.2-ml amounts
water bath equilibrated at 42C. Allow the tube to stand into prechilled sterile microcentrifuge tubes, freeze in a dry
without shaking for 60 s. ice/ethanol bath or on liquid nitrogen and store at - 70C.
748 w MOLECULAR GENETICS

When needed for electroporation, the frozen cells can be identified. Interestingly, a novel genetic strategy was used
thawed on ice and used as described below. to isolate a hypercompetent Legionella pneumophila strain
7. Prechill all tubes and electroporation cuvettes on ice, from a normal noncompetent background (112). It is also
preparing enough supplies for each sample and the positive worth noting that natural competency can be of limited
and negative controls (see below). Mix 40 pl of cells with 1 value when transforming plasmids because in many of
or 2 pl of DNA in a chilled microcentrifuge tube (note that these systems the DNA is imported as single-stranded frag-
DNA should be in H 2 0or a low-ionic-strength buffer such as ments.
Tris-EDTA [TE]).Mix well, but gently, and incubate on ice
-
for 1 min. Transfer the cell-DNA mixture to a cold 0.2-cm 31.4.3.1. Procedure for Chromosomal DNA
cuvette, shake to the bottom, and place in the Gene Pulser Isolation
cuvette holder. Apply one pulse with the gene pulser set at Isolation of plasmid DNA is covered in depth elsewhere in
2.5 kV and 25 pF and the pulse controller set at 200 s1. If this text. This procedure is appropriate for preparing mul-
using a 0.1-cm cuvette, set the apparatus at 1.5 to 1.8 kV. tiple small samples by using a microcentrifuge (7). Many
8. Immediately suspend the cells in 1 ml of SOC. It is commercial kits are also available for isolating plasmid and
important to add SOC immediately, since waiting will re- chromosomal DNA.
sult in lower transformation frequencies. Transfer the cells
to a tube, and incubate at 37C with aeration for 1 h to 1. Pellet 1.5 ml of an overnight culture of bacteria for 2
allow recovery of the transformants and expression of the min at top speed in a microcentrifuge. Remove all traces of
genes encoding antibiotic resistance. The apparatus should media.
allow you to read an expression of the pulse strength for 2. Use a vortex mixer and/or pipetting to completely
each sample called the time constant. Ideally the time resuspend the cell pellet in 567 pl of TE buffer.
constant should be between 4 and 5 ms for a 0.2-cm cuvette 3. Add 30 pl of 10% sodium dodecyl sulfate (prepared
containing 40 pl of cells. in HzO) and mix by inversion 10 times.
9. Plate out the cells on selective medium in a volume of 4. Add 4 pl of proteinase K (prepared at a concentra-
100 to 200 p1. tion of 10 mg/ml and stored at -20C as aliquots that are
never used twice) and mix by inversion 10 times.
I t should be noted that electroporation of E. coli with 5. Incubate the samples for 1 h at 37C, mixing every
ligated DNA results in low transformation frequencies un- 15 min by inverting several times.
less the DNA is first transferred to a low-ionic-strength 6. Add 100 p l of 5 M NaCl and mix by extensive mix-
buffer (62, 132, 138). This can be accomplished by precipi- ing (200 times); to avoid shearing of the DNA in this and
tating the reaction with ethanol or by diluting the reaction all subsequent steps, mix by flicking or shaking the tubes; do
1/10 to 1/20. For ethanol precipitation add 2.5 volumes of not vortex or use a pipette for mixing.
ethanol and 0.1 volume of 3 M sodium acetate (pH 5.2) fol- 7. Add 80 pl of prewarmed (65C) hexadecyltrimethyl-
lowed by centrifugation and a 70% ethanol wash (109). ammonium bromide-NaC1 solution (dissolve log of hexa-
Cuvettes may be recycled by soaking them in bleach for decyltrimethyl-ammonium bromide and 4.1 g of NaCl into
a few minutes and washing extensively with tap water, then 80 ml of H 2 0 with stirring with low heat. Adjust the final
in distilled water, and finally with 70% ethanol. Allow the volume to 100 ml and store at room temperature). Mix very
cuvettes to dry before using. Do not use acetone and do not well by flicking or shaking the tube (200 times). Incubate at
autoclave the cuvettes. 65C for 10 min, mixing 10 times at 5 min.
8. Add 780 p l of chloroform and isoamyl alcohol solu-
31.4.2.2. Adaptations for Electroporation tion (24:1), mix by flicking or shaking the tube 100 times,
There are additional considerations for some strains of bac- and centrifuge for 5 min at 9,000 X g in a microcentrifuge.
teria that are unable to withstand the change in conditions 9. Transfer 700 pl of the aqueous phase (upper layer) to
associated with preparing the cells (35,93, 129). For exam- a new tube. Add 700 p l of phenol-chloroform-isoamyl al-
ple, with Geobacter sulfurreducens an optimized electropora- cohol solution (24:24:1), mix 100 times, and centrifuge for
tion buffer was required consisting of 1 mM HEPES (pH 5 min at 9,000 X g in a microcentrifuge.
7.0), 1 mM MgC12, and 175 mM sucrose (35). In G. sul- 10. Transfer 600 p l of the aqueous phase (upper layer) to
furreducens the addition of MgClz prevents cell lysis, pre- a new tube and precipitate with 360 p l of isopropanol.
sumably by stabilizing the outer membrane. Sucrose was High-molecular-weight DNA and RNA should be visible.
added to abolish the change in osmolarity from the growth Pellet briefly in a microcentrifuge and wash once with 70%
media. Finally, high viability during storage at -70C re- ethanol.
quired that the cells be frozen in 10% DMSO and not glyc- 11. Dry briefly and dissolve in 50 p1of sterile water.
erol. Other variations may be required to allow this proce-
dure to be optimized for other nonenteric bacteria (93). 31.4.4. Controls and Precautions for Transformation
Strategies
31.4.3. Natural Competency In every transformation experiment it is important to in-
Natural competence is perhaps the easiest way to introduce clude both positive and negative controls. For the negative
DNA into bacteria. Well-studied systems for natural com- control a sample containing water or TE and lacking DNA
petence exist in H. pylori (130), Haemophilus influenzae should be included. The negative control is important to
( l l ) ,and Neisseria sp. (51, 123). Some of the cyanobacte- rule out contamination and defective selection plates. A
ria are also naturally competent (68). Interestingly, the positive control should be included where 10 picograms of
genes encoding natural competence are fairly widespread circular superhelical plasmid DNA is transformed to deter-
in many bacteria that are not generally considered natu- mine the transformation efficiency with the electroporation
rally competent (29). One possibility is that the appropri- and chemical competency procedures (lower-efficiency
ate signal or conditions that are needed to allow these competency procedures may require up to 1 nanogram of
organisms to be made naturally competent have not been DNA). A n additional control to consider is doping one of
31. Gene Transfer in Gram-Negative Bacteria 749

the samples with circular superhelical plasmid DNA. This set of strains and plasmids that allow cloning of genes, into
will control for the very real possibility that a contaminant conditionally replicating R6K plasmids, and mobilization
in your DNA preparation is inhibiting transformation. For with the RP4 system is available (88). These vectors are
example, residual salts, agarose, or enzyme present in the also capable of both positive and negative selection.
DNA sample could be affecting transformation efficiencies Conjugation occurs at highest efficiency on solid media.
in cells that would otherwise be highly competent. Recipient and donor cells can be mixed directly on non-
selective agar plates and allowed to mate from 1 h to
overnight. Transconjugants are isolated on a medium that
31.5. C O N J U G A T I O N selects for a marker encoded on the conjugal plasmid and a
Conjugation remains an important tool to introduce DNA marker found only in the recipient cells (usually carried on
into proteobacteria and cyanobacteria. The process of con- the chromosome). Controls include plating the donor and
jugation is very efficient and can reach 100% in many or- recipient cells individually. It is sometimes more conven-
ganisms. The high efficiency of this process likely over- ient to do a triparental mating. In a triparental mating
whelms the restriction and modification systems that might three strains are used; one donor strain contains a self-
otherwise limit other methods for introducing DNA. transmissible plasmid which can transfer to a second strain
Historically conjugation was critical in the mobilization of and mobilize the resident plasmid to transfer to the final re-
chromosomal genes for genetic mapping. The topic of con- cipient. A strategy is normally used to prevent the self-
jugal mapping has been covered in great detail in another transmissible plasmid from being maintained in the final re-
review (80). cipient (e.g., the use of a narrow-host-range conjugal
Conjugation requires direct cell-to-cell contact between plasmid). An advantage of triparental mating is that the
the plasmid-containing cell and the recipient bacterium. mobilizable plasmid does not need to be separately intro-
During conjugation, one strand of the plasmid is delivered duced into the special donor cell.
into the recipient cell in a process that resembles rolling-
circle DNA replication. Plasmids can be characterized as
either self-transmissible or mobilizable. Self-transmissible 31.6. BARRIERS T O D N A TRANSFER
plasmids encode all of the functions required to move be- Many bacterial host-encoded endonucleases can inhibit the
tween bacteria. Self-transmissible plasmids can also facili- transfer of DNA between bacteria. Multiple genetic and
tate the movement of other plasmids (and chromosomes) technical strategies exist for overcoming these barriers.
by a process called mobilization. Mobilizable plasmids con-
tain only a subset of the functions required for transfer, such 31.6.1. Restriction of Foreign DNA by E. cob
as a cis-acting origin of transfer (oriT) and perhaps the pro- Many standard laboratory strains of E. coli have multiple
teins (sometimes called Mob functions) that specifically systems that allow the destruction of foreign DNA. These
recognize this oriT. Mobilizable plasmids can move between systems can form a barrier to introducing DNA. E. coli K-12
bacteria only when specific transfer functions are provided strains naturally carry the EcoKI restriction system in the
in trans in the cell. Mobilizable plasmids can be produced de hsdRMS genes. DNA that is not modified by adenine
novo but also occur naturally. methylation on the appropriate recognition site will be
Conjugation can be used as a tool to deliver plasmids cleaved by the EcoKI system. Strains bearing mutations in
that are capable of being stably maintained in the target the hsdR gene are useful because they will not restrict DNA
host or as a tool to deliver suicide vectors that cannot without the appropriate methylation, but they will methyl-
replicate in the target host. Suicide vectors can be used to ate DNA at the appropriate recognition sites so that the
deliver transposons or they can be integrated into the host DNA can subsequently be used to propagate DNA for in-
chromosome if they contain sequences that are homologous troduction into hsdRt hosts. Strains bearing mutations in
to the chromosome. the hsdS gene will not restrict or methylate DNA at the ap-
Two widely used strains for mobilizing the transfer of propriate recognition site.
plasmids containing RP4 origins of transfer are SMlO and E. coli also has a series of systems for restricting DNA
S17-1 (117). To provide the RP4 transfer functions without that is methylated at specific cytosine and adenine posi-
having the normally self-transmissible RP4 vector itself tions. These systems are encoded by the mcrA, mcrBC, and
being transferred into recipient cells, the RP4 vector was in- mrr genes. Strains that are mutant in the mcr and mrr sys-
tegrated into the E. coli chromosome. Strain SMlO was cre- tems should be considered when difficulty is encountered in
ated from E. coli strain C600 by integrating the plasmid cloning DNA that has not been propagated in E. coli. The
RP4-2-Tet::Mu (kanamycin resistant and tetracycline sensi- mcrA gene is carried on the excisable genetic element el 4.
tive) into the chromosome by forcing its maintenance in Strains that are ef4-negative do not have the mcrA gene
the presence of the incompatible RP4 derivative pSR120 (14) (Table 2).
(tetracycline resistant); the cell was later cured of pSR120.
The integrated RP4-2-Tet::Mu plasmid imparts kanamycin 31.6.2. Barriers That Inhibit Moving DNA from
resistance and can excise and transfer only at a very low fre- E. coli into Other Bacteria
quency (- (1 17). The same strategy was used to pro- As mentioned, most bacteria encode restriction systems
duce S17-1 from the E. coli strain 294, in which an RP4-2- that can inhibit the introduction of exogenously replicated
Tet::Mu derivative was used where a T n 7 insertion DNAs. Various strategies can be used to limit the effects of
inactivated the kanamycin resistance gene (RP4-2-Tet::Mu these systems. In cases where the actual restriction system is
Kan::Tn7), thereby imparting a strain that is kanamycin not known, genetic strategies can be used to directly select
sensitive, but spectinomycin, streptomycin, and trimetho- strains with a lower restriction barrier (84, 129). It is also
prim resistant. E. coli strain BW20767 is a more recent con- possible to specifically mutate endonucleases that can be
struction containing the RP4-2-Tet::Mu Kan::Tn7 region in identified from an available genome sequence. It was found
a strain that is also capable of maintaining plasmids with that deleting the restriction endonuclease Yen1 vastly in-
the R6K origin of replication (Table 2) (88). A convenient creased the transformability of Yersina enterocolitica (66). A
750 MOLECULAR GENETICS

similar strategy was utilized for Vibrio cholerae strains, Many bacterial strains have resistance to various antibi-
whereby deleting multiple DNases allowed for a modest in- otics. Many mutations in the rpsL gene encoding the ribo-
crease in transformation efficiency (48). somal protein, small S12 subunit, can impart resistance to
Various technical strategies can also deal with barriers streptomycin (e.g., rpsL20, rpsWf, rpsLf 04, and rpsLf36).
that inhibit moving DNA from E. coli into other bacteria. Mutations in gyrA can impart resistance to nalidixic acid
One simple strategy is to use more DNA along with the (e.g., o r 2 9 and gyr96). Transposons often encode antibiotic
highest-efficiency mechanism to introduce DNA in order resistance determinants. Commonly used transposons in-
to overwhelm the restriction systems. This technique has clude Tn5 (encoding kanamycin resistance), Tn7 (encod-
been found to be successful in Neisseria sp. (51). There are ing streptomycin, spectinomycin, and trimethoprim resis-
anecdotal reports that the addition of sheared salmon sperm tance), Tn9 (encoding chloramphenicol resistance), and
DNA can allow a 100-fold increase in transformation eFi- T n l 0 (encoding tetracycline resistance). Transposons that
ciency by electroporation in Pseudomoms sp., possibly by have localized to a certain gene are indicated with a specific
acting as a nonspecific target for endogenous nucleases. nomenclature; for example, a TnlO insertion in the srlD
Because methylation can also make DNA a target for degra- gene would be indicated as srlD::TnlO.
dation, a property that is common in gram-positive bacte-
ria, it might be appropriate in some cases to propagate DNA 31.7.2. How To Get Bacterial Strains
in strains of E. coli that do not methylate DNA (Table 2). E. coli strains can be obtained from a variety of sources. The
Strategies also exist for methylating DNA in vitro, and it is most comprehensive collection of strains can be found at
formally possible that commercially available methylases the E. coli Genetic Stock Center at Yale University
such as the CpG methylase (MSssI) could protect DNA (http://cgsc2.biology.yale.edu). Large E. coli genome proj-
from restriction. A n interesting strategy that was used in H. ects are currently under way to isolate selectable null muta-
pylori involves the in vitro treatment of DNA with cell-free tions and conditional alleles for every open reading frame in
extracts from the recipient strain to apply the appropriate E. coli. Plasmid clones for every open reading frame in E.
methylation pattern (44). This strategy could provide an coli are also being constructed. These reagents should be
important option when working with strain backgrounds available in the near future from the E. coli Genome Project
with multiple restriction-modification systems. at University of Wisconsin-Madison (http://www.genome
.wisc.edu/resources.htm) and the E. coli National
BioResource Project in Japan (http://shigen.lab.nig.ac
31.7. BACTERIAL STRAINS .jp/ecoli/strain/top/top.jsp). Other sources of bacterial
For historical reasons many laboratories work with different strains include the American Type Culture Collection
derivatives of the original E. coli K-12 bacterium (9). Only (http://www.atcc.org), biotechnology companies, and inde-
two laboratory strains of E. coli have been sequenced, pendent investigators.
MG1655 and W3110 (15). MG1655 is an early derivative
of the original E. coli K-12 strain that was cured of the en-
dogenous A lysogen and Fertility plasmid. Many E. coli K-12 31.8. MEDIA
derivatives continue to be used including AB1157,
MC4100, and HBlOl (Table 2). However, there is a general 31 .&I. LB Medium
move towards doing experiments with MG1655. Dissolve 10 g of Bacto tryptone, 5 g of Bacto yeast extract,
and 10 g of NaCl in 1 liter of deionized HzO. Adjust to pH
31.7.1. Basics of F. coli Strain Genotypes 7.0 with 5 N NaOH and sterilize by autoclaving.
There are a few common genotypic features that should be
checked when choosing strains for experimentation. Genes 31.8.2. SOB Medium
in the lactose operon are often important in strains used for Dissolve 20 g of Bacto tryptone, 5 g of Bacto yeast extract,
cloning because many cloning vectors have special genetic and 0.5 g of NaCl in 950 ml of deionized HzO. Add 10 ml
requirements. As described above in the section on plas- of 250 mM KC1, adjust to pH 7.0 with 5 N NaOH, and ad-
mids, the LacZa fragment located in many useful multi- just the volume to 1 liter. Sterilize by autoclaving. Just prior
cloning sites requires that a special deletion derivative of to use add 5 ml of sterile 2 M MgClz per liter (sterilized by
lac2 reside in the cell called A(lac2)MIS. The A(kZ)M15 autoclaving) .
allele is often not carried at its natural position on the E.
coli chromosome. Most cloning strains have extensive dele- 31.8.3. SOC Medium
tions that remove the lactose operon on the chromosome SOC is identical to SOB medium except that it contains 20
such as the 97-kb A(ykfD-mhpD)169 deletion allele, which mM glucose. Glucose is added to SOB after autoclaving and
for historical reasons is called A(argF-lac) I69 (99). cooling as 20 ml of a 1 M filter-sterilized solution.
Typically the A(lacZ)M15 allele is carried on an F plasmid
derivative as in JP109 and XL1-Blue or in the chromosome 31.8.4. Psi Medium
on th; A-like bacteriophage lysogen $80 as in the case of Dissolve the following into 800 ml of deionized HIO: 5g of
DH5a (Table 2). Cloning strains may have mutations in Bacto yeast extract, 20 g of Bacto tryptone, 5 g of MgS04.
various restriction systems to remove barriers to cloning Adjust the pH to 7.6 with 5 M NaOH and adjust volume to
some DNAs (see above). Ideally cloning strains should also 1 liter. Aliquot and sterilize by autoclaving.
be endA-negative, as this gene encodes a stable nonspecific
endonuclease that contaminates plasmid DNA prepara- 31 3 . 5 . M 9 Medium
tions. When attempting to use expression plasmids that re- Make a lox stock of M9 salts by adding the following to
quire arabinose for induction, it is important to note that 800 ml of deionized HzO: 60 g of NaZHPO, (anhydrous), 30
the araD139 allele found in strains such as MC4100 causes g of KHzP04 (anhydrous), 5 g of NaC1, 10 g of NH4C1.
the strain to be sensitive to arabinose (52, 65). Adjust the volume to 1 liter and sterilize by autoclaving.
31. Gene Transfer in Gram-Negative Bacteria 751

Combine the 1OX salts with sterile distilled water to a 1X 31.8.9. Tetracycline-Sensitive-Selective(TSS) Agar
solution in 1 liter with CaC12 (10 ml of 0.01 M CaC12), Plates
MgS04 (1 ml of 1 M MgS04-7H20), vitamin Bl/thiamine Combine 2.5 g of Bacto tryptone, 2.5 g of Bacto yeast ex-
hydrochloride (0.5 ml of a 1% stock), and a carbon source tract, 5 g of NaCl, 7.5 g of agar, and 25 mg of chlorotetracy-
(10 ml of a 20% solution of sugar or glycerol). If needed, add cline HCl (Sigma) with 400 ml of H 2 0and autoclave. Cool
40 kg/ml of the required L-amino acid. to -45C and add sterile solutions of 5 g of NaH2P04-H20
31.8.6. M63 Medium in 100 ml of H 2 0 , 3 ml of 2 mg/ml fusaric acid, and 2.5 mi
of 20 mM ZnC12. TSS plates must be used within a few days
Make a lox stock of M63 salts by adding the following to of preparation (17, 88).
800 ml of deionized H 2 0 : 136 g of KH2P04, 20 g of
(NH4)2S04,5 mg of FeS04-7H20. Adjust the pH to 7.0
with KOH, adjust the volume to 1 liter, and sterilize by au- 31.9. BUFFERS
toclaving. Combine with sterile distilled water to a 1x so-
lution in l liter with MgS04 (lml of l M MgS04-7H20), 31.9.1. lnoue Transformation Buffer
vitamin Bl/thiamine hydrochloride (0.5ml of a 1% stock), Dissolve all of the following buffer constituents in 800 ml of
and a carbon source (10 ml of a 20% solution of sugar or pure H 2 0 : 55 mM MnC12 (10.88 g of MnC12-4H20), 15
glycerol). If needed, add 40 kg/ml of the required L-amino mM CaC12(2.20 g of CaC12-2Hz0), 250 mM KC1 (18.65 g
acid. of KC1). Add 20 ml of 0.5 M PIPES (piperazine-N,N'-bis
[a-ethanesulfonic acid]) to give a final concentration of 10
31.8.7. Supplements mM (prepare as a 0.5M solution by dissolving 15.1 g of
Antibiotics are prepared, stored, and added to media prior PIPES in 80 ml of Milli-Q H 2 0 [with pH adjusted to 6.7
to use according to Table 4. Stock solutions of 5-bromo-4- with 5 M KOH] and final volume brought to 100 ml, and
chloro-3-indolyl-f3-~-galactopyranoside (X-Gal) and 5- stored in 20-ml aliquots at -20C). Adjust the volume to 1
bromo-4-chloro-3-indolylphosphate (XP) are prepared at liter, filter sterilize, aliquot, and store at -20C.
20 mg/ml in dimethylformamide. Each is light sensitive and
should be stored in an amber glass bottle or wrapped in alu- 31.9.2. Tfbl
minum foil at -20C. A stock solution of isopropyl-P-D- Dissolve all of the following buffer constituents in 150 ml of
thiogalactopytanoside (IPTG) is 200 mg/ml in H 2 0 . This distilled H 2 0 ; 30 mM CH3COOK (0.588 g of CH3COOK),
solution is filter sterilized and stored in aliquots at -20C. 100 mM RbCl (2.42 g of RbCl), 10 mM CaC12 (0.294 g of
X-Gal (80 pl) and IPTG (8 pl) can be spread on plates just CaC12-2H20), 50 mM MnC12 (0.100 g of MnC12-4H20).
prior to use, but the solution should be allowed to dry com- Add 30 ml of glycerol (15% [vol/vol] final) and adjust the
pletely. XP and X-Gal are added to agar media at a concen- pH to 5.8 with dilute acetic acid. Adjust volume to 200 ml,
tration of 40 mg/l. filter sterilize, aliquot, and store at -20C.
31.8.8. Preparation of Agar Media 31.9.3. Tfbll
LB agar plates are prepared by including 15 g of Bacto agar Dissolve all of the following buffer constituents in 70 ml of
per liter of medium prior to autoclaving. Soft agar solutions distilled HzO: 10 mM MOPS (morpholinepropanesulfonic
for agar overlays are made by adding 8 g of Bacto agar per acid) (0.21 g of MOPS), 75 mM CaC12 (1.1g of CaClZ),10
liter of medium. Soft agar can be made and stored in 100- mM RbCl (1.1 g of RbCl). Add 15 ml of glycerol (15%
ml aliquots. Soft-agar aliquots can be carefully melted in the [vol/vol] final) and adjust the pH to 6.5 with dilute NaOH.
microwave prior to use (with the bottle cap removed) and Adjust the volume to 100 ml, filter sterilize, aliquot, and
then stored in a wet or dry bath for many days. store at -20C.

TABLE 4 Antibiotic concentrations for stock solutions and medium supplementation


Antibiotic Solvent Storage temp ("C) Stock concn (mg/ml) Plate concn (Fg/ml) Amount (ml/liter)
Ampicillin Watera - 20 100 100 1
Chloramphenici31 EtOHb - 20 10 30 3
Gentamicin WateP - 20 10 10 1
Kanamycin WateP - 20 50 50 1
Nalidixic acid NaOH' 4 5 20 4
Rifampicin MeOHd - 20 25 100 4
Spectinomycin WateP - 20 50 100 2
Streptomycin WateP - 20 50 50 1
Tetracycline' M~OH~ - 20 5 20 4
Trimethoprim D M S ~ - 20 50 100 2
'Filter sterilize the solution with a 0.2-pm-pore-size filter.
bSuspend in 100% ethanol.
'Suspend in 0.1 N NaOH, which converts the acid to the sodium salt.
dSuspend in 100% methanol; store in the dark.
eTetracycline is reported to be incompatible with magnesium ions and therefore should be tested before being used in minimal media
fSuspend in DMSO.
752 MOLECULAR GENETICS

31.1 0. FINAL CONSIDERATIONS caused by indirect suppression of rec- mutations. Proc.


Natl. Acud. Sci. USA 67:128-135.
Many of the compounds used in these experiments are 11. Barcak, G. J., M. S. Chandler, R. J. Redfield, and 1.-F.
known to be toxic, caustic, and/or carcinogenic. In all cases, Tomb. 1991. Genetic systems in Haemophilus influenwe.
before using a compound the material safety data sheet Methods Enxymol. 204:321-342.
should be read and all safety recommendations followed. In 12. Baudin, A., 0. Ozier-Kalogeropoulos, A. Denouel, F.
addition, laboratory personnel should work with their local Lacroute, and C. Cullin. 1993. A simple and efficient
environmental safety group to ensure proper disposal of all method for direct gene deletion in Saccharomyces cere-
waste. Investigators should consult the latest National visiae. Nucleic Acids Res. 21:3329-3330.
Institutes of Health guidelines for work with recombinant 13. Bender, R. A. 1981. Improved generalized transducing
DNA before initiating any experiments and work closely bacteriophage for Caulobucter crescentus.J. Bacteriol. 148:
with their local institutional biosafety committee. 734-735.
14. Berlyn, M. K., K. B. Low, and K. E. Rudd. 1996. Linkage
I owe thanks to Matthew DeLisa and Steve Winuns for comments map of Escherichia coli K-12. In E C. Neidhardt, R. I.
on the chapter and to Jose Carlos Huguet-Tupiu, Qiaojuan Shi, and Curtiss, J. L. Ingraham, E. C. C. Lin, K. B. Low, B.
Adum Parks, who trial ran experiments. Special thanks to the many in- Magasanik, W. S. Reznikoff, M. Riley, M. Schaechter, and
dividuals who provided important species-specific information: Andrew H. E. Umbarger (ed.), Escherichia coli and Salmonella:
Camilli, Maddalena Coppi, Andrew Darwin, Brian Kvitko, Nina Cellular and Molecular Biology, 2nd ed. American Society
Salama, James Shapleigh, Howard Shuman, Daniel Stein, Chris for Microbiology, Washington, DC.
Waters, Patrick Viollier, and Steve Winans. Work in the lab is s u p
15. Blattner, F. R., G. Plunkett 111, C. A. Bloch, N. T.
ported by the National Science Foundation (MCB-03153 16).
Perna, V. Burland, M. Riley, J. Collado-Vides, J. D.
Glasner, C. K. Rode, G. F. Mayhew, J. Gregor, N. W.
3 1.11. REFERENCES Davis, H. A. Kirkpatrick, M. A. Goeden, D. J. Rose, B.
Mau, and Y. Shao. 1997. The complete genome sequence
31 .I 1 . I . General References of Escherichia coli K-12. Science 277:1453-1474.
1. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. More, 16. Blomfield, I. C., V. Vaughn, R. F. Rest, and B. I.
J. G. Seidman, J. A. Smith, and K. Struhl. 1988. Current Eisenstein. 1991. Allelic exchange in Escherichia coli using
Protocols in Molecular Biology. Greene Publishing the Bacillus subtilis sucB gene and a temperature-sensitive
Associates Inc. and John Wiley & Sons, Inc., New York, pSClOl replicon. Mol. Microbiol. 5:1447-1457.
NY. 17. Bochner, B. R., H. C. Huang, G. L. Schieven, and B. N.
2. Miller. 1. H. 1991. A Short Course in Bacterial Genetics: A Ames.
~ ~.
~~~~ 1980. Positive selection for loss of tetracvcline re-
Labor&& Manual and Handbook for Escherichia coli and sistance. J. Bacteriol. 143:926-933.
Related Bacteria. Cold Spring Harbor Laboratory Press, 18. Bolivar, F., R. L. Rodriguez, P. J. Greene, M. C. Betlach,
Cold Spring Harbor, NY. H. L. Heyneker, and H. W. Boyer. 1977. Construction
3. Miller, J. H. (ed.). 1991. Methods in Enzymology, vol. 204. and characterization of new cloning vehicles. 11. A multi-
Bacterial Genetic Systems. Academic Press, San Diego, CA. purpose cloning system. Gene 2:95-113.
4. Sambrook, J., and D. W. Russell. 2001. Molecular 19. Boyd, D., D. S. Weiss, J. C. Chen, and J. Beckwith.
Cloning: A Laboratory Manual, 3rd ed. Cold Spring Harbor 2000. Towards single-copygene expression systems making
Laboratory Press, Cold Spring Harbor, NY. gene cloning physiologically relevant: lambda Inch, a sim-
5. Silhavy, T. J., M. L. Berman, and L. Enquist. 1984. ple Escherichia coli plasmid-chromosome shuttle system.
Experiments with Gene Fusions. Cold Spring Harbor J. Bacteriol. 182:842-847.
Laboratory Press. Cold Spring Harbor, NY. 20. Budzik, J. M., W. A. Rosche, A. Rietsch, and G. A.
O'Toole. 2004. Isolation and characterization of a general-
3 1 .I 1.2. Specific References ized transducing phage for Pseudomom mruginosa strains
6. Anderson, D. G., and S. C. Kowalczykowski. 1997. The PA01 and PA14. J. Bacteriol. 186:3270-3273.
translocating RecBCD enzyme stimulates recombination 21. Burrus, V., and M. K. Waldor. 2004. Shaping bacterial
by directing RecA protein onto ssDNA in a chi-regulated genomes with integrative and conjugative elements. Res.
manner. Cell 90:77-86. Micmbiol. 155:376-386.
7. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, 22. Cai, Y. P., and C. P. Wolk. 1990. Use of a conditionally
J. G. Seidman, J. A. Smith, and K. Struhl. 1988. Current lethal gene in Anabaena sp. strain PCC 7120 to select for
Protocols in Molecular Biology. Greene Publishing double recombinants and to entrap insertion sequences.
Associates Inc. and John Wiley & Sons, Inc., New York, J. Bacteriol. 172:3 138-3 145.
NY. 23. Campos, J. M., J. Geisselsoder, and D. R. Zusman. 1978.
8. Ayres, E. K., V. J. Thomson, G. Merino, D. Balderes, Isolation of bacteriophage MX4, a generalized transducing
and D. H. Figurski. 1993. Precise deletions in large bac- phage for Myxococcw xanthus. J. Mol. Bid. 119:167-178.
terial genomes by vector-mediated excision (VEX). The 24. Cangelosi, G. A., E. A. Best, G. Martinetti, and E. W.
trfA gene of promiscuous plasmid RK2 is essential for repli- Nester. 1991. Genetic analysis of Agrobacterium. Methods
cation in several gram-negative hosts. J. Mol. Bid. 230: Enzymol. 204:3 84-39 7.
174-185. 25. Casadaban, M. J. 1976. Transposition and fusion of the lac
9. Bachmann, B. J. 1996. Derivations and genotypes of some genes to selected promoters in Escherichia coli using bacte-
mutant derivatives of Escherichia coli K-12, p. 2460-2488. riophage lambda and Mu. J. Mol. Biol. 104541-555.
In F. C. Neidhardt, R. I. Curtiss, J. L. Ingraham, E. C. C. 26. Chang, A. C., and S. N. Cohen. 1978. Construction and
Lin, K. B. Low, B. Magasanik, W. S. Reznikoff, M. Riley, characterization of amplifiable multicopy DNA cloning
M. Schaechter, and H. E. Umbarger (ed.), Escherichia coli vehicles derived from the p15A cryptic miniplasmid.
and Salmonella: Cellular and Molecular Biology, 2nd ed. J. Bucteriol. 134:1141-1156.
American Society for Microbiology, Washington, DC. 27. Cherepanov, P. P., and W. Wackernagel. 1995. Gene dis-
10. Barbour, S. D., H. Nagaishi, A. Templin, and A. J. ruption in Escherichia coli: TcR and KmR cassettes with
Clark. 1970. Biochemical and genetic studies of recombi- the option of Flp-catalyzed excision of the antibiotic-
nation proficiency in Escherichia coli. 11. Recf revertants resistance determinant. Gene 158:9-14.
31. Gene Transfer in Gram-Negative Bacteria w 753

28. Chun, K. T., H. J. Edenberg, M. R. Kelley, and M. G. velopment of a transformation system. Mol. Microbiol. 5:
Goebl. 1997. Rapid amplification of uncharacterized 2547-2555.
transposon-tagged DNA sequences from genomic DNA. 49. Gay, P., D. Le Cog, M. Steinmetz, T. Berkelman, and
Yeast 13:233-400. C. I. Kado. 1985. Positive selection procedure for entrap-
29. Claverys, J. P., and B. Martin. 2003. Bacterial compe- ment of insertion sequence elements in gram-negative
tence genes: signatures of active transformation, or only bacteria. J. Bucteriol. 164:918-921.
remnants? Trends Microbiol. 11:161-165. 50. Glazebrook, J., and G. C. Walker. 1991. Genetic tech-
30. Close, T. J., D. Zaitlin, and C. I. Kado. 1984. Design and niques in Rhizobium meliloti. Methods Enzymol. 204:398-
development of amplifiable broad-host-range cloning vec- 418.
tors: analysis of the vir region of Agrobucterium tumefuciens 51. Gunn, J. S., and D. C. Stein. 1996. Use of a non-selec-
plasmid pTiC58. Plasmid 12:lll-118. tive transformation technique to construct a multiply re-
31. Cohen, M. F., J. C. Meeks, Y. A. Cai, and C. P. Wolk. striction/modification-deficientmutant of Neisseria gonor-
1998. Transposon mutagenesis of heterocyst-forming fila- rhoea. Mol. Gen. Genet. 251509-517.
mentous cyanobacteria. Methods Enzymol. 305:3-17. 52. Guzman, L.-M., D. Belin, M. J. Carson, and J.
32. Cohen, S. N., A. C. Chang, and L. Hsu. 1972. Non- Beckwith. 1995. Tight regulation, modulation, and high-
chromosomal antibiotic resistance in bacteria: genetic level expression by vectors containing the arabinose PBAD
transformation of Escherichia coli by R-factor DNA. Proc. promoter. J. Bucteriol. 177:41214130.
Natl. Acud. Sci. USA 69:2110-2114. 53. Haldimann, A., and B. L. Wanner. 2001. Conditional-
33. Conchas, R. F., and E. Carniel. 1990. A highly effkient replication, integration, excision, and retrieval plasmid-
electroporation system for transformation of Yersinia. Gene host systems for gene structure-functionstudies of bacteria.
87:133-137. J . Bucteriol. 183:6384-6393.
34. Copeland, N. G., N. A. Jenkins, and D. L. Court. 2001. 54. Hamilton, C. M., M. Aldea, B. K. Washburn, P.
Recombineering: a powerful new tool for mouse functional Babitzke, and S. R. Kushner. 1989. New method for gen-
genomics. Nut. Rev. Genet. 2:769-779. erating deletions and gene replacements in Escherichia coli.
35. Coppi, M. V., C. Leang, S. J. Sandler, and D. R. Lovley. 1.Bucteriol. 17146174622.
2001. Development of a genetic system for Geobucter sul- 55. Hanahan, D. 1983. Studies on transformation of Esche-
fuweducens. Appl. Enwiron. Microbiol. 67:3 180-3 187. richia coli with plasmids. J. Mol. Biol. 166557-580.
36. Costantino, N., and D. L. Court. 2003. Enhanced levels 56. Hanahan, D., J. Jessee, and F. R. Bloom. 1991. Plasmid
of lambda Red-mediated recombinants in mismatch repair transformation of Escherichia coli and other bacteria.
mutants. Proc. Natl. Acud. Sci. USA 100:15748-15753. Methods Enzymol. 204:63-113.
37. Court, D. L., J.A. Sawitzke, and L. C. Thomason. 2002. 57. Hava, D. L., and A. Camilli. 2001. Isolation and charac-
Genetic engineering using homologous recombination. terization of a temperature-sensitive generalized transduc-
Annu. Rev. Genet. 36:361-388. ing bacteriophage for Vibno cholera J. Microbiol. Methods
38. Court, D. L., S. Swaminathan, D. Yu, H. Wilson, T. 46:2 17-225.
Baker, M. Bubunenko, J. Sawitzke, and S. K. Sharan. 58. Herring, C. D., J.D. Glasner, and F. R. Blattner. 2003.
2003. Mini-lambda: a tractable system for chromosome Gene replacement without selection: regulated suppres-
and BAC engineering. Gene 315:63-69. sion of amber mutations in Escherichia coli. Gene 311:153-
39. Datsenko, K. A., and B. L. Wanner. 2000. One-step in- 163.
activation of chromosomal genes in Escherichia coli K-12 59. Heuermann, D., and R. Haas. 1998. A stable shuttle vec-
using PCR products. Proc. Natl. Acud. Sci. USA 97:6640- tor system for efficient genetic complementation of
6645. Helicobucter pylori strains by transformation and conjuga-
40. Davison, J., M. Heusterspreute, N. Chevalier, V. Ha-Thi, tion. Mol. Gen. Genet. 257619-528.
and F. Brunel. 1987. Vectors with restriction site banks. V. 60. Hong, J. S., and B. N. Ames. 1971. Localized mutagenesis
pJRD215, a wide-host-range cosmid vector with multiple of any specific small region of the bacterial chromosome.
cloning sites. Gene 51:275-280. Proc. Natl. Acud. Sci. USA 68:3158-3162.
41. de Lorenzo, V., M. Herrero, J.M. Sanchez, and K. N. 61. Inoue, H., H. Nojima, and H. Okayama. 1990. High ef-
Timmis. 1998. Mini-transposons in microbial ecology and ficiency transformation of Escherichia coli with plasmids.
environmental biotechnology. FEMS Microbiol. Ecol. 27: Gene 96:23-28.
21 1-224. 62. Jacobs, M., S. Wnendt, and U. Stahl. 1990. High-
42. Derbise, A., B. Lesic, D. Dacheux, J. M. Ghigo, and E. efficiency electro-transformation of Escherichia coli with
Carniel. 2003. A rapid and simple method for inactivating DNA from ligation mixtures. Nuckic Acids Res. 18:1653.
chromosomal genes in Yersinia. FEMS Immunol. Med. 63. Jasin, M., and P. Schimmel. 1984. Deletion of an essential
Microbiol. 38:113-116. gene in Escherichia coli by site-specificrecombination with
43. DeWitt, S. K., and E. A. Adelberg. 1962. The occurrence linear DNA fragments. J. Bucteriol. 159:783-786.
of a genetic transposition in a strain of Escherichia coli. 64. Kaiser, D., and M. Dworkin. 1975. Gene transfer to My-
Genetics 47:5 7 7-585. xobucterium by Escherichia coli phage P1. Science 187:653-
44. Donahue, J. P., D. A. Israel, R. M. Peek, M. J.Blaser, 654.
and G. G. Miller. 2000. Overcoming the restriction bar- 65. Khlebnikov, A., K. A. Datsenko, T. Skaug, B. L.
rier to plasmid transformation of Helicobucter pylori. Mol. Wanner, and J. D. Keasling. 2001. Homogeneous expres-
Microbiol. 37:1066-1074. sion of the P(BAD) promoter in Escherichia coli by consti-
45. Donohue, T. J., and S. Kaplan. 1991. Genetic techniques tutive expression of the low-affinity high-capacity AraE
in Rhodospirillaceae. Methods Enzymol. 204:459-485. transporter. Microbiology 147:324 1-3 247.
46. Ely, B. 1991. Genetics of Caulobucter crescentus. Methods 66. Kinder, S. A., J. L. Badger, G. 0. Bryant, J. C. Pepe, and
Enzymol. 204:372-384. V. L. Miller. 1993. Cloning of the Yen1 restriction en-
47. Finan, T. M., E. Hartweig, K. LeMieux, K. Bergman, donuclease and methyltransferase from Yersiniu enterocolit-
G. C. Walker, and E. R. Signer. 1984. General transduc- ica serotype 0 8 and construction of a transformableR-M+
tion in Rhizobium meliloti. J. Bacteriol. 159:120-124. mutant. Gene 136:271-275.
48. Focareta, T., and P. A. Manning. 1991. Distinguishingbe- 67. Koch, B., L. E. Jensen, and 0. Nybroe. 2001. A panel of
tween the extracellular DNases of Vibrio chokrae and de- Tn7-based vectors for insertion of the sfp marker gene or
754 w MOLECULAR GENETICS

for delivery of cloned DNA into Gram-negative bacteria 86. Marx, C. J., and M. E. Lidstrom. 2001. Development of
at a neutral chromosomal site. J. Microbiol. Methods 45: improved versatile broad-host-range vectors for use in
187-195. methylotrophs and other Gram-negative bacteria. Micro-
68. Koksharova, 0. A., and C. P. Wolk. 2002. Genetic tools biology 147: 2065-2075.
for cyanobacteria. Appl. Microbiol. Biotechnol. 58: 123- 87. Maxson, M. E., and A. J. Darwin. 2004. Identification
137. of inducers of the Yersinia enterocolitica phage shock pro-
69. Kolisnychenko, V., G. Plunkett 111, C. D. Herring, T. tein system and comparison to the regulation of the RpoE
Feher, J. Posfai, F. R. Blattner, and G. Posfai. 2002. and Cpx extracytoplasmic stress responses. J. Bacteriol.
Engineering a reduced Escherichia coli genome. Genome 186:41994208.
Res. 12:640-647. 88. Metcalf, W. W., W. Jiang, L. L. Daniels, S. K. Kim, A.
70. Kolter, R., M. Inuzuka, and D. R. Helinski. 1978. Trans- Haldimann, and B. L. Wanner. 1996. Conditionally
complementation-dependent replication of a low molecu- replicative and conjugative plasmids carrying lac2 alpha
lar weight origin fragment from plasmid R6K. Cell 15: for cloning, mutagenesis, and allele replacement in bac-
1199-1208. teria. Plasmid 35:1-13.
71. Kovach, M. E., P. H. Elzer, D. S. Hill, G. T. Robertson, 89. Metcalf, W. W., W. Jiang, and B. L. Wanner. 1994. Use
M. A. Farris, R. M. Roop 11, and K. M. Peterson. 1995. of the rep technique for -allele replacement to construct
Four new derivatives of the broad-host-range cloning vec- new Escherichia coli hosts for maintenance of R6K gamma
tor pBBRlMCS, carrying different antibiotic-resistance origin plasmids at different copy numbers. Gene 138:l-7.
cassettes. Gene 166:175-176. 90. Miller, J. H. 1992. A Short Course in Bacterial Genetics.
72. Kowalczykowski, S. C. 2000. Initiation of genetic recom- Cold Spring Harbor Laboratory Press, Cold Spring
bination and recombination-dependent replication. Trends Harbor, NY.
Biochem. Sci. 25:156-165. 91. Muramatsu, K., and H. Matsumoto. 1991. Two general-
73. Kowalczykowski, S. C., D. A. Dixon, A. K. Eggleston, ized transducing phages in Vibrio parahuemolyticus and
S. D. Lauder, and W. M. Rehrauer. 1994. Biochemistryof Vibrio alginolyticus. Microbiol. Immunol. 35:1073-1084.
homologous recombination in Escherichia coli. Microbiol. 92. Nichols, B. P., 0. Shafiq, and V. Meiners. 1998.
Reo. 58:401-465. Sequence analysis of TnlO insertion sites in a collection
74. Kuzminov, A. 1999. Recombinational repair of DNA of Escherichia coli strains used for genetic mapping and
damage in Escherichia coli and bacteriophage lambda. strain construction. J. Bucteriol. 180:6408-6411.
Microbiol. Mol. Biol. Reo. 63:751-813. 93. Nickoloff, J. 1995. Electroporation Protocols for Micro-
75. Lacks, S., and B. Greenberg. 1977. Complementary organisms, vol. 47. Humana Press, Totowa, NJ.
specificity of restriction endonucleases of Diplococcuspneu- 94. OConnor, K. A., and D. R. Zusman. 1983. Coliphage
moniue with respect to DNA methylation. J. Mol. Biol. P1-mediated transduction of cloned DNA from Esche-
114: 153-1 68. richia coli to Myxococcus xanthus: use for complementation
76. Lafontaine, D., and D. Tollervey. 1996. One-step PCR and recombinational analyses. J. Bucteriol. 155:317-329.
mediated strategy for the construction of conditionally ex- 95. OConnor, M., M. Peifer, and W. Bender. 1989. Con-
pressed and epitope tagged yeast proteins. Nucleic Acids struction of large DNA segments in Escherichia coli.
Res. 24:3469-3471. Science 244: 1307-1 3 12.
77. Lederberg, J., and E. L. Tatum. 1946. Gene recombina- 96. Oppenheim, A. B., A. J. Rattray, M. Bubunenko, L. C.
tion in Escherichia coli. Nature 158658. Thomason, and D. L. Court. 2004. In vivo recombi-
78. Lee, E. C., D. Yu, J. Martinez de Velasco, L. Tessarollo, neering of bacteriophage lambda by PCR fragments and
D. A. Swing, D. L. Court, N. A. Jenkins, and N. G. single-strand oligonucleotides. Virology 3 19: 185-189.
Copeland. 200 1. A highly efficient Escherichia coli-based 97. Orr-Weaver, T. L., J. W. Szostak, and R. J. Rothstein.
chromosome engineering system adapted for recombino- 1981. Yeast transformation:a model system for the study of
genic targeting and subcloning of BAC DNA. Genomics recombination. Proc. Natl. Acd. Sci. USA 78:6354-6358.
73:56-65. 98. Peters, J. E., and N. L. Craig. 2001. Tn7: smarter than
79. Link, A. J., D. Phillips, and G. M. Church. 1997. we thought. Nut. Reo./Mol. Cell Biol. 2:806-814.
Methods for generating precise deletions and insertions in 99. Peters, J. E., T. E. Thate, and N. L. Craig. 2003.
the genome of wild-type Escherichia coli: application to Definition of the Escherichia coli MC4100 genome by use
open reading frame characterization. J. Bacteriol. 179: of a DNA array. J. Bacteriol. 185:2017-2021.
6228-623 7. 100. Phillips, G. J. 1999. New cloning vectors with tempera-
80. Low, K. B. 1991. Conjugational methods for mapping ture-sensitive replication. Plasmid 41:78-81.
with Hfr and F-prime strains. Methods Enxymol. 204:43-62. 101. Posfai, G., V. Kolisnychenko, Z. Bereczki, and F. R.
81. Low, K. B., and D. D. Porter. 1978. Modes of gene trans- Blattner. 1999. Markerless gene replacement in Esche-
fer and recombination in bacteria. Annu. Reo. Genet. 12: richia coli stimulated by a double-strandbreak in the chro-
249-287. mosome. Nucleic Acids Res. 27:4409-4415.
82. Mandel, M., and A. Higa. 1970. Calcium-dependent bac- 102. Price-Carter, M., J. Tingey, T. A. Bobik, and J. R. Roth.
teriophage DNA infection. J. Mol. Biol. 53:159-162. 2001. The alternative electron acceptor tetrathionate
83. Margolin, P. 1987. Generalized transduction, p. 1154- supports B12-dependent anaerobic growth of Salmonella
1168. In F. C. Neidhardt, J. L. Ingraham, K. B. Low, B. enterica serovar Typhimurium on ethanolamine or 1,2-
Magasanik, M. Schaechter, and H. E. Umbarger (ed.), propanediol. J. Bucteriol. 183:2463-2475.
Escherichia coli and Salmonella typhimurium: Cellular 103. Priefer, U. B., R. Simon, and A. Puhler. 1985.
and Molecular Biology, vol. 2. American Society for Extension of the host range of Escherichia coli vectors by
Microbiology, Washington, DC. incorporation of RSFlOlO replication and mobilization
84. Marra, A., and H. A. Shuman. 1989. Isolation of a functions. 1.Bacteriol. 163:324-330.
Legionella pneumophila restriction mutant with increased 104. Raibaud, O., M. Mock, and M. Schwartz. 1984. A tech-
ability to act as a recipient in heterospecific matings. J. nique for integrating any DNA fragment into the chro-
Bacteriol. 17 1:2238-2240. mosome of Escherichia coli. Gene 29:231-241.
85. Marx, C. J., and M. E. Lidstrom. 2002. Broad-host-range 105. Rong, R., M. M. Slupska, J. H. Chiang, and J. H.
cre-lox system for antibiotic marker recycling in gram- Miller. 2004. Engineering large fragment insertions into
negative bacteria. BioTechniques 33: 1062-1067. the chromosome of Escherichia coli. Gene 336:73-80.
31. Gene Transfer in Gram-Negative Bacteria w 755

106. Rothmel, R. K., A. M. Chakrabarty, A. Berry, and A. 124. Sternberg, N. L., and R. Maurer. 1991. Bacteriophage-
Darzins. 1991. Genetic systems in Pseudomom. Methods mediated generalized transduction in Escherichiu coli and
Enzymol. 204:485-514. Salmonella typhimurium. Methods Enzymol. 204: 18-43.
107. Russell, C. B., and F. W. Dahlquist. 1989. Exchange of 125. Stibitz, S., W. Black, and S. Falkow. 1986. The con-
chromosomal and plasmid alleles in Escherichia coli by se- struction of a cloning vector designed for gene replace-
lection for loss of a dominant antibiotic sensitivity ment in Bordetella pertussis. Gene 50:133-140.
marker. J. Bacteriol. 171:2614-2618. 126. Szostak, J. W., T. Orr-Weaver, R. J. Rothstein, and
108. Russell, C. B., D. S. Thaler, and F. W. Dahlquist. 1989. F. W. Stahl. 1983. The double-strand-breakrepair model
Chromosomal transformation of Escherichia coli recD for recombination. Cell 33:25-35.
strains with linearized plasmids. J. Bacteriol. 171:2609- 127. Trulzsch, K., T. Sporleder, E. I. Igwe, H. Russmann,
2613. and J. Heesemann. 2004. Contribution of the major se-
109. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Mo- creted Yops of Yersinia enterocolitica0:8to pathogenicity
lecular Cloning: a Laboratory Manul, 2nd ed. Cold Spring in the mouse infection model. Infect. Immun. 725227-
Harbor Press, Cold Spring Harbor, NY. 5234.
110. Sauer, B. 1998. Inducible gene targeting in mice using 128. Uzzau, S., N. Figueroa-Bossi, S. Rubino, and L. Bossi.
the Crellox system. Methods 14:381-392. 2001. Epitope tagging of chromosomal genes in
111. Seifert, H. S., and M. So. 1991. Genetic systems inpath- Salmonella. Proc. Natl. Acud. Sci. USA 98:15264-15269.
ogenic Neisseriue. Methods Enzymol. 204:342-357. 129. Visick, K. G., and E. G. Ruby. 1996. Construction and
112. Sexton, J. A., and J. P. Vogel. 2004. Regulation of hy- symbioticcompetence of a lud-deletion mutant of Vibrio
percompetence in Legionellapneumophila.J. Bacteriol. 186: fischeri. Gene 175:89-94.
3814-3825. 130. Wang, Y., K. P. ROOS, and D. E. Taylor. 1993.
113. Shi, Z. X., H. L. Wang, K. Hu, E. L. Feng, X. Yao, Transformation of Helicobacter pylori by chromosomal
G. F. Su, P. T. Huang, and L. Y. Huang. 2003. metronidazole resistance and by a plasmid with a selec-
Identification of alkA gene related to virulence of Shigella table chloramphenicol resistance marker. J. Gen.
fixneri 2a by mutational analysis. World J. Gumoenterol. Mimobid. 139(Pt. 10)~2485-2493.
9:2720-2725. 131. Weiss, B. D., M. A. Capage, M. Kessel, and S. A.
114. Shizuya, H., B. Birren, U. J. Kim, V. Mancino, T. Benson. 1994. Isolation and characterization of a gener-
Slepak, Y. Tachiiri, and M. Simon. 1992. Cloning and alized transducing phage for Xunthomom campescris pv.
stable maintenance of 300-kilobase-pair fragments of cumpescris.J. Bucteriol. 176:3354-3359.
human DNA in Escherichia coli using an F-factor-based 132. Willson, T. A., and N. M. Gough. 1988. High voltage
vector. Proc. Natl. Acud. Sci. USA 89:8794-8797. E. coli electro-transformationwith DNA following liga-
115. Sik, T., J. Horvath, and S. Chatterjee. 1980. Gen- tion. Nucleic Acids Res. 16:11820.
eralized transduction in Rhizobium meliloti. Mol. Gen. 133. Winans, S. C., S. J. Elledge, J. H. Krueger, and G. C.
Genet. 1 7 8 511-5 16. Walker. 1985. Site-directed insertion and deletion muta-
116. Silverman, M., R. Showalter, and L. McCarter. 1991. genesis with cloned fragments in Escherichia coli. J. Buc-
Genetic analysis in Vibrio. Methods Enzymol. 204515- teriol. 161:1219-1 22 1.
536. 134. Woodcock, D. M., P. J. Crowther, J. Doherty, S.
117. Simon, R., U. B. Priefer, and A. Puhler. 1982. A broad Jefferson, E. DeCruz, M. Noyer-Weidner, S. S. Smith,
host range mobilization system for in vivo genetic engi- M. Z. Michael, and M. W. Graham. 1989. Quantitative
neering: transposon mutagenesis in gram-negative bacte- evaluation of Escherichia coli host strains for tolerance to
ria. Biotechnology 1:784-791. cytosine methylation in plasmid and phage recombi-
118. Singer, M., T. A. Baker, G. Schnitzler, S. M. Deischel, nants. Nucleic Acids Res. 17:3469-3478.
M. Goel, W. Dove, K. J. Jaacks, A. D. Grossman, J. W. 135. Yanisch-Perron, C., J. Vieira, and J. Messing. 1985.
Erickson, and C. A. Gross. 1989. A collection of strains Improved M13 phage cloning vectors and host strains:
containing genetically linked alternating antibiotic re- nucleotide sequences of the M13mp18 and pUC19 vec-
sistance elements for genetic mapping of Escherichia coli. tors. Gene 33:103-119.
Microbiol. Rev. 53: 1-24. 136. Yu, D., H. M. Ellis, E. C. Lee, N. A. Jenkins, N. G.
119. Skorupski, K., and R. K. Taylor. 1996. Positive selec- Copeland, and D. L. Court. 2000. An efficient recombi-
tion vectors for allelic exchange. Gene 169:47-52. nation system for chromosome engineering in Escherichia
120. Slauch, J. M., and T. J. Silhavy. 1991. Genetic fusions as coli. Proc. Natl. Acud. Sci. USA 976978-5983.
experimental tools. Methods Enzymol. 204:213-248. 137. Yu, D., J. A. Sawitzke, H. Ellis, and D. L. Court. 2003.
121. Smeets, L. C., J. J. Bijlsma, S. Y. Boomkens, C. M. Recombineering with overlapping single-stranded DNA
Vandenbroucke-Grauls, and J. G. Kusters. 2000. comH, oligonucleotides: testing a recombination intermediate.
a novel gene essential for natural transformation of Proc. Natl. Acud. Sci. USA 100:7207-7212.
Helicobacter pylori. J. Bacteriol. 182:3948-3954. 138. Zabarovsky, E. R., and G. Winberg. 1990. High effi-
122. Stalker, D. M., R. Kolter, and D. R. Helinski. 1979. ciency electroporation of ligated DNA into bacteria.
Nucleotide sequence of the region of an origin of replica- Nucleic Acids Res. 18:5912.
tion of the antibiotic resistance plasmid R6K. Proc. Natl. 139. Zhang, Y., F. Buchholz, J. P. Muyrers, and A. F.
A c d . Sci. USA 76:1150-1154. Stewart. 1998. A new logic for DNA engineering using
123. Stein, D. C. 1991. Transformation of Neisseria gonor- recombination in Escherichia coli. Nut. Genet. 20:123-128.
rhoea: physical requirements of the transforming DNA. 140. Zinder, N. D., and J. Lederberg. 1952. Genetic ex-
Can. j . Microbiol. 37:345-349. change in Salmonella. J. Bucteriol. 64:679-699.
32
Genetic Exchange in Gram-Positive Bacteriat
CHRISTOPHER J . KRISTICH. CHRISTINE E. SALOMON.
AND GARY M. DUNNY

32.1. INTRODUCTION AND OVERVIEW OF GENE TRANSFER


MECHANISMS AND THEIR APPLICATIONS ................... 757
32.1.1. Conjugation ......................................... 758
.
2. Transduction ........................................ 761
.
3. Transformation ...................................... 761
.
4. Applications ........................................ 762
32.2. GENE TRANSFER IN VARIOUS GROUPS OF GRAM-POSITIVE
BACTERIA .............................................. 762
32.2.1. Streptococci ......................................... 762
32.2.1.1. Plasmids .................................... 763
.
2. Conjugative Elements ........................... 763
.
3. Bacteriophage and Transduction ................... 763
.
4. Transformation ............................... 764
.
5. Applications of Gene Transfer.................... 765
32.2.2. Staphylococci ....................................... 767
32.2.2.1. Plasmids .................................... 767
.
2. Conjugative Elements ........................... 767
.
3. Bacteriophage and Transduction ................... 767
.
4. Transformation ............................... 768
.
5. Applications of Gene Transfer.................... 768
32.2.3. Enterococci ......................................... 769
32.2.3.1. Plasmids .................................... 769
.
2. Conjugative Elements ........................... 770
.
3. Bacteriophage and Transduction ................... 771
.
4. Transformation ............................... 771
.
.5 Applications of Gene Transfer .................... 771
32.2.4. Lactococci and Related Food-Fermenting Bacteria .............. 772
32.2.4.1. Plasmids .................................... 773
.
2. Conjugative Elements ........................... 774
.
3. Bacteriophage and Transduction ................... 774
.
4. Transformation ............................... 774
.
5. Applications of Gene Transfer.................... 775
32.2.5. Bacilli ............................................. 776
32.2.5.1. Plasmids .................................... 776
.
2. Conjugative Elements ........................... 776
.
3. Bacteriophage and Transduction ................... 777
.
4. Transformation ............................... 777
.
5. Applications of Gene Transfer.................... 778
32.2.6. Clostridia .......................................... 779
32.2.6.1. Plasmids .................................... 779
.
.2 Conjugative Elements ........................... 779
.
3. Bacteriophage and Transduction ................... 779
.
4. Transformation ............................... 779
.
5. Applications of Gene Transfer.................... 780
32.2.7. Mycobacteria ........................................ 780
32.2.7.1. Plasmids .................................... 780
.
.2 Conjugative Elements ........................... 780
.
3. Bacteriophage and Transduction ................... 780
.
4.Transformation ............................... 781
.
5. Applications of Gene Transfer.................... 781
A chapter titled Gene Transfer in Gram-Positive Bacteria in Methods for General and Mokcular Bacteriology was authored
by Simon M . Cutting and Philip Youngman. This chapter is a completely revised. reorganized. and expanded presentation by
the current authors.

75 6
32. Genetic Exchange in Gram-Positive Bacteria 757

....................................... 782
32.2.8. Actinomycetes
.................................... 782
32.2.8.1. Plasmids
........................... 782
.2. Conjugative Elements
................... 783
.3. Bacteriophage and Transduction
.4. Transformation ............................... 783
.5. Applications of Gene Transfer .................... 785
32.3. REFERENCES ............................................ 786

into two main groups on the basis of DNA base composi-


32.1. INTRODUCTION AND OVERVIEW OF tion. In terms of gene regulation, promoter structure/func-
GENE TRANSFER MECHANISMS AND THEIR tion, transcription, and translation, there are major differ-
APPL I CATI 0NS ences between the high-GC and low-GC groups
The gram-positive (gramf) bacteria represent an extremely resulting from codon usage, DNA secondary structure, and
diverse group of organisms (Fig. l ) , many of which are of other phenomena directly related to DNA base composi-
significant medical, environmental, or industrial impor- tion. As noted later in this chapter, certain members of the
tance. From the basic biology perspective, it is noteworthy high-GC group carry out forms of conjugation whose mech-
that both horizontal gene transfer and the existence of anisms may be fundamentally different from that observed
DNA as the genetic material were first discovered in an in other bacteria. The other overriding issue affecting the
organism from this group, Streptococcus pneumoniae (7, manner in which natural and engineered genetic changes
149). The three known mechanisms of gene transfer in bac- occur is whether the organism in question is naturally com-
teria (conjugation, transformation, and transduction) are petent for genetic transformation; this affects not only the
widespread in gram+ bacteria, although the relative preva- entry of transferred DNA into a new host but also the sub-
lence of each of these processes in different organisms is sequent recombination events that ultimately determine
highly variable, and in most cases not clearly documented. whether a heritable change results from intercellular trans-
These differences in the preferred modes of genetic trans- fer of genetic material. While the ability to undergo genetic
fer have significant implications both for the natural evolu- transformation is encoded within the core genome of com-
tion of the organisms and for the development of successful petent organisms, both conjugation and transduction abili-
approaches to their genetic manipulation in the laboratory. ties are generally encoded by accessory elements (plasmids,
Thus, when designing schemes for manipulation of gram+ conjugative transposons, and bacteriophages) which have a
bacteria, a useful rule of thumb is that approaches known to degree of genetic autonomy from their bacterial hosts.
be successful in the most closely related gram bacteria are These elements may be viewed as parasitic selfish DNAs
the most likely to work (see Fig. l), although there certainly whose long-term evolutionary goals may not completely
are exceptions. overlap with those of the host bacteria, although a prepon-
There are several important operational constraints derance of evidence clearly indicates that lateral transfer
strongly affecting natural gene transfer events and the use of events mediated by mobile elements have had a tremendous
genetic transfer methods in the laboratory. The first impor- influence on microbial evolution and on how phylogenetic
tant consideration is that the gram+ bacteria can be divided relationships are determined (89, 99).

Streptomyces
coelicolor
P

Leuconostoc
mesenteroides

I Lactobacillus

Bacillus
subtilis4r Enterncoccus
Streptococcus
pyogenes
faecalis Streptococcus

Staphylococcus
aureus
pneumoniae

FIGURE 1 Unrooted phylogenetic tree of selected gram+ bacteria based on 16s rRNA se-
quences. The sequences for each organism were obtained from the NCBI database and subjected to
phylogenetic analysis assuming maximum parsimony and bootstrapping (500 times resampling).
The length of the lines between the species corresponds to phylogenetic distance.
758 MOLECULAR GENETICS

This chapter presents an overview of gene transfer sys- siderable conservation in the protein components of the
tems and their applications in several important groups of DNA processing systems and coupling proteins of gram' and
gram' bacteria. In the following paragraphs, a summary of gram- systems, whereas the channel components seem to be
each of the known mechanisms of transfer as they have less related and are largely uncharacterized in gram+ bacte-
been described for gram+ genera is presented. In the re- ria. Conjugative elements transferred by such systems con-
maining sections of the chapter, transfer systems and their tain an on'T (origin of transfer) DNA sequence that serves as
application in various groups of related gram+ bacteria are a site for binding and strand-specific and nucleotide-specific
summarized. We have provided extensive, though not com- nicking of the DNA strand destined for transfer. Small non-
prehensive, citations both to the primary literature and to conjugative plasmids often carry on'T and DNA processing
review articles. The use of various transfer systems for ge- components that enable them to be efficiently mobilized by
netic manipulation of gram+ bacteria is described, and rep- coresident conjugative plasmids whose transfer machinery
resentative protocols are presented as illustrations of gen- can interact with the relaxosome complex of the small plas-
eral approaches that can be applied to gram+ bacteria. In mid. Mobilization of small plasmids in the laboratory can
addition, a table describing selected plasmids (Table 1) that also be effected by insertion of oriT sequences using recom-
have found use in genetic manipulation of gram' bacteria is binant DNA techniques; these manipulations can be very
provided. While it is not feasible to provide complete pro- useful in genetic engineering applications for bacterial
tocols or comprehensive plasmid lists for every individual species refractory to introduction of DNA by transforma-
organism discussed, the material presented provides re- tion, a common problem for researchers studying gram' bac-
sources to enable investigators to identify or design the ap- teria.
propriate protocols for transfer studies with their respective In the past decade, analysis of conjugation systems in the
organism of choice. In many cases, genetic tools used for high-GC gram+ genera, particularly the filamentous organ-
manipulation of one gram' species can be readily adapted isms and the mycobacteria, has suggested that these organ-
for use in others, particularly among the low-GC gram' isms carry out novel forms of transfer utilizing one or more
species. For example, tools useful for genetic manipulation sets of molecular machinery which show no apparent evo-
of the low-GC gram+ Listeria monocytogenes, although not lutionary relatedness to the more conventional systems de-
discussed explicitly in this chapter, have been derived from scribed above. In Streptomyces spp., a number of conjugative
tools applicable in other low-GC gram+ species, including elements have been identified, including small and large el-
some described herein (130). ements, high- and low-copy-number elements, and linear
and circular elements; some of these are described later in
32.1 . l . Conjugation this chapter. In some cases a single protein encoded by the
Conjugation in bacteria is defined as a lateral transfer mech- mobile element has been shown to be sufficient for transfer;
anism where the transmission of genetic material from one these Tra proteins are related to septa1 DNA translocator
cell to another requires direct cell-to-cell contact. During proteins of the SpoIIIE/FtsK family. This suggests that the
conjugation a channel is formed between the donor and re- core genome of these organisms encodes functions involved
cipient cells. This channel serves as a direct conduit for the in mycelial fusion events that must be required to form a
transferred DNA, such that the process cannot be disrupted functional mating contact used by the Tra protein to effect
by exogenous nucleases. Transfer is generally unidirectional transfer. A small cis-acting DNA site called clt has been im-
(although multiple rounds of transfer may give the appear- plicated in transfer of certain plasmids (244). Although clt
ance of bidirectionality) and requires viable, metabolically would appear to play a role similar to oriTs in other systems,
active donor cells. Conjugation has a tremendous impact on attempts to demonstrate strand-specific nicking have been
natural evolution of microorganisms (and probably higher unsuccessful, and it is possible that double-stranded DNA is
organisms as well) because the transfer systems can allow ge- transferred by these systems. Very recently Derbyshire and
netic exchange between very distantly related species, be- colleagues have presented striking evidence for a novel
cause it can occur very efficiently under optimal conditions, form of conjugation in mycobacterial species (123, 394,
and because the number of genes transferred can be very 395). As in the Streptomyces systems, the evidence suggests
large. While many conjugative transposons and some con- that the core genome of these organisms may encode func-
jugative plasmids can be maintained stably in a very broad tions that facilitate mating pair formation, and that the
range of bacterial hosts, other conjugative plasmids show a DNA processing mechanism( s) is fundamentally distinct
very narrow host range; the latter elements may still effect from the oriT-based systems.
lateral transfer across species barriers by carrying transpos- Significantly, the genes encoding the mating pair forma-
able elements or by mobilizing plasmids that may survive in tion machinery of several gram- conjugation systems have
a new host independently of the conjugative element itself. been shown to belong to the type IV protein secretion sue
In gram+ bacteria, there seem to be at least two fundamen- perfamily (51). Type IV systems are involved in export of
tally different forms of genetic exchange that fit the defini- virulence factor proteins from bacterial pathogens into host
tion of conjugation. As described in a recent review (150), a target cells (61). Even among the better-studied gram'
variety of plasmids and conjugative transposons identified in transfer systems, very little is known about the mechanism of
gram' species encode three essential transfer functions that mating pair formation or about whether these systems could
are also found in well-characterized conjugative elements be involved in export of virulence factors. This is an unad-
from gram-negative (gram-) organisms. These include a dressed question that is of great importance relative to the
proteinaceous DNA processing machinery that specifically genetic basis of virulence in these organisms. Interestingly, a
nicks and unwinds the transferred DNA while remaining recent genetic study of mycobacterial conjugation showed
attached to the 5' end of the nick. The resulting relaxosome that genes important for conjugation mapped to a locus be-
complex interacts with a coupling protein to guide the trans- lieved to be essential for secretion of virulence factors (123),
ferred DNA to a mating channel extending from the inner suggesting that the functional link between conjugative
cytoplasmic membrane through the cell envelopes of both mating pair formation and virulence may extend through all
the donor and recipient. Sequence analysis has shown con- the diverse transfer systems identified in gram' microbes.
TABLE 1 Representative plasmids of use for genetic manipulation of gram+ bacteriaa
Gram+ Gram-
Plasmid Gram+ origin Gram- origin
selectable selectable Property Host Reference(s)
name of replication of replication range
marker marker

pAM40 1 pIP501 Cm p15A (pACYC184) Cm, Tc Shuttle vector for general cloning in LGC LGC 406
pAT18 PAMPI Em ColEl (pBR322) Em Shuttle vector for general cloning in LGC; carries RK2 oriT LGC 378
for mobilization to gram+ hosts from E. coli
pAT28 PAMPI SP ColEl (pBR322) SP Shuttle vector for general cloning in LGC; carries RK2 oriT LGC 377
for mobilization to gram+ hosts from E. coli
pCJK47 Suicide Em pWVOl (requires Em Conjugative delivery vector for allelic exchange carrying E. faecalis 210a
provision of gram+ counterselectable marker
RepA in trans)
pCL52.1 pE 194 Tc pSCl0l SP TS; delivery vector for allelic exchange in Stuphylococcus S 230
pCL84 Suicide Tc pSCl0l SP Site-specific integration into attB used by phage L54a in S 22 1
S. uureus
pCN series pT181 or SP, Tc, Cm, ColEl (pUC19) AP Interchangeable cassette-based shuttle vectors for S 54
pI258 Kn, or Em complementation, operon fusions in Sta~hylococcus
pEG1661- Suicide Cm, SP, ColEl AP Vectors for ectopic single-copy integration into B. subtilis B 152
pDG1664 or Em umyE locus for complementation, operon fusion analysis
series
pDGl728-
pDG1731
series
pDL276 pVA380-1 Kn ColEl (pUC18) Kn Shuttle vector for general cloning in LGC; RCR replicon LGC 104
pDL278 pVA380- 1 SP ColEl (pUC18) SP Shuttle vector for general cloning in LGC; RCR replicon LGC 219
pFW series Suicide SP, Tc, Cm, ColEl (pUC18) SP, Tc, Cm, Delivery vector for gene inactivation or allelic exchange LGC 306
Kn, or Em Kn, or Em in LGC
pG+host4 pWV0l Cm, Em pWV0l Cm, Em TS; delivery vector for allelic exchange in LGC LGC 253
pG+host5 pWV0l Cm, Em ColEl (pBR322) Cm, Em TS; delivery vector for allelic exchange in LGC; contains LGC 30
pBR322 origin for replication at 37C in E. coli
pIJ486 pIJlOl Nm, Kn, Th Promoter-probe plasmid A 397
pIJ698 SCP2 Th, HY p15A (RK2) Vm, Kn Bifunctional E. coli/Streptomyces replacement vector A 199
PWO SCP2* Nm Unstable Streptomyces plasmid used to cure SCP2* A 370
derivatives from host strains
pIJ941 SCP2* Th, HY SCP2*-derived Streptomyces vector with transfer functions A 240
used for cloning and analysis
pJIR series pIP404 Cm or Em ColEl (pUC18) Cm or Em Shuttle vectors for general cloning in C. pegringens; some C 243
versions carry RP4 oriT for mobilization from E. coli
pJOE2577 pSG5 Th, Kn ColEl (pUC19) AP TS transposon delivery shuttle vector containing the high- A 388
frequency transposase gene tnpA
(Continued on next page)
TABLE 1 Representative plasmids of use for genetic manipulation of gram+ bacteriaa (Continued) rn

Gram+ Gram-
Gram- r
Plasmid
name
Gram+ origin
of replication
selectable
marker
Gram- origin
of replication
selectable
marker
Property Host
range
Reference(s) k%
L
pKCll39 pSG5 Am ColEl (pBR322) Am, lac2 Bifunctional conjugative E. coli/Saeptomyces plasmid A 28 rn
i
pKCl2 18 SCP2" Am ColEl (pUC18) lac2 Bifunctional conjugative E. coli/Saeptomyces plasmid A 28
G
pMSP3535 PAMPI Em ColEl (pSP73) Em Shuttle vector for inducible gene expression in LGC; nisin- LGC 41
inducible promoter for operon fusions
pMSP3545 PAMPI Em ColEl (pSP73) Em Shuttle vector for inducible gene expression in LGC; nisin- LGC 41
inducible promoter for gene fusions
pMUTIN Suicide Em ColEl AP Insertional inactivation of Bacillus genes; operon fusions of B 383
inactivated gene to lacZ; IPTG-regulatable expression of
downstream genes
pOJ446 SCP2* Am ColEl (pUC) Bifunctional conjugative E. coli/Streptomyces cosmid vector A 28
pPR27 pAL5000 Investigator p15A (pACYC184) Gm TS; delivery vector for allelic exchange or transposon M 298
determined mutagenesis in mycobacteria
pSET152 Am ColEl (pUC18) lac2 Conjugative vector that integrates into the chromosome at A 28
the +C31 attachment site
pSG series pC194 or Cm or Sp ColEl (pSK-) AP Shuttle vectors (or delivery vectors for integration at amyE B 116,228
suicide locus) for construction of gfe,cfp, yfp, or dsRed fluorescent
fusion proteins in Bacillus
pSWEET Suicide Cm ColEl AP Vector for ectopic single-copy integration into B. subtilis B 24
amyE locus for xylose-inducible gene expression
pTCV-lac PAMW Em, Kan p15A (pACYC184) Em, Kan Shuttle vector for construction of operon fusions to lac2 in LGC 310
LGC; carries RK2 oriT for mobilization to gram+ hosts
from E. coli
pVA838 PAMPI Em p15A (pACYC184) Em, Cm Shuttle vector for general cloning in LGC LGC 249
pVE6 16 Th ColEl (pBR322) Am Integrating cosmid vector A 246
pYUB657 Suicide HY ColEl AP Delivery vector for allelic exchange in mycobacteria carrying M 295
sacB counterselectable marker
'Abbreviations: LGC, low-GC gram' bacteria; S, staphylococci; B, bacilli; M, mycobacteria; C, clostridia; A, actinomycetes; Ap, ampicillin resistance; Kn, kanamycin resistance; Cm, chloramphenicol resistance; Sp, spectino-
mycin resistance; Em, erythromycin resistance; Gm, gentarnicin resistance; Tc, tetracycline resistance; Th, thiostrepton resistance; Nm, neomycin resistance; Vm, viornycin resistance; Am, apramycin resistance; Hy, hygromycin re-
sistance; bla, p-lactamase; ds, double stranded.
32. Genetic Exchange in Gram-Positive Bacteria D 761

32.1.2. Transduction ciency with which natural transformation occurs among


Transduction is a form of lateral genetic transfer mediated various strains of competent species varies considerably.
by bacteriophage. Traditionally, transduction is envisioned However, genome sequencing projects have revealed appar-
to occur as a result of errors in packaging of phage particles ently functional competence genes in organisms which
during a lytic phage replication cycle. In generalized trans- have not been shown to undergo natural transformation
duction, a random segment of the bacterial chromosome is (66), and recent reports suggest that even heavily studied
inserted into a nascent phage particle. Upon release from organisms like Escherichia coli may be naturally trans-
the infected donor bacterium by lysis, this transducing par- formable under certain conditions (252), so the prevalence
ticle can bind to a new bacterial host cell displaying the ap- of natural transformation may be higher than is currently
propriate surface receptor for phage attachment and inject appreciated.
its DNA, which can recombine into the host chromosome Given the differences in the cell envelope of gram+ and
or, in the case of transduced plasmids, can replicate au- gram- bacteria, the existence of significant differences in
tonomously. Specialized transduction involves an aberrant the transformation machinery utilized by these two groups
excision of an integrated prophage from the chromosome is perhaps not surprising. However, the competent bacterial
during induction of a lysogenic bacterial strain. The excised species examined to date seem to utilize a core group in-
DNA contains a segment of chromosomal DNA adjacent to cluding binding proteins, nucleases, channel-forming pro-
the phage attachment site, and a segment of the phage teins, and nucleoside triphosphate-dependent helicases to
genome is usually left behind in this process, again render- move transforming DNA across the cytoplasmic membrane
ing the transducing particle defective. In specialized trans- into the recipient cell. Among these DNA transport com-
duction, the chromosomal genes linked to the phage at- ponents there is significant conservation of function and
tachment site are transmitted at significant frequencies amino acid sequence across both groups of bacteria. In
since nearly all transducing particles are generated during gram- bacteria, pili are generally parts of the transforma-
the initial excision of the prophage following induction of tion apparatus, and they may serve as the conduit for import
a lysogen. While numerous examples of transduction of of the transforming DNA across the outer membrane into
gram+ bacteria have been reported, it is somewhat surpris- the periplasmic space. Some of the components of the com-
ing that refined transduction systems for allelic exchange petent cell transformation apparatus also show similarity to
and chromosomal gene mapping have only been fully de- conjugative DNA export components and other secretion
veloped and utilized with a small number of species, such systems (55). In competent cell transformation of both B.
as Bacillus subtilis and Staphylococcus aureus. O n the other subtilis and Streptococcus pneumoniae, the two most thor-
hand, the upswing in bacterial and bacteriophage genome oughly studied systems of gram species, the transforming
sequencing projects in recent years has sparked a new ap- material is bound by a secreted cell wall DNA binding pro-
preciation for the important role of phages in natural lateral tein and then subjected to both single-stranded and double-
transfer events and in the evolution of many industrially stranded nucleolytic attack during its transport into the
and medically important bacteria, including numerous cytoplasm. Ultimately, a double-stranded cleavage point
gram species. In addition to transduction of host genes, seems to serve as the substrate for import, and as the DNA
the related phenomenon of phage conversion (where genes is taken into the recipient cell, one strand is completely di-
actually carried by the wild-type temperate phage change gested as the opposite strand is imported. In contrast to
the phenotype of the host bacterium) plays an essential role some gram- systems, uptake of DNA by competent gram+
in the evolution of bacteria, especially in the case of strains is not sequence specific, but any heterologous DNA
pathogens. Phage-mediated inversions, integrations, and that enters is unlikely to recombine into the chromosome
other forms of DNA recombination can also impact ge- and is subject to rapid degradation. Likewise, autonomously
nomic evolution in bacteria; since the numbers of phage replicating plasmids are taken up efficiently by competent
species are probably orders of magnitude higher than the bacteria but are subject to complete degradation of one
number of bacterial species, phage-mediated transfer events strand and partial degradation of the other; thus, these ele-
have been estimated to occur at rates approaching 1015/s in ments transform with multihit kinetics, which places con-
the marine environment, where the phage titers exceeding straints on certain recombinant DNA manipulations such
107/ml make these elements the most abundant biological as construction of genomic libraries. O n the other hand,
entities on the planet (293). Because phage generally ex- chromosomal DNA from a strain closely related to the re-
hibit a high degree of specificity for host receptors, one pre- cipient is stably incorporated extremely efficiently, because
diction is that natural transfer events mediated by phage the induction of competence (see next paragraph) generally
might have their largest impact on the evolution of closely results in increased expression of homologous recombina-
related bacterial species or strains. While this is probably tion functions. This makes natural transformation the
true, clearly there is significant genetic exchange between method of choice for allelic exchange and insertion/dupli-
distantly related phages infecting distinct bacterial hosts (4, cation mutagenesis (83) in competent organisms.
93, 163, 164). A n important aspect of competent cell transformation
in gram+ species is that development of competence is gen-
erally subject to control by peptide-mediated quorum sens-
32.1.3. Transformation ing mechanisms (65). In addition to serving as the first ex-
Lateral genetic transfer mediated by the uptake and recom- perimental system for demonstration of horizontal genetic
bination of naked DNA into the recipient cell genome (i.e., transfer by DNA-mediated transformation, S. pneumonia is
transformation) was the first form of horizontal transfer the organism for which the first direct experimental evi-
documented experimentally (7, 149). Although natural or dence for release of a bacterial signaling molecule into the
competent cell transformation has been described for growth medium was presented (373). More recently, analy-
more than 40 microbial species, including gram+ and gram- ses of cell-cell signaling in the control of competent cell
organisms, investigators have failed to demonstrate trans- transformation of B. subtilis and of several streptococcal
formation for a much larger number of species, and the effi- species have contributed very significantly to the overall
762 MOLECULAR GENETICS

understanding of multicellular microbial activities (15, 65, replication in gram- bacteria generally do not replicate in
83, 154). Interestingly, transformation and its stimulation gram' bacteria. Therefore, common gram- replicons (e.g.,
by peptide-mediated cell-cell signaling occur efficiently in pUC) are frequently used as suicide plasmids in gram+ bac-
biofilms. Even more striking are recent results suggesting teria for the delivery of DNA as substrates for homologous
that competence-stimulating peptides themselves are also recombination. Additionally, shuttle plasmids (i.e., plas-
involved directly in biofilm development (83), and that mids capable of replication in both a gram- and a gram+
both biofilm formation and competence development are species) are typically constructed using molecular tech-
actually components of global stress response pathways in niques to fuse a gram+ replicon with a gram- replicon,
these organisms (65, 67, 83, 84). yielding a recombinant plasmid carrying multiple origins of
In terms of the effects of natural transformation on evo- replication that is therefore capable of replication in both
lution via lateral transfer, clearly natural transformation classes of bacteria. This approach allows investigators to use
contributes substantially to processes such as antigenic vari- the vast array of tools available for general manipulation of
ation, and antibiotic resistance dissemination within DNA in E. coli, prior to insertion of the modified DNA
species and between closely related species, while being less into the gram' organism of interest for analysis. Many such
involved in lateral transfer of large amounts of DNA across shuttle vectors have been constructed using the broad-host-
substantial phylogenetic gaps. Finally, of note is the fact range gram+ replicon pAMP 1. Accordingly, these vectors
that the development of physical methods for introduction can often be applied in a wide spectrum of different gram'
of DNA into living bacterial cells by techniques such as species, even though their use may have been described
treatment with divalent cations, protoplast transformation, only for one or a small number of species. Other tools used
and especially electroporation has been at least partially for genetic analysis of gram- species can sometimes be
successful in many, if not most, strains examined. None of adapted for use in gram+ bacteria. For example, many re-
these techniques shows much resemblance to natural trans- porter genes that are used to analyze gene expression in
formation. Recently the molecular basis of electroporation gram- bacteria can also be used in gram' bacteria by
was investigated in phospholipid bilayers, with the main cloning them into shuttle vectors and creating operon h-
conclusion being that the high-voltage pulse across the sions with promoters of interest. However, the machinery
membrane drives the formation of electric field gradients at for gene expression in gram+ bacteria (particularly for trans-
the lipid/water interface. This results in penetration of the lation) tends to exhibit somewhat more stringent require-
bilayer by water, generating hydrophilic pores lined with ments for efficient function. Therefore, modification of the
phospholipid head groups (372). In all of these methods, translational initiation signals present in genes originating
exogenously added DNA enters by diffusion through artifi- from gram- bacteria is often necessary for them to be rec-
cially generated pores. Since competence-associated nucle- ognized efficiently by the translational apparatus in gram+
ase and recombinase activities are not induced, there is no species. Conversely, the more "promiscuous" gene expres-
specific mechanism operating to promote recombination of sion machinery in gram- bacteria can sometimes cause
added DNA into the genome. O n the other hand, recombi- problems when shuttle vectors bearing cloned gram' genes
nant plasmids can potentially be established very efficiently, are propagated in gram- species (i.e., E. coli), particularly if
making artificial transformation the preferred method for the cloned genes encode regulatory or secreted proteins.
many recombinant DNA applications. Aberrant overexpression of the gram' genes can lead to
toxicity for the E. coli host, which may result in growth in-
32.1.4. Applications hibition and/or production of deletions or mutations in the
All three naturally occurring forms of genetic exchange cloned gram+ genes.
have been harnessed for manipulation of gene content and
organization of gram+ bacteria, although the relative appli-
cability of any particular method is substantially species de- 32.2. GENE TRANSFER IN VARIOUS GROUPS
pendent. Genetic manipulation in gram' bacteria is con- OF GRAM-POSITIVE BACTERIA
ceptually very similar to genetic manipulation of gram-
bacteria, although in almost every case not nearly as tech- 32.2.1. Streptococci
nically advanced as for the familiar gram- genetic work- The streptococci are a heterogeneous and diverse group of
horse, Escherichia coli, or for many other gram- bacteria. low-GC, gram' bacteria. This is manifest in the variety of
Mutant construction, analysis, and complementation in their habitats, in their roles as human commensals and
gram+ bacteria utilize many of the same procedures fre- pathogens, and in the differences in types of disease they
quently employed in gram- bacteria (e.g., the use of conju- cause. Although there are some similarities in the mecha-
gation, transduction, and transformation to introduce DNA nisms of genetic exchange and other genetic approaches
into strains; the use of replicating and nonreplicating plas- that are useful for all streptococci, there also exist substan-
mids as vehicles for substrate DNA; and RecA-dependent tial differences among different species. For example, ge-
homologous recombination). Other approaches commonly netic exchange by natural transformation is prevalent in
used for manipulation of gram- bacteria (e.g., lambda red- members of the mitis and anginosus groups of streptococci
mediated recombination) are notably absent from the arse- (e.g., S. pneumoniae, Streptococcus gordonii, Streptococcus
nal of genetic tools available for manipulation of gram' mitis, Streptococcus infantis, Streptococcus cristatus, Strep-
bacteria. Chapter 27 of this volume (and references tococcus anginosus) and is also present to various extents in
therein) describes the mechanisms of basic genetic manipu- the some members of the mutans group of streptococci (e.g.,
lations and some general considerations important for their Streptococcus mutans) but not other mutans group strepto-
application. We do not recapitulate this information in de- cocci (e.g., Streptococcus sobrinus). However, the property of
tail here, but instead we merely reference specific tech- natural competence is apparently absent from members of
niques when appropriate and focus on aspects of genetic ex- the salivarius, bovis, and pyogenic groups of streptococci
change that are specifically relevant to their application (e.g., Streptococcus pyogenes, Streptococcus agalactiae,
in gram+ bacteria. For example, plasmids that are capable of S treptococcus dysgalactiae, Streptococcus salivarius, and
32. Genetic Exchange in Gram-Positive Bacteria 763

Streptococcus equinus), although one strain of Streptococcus teria, including a host of streptococci and lactobacilli (371),
bovis has been found to exhibit natural competence (266). lactococci (216), Listeria spp. (45), staphylococci (335),
The existence of natural transformation facilitates certain bacilli (213), and pediococci (143). Recently, the remark-
genetic manipulations that are difficult or impossible in ably broad host range of pIP501 was expanded to include
bacteria that lack this property. Therefore, we have com- the high-GC gram+ bacterium Streptomyces lividans and the
pared and contrasted two widely studied species of strepto- gram- bacterium E. coli (213). Conjugative plasmids in-
cocci, with the idea that genetic methods applicable to any digenous to S . pneumoniue have not been experimentally
given species of Streptococcus can be developed by applying characterized, but as in the case of GAS, several conjuga-
concepts from one or both of these diverse groups. Below we tive plasmids from heterologous streptococci have been suc-
discuss genetic exchange in the pyogenic group A strepto- cessfully transferred into S. pneumoniae (e.g., pIP501 [350]
cocci (i.e., S . pyogenes; also known as GAS) and in the mitis and pAMPl [ l l l ] ) .
group streptococcus S. pneumonia. Conjugative transposons belong to a large class of mo-
bile genetic elements now known as integrative and con-
32.2.1 .I. Plasmids jugative elements (ICES) (44) that normally reside inte-
grated in the chromosome of their host (see the section on
GAS can serve as a host for several classes of plasmids, in-
enterococci for more details). In GAS, one naturally occur-
cluding the rolling-circle-replication (RCR) plasmids (dis-
ring conjugative transposon, Tnl207.3 (carrying the mefA
cussed in more detail in the section on staphylococci) and
macrolide efflux determinant), has recently been described
the &replicating plasmids of the Incl8 family (discussed in
more detail in the section on enterococci). Members of
(330). This element has been transferred by conjugation to
other streptococcal species and was found to insert at a spe-
these plasmid classes were originally identified in other
cific site in the streptococcal chromosome. Other conjuga-
gram' species, and they are generally capable of replication
tive transposons can also be transferred into GAS. For ex-
in a wide variety of gram+ bacteria, but they are mentioned
ample, the broad-host-range conjugative transposon Tn916
here because they form the basis of a number of tools for ge-
(see the section on enterococci for more detail on Tn916)
netic analysis in GAS. It is worth noting that the common
can be used for mutagenesis in GAS, even in isolates that
staphylococcal RCR replicons pC194, pE194, and PUB1 10
are difficult to transform with plasmid delivery vectors bear-
do not replicate reliably in GAS (46), unlike in many other
ing transposons. However, the use of Tn916 as a mutagen is
gram+ cocci. However, the lactococcal RCR replicon
subject to certain disadvantages: the element is quite large
pWVOl (227) is functional in GAS, and a temperature-
sensitive-for-replication (TS) pWVOl derivative (known
(>18 kb), it exhibits some degree of site specificity for in-
sertion (the degree of insertion site specificity varies by
as the pG'host series) (253) has been used for genetic ma-
species and sometimes even by strain), and it frequently
nipulation in this organism. Plasmid vectors based on the 8-
generates multiple insertions per host. Despite these limita-
replicating Incl8 replicon of pAMPl (see the section on
tions, Tn916 has been used to identify a number of mutants
enterococci for more details) have also been widely used in
of GAS (22, 48, 100, 233, 276,403). A detailed procedure
GAS (e.g., the shuttle vector pAT28, containing the pUC
describing the use of Tn916 as a mutagen in GAS was pub-
replicon derived from ColEl [377]). Plasmids bearing
lished by Caparon and Scott (49).
Incl8-type replicons transform GAS at a lower frequency
than plasmids with pWVO1-based replicons (46), but the
In S. pneumoniae, two types of conjugative transposons
have been characterized. The first, Tn1545, carries resis-
Incl8-based vectors have been found to be useful for ob-
tance determinants for tetracycline, kanamycin, and eryth-
taining expression of cloned genes that were only poorly ex-
romycin. Tn1545 is closely related to Tn916 and, accord-
pressed by pWVO1-based vectors (124).
ingly, has been shown to have a similarly broad host range
In S. pneumonia, the most common indigenous plasmid
is pDP1, a cryptic 3-kb RCR plasmid that belongs to the
(78). A second type of conjugative transposon, designated
Tn5253, is a composite of two unrelated conjugative trans-
pC194 family (338, 349). However, pDPl has not been
posons that are capable of transferring themselves to recip-
widely exploited for genetic manipulation. The S. agalactiae
ients independently of each other. This complex genetic el-
plasmid pMV158 has been used as a cloning vector in S.
ement is composed of a copy of Tn5252 into which Tn525 1
pneumoniae (354). Another broad-host-range plasmid,
(a Tn916-like conjugative transposon) has inserted (9).
pVA838, which carries a derivative of the pAMPl replicon
(248, 249), has been used in S. pneumonia (412). 32.2.1.3. Bacteriophage and Transduction
Bacteriophage-mediated generalized transduction is an im-
32.2.1.2. Conjugative Elements portant method of genetic manipulation that can be ac-
Conjugative elements encode the machinery required for complished using either lytic or temperate phages. A vari-
self-transmission to neighboring bacteria by conjugation ety of lytic GAS phages have been described, including
(see the section on enterococci for more details). Con- phages A6, A12, A25, and A27 (261). A derivative of A25
jugative elements exist in two varieties broadly defined (255) has been widely used for generalized transduction; a
based on their mode of replication: (i) autonomously repli- detailed procedure for A25-mediated transduction has been
cating plasmids and (ii) conjugal elements that depend on described by Caparon and Scott (49). Temperate phages of
integration into the host chromosome for replication. GAS have also been described, some of which result in tox-
Although no conjugative plasmid indigenous to GAS has igenic conversion of their GAS host upon the establish-
been experimentally characterized, the enterococcal con- ment of lysogeny. Toxigenic conversion is the introduction
jugative plasmid pAMPl (68) exhibits a broad host range of a temperate phage-borne gene that functions as a viru-
and has been transferred into GAS. A relative of pAMP1, lence factor into the genome of its host bacterium, follow-
known as pIP501, was originally identified in a clinical iso- ing the establishment of lysogeny. For example, GAS phage
late of Streptococcus ugulactiae (group B streptococcus) (170) T12 carries the gene encoding speA pyrogenic exotoxin
and has been transferred to GAS. In fact, pIP501 has been (186, 401), and phage CS112 carries the gene encoding
transferred by conjugation into a wide variety of gram' bac- spec pyrogenic exotoxin (187). Thus, genetic exchange by
764 1 MOLECULAR GENETICS

bacteriophage appears to play an important role in the evo- THY broth


lution of GAS and in the emergence of virulent pathogenic
variants. Todd-Hewitt broth (Difco) . . . . . . . . . . . . . . 30 g/liter
It also seems likely that bacteriophage play an important Yeast extract (Difco) . . , . .. , .. 2 g/liter
role in the evolution of S. pneumoniae, as 70% of clinical S.
pneumoniae isolates carry putative prophages (235). A num- Electroporation buffer
ber of lytic phage capable of infecting S. pneumoniae have
been experimentally identified (e.g., phages Cp-1 [258]and Glucose
Dp-1 [133]), as have temperate phage (e.g., phages MM1 MgClz *
[282] and EJ-1 [323]). However, in contrast to GAS, no sys-
tem for genetic exchange by generalized transduction using Adjust pH to 6.5 with NaOH.
any of these or other phages has been described, perhaps in
part due to the ease of performing genetic manipulations by Transformation of S. pneumoniae
natural transformation in S. pneumoniae (see below). Natural genetic transformation (or competence) has been
extensively investigated in S. pneumoniae and exhibits
32.2.1.4. Transformation many similarities with that of B. subtilis. The competent
state is transiently induced when a culture of S. pneumonia
Competence for transformation, determined by a geneti-
achieves a critical cell density. Induction of the competent
cally encoded mechanism, has not been described for GAS.
state is dependent on the accumulation of a competence-
However, a procedure to introduce DNA into GAS based
stimulating peptide (CSP) in the culture medium; CSP is
on electroporation has been developed. This procedure re-
actively secreted by the pneumococci during growth, using
lies on growth of the bacteria in the presence of high con-
a dedicated processing and export apparatus. When extra-
centrations of glycine to weaken the cell wall prior to elec-
cellular CSP achieves a suitably high concentration, its
troporation, an approach that is often used for various
presence is detected by a two-component sensory transduc-
gramf species. A detailed protocol, as reported by Caparon
tion system (encoded by the genes comDE), initiating a sig-
and Scott (49), is described below. A n alternative set of
nal transduction cascade that ultimately results in en-
conditions for electroporation of GAS has also been de-
hanced transcription of a collection of genes involved in
scribed (346) that may be more effective for some strains.
binding, processing, uptake, and recombination of exoge-
Although a mechanistically different method for artificial
nous DNA. Thus, the addition of spent culture medium
transformation, known as protoplast transformation, has
containing CSP (or addition of synthetic CSP) to a low-
been developed for some related gram' cocci, for unknown
reasons this approach was not found to be effective for GAS
s.
density population of pneumoniae results in the premature
development of the competent state. In addition to cell
(49). It is worth noting that growth of streptococci in the density-dependent control mediated by CSP, development
presence of high concentrations of glycine can sometimes
of competence in streptococci also depends on a variety of
be irreproducible or erratic; an alternative procedure for
other factors, some of which are poorly understood. The
electroporation of streptococci has been developed (42)
regulation of competence for transformation has been re-
that limits glycine exposure of bacteria to 1 h, and it was
cently reviewed (65), as has the process of DNA transport
found to be effective for electroporation of a variety of dif-
by gram+ bacteria (55, 56). From a practical perspective,
ficult-to-transform streptococci.
the ability to easily and routinely obtain reproducibly high
Procedure for Transformation of GAS by frequencies of transformation has been greatly enhanced by
Electroporation the availability of synthetic CSP, which can be added to
The strain to be transformed is cultivated overnight in THY cultures of S. pneumoniae to induce the competent state at
broth supplemented with 20 mM glycine at 37C. For some will. A protocol for transformation of S. pneumonia using
strains of GAS, or for other species of streptococci, the con- synthetic CSP, as reported by Luo et al. (239), is described
centration of glycine in the growth medium may require below. However, it is important to note that not all strains
empirical optimization. Dilute the overnight culture 20-fold of S. pneumoniae are equally transformable. A modified pro-
in fresh THY broth plus glycine to yield 100 ml total. cedure for reproducible transformation of a type 4 encapsu-
Continue incubating as before without aeration until the lated strain of S. pneumoniae has been described (34).
A600of the culture reaches 0.2 (-2 h). Harvest the bacteria
by centrifugation (5,000 x g, 10 min, 10C) and wash twice
Procedure for Transformation of S. pneumoniae
The S. pneumoniae strain of interest (e.g., CP1250 [300]) is
in 20 ml of ice-cold electroporation buffer. The cell pellet
cultured at 37C in CAT medium (269) supplemented with
obtained from 100 ml of culture is resuspended in 0.75 ml of
0.2% bovine serum albumin, 0.5 mM CaC12, and 10 mM
ice-cold electroporation buffer and stored on ice. Aliquots HCl to an optical density at 550 nm (OD550) of 0.04.
of this cell suspension (0.2 ml) are transferred to a chilled
Aliquots of this culture are exposed to transforming DNA (1
0.2-cm electroporation cuvette. DNA (1 to 5 kg) is added
pg/ml) at 30C for 30 min in the presence of CSP (250
to the experimental samples and the cuvettes are pulsed ng/ml). The bacteria are then shifted to 37C and incubated
using the Bio-Rad Gene Pulser apparatus (voltage set at
for 1 h before plating in CAT agar with appropriate drugs.
1.75 kV, capacitance at 25 pF, and pulse controller at 400
0).Immediately following the pulse, the cell suspension is Casein hydrolysate yeast extract medium (CAT
withdrawn and placed directly into 10 ml of nonselective medium)
THY broth (without glycine) and incubated for 2 h at 37C
to allow for recovery of the cells and expression of the re-
sistance determinants. Finally, the culture is centrifuged as
described above, washed once in THY broth, and resus-
pended in 1 ml of THY broth prior to plating of aliquots on
appropriate selective agar plates. NaCl .............................. 5 g/liter
3 2 . Genetic Exchange in Gram-Positive Bacteria H 765

Autoclave; after cooling, supplement to the following con- recombinants in which the plasmid has been integrated
centrations. by Campbell-type homologous recombination (single-
crossover recombination) at the duplicated sequence
Glucose ............................. 0.2% (thereby inactivating the gene). However, one disadvantage
KlHP04 ............................. 0.167M of this approach is that it inherently generates mutations that
are polar on the expression of downstream genes. The signif-
Synthetic CSP (peptide sequence: EMRLSKFFRD- icance of polar effects can be assessed, to some extent, by
FILQRKK) can be synthesized by core service facilities at constructing a second, analogous strain using a recombinant
most major research universities. CSP is diluted in sterile vector carrying the 3' end of the target gene, instead of an in-
water to final concentration 250 Fg/ml, filter sterilized, and ternal gene fragment. After integration, such an arrangement
stored at -20C. confers polarity on the expression of downstream genes but
does not disrupt the target gene, thereby allowing the inves-
32.2.1.5. Applications of Gene Transfer tigator to determine if polar effects are resulting in any ob-
served phenotype. However, this approach is often not opti-
32.2.1.5.1. Construction of Mutants mal, as the phenotypic effects of polarity may obscure the
effect of inactivation of the gene of interest.
32.2.1.5.1.1. Transposon mutagenesis. Use of trans- Recently, alternative strategies to inactivate genes have
posons as agents of insertional mutagenesis is a common been developed to circumvent such complications. For ex-
and effective means of identifying genes involved in a bio- ample, using standard molecular techniques, recombinant
logical process of interest. In GAS, transposon mutagenesis plasmids can be constructed that carry cloned chromosomal
has been conducted using a variety of transposable ele- segments flanking a gene of interest. A n antibiotic resist-
ments. As described above, the conjugative transposon ance cassette is then inserted between these two flanking
Tn916 has been used to identify genes of interest. A more fragments, creating a cloned allele in which the gene se-
conventional transposon isolated from a gram+ bacterium, quence has been replaced with a selectable antibiotic
Tn917, has been cloned into the TS delivery vector marker. If this allele is carried on a suicide plasmid, electro-
pG+host4 (109) and used as a tool for transposon mutagen- poration of a linear form of this recombinant plasmid into a
esis (Tn917 is discussed in the section on enterococci). In GAS strain and selection for the appropriate antibiotic re-
addition, an engineered derivative of the staphylococcal sistance will yield viable recombinants only after double-
transposable element Tn4OOI (known as TnSpc) has also crossover recombination in the flanking DNA on both sides
been used to identify genes of interest in GAS (241), al- of the antibiotic marker. In such mutants, the wild-type al-
though a significant percentage of transposon insertion mu- lele will have been exchanged with the plasmid-borne mu-
tants generated with this element carry transposon inser- tant allele. Because no other plasmid-specific sequences are
tions at multiple loci. Thus, mutants generated by TnSpc incorporated in the mutant chromosome, this approach can
mutagenesis must be analyzed carefully to determine that sometimes be used to construct mutations that are not polar
the mutant phenotype is due to a particular transposon in- on the expression of downstream genes, although this de-
sertion. Another derivative of Tn4001 carrying the pends on the particular antibiotic resistance cassette and
Enterococcus fmcalis gene encoding alkaline phosphatase should be verified experimentally. Transforming DNA frag-
(phoZ), an enzyme that must be secreted to the extracellu- ments that carry at least 1 kb of flanking DNA sequence on
lar space to become active, has been used to identify se- either side of the gene targeted for mutagenesis yield the
creted proteins of GAS (141). highest frequencies of recombination. A rapid approach for
As noted above, S. pneumonim exhibits natural genetic PCR-based generation of linear recombinant fragments to
competence and therefore readily acquires exogenous DNA be used as substrates for transformation has been developed
from its environment. If this DNA contains regions of ho- which eliminates the requirements for extensive in vitro
mology to the streptococcal chromosome, it can be inte- cloning steps (217). The use of linear fragments for trans-
grated by homologous recombination. This property has formation is most effective with naturally transformable
been exploited to develop an in vitro strategy for transposon host bacteria (e.g., S. pneumoniae and S . mutans), although
mutagenesis, using a derivative of the mariner transposable they can be used with GAS as well (the frequency at which
element (2, 159), which requires only a TA dinucleotide at recombinants arise is substantially reduced). For more de-
its site of insertion. The mutagenesis procedure is divided tail, see the description of long-flanking homology PCR in
into two parts: high-efficiency insertion of a mini-mariner the section on bacilli (below).
transposable element is performed in vitro with purified An allelic exchange method to construct mutants of
pneumococcal chromosomal DNA, purified transposon GAS carrying in-frame deletions of target genes has been
DNA, and mariner transposase; in the second step, the re- developed (184). This approach has the advantage that the
sulting DNA (carrying transposon insertions) is efficiently mutations are stable and nonpolar on the expression of
transformed into S. pneumonim cells and recombined into downstream genes, but it suffers from the disadvantage that
the genome to generate a pool of transposon mutants. such mutants are more difficult and time-consuming to con-
struct. This strategy employs a two-step insertion-excision
32.2.1.5.1.2. Directed mutagenesis and allelic ex- sequence of recombination events to exchange a plasmid-
change. Insertion mutations to inactivate specific genes in borne allele of a gene of interest with the wild-type chro-
GAS (and most other bacteria) are typically constructed by mosomal copy. Initially, a plasmid-borne in-frame deletion
taking advantage of bacterial homologous recombination allele must be constructed, using standard molecular tech-
systems (see chapter 27 for more details). One common ap- niques, in an appropriate conditionally replicating vector
proach is to introduce a circular suicide plasmid, carrying a (e.g., the TS vector pGfhost5 [30]).As noted above, clones
cloned internal fragment of the target gene, into the strain that contain at least 1 kb of flanking DNA on either side of
of interest by electroporation. Subsequent selection for a the deletion yield the highest recombination frequencies.
plasmid-encoded antibiotic resistance determinant yields After introducing the recombinant plasmid into the GAS
766 MOLECULAR GENETICS

strain of interest, growth under conditions that are nonper- ficient expression. Thus, insertion of such a resistance de-
missive for plasmid replication and selection for the plas- terminant at a given chromosomal locus will yield viable,
mid-encoded antibiotic resistance determinant will yield re- antibiotic-resistant recombinants only when inserted in one
combinants in which the plasmid has integrated into the of the two possible orientations. To circumvent this prob-
host chromosome by single-crossover homologous recombi- lem, alleles of some markers have been developed that en-
nation at either the upstream or the downstream segment of code synthetic promoters, rendering their expression inde-
flanking DNA, resulting in duplications of these sequences. pendent of chromosomal context (64). It should be noted
A second single-crossover recombination event (during that DNA encoding a p-lactamase should not be intro-
subsequent growth under nonselective conditions) between duced into GAS, because p-lactam antibiotics remain clin-
corresponding duplicated fragments can lead to the excision ically relevant therapeutic agents for these organisms.
of the integrated plasmid from the chromosome, either A counterselectable marker (i.e., a marker whose ab-
leaving the mutant allele on the chromosome or regenerat- sence can be selected for, also known as a negative selec-
ing the wild-type allele, depending on the site of recombi- tion) that can be applied in GAS has not been described.
nation. The mixed population of cells that results must then However, a counterselectable marker has been developed
be screened to determine which allele has been retained on for use in negative selections during genetic manipulation
the host chromosome. Because the recombination event of S. pneumoniae (362). This strategy is analogous to a sim-
that leads to the excision of the integrated plasmid occurs ilar procedure used in gram- bacteria and is based on the
at a relatively low frequency, it is highly advantageous to in- well-known recessive properties of E. coli rpsL mutant alle-
corporate a counterselectable marker into the plasmid that les that confer streptomycin resistance (Smr) (220, 327,
permits a selection to be used to identify such recombi- 358). When such an allele is present in diploid with a wild-
nants. However, no such marker is available for GAS. type rpsLf allele, the cells exhibit sensitivity to strepto-
Therefore, either the deletion mutant to be identified must mycin (thus, the mutant is recessive to the wild-type allele).
have a known and selectable phenotype or a time-consuming Consequently, introduction of rpsL+ (carried on the back-
and laborious screening process must be used to identify the bone of an integrating plasmid) into an rpsL-containing
appropriate recombinants. (Sm') host in the first step of a two-step allelic exchange
The ability to naturally transform S. pneumonia facili- manipulation (as described above) generates an Sms
tates the construction of directed mutants but also requires merodiploid strain. The investigator then has the ability to
that care be exercised in the interpretation of the results. select for the relatively rare Sm' recombinants that result
The frequency of spontaneous mutation increases during from the excision of the integrated plasmid (and the con-
transformation with homologous DNA, such that muta- comitant loss of the rpsL+ allele) in the second step of the
tions other than those desired are often observed to occur procedure, rather than having to screen for such recombi-
(411). Two methods, based on the same principles described nants.
above for GAS, are generally used for gene inactivation in
S. pneumoniae: (i) insertional mutagenesis via a single- 32.2.1.5.2. Analysis of Mutants
crossover homologous recombination with a circular suicide
plasmid and (ii) gene replacement using double-crossover 32.2.1.5.2.1. Complementation. Complementation
recombination of linear DNA fragments. For insertional analysis requires the introduction of a wild-type copy of a
mutagenesis using a circular suicide plasmid, a strong de- gene of interest into the corresponding mutant genetic
pendence of recombination frequency on the length of the background to determine if the mutant phenotype can be
cloned segment has been observed (225). By constructing a rescued. This is accomplished either by introducing a re-
plasmid library containing random fragments of the pneu- combinant, autonomously replicating plasmid carrying the
mococcal genome, this type of insertional-mutagenesis a p gene of interest into the mutant strain or by using recombi-
proach was used to create an insertion mutant library of S . nation to introduce the gene of interest at an ectopic locus
pneumonia strains (224). Transformation by linear DNA in the chromosome of the mutant strain. Plasmid vectors
fragments and double-crossover recombination of these based on the pWVOl or pAMPl replicons can be used to
fragments into the genome is highly efficient in S . pneumo- express cloned genes in GAS for complementation or
nia and probably the most common method of directed analysis (124). In S. pneumoniae, plasmids based on
mutant construction, due to the action of the natural ge- pMV158 and pVA838 have been used to express cloned
netic competence system (for more detail on this method, genes. Other plasmid vectors based on the pAMPl replicon
see the description of long-flanking homology PCR, in the are also functional. A series of suicide plasmid vectors (the
section on Bacillus below). pVIT series) designed to allow the integration of cloned
genes or promoter fusions into a resident chromosomal copy
32.2.1.5.1.3. Selection and counterselection. As with of Tn916 have been developed for use in GAS (140). A n
many other members of the gram+ cocci, nutritional re- equivalent approach has been employed in S . pneumoniae
quirements of most streptococci, including GAS and S. (311).This approach facilitates the transfer of such recom-
pneumoniae, are complex. Therefore, nutritional markers binant Tn916 elements to other streptococcal strains that
(e.g., selection for prototrophy or carbon source utilization) may not be readily transformable themselves (because the
are generally not used for genetic manipulation in these or- recombinant Tn916 elements can be transferred by conju-
ganisms. Instead, a variety of antibiotic resistance determi- gation to such strains), and it provides the advantage of ex-
nants serve as selectable markers to facilitate genetic ma- pressing genes at the single-copy level, which may help
nipulation. Most common resistance determinants that avoid some complications resulting from overexpression of
function in other gram+ bacteria are also functional in the the cloned gene. However, this approach suffers from the
streptococci, e.g., e m (erythromycin), cat (chlorampheni- obvious disadvantage that a Tn916-containing derivative of
col), tet (tetracycline), and kan (kanamycin). However, the mutant strain must be constructed and characterized be-
some alleles of these determinants, originally identified on fore complementation can be performed.
multicopy plasmids, are known to require transcriptional Often, the ability to regulate the expression of cloned
read-through from adjacent chromosomal promoters for ef- genes in a defined manner is desirable when performing
Next Page

32. Genetic Exchange in Gram-Positive Bacteria 767

complementation analysis. Expression of cloned genes in a when a plasmid based on an RCR replicon is used for
controlled manner in both GAS and S. pneumoniue is pos- cloning or expression purposes, it is important to monitor
sible using the lactococcal nisin-inducible promoter system the integrity of the plasmid throughout the study.
that is functional in a variety of gram+ bacteria (108, 238). Replication of RCR plasmids has been reviewed in detail
In GAS, the isopropyl-P-D-thiogalactopyranoside(1PTG)- ( 193, 194). (ii) Staphylococcal multiresistance plasmids
inducible Pspacpromoter ( 108) that was originally devel- (e.g., pI524, pSK23, and pI258) are typically 15 to 40 kb in
oped for use in B. subtilis has also been used. Other options size and exist at copy numbers of 4 to 6 per cell. These are
are available for use in S. pneumonia. These include the thought to replicate via a 0-type mechanism, which confers
promoter for a-galactosidase (inducible by raffinose [238, enhanced stability. They typically carry some combination
3251) and a fucose-regulated promoter (53) that has been of resistance determinants for p-lactam antibiotics,
used for analysis of essential genes in S. pneumonia. macrolides, or heavy metal ions, some of which can be en-
coded on transposable elements that reside on the plasmids.
32.2.1.5.2.2. Expression analysis. Reporter genes Although the transposable elements and any associated re-
that specify easily assayed gene products are routinely used sistance genes may be able to transpose to heterologous ge-
as surrogate indicators of the level of expression for genes netic elements, the plasmids themselves in this class are not
of interest. This is usually done by using molecular cloning mobile by conjugation. (iii) Conjugative plasmids are dis-
techniques to fuse the promoter of the gene of interest to cussed in detail below. (iv) The fourth class of plasmids in-
a suitable reporter gene and then introducing the recom- cludes those that do not obviously belong to any of the pre-
binant construct into appropriate strains for analysis, either ceding three classes, including pSK639 (226) and related
on an autonomously replicating plasmid or after integra- plasmids. Plasmids of this class have not been characterized
tion into a chromosomal locus. A wide variety of reporter in as much detail as those of the other classes.
genes are available for use in GAS to monitor promoter
activity. These include the genes for chloramphenicol 32.2.2.2. Conjugative Elements
_ I

acetyltransferase (47, 264), P-galactosidase (124, 333), P- Conjugative plasmids represent the primary type of con-
glucuronidase (3l ) , alkaline phosphatase (147), luciferase jugative element described for staphylococci. This class of
(110, 210, 307), and green fluorescent protein (151). Most plasmids is typically 30 to 60 kb in size. Members encode
of these reporters have also been used to monitor gene ex- the machinery required for conjugative self-transfer, as well
pression in S. pneumonia, including the genes for chloram. as some (variable) combination of resistance determinants.
phenicol acetyltransferase (96), alkaline phosphatase (296, For example, plasmid pSK41 encodes resistance to amino-
297), luciferase (16, 127), and green fluorescent protein glycosides, gentamicin, tobramycin, and kanamycin as well
(90, 189). The lac2 reporter can be used in S. pneumoniae as neomycin, and to antiseptics and disinfectants (19). The
but requires a genetic background that carries a mutation best-studied family of conjugative plasmids contains pSK41
reducing the endogenous production of P-galactosidase (19, 117), pGOl (270), and pJEl (113). Multiple copies of
(64). insertion sequences, such as IS2.57, are typically present in
these plasmids, and the IS elements are often associated
32.2.2. Staphylococci with antibiotic resistance determinants, suggesting that the
The significance of some species of staphylococci as human plasmids acquired their resistance determinants through a
pathogens has prompted considerable study of these bacte- series of transposition events. Conjugative transfer of
ria, particularly Staphylococcus aureus. As a result, it is now pSK41 family plasmids occurs on solid surfaces at relatively
clear that genetic exchange plays a significant role in low frequencies ( lop5 to transconjugants per donor
staphylococcal evolution and the emergence of antibiotic- cell) and does not involve a pilus-like structure. These plas-
resistant and pathogenic variants. For example, lysogenic mids can be transferred to other staphylococcal species.
staphylococcal phage can endow their hosts with the ability Although not originally discovered in staphylococci, the
to produce toxins. Furthermore, plasmids are widespread in broad-host-range conjugative plasmids pAMpl and pIP501
the staphylococci and are associated with the emergence of can be transferred to S. uureus (111, 247,334).
antibiotic-resistant strains in health care settings. Members Conjugative transposons indigenous to S. aureus have
of one class of plasmids discovered in staphylococci, the not been experimentally characterized. However, a putative
RCR plasmids, are capable of replication in a wide variety Tn916-like conjugative transposon has been identified by
of gram+ bacteria and have thus been developed as tools for comparative genomics in the completely sequenced genome
genetic manipulation in many species. of strain Mu50 (214). Whether or not this element retains
the ability to transfer itself to other staphylococci or has be-
32.2.2.1. Plasmids come inactivated by mutation remains to be determined.
Staphylococcal plasmids can be grouped into four general The broad-host-range conjugative transposon Tn916 can
classes. (i) Small RCR plasmids utilizing an asymmetric be transferred into S. aureus and has been used as a tool for
single-stranded replication mechanism are widespread in mutagenesis (188).
staphylococci. Four families of RCR plasmids have been
identified: the pT181 family, pC194 family, pSN2 family, 32.2.2.3. Bacteriophage and Transduction
and pE194 family. These are normally less than 5 kb in size, Most natural isolates of S. aureus carry one or more pro-
generally maintain themselves at relatively high copy num- phages. The repertoire of prophages present in a given strain
ber (10 to 50 per cell), typically are cryptic or carry only a influences susceptibility to further phage infection, thereby
single antibiotic resistance determinant, and are rarely forming the basis for a historically widely used phage typing
found to contain transposable elements (278). RCR plas- scheme (5). This procedure utilizes a standard reference set of
mids can replicate in many heterologous gramf bacteria phages and has been employed for epidemiological analysis,
(280) and have therefore been widely employed as cloning although it has now been largely supplanted by molecular
and expression vectors, but derivatives of RCR plasmids techniques (5). As with many other species of bacteria, tem-
carrying cloned genes can sometimes be structurally unsta- perate phage integration results in lysogenic conversion for
ble, leading to deletions of cloned gene segments. Thus, staphylococci. For example, the staphylococcal enterotoxin
Genetics of Archaea
KEVIN R . SOWERS. PAUL H. BLUM. AND SHILADITYA DAsSARMA

33.1. HALOPHILIC EURYARCHAEOTA ........................... 801


........................................ 801
33.1.1. Introduction
. .................................. 802
2. Strain Characteristics
. .......................
33.1.2.1. Halobacterium sp NRC-1 802
. . .................................. 802
2. H volcunii
............................ 802
33.1.3. Genetic Markers for Selection
. .............................. 803
4. Vectors for Gene Transfer
...................... 803
33.1.4.1. Shuttle and Cloning Vectors
. ................. 804
2. Gene Replacements and Knockouts
. .............. 804
3. Insertion and Transposon Mutagenesis
. ............................... 804
4. Gene Reporters
. ........... 805
5. Promoter Analysis and Expression Vectors
...................................... 806
33.1.5. Transformation
....................... 806
33.1.5.1. Transformation with PEG
.................................... 806
33.1.6. Media and Growth
............................. 806
33.1.6.1. Media Preparation
. ............................. 807
2. Storage of Strains
...................................... 807
33.1.7. Troubleshooting
. .................................. 807
8. Recent Developments
33.2. METHANOGENIC EURYARCHAEOTA ........................ 807
........................................
33.2.1. Introduction 807
. ..................................
2. Strain Characteristics 808
. ...........................
33.2.2.1. M acetivorans C2A 808
. . .............................
2. M maripaludis JJ 810
33.2.3. Genetic Markers for Selection ............................ 810
.
4. Vectors for Gene Transfer .............................. 810
......................
33.2.4.1. Shuttle and Cloning Vectors 810
. ........
2. Transposon-Mediated Random Gene Disruption 810
. .......
3. Directed Gene Disruption and Complementation 812
. 4. Gene Reporters............................... 812
33.2.5. Transformation ...................................... 813
.
33.2.5.1. Transformation of M acetivorans with Liposomes ...... 813
. .
2. Transformation of M maripuludis with PEG .......... 813
. ......... 814
3. Colonization of Transformants on Agar Plates
33.2.6. Media and Growth .................................... 814
33.2.6.1. Media Preparation ............................. 814
. .............................
2. Storage of Strains 816
33.2.7. Troubleshooting ...................................... 816
.
8. Recent Developments .................................. 817
33.3. HYPERTHERMOPHILIC CRENARCHAEOTA ................... 817
33.3.1. Introduction ........................................ 817
.
2. Strain Characteristics.................................. 817
. ............. 817
33.3.2.1. S solfataricus Strains for lacs Selection
. . ............ 817
2. S solfataricus Strains for malA Selection
33.3.3. Genetic Markers for Selection ............................ 818
33.3.3.1. lacs and malA as Selectable Markers............... 818
. ...................... 819
2. Screening for Recombinants
33.3.4. Vectors for Gene Transfer .............................. 819
33.3.4.1. DNA Methylation ............................. 819
33.3.5. Transformation ...................................... 819
. ..... 819
33.3.5.1. Transformation of S solfataricus by Electroporation
33.3.6. Media and Growth .................................... 819
33.3.6.1. Media Preparation ............................. 819
. .............................
2. Storage of Strains 820
33.3.7. Troubleshooting ...................................... 820
.
8. Recent Developments .................................. 820
33.4. REFERENCES ............................................ 820

800
33. Genetics of Archaea 801

The Archaea represent a phylogenetic lineage of microor- ditions, with normal atmospheric temperature and pressure
ganisms that are distinct from both the Bacteria and (29, 42). In contrast to methanogens and thermophiles,
Eukarya. Although this phylum of microorganisms exhibits both of which require specialized culturing facilities, haloar-
a wide range of morphological and physiological diversity, chaea can be grown in any microbiology laboratory by using
which includes sulfur-metabolizing hyperthermophiles, standard media supplemented with 12 to 25% salt. As a re-
methanogens, and extreme halophiles, all Archaea have sult, many methods for genetic manipulation of haloarchaea
one characteristic in common: the requirement for growth have been developed and standardized during the past 20
conditions that have periodically redefined the limits for years (see reference 31). With the recent availability of com-
extant life as we know it. These include saline concentra- plete or nearly complete genome sequences for several
tions up to saturation, highly reduced anoxic environments haloarchaea, these microorganisms have come to represent
poised below -350 mV, and extremely high temperatures of very tractable model systems for genetic studies.
up to 113C.Numerous reports have been presented on the Haloarchaea are found in evaporatic brine pools in the
physiology and biochemistry of all three groups of Archaea, tropical, temperate, and arctic regions of the world, usually
but several unique characteristics of this phylum have pre- at the surface, but also under the sea in submarine pools and
cluded the ability to apply standard bacterial or eukaryal ge- underground in ancient salt deposits (29). Many hyper-
netic protocols. Traditional colony growth of many of the saline environments are dynamic with respect to tempera-
fastidious anaerobes in roll tubes is not as practical as plat- ture, pH, oxygen, sunlight, etc.; therefore, haloarchaea dis-
ing for screening large numbers of clones; unique cell wall play a wide variety of biological responses (28, 59). To
structures prevent the use of commonly used antibiotic ge- balance the high sodium chloride concentration in the
netic markers that target cell wall synthesis; bacterial gene medium, haloarchaea accumulate potassium chloride as a
promoters associated with many of the commonly used ge- compatible solute, and all of their metabolic activities take
netic markers are not recognized by the archaeal transcrip- place in a hypersaline cytoplasm. Haloarchaea also exhibit
tional apparatus; bacterial plasmids and phages will not a high level of resistance to UV light and y-radiation as pro-
replicate in archaeal species. In recent years several labora- tection against the intense solar radiation usually found in
tories have overcome these difficulties by developing effec- their environment. Many species are highly motile, display-
tive plating techniques, identifying genetic markers that do ing phototaxis, chemotaxis, and gas-vesicle-mediated flota-
not target cell wall synthesis, fusing archaeal promoters tion. Some are capable of phototrophic growth, and most
with recombinant genes, and isolating native vectors and display facultative anaerobic capabilities.
promiscuous nonnative vectors. All of the haloarchaea described thus far belong to the
This chapter does not attempt to describe historical as- Halobacteriaceae family (42). Of the 19 different genera of
pects or gene transfer systems under development, but focuses haloarchaea that have been described, genetic and genomic
instead on tractable systems that are currently available for analyses have been concentrated on a few species and strains.
the Archaea. Because of fundamental differences between The most intensively studied are Halobacterium spp., which
gene transfer systems for each archaeal branch, the chapter is are well known for their production of purple membrane, and
divided into three inclusive sections covering the halophilic Haloferax oolcanii, which can grow on minimal medium.
and methanogenic Euryarchaeota and the hyperthermophilic Halobacterium spp. and H. volcanii have well-developed ge-
Crenarchaeota. All three Archaea require methodologies netic tools, including a facile transformation system, selec-
that are specialized to varying degrees. The halophilic table markers, cloning and expression vectors, reporter genes,
Euryarchaeota require only media with high concentrations and gene replacement and knockout systems (31).
of salt, utilizing otherwise standard protocols for mesophilic, Genome sequences are available for some of the com-
aerobic bacteria. The hyperthermophilic Crenarchaeota re- mon laboratory strains of haloarchaea. The first complete
quire incubators adapted for higher temperatures (8OOC) and genome sequence became available in 2000 for Halobac-
sealed containers to prevent evaporation of medium, but terium sp. NRC-1 (78, 80). Several additional genome se-
otherwise utilize standard protocols for aerobic bacteria. quences are under investigation (Table l),including H. vol-
Additional information on methods for growing halophilic canii and Haloarcula marismortui and also Halorubrum
Euryarchaeota and hyperthermophilic Crenarchaeota can be lacusprofundi, Natronomonas pharaonensis, and Haloqua-
found elsewhere (31,95). The methanogenic Euryarchaeota drutum walsbyi. The genomes of haloarchaea are relatively
require specialized apparatus and methods to maintain anaer- G+C-rich (58 to 68%), with the exception of some large
obic media at a low redox potential and prevent exposure of and small extrachromosomal species that are rich in inser-
cultures to oxygen. Because details of these methods are be- tion sequence (IS) elements. These extrachromosomal
yond the scope of this chapter, the reader should refer else- species have been studied in Halobacterium spp. and shown
where for a comprehensive description of the growth of to rearrange at high frequency. This is in contrast to the
methanogenic Euryarchaeota before beginning genetic ex- Halobacterium chromosome, which is quite stable and in
periments with this system (108). Despite varying degrees of fact contains a smaller number of transposable IS elements
difficulty growing Archaea, all three systems are routinely than some other archaea, e.g., Sulfolobus solfataricus (28).
used by laboratories conducting research on archaeal genet- The combination of easy culturing, highly developed ge-
ics and can be mastered by anyone with a fundamental netic tools, and interesting biology makes haloarchaeal sys-
knowledge of microbial genetic techniques. For reviews on tems attractive for study. Past studies have been extremely
archaeal genetics see references 2, 69, 98, and 110. fruitful, providing a deeper understanding of prokaryotic
genome structure and dynamics, lateral gene transfer, ge-
netic regulatory mechanisms, and DNA replication, tran-
33.1. HALOPHILIC EURYARCHAEOTA scription, and translation. With the recent availability of
complete genome sequences, systematic knockout strategies,
33.1 . I . Introduction DNA arrays, and proteomics, this system has the potential to
Halophilic Archaea grow optimally in hypersaline environ- provide tremendous opportunities for advancing the biology
ments containing 2 to 5 M NaC1, usually under aerobic con- of haloarchaea.
802 MOLECULAR GENETICS

TABLE 1 Archaeal genomic sequences


Archaeal strains Website Reference
Halophilic Euryarchaeota
Halobacterium sp. NRC-1 http://zdna2.umbi.umd.edu/ 80
Haloferax volcanii DS2 http://zdnaZ.umbi.urnd.edu/-haloweb/hvo.html Unpublished
Haloarcula marismortui http://zdna2.umbi.umd.edu/-haloweb/hma.html Unpublished
Methanogenic Euryarchaeota
Methanococcus maripaludis LL http://www.genome.washington.edu/UWGC/methanococus/Methanococcus.html 50
Methanosarcina acetiworuns C2A http://www.broad.mit.edu/annotation/microbes/methanosarcina/ 39
Methanosarcina barkeri Fusaro http://genome.jgi-psf.org/finished_microbes/metba/metba.home.html 64a
Hyperthermophilic Crenarchaeota
Sulfolobus solfataricus P2 http://www-archbac.u-psud.fr/projects/sulfolobus/ I03

33.1.2. Strain Characteristics deletion of pNRClOO resulting from intramolecular trans-


position of ISH8. A second strain, SD112, has a 59-kbp
33 .I .2 .I. Halobacterium sp. N RC-1 deletion resulting from an ISH2 transposition.
Halobacterium sp. strain NRC-1 (ATCC 700922) is a com-
mon, rod-shaped strain which grows well in complex media 33.1.2.2. H. volcanii
containing 4.5 M NaCl with peptone, yeast extract, or H. volcanii DS2 (ATCC29605), isolated from the Dead Sea,
Casamino Acids as carbon and energy sources (Color Plate is disk or cup shaped and grows optimally at 45C in 1.5 to
13). Optimal growth (6-h generation time) is observed at 2.5 M NaC1. It is tolerant to high concentrations of Mg2+,
42C. Growth on defined medium containing 15 amino up to 1.5 M, reflecting the composition of its natural envi-
acids and vitamins has also been reported, and cell lysis is ronment. H. volcanii can grow in minimal medium on glu-
minimized in the presence of trace quantities of metal ions. cose as sole carbon and energy source. However, the growth
Under low oxygen tension, Halobacterium sp. NRC-1 in- rate is slow compared with rich medium, where the genera-
duces purple membrane patches in the cell membrane and tion time is 4 h. A derivative of DS2, H. volcanii WFDl l, is
buoyant gas vesicles intracellularly, which increases the cured of the natural cryptic miniplasmid pHV2 (63).
availability of light and oxygen and allows a period of light- Several pHV2 derivatives are popular cloning and shuttle
driven proton pumping and phototrophic growth (13). vectors. A n H. piolcanii ApyrElApyrEZ mutant, WR479, can
One of the most useful mutant strains of NRC-1 has a be used for gene knockout with a method similar to that de-
deletion of the ura3 gene (NRC-1 [Aura3]) and is auxo- veloped for Halobacterium sp. NRC-1 (16).
trophic for uracil and 5-fluoroorotic acid (5-FOA) resistant Genotypic characteristics for some haloarchaeal strains
(Ura-, Foa) (86). The NRC-1 (Aura3) strain is used for are summarized in Table 2.
generating knockouts using either selection for mevinolin
resistance (MeV) and counterselection against ura3 (Foa) 33.1.3. Genetic Markers for Selection
or both selection and counterselection for ura3 (62, 86). One of the most commonly used selective markers for the
Several single and multiple gene mutants have been suc- haloarchaea confers resistance to mevinolin (62).
cessfully constructed by using these procedures (e.g., Ahp, Mevinolin is an inhibitor of 3-hydroxy-3-methylglutaryl
Ablh, Ahp/blh, and AcrtY, coding for enzymes of retinal coenzyme A reductase, an enzyme involved in the produc-
biosynthesis, and AarsADRC, AarsB, and AarsM, coding for tion of sterols in eukaryotes and isoprenoid side chains of
heavy metal resistance), indicating that knockout of any lipids in archaea. Selection of mevinolin-resistant (MeV)
nonessential gene is possible (87, 88). Numerous studies mutants of H. volcanii resulted in the finding of up-promoter
have been conducted on the purple membrane protein, bac- mutations, an allele that has been used for selection in
teriorhodopsin (BR), and the bop gene. The commonly used H . uolcanii, Halobacterium spp., and other haloarchaea. The
BR overproducer (Pum+++)strain S9 is not a derivative of background mutation rate to MeVr is relatively low
or isogenic with NRC-1 (13). It was isolated from another (<lop7),making this an excellent selection system for the
natural Halobacterium strain by extensive chemical mutage- haloarchaea.
nesis. S9 likely contains dozens of mutations and allelic dif- Selectable markers for halophilic archaea allowing both
ferences from NRC-1. The mutation responsible for the selection and counterselection have been developed re-
Purn+++ (deep purple) phenotype of S9 has been shown to cently. These are the Halobacterium sp. NRC-1 ura3 gene,
be a double frameshift in the bat gene (bat allele), which encoding orotidine-5-monophosphatedecarboxylase, and
results in a 4eamino-acid change in the encoded protein. H . volcanii pyrE2 genes, coding for orotate phosphoribosyl-
Mutants of S9 lacking BR, e.g., SD23, which is purple mem- transferase (16, 86). These markers are extremely useful for
brane and bacterioruberin deficient (Pum-, Rub-) as a re- both gene replacements and gene knockouts. Forward se-
sult of an ISHl insertion in bat, and SD20, which is Pum- lection can be carried out in a Aura3 or ApyrE2 background
as a result of an ISHl insertion in bop, have been character- by selection for uracil prototrophy (Ura+), using uracil-
ized extensively (13,27). Other strains lacking BR have also dropout medium for Halobacterium sp. and Casamino Acids
been isolated and characterized (e.g., RlmR and L33) (27). for H. uolcanii. Counterselection can be carried out using re-
Numerous genetic studies have also been conducted on sistance to 5-FOA (Foa), which is metabolized to a toxic
gvp genes with a gas vesicle-deficient (Vac-) NRC-1 strain intermediate in a Ura+ background.
deleted for the pNRClOO gup gene cluster (30, 77). The A variety of other selectable markers have also been de-
most common strain, SD109 (Color Plate 13), has a 67-kbp scribed. The gyrB gene allele has been used for selection of
3 3 . Genetics of Archaea rn 803

TABLE 2 Archaeal strain characteristics and sources'


Archaeal strains Characteristics Reference
Halophilic Euryarchaeota
Halobacterium sp. NRC-1 Wild-type ATCC 700922, JCM 11081 80
Halobacterium sp. SK400/MPK414 NRC-1 A u ~ 123
Halobacterium sp. S9 Purple membrane overproducer 13, 113
Halobacterium sp. R1 Gas-vesicle-deficientmutant ATCC 19700 113
Haloferax volcanii DS2 Wild-type ATCC 29605, DSMZ 3757, JCM 8879 24
Halobacterium sp. SD20 Purple-membrane-deficientmutant of S9 bat::ISHl 9
Halobacterium sp. SD23 Purple-membrane-deficient mutant of S9 bop::ISHl 9
Halobacterium sp. SD109 NRC-1 with 67-kb deletion of gvp gene cluster 61
Halobacterium sp. SD112 NRC-1 with 59-kb deletion of gwp gene cluster 61
Halobacterium sp. RlmR Purple membrane mutant of R1 bop::ISH2 24
Halobacterium sp. L33 Purple membrane mutant of S9 bop::ISH2 24
Haloferax volcanii WFDl 1 DS2 cured of pHV2 63
Haloferax wolcanii WR479 WR341 ApyrElApyrE2 16
Methanogenic Euryarchaeota
Methanococcus mripaludis JJ Wild-type ATCC 43000, DSMZ 2067, JCM 10722, OCM 175 55
Methanosarcina acetiworans C2A Wild-type ATCC 35395, DSMZ 2834, JCM 12185, OCM 95 105
Methanosarcina barkeri Fusaro Wild-type ATCC 29787, DSMZ 804, OCM 83 58
Methanosarcina mazei GO1 Wild-type ATCC BAA-159, DSMZ 3647, JCM 11833, OCM 88 57
Hyperthermophilic Crenarchaeota
Sulfolobus solfataricus 9812 Wild type 96
Sulfolobus solfataricus PBL2025 98/2 ASSO3004SSO3050,lacs deletion 101
Sulfolobus solfataricus PBL2030 98/2 ASSO30133303037, ASSO3051, mlA deletion Hoang and Blum,
unpublished
Sulfolobus solfataricw Gtheta Wild-type derivative of MT3 23
Sulfolobw solfataricus P2 Wild-type ATCC 35092, DSMZ 1617 130
Sulfolobus solfataricus P1 Wild-type ATCC 35091, DSMZ 1616 130
Sulfolobus acidocaldarius Wild-type ATCC 33909 22
"ATCC, American Type Culture Collection (http://www.atcc.org/); DSMZ, Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (http://www
OCM, Oregon Collection of Methanogens (http://methanogens.pdx.edu/).
.dsmz.de/); JCM, Japan Collection of Microorganisms (http://www.jcm.riken.go.jp/);

resistance to the DNA gyrase inhibitor novobiocin (53). ated together to form the shuttle vector pWL102 (63).This
Several protein synthesis inhibitors, including anisomysin, plasmid was reduced in size by minimizing the mevinolin re-
thiostrepton, and chloramphenicol, have also been used as sistance region, and a multiple cloning region with unique
selectable markers. These are particularly useful for gene re- ApaI, EcoRI, SmaI, S a d , SphI, KpnI, XbaI, HindIII, and
placements of rRNA genes, especially for Halobacterium Not1 sites was inserted to form the 8.8-kbp shuttle vector
spp., which have a single copy of the rRNA operon (65). pWL 104.
A Halobacterium pNRC100-derived vector, pNG168,
33.1.4. Vectors for Gene Transfer 8.9 kbp (Fig. l), contains the pTZ18 vector, including the
Amp' determinant for selection in E. coli; multiple cloning re-
33.1.4.1. Shuttle and Cloning Vectors gion with unique ApaI, HindIII, EcoRI, PstI, SmaI, BamHI,
Halophilic archaea are rich in plasmid diversity, and many SpeI, and No1 restriction sites; the pNRCl00 minimal repli-
recombinant vectors have been constructed (3 1). Some of con pNG101; and the minimal H . volcanii MeVr marker for
the most common vectors are derivatives of H . volcanii mini- selection in halophiles. pNG168 is capable of replication in
plasmid pHV2 and Halobacterium spp. minichromosomes or both Halobacterium sp. NRC-1 and H. vokanii and has been
megaplasmids, such as pNRClOO and pHHl or miniplas- used for a variety of genetic experiments (30,79).
mids pGRB and pHSB. In general, these vectors replicate in A vector similar to pNG168, pUBP2, is somewhat larger
all commonly used strains of halophilic archaea tested. (12.3 kbp) and was constructed from the related Halo-
Vectors derived from the H. volcanii miniplasmid pHV2 bacterium sp. PHHl plasmid pHHl (18). Th'is vector con-
are in wide use and can be selected and maintained in tains the E. coli vector pIBI, with the H. volcunii hmg marker
Haloferax spp., Halobacterium spp., or Escherichia coli. These for MeVr selection in halophiles, and pHH9, a natural dele-
vectors were constructed by first cloning the hmg gene, en- tion derivative of pHH1. Several unique restriction sites are
coding resistance to the 3-hydroxy-3-methylglutaryl coen- available for cloning in this vector.
zyme A reductase inhibitor mevinolin, into the pHVZ repli- Three related Halobacterium spp. miniplasmids, pGRBl ,
cation region to form pWL2. The resistance determinant, pHSB1, and pHGN1, which replicate by a single-stranded
portions of pHV2, and an ampicillin- and tetracycline- intermediate, have been used for vector construction by
resistance-conferring pBR322 derivative, pAT153, were lig- inserting the MeVr marker. Another vector, pMSDZO, was
804 MOLECULAR GENETICS

Hindlll 33.1.4.3. Insertion and Transposon Mutagenesis


Insertion mutagenesis is conducted by employing natural IS
element insertions, gene cassettes and oligonucleotide link-
ers, and transposons constructed with haloarchaeal IS ele-
ments. Natural phenotypic variants were used in early stud-
ies to identify bop and gvp regulatory genes and coordinately
regulated genes. IS element insertions identified the brp and
the bat genes necessary for purple membrane production up-
stream of bop (27). Similarly, IS element insertions identi-
fied gupD, gupE, and other genes necessary for wild-type gas
vesicle synthesis (30). More recently, IS element insertions
have been found in the ura3 gene in Foa' mutants (86).
These studies represent a valuable natural transposon muta-
genesis system in haloarchaea that has provided significant
insights into gene regulation.
Gene disruptions made by using gene cassettes and
oligonucleotide linkers are also useful for genetic analysis.
To study the functions of the gup gene cluster on pNRClOO
\. in gas vesicle formation, a kanamycin resistance ( K ) gene
cassette was used for scanning mutagenesis of the gene clus-
hmg ter in pFL2, a 24.5-kb E. coli-Halobacterium shuttle plasmid
(30). Transformation of Halobacterium sp. SD109, deleted
FIGURE 1 Haloarchaeal shuttle vector pNG168. This plas- for the entire gup gene cluster, with pFL2 and mutated pFL2
mid contains the E. coli pTZl9r replicon and the Halobacterium derivatives showed that, while the unmutated gene cluster
NRC-1 pNRClOO minimal replication region. The bla gene successfully programmed gas vesicle formation (Color Plate
provides selection with ampicillin in E. coli, and the hmg gene 13),derivatives with insertion of the K cassette in nearly all
provides selection with mevinolin in haloarchaea. The multi- of the gup genes lacked normal gas vesicles. In most cases,
ple cloning site is located in the lacZcl fragment gene and per- the block in gas vesicle synthesis did not result from polar
mits blue-white screening in E. coli. The plasmid is available effects, since similar results were obtained for derivatives of
from ATCC (catalog no. MBA-77) and the sequence is avail- the insertion mutants in which most of the internal portion
able in GenBank (accession no. AY291460). of the K cassette was deleted and only small (15- to 54-bp)
insertions remained.
Similarly, one pNRClOO minireplicon, pNGllA12, was
analyzed by linker-scanning mutagenesis by using a short
constructed from cryptic Haloferax sp. plasmid pHK2 by oligonucleotide (79). Insertions in the repH gene knocked
cloning the novobiocin resistance (Nov') gyrB gene from out the capability of the minireplicon for autonomous repli-
H. uolcanii. This plasmid was used to transform H. uolcanii cation, while a second insertion at the same site, restoring
at high frequency. the repH reading frame, resulted in reversion to replication
proficiency.
33.1.4.2. Gene Replacements and Knockouts
Some effort has been directed toward construction of re-
A n exciting recent development is a new gene replace- combinant transposons with selectable markers from natu-
ment and knockout method in the sequenced strain Halo- ral IS elements (33, 122). Synthetic transposons were con-
bacterium sp. NRC-1 using the ura3 gene, which can be structed consisting of haloarchaeal ISH elements (ISH2,
both selected for and counterselected against (86). This ISH26, or ISH28) flanking a mevinolin resistance determi-
approach is diagrammed in Fig. 2. A gene allele of interest nant. Introduction of an ISH28-based transposon (ThD28)
(e.g., a precise deletion) is first cloned into an E. coli plas- into Haloarcula hispanica on nonreplicating plasmids pro-
mid, and the suicide plasmid is introduced into the halo- duced numerous non-site-specific transformants, with some
archaeon host by transformation. Integrants are selected bias toward low G + C content regions. However, when
by uracil prototrophy (Ura+, using commercially available transposons were introduced into haloarcheal strains with
uracil-dropout media components available from Sigma- perfectly homologus DNA (e.g., H . uolcanii),plasmids inte-
Aldrich Corp., St. Louis, MO). Alternatively, mevinolin grated at high frequency by homologous recombination.
resistance (MeV') may also be used. Subsequently, ex-
cisants are selected by counterselecting for 5-FOA resis- 33.1.4.4. Gene Reporters
tance (Foa'), giving rise to both the replaced allele and the Several gene reporters are used for haloarchaea, including
original, which can be distinguished by PCR or pheno- the Aequorea victoria green fluorescent protein (GFP) (83).
typic analysis. A similar approach has proved successful for The gfp gene was fused to the bop gene and expressed as a
H . uolcanii, but substituting the pyrE2 gene (in place of BR-GFP fusion in a Halobacterium sp. The fusion protein
ura3 ) . preserved the intrinsic function of each component under
In the past, gene deletions and replacements were con- conditions with an extremely high-salt concentration and
structed, but they required extensive manual screening. The was shown to be properly localized in the plasma mem-
largest number of mutants was characterized for the purple brane. The results indicate that GFP can be used as a versa-
membrane bop gene, employing the mevinolin resistance tile reporter of gene expression in Halobacterium for investi-
marker (e.g., reference 60). Another interesting application gations of halophilic membrane proteins.
of gene replacement was for the single rRNA operon of A P-galactosidase (bguH) gene from Haloferax alicantei
Halobacterium spp., where dominant mutations resulting in has also become available as a reporter in haloarchaea (84).
antibiotic resistance could be selected (65). When introduced into a Halobacterium sp. on a plasmid
33. Genetics of Archaea w 805

f
Recircuiarize Transform Aura3 host
/ I lntegrant

1
Select for recombinants

or
I2
v AgeneX

FIGURE 2 Gene knockout and replacement in the halophilic Euryarchaeota. The example
shown is for selection and counterselection with ura3. A cloned haloarchaeal target gene (geneX)
in a plasmid vector, which does not replicate in haloarchaea, is used for PCR amplification (primers
designated by arrowheads) and recircularization to provide for a precisely deleted gene. The plasmid
is introduced into a Aura3 haloarchaeon by transformation. Integrants are selected by uracil pro-
totrophy using uracil dropout plates. Excisants are selected for by plating on plates containing 5-
FOA. Depending on the site of the recombination (1 or 2), different outcomes are possible.
Alternatively, mevinolin selection can also be used for integration and the pyrE2 gene can be used
for selection and counterselection.

vector-carrying bgaH, the enzyme activity in cell lysates can Fusions of the bop promoter and N-terminal coding re-
be determined by a colorimetric assay and colonies screened gion have been used successfully for overproduction of bac-
for activity on plates containing X-Gal substrate. teriorhodopsin mutants, halorhodopsin (the chloride
Expression of bgaH under the control of various halobacte- pump), several sensory rhodopsins (the phototactic recep-
rial promoters of known strength leads to different specific tors), and two halotransducers (e.g., references 51, 56, 60,
P-galactosidase activities in the lysates. In one study, the 81). Additionally, foreign membrane proteins, e.g., mam-
bgaH gene was used as a reporter to investigate three differ- malian G-coupled protein receptors (GPCRs), such as the
ent haloarchaeal promoter regions derived from gvpA (43). human muscarinic acetylcholine (MI) and adrenergic (a2b,
pz)receptors, have been expressed in haloarchaea (14,85).
33.1.4.5. Promoter Analyskand Expression Vectors However, both proteolysis and uncharacterized determi-
The two main promoters commonly utilized for heterolo- nants at the 3 end of the bop gene were found to limit the
gous or homologous gene expression in haloarchaea are the usefulness of this heterologous gene expression system.
ferredoxin (fdx) promoter, mainly for soluble proteins, and The fdx promoter has been used to express a variety of
the bop promoter, usually for membrane proteins. Both pro- soluble proteins (e.g., references 56 and 89). These expres-
moters have been characterized by saturation mutagenesis sion systems have sometimes been coupled to His-tagged
and up-promoter mutations have been reported (11, 12, proteins, which have been successfully purified using metal
26). Vectors containing 200 bp upstream of the fdx coding affinity chromatography in a variety of systems. However,
region are sufficient for constitutive expression. For bop, difficulties have also been encountered in haloarchaea due
only 53 bp upstream of the coding region is sufficient, but to binding of endogenous proteins with metal affinity (e.g.,
regulation is complex, and expression is highly dependent Halobacterium sp. NRC-1 protein VNG2021 is a frequent
on the specific gene and construct. contaminant).
806 w MOLECULAR GENETICS

In addition to the fdx and bop promoter vectors, a few 9. After overnight incubation, add 1 ml of CM+
other specialized expression systems have also been used medium (see below) and grow with shaking overnight.
successfully. The gas vesicle gene cluster has been modi- 10. Check cultures under a phase-contrast microscope
fied to incorporate a cloning site in the gupC gene in the for regeneration. When >80% of cells have returned to rod
pFM104D shuttle vector and used to produce fusion pro- shape (in general, 1 to 2 days after addition of CM+ media),
teins attached to the surface of floating vesicles (114). inoculate transformants on selective plates by spreading and
This system has been used for antigen expression and incubate at 42C. Visible transformant colonies develop
delivery and for development and testing of vaccine can- over 5 to 10 days.
didates. Also, WL204, a derivative of pWL102 with a H.
volcanii tRNAE S gene promoter cloned in, has been used NOTE. Similar procedures can also be used to transform
H . volcanii, H. rnediterranei,H. hispanica, and H. vallismortis,
for expression of tRNA genes to study RNA processing
with changes in the spheroplasting and regeneration media
(82). to reflect the different ionic optima for the different species
33.1.5. Transformation (21). Many of the details may be obtained from Halophiles,
Both Halobacterium and Haloferax spp. and several other volume 1 of Archaea: A Laboratory Manual (31).
haloarchaea can be transformed using a procedure involv-
33.1.6. Media and Growth
ing spheroplast formation, by chelating MgZf with EDTA,
generating competent cells with PEG treatment, DNA up- In general, culturing of halophiles is relatively simple and
take, cell regeneration, and plating (31). The routine effi- conducted in the same way as for common aerobic bacteria.
ciency of the procedure is about lo5 to lo6 transformants Liquid cultures are grown at 37 to 42C in test tubes or
per microgram of common shuttle plasmid DNA. The main Erlenmeyer-Fernbach flasks with shaking at 200 to 300 rpm.
procedural variations for transformation of different halo- For phototrophic strains, fluorescent lighting may be used
philes reflect the specific ionic concentrations of the media. but is not essential. Plates contain 2% agar, which is re-
An alternate procedure for Halobacterium sp. has been re- quired for solidifying under hypersaline conditions, and are
ported by using freeze-thaw of cells, but the efficiency is rel- placed in airtight containers or humidified environment to
atively low in comparison with the EDTA-PEG procedure avoid crystallization of salt.
(128).
33.1.6.1. Medium Preparation
Most of the haloarchaeal species tested have a restric-
tion system, which reduces the efficiency of transformation Descriptions are provided here for rich media for the three
with DNA from a heterologous host by a factor of lo3 or model haloarchaea, Halobacterium sp. NRC-1, and H. uol-
greater (31). The modification is evident by the inability of canii.
some restriction enzymes to digest chromosomal DNA from
Halobacterium sp. NRC-1 CM+ Complex Medium
haloarchaea, and corresponding haloarchaeal restriction-
(per Liter)
modification activities have been reported (16, 51, 62).
Purification of transforming DNA from an E. coli DamC
NaCl ................................. 250g
strain can reduce or eliminate the restriction problem.
MgS04 . 7H2O ......................... 20 g
33.1.5.1. Transformation with PEG KCl .................................. 2g
Sodium citrate ......................... 3g
1. Grow a 50-ml culture of Halobacterium sp. NRC-1 at Oxoid peptone (Oxoid L34) . . . . . . . . . . . . . . . 10 g
42C to an OD600 of 1.0 in a flask with good aeration. Trace metal stock (2,000X ) . . . . . . . . . . . . . . . 0.5 ml
2. Gently pellet cells by centrifugation at 6,000 X g for
10 min. pH to 7.2 with NaOH.
3. Gently resuspend cells in 5 ml of spheroplasting so-
lution (2 M NaC1, 27 mM KC1, 50 mM Tris-C1 [pH 8.751, Trace Metal Stock (2,000 X )
15% [wt/vol] sucrose).
4. After 5 min, examine cells in a phase-contrast mi- FeS04 . 7 H 2 0 ...................... 3.50 mg/ml
croscope to verify spheroplast formation, which is charac- ZnS0, . 7H20 ...................... 0.88 mg/ml
terized by cell morphology with perfectly circular spheres. MnS0, . H 2 0 ...................... 0.66 mg/ml
5.Add 10 pl of DNA in spheroplasting solution or 10 c u s o 4 . 5 H 2 0 ...................... 0.02mg/ml
mM Tris-HC1-1 mM EDTA to 200 p l of spheroplasts.
Examine cell morphology under a phase-contrast micro- NOTE. Oxoid peptone should be used for optimum pur-
scope to check for cell lysis. Cells should appear spherical or ple membrane production. Peptone may be substituted with
pleomorphic, and the number of visible cells should not 7.5 g of Casamino Acids and 10 g of yeast extract. Trace
change substantially after addition of DNA. Cell lysis re- metal stock is filter sterilized and added to cooled auto-
sults in increased viscosity and greatly reduces the transfor- claved medium.
mation efficiency.
6. Add 200 p1 of PEG solution (6 ml of PEG600 in 4 Halobacterium sp. NRC-1 Uracil-Dropout Medium
ml of spheroplasting solution) and mix gently by tapping. (per Liter)
7. After 20 min, add 1 ml of spheroplast dilution
medium (4.3 M NaC1,27 mM KC1,80 mM MgSO,, 10 mM NaCl ................................. 250g
sodium citrate, 1.4 mM CaC12, 50 mM Tris-C1 [pH 7.41, MgSO, . 7H2O ......................... 20 g
15% [wt/vol] sucrose, 1% [wt/vol] Oxoid Peptone) to the Sodium citrate .......................... 3g
mixture and gently invert tubes to mix. KCl .................................. 2g
8. Centrifuge at 6,000 X g for 4 min and remove super- Nitrogen base (Sigma Y0626) . . . . . . . . . . . . . . 10 g
natant. Gently resuspend cell pellets in 1 ml of spheroplast Dropout formula (Sigma Y1501) . . . . . . . . . . . . 1.92 g
dilution medium, transfer to sterile test tubes, and incubate
the tubes with shaking at 42C overnight. pH 7.0 with NaOH
33. Genetics of Archaea 807

H. volcanii Complex Medium (per Liter) esting application for the GFP reporter system has been to
study proteolysis in H. volcanii (94). Finally, several mi-
NaCl ................................. 206g croarray studies have been published for Halobacterium sp.
MgS04 . 7H20 .......................... 37 g NRC-1, including one for anaerobic growth and another for
KC1 .................................. 3.7g UV response (68, 76).
Yeast extract ........................... 3g
Tryptone .............................. 5g
CaClz . 2 H z 0 (10%) .................... 5.0 ml 33.2. METHANOGENIC EURYARCHAEOTA
MnClz (75 mg/ml) ...................... 1.7 ml
1 M Tris-HC1 (pH 7.2) . . . . . . . . . . . . . . . . . . . 50 ml 33.2.1. Introduction
The methanogenic Archaea have a significant role in the
NOTE. global carbon cycle as microbial catalysts in anaerobic
degradative processes in habitats where 0 2 is not available.
For agar plates, add 20 g of Bacto-Agar per liter of The methanogens are a phylogenetically coherent group
medium. that currently includes more than 60 described species in
Autoclave at 15 lb/in2 for 20 min. five orders within the archaeal kingdom Euryarchaeota.
Cool agar media to 65C before addition of antibiotics They range from psychrophilic species from Antarctica that
and/or trace metals. grow at 1.7Cto extremely thermophilic species from deep
submarine vents that grow at 113C; acidophiles from ma-
33.1.6.2. Storage- of Strains rine vents that grow at pH 5.0to alkaliphiles from alkaline
Cultures may be stored for weeks to several months without lake sediments that grow at pH 10.3; species from freshwa-
extreme loss of viability, although extended storage may re- ter lake sediments that grow at saline concentrations less
sult in accumulation of mutants from IS element transposi- than 0.1 M to extreme halophiles from solar salterns that
tions and other DNA rearrangements. For long-term stor- grow at nearly saturated NaCl concentrations; autotrophs
age, cultures may be frozen at -70C after addition of that use only COz for cell carbon and methylotrophs that
glycerol to 15% (vol/vol) in liquid medium. Culturing con- utilize methylated carbon compounds. Despite the range
ditions and growth media for additional haloarchaeal and diversity of growth habitats where methanogens are
strains may be found in Halophiles (31). found, methanogens have one common attribute: they all
generate CH4 during growth.
33.1.7. Troubleshooting - Two tractable genetic systems are currently available for
High salinity of halophile media presents some unique the methanogenic Archaea within the Methanococcales and
problems for genetic studies. Salts inhibit gelling of agar, Methanosarcinales. The order Methanococcales includes ma-
making a relatively high concentration essential to obtain rine autotrophs that grow exclusively by C02 reduction
a solid surface. Salts also reduce surface tension of the with Hz. Morphologically, these species form irregularly
medium, making removal of bubbles more difficult. Extra shaped cocci. Instead of a rigid cell wall typical of the
care must be taken to avoid formation of bubbles when Bacteria, these species form an S-layer composed of a
pouring plates since their removal by flaming is less effec- paracrystalline array of glycoproteins and are subject to os-
tive. Another problem in culturing halophiles arises from motic lysis at NaCl concentrations below seawater. This
the contamination of some batches of peptone with bile order includes several mesophilic species, the moderate
acids, leading to growth inhibition and even cell lysis. thermophile Methanothernococcus thernolithotrophicus, and
Synthesis of purple membrane is especially sensitive to bile the extreme thermophiles Methanotomis igneus, Methano-
acids. For suspension of cells and transformation, the ionic caldococcus junnaschii, and Methanocaldococcus infernus. A
strength of the medium is critical to maintain integrity of genetic system has been developed for the mesophilic
spheroplasts. Even a small amout of lysis will result in sig- species Methanococcus mripaludis . Although genetic ap-
nificant reduction in transformation efficiency through the proaches were first developed with Methanococcus voltae,
release and precipitation of DNA. later investigators focused on M. mripaludis after the dis-
The selections for transformation work well, in general, covery of a native plasmid. As the genome of this species
although rare cultures may contain large numbers of drug- will soon be completed (http://www.genome.washington
resistant mutants and high background, resulting from .edu/UWGC/), this has become the preferred obligate
Luria-Delbruck fluctuations. This is true for both mevinolin hydrogen-utilizing species for genetic research.
and 5-FOA resistance. The presence of homologous DNA The order Methanosarcinales is the most catabolically di-
will lead to the integration of transforming plasmids at high verse species of methanogens. In addition to growth and
frequency, with a greater problem for larger regions of iden- methanogenesis by COz reduction with H2, some species
tity. Finally, the high mutation rate of some species and grow by the dismutation or splitting of acetate and by
strains can lead to genetic variation among populations. methylotrophic catabolism of methanol, methylated amines,
Strict microbiological practice, including purification of pyruvate, and dimethyl sulfide. Species in most genera are
strains and their long-term storage in frozen stocks, is es- either obligate methylotrophs or, in the case of Methanothrix
sential for genetic studies. spp., obligate acetotrophs. However, species of Methano-
sarcina can grow by all three catabolic pathways. All species
33.1.8. Recent Developments have a protein S-layer cell wall and grow as irregularly
The genome sequences of H. mrismortui and N. phrao- shaped cocci, but in low-saline medium some species also
nensis have recently been published (10, 36). Also, the grow as multicellular aggregates embedded in a heteropoly-
number of interesting mutants is increasing, e.g., mutants of saccharide matrix synthesized adjacent to the S-layer.
dimethyl sulfoxide reductase and its regulator dmsR (76); The discovery of a native plasmid in Methanosarcina
photolyase (phrl and phr2) gene mutants (9, 67) of Halo- acetivorans led to the development of a genetic system for this
bacterium sp. NRC-1; a radB mutant of H. volcanii (46); and metabolically diverse genus of hydrogen-utilizing, acetic-
rRNA operon deletions of H. mrismortui (115). An inter- lastic, and methylotrophic species. Although optimized for
808 MOLECULAR GENETICS

M. acetiworans, the genetic system can be used with most like (methanochondroitin) outer layer, causing them to
species. A slightly modified transformation system has grow as multicellular aggregates, which would normally
been developed for Methanosarcina maqei (34). Genomes preclude genetic manipulation. However, when t h e
for M. acetivorans C2A (39), M. mazei GO1 (32), and methanochondroitin-producing species are adapted and
Methanosarcina barkeri Fusaro have been completed grown at higher salt concentrations, they no longer gener-
(http://www.jgi.doe.gov). For overviews of genetic systems ate methanochondroitin and grow as individual cells with
for the methanogenic Archaea, see Lang and Ahring (64) only a n S-layer (106). For transformation of Methan-
Sowers and Schreier (110), and Tumbula and Whitman osurcina s spheroplasts are generated by suspending cells
(118).
Pi
in a Mg2 - ree sucrose buffer, which disrupts the integrity
of the S l a y e r (37). After transformation, the S l a y e r is re-
33.2.2. Strain Characteristics generated by resuspension of the cells in medium that con-
tains Mg2+. Although a functional restriction system has
33.2.2.1. M. acetivorans C2A not been reported in this species, a putative restriction
M. acetivorans C 2 A is an irregularly shaped, coccus-shaped modification has been identified in the annoted genome
cell isolated from a submarine canyon; it grows in a defined sequence. M. ucetivorans contains a 5.4-kb plasmid open
marine mineral medium with acetic acid, methanol, or reading frame encoding a putative RecA replication initia-
methylated amines as sources of energy for growth and tion protein associated with a rolling-circle mechanism of
methanogenesis (105). No exogenous growth factors are replication and a putative site-specific recombinase (7 1).
required. This species generates a protein Slayer rather This plasmid has been recombinantly modified to serve as
than a rigid cell wall. In addition to a n Slayer, several a shuttle vector for genetic transformation of most
other Methunosurcinu spp. synthesize a rigid chondroitin- Methunosurcinu spp (Table 3).

TABLE 3 Archaeal genetic vectors


Plasmids Features Reference
Halophilic Euryarchaeota
pWL104 Haloarchaeal-E. coli shuttle plasmid for Halobacterium and Haloferax spp.: has 63
multiple cloning sites and confers mevinolin resistance in haloarchaea and
ampicillin resistance in E. coli
pNG168 Haloarchaeal-E. coli shuttle plasmid for Halobacterium and Haloferax spp.: has 31
multiple cloning sites and confers mevinolin resistance in haloarchaea and
ampicillin resistance in E. coli
pUBP2 Haloarchaeal-E. coli shuttle plasmid for Halobacterium and Haloferax spp.: con- 18
fers mevinolin resistance in haloarchaea and ampicillin resistance in E. coli
pMSD2O Haloarchaeal-E. coli shuttle plasmid for Haloferax spp.: confers mevinolin- 53
resistance in haloarchaea and ampicillin resistance in E. coli
pHRZH Haloarchaeal rRNA gene replacement vector for Halobacterium spp.: confers 65
anisomycin and thiostrepton resistance
pMPK408/pSK400 Haloarchaeal ura3 suicide vector for Halobacterium gene knockouts for Halobac- 86, 119
cerium spp.: integrates into genome and confers uracil prototrophy and 5-FOA
sensitivity
pGB70 Haloarchaeal pyrE suicide vector for Haloferax spp.: integrates into genome and 16
confers uracil prototrophy and 5-FOA sensitivity
pXLNov Haloarchaeal bop gene expression vector for Halobacterium spp. used for green 83
fluorescent protein (GFP) fusion: confers resistance to novobiocin
pS07 Haloarchaeal sensory rhodopsin SRI (sop1) gene expression vector for 61
Halobacterium spp.: confers resistance to mevinolin
pBBEVl Haloarchaeal bop promoter expression vector for Halobacterium spp.: confers re- Berquist & DasSarma,
sistance to mevinolin unpublished data
pKJ408sfdx Haloarchaeal HtrI expression vector using super fdx promoter for Halobacterium 56
spp.: confers resistance to mevinolin
pSEl Haloarchaeal fdx-DHFR fusion gene for promoter analysis in Haloferax uolcanii: 26
confers resistance to trimethoprim
pSDl Haloarchaeal GPCR expression plasmid for Haloferax spp.: confers resistance to 85
trimethoprim
pENDS Haloarchaeal bop promoter-based GPCR expression plasmid in Halobacterium 14
spp.: confers resistance to mevinolin
pFL2 Haloarchaeal gas vesicle expression plasmid for Halobacterium spp.: confers re- 30
sistance to mevinolin
(Continued on next page)
Next Page

33. Genetics of Archaea 809

TABLE 3 Continued
Plasmids Features Reference

pFM104D Antigen display vector on gas vesicles for Halobacterium spp.: confers resistance 114
to mevinolin
pWL204 Haloarchaeal tRNA gene expression vector for Haloferax spp.: confers resist- 82
ance to mevinolin
Methanogenic Euryarchaeota
pJK 28 through -41 Methanoarchaeal-E. coli shuttle plasmids for site-directed gene disruption in 71
Methanosurcina spp.: do not replicate in Methanosarcina; have different h Z c l
polylinker cloning sites for cloning DNA homologous to the target gene;
confer puromycin resistance in methanoarchaea and ampicillin resistance in
E. coli
pDLT44 Methanoarchaeal-E . coli shuttle plasmid for gene expression/complementation 117
in M. maripuludis: autonomously replicates in M. maripuludis, has unique XbuI
and Sac1 restriction sites for cloning heterologous DNA, and confers puromycin
resistance in methanoarchaea and ampicillin resistance in E. coli
pWM309 through Methanoarchaeal-E. coli shuttle plasmids for gene expression-complementation 71
-321 in Methanosurcina spp.: autonomously replicate in Methanosurcina spp., have
different lac.21~polylinker restriction sites for cloning heterologous DNA, and
confer puromycin resistance in methanoarchaea and ampicillin resistance in
E. coli
pJK60 Methanoarchaeal-E. coli shuttle plasmid for in vivo random gene disruption in 126
Methanosurcina spp.: contains the mariner transposon and confers puromycin
resistance in methanoarchaea and kanamycin resistance in E. coli
pEA105 Methanoarchaeal-E. coli shuttle plasmid for gene expression-complementation Apolinario and
in Methanosurcina spp.: derivative of pWM3 15 with greater transformation Sowers,
efficiency in E. coli unpublished
pJK5 Methanoarchaeal-E. coli shuttle plasmid for site-directed gene disruption in 127
Methanosarcina spp.: pac-oriR6K-aph cassette plasmid with symmetrical XbuI
and EcoRI flanking restriction sites for cloning DNA homologous to the tar-
get gene.
pPB12 Methanoarchaeal-E. coli shuttle plasmid for gene expression-complementation 19
in Methanosurcina spp.: autonomously replicates in Methanosurcina spp. and
confers pseudomonic acid resistance in methanoarchaea and ampicillin resist-
ance in E. coli
pMipuid Methanoarchaeal-E. coli shuttle plasmid for assaying gene expression in M. 15
maripuludis: integration vector with EcoRI and NdeI restriction sites upstream
of uidA for detecting archaeal promoter strength by P-glucuronidase activity;
confers puromycin resistance in methanoarchaea and ampicillin resistance in
E. coli
pMudpur Methanoarchaeal-E. coli shuttle plasmid for in vitro directed gene disruption in 17
M. manpaludis genomic DNA: contains the Mu transposon and confers
puromycin resistance in methanoarchaea and chloramphenicol resistance in
E. coli
+
pWLG3O lac2 Methanoarchaeal-E. coli shuttle plasmid for assaying gene expression in M. 40
maripaludis: autonomously replicating reporter vector with NsiI restriction site
upstream of lac2cl for detecting archaeal promoter strength by P-galactosidase
activity
Hyperthermophilic Crenarchaeota
pAG I Ap' E. coli shuttle vector: butanol' benzyl alcohol' (alcohol dehydrogenase gene 5
from S . solfutaricus) Pyrococcus cryptic plasmid (pGT5) for S . ucidocaldarius
pEXSs Ap' E.coli shuttle vector: hygromycin B'(mutant hygromycin phosphotrans- 20
ferase gene from E. coli), SSVI viral origin for S. solfataricus Gtheta
pNOB8-lacs A self-transmissible plasmid derived from pNOB8 with the P-glycosidase gene 35
(lacs)fused to ribosomal protein S12 promoter from S. acidocaldariusand in-
serted at unique Sul I site; for use in s. solfataricus P1
pKMSD48 Ap' E. coli shuttle vectors: SSVI virus hybrid. For use in S. solfatukus 112
Genetic Manipulations Using Phages
GRAHAM F. HATFULL. DEBORAH JACOBSeSERA. MICHELLE H . LARSEN.
AND WILLIAM R. JACOBS. JR.

34.1. INTRODUCTION AND OVERVIEW OF PHAGE GENETICS AND


ITS APPLICATIONS ....................................... 825
34.1.1. The World of Bacteriophages ............................ 825
.
2. The Power of Phage Genetics ............................ 826
. .....
3. The Role of Phages in Mycobacterial Genetics: A Case Study 827
34.2. PHAGE ISOLATION ....................................... 827
34.2.1. Sample Collection .................................... 827
.
2. Identification of Plaques................................ 828
.
3. Plaque Purification ................................... 828
.
4. Preparation of a Working Stock Lysate ..................... 828
.
5. Growth of High-Titer Stocks ............................ 828
.
6. Precipitation with PEG 8000 ............................ 828
.
7. CsCl Equilibrium Density Centrifugation ................... 829
34.3. PHAGE GENOMIC CHARACTERIZATION .................... 829
34.3.1. Preparing Phage DNA ................................. 829
.
.2 Restriction Analysis ................................... 829
.
3. Construction of Genomic Libraries ........................ 829
.
4. Sequencing and Assembly ............................... 829
.
5. Genome End Analysis ................................. 830
34.4. SHUTTLE PHASMID AND THEIR CONSTRUCTION ............ 830
34.4.1. Overview .......................................... 830
.
2. Generation of a Library of Cosmid Insertions in a
Mycobacteriophage Genome ............................. 831
.
3. Identification of Shuttle Phasmids Following Transfection of
M.smegmatis ....................................... 831
.
4. Cloning Genes into Shuttle Phasmids ...................... 831
34.5. GENE DELIVERY USING PHAGES ........................... 832
34.5.1. Reporter Gene Delivery ................................ 832
.
2. Conditionally Replicating Mycobacteriophages and Transposon
Delivery ........................................... 832
.
3. Allelic Exchange by Specialized Transduction ................. 832
34.6. OTHER PHAGE-DERIVED GENETIC TOOLS ................... 832
34.6.1. Construction of Integration-Proficient Plasmid Vectors .......... 832
.
2. Recombineering: Phage-Mediated Recombination for Allelic
Exchange ........................................... 833
.
3. Generalized Transduction............................... 834
34.7. RECIPES ................................................ 834
.
34.7.1. Culture of M smegmatis ............................... 834
. . .....................
2. Preparation of E coli Competent Cells 834
.
3. Transformation of M. smegmatis.......................... 834
.
4. Media and Reagents ................................... 835
34.8. REFERENCES ............................................ 835

34.1. INTRODUCTION A N D OVERVIEW OF are particulate in nature. Thus at the appropriate concentra-
PHAGE GENETICS A N D ITS APPLICATIONS tion. individual areas of killing or plaques appear on bacte-
rial lawns. Each plaque is derived from a single submicro-
.
34.1 1. The World of Bacteriophages scopic entity that had the ability to replicate on its bacterial
Bacteriophages were discovered by Frederick Twort ( 1916) host. The next few decades saw continued advances in un-
and Felix dHerelle (1917) as agents that have the ability to derstanding these viruses. although it was not until the
kill bacteria (49). The nature of the killing property re- development of the electron microscope in the 1940s that
mained unclear for many years. although the dilution exper- the morphologies of phages became apparent (44). With the
iments of dHerelle provided compelling evidence that they description of the structure of DNA following shortly after

825
826 rn MOLECULAR GENETICS

(Sl), it became clear that the ability to quickly and simply comes established and maintained within the cell for many
propagate phages at high concentrations would help to un- generations (and may either integrate or replicate extra-
ravel the secrets of the genetic code and provide invaluable chromosomally) and the lytic phage functions are switched
tools for manipulating DNA molecules. off. Temperate phages form turbid plaques that arise from a
More recently, the vastness of the phage population has combination of killed cells and lysogens that are now im-
come into sharper focus (15). Determination of the con- mune to subsequent reinfection by the phage. A variety of
centrations of phages in a multitude of environmental Sam- plaque morphologies are shown in Fig. 1. In practice, a s i p
ples shows that they are commonly present at lo6 to 107/ml, nificant number of phages form hazy plaques (intermedi-
and usually about 10-fold more prevalent than bacteria ate between turbid and clear) and cannot readily be de-
(15). Extrapolation of these numbers suggests that there are scribed as either truly temperate or lytic. For example, many
about lo3 phage particles in the biosphere, making them mycobacteriophages form hazy plaques and encode integra-
an absolute majority of all biological forms (52). Individual tion functions in their genomes. A plausible explanation is
phages show specificity for particular hosts, and the spec- that such phages are competent to form stable lysogens, but
trum of available hosts is defined as the host range; some perhaps not necessarily in all bacterial hosts. Thus phages
phages have extremely narrow host ranges, such as a single may exhibit different growth cycles depending on which
serovar, while others are broad and can infect many differ- bacterial host they are infecting.
ent bacterial species. The genetic complexity of this huge
population remains unclear, although the genomic charac- 34.1.2. The Power of Phage Genetics
terization of about 300 phage genomes shows that they are The term phage genetics comes with multiple meanings
extremely diverse and are typically constructed with mosaic depending on the perspectives of the reader. In the more
architectures (10, 12, 20, 37). This unusual genetic texture traditional and narrower application of the term, it is used
suggests that phages have existed for several billion years to describe the isolation, mapping, and functional analysis
and that horizontal genetic exchange has played a major of bacteriophage mutations. In the broader context, it refers
role in their evolution (18-20). Particular phage types thus to the use of bacteriophages to genetically manipulate a
cannot easily be classified within any generally accepted bacterial host, and as sources for the development of genetic
species concept, and their taxonomy is fraught with com- tools. It is in this broader context that we will use the term
plications (31). phage genetics in this chapter.
While phage morphologies are diverse, the population is The role of phage genetics in microbiology has expanded
dominated by doubled-stranded DNA (dsDNA) tailed with the realization that we understand only a small frac-
phages. Typically, these have a DNA-containing head with tion of bacterial diversity. Microbes such as Escherichia coli
icosahedral symmetry attached to a tail, and the viruses can and Salmonella have been deeply studied and to great effect,
be grouped into three main classes depending on the type of but as our attention has turned to the broader bacterial
the tail: Podoviridue contain short stubby tails, Siphoviridae community, new bacteria have brought new genetic chal-
have long flexible tails, and Myoviridue have contractile lenges. Fortunately, phages have proven to be great friends
tails (1). Most phages can also be divided into those that are of the microbiologist for a multitude of reasons. First,
temperate and those that are lytic. Lytic (or virulent) phages can be isolated for virtually any bacterial species
phages usually have only a single outcome upon infection of that can be propagated in the laboratory and from which
a bacterial host: reproduction of the virus and lysis of the smooth lawns can be grown on agar plates. Second, these
bacterium. As a consequence, plaques appear as perfectly phages can be propagated to high titer (-1O1/m1) without
clear areas of infection, within which all of the bacterial any detailed knowledge of replication systems, antibiotic
cells are dead. In contrast, there are two alternative out- markers, or transformation methods. Third, DNA can be
comes of infection by a temperate phage. One is the same extracted from these particles and characterized genomi-
process as occurs with lytic phages, but the second is the es- cally. Fourth, phage genomes are replete with genetic fea-
tablishment of lysogeny, in which the phage genome be- tures that are of great utility to the genetic engineer, and

FIGURE 1 Mycobacteriophage plaque morphologies on M. srnegmutis lawns. (A) Small clear


plaques (Mycobacteriophage Oasis). (B) Large clear plaques (Mycobacteriophage Hammer). (C)
Turbid plaques (MycobacteriophageRedRock).
34. Genetic Manipulations Using Phages a 827

their modular nature means that these can be harvested ria. Furthermore, genes can be introduced this way and
from the genome and employed to develop genetic tools. their expression and phenotypic consequences evaluated by
Last, the high efficiency of phage infection provides a screening without any need for selection, especially when
means of introducing recombinant molecules to every cell temperate phages are used (47). A notable example is that
within a bacterial population. antibiotic resistance genes can be introduced and their util-
In this chapter we will describe some common phage ity can be determined without any prior knowledge of
methods and approaches that can be applied to virtually any whether and how they work in that bacterial host. Shuttle
bacterial host. However, we will utilize an example that we phasmids thus helped in showing how DNA can be effi-
are familiar with-the development of mycobacterial genet- ciently introduced into mycobacteria and what drug resist-
ics-to illustrate this awesome power of phage genetics. ance genes might be useful and how. Further details in shut-
tle phasmid construction are provided below.
34.1.3. The Role of Phages in Mycobacterial The general utility of shuttle phasmids is illustrated by
Genetics: A Case Study three key applications. First, they can be used to efficiently
The predominant role of the mycobacteria in human health introduce transposons, taking advantage of the high effi-
at the global scale demands the development of a facile ge- ciency of infection to overcome the inherent low frequency
netic system for understanding and manipulating them; of transposition (4). Second, they can be used to introduce
thus it is an obvious system for phage genetics. The World reporter genes such as the firefly luciferase, where the effi-
Health Organization estimates that one in three of all peo- ciency of infection has the potential to enable the simple
ple are infected with Mycobacterium tuberculosis, and each diagnosis of mycobacteria (and drug susceptibilities) within
year more people die of these infections than because of any a clinical sample (23,45). Third, the efficiency of infection
other single infectious agent (11, 13). However, these num- can be utilized to overcome the inherently low frequencies
bers also illustrate what a formidable opponent M. tubercu- of homologous recombination when constructing mutants
losis is; most people that it infects do not die of the disease by allelic exchange (3).
or even get serious symptoms. Instead, a lifelong latent in- While mycobacteriophage shuttle phasmids have
fection is established, a time bomb that can explode into proven critical to the development of mycobacterial genet-
full-blown tuberculosis whenever the immune system loses ics, they represent only one of many examples of phage
its full proficiency. genetics in the mycobacterial system. The field of myco-
Twenty years ago, two main challenges faced the myco- bacteriophage genetics has been significantly fueled by the
bacterial geneticist. The first was the pathogenicity of M. isolation and genomic characterization of more than 30 in-
tuberculosis that warrants growing it under strict biosafety dividual phages, and these have served as a genetic reser-
conditions (BSL-3), and the second was its extremely slow voir for tool development (17). For example, phage-derived
growth rate with a doubling time of 24 h. (Consider that a integration vectors are useful for constructing single-copy
typical colony has lo8 bacterial cells, which would require stable recombinants (28, 32), and recombinase proteins
30 generations of growth from a single cell, thereby requir- can be identified and adapted for mycobacterial recombi-
ing 30 days to obtain a M. tuberculosis colony.) The good neering (50a).
news is that it grows on defined medium and there are a va- In this chapter, we will describe how phages can be iso-
riety of possible surrogate bacteria that grow much faster lated, purified, and genomically characterized, how shuttle
and are nonvirulent (such as Mycobacterium smegmatis). But phasmids can be constructed and utilized, and how phages can
in 1986 no simple and efficient methods existed for intro- be used to construct a variety of genetic tools. The mycobac-
ducing and propagating recombinant DNA molecules or for terial context provides specific examples, but the approaches
the efficient isolation of mutants. Bacteriophages played a are adaptable to many other bacterial systems. Finally, it will
central role in overcoming these hurdles (22). become apparent that the potential for phage genetics in any
Several collections of mycobacteriophages had been as- bacterial host is vast, and the approaches and methods de-
sembled previously with a view to using their host-range scribed here represent just a small part of what bacteriophages
specificities for the typing of clinical isolates (26, 27). One have to offer to the microbial genetic engineer.
of these phages had been shown to mediate generalized
transduction in M. smegmatis (40) (although none in M. tu-
berculosis), and Tokunaga and colleagues achieved uptake of 34.2. PHAGE ISOLATION
phage DNA (i.e., transfection) by M. tuberculosis (50).
However, these did not immediately facilitate the develop- 34.2.1. Sample Collection
ment of methods for introducing foreign DNA into my- The process used to isolate phages from environmental sam-
cobacteria. ples naturally depends on the ecology of the bacterial host
This important advance was accomplished through the and the environments where the host and its phages usually
creation of novel chimeric molecules termed phasmids, reside. To isolate phages, simply collect samples from a va-
which have the ability to grow in E. coli as large extrachro- riety of places where the bacterial host is likely to be found,
mosomal plasmid molecules and as bacteriophages in my- extract the phages using a simple buffer, and plate a portion
cobacteria (25). These can be generated from any phage of this directly on lawns of the bacterial host. A n amplifi-
genome that is appropriately sized (40 to 50 kbp) by ligation cation step can be included if desired, but we typically avoid
into an E. coli cosmid vector, packaging into phage lambda it to prevent biasing the collection of phages toward those
particles in vitro, and infection of E. coli; DNA can then that flourish under laboratory conditions and out-compete
be prepared from this library of recombinants and used to all others. Detailed protocols for isolating phages by ampli-
transfect mycobacteria (see Fig. 2) (24). Phage lysates can fication have been described elsewhere and will not be pre-
then be prepared and used to infect any suitable myco- sented here in any detail.
bacterial host. The key advantage of these phasmids is that A typical procedure for isolating mycobacteriophages is
they can simply be genetically manipulated in E. coli and as follows. First, gather a small sample from the environ-
used as vehicles to introduce foreign DNA into mycobacte- ment (such as from soil, compost, ponds, etc.) into a 15-ml
828 w MOLECULAR GENETICS

screw-cap conical tube, typically enough to fill about one- dividual plaques within the spot will be observed. This is con-
third of the tube. If the sample is solid, add an approximately venient if testing large numbers of samples, since many can
equal volume of phage buffer (make sure that the buffer in- be added onto a single agar plate. A positive spot test will
cludes 1 mM CaC12). It is helpful to avoid adding too much usually show complete cell killing in the entire spotted drop.
liquid since this will dilute the sample too much; if too little
buffer is added, it will be difficult to subsequently clarify the 34.2.3. Plaque Purification
sample. Thoroughly mix the sample by shaking and then let If the preceding procedures are successful, then it is neces-
it sit for about 20 min; most of the solid material will have sary to ensure the homogeneity of the phage sample by per-
settled to the bottom of the tube. Remove about 1 ml of liq- forming at least three rounds of plaque purification-more
uid from the liquid phase by drawing up the sample into a won't do any harm! To do this, pick a single plaque from a
1-ml syringe, attach a 0.22-pm-pore-size filter to the syringe, plate by using a plastic pipette tip (or from a spot from a
and collect the filtered sample in a 1.5-ml microcentrifuge spot test) and twirl into 100 p1 of phage buffer. (Be cautious
tube. The sample is now clear and sterile. if the spot test plate is used for this step. I t may be best to
go back to the sample used to make the spot test to avoid
34.2.2. Identification of Plaques contamination from other spots). Prepare serial dilutions of
The specific conditions that are used to grow a bacterial the sample in phage buffer-typical dilutions are lop2,
host will obviously depend on what host is used. The fol- 10-4-and add 10 pl of each dilution to 0.5 ml of
lowing description details the process of isolating mycobac- plating cells. After allowing 15 to 30 minutes for adsorp-
teriophages by using M. smegmatis as a host, but it can be tion, add 4.5ml of top agar and pour onto plates. Allow agar
readily adapted to other hosts. to harden, invert plates, and incubate until a lawn has
In sterile tubes, add about 50 pl of sterile phage sample grown and plaques are visible. When starting with a single
to 0.5 ml of a culture of saturated M. smegmatis. Set up a plaque, between lo4 and 106particles are transferred into
parallel sample to act as a negative control to which no the original tube. One reason why it is important to do
phage sample is added; simply add 50 p1 of phage buffer. Let many rounds of plaque purification is that the phage parti-
these sit on the bench for 15 to 30 min to allow the phage cles can continue to diffuse through the top agar layer after
to adsorb to the bacteria. During this time, melt some top the lawns have fully grown and the plaques do not appear to
agar by microwaving a 50-ml bottle of 0.7% top agar until get any larger. Thus, there is always a risk of picking up par-
boiling, then adding an equal volume (50 ml) of medium to ticles other than those that are in the intended plaque.
bring the final agar concentration to about 0.35%; maintain Avoid extended incubation or wait times between rounds of
the temperature at about 55C; we typically add CaClz to a purification to prevent this problem.
final concentration of 1 mM after the agar has been boiled
and diluted. Approximately 4.5 ml of this top agar is then 34.2.4. Preparation of a Working Stock Lysate
added to each sample tube, and the entire contents is To prepare a lysate as a working stock, use a freshly prepared
poured onto a standard agar plate. For this step to work well, plate from the final plaque purification process that con-
it is important that the plates are relatively freshly poured, tains just enough phage to have cleared the entire plate. To
are prewarmed to either room temperature or 37"C, and are this add 5 ml of phage buffer and swirl it so that the whole
not too wet (i.e., fresh but not too fresh!). Immediately after plate is covered; let this stand at room temperature for 2 to
pouring the contents out, swirl the plate to make sure it is 3 h (or overnight in the cold). Collect the liquid using a
evenly covered and then let stand until the top agar layer Pasteur pipette and filter through a 0.22-pm-pore-size filter
hardens (usually about 15 to 30 min). Although neither of into a clean sterile tube. Determine the titer of this by mak-
these actions appears to be difficult, they are critical to suc- ing and plating serial dilutions as described above.
cess. A minimal amount of swirling will distribute the agar
and sample over the entire agar surface, although it is im- 34.2.5. Growth of High-Titer Stocks
perative that the plates are not moved during cooling, to For isolation and characterization of phage particles and
ensure that the top agar solidifies uniformly. Turn the plate phage DNA, it is usually necessary to prepare a high-titer
over carefully, and incubate at 37C Overnight or until a stock and then to purify and concentrate the phage. While
bacterial lawn has grown across the entire plate. there are many possible ways to do this, a typical approach
When the bacterial lawn is fully grown, examine it for is to prepare a plate lysate as described above but using 30
plaques. Since amplification has not been done in this pro- large petri plates. For each of these large plates, use 1 ml of
cedure, the number of plaques is usually small and highly host bacterial culture, mixed with just enough phage parti-
variable. Frequently, only about 3 to 10% of all the samples cles to give complete clearing of the bacterial lawn, and 9
give any plaques at all, and when they do, it is usually only ml of top agar. After the plates have grown, flood each plate
one or two plaques; occasionally, a sample gives a couple of with 10 ml of phage buffer and collect the phage as de-
dozen plaques or more. It is usual to also see a few bubbles scribed above, typically generating about 300 ml of lysate.
in the top agar, and these can easily be mistaken for Transfer this to a centrifuge bottle, spin for 5 min at 3,500
plaques! If several plaques appear to have different plaque x g, and collect the clear supernatant.
morphologies (such as large, small, clear, turbid) (Fig. l ) ,
then these may be different and can be picked for further 34.2.6. Precipitation with PEG 8000
testing. To precipitate the phage particles, add polyethylene glycol
To confirm that plaques are real (rather than bubbles), use 8000 (PEG 8000) to a final concentration of 10% and NaCl
a plastic pipette tip to touch the putative plaque, and then to a final concentration of 1 M. To do so, measure the
twirl the tip in 100 pl of phage buffer and perform a spot test amount of the lysate (e.g., 275 ml) and then add the appro-
on a fresh lawn of bacteria. To do the spot test, prepare the priate amount of solid PEG 8000 (e.g., 27.5 g in this exam-
lawn as described above for the no-phage control, let the agar ple) and solid NaC1. Add a stirrer bar and stir overnight in
harden, and then spot with a 5-pl drop of phage sample. the cold.
Allow to dry and incubate at 37C overnight. If the sample The following morning, collect the phage pellet by cen-
contains phage particles, then either complete killing or in- trifugation at 3,500 x g for 5 min and pour off the super-
34. Genetic Manipulations Using Phages 829

natant; drain as well as possible. Resuspend the pellets in 10 recommended conditions, and the products can be analyzed
ml of phage buffer by running the sample up and down a by agarose gel electrophoresis. Note several points. First,
pipette. When as much phage sample is resuspended as pos- many phage genomes contain short single-stranded DNA
sible, clarify by spinning the sample again at 3,500 X g for complementary ends, which can anneal and remain an-
10 min. There is always a significant amount of insoluble nealed during electrophoresis. Thus different restriction
debris, but the phage usually suspend well. patterns may be seen depending on how you treat the sam-
ple prior to electrophoresis. If you specifically want to see
34.2.7. CsCl Equilibrium Density Centrifugation the end fragments, then after restriction, heat the sample to
Once the precipitated phage is resuspended, it can be fur- 75C for 10 min and snap cool it on wet ice. Second, some
ther purified and concentrated using CsCl equilibrium den- or many enzymes may not digest phage DNA if the DNA is
sity centrifugation, although there are several reasonable al- modified, and using a variety of different enzymes is helpful.
ternative approaches (such as step gradients and high-speed Last, since phage genomes can be quite large, it is helpful to
centrifugation). To do this, add solid CsCl to the lysate and use agarose gel electrophoresis conditions that can separate
dissolve, adding about 8.5 g of CsCl for each 10 ml of lysate, relatively large DNA fragments.
and check the density using a refractometer. Alternatively,
the density can be determined by simply weighing 1 ml of 34.3.3. Construction of Cenomic libraries
the sample. Adjust the density to 1.5 by adding either a For genomic sequencing, prepare a plasmid library of DNA
small amount of a saturated CsCl solution (to increase the fragments derived by hydrodynamic shearing of phage genomic
density) or phage buffer (to reduce it). DNA. We use a Hydroshear instrument (GeneMachines
Transfer the solution to an appropriate centrifuge tube Inc., San Carlos, CA) that has been calibrated to shear the
(such as heat-sealable tubes) and spin at 38,000 rpm at DNA predominantly into 1- to 3-kbp pieces. After the
10C for 16 to 24 h. The phage should appear as an opaque shearing, the DNA is repaired by incubation with both T4
band positioned approximately one-third or one-half of the DNA polymerase and Klenow according to the manufac-
way up the tube; there is usually a small pellet of debris at turers instructions, and ethanol precipitated. After washing
the bottom. Collect the phage band by carefully placing the (70% ethanol), drying, and resuspending in a small volume
tube in a clasp on a ring stand, excising the top of the tube of TE, the entire sample is loaded onto a wide-slot 0.7%
by using a hot scalpel, and inserting a needled syringe (18 agarose gel along with a size marker. After electrophoresis,
gauge) through the side of the tube about 4 to 5 mm below a gel slice containing 1- to 3-kbp fragments is excised and
the band. With the needle turned so that the bevel is facing the DNA is recovered.
upward and the needle of the syringe is pointing upward, For generating genomic libraries, we typically use one of
carefully draw off the band from beneath. It is helpful to try the pBluescript vectors, such as pBluescriptI1 SK+,that has
to remove as small a sample as possible to avoid diluting it. been cleaved to completion with EcoRV and dephosphory-
Phage particles usually store well in CsCl at 4C,and this is lated with CIAP. Ligations are performed in small volumes
a reasonable option for long-term storage. But for further under conditions favoring blunt-end ligation, using differ-
processing, dialyze away the CsCl by transferring a portion ent quantities of the size-fractionated DNA (such as 0.5 ~ 1 ,
to a dialysis tube and dialyze against two changes of cold 1 pl, or 2 pl from a total of 15 to 20 ~ 1 and ) then trans-
phage buffer for several hours. formed by electroporation into a suitable E. coli strain such
as E. coli XL1-Blue. The degree of success can be deter-
mined by the ratio of white and black colonies on selective
34.3. PHAGE G E N O M I C CHARACTERIZATION plates containing S-Gal indicator (or blue colonies using X-
Gal). With a good preparation of vector DNA, self-ligation
34.3.1. Preparing Phage DNA in the absence of phage DNA generates only a few black
DNA can be recovered from a concentrated phage stock by colonies and no white ones. In the presence of phage DNA,
several rounds of phenol extraction. Typically, begin with a similar number (or a small increase) of black colonies and
about 0.5 ml of concentrated phage, add an equal volume of a significant number (dozens or hundreds) of white colonies
buffer-saturated phenol, and mix by inversion. The phases is desired. If one particular amount of phage DNA yields op-
can then be separated by centrifugation in a microfuge for timal numbers of white colonies, the remainder of the liga-
about 2 min at top speed (-14,000 K rpm). Remove the top tion mix can be electroporated to generate sufficient white
aqueous phase to a clean tube; avoid as much of the white colonies to complete a genome-sequencing project, typi-
particulate material at the interface as possible. Repeat the cally 300 to 600 depending on the size of the genome. The
phenol extraction several more times, and also backextract presence of recombinants among the white colonies can be
the phenol layers. All aqueous phase samples are then com- determined by growing up a small number (e.g., 12), isolat-
bined, and the DNA is precipitated by adding 0.1 volume of ing plasmid DNA, and analyzing by restriction and agarose
3 M sodium acetate and 2.5 volumes of cold ethanol. Mix gel electrophoresis.
well and freeze at -20C for several hours. Collect the
DNA by centrifugation, remove the supernatant, and wash 34.3.4. Sequencing and Assembly
at least once with 70% ethanol. Allow the pellet to air dry Large numbers of recombinant plasmids can be prepared by
and resuspend the DNA in a small volume (100 to 200 pl) growing them in blocks of 96 samples, and the DNA
of Tris-EDTA (TE).The DNA can be slow to dissolve, and preparation is greatly simplified by using a robot such as the
if necessary you can speed it up by a brief incubation at Qiagen BioRobot 9600. Blocks of DNA samples can then
60C. Determine the DNA concentration by using a spec- be used to perform standard sequencing reactions as de-
trophotometer. scribed by the manufacturer, cleaned up to remove excess
dyes and/or primers, and analyzed using an automating
34.3.2. Restriction Analysis DNA sequencer such as AB13730. Each block can be se-
Restriction enzyme analysis is useful for evaluating the quenced using both forward and reverse primers. It is im-
uniqueness of the phage genome. Phage DNA can be cut portant to sequence sufficient blocks to provide good cov-
with restriction enzymes according to the manufacturers erage and to minimize the number of weak areas that need
830 MOLECULAR GENETICS

to be worked on subsequently. When the sequence coverage bases away) and pointing outward; use these to extend on
is good (-sevenfold redundant), oligonucleotide primers phage DNA templates.
extended on phage DNA template is normally the quickest
route to completion of the project.
A variety of programs are available for the assembly of se- 34.4. SHUTTLE PHASMIDS AND THEIR
quence fragments into a final single contiguous sequence. CONSTRUCTI 0N
The Phred/Phrap/Consed package is one that we have found
to be simple and effective to use; besides providing a reliable 34.4.1. Overview
assembly, it can be used to simply design primers for com- Shuttle phasmids (part phage, part plasmid) are phage-
pletion of the weaker areas as well as the genome termini. cloning vectors for mycobacteria that also shuttle to E. coli
(Fig. 2). They are chimeric molecules made up of a trun-
34.3.5. Genome End Analysis cated mycobacteriophage genome cloned into an E. coli-
Some phage genomes contain defined ends, while others bacteriophage cosmid. They can be packaged into bacterio-
have multiple possible ends such that the genome assembles phage heads as cosmids, thereby allowing for the use of
into a circle. If the assembly circularizes, then it is helpful to bacteriophage in in vitro packaging mixes. Upon transfec-
organize the final sequence into a linear array with an arbi- tion of the fast-growing M. smegmatis, these chimeric DNA
trarily defined left end; if a terminase gene can be identi- molecules become packaged into mycobacteriophage parti-
fied, then its left end is a suitable choice for this. If the cles that can then infect M. tuberculosis, Mycobacterium
genome has defined ends, then this often becomes apparent bouis, BCG, and many other mycobacteria. TM4 mycobac-
from the assembly, since the ends tend to be overrepre- teriophage shuttle phasmids enabled the introduction of
sented (as there are two fixed ends among the sheared DNA foreign DNA into both fast- and slow-growing mycobacteria
population). The simplest way to define the ends precisely for the first time (25). Moreover, the generation of temper-
is to design primers that are close to the ends (50 to 100 ate shuttle phasmids from mycobacteriophage L5 allowed a

Pacl

Purify cosmid
DNA

Transduce E. coli

Plaques on M. s ~ e ~ ~ a f ~ s

FIGURE 2 Construction of shuttle phasmids. Shuttle phasmids are chimeric molecules that
replicate as extrachromosomal plasmids in E. coli and as phages in another bacterial host.
Construction details are provided in the text.
34. Genetic Manipulations Using Phages 831

functional selectable marker to be identified for mycobacte- in a mycobacteriophage genome. A n easy way to think
ria, which led to the development of the first transforma- about this is to imagine the phage genome as representing
tion system for mycobacteria (47). For organisms for which the outline of a clock, with the mycobacteriophage cohe-
genetics is nonexistent or limited, shuttle phasmid con- sive end site (cos) positioned at 12 oclock. Then imagine
struction provides a powerful approach. that each cosmid is inserted at some minute position around
Shuttle phasmids cannot be made from all phages. For the clock. Cosmid A may have the cosmid inserted at 2
example, they cannot be easily made with a phage whose oclock. Cosmid B may have the cosmid inserted at 7 oclock
genome is readily expressed in E. coli, such as E. coli bacte- on the genome. A viable shuttle phasmid will be one in
riophage or Salmonella phage P22, since the multicopy plas- which the cosmid is inserted in a nonessential region of the
mid is usually toxic to E. coli due to the expression of phage phage, and this should produce plaques upon transfection of
genes. Mycobacterial chromosomal DNA and most myco- M. smegmatis. If cosmid A has an insertion in a DNA repli-
bacteriophage genomes are not readily expressed in E. coli cation gene, the phage will not likely yield viable phages.
because of the inability of the E. coli transcriptional ma- However, if cosmid B has the cosmid inserted at a nonessen-
chinery to recognize high G +C% mycobacterial promoters tial accessory region, it should yield plaques..From a library
(21). Based on this attribute, it should be possible to make of thousands of insertions, one can simply identify those in-
shuttle phasmids from any high G + C % phage, including sertions in the nonessential region by transfecting the cos-
those from any Actinomycetes family members including mids into M. smegmatis and looking for plaques (Fig. 2).
Mycobacterium, Streptomyces, Rhodococcus, Norcardia, and A general protocol for characterizing shuttle phasmids
Corynebacterium. In addition, this might also work for or- after transduction of M. smegmatis includes: (i) isolate cos-
ganisms from the high-G+C% gram-negative bacteria like mid DNA from pooled E. coli transductants as described in
Pseudomonas or Caulobacter. In contrast to a similar phas- section 34.4.2, (ii) electroporate pooled cosmid DNA into
mid +C31 vector (42), shuttle phasmids also incorporate a M. smegmatis and mix electroporated cells with M. smegma-
bacteriophage cos sequence, which allows for their packag- tis cells on a phage plaque lawn, (iii) pick and purify result-
ing in vitro into bacteriophage heads. This addition facili- ing plaques, (iv) make phage stocks and isolate the result-
tates the genetic manipulation of shuttle phasmids in E. ing phage DNA, (v) ligate the putative shuttle phasmid
coli. Shuttle phasmids have been successfully constructed DNA to form concatemers and in vitro package with
from the lytic mycobacteriophages TM4 (25) and D29 (36) lambda mix, and (vi) transduce E. coli and select for
and the temperate mycobacteriophages L5 and L1 (47). ampicillin-resistant colonies. Ampicillin-resistant colonies
in E. coli indicate that the DNA fragment isolated from a
34.4.2. Generation of a library of Cosmid phage particle can replicate in E. coli as a cosmid.
Insertions in a MycobacteriophageGenome Surprising differences occurred in the plaques obtained
The successful generation of a shuttle plasmid from a my- from TM4 compared with L5 and D29. The library of TM4
cobacteriophage genome requires the insertion of an E. coli cosmids yield hundreds of plaques, but most of these plaques
cosmid in a nonessential region of the phage genome (Fig. resulted from the propagation of wild-type phage. Plaque
2). This can be achieved by generating a library of phage hybridization with the cosmid revealed that only 1 in 400
genome insertions into a cosmid vector in E. coli, followed plaques had cosmid DNA. It is likely that the wild-type
by selection for those recombinants that yield viable phage TM4 phages had arisen by recombination, since it was
particles following transfection of the library (25). A suit- highly likely that any one of the transfected M. smegmatis
able phage genome must contain cohesive ends (a headfull cells got more than one cosmid molecule. Consider if a cell
packaging phage would be difficult to use) and a genome received two cosmids, each with an insert in an essential re-
size of 40 to 60 kb (capable of being packaged into bacte- gion. If the nonessential regions did not overlap and re-
riophage lambda heads in vitro). The steps to construct a li- combination occurred, one recombination product would
brary of cosmid insertions in a mycobacteriophage genome be a wild-type TM4 phage. This possibility has been rigor-
include: (i) ligation of purified mycobacteriophage genomic ously demonstrated (]. C. van Kessel and G. E Hatfull, un-
DNA to form concatemers, (ii) partial digestion of con- published communication). In contrast, if L5 or D29 are
catemers with Sau3A to generate 40- to 50-kb-long frag- used, the number of plaques obtained is less, but more than
ments, (iii) ligation of large Sau3A fragments to arms of a 95% have cosmid inserts. The working hypothesis is that
double cos vector, (iv) package ligated DNA with lambda in TM4 possesses a phage-mediated recombination system.
vitro packaging mix, (v) transduction of packaging mix into This system may contribute to the success of allelic ex-
lambda-susceptible E. coli strain, and (vi) characterization change in M. tuberculosis, though these studies are ongoing.
of DNA recovered from individual transductants to confirm Regardless of the phage recombination system, shuttle
the presence of phage DNA. A good shuttle phasmid library phasmids have been obtained from three different myco-
will have two key features: most of the transductants will bacteriophages to date.
contain phage DNA, and cosmid insertions into the my-
cobacteriophage genome will occur around the entire 34.4.4. Cloning Genes into Shuttle Phasmids
genome. Each insertion point for an E. coli cosmid on the Phage delivery of DNA represents the absolute most eRi-
mycobacteriophage genome should also be accompanied by cient way to introduce DNA into any bacterial cell. Based
a small deletion of nonessential phage sequences. See on the ease of cloning genes into shuttle phasmids in E. coli,
Jacobs et al. (23) for further explanation. The end result of and the ability to generate TM4 phage particles of packaged
this genomic cloning is a library of insertions of an E. coli shuttle phasmid DNA, shuttle phasmids are ideal for deliv-
cosmid around the mycobacteriophage genome (Fig. 2). ery of DNA to mycobacterial cells. Shuttle phasmids have
been useful for delivering reporter genes, transposons, or al-
34.4.3. Identification of Shuttle Phasmids Following lelic exchange substrates. The cloning of any gene into a
Transfection of M. smegmafis shuttle phasmid is a straightforward task that involves the
In section 34.4.2, we described a means of generating a li- replacement of the existing cosmid with a cosmid of choice.
brary of cosmid insertions with simultaneous small deletions The steps involved in cloning genes into shuttle phasmids
832 I MOLECULAR GENETICS

include: (i) preparation of the plasmid form of the shuttle mids, chemical modification of allelic exchange substrates,
phasmid in E. coli, (ii) digestion of both the shuttle phasmid and specialized phage transduction (3). Both suicide plas-
and the new cosmid with Pad, (iii) ligation of equimolar mids and chemical modifications of plasmids have the ad-
amounts of phasmid and new cosmid, (iv) packaging of lig- vantage that only a plasmid construction and subsequent
ated molecules using lambda in vitro packaging mix, (v) M. tuberculosis strain electroporation is necessary. The dis-
transduction of lambda-sensitive E. coli strain with selec- advantage of using suicide plasmids or chemically modified
tion for the antibiotic resistance gene on the new cosmid, plasmids is that electroporation can be very inefficient and
(vi) analysis of E. coli propagated shuttle phasmid by re- electroporation efficiencies vary widely between M. tuber-
striction enzymes to verify exchange of previous cosmid for culosis strains. Specialized phage transduction has the disad-
the new cosmid, and (vii) transfection of M. smegmatis vantage of more preparation steps, but the advantage of
and preparation of TM4-packaged shuttle phasmids. higher efficiencies of substrate delivery than electropora-
tion as well as less variability in efficiency for many differ-
ent strains of M. tuberculosis.
34.5. GENE DELIVERY USING PHAGES Generation of knockout mutants with specialized trans-
duction entails five steps (see Fig. 3): (i) construction of an
34.5.1. Reporter Gene Delivery allelic exchange cosmid; (ii) phasmid transduction; (iii)
Luciferase reporter phages were first constructed with the phage lysate preparation; (iv) phage transduction; and (v)
goal of rapidly assessing drug susceptibilities in clinical iso- generation of double-crossover alleles (Fig. 3). In brief, the
lates of M. tuberculosis (23). Drug resistance could be dis- plasmid for constructing allelic exchange substrates (AES)
tinguished from drug susceptibility by treating M. tuberculo- has multiple cloning sites (MCS)upstream and downstream
sis cells with or without different concentrations of drugs and of an antibiotic resistance marker (hygromycin), as well as
infecting with a luciferase reporter phage. Drug-sensitive a counter-selectable marker (sacB); also on the AES plas-
cells would fail to generate any luciferase activity compared mid is oriE, an E. coli plasmid origin of replication, lambda-
with the non-drug-treated control. Drugresistant cells cos site for lambda packaging, and a unique PacI restriction
would glow like untreated cells. TM4 phages are particu- site for eventual ligation to a modified temperature-
larly attractive since hundreds of M. tuberculosis isolates sensitive mycobacteriophage such as TM4. DNA segments
have been infected and no strain has yet been found to be (typically SO0 bp) derived from each side of the gene to be
resistant to TM4 phage. Luciferase reporter phages have replaced are generated by PCR amplification and cloned
also been constructed from both phages D29 and L5 (36, into convenient restriction sites in the AES plasmid on ei-
45) and several studies have used this methodology (2,41). ther side of the resistance gene (3). Detailed protocols for
these manipulations can be found at www.tbvaccines.org.
34.5.2. Conditionally Replicating
Mycobacteriophages and Transposon Delivery
Historically, conditionally replicating phages, usually amber 34.6. OTHER PHAGE-DERIVED GENETIC
mutants, have been used to efficiently deliver transposons TOOLS
(29). To achieve such a goal with mycobacteriophages, a
battery of mutants of TM4 or D29 shuttle phasmids were 34.6.1. Construction of Integration-Proficient
isolated that permit replication of the phage in M. smegma- Plasmid Vectors
tis at 30C, but not at 38 or 39C (4, 5). It is noteworthy Since many phages carry an apparatus for integrating their
that phasmid phAE87, derived from the TM4 phage phlOl DNA into the host chromosome, it is normally not hard to
(S), has a reversion frequency of lo- to lop9in contrast to find this feature for use in the construction of integration-
most thermosensitive mutants, which have a reversion fre- proficient plasmid vectors. Conceptually, this is straightfor-
quency of lop4 to this low reversion frequency is ward and requires the incorporation of a segment of phage
likely the result of two independent thermosensitive muta- DNA containing the integrase gene and the attP site into a
tions. This low level of reversion makes this phage the ve- plasmid vector; this vector should then transform the host
hicle of choice for delivery of transposons to M. tuberculosis by integrase-mediated integration into the host chromo-
(4), M. smegmatis (4), and Mycobacterium aviurn (16). some. The advantages of such vectors are that they should
Initially this phage was used to deliver IS1096-derived be stably maintained and they provide single-copy recombi-
transposons (4, 6,35). Such transposon libraries have been nants.
useful in identifying many previously unknown virulence Although conceptually easy, there are several consider-
genes including pcaA (14), PDIM biosynthetic genes (8), ations when constructing integration-proficient vectors.
and PE PGRS (9). More recently, by incorporating the First, it is important to carefully define the segment of
Himar mariner transposon, a transposon that displays little phage DNA to be used. Unless experimental data are avail-
sequence specificity (30, 43), newer transposon delivery able, the integrase gene can usually be identified bioinfor-
phages have been constructed. matically and usually belongs to one of two groups, the ty-
rosine integrases (such as phage lambda) or the serine
34.5.3. Allelic Exchange by Specialized integrases (such as +C31 or Bxbl). While these can be
Transduction readily identified, the phage attachment sites (attP) are
The ability to generate null mutants in M. tuberculosis is often more difficult, although in almost every known case
critical for rigorous genetic analysis. In any knockout the attP site is located adjacent to the integrase gene, either
scheme, an allelic exchange substrate containing the modi- to the 5 or 3 side. Since members of the tyrosine integrase
fied gene of interest needs to be delivered to the cells, fol- family predominantly utilize chromosomal attachment sites
lowed by selection for recombinants that have the knock- (attB) that overlap tRNA genes, which are reconstituted
out allele. Several methods have been used to successfully following integration, then the corresponding attP sites in-
generate knockout strains of M. tuberculosis (3), including clude portion of tRNA genes corresponding to their 3
transformation with linear DNA substrates, suicide plas- ends. Thus, if the genome sequence of the host is available
-
34. Genetic Manipulations Using Phages 833

Transfection

M. smegmatis at 30C

Transduction

High Titer Phage


M. smegmatis
M. bovis BCG
M. tuberculosis
a t 37C
FIGURE 3 Specialized transduction. Regions flanking the upstream and downstream regions of
Your favorite gene (yfg) are cloned into a hygromycin-containing cassette on a plasmid contain-
ing both lambda cos sites and mycobacteriophage cos sites. M. srnegmatis is transfected at 30C to
allow for production of a high-titer phage lysate containing the AyfgX::H$ allele. The high-titer
phage lysate is then used to transduce M. tuberculosis at 37C (nonpermissive temperature for phage
lysis) to deliver AyfgX::Hy$ substrate to the cell for subsequent allelic exchange.

(or a near relative) then a BLAST search with sequences nals (28,32,39). Since it is possible that integrase can pro-
flanking the integrase genes can reveal 20 to 40-bp regions mote RDF-independent excision at low frequency include
of near-sequence identity. If this corresponds to an anno- and that this frequency will be elevated with high integrase
tated host tRNA gene, then this almost certainly indicates expression (48), it may be desirable to avoid the use of
the position of attP. Unfortunately, this approach normally strong heterologous promoter systems. It is also important
does not work for serine integrases because they do not use to avoid the inclusion of plasmid replication origins that
tRNA genes as integration sites and the extent of sequence function in the host, since these are likely to be toxic to the
identity with the host is small, often just 3 to 8 bp (46). host when integrated into the chromosome.
The second important consideration involves the loca- An alternative approach to avoiding undesirable excision
tion of the excise gene. Most phage genomes that integrate is to express the integrase transiently from a nonreplicating
usually also encode a recombination directionality factor plasmid that is coelectroporated with an attP-containing
(RDF) that facilitates an integrase-mediated excision reac- plasmid (38). This generates far fewer transformants, but
tion; sometimes this is encoded nearby the integrase gene, those recovered are no longer expressing integrase and the
but several examples exist where it is not (34). Un- integrated sequences are maintained without any significant
fortunately, these RDFs are highly varied in their sequences loss. If serine integrase systems are used, then the extent of
and they can be difficult to identify if relying solely on sequence identity shared by the two junction sites attL and
bioinformatic approaches. When defining a segment of attR flanking the integrated sequence is short (3 to 8 bp),
phage DNA for constructing integration-proficient vectors, and therefore undesirable excision by the host recombina-
it is helpful to try to avoid the RDF gene, since it will con- tion system is infrequent.
tribute to either low transformation frequencies or instabil-
ity of the integrated DNAs (32). If the RDF gene can be 34.6.2. Recombineering: Phage-Mediated
identified, it is usually simple to exclude it; if not, it is best Recombination for Allelic Exchange
to use as small a segment as possible that includes both the Two phage-derived recombination systems-encoded by
integrase gene and the attP site. the lambda Red system and the Rac prophage-have been
A third consideration is the expression of the integrase developed for high-frequency allelic exchange and mutage-
gene. While it is possible that a promoter may be needed to nesis in E. coli. This process is referred to as recombineering
promote integrase expression, this has not been necessary (7). While these systems work well in E. coli-and can be
for any of the mycobacteriophage-derived vectors that have used to construct point mutations in the absence of selec-
been constructed; presumably the gene is expressed either tion-they may not function as well in other bacteria, es-
from vector sequences or from closely associated phage sig- pecially gram-positive bacteria. While the reasons for this
834 MOLECULAR GENETICS

are not clear, it could be due to differences in G+C% con- duce bacterial contamination; it is also common to include
tent, the activities of small DNA-binding proteins, or alter- cycloheximide (CHX) at 10 (g/ml to eliminate fungal con-
native modes of DNA replication. However, a reasonable tamination. Since it has a strong tendency (like most my-
solution to this problem is to develop host-specific recom- cobacteria) to clump when grown in liquid media, addition
bineering systems using analogous (or homologous) systems of 0.05% Tween@ 80 is typically added to help form a dis-
encoded by phages that infect the host of interest. This ap- persed culture. However, this causes a serious problem for
proach has been successful for mycobacterial recombineer- phage infections, since mycobacteriophages are frequently
ing and should be applicable in general to other bacteria sensitive to these detergents. Since mycobacteria hydrolyze
(50a). Tween as they grow, this problem can usually be circum-
Homologues of the RecET proteins encoded by the Rac vented either by using mid- or late-logarithmic cultures
prophage are rare among some groups of phages. For exam- or by subculturing a stationary-phase culture into fresh
ple, only one (Che9c) of 30 completely sequenced my- medium lacking detergent. M. smegmatis cultures are grown
cobacteriophage genomes encodes homologues of both the in standard test tubes placed at an angle on a shaker, but for
RecE exonuclease and the RecT DNA-paring enzyme. bigger volumes baffled flasks with agitation are appropriate.
However, these can be adapted for recombineering by ex- Since many phages require calcium for infection, CaCll at
pressing both proteins from an inducible acetamidase pro- a final concentration of 1 mM is routinely included in all
moter and then preparing electrocompetent cells. These media. We typically use 0.35% top agar to screen for new
can then be transformed using a linear DNA substrate that phages (since large phages do not produce visible plaques
contains approximately 500 bp of chromosomal DNA on thicker agar), but 0.7% agar can be used in subsequent
flanking a drug resistance gene. If these 500-bp segments steps.
correspond to sequences flanking gene X, then selection for
the drug marker generates transformants in which gene X is 34.7.2. Preparation of E. coh Competent Cells
now replaced by that marker. In the mycobacterial system, To prepare E. coli HBlOl for transduction, grow the HBlOl
approximately 100 transformants can be recovered using strain overnight in Luria Broth (LB) supplemented with 10
only 100 ng of DNA in both fast-growing (e.g., M. smegma- mM MgS04 and 0.2% maltose. Inoculate 25 ml of fresh
t i s ) and slow-growing (e.g., M. tuberculosis) mycobacterial medium with 0.5 ml of overnight culture. Shake culture at
strains; approximately 90% or more of these are the prod- 200 rpm at 37C until OD600 reaches 0.8 to 1.0. Transfer
ucts of allelic exchange. Such a recombineering system cells to a 50-ml conical tube and pellet cells in a tabletop
could likely be further adapted to function with shorter seg- centrifuge at 3,000 rpm for 10 min at 4C. Decant super-
ments of DNA homology or with single-stranded DNA sub- natant and resuspend the cell pellet in 12.5 ml of 10 mM
strates that require only the pairing enzyme. MgS04. Store cells at 4C until ready for use.

34.6.3. Generalized Transduction 34.7.3. Transformation of M. smegmafis


Generalized transduction is an essential component of any 1. Grow fresh 50 ml of culture of M. smegmatis mc155
well-developed system for bacterial genetics, since it pro- in either Middlebrook 7H9 broth or LB with Tween
vides a simple method for constructing isogenic strains. (0.05%) to mid-log phase (OD6000.5to 1.0).
Generalized transducing phages have thus been identified 2. Incubate cells on ice for 10 min but no longer than
and described for a wide variety of bacterial hosts, and they 2 h.
can be used similarly. Methods have long been established NOTE: For the following steps, keep cells as close to 0C
for generalized transduction and usually involve the prepa- (ice bath) as possible. The electroporation cuvettes, 50-ml
ration of a phage lysate on the donor bacterial strain, fol- conical tubes, and 10% glycerol should be prechilled to
lowed by infection of a recipient strain with this lysate and 4C.
selection for the desired genetic marker. Transduction fre- 3. Transfer chilled cells to a 50-ml prechilled conical
quencies vary greatly, but even if they are fairly low and pro- tube. Spin down cells in a tabletop centrifuge at 2,000 X g
duce only a small number of colonies, this is usually useful for 10 min. Decant supernatant from cell pellet.
provided that the marker does not have a high reversion 4. Wash cells two times with 40 ml of ice-cold 10%
frequency. A primary concern is that there may be signifi- glycerol.
cant levels of killing by the phage lysate, which may need NOTE: For optimal results, prepare 10% glycerol fresh
to be minimized following initial adsorption by addition of weekly with distilled water; sterilize by filtration through
antiphage antisera or chelating agents that lower the con- 0.2-pm membrane pore; do not autoclave.
centration of available divalent cations such as calcium. In 5. Resuspend the washed cell pellet in 5 ml of ice-cold
the mycobacteria, several transducing phages have been de- 10% glycerol ( l / l O culture volume) and store on ice (will
scribed (e.g., I3 and Bxzl), but none for M. tuberculosis (33, use 0.4 ml per sample).
40). 6. Aliquot transforming DNA (1 to 10 pl) in a
prechilled GenePulser electroporation cuvette with a 0.2-
cm electrode gap (Bio-Rad, catalog no. 165-2086) and in-
34.7. RECIPES cubate on ice. To each aliquot of DNA, add 400 p1 of pre-
pared cell suspension and mix by gently pipetting up and
34.7.1. Culture of M. smegmafis down (use prechilled Pipetman tips for cell transfer).
M. smegmatis mc155 is a suitable host bacterium for phage 7. Pulse one time with GenePulser apparatus (Bio-Rad
propagation. It has a doubling time of approximately 3 h at catalog no. 1652076) set at 2.5 V: resistance, 1,000 Ohms;
37C and grows to a colony on solid medium in 3 to 4 days; capacitance, 25 pFD. The time constant reading should be
bacterial lawns typically grow in about 24 h, and plaques about 19 to 21 for control cells with no DNA added.
can be seen after about 16 h. It grows in defined media as 8. Immediately dilute transformed cell mixture with 2
described below and is resistant to carbenicillin (CB), ml of medium. Transfer cell suspension to a 15-ml snap-cap
which is typically included in media (50pg/ml) to help re- Falcon tube (Falcon catalog no. 2059).
34. Genetic Manipulations Using Phages 835

9. Incubate at 37C (with or without shaking) for 2 h NaC1 ................................ 5g


to allow cell recovery and expression of antibiotic resist- dH20 .............................. .to 1 liter
ance.
10. Spin down cells in tabletop centrifuge at 2,000 X g Autoclave to sterilize.
for 10 min. Aspirate supernatant from cell pellet and resus- NOTE: For LB Agar, add 15 g of agar. For L-Kan drug
pend cell pellet in 0.5 ml of medium. plates, add 0.8 ml of kanamycin per liter after temperature
11. Plate desired volume of cell resuspension on plates is less than 60C. For L-CB drug plates, add 1 ml of car-
containing the antibiotic of choice (kanamycin at 10 to 20 benicillin after temperature is less than 60C.
pg/ml; hygromycin B at 50 pglml).
Middlebrook 7H9 liquid Medium
34.7.4. Media and Reagents (Autoclave and add antibiotics, ADC, and calcium prior to
use.)
ADC (NO HEAT, filter sterilize)
7H9 broth base ......................... 4.7 g
Dextrose ............................. 60 g 40% glycerol ........................... 5 ml
NaCl ............................... 25.5g ddHzO ............................... 900ml
Albumin. ............................ 150 g
....................... .2,850 ml
Middlebrook 7H10 Solid Medium
Be sure to add water before adding albumin. Mix on hot
plate on the lowest setting (or not at all) to dissolve. Filter 7H10 agar ............................ 19 g
sterilize using a 0.22-pm-pore-size filter. 40% glycerol .......................... 12.5 ml
ddHzO ............................... 890ml
0.1 M CaCI2 (Stock Solution)
Calcium is added to mycobacterial growth medium to en- Autoclave, cool to 55"C, and add 100 ml of ADC and
sure adequate calcium is available for necessary cellular 10 ml of 0.1 mM CaC12 , CB, CHX, and then pour.
metabolic processes. Note that, for the most part, calcium is
added to obtain a 0.1 mM concentration in the final solu- MBTA (Middlebrook Top Agar)
tions of each medium or reagent. Therefore, add lml of the
1 mM CaClz stock solution to phage buffer, but 2 ml of 7H9 ............................... 4.7 g
CaClz to 7H9, because it will be used to dilute top agar by Agar ............................... 7s
an additional 50%. H 2 0 ............................... to900ml

Calcium chloride (a Prepared at 0.7% concentration, melted, diluted to


dHzO .. 0.35% with 7H9 (plus 2 ml of 0.1 M CaC&).

Carbenicillin (CB)(50 pg/ml) Phage Adsorption Medium

... 2.5 g 10% albumin dextrose saline . . . . . . . . . . 2% glycerol


. . . . . . t o 50ml Phage Buffer
Filter sterilize. Store in the refrigerator. 1 M Tris, pH 7.5 ......................... 10 ml
Cycloheximide (CXH) (10 pg/ml) 1MMgS0 ............................. 10ml

....................... Autoclave or filter sterilize. Add 0.1 mM CaClz prior


Cycloheximide 1g to use.
dHzO to 100 ml
20% Tween@
Filter sterilize. Store in the refrigerator.
Hygromycin (100 pg/ml) TweenB80 ............................. 20ml
ddH20 ................................ 80ml
Hygromycin .......................... 1g Warm to 50C to dissolve and filter sterilize.
dHzO . . . . . . . . . .to 10 ml
We wish to thank all former and current members of the Hatfull and
Filter sterilize. Store at 4C. Jacobs laboratories for developing the methods and approaches described
in this chapter.
Kanamycin (50 mg/ml)

Kanamycin ........................... 500 mg 34.8. REFERENCES


dH2O . . . . . . . . . . ........ .to 10 ml 1. Ackermann, H. W. 2001. Frequency of morphological
phage descriptions in the year 2000. Brief review. Arch.
Filter sterilize. Store at 4C. Virol. 146~843-857.
2. Banaiee, N., M. Bobadilla-del-Valle, P. F. Riska, S.
Luria Broth Bardarov, Jr., P. M. Small, A. Ponce-de-Leon, W. R.
Jacobs, Jr., G . F. Hatfull, and J. Sifuentes-Osornio. 2003.
Tiyptone ............................. 10 g Rapid identification and susceptibility testing of Myco-
Yeast extract .......................... 5s bacterium tuberculosis from MGIT cultures with luciferase
836 MOLECULAR GENETICS

reporter mycobacteriophages.J. Med. Microbiol. 52:557- 18. Hendrix, R. W. 1999. The long evolutionary reach of
561. viruses. Cum. Biol. 9:914-917.
3. Bardarov, S., S. Bardarov, Jr., M. S. Pavelka, Jr., V. 19. Hendrix, R. W. 2003. Bacteriophage genomics. Cum.
Sambandamurthy, M. Larsen, J. Tufariello, J. Chan, G. Opin. Microbiol. 6506-51 1.
Hatfull, and W. R. Jacobs, Jr. 2002. Specialized transduc- 20. Hendrix, R. W., M. C. Smith, R. N. Burns, M. E. Ford,
tion: an efficient method for generating marked and un- and G. F. Hatfull. 1999. Evolutionary relationships among
marked targeted gene disruptions in Mycobucterium tuber- diverse bacteriophages and prophages: all the worlds a
culosis, M. bovis BCG and M. smegmatis. Microbiology 148: phage. Proc. Natl. Acud. Sci. USA 96:2192-2197.
3007-3017. 21. Jacobs, W. R., M. A. Docherty, R. D. Curtiss, and J. E.
4. Bardarov, S., J. Kriakov, C. Carriere, S. Yu, C. Clark-Curtiss. 1986. Expression of Mycobacterium leprue
Vaamonde, R. A. McAdam, B. R. Bloom, G. F. Hatfull, genes from a Streptococcus mutuns promoter in Escherichia
and W. R. Jacobs, Jr. 1997. Conditionally replicating my- coli K-12. Proc. Natl. Acud. Sci. USA 83:1926-1930.
cobacteriophages: a system for transposon delivery to 22. Jacobs, W. R., Jr. 2000. Mycobucterium tuberculosis: a once
Mycobacterium tuberculosis. Proc. Natl. Acud. Sci. USA 94: genetically intractable organism, p. 1-16. In G. F. Hatfull
10961-10966. and W. R. Jacobs Jr. (ed.), Molecular Genetics of the Myco-
5. Carriere, C., P. F. Riska, 0. Zimhony, J. Kriakov, S. bacteria. ASM Press, Washington, DC.
Bardarov, J. Burns, J. Chan, and W. R. Jacobs, Jr. 1997. 23. Jacobs, W. R., Jr., R. G. Barletta, R. Udani, J. Chan, G.
Conditionally replicating luciferase reporter phages: im- Kalkut, G. Sosne, T. Kieser, G. J. Sarkis, G. F. Hatfull,
proved sensitivity for rapid detection and assessment of and B. R. Bloom. 1993. Rapid assessment of drug suscep-
drug susceptibility of Mycobacterium tuberculosis. J. Clin. tibilities of Mycobacterium tuberculosis by means of luci-
Microbiol. 35:3232-3239. ferase reporter phages. Science 260:819-822.
6. Cirillo, J. D., R. G. Barletta, B. R. Bloom, and W. R. 24. Jacobs, W. R., Jr., S. B. Snapper, M. Tuckman, and
Jacobs, Jr. 1991. A novel transposon trap for mycobacte- B. R. Bloom. 1989. Mycobacteriophage vector systems.
ria: isolation and characterization of IS1096. J. Bacteriol. Rev. Infect. Dis. 11 (Suppl 2):S404-S410.
173 :7 7 7 2-7 7 80. 25. Jacobs, W. R., Jr., M. Tuckman, and B. R. Bloom. 1987.
7. Court, D. L., J. A. Sawitzke, andL. C. Thomason. 2002. Introduction of foreign DNA into mycobacteria using a
Genetic engineering using homologous recombination. shuttle phasmid. Nature 327:532-535.
Annu. Rew. Genet. 36:361-388. 26. Jones, W. D., Jr. 1975. Phage typing report of 125 strains
8. Cox, J. S., B. Chen, M. McNeil, and W. R. Jacobs, Jr. of Mycobucterium tuberculosis.Ann. Sclawo 17599-604.
1999. Complex lipid determines tissue-specificreplication 27. Jones, W. D., Jr., and J. Greenberg. 1978. Modification
of Mycobacterium tuberculosis in mice. Nature 402:79-83. of methods used in bacteriophage typing of Mycobacterium
9. Delogu, G., C. Pusceddu, A. Bua, G. Fadda, M. J. tuberculosis isolates. J. Clin. Microbiol. 7467-469.
Brennan, and S. Zanetti. 2004. Rvl818c-encoded 28. Kim, A., P. Ghosh, M. A. Aaron, L. A. Bibb, S. Jain,
PE-PGRS protein of Mycobucterium tuberculosis is surface and G. F. Hatfull. 2003. Mycobacteriophage Bxbl inte-
exposed and influences bacterial cell structure. Mol. Micro- grates into the Mycobacterium smegmatis groELl gene. Mol.
bio-1. 52:725-733. Microbiol. 50:463473.
10. Desplats, C., and H. M. Krisch. 2003. The diversity and 29. Kleckner, N., J. Bender, and S. Gottesman. 1991. Uses of
evolution of the T4-type bacteriophages. Res. Microbiol. transposons with emphasis on TnlO. Methods Enzymol.
- - . -.
154:259-267.
. - .. 204:139-180.
11. Dye, C., S. Scheele, P. Dolin, V. Pathania, and M. C. 30. Lamichhane, G., M. Zignol, N. J. Blades, D. E. Geiman,
Raviglione. 1999. Consensus statement. Global burden of A. Dougherty, J. Grosset, K. W. Broman, and W. R.
tuberculosis: estimated incidence, prevalence, and mortal- Bishai. 2003. A postgenomic method for predicting essen-
ity by country. WHO Global Surveillance and Monitoring tial genes at subsaturation levels of mutagenesis: applica-
Project. JAMA 282:677-686. tion to Mycobacterium tuberculosis. Proc. Natl. Acud. Sci.
12. Edwards, R. A., and F. Rohwer. 2005. Viral metage- USA 100:7213-7218.
nomics. Nut. Rew. Microbiol. 3:504-510. 3 1. Lawrence, J. G., G. F. Hatfull, and R. W. Hendrix. 2002.
13. Frieden, T. R., and C. R. Driver. 2003. Tuberculosis con- Imbroglios of viral taxonomy: genetic exchange and fail-
trol: past 10 years and future progress. Tuberculosis (Edinb) ings of phenetic approaches. ]. Bacteriol. 1844891-4905.
83:82-85. 32. Lee, M. H., L. Pascopella, W. R. Jacobs, Jr., and G. F.
14. Glickman, M. S., J. S. Cox, and W. R. Jacobs, Jr. 2000. Hatfull. 1991. Site-specific integration of mycobacterio-
A novel mycolic acid cyclopropane synthetase is required phage L5: integration-proficient vectors for Mycobucterium
for cording, persistence, and virulence of Mycobucterium smegmatis, Mycobucterium tuberculosis, and Bacille Calmette-
tuberculosis. Mol. Cell. 5:717-727. Guerin. Proc. Natl. Acud. Sci. USA 88:3111-3115.
15. Hambly, E., and C. A. Suttle. 2005. The viriosphere, di- 33. Lee, S., J. Kriakov, C. Vilcheze, Z. Dai, G. F. Hatfull,
versity, and genetic exchange within phage communities. and W. R. Jacobs, Jr. 2004. Bxzl, a new generalized trans-
Cum. Opin. Microbiol. 8:444-450. ducing phage for mycobacteria. FEMS Microbiol. Lett.
16. Harris, N. B., Z. Feng, X. Liu, S. L. Cirillo, J. D. Cirillo, 24 1:27 1-276.
and R. G. Barletta. 1999. Development of a transposon 34. Lewis, J. A., and G. F. Hatfull. 2001. Control of direc-
mutagenesis system for Mycobacterium awium subsp. paratu- tionality in integrase-mediated recombination: examina-
berculosis. FEMS Microbiol. Lett. 175:21-26. tion of recombination directionality factors (RDFs) in-
17. Hatfull, G. F., M. L. Pedulla, D. Jacobs-Sera, P. M. cluding Xis and Cox proteins. Nucleic Acids Res. 29:2205-
Cichon, A. Foley, M. E. Ford, R. M. Gonda, J. M. 2216.
Houtz, A. J. Hryckowian, V. A. Kelchner, S. Namburi, 35. McAdam, R. A., S. Quan, D. A. Smith, S. Bardarov,
K. V. Pajcini, M. G. Popovich, D. T. Schleicher, B. Z. J. C. Betts, F. C. Cook, E. U. Hooker, A. P. Lewis, P.
Simanek, A. L. Smith, G. M. Zdanowicz, V. Kumar, Woollard, M. J. Everett, P. T. Lukey, G. J. Bancroft,
C. L. Peebles, W. R. Jacobs, J. G. Lawrence, and R. W. W. R. Jacobs, Jr., and K. Duncan. 2002. Characterization
Hendrix. 2006. Exploring the mycobacteriophage of a Mycobucterium tuberculosis H37Rv transposon library
metaproteome: phage genomics as an educational plat- reveals insertions in 351 ORFs and mutants with altered
form. PLoS Genetics 2,:e92. virulence. Microbiology 148:2975-2986.
34. Genetic Manipulations Using Phages 837

36. Pearson, R. E., S. Jurgensen, G. J. Sarkis, G. F. Hatfull, 44. Ruska, H. 1940. Die Sichtbarmachung der Bakterio-
and W. R. Jacobs, Jr. 1996. Construction of D29 shuttle phagenLyse im Ubermikroskop. Naturevissenschaften 28:
phasmids and luciferase reporter phages for detection of 4546.
mycobacteria. Gene 183:129-136. 45. Sarkis, G. J., W. R. Jacobs, Jr., and G. F. Hatfull. 1995.
37. Pedulla, M. L., M. E. Ford, J. M. Houtz, T. Karthikeyan, L5 luciferase reporter mycobacteriophages: a sensitive tool
C. Wadsworth, J. A. Lewis, D. Jacobs-Sera, J. Falbo, J. for the detection and assay of live mycobacteria. Mol.
Gross, N. R. Pannunzio, W. Brucker, V. Kumar, J. Microbiol. 15:1055-1067.
Kandasamy, L. Keenan, S. Bardarov, J. Kriakov, J. G. 46. Smith, M. C., and H. M. Thorpe. 2002. Diversity in the
Lawrence, W. R. Jacobs, R. W. Hendrix, and G. F. serine recombinases. Mol. Microbiol. 44:299-307.
Hatfull. 2003. Origins of highly mosaic mycobacterio- 47. Snapper, S. B., L. Lugosi, A. Jekkel, R. E. Melton, T.
phage genomes. Cell 113:171-182. Kieser, B. R. Bloom, and W. R. Jacobs, Jr. 1988.
38. Peiia, C. E., M. H. Lee, M. L. Pedulla, and G. F. Hatfull. Lysogeny and transformation in mycobacteria: stable ex-
1997. Characterization of the mycobacteriophage L5 at- pression of foreign genes. Proc. Natl. Acad. Sci. USA 85:
tachment site, attP. J. Mol. Biol. 266:76-92. 6987-6991.
39. Pena, C. E., J. Stoner, and G. F. Hatfull. 1998. Myco- 48. Springer, B., P. Sander, L. Sedlacek, K. Ellrott, and E. C.
bacteriophage D29 integrase-mediated recombination: Bottger. 2001. Instability and site-specific excision of inte-
specificity of mycobacteriophage integration. Gene 225: gration-proficient mycobacteriophage L5 plasmids: devel-
143-15 1. opment of stably maintained integrative vectors. Int.
40. Raj, C. V., and T. Ramakrishnan. 1970. Transduction in 1. Med. Microbiol. 290:669-675.
Mycobacterium smegmatis. Nature 228:280-281. 49. Summers, W. C. 1999. Felix dHerelle and the Origins of
41. Riska, P. F., and W. R. Jacobs, Jr. 1998. The use of Molecular Biology. Yale University Press, New Haven, CT.
luciferase-reporterphage for antibiotic-susceptibility test- 50. Tokunaga, T., and R. M. Nakamura. 1967. Infection of
ing of mycobacteria. Methods Mol. Biol. 101:431455. Mycobacterium tuberculosis with deoxyribonucleic acid ex-
42. Rodicio, M. R., C. J. Bruton, and K. F. Chater. 1985. tracted from mycobacteriophageB1. J. Virol. 1:448449.
New derivatives of the Streptomyces temperate phage phi 50a.van Kessel, J. C., and G. E Hatfull. 2007. Recombineering
C31 useful for the cloning and functional analysis of in Mycobacterium tuberculosis. Nut. Meth. 4:147-152.
Streptomyces DNA. Gene 34:283-292. 51. Watson, J. D., and F. H. Crick. 1953. Molecular structure
43. Rubin, E. J., B. J. Akerley, V. N. Novik, D. J. Lampe, of nucleic acids; a structure for deoxyribose nucleic acid.
R. N. Husson, and J. J. Mekalanos. 1999. In vivo trans- Nature 171:737-738.
position of mariner-based elements in enteric bacteria 52. Wommack, K. E., and R. R. Colwell. 2000. Virio-
and mycobacteria. Proc. Natl. A c d . Sci. USA 96:1645- plankton: viruses in aquatic ecosystems. Microbiol. Mol.
1650. Biol. Rew. 64:69-114.
COMMUNITY AND
GENOMIC ANALYSIS

Introduction to Community and 39. Single Cell Identification by


Genomic Analysis 841 Fluorescence In Situ Hybridization 886
THOMAS M. SCHMIDT, Editor BERNHARD M. FUCHS, JAKOB PERNTHALER,
AND RUDOLF AMANN
35. Characterization of Bacterial Genome
Sequences by Similarity Searching 842 40. Measurement of rRNA Abundance by
WILLIAM R. PEARSON Hybridization with
Oligodeoxynucleotide Probes 897
36. Reconstructing and Interpreting DANIEL H. BUCKLEY AND
Evolutionary Relationships 856 THOMAS M. SCHMIDT
CHRISTOPHE J. DOUADY AND
CAMILLA L. NESB0 41. Analysis of Microbial Communities with
Denaturing Gradient Gel Electrophoresis
37. Microbial Nucleotide Fingerprints in and Terminal Restriction Fragment
Nature 869 Length Polymorphism 909
DAVID M. KARL CINDY H. NAKATSU AND TERENCE L. MARSH

38. Construction of BAC and Fosmid


Libraries from Naturally Occurring
Microbial Populations 879
EDWARD F. DELONG
Introduction to Community and Genomic Analysis
THOMAS M. SCHMIDT

Remarkable advances in DNA-sequencing technology and specialized sequencing facilities, probing the genomic
the computational capacity to analyze sequences has had a content of microbial communities through large insert li-
tremendous impact on our understanding of microbes and braries requires the construction of the clone libraries.
microbial communities. No longer is microbiology con- DeLong (chapter 38) provides a discussion of the cloning
strained by the absolute requirement for cultivation of mi- vectors appropriate for this task, along with the tricks of
crobes since genome-based approaches have extended the the trade resulting from his years of experience as a pio-
reach of microbiologists to include microbes that have not neer of this approach.
yet been cultured. While physiological and genetic studies DNA sequences are the foundation for the design of nu-
of cultivated microbes and biochemical analyses of cloned cleic acid probes that serve as pivotal tools in a series of
genes continue to provide the ground truth necessary to techniques that provide a window into the structure and
interpret DNA sequences, the capacity to determine com- activities of microbial communities. Microbial ecologists
plete genome sequences and meta-genomes from microbial have the choice of probing intact cells with fluorescent or
communities has helped usher in a new era of microbiology radioactive probes, or sometimes in combination to provide
that has been dubbed The Third Golden Age of microbi- phylogenetic and physiological information simultane-
ology (1). In this era, the subdisciplines of microbiology are ously. This collection of techniques is presented in chapter
being integrated, due in part to the common use of nucleic 39 by Fuchs, Pernthaler, and Amann, who have developed
acid sequences to study the genetics, evolution, physiology, and applied these approaches to numerous microbial com-
and ecology of diverse microbes. munities. Community analysis can also be accomplished
The advances in genomics underlying this new era of through the probing of purified RNA to target metaboli-
microbiology have warranted the creation of a new section cally active populations. Buckley and Schmidt present this
in this manual that is focused on nucleic acid-based meth- method (chapter 40) as one strategy to avoid the potential
ods for community and genome analysis. This section in- bias introduced by the amplification of nucleic acids.
cludes descriptions of the methods to analyze gene se- However, phylogenetically informative genes or functional
quences, beginning with effective strategies for searching genes can be successfully targeted for analysis through PCR
genome databases for homologs (chapter 35). Although it amplification and then separation of the amplicons on de-
is common practice to search databases using nucleic acid naturing gels or analysis of the terminal fragments of the
sequences, Pearson describes more effective approaches for amplicons (chapter 41). Nakatsu and Marsh provide a use-
finding homologs that include a concise explanation of the ful explanation of these approaches and discuss their po-
advantages of searching with protein sequences. Once ho- tential pitfalls.
mologs have been identified, additional information can be Each of the methods presented in this section offers op-
extracted from the sequence data through phylogenetic portunities and limitations in the analysis of genomes and
analyses. Douady and Nesba provide a thoughtful overview community structure, and the authors provide the thought-
of molecular phylogenetics and specific recommendations ful commentary necessary for readers to make an appropri-
for constructing and interpreting phylogenetic trees in ate selection of the most reasonable approaches to their re-
chapter 36. search questions. Since methods that utilize nucleic acid
While the capacity to study complex microbial com- sequences are rapidly expanding, the collection of chapters
munities is expanding rapidly, basic questions still fre- in this section provides a strong foundation of methods for
quently top the list when studies of a new microbial com- the microbiologists planning to probe the complexity of
munity are initiated. Those questions include How much microbial communities and genomes. The references pro-
biomass is present? and What is the metabolic activity vided in each chapter provide the reader with useful entry
of the total microbial community? In chapter 37, Karl points into this burgeoning literature, including many com-
provides a review of the methods for answering these plementary laboratory manuals for more specialized ap-
questions, including detailed methodology for using nu- proaches to community and genomic analyses.
cleotide fingerprints to assess biomass and metabolic ac-
tivity. A more detailed look at the structure of microbial
communities is made possible by analysis of the genes and REFERENCE
genomes present in a community. While genome se- 1. Schaechter, M. 2003. Integrative microbiology-the
quencing and sequence assembly is conducted primarily at Third Golden Age. J. Biosci. 28:149-154.

841
35
Characterization of Bacterial Genome Sequences
by Similarity Searching
WILLIAM R. PEARSON

35.1. INTRODUCTION ......................................... 842


.2. SIMILARITY, HOMOLOGY, AND STATISTICS .................. 842
35.2.1. Homology and Statistical Significance ...................... 843
.2. Homology and Nonhomology ............................ 844
.3. Expectation and Percent Identity .......................... 844
.4. Improving Sensitivity with Smaller Databases-Part I ........... 845
.5. Homology and Function-Orthology ....................... 845
....... 845
.6. Statistical Thresholds and Large-Scale Similarity Searching
35.3. SIMILARITY SEARCHING PROGRAMS ....................... 846
.4. PROTEIN VERSUS DNA SEQUENCE COMPARISON ............. 847
.5. IMPROVING SEARCH SENSITIVITY ......................... 848
35.5.1. Improving Pairwise Similarity Scores ....................... 848
.2. Adjusting Scoring Matrices for Evolutionary Distance .......... 849
.3. Improving Sensitivity with Smaller Databases-Part I1 .......... 85 1
35.6. IDENTIFYING ANONYMOUS SEQUENCES FROM BACTERIAL
POPULATIONS .......................................... 85 1
.7. PATHOLOGICAL CASES ................................... 853
35.7.1. Low-Complexity Regions-SEG .......................... 853
.2. Confirming Statistical Significance with Shuffled Comparisons .... 853
35.8. SUMMARY .............................................. 854
.9. REFERENCES ............................................ 855

35.1. INTRODUCTION basis for inferring homology; from this statistical perspec-
With more than 100 fully sequenced microbial genome se- tive, several strategies emerge for improving the effective-
quences publicly available from many of the deep divisions ness of similarity searches.
of bacteria and archaea, protein and DNA sequence simi- Indeed, the four most common errors made when search-
larity searching is one of the most widely used methods for ing with BLAST and FASTA involve statistics: (i) compar-
analyzing bacterial genes and genomes. Most molecular ing DNA sequences to a DNA sequence database, instead
bacteriologists have used similarity searches to identify in- of using protein or translated DNA sequence comparisons;
dividual genes, and with the decreasing cost of DNA se- (ii) focusing on percent identity, rather than expectation
quencing, it is becoming more routine to consider large- value, when inferring homology; (iii) searching databases
scale sequencing of diverse bacterial populations. While the that are much larger than necessary; and (iv) using expec-
common practice of BLAST-ing bacterial sequences tation value thresholds that are too stringent. The sections
against the NCBIs comprehensive NR (nonredundant) below present the philosophical basis for the relationship
database can provide important new scientific insights, it is between homology and statistical significance and then
rarely the most effective strategy for characterizing bacterial present the statistical equations that describe local similar-
sequences. In this chapter, I focus on more effective meth- ity scores. From these equations, several strategies for im-
ods for similarity searching with protein and DNA se- proving statistical significance, and thus the ability to rec-
quences, with the goal of identifying distantly related genes ognize homologs, emerge. Finally, potential failures in the
in bacteria and bacterial populations. statistical model are discussed.
While strategies for similarity searching are often pre-
sented as a process of selecting a sequence comparison pro-
gram, and possibly the scoring matrix and gap penalties, this 35.2. SIMILARITY, HOMOLOGY, AND
chapter provides a different perspective. The inference of STATISTICS
homology is based on parsimony and statistics. If two se- Effective strategies for sequence similarity searching are best
quences share much more similarity than is expected by understood by exploring the statistical basis for inferring
chance, the most parsimonious explanation for the excess homology. Sequence comparison programs like BLAST and
similarity is that they derived from a shared ancestor. FASTA are designed to find homologous sequences-se-
Homologous sequences have similar three-dimensional quences that share excess similarity because they descended
structures, and often, but not always, they have similar from a common ancestor. Over the past 20 years, the term
functions. The goal of sequence similarity searches is to find homology has emerged from evolution journals to com-
homologous sequences, and the inference of homology is mon parlance, but the term is still frequently misused (two
statistical. Thus, this chapter summarizes the statistical sequences are never 50% homologous, though they may be

842
35. Genome Similarity Searching 843

50% identical [14]). Homology is a qualitative inference- 1 parameter; if the scoring matrix values are doubled, 1 is
two proteins (or protein domains) either are homologous or halved so that 1s remains the same. The K parameter cor-
are not-and it is a statement about the evolutionary his- rects for the lengths of the sequences, and there are more
tory of a protein that existed hundreds of millions or bil- places to start an alignment between two long sequences
lions of years in the past-the common or shared ancestor than between two short sequences, and the normalized
of the homologous proteins. score S must be adjusted to correct for this effect. Once the
The great success of computational genome analysis re- raw alignment score S,, has been normalized to S using
flects the fact that for many protein families, their se- equation 2, the probability of the score can be calculated
quences change so slowly that statistically significant align- using equation 1.
ments can be found after billions of years of divergence. For Equation 1 can also be rewritten as
example, the human mismatch repair mutS homolog MSH2
P(Sbit > x) = 1 - exp(mn2-) (3)
shares more than 30% protein sequence identity with most
bacterial mutS proteins; alignments between the human where
and prokaryotic proteins have similarity scores that are ex-
pected less than once in lo4 database searches. The most (4)
parsimonious explanation for this high level of sequence Sbit is the bit score reported by current versions of the
similarity is descent from a common ancestor. A n alterna- BLAST and FASTA programs. For statistically significant
tive explanation would require that the bacterial and eu- bit scores (P < 0.01), equation 3 can be rewritten as
karyotic mismatch repair proteins arose independently in at
least two separate events; it is much simpler to assume,
P(Sbit > x ) = mn2- (5)
when presented with two sequences that are much more Sbit similarity scores have the convenient property that in-
similar than expected by chance, that they were copied creasing the score by 1 bit halves the probability (a 3-bit in-
from a single common ancestor. Thus, the inference of ho- crease improves it eightfold), and the probability of the
mology is statistical; we need to have confidence that we score can be calculated knowing only m and n, the lengths
know how often a similarity score might occur by chance, of the two sequences, and without knowing A or K.
so that we can recognize excess similarity that is signifi- The extreme value distribution (equations [REF: extv-
cantly greater than that expected by chance. eqnll and [REF: s-norm12) reports the probability that two
sequences, picked independently, will produce a given pair-
35.2.1. Homology and Statistical Significance wise similarity score. However, the high-scoring sequences
Most biologists are comfortable concluding that when two from a similarity search are not independent; the sequences
sequences are 70 to 100% identical over their entire length, of interest are usually examined because they were the
they must be homologous. But many investigators are less highest-scoring sequences in a comprehensive similarity
comfortable with this inference when the level of similarity search. Thus, one must correct for the fact that tens or hun-
drops below 40% identity. Because of the complex relation- dreds of thousands of sequences were examined to find the
ship between sequence similarity, sequence identity, align- highest-scoring sequences. (While the odds of a fair coin
ment length, and statistical significance, statistical intu- coming up heads 10 times in exactly 10 tosses is about
ition often fails us. Fortunately, modern sequence databases 1/1,000, one expects to see 10 heads in a row after about
are large and very comprehensive, with hundreds of thou- 1,000 tosses, and one would be very surprised not to see 10
sands of unrelated sequences. Thus, when analyzing the re- heads in a row after 3,000 tosses.) As a result, modern sim-
sults from a sequence similarity search, we need not specu- ilarity searching programs report the expectation that a
late whether a score is much greater than expected by score would occur by chance:
chance; we can examine the distribution of scores, particu-
larly the high-scoring sequences, to evaluate the accuracy of E(S) = P(S)D (6)
the statistical estimates. With rare exceptions, these esti- where D is the size of the database searched (the number of
mates are accurate, so that statistically significant similarity, alignment scores calculated). [The BLAST programs calcu-
even between sequences that share less than 20% identity, late the expectation using a slightly different form: E(S) =
can be used to infer homology. P(S)N/n, where N is the total number of residues in the
Thus, to infer homology reliably, we need accurate esti- database and n is the number of residues in the library se-
mates of how likely an alignment score will occur by quence.]
chance. The probability that two random sequences will To illustrate the use of the various statistical measures of
produce local similarity score S is given by the extreme similarity and the effects of different search strategies on
value distribution (2; reviewed in reference 13): search sensitivity, I will use two example query sequences from
Methanococcus jannaschii: (i) hypothetical protein MJ0050
P (S > x ) = 1 - exp(e-) (1) (SwissProt YOSO-METJA, NCBI RefSeq NP-247014), a
where group I1 decarboxylase homolog (Table lA), and (ii) hypo-
thetical protein MJ1633 (SwissProt YG33_METJA, RefSeq
S = XS,,, - ln(Kmn) (2) NP-248643), which shares a potassium efflux antiporter do-
Equation 1 is a simplified form of the extreme value distri- main with several Eschenchia coli proteins (Table 1B). The
bution; equation 2 reflects the fact that sequence raw simi- MJ0050 decarboxylase homolog is an example of a protein
larity scores for random alignments depend on the scoring that shares global similarity with most of its homologs, i.e.,
matrix and the lengths of the two sequences. The role of the similarity that extends from the beginning of the query se-
scoring matrix is apparent from the following argument: if quence and extends to the end, so that the alignment length
all the values in the scoring matrix were doubled, then is similar to the query sequence length. In contrast, only a por-
alignment scores using the doubled matrix would be twice tion of MJ1633 aligns with the potassium transport domain;
as high, yet their statistical significance would be un- here the alignment length is only about 40% of the query se-
changed. Thus, raw similarity scores S,,, are scaled with the quence length.
844 w COMMUNITY AND GENOMIC ANALYSIS

For the alignment between MJ0050 and glutamate de- [the parentheses in the E() notation indicate that the ex-
carboxylase (NP-416010) in Table lA, one can calculate pectation depends on the number of sequences, in this case
the probability that the 66-bit alignment score would occur the 4,311 E. coli proteins] would not generally be consid-
by chance in a single pairwise comparison using equation 5: ered statistically significant (similar scores will occur by
chance every 40 database searches; a more reliable signifi-
P(Sbic 2 x) = mn2-"
cance threshold is 0.001 to 0.01), but MJ1633 and
P(sbit 2 66) = 396 X 466 X 2? NP-415011 do share a homologous domain, which can be
readily identified by searches with members of the family
= 184,536 X 1.36 X lop2' from E. coli.
= 2.4 x 10-l~ Homologous sequences that do not share significant pair-
wise similarity are generally identified using one of three
However, this pairwise probability does not reflect the strategies. (i) The first is significant tertiary structural simi-
chance of seeing a 66-bit match after examining each of the
larity (when three-dimensional structures are available for
4,311 sequences in this E. coli proteome, which is
the sequences or their clear homologs). (ii) The second is
E(Sbi, 2 X) = P(sbit 2 x)D significant similarity to intermediate homologs (transitive
homology)-in Table lB, MJ1633 does not share significant
E(Sbit 2 66) = 2.4 X X 4,311
similarity to NP-415011, but it does share significant simi-
= 1.1 x lo-" larity with NP-414589 [E() < which in turn shares
very strong similarity with NP-415011 [E() < 1Ow2Y.
This 66-bit similarity score is expected to occur by chance
Homology is transitive; if A is homologous to B and B is ho-
only once in 10" database searches; the most parsimonious
mologous to C over the same domain, then A can be inferred
explanation for this strong similarity is descent from a
to be homologous to C, even when they do not share signif-
shared ancestor.
icant pairwise similarity. (iii) The third strategy is significant
35.2.2. Homology and Nonhomology similarity to a profile, a position-specific scoring matrix (4),
or Hidden Markov model (16) for the protein family. Thus,
One of the most confusing aspects of homology inference is
conventional pairwise similarity searches can establish ho-
its nonreversibility. Sequences that share statistically signif-
mology based on statistically significant similarity, but they
icant similarity are almost always homologous (some patho-
cannot be used to establish nonhomology.
logical cases are considered in section 35.7), but the inverse
is not true: sequences that do not share statistically signifi-
cant similarity are not always nonhomologous. For example, 35.2.3. Expectation and Percent identity
in Table lB, the alignment between MJ1633 and the puta- The MJ0050 glutamate decarboxylase alignment illustrates
tive transport protein (NP-415011) with an E() of <0.025 a common error in inferring homology from sequence simi-

TABLE 1 Sample search with query sequences from M. jannaschii"


Sequence Length s-w Bits E(4,311) % ID AL
A. MJ0050, 396 aa, best scores
NP-416010 glutamate decarboxylase 466 250 66.0 1.1 x lo-" 24.9 40 1
NP-417379 glycine decarboxylase 957 169 46.2 2 x 10-~ 22.1 420
NP-417025 putative aminonansferase 404 122 35.3 0.016 23.6 254
NP-414772 aminoacyl-histidine 485 110 32.3 0.15 23.4 188
NP-415139 alkyl hydroperoxide 53 1 99 29.6 1.o 26.9 156
NP-417345 putative transcription 592 92 27.9 4.0 25.2 107

B. MJ1633,478 aa, best scores


NP-417809 KefB (E. coli) 601 196 49.0 2.2 x 28.2 177
NP-414589 K+ efflux antiporter 620 175 44.4 5.4 x 1 0 - ~ 25.4 142
NP-415011 putative transport protein 558 133 35.4 0.025 23.2 142
NP-417748 TrkA (E. coli) 458 128 34.4 0.041 23.7 135
NP-416807 putative sugar nuclease 297 103 29.2 0.98 26.1 92
NP-418046 putative oxidoreductase 382 104 29.3 1.2 22.2 171
NP-416911 PEP-protein phosphotransferase 575 106 29.6 1.5 22.2 234
NP-418023 xylose binding protein 330 100 28.5 1.8 21.3 253
NP-417468 putative hydrogenase 567 104 29.2 1.9 23.4 316
NP-417367 peptide chain release 365 99 28.3 2.3 22.3 260
""Typical" sequence similarity searching results for comparison of MJ0050,SwissProt YO50_METJA,or MJ1633,SwissProt YG33_METJA,with the E. coli proteome.
Comparisons used the Smith-Waterman algorithm (4) as implemented by the SSEARCH program from the FASTA package (5). Results include the description of the
E. coli library sequence, the length of the library sequence, and four measures of sequence similarity: the raw Smith-Waterman alignment (S-W) score using the
BLOSUMSO matrix and a penalty of -10 for a gap open and -2 for each residue in the gap; the bit score (bits) as defined in equation 5; E(4,311),the expectation, as
defined in equation 6; the percent identity (% ID); and alignment length (AL). aa, amino acids.
3 5 . Genome Similarity Searching 845

larity-focusing on the percent identity and alignment length to remember that it is essentially a structural method.
rather than the bit xoye or expectation. In this example, the Homologous proteins have similar structures, but not nec-
closest homolog (NP-416010) has a very significant align- essarily similar functions. Homology can be subdivided into
ment score [E() < lo-], yet shares less than 25% identity two types: orthology and paralogy. Orthologous sequences
over 400 amino acids, a level of identity often considered differ because of speciation events; for example, cytochrome
marginal. For example, the nonhomologous NP-415 139 c in humans is both homologous and orthologous with cy-
shares higher percent identity (26.9%) with MJ0050 tochrome c in fish, insects, yeasts, and plants. Orthology is
(though over a shorter aligned region, 156 residues). And most easily established for genes like the cytochrome c gene
the alignment between MJ0050 and glycine decarboxylase which are found once per haploid genome, and are found in
has even lower identity (22%), but it is also very statistically most organisms. The differences in the cytochrome c se-
significant [E() < 2 x lop5].While the widely used rule of quences of humans, insects, plants, and fungi can be used to
thumb-that two sequences that share more than 30% produce an accurate speciesphylogenetic tree that reflects
identity over their entire lengths are homologous-is usue the evolutionary history of the organisms.
ally correct in predicting homologs, it is much too conser- In contrast, paralogous sequences (which are also ho-
vative. In a large-scale comparison of human proteins to E. mologous) have diverged from one another after a gene du-
coli proteins, the median percent identity for alignments plication event. Thus, hemoglobin a,P-globin, and myo-
with E() was 28.5%, one-fifth of the align- globin are all paralogous sequences; they appeared after
ments shared less than 25% identity and one 19.2% identi- successive gene duplication events in the vertebrate line-
cal alignment has an E() While high identity age. Evolutionary trees of paralogous sequences do not re-
(>30%) over long regions is a reliable indicator of homol- flect the evolutionary history of the organisms where they
ogy, many biologically interesting homologs are readily are found; they reflect the evolutionary history of the gene
found with very significant expectation values despite shar- family.
ing less than 25% identity. Statistical significance-the ex- While homologous sequences need not share a common
pectation value-is the most sensitive and most reliable in- function, orthologous sequences are very likely to have very
dicator of homology. similar or identical functions. In the case of proteins like
cytochome c, which are present only once per haploid
35.2.4. Improving Sensitivity with Smaller genome, the critical function of the gene must be preserved
Databases-Part I in other organisms. Thus, databases like COGS (Clusters of
Equation 6 provides a simple strategy for dramatically im- Orthologous Groups [18]) can be used to provide more reli-
proving the sensitivity of a similarity search: search smaller, able functional inferences than similarity searches alone.
genome-sized, databases. For example, an alignment with Alternatively, paralogous sequences, because they are dupli-
an Sblt of 40 between two 200-residue proteins would not be cated genes, are more likely to acquire different functions.
significant in a search of the NCBI NR database, with about Reliably distinguishing orthology from paralogy over long
1.6 million sequences: evolutionary distances can be very difficult. A common
strategy for assigning orthology is by finding the reciprocal
E(Sblt 2 40) = P(Sb,, 2 40)D best BLAST hit; sequence b in genome B is orthologous to
= ZOO x ZOO x 2-40 x 1.6 x 106 sequence a in genome A if b is the best match to a in
genome B, and a is the best match to b in genome A. More
= 3.64 X lo-* X 1.6 X lo6 robust strategies for inferring orthology build evolutionary
= 0.06 trees (chapter 36) for the protein families of interest (20).
However, in a search of either the E. coli or human genome 35.2.6. Statistical Thresholds and large-Scale
alone, the expectation would be Similarity Searching
E(Sblt 2 40) = P(Sblt 2 40)D The expectation value of an alignment score is the most
sensitive and most reliable indicator of homology (section
lop8 X 4,300 (E. coli)
= 3.64 X 35.2.3). The expectation in equation 6 reports the number
1.6 x 1 0 - ~
= of times a score is expected in a single similarity search. For
protein sequences, the expectation value calculated by the
= 3.64 X lop8 X 40,000 (human) BLAST and FASTA programs is quite accurate; random se-
= 1.5 x 1 0 - ~ quences will receive expectation values of 0.02 about 1 time
in 50, and real, unrelated protein sequences will typically
The comprehensive NR database has almost 400 times as receive expectation values between 0.004 and 0.01 about 1
many sequences as most bacterial genomes, so any search time in 50 (6, 10). Thus, with real sequences, reported ex-
against the NR database is 400 times less sensitive than a pectation values sometimes overestimate statistical signifi-
search against a single prokaryotic genome. Thus, while the cance by a small factor, of 2 to 5. To adjust for this error,
most common BLAST search strategy-searching the de- conservative statistical significance thresholds between
fault NR database-provides the most comprehensive 0.001 and 0.01 are generally used to infer homology.
search, it also provides the least sensitive search. A taxo- The relationship between statistical significance and
nomically diverse selection of sequenced genomes can pro- homology is not symmetric; while proteins with pairwise
vide a much more sensitive search strategy (e.g., 10 prokary- alignment scores with an E() of <0.001 are almost always
otic genomes might contain 40,000 sequences) with little homologous, proteins with worse E() values are not neces-
loss in diversity (section 35.5.3). sarily nonhomologous. Proteins with alignment scores be-
tween 0.01 < E() < 0.05 are probably homologous as well,
35.2.5. Homology and Function-Orthology but unrelated sequences will obtain scores this good by
While similarity searching is a very sensitive and reliable chance in 5 to 10% of searches. Indeed, since very distantly
technique for characterizing new sequences, it is important related homologous sequences need not share statistically
846 COMMUNITY AND GENOMIC ANALYSIS

significant pairwise similarity, homologous sequences quence databases: BLAST (4) and FASTA (11).BLAST is
sometimes have an E() of >1.0 (and homology must be in- by far the more popular; BLAST is considerably faster than
ferred from other analyses). The E() -1 criterion is useful FASTA and integrated into the comprehensive sequence,
for evaluating statistical estimates because, on average, the protein family, and structure databases at the NCBI website
highest-scoring unrelated sequence should have E() -1. In (http://www.ncbi.nih.gov). The FASTA programs are avail-
Table 1, the highest scoring sequences that are clearly un- able at the European Bioinformatics Institute website
related to the query have an E() of <0.15 (MJOOSO versus http://www.ebi.ac.uk/fasta; source code for the FASTA
NP-414772, aminoacyl-histidine dipeptidase) and an E ( ) package is available from ftp://ftp.virginia.edu/pub/fasta.
of <0.98 (MJ1633 versus NP-416807, a putative sugar nu- Both the BLAST and FASTA packages provide a vari-
cleotide epimerase). Although one cannot infer nonho- ety of different programs for protein, DNA, and translated-
mology based on a lack of statistical significance in a single DNA searches (Table 2). As discussed in the next section,
similarity search, additional searches can confirm that li- all similarity searching programs are most effective when
brary sequences with expectations around 1 are probably comparing sequences at the protein or translated-protein
not homologous. The decarboxylases in Table 1A and the level, and modern versions of the BLAST and FASTA pro-
transport proteins in Table 1B belong to large protein fam- grams provide efficient algorithms for translated searches
ilies; the E. coli glutamate decarboxylase (NP-416010) with sequences that may include frameshift errors and ter-
shares statistically significant similarity [E() < 0.011 with mination codons.
more than 20 glutamate decarboxylases in SwissProt (5) For protein sequence comparison, the major difference
from a broad range of bacteria, archaea, yeasts, plants, and between BLASTP and FASTA is the strategy used to esti-
animals. None of the members of this family share signifi- mate statistical significance. (The two programs also differ
cant similarity with NP-414772, strongly suggesting non- in their default amino acid scoring matrices; BLASTP uses
homology. Identification of the highest-scoring candidate BLOSUM62, while FASTA uses BLOSUMSO; see section
nonhomolog, and confirming the nonhomology with ad- 35.5.1). The original BLAST programs (3) used analytical
ditional sequence database searches, provides empirical formulas to calculate the X and Kparameters for equation 2.
confirmation of the accuracy of the statistical estimates Unfortunately, these equations applied only to local align-
and allows a less stringent significance threshold [E() ments without gaps, and alignments with gaps are significantly
< 0.011. more sensitive. With the introduction of gapped-BLAST
The statistical thresholds discussed in this section apply (4), A and K were calculated for specific combinations of
to one or a few sequence similarity searches. When a very scoring matrices and gap penalties using a simulation strat-
large number of searches are performed, more conservative egy. Several recent improvements to BLAST involve im-
thresholds must be used. An accurate E() of <0.01 states provements to the statistical estimates.
that the alignment score will occur one time in 100 The FASTA programs calculate statistical estimates
searches by chance; thus, in a large-scale genome analysis from the distribution of unrelated-sequence scores that are
that searches with 2,000 candidate open reading frames, an produced in any comprehensive database search (10).
E() of <0.01 should occur 2,000 X 0.01 = 20, i.e., times by (When unrelated scores are unavailable, fasta can use shuf-
chance. A conservative correction for the large number of fled sequences to estimate the statistical parameters.) Thus,
searches reduces the statistical significance threshold by a FASTA effectively calculates different X and K parameters
factor based on the number of searches. It is common to use for each query sequence in each database search. This strat-
a statistical thresholds of an E() of <lop6 when thousands egy can be more robust to unusual properties in query se-
of searches are done. This conservative approach seeks to quences.
reduce the number of false positives-sequences tentatively For DNA sequence comparison, the empirical statistical
identified as homologous but that actually obtain their high estimation approach used by FASTA is often more accurate
scores by chance because of the large number of searches. than BLASTN estimates because unrelated DNA sequences
Unfortunately, particularly in bacterial genome analysis, are highly nonuniform and differ considerably from simple
where the evolutionary distances to the nearest phyloge- random sequences. (In contrast, real protein sequences are dif-
netic neighbor can be quite large, increasing the statistical ficult to distinguish from random sequences.) In addition, the
stringency from lop3to lop6can also dramatically increase default BLASTN parameters for DNA alignments, + I for a
the number of false negatives, sequences that are homolo- match and -3 for a mismatch, optimally identify sequences
gous but no longer meet the statistical criterion. For exam- that are more than 99% identical. DNA comparison can
ple, in a comparison of 1,731 M. jannaschii sequences with identify significant alignments between sequences that share
the E. coli proteome using the Smith-Waterman algorithm, as little as 70% identity; the FASTA program uses +5/-4
there were 3,251 alignments with anE(4,311) of but match/mismatch values, which are targeted for 70% identical
only 2,227 with an E() of Because there were 1,711 alignments (17). For more sensitive comparison with the
queries, we might expect that 2 of the 1,024 alignment BLAST package, the TBLASTX program, which compares
scores with < E() < occurred by chance, but six-frame translations of both the DNA query and DNA li-
more than 1,000 of these alignments probably reflect gen- brary sequence, is sometimes used. In general, FASTA is both
uine distant homologs that become false negatives at the more sensitive and more selective (because of more accurate
stricter statistical threshold. The statistical threshold for de- statistics) than BLASTN for DNA sequence comparison.
finitively annotating sequences based on significant similar- Unfortunately, FASTA is also considerably slower.
ity is very different from the appropriate threshold for ten- For translated-DNA sequence comparison, both
tatively identifying a sequence as unique, or unrelated to BLASTX/TBLASTN and FASTX/TFASTX use sophisti-
known proteins. cated alignment algorithms that provide almost as much
sensitivity as protein-protein sequence comparison. The
major difference between the two programs is in the treat-
35.3. SIMILARITY SEARCHING PROGRAMS ment of frame shifts; FASTX and TFASTX use a more so-
As this chapter is written in late 2003, there are two widely phisticated alignment algorithm that allows alignments to
used sets of programs for searching protein and DNA se- continue across nucleotide insertions and deletions that
35. Genome Similarity Searching 847

TABLE 2 BLAST and FASTA similarity search programs


Query Library BLAST Matrix FASTA Matrix
Protein Protein BLASTP BL62 FASTA BL50
SSEARCH BL50
DNA DNA BLASTN +11-3 FASTA +5/-4
DNA Protein BLASTX BL62 FASTX BL50
Protein DNA TBLASTN BL62 TFASTX BL50
DNA DNA TBLASTX BL62
Protein Fragments Protein FASTS MD20
Protein Fragments DNA TFASTS MDlO

cause translation frameshifts. Frameshifts cause BLASTX/ DNA-protein alignments with bit scores of >330, or ex-
TBLASTX to break alignments into separate segments, pectation values of an E() of with 40% identity.
which can have incorrect boundaries. The BLASTX alignments with bit scores between 45 and
The FASTA package also includes SSEARCH, an im- 200 correspond to much more distant homologs, with
plementation of the optimal Smith-Waterman algorithm amino acid percent identities as low as 25%, which are
(15). Although it is as much as 100 times slower than missed with DNA sequence comparison. Indeed, using a
BLASTP, the Smith-Waterman algorithm can be more sen- translated-DNA search against archaeal proteins in
sitive than the heuristic BLAST and FASTA algorithms, Genpept, homologs with bits scores of >50 are found in 12
and on modern gigahertz computers, searches against bacte- euryarchaeotal organisms and Aeropyrum pernix, a crenar-
rial genomes take only tens of seconds. In addition, network chaeota. None of these homologs are found with BLASTN
parallel versions of the FASTA programs (including using the more sensitive +3/-3 scoring parameters.
SSEARCH) are available that allow large-scale bacterial Protein or translated-DNA sequence comparison is far
genome comparisons using the Smith-Waterman algorithm more sensitive than DNA sequence comparison for several
to finish in a few hours. reasons. (i) Protein substitution matrices distinguish con-
servative (e.g., lysine to arginine) and nonconservative
(e.g., leucine to glutamate) amino acid replacements. (ii)
35.4. PROTEIN VERSUS DNA SEQUENCE This distinction is lost in DNA scoring matrices, and the
COMPARISON problem is compounded by the fact that some DNA substi-
The most common sequence similarity search performed at tutions are silent at the codon level, while others cause
the NCBI website, using BLASTN to compare a DNA se- dramatic amino acid replacements. Protein-based compar-
quence to a DNA sequence database, is also the least effec- ison is also more sensitive because of the larger alphabet
tive search strategy. O n average, DNA-DNA sequence com- and the shorter sequences. (iii) In addition, protein align-
parison is 5 to 10-fold less sensitive than the equivalent ment statistics are far more accurate than DNA statistics.
protein or translated-DNA similarity search. In practice, A n E() of is a very safe threshold for statistical sig-
the most sensitive DNA searches for protein coding see nificance for protein alignments; for DNA comparisons,
quences provide a 200 million- to 500 million-year look- thresholds of an E() of or even are required to
back time; similar protein or translated-DNA searches rou- avoid false positives. (iv) Moreover, low-complexity regions,
tinely identify homologs that diverged 1 billion to 2 billion which are the most common cause of statistical errors, are
years ago. easily removed from protein sequences; many DNA se-
The dramatic difference in search effectiveness with quences that encode low-complexity protein sequences are
DNA versus translated-DNA or protein searches is illus- not easily recognized.
trated in Table 3. Here, the taxonomic results from three A common excuse for searching with DNA sequences,
searches are shown, each with exactly the same DNA query rather than protein sequences, is that the sequences of in-
(the DNA sequence encoding E. coli glutamate decarboxy- terest are of relatively low quality and may contain
lase): a BLASTX translated-DNA search against the pro- frameshifts or codon errors that would produce incorrect
teins in the bacterial division of Genpept, and two DNA translations. However, it is not necessary to first translate a
searches, one with relatively sensitive BLASTN parame- DNA sequence before searching a protein database; both
ters, + 3 for a match and -3 for a mismatch, and a second FASTX and BLASTX use algorithms that are robust to
with the default (+ 1/-3) BLASTN scoring values. With both frameshifts and codon errors. These errors can de-
the translated BLASTX search, the DNA query can identify crease the sensitivity of the search substantially, but not
homologs in a broad variety of organisms, including several nearly to the level of a DNA-DNA search.
that are very distantly related to the proteobacteria. In cone The most effective strategy for improving sequence sim-
trast, when BLASTN is used with its default scoring param- ilarity search effectiveness is to search with protein or
eters, only very closely related homologs are found. These translated-DNA sequences. Protein-based alignments pro-
genes probably diverged within the past 100 million years vide 5- to 10-fold-deeper evolutionary look-back time. For
(the a-proteobacterial homolog is likely to have arisen by studies of Bacteria sequences, where the most closely re-
horizontal gene transfer; the E. coli and Brucella melitensis se- lated organism may have diverged more than 2 billion
quences are 77% identical at the protein sequence level). years ago, DNA sequence comparison is impractical for all
BLASTN searching is far more sensitive when less strin- but the most highly conserved sequences. Even in those
gent scoring parameters are used (+3/-3), but even then, cases, the more sensitive (+3/-3) BLASTN parameters
the most distant DNA matches correspond to translated- should be used.
848 COMMUNITY AND CENOMIC ANALYSIS

TABLE 3 DNA and translated-DNA similarity searches'


BLASTN
Taxonomic group BLASTX TAXON
+3/-3 + 1/-3
Bacteria eubacteria
Proteobacteria
y -Proteobacteria
Enterobacteriaceue
Shigella enterobacteria
Shigella flexneri 2a 979 2,165 2,595 Enterobacteria
Escherichia coli CFT073 976 2,130 2,508 Enterobacteria
Escherichia coli 0157:H7 959 2,184 2,642 Enterobacteria
Escherichia coli 758 2,253 2,817 Enterobacteria
Edwardsiella tardu 784 1,102 180 Enterobacteria
Brucella melitensis 16M 496 854 113 a-proteobacteria
Mesorhizoobium loti 60 a-proteobacteria
Bordetella bronchiseptica RB 330 217 P-proteobacteria
Geobacter metullireducens 53 6-proteobacteria
Geobacter sulfurreducens PCA 53 6-proteobacteria
Prochlorococaa marinus MIT 517 458 Cyanobacteria
Synechocystis sp. strain PCC 6803 466 284 Cyanobacteria
Closcridium perfringens strain 13 427 Eubacteria
Streptomyces coelicolor A3(2) 41 7 High-GC gram+
Mycobacterium tuberculosis 414 311 High-GC gram'
Listeria innocuu 414 257 Eubacteria
Listeria monocytogenes 414 234 Eubacteria
Enterococcus faecium 41 1 Eubacteria
Streptomyces uvermitilis MA4680 409 High-GC gram+
Lactococcus lactis 405 183 Eubacteria
Lactobacillus plantarum WCFSl 390 23 1 Eubacteria
Bacteroides thetuiotaomicron VPI 387 233 CFB group bacteria
Chloroflexus aurantiacus 72 GNS bacteria
Gloeobacter violaceus PCC 7421 48 Cyanobacteria
Streptomyces viridifaciens 45 High-GC gram+
Clostridium tetani E88 45 Eubacteria
"Bit scores from BLASTX and BLASTN searches presented using the BLAST taxonomy summary option. The DNA sequence (M84025) encoding E . coli
glutamate decarboxylase was used to search the bacterial division of GenBank or Genpept. Species that contain a homolog with a bit score of 245 [E() < lo-' for
BLASTX] are shown. The numbers under the BLASTX and BLASTN columns indicate the highest bit score obtained for that taxonomic group. gram+, gram positive;
CFB, Cytophaga-FLcuobacterium-Bacteroides;GNS, green nonsulfur.

35.5. IMPROVING SEARCH SENSITIVITY more effective at identifying shorter domains than the
Because homology is a statistical inference, the terms in default matrix (BLOSUMSO) and gap penalties used by
equations 1, 2, and 6 are the basis for any strategy to im- FASTA and SSEARCH; the latter scoring parameters are
prove search sensitivity. From equations 1 and 2, the prob- more effective for long alignments (>lo0 amino acids).
ability of a pairwise score can be increased by increasing S' The relationship between scoring matrix, evolutionary dis-
= AS - ln(Kmn); from equation 6, one can improve the ex- tance, and alignment length was described in detail in ref-
pectation by searching smaller databases. erence 1; for the BLOSUM matrices (8), matrices with
higher numbers (BLOSUM6LBLOSUM80) are more ef-
35.5.1. Improving Pairwise Similarity Scores fective at finding shorter homologous domains while the
Improving S' largely involves increasing XS, the similarity BLOSUM45-BLOSUM50 matrices can be used for longer,
score. [The ln(Kmn) term in equation 2 is usually much more distant, relationships.
smaller than XS, and m and n are fixed.] Similarity scores The effects of different scoring matrices and gap penal-
can be improved by selecting a scoring matrix and gap ties are shown in Table 4. The MJOOSO query shares global
penalties that are most appropriate for the evolutionary dis- similarity with most of its homologs, and the BLOSUM50
tance and length of the homologous domain. For example, matrix is the most effective at identifying homologs. When
the BLOSUM62 matrix and gap penalties ( - 11 to open a the default BLASTP scoring parameters (BLOSUM62,
gap; -1 for each residue in the gap) used by BLASTP are - 11/- 1) are used, the statistical significance and the align-
35. Genome Similarity Searching 849

ment length of both homologs decrease substantially. When


lower gap penalties (-7/-1) are used with BLOSUM62,
the alignment length increases to that found with
BLOSUM50, presumably the entire length of the homolo-
gous domain.
For the local alignments of MJ1633 with its homologous
domains, BLOSUM62, - 11/- 1, performs dramatically bet-
ter than the other scoring parameters, even though some-
times a shorter aligned domain is found. Indeed, it seems
likely that the alignment between MJ1633 and kefB pro-
duced by BLOSUM50 is too long; other alignments of this
-
domain are 140 residues.
Thus, for weak or mrgimlly significant alignments
< E() < 0.11, the length of the alignment can serve a guide
for the most appropriate scoring matrix. If the alignments are
short (<ZOO residues), then the BLOSUM62 matrix with
stringent gap penalties (- 11/- 1)may improve statistical sig-
nificance; if the alignments are even shorter (<lo0 residues),
BLOSUM80 may be more appropriate. BLOSUM62 and
BLOSUM80 are particularly effective when searching with
short query sequences, as when searching for unannotated
proteins in short intergenic regions. However, these examples
were selected to highlight differences in performance with
different scoring matrices. In most cases, particularly for less
distant relationships, the scoring matrix does not matter; the
homologs will have very similar bit scores and E() values
with either BLOSUM50 and BLOSUM62.
35.5.2. Adjusting Scoring Matrices for Evolutionary
Distance
The default scoring matrices and gap penalties used by
BLASTP and FASTA are designed for maximum sensitiv-
ity-detecting the most distant evolutionary relations pos-
sible. While this is certainly the most common task, in
some cases we seek to restrict our focus to a particular evo-
lutionary distance. This is particularly true when trying to
identify orthologs of a protein family in relatively closely re-
lated species ( 4 0 0 My).Table 5 shows a typical case,
where I seek to identify the ortholog of the E. coli KefB pro-
tein in Vibrio cholerae and Yersinia pestis.
While the widely used BLOSUM (8) scoring matrices
are the most effective at finding distant homologs, the PAM
series of matrices (7) are built using an evolutionary model
that can produce matrices appropriate for any evolutionary
distance. (A set of PAM matrices based on a more modern
and comprehensive protein set is described in reference 9.)
The number associated with each PAM matrix, e.g.,
PAM20 or PAM120, indicates the amount of evolutionary
change for which the matrix is targeted; PAM20 is targeted
for 20% change, or sequences that are 80% identical; 120%
change produces sequences that are about 40% identical.
(The 120% change, or PAM120, denotes 120 changes per
100 residues; after 120 changes in 100 positions, the two se-
quences will be 40% identical, on average, because of mul-
tiple hits at some sites and reversions at other sites. After
250% change [PAM250], homologous sequences remain
20% identical [7].) Table 5 shows that the scoring matrix
that most closely approximates the correct evolutionary dis-
tance produces the most statistically significant alignment
(1). For example, E. coli KefB and the E. coli paralog
NP-414589 are about 45% identical, and PAM120 pro-
duces the most statistically significant alignment between
these two sequences. While the BLOSUM50 alignment is
about the same length and identity, it is much less statisti-
cally significant; BLOSUMSO is targeted to much more dis-
tant (<25%) identical alignments.
TABLE 5 Identifying orthologs

BLOSUMSO PAM120 PAM20


Sequence
E(4.3 11) % ID AL E(4.3 11) % ID AL E(4.311) % ID AL
E. coli orthologs and paralogs, best scores
NP-417809 KefB ( E . coli) 5 x 10-173 100 601 0 100.0 60 1 0 100.0 601 >
NP-414589 Kf efflux antiporter 4 x 10-72 44.2 608 10-143 45.3 563 47.8 502 Z
0
NP-415011 putative transport protein 6X 30.5 525 2 x 10-43 30.2 493 0.045 47.1 51
NP-415709 hypothetical protein 0.013 22.5 427
NP-418489 hypothetical protein 0.12 22.5 306
NP-417959 arsenical pump 0.22 23.9 364
NP-417364 putative permease ( E . coli) 0.24 24.2 397
NP-417559 putative transport protein 0.3 25.9 263

Y. pestis orthologs and paralogs, best scores


NP-671265 K+ efflux; NEM-act 3x 77.9 602 0 77.9 602 0 78.1 603
NP-668414 putative K+ efflux 7x 31.1 534 2 x 10-40 30.4 493 3.5 49.0 51
NP-668178 arsenical pump 0.12 23.7 375
NP-669263 putative transport protein 0.3 1 25.3 340
NP-668704 putative permease 0.56 25.7 296
NP-67 1240 permease (major) 0.71 23.6 296
NP-671313 potassium uptake protein 0.76 29.3 99 0.059 28.3 99
NP-671412 putative membrane 0.85 24.0 329

V. cholerae orthologs and paralogs, best scores


NP-232234 glutathione regulator 4 x 1o-lo7 58.3 597 6X 58.9 589 0 59.6 589
NP-230638 glutathione regulator 3 x 10-l6 25.2 527 3 x 10-~ 30.7 137 9.6 80.0 10
NP-232330 conserved hypothetical 3 x 10-~ 24.5 335 2.1 33.3 48
NP-229702 potassium uptake protein 0.024 28.9 114
NP-230338 conserved hypothetical 0.051 20.6 45 1
NP-23 1264 NaC/Hf antiporter protein 0.29 26.2 294
NP-229728 multidmg resistance 0.38 23.7 354
NP 232555 NADH dehvdroeenase 0.55 23.0 357
High,scoring sequences found in searches of the E. cob, Y.pestis, and V. cholerae genomes using the E. coli KetB protein sequence (NP-417809). Results with three different scoring matrices are shown: BLOSUM50 (the FASTA
default), PAM120 (appropriate for sequences that are about 40% identical), and PAM20 (appropriate for sequences that are about 80% identical). For abbreviations, see Table 1, footnote u.
35. Genome Similarity Searching 851

The E. coli KefB alignment with Y. pestis NP-668414 produced by selecting all bacterial sequences from the NR
shows how a shallow scoring matrix can focus on more re- database.
cent evolutionary events. The most significant alignment This strategy can also be implemented on local comput-
uses the PAM120 matrix, and because the sequences share ers by downloading the appropriate microbial proteome
about 30% identity, the PAM20 matrix misses the homol- databases from the NCBI and searching with BLASTP (or
ogy altogether. Conversely, the V. cholera KefB ortholog, BLASTX), FASTA (FASTX), or S E A R C H . Complete
NP-232234, shares 60% identity and has the best expecta- bacterial genomes and protein sets are available from
tion value with PAM2O. Targeted scoring matrices, e.g., ftp://ftp.ncbi.nih.gov/genomes/bacteria.Both the BLAST
PAM120 for sequences that have diverged in the past 400 and FASTA programs can be downloaded and run on local
million years or PAM20 for sequences that have diverged in computers. Versions of the programs are available for Unix,
the past 100 million years allow one to focus on recent evo- Linux, Windows, and MacOS operating systems, but it is
lutionary events. typically much easier to manage the various downloading
and searching processes on a Unix/Linux platform.
35.5.3. Improving Sensitivity with Smaller
Databases-Part I I
A second strategy for improving search sensitivity is to 35.6. IDENTIFYING ANONYMOUS
reduce the size of the database being searched (equation 6). SEQUENCES FROM BACTERIAL
The most comprehensive database at the NCBI website, the POPULATIONS
NR database, contained about 1.6 million sequences at the rRNA sequences are widely used to study bacterial diversity
end of 2003. In contrast, bacterial genomes typically con- from uncultured samples. While this has proved to be a very
tain 2,000to 4,000 proteins, and metazoan genomes typi- powerful strategy, and more than 7,000 rRNA sequences are
cally contain 15,000 to 30,000 protein coding genes. When available in current databases, an alternative strategy in-
the taxonomy of a bacterial sequence is approximately volves simply cloning and sequencing DNA from uncul-
known, the most efficient and sensitive characterization tured, anonymous bacterial samples. The current taxo-
strategy would initially search completed genomes from the nomic distribution of sequenced bacteria and archaea is
same class (e.g., y-Proteobacteria) or phylum (Actinobacteria, sufficiently complete that, in general, one would expect
Proteobacteria, etc.). that most bacterial DNA sequences longer than 5,000 nu-
The NCBI BLAST server (http://www.ncbi.nlm.nih cleotides should contain at least one gene, and typically
.gov/BLAST) provides a powerful facility for limiting data- several, that is recognizable based on similarity to se-
base searches to a phylogenetic subset. Under the options for quenced prokaryotic genomes.
advanced blasting,any taxonomic division can be specified, To test the ability of translated-DNA similarity search-
e.g., Actinobacteria[orgn] or txid68525[Organism:exp] ing to identify anonymous DNA sequences, I selected either
(the latter selects S/Eproteobacteria; the txid can be found 10 10,000-nucleotide sequences or 100 1,000-nucleotide se-
with the NCBI taxonomy browser; many NCBI/Entrez quences at random from the completed Archaeoglobus
search term strategies can be found by selecting the Details fulgidus, E. coli 0157:H7, or Screptococcus pyogenes genome.
option). By selecting Actinobacteria, one reduces the size of These DNA sequences were used to search the NR database,
the database search from 1.6 million entries and 520 million excluding all sequences from either the phylum (e.g., Pro-
residues to about 20 million residues. Thus, a search for teobacteria, Firmicutes, Euryarchueota, etc.) or superkingdom
Actinobacteria sequences from the taxonomic subset is about (Bacteria, Archaea [Table 61). Searches were also performed
26 times more sensitive than searching all of NR. using DNA sequences with random mutations (substitu-
While the NCBI BLAST/Entrez taxonomy subset op- tions, insertions, or deletions) introduced at 5% of the posi-
tions can reduce the database size 10- to 20-fold, the taxo- tions. This analysis sought to simulate the analysis of
nomic subset is still very redundant. The NR database does anonymous DNA sequences, with sequencing errors that
not contain any identical sequences, but it contains many might be found in an environmental sample.
sequences that differ by a single residue, and thus the NR Anonymous DNA sequences with average lengths of
database is very redundant. For example, while the anno- 10,000 or 1,000 nucleotides were selected at random from
tated E. coli genome contains 4,311 protein sequences, the A. fulgidus (RefSeq NC-000917), E. coli 0157H7
NR database contains 9,522 E. coli K-12 sequences (simply (NC-002655), and S. pyogenes (NC-004070). These se-
restricting the search to Escherichia coli would select 87,754 quences were also randomly mutated at 5% of sites, with re-
sequences). placements twice as likely as insertions or deletions. Using
A more sensitive strategy searches against the proteomes the FASTX program, both the correct and mutated se-
of individual genomes. The NCBI provides a very powerful quences were compared with subsets of the NR database
facility for searching individual genomes, and combinations that excluded the taxonomic divisions indicated. The frac-
of genomes, through their Genomic BLAST pages, avail- tion of the anonymous query sequences that had a statisti-
able from the NCBI Genome pages. Using http://www.ncbi cally significant alignment with an NR sequence, the me-
.nlm.nih.gov/sutils/genomtable.cgi,one can select taxo. dian length of the fraction of the query that aligned for
nomic sets of genomes, and include or exclude specific queries that matched the NR sequence, and the median
genomes (for example, when fully sequenced Actinobacteria number of NR sequences that matched the query (coverage
proteomes are selected, two strains of Mycobacterium tuber- depth) are shown.
culosis and Tropheryma whipplei are included). When The data in Table 6 illustrate the effectiveness of char-
Genomic BLAST is used to search the Actinobacteria, only acterizing anonymous DNA sequences by similarity search-
35,000 sequences, with 10 million residues, from 10 ing. If long (- 10,000-nucleotide) sequences are available
genomes are examined. Indeed, one can select a set of 70 with 5% errors, then 50 to 60% will align with sequences
bacterial genomes that represents every different genus only from different phyla in the NR database, with 20 statis-
once. This set of genomes includes 221,223 sequences (70 tically significant alignments per open reading frame.
million residues), about 40% of the number of sequences This depth of coverage-20 homologs per query-should
852 COMMUNITY A N D CENOMIC ANALYSIS

TABLE 6 Identification of anonymous DNA sequences at different evolutionary distances


Unknown DNA Excluded sequences Query length E() threshold Fraction found Length Coverage (X)
A. fulgidis Euryarchaeota 10,000 1.oo 0.785 632
1.oo 0.781 344
1,000 0.64 0.811 30
0.64 0.748 21
A. fulgidis, 5% mutated Euryarchaeota 10,000 1.00 0.657 260
1.oo 0.648 148
1,000 0.64 0.811 30
0.64 0.748 21
A. fulgidis Archaea 10,000 1.00 0.725 607
1.00 0.781 344
1,000 0.57 0.746 33
0.52 0.733 21
A . fulgidis, 5% mutated Archaea 10,000 1.oo 0.553 240
1.00 0.781 344
1,000 0.57 0.746 33
0.52 0.733 21
E. coli Proteobacteria 10,000 1.oo 0.727 336
1.oo 0.679 281
1,000 0.57 0.788 28
0.52 0.746 17
E. coli, 5% mutated Proteobacteria 10,000 1.oo 0.636 256
1.oo 0.619 171
1,000 0.57 0.788 28
0.52 0.746 17
E. coli Bacteria 10,000 1.oo 0.430 102
0.90 0.392 109
1,000 0.44 0.665 17
0.36 0.682 11
E. coli, 5% mutated Bacteria 10,000 0.90 0.375 92
0.70 0.396 61
1,000 0.44 0.665 17
0.36 0.682 11
S. pyogenes Firmicutes 10,000 1.oo 0.723 570
1.oo 0.695 377
S . pyogenes, 5% mutated Firmicutes 10,000 0.90 0.628 332
0.90 0.524 195
S. pyogenes Bacteria 10,000 1.oo 0.480 150
0.90 0.475 89
S . pyogenes, 5% mutated Bacteria 10,000 0.90 0.433 72
0.80 0.433 43

provide considerable phylogenetic detail about the likely error. And 30% of mutated 1,000-nucleotide sequences
origin of the sequence. When shorter (1,000-nucleotide) found a homolog, with about 20 significant alignments. The
reads were examined, 40 to 60% of the sequences pro- A. fdgidus case is perhaps the easiest; the S. pyogenes data
duced significant alignments to sequences from other phyla are probably more representative, but even in this case, if
or other superkingdoms. long sequences are available, many homologs are found.
As one might expect, the effectiveness of this strategy Naturally, over the next several years, the phylogenetic
depends on the origin of the anonymous DNA sample, sampling of microbial genomes will become much more in-
but the taxonomic exclusions used in Table 6 are very chal- formative. Today, one expects to be able to identify more
lenging. Because of the large number of diverse bacterial se- than half of long anonymous DNA sequences simply by
quences available, all of the 10,000-nucleotide archaeal translated-DNA-protein similarity searching. This number
sequences samples shared a statisticallysignificant match with will only increase as the microbial kingdoms are better
an NR sequence with an E() of <lo-, even with 5% sampled.
35. Genome Similarity Searching 853

35.7. PATHOLOGICAL CASES Removing low-complexity domains is especially impor-


tant when comparing translated-DNA sequences to protein
35.7.1. Low-Complexity Regions-SEC sequence databases with BLASTX or FASTX. Out-of-frame
This chapter has focused on exploiting our understanding translations of DNA sequences frequently produce low-
of local similarity statistics to develop more sensitive and complexity regions (12), which show up as statistically sig-
selective sequence comparison strategies. This strategy as- nificant matches to low-complexity protein domains.
sumes, however, that the similarity statistics are accurate: Unfortunately, the current web interface to the BLAST
that an alignment with an E() of will occur, by programs at the NCBI does not offer this option, though
chance, about once in 1,000 searches. While statistical es- one can search a SEGed database with a local BLAST in-
timates for pairwise alignment scores are generally accu- stallation.
rate, alignments that appear to be significant can occur
between nonhomologs when the statistical assumptions 35.7.2. Confirming Statistical Significance with
fail. The most common failure is caused by low-complexity Shuffled Comparisons
domains in proteins-regions with a highly biased amino The statistical estimates reported by the BLAST and
acid composition (19). Figure 1 shows an example of a FASTA programs report the number of times an alignment
protein with several low-complexity domains, as identi- score is expected by chance, from sequences with composi-
fied by the SEG program. These small domains of biased tions and lengths similar to those in the sequence database.
amino acid composition can produce unexpectedly high For sequences with average compositions, these estimates
similarity scores, not because of homology but because of are very accurate, but as discussed above, for proteins with
shared composition bias. The BLASTP and TBLASTN low-complexity domains having very biased composition,
programs automatically applies SEG filtering to protein statistical estimates may be off by many orders of magni-
query sequences, where it is extremely effective in ex- tude. However, there are intermediate cases as well; some
cluding significant alignments caused by low-complexity sequences may have only slightly biased regions (the most
domains. common examples are transmembrane proteins). These
BLAST and FASTA use slightly different strategies for proteins do not have average compositions, and their high
excluding low-complexity domains from alignment scores. alignment scores may reflect shared composition bias rather
By default, BLASTP and TBLASTN run the SEG (19) pro- than shared ancestry. (In my experience, nonhomologous
gram on all query sequences before the search begins. membrane proteins never share statistically significant se-
BLAST uses a SEG option that masks the low-complexity quence similarity, though often the high-scoring nonsignif-
regions by converting the residues to Xs. The FASTA pro- icant alignments include other membrane proteins.)
grams use a slightly different strategy. SEG can be run on a The PRSS and PRFX programs in the FASTA package
protein library using a mode (pseg -q -z 1) that converts can be used to confirm that statistical estimates do not sim-
low-complexity domains into lowercase letters, rather than ply reflect shared composition bias. PRSS is a derivative of
Xs. Then, FASTA or FASTX can be run with the -Sop- the SSEARCH (Smith-Waterman) program that compares
tion, and the lowercase residues in library sequences are two sequences to produce an optimal local alignment score
masked out for the initial score calculation but are included and then shuffles the second sequence 200 to 1,000 or more
in the final alignment so that homologous low-complexity times, calculating Smith-Waterman alignment scores be-
regions are displayed. (The same strategy can be used to tween the first sequence and each shuffled sequence. The
lowercase low-complexity regions in query sequences with 200 to 1,000 shuffled sequence scores are used to estimate a
TFASTX.) Figure 2 shows similarity scores obtained with X and K (equation 1) that are specific for the length and
the NR database when low-complexity domains are ex- composition of the sequence pair of interest. In addition, the
cluded as well as when they are included. shuffling method can be modified to increase the stringency

>gi1219095501reflNP663818.11 putative secreted protein


1-46 MKKRILSAVLVSGVTLGAATTVGAEDLSTK
IAKQDSIISNLTTEQK
aaqnqvsalqaqvsslqs 47-64 EQDKLTARNTELEALSKRFEQEIKALTSQI
VARNEKLKNQARSAYKNNETSGYINALLNS
KSISDVVNRLVAINRAVSANAKLLEQQKAD
KVSLEEKQ
aanqtaintiaanmamaeenqn 163-184
185-187 TLR
tqqanleaatanlalqlasat 188-208
209-213 EDKAN
lvaqkeaaekaaaealaqeqaakvkaqeqa 214-288
aqqaasveaaksaitpapqatpaaqssnai
epaaltapaapsarp
289-310 QTSYDSSNTYPVGQCTWGAKSL
apwagnnwgnggqwa 311-325
326-354 YSAQAAGYRTGSTPMVGAIAVWNDGGYGH
vavvvevqsass 355-366
367-398 IRVMESNYSGRQYIADHRGWFNPTGVTFIY
PH

FIGURE 1 Low-complexity regions identified in a putative secreted protein from S. pyogenes


(NP-663818) by the SEG program (19). Low-complexity regions are shown to the left of the
residue numbers.
854 COMMUNITY AND GENOMIC ANALYSIS

1 0 0 0 0 0 1000 ~ timate A and K for the translated sequence comparison. As


with PRSS, either a uniform or segmental shuffle can be
used, but a uniform shuffle is usually sufficient to identify
750
significant scores that reflect shared composition bias,
rather than homology. FASTX statistical estimates for
alignments that exclude low-complexity regions (SEGed
proteins) are generally very accurate; 5 to 10 per thousand will
500
have significance estimates that drop lo3- to 106-foldwhen
PRFX shuffles are done. For large-scale translated-DNA se-
quence comparison, PRFX analysis should be used routinely
250 to confirm biologically unexpected observations.
Statistical estimates from shuffled protein sequences are
very reliable because, with the exception of composition
0 effects, shuffled protein sequences appear to be very similar
to unrelated protein sequences. This is not true for DNA se-
quences; DNA sequences have higher-order dependencies
(e.g., codon biases) that are usually disrupted by sequence
20 40 60 80 100 120 shuffling, so that 1,000 shuffled DNA sequences may have
Similarity Score alignment properties that are very different from 1,000 real,
unrelated sequences. Thus, in general, the DNA-DNA sta-
FIGURE 2 Low-complexity domains increase similarity tistical significance reported by FASTA are probably more
scores. Distribution of scores obtained searching the NR data- accurate than those produced by PRSS.
base excluding Firmicutes sequences with a putative secreted
protein from S. pyogenes (NP-663818), a 398-amino-acid pro-
tein that is about 30% low complexity. The filled symbols show 35.8.SUMMARY
the distribution of scores obtained when low-complexity BLAST and FASTA similarity searches against comprehen-
regions are excluded from the alignment scores; the open dia- sive sequence databases are among the most sensitive and
monds show the scores obtained when low-complexity do- reliable tools available for characterizing bacterial genes
mains are included. The inset (upper right) shows that about and genomes. Similarity searches are most effective when
1,000 additional sequences have high scores if low-complexity used to identify homologous genes; with the exceptions
domains are included; three of these sequences have an E() of noted above, protein and translated-DNA sequences that
< and six more have an E() of <0.01. There are no sta- share statistically significant similarity are homologous,
tistically significant matches when low-complexity domains with similar three-dimensional structures.
are excluded. When characterizing bacterial genomes, a few strategies
can improve dramatically the effectiveness of similarity
searches. (i) Whenever possible, compare protein sequences
or perform translated-DNA comparisons; DNA-DNA com-
of the analysis: the second sequence can be shuffled uni- parisons are 5- to 10-fold less sensitive than protein com-
formly, so that each residue from the unshuffled sequence is parisons. (ii) Search the smallest comprehensive database
placed anywhere in the shuffled copy; or the second se- that is likely to contain the sequences of interest. If ho-
quence can be shuffled in segments (window shuffle), so that mologs are not found in the smaller database, a larger data-
residues 1 to 20 of the unshuffled sequence are found only in base can be searched, but searches against comprehensive
positions 1 to 20 of the shuffled sequences, residues 21 to 40 databases like the NR database are 100- to 1,000-fold less
remain in positions 21 to 40, etc. The segmental or window sensitive than searching bacterial genomes. (iii) Focus on
shuffling preserves not only the global but also the local the statistical estimates when inferring homology; infer-
composition in the shuffled sequences, so that transmem- ences based on percent identity and alignment length are
brane domains in the unshuffled sequence will be placed in either much less sensitive or much less reliable. (iv)
the same position, but in shuffled order, in the shuffled se- Sequence alignments with expectation values between
quences. lop3 and lop6 are almost always homologous, except for
For proteins with average amino acid composition, low-complexity domains or out-of-frame translations. Low-
there is rarely more than a twofold difference between sta- complexity domains can usually be removed with the SEG
tistical estimates reported from similarity search and esti- program, and statistical estimates can be confirmed with
mates based on PRSS shuffles. For proteins containing PRSS and PRFX.
multiple transmembrane domains (e.g., G protein-coupled The inference of homology from statistically significant
receptors or ion channels), the uniform shuffle estimates similarity is a powerful strategy for characterizing sequences.
differ only slightly from the similarity search statistical es- Current versions of the BLAST and FASTA comparison
timates, but segmental shuffle estimates may differ 10- to programs produce very reliable statistics. But homology re-
100-fold (the difference is generally greater for more signif- flects the evolutionary history of a sequence, preserved
icant scores). through constraints on structure. Today, the most common
While protein-protein statistical estimates are generally errors attributed to similarity searches and homology infer-
very accurate, translated-DNA-protein estimates can be ence involve assertions about biological function. Never-
more misleading because of the unusual properties of out- theless, searching for homologs remains the most informa-
of-frame alignments. The PRFX program performs a tive analysis available for sequences and promises to remain
FASTX translated-DNA-protein alignment and then shuf- so as more genomes are sequenced and sequence databases
fles the protein sequence 200 to 1,000 or more times to es- expand.
35. Genome Similarity Searching 855

35.9. REFERENCES 11. Pearson, W. R. 1999. Flexible similarity searching with


1. Altschul, S. F. 1991. Amino acid substitution matrices the fasta3 program package, p. 185-219. In S. Misener and
from an information theoretic perspective. J. Mol. Biol. S. A. Krawetz (ed.), Bioinfomatics Methods and Protocols.
219555-565. Humana Press, Totowa, NJ.
2. Altschul, S. F., M. S. Boguski, W. Gish, and J. C. 12. Pearson, W. R., T. Wood, Z. Zhang, and W. Miller. 1997.
Wootton. 1994. Issues in searching molecular sequence Comparison of DNA sequences with protein sequences.
databases. Nut. Genet. 6:119-129. Genomics 46: 24-36.
3. Altschul, S. F., W. Gish,W. Miller, E.W. Myers, and 13. Pearson, W. R., and T. C. Wood. 2001. Statistical signif.
D. J. Lipman. 1990. Basic local alignment search tool. J. icance in biological sequence comparison, p. 39-65.
Mol. Biol. 215:403-410. In D. J. Balding, M. Bishop, and C. Cannings (ed.), Hand-
4. Altschul, S. F., T. L. Madden, A. A. Schaffer, J. Zhang, book of Statistical Genetics. Wiley, London, United King-
Z. Zhang, W. Miller, and D. J. Lipman. 1997. Gapped dom.
BLAST and PSI-BLAST a new generation of protein data- 14. Reeck, G. R., C. de Haen, D. C. Teller, R. F. Doolittle,
base search programs. Nucleic Acids Res. 25:3389-3402. W. M. Fitch, R. E. Dickerson, P. Chambon, A. D.
5. Boeckmann, B., A. Bairoch, R. Apweiler, M. Blatter, A. McLachlan, E. Margoliash, T. H. Jukes, and E.
Estreicher, E. Gasteiger, M. J. Martin, K. Michoud, C. Zuckerland. 1987. Homology in proteins and nucleic
ODonovan, I. Phan, S. Pilbout, and M. Schneider. acids: a terminology muddle and a way out of it. Cell 50:667.
2003. The Swiss-Prot protein knowledgebase and its sup- 15. Smith, T. F., and M. S. Waterman. 1981. Identification of
plement trembl in 2003. Nucleic Acids Res. 31:365-370. common molecular subsequences.1. Mol. Biol. 147: 195-
6. Brenner, S. E., C. Chothia, and T. J.Hubbard. 1998. 197.
Assessing sequence comparison methods with reliable 16. Sonnhammer, E. L., S. R. Eddy, and R. Durbin. 1997.
structurally identified distant evolutionary relationships. Pfam: a comprehensive database of protein domain fami-
Proc. Natl. Acud. Sci. USA 95:6073-6078. lies based on seed alignments. Proteins 28:405-420.
7. Dayhoff, M., R. M. Schwartz, andB. C. Orcutt. 1978. A 17. States, D. J., W. Gish, and S. F. Altschul. 1991. Im-
model of evolutionary change in proteins, p. 345-352. In proved sensitivity of nucleic acid database searches using
M. Dayhoff (ed.), Atlas of Protein Sequence and Structure, application-specificscoring matrices. Methods Companion
vol. 5, suppl. 3. National Biomedical Research Founda- Methods Enzymol. 3:66-70.
tion, Silver Spring, MD. 18. Tatusov, R. L., E. V. Koonin, and D. J. Lipman. 1991. A
8. Henikoff, S., and J.G. Henikoff. 1992. Amino acid sub- genomic perspective on protein families. Science 278:63 1-
stitutions matrices from protein blocks. Proc. Natl. Acud. 637.
Sci. USA 89:10915-10919. 19. Wootton, J. C., and S. Federhen. 1993. Statistics of local
9. Jones, D. T., W. R. Taylor, and J. M. Thornton. 1992. complexity in amino acid sequences and sequence data-
The rapid generation of mutation data matrices from pro- bases. Comput. Chem. 17:149-163.
tein sequences. Comput. Appl. Biosci. 8:275-282. 20. Zmasek, C . M., and S. R. Eddy. 2002. Rio: analyzing pro-
10. Pearson, W. R. 1998. Empirical statistical estimates for se- teomes by automated phylogenomics using resampled in-
quence similarity searches. J. Mol. Biol. 276:7 1-84. ference of orthologs. BMC Bioinfomatics 3: 14.
36
Reconstructing and Interpreting Evolutionary
Relationships
CHRISTOPHE J . DOUADY AND CAMILLA L. NESBIZ)

36.1. INTRODUCTION ......................................... 856


.2. CONSTRUCTION OF PHYLOGENETIC TREES ................. 857
36.2.1. Alignment .......................................... 857
36.2.1.1. Automated Alignment .......................... 857
.2. Alignment Editing ............................. 857
.3. Codon Alignment ............................. 857
36.2.2. Tree Building Options ................................. 858
36.2.2.1. What Type of Tree Can Be Expected from the Data? .... 858
.2. Tree Search: Exhaustive versus Heuristic Searches ..... 859
.3. Type of Data ................................. 859
.4. Tree Selection: Optimality Criterion................ 860
.5. Models of Evolution ............................ 861
36.2.3. How To Decide? ..................................... 861
36.2.3.1. Need for Compromise .......................... 861
.2. Trpe of Search ............................... 861
.3. Type of Data ................................. 861
.4. Type of Optimality Criterion ..................... 862
.5. Model of Evolution ............................ 862
36.2.4. Clustering Algorithms: a Shortcut ......................... 862
36.3. INTERPRETATION OF PHYLOGENETIC TREES ................ 862
36.3.1. Reading of the Tree ................................... 862
36.3.1.1. Rooting and Polarizing the Tree ................... 862
.2. Phylogenetic Terminology ....................... 862
36.3.2. Robustness ......................................... 864
36.3.2.1. Initial Matrix without Resampling .................. 864
.2. Resampling .................................. 864
36.4. SUMMARY AND CONCLUSION: W H Y DO WE CONSTRUCT
PHYLOGENETIC TREES? .................................. 864
.5. SOMEDEFINITIONS ...................................... 865
.6. FREQUENTLY ASKED QUESTIONS .......................... 865
.7. REFERENCES ............................................ 866
36.7.1. Further Reading ...................................... 866
.2. General References ................................... 867

36.1. INTRODUCTION should be able to use this chapter as a guide to the options
Phylogenetics is the study of evolutionary relationships available for different types of data as well as to software
among organisms or genes. and phylogenetic analyses aim at available for doing the analysis. Such software is becoming
estimating or reconstructing the evolutionary relationships increasingly user friendly. Our goal. then. is to provide some
among such operational taxonomic units (OTUs). These background and understanding of what methods may be
relationships are usually visualized as a phylogenetic tree. useful for what purpose .
which portrays the evolutionary history of the OTUs. In All phylogenetic tree-building methods inherently rely
this chapter the OTUs of interest are represented either by on a common set of assumptions. For example. all analyses
DNA or amino acid sequences. We will give a general in- assume that the sequences to be analyzed are homologs. that
troduction to methods for reconstructing phylogenetic trees they are not a mixture of different sets of paralogs (see sec-
from a set of homologous DNA or amino acid sequences tion 36.5), and that gene flow between taxa is nonexistent
(see section 36.5), obtained for example by doing database or extremely low (see below). There are four steps in the
searches. Given space limitations. it is impossible to guide phylogenetic analysis of molecular sequence data: (i) build-
inexperienced users across all or even most of the steps of ing an alignment. (ii) determining an appropriate substitu-
tree reconstruction. Additionally. this task has already been tion model. (iii) tree building. and (iv) tree evaluation. It is
accomplished by several books and book chapters (28. 47. important to remember that all subsequent steps in the
52). A reader with little or no prior experience. however. analysis are highly dependent on the assumption that the

856
36. Interpreting Evolutionary Relationships 857

initial alignment is correct, i.e., that each position in the documentation [63, 641). ClustalX can accept raw data in
alignment is composed of homologous residues (positional FASTA format and other formats (Fig. 2) and lets the oper-
homology; see section 36.5). Even the most sophisticated ator decide the penalty that should accompany the opening
phylogenetic analysis software cannot correct for bad input and extension of a new gap, compared with creating a mis-
data. Similarly, obtaining the best model for the data is also match. ClustalX starts by generating painvise alignments
important, and a phylogenetic analysis should be viewed as between all the sequences to create a dendrogram that is
a search for the optimal evolutionary model as well as for used during the final, multiple alignment step. It should be
the optimal tree. noted that the dendrogram produced during the second
phase is treelike but is not a valid phylogenetic tree.
Furthermore, no objective reason exists for choosing one set
36.2. CONSTRUCTION OF PHYLOGENETIC of penalties over another, and often many should be tried
TREES and the alignment inspected manually to determine the
most suitable set for the data. This step is crucial for all sub-
36.2.1. Alignment sequent inference. More practical information can be found
The primary input required by phylogenetic software is an in reference 28 (p. 21-37).
alignment. A n alignment is a matrix (usually an electronic For rRNA analyses, alignments based on structural data
file) where each sequence of interest occupies a line and can be downloaded from several databases, such as the
each homologous site or position in these sequences occu- European Ribosomal RNA Database (66, 67) (http://www
pies the same column. Selecting the relevant sequences to .psb.ugent.be/rRNA/), the Ribosomal Database Project I1
be studied is an important process, and pertinent informa- (RDP 11) ( 7 ) (http://rdp.cme.msu.edu), and the Compar-
tion can be found in recent literature (see chapter 35, this ative RNA Web Site (5) (http://www.rna.ccbb.utexas
volume, and reference 28, p. 8-20). The next step consists .edu/). Sequences can be added to these alignments online,
of generating columns of homologous sites and would be manually (see below) or in ClustalX, and should be in-
trivial if not for insertion and deletion mutations (indels). spected and edited by eye (see below). These databases
These indels can be compensated for in the alignment by also offer other services such as BLAST analyses, primer
introducing gaps (Fig. la, b). However, as shown in Fig. 1, construction, or distance-based phylogenetic analyses
multiple options for introducing gaps usually exist. Hence, using clustering algorithms such as neighbor joining (see
generating an optimal alignment can be summarized as re- section 36.2.4). For the online phylogenetic analyses at
ducing the number of mismatches (putatively homologous RDPII, it should be noted that the user is less free to ma-
positions that do not share the same base or nucleotide) and nipulate the different parameter settings than in, for in-
adding as few (in terms of both number and size) gaps as stance, PAUP*, and trees should be built using other phy-
possible. logenetic software as well. The trees are useful as a starting
There are several different alignment file formats; point, however. The Quick phylogeny search at the
FASTA, NEXUS, and PHYLIP formats are the most com- European Ribosomal database, which implements the BIB1
monly used. The simplest format is the FASTA format, program (http://pbil.univ-lyonl .fr/bibi/), is based on Clustal
shown in Fig. 2. Information on alignment format options alignments (not structural alignments), and as stated on the
for different phylogenetic analysis programs is available in web page, the tool should be used with care. However, the
their respective user manuals. tree produced is a true neighbor-joining (NJ) tree (even if
Clustal produced it) and not the dendrogram produced dur-
36.2.1 . I . Automated Alignment ing the alignment process. Alignments can also be built
When only a few relatively similar sequences are investi- using ARB (http://www2.mikro.biologie.tu-muenchen.de/arb/
gated, visual inspection in a text editor or sequence editor about.htm1). This program package also offers phylogenetic
such as BIOEDIT, Se-A1, or MacClade (see Table 1) gener- analyses and probe design. The trees are useful as a starting
ally allows the unambiguous introduction of gaps. When point.
the number and divergence of the sequences increases, the
operation can become extremely tedious. Phylogeneticists 3 6.2.1.2. Alignment
- Editing
-
usually use software such as ClustalX, which applies a Automatic alignments in general require subsequent human
heuristic algorithm to generate an alignment (for more in- editing. Most common actions include (i) correction of ob-
formation see p. 86-98 in reference 27 and the ClustalX vious alignment errors, (ii) edge trimming of the alignment
(it is unlikely that all the sequences in the file will start and
end at the same position), and (iii) removal of inherently
ambiguous sections. Ambiguous portions occur when se-
A Sequencel A C G C T A C T T G A C T A G quences share zones of weak similarity and much length
Sequence2 A C G C A C A T G variation, forcing an arbitrary choice between equally likely
alignments (i.e., the gap could be introduced at several
places within a small range of sites). Recently developed
B Sequencel A C G C T A C T T G A C T A G
software can help perform this final editing (e.g., G blocks
Sequence2 A C G C - A C A T G [6]; SOAP [42]), but even if such software represent
progress toward objectivity, caution should prevail and a
C Sequencel A C G C T A C T T G A C T A G final check by eye is advisable.
Sequence2 A C G C - A C - - - A - T - G
36.2.1.3. Codon Alignment
FIGURE 1 Three possible alignments between the same two When aligning protein-coding DNA sequences, gaps
sequences, showing the compromises between the number of should not only be introduced to decrease the number of
gaps and the number of mismatches. Positions in bold indicate mismatches, but they should also try to respect the triplet
matching sites. structure of the code (i.e., usually gaps are introduced by
858 H COMMUNITY AND CENOMIC ANALYSIS

>gi164682941emblAJ251082.llHMA251082 Haloarcula marismortui 5s rRNA


gene, rrnB operon
TCATTCATACGCACTGTGACTCATTCACCGACGATTTAACTCGTCGCTGAACGAGTCCAGGCGCAAACTG
GATCGCACGTAATCACACGGTGGAAGAGTTAATCGAGTTAATCGAGACTGGTACTATCGCGGTTCGATTCCGTGACTCG
ACGTTAGGCGGCCACAGCGGTGGGGTTGCCTCCCGTACCCATCCCGAACACGGAAGATAAGCCCACCAGC
GTTCCGGGGAGTACTGGAGTGCGCGAGCCTCTGGGAAACGCGGTTCGCCGCCACCATTCATACCTTTCAT
AGCCCACTCAGGAGAGATATCTCTCCCGAGTGGGCTTTCCGTATTTAAACAGAGCCGAACCACTCAGTAA
ATGACCGGTTCTCGCACTCTGTGGAATACGGCTTCAATCGGCTTCAATCGGTGAGATCAGACGTGCGACTAGCGATCGTG
ATCGAGTCGTTGA
>gi14391981gblL27163.l~HACRRAHaloarcula vallismortis 5s ribosomal RNA
GTAGCGGCCACAGCGGTGGGGTTCCTCCCGTACCCATCCCGAACACGGAAGATAAGCCCACCAGCGTTCC
GGGGAGTACTGGAGTGCGCGACCCTCTGGGAAACCGGGTTCGCCGCTACYY
>gi1435541emblX03407.llHHRRNA Halobacterium halobium rRNA operon with
genes for 16s rRNA, 235 rRNA, 5s rRNA and tRNA-Ala (UGC)
GGTACCACTCGGCCCGACCGAACGCACTCGCGCGGGATGACCGGCCGACCTCCGCCTACGCAATACGCTG
TGGCGTGTGTCCCTGGTGTGGGCCGCCATCACGAAGCGCGCTGCTGGTTCGACGGTGTTTTATGTACCCCAC
CACTCGGATGAGATGCGAACGACGTGAGGTGGCTCGGTGCACCCGACGCCACTGATTGACGCCCCCTCGT
CCCGTTCGGACGGAACCCGACTGGGTTCAGTCCGATGCCCTTAAGTACAACAGGGTACTTCGGTGGAATG
CGAACGACAATGGGGCCGCCCGGTTACACGGGTGGCCGACGCATGACTCCGCTGATCGGTTCGGCGTTCG
GCCGAACTCGATTCGATGCCCTTAAGTAATAACGGGTGTTCCGATGAGATGCGAACGACAATGAGGCTAT
CCGGTTCGTCCGGGTGGCTGATGCATCTCTTCGACGCTCTCCATGGTGTCGGTCTCACTCTCAGTGAGTG
TGATTCGATGCCCTTAAGTAATAACGGGCGTTACGAGGAATTGCGAACGACAATGTGGCTACCTGGTTCT
>Hb halobi
GGTACCACTCGGCCCGACCGAACGCACTCGCGCGGGATGACCGGCCGACCTCCGCCTACGCAATACGCTG
TGGCGTGTGTCCCTGGTGTGGGCCGCCATCACGAAGCGCTGCTGGTTCGACGGTGTTTTATGTACCCCAC
CACTCGGATGAGATGCGAACGACGTGAGGTGGCTCGGTGCACCCGACGCCACTGATTGACGCCCCCTCGT
CCCGTTCGGACGGAACCCGACTGGGTTCAGTCCGATGCCCTTAAGTACAACAGGGTACTTCGGTGGAATG
CGAACGACAATGGGGCCGCCCGGTTACACGGGTGGCCGACGCATGACTCCGCTGATCGGTTCGGCGTTCG
GCCGAACTCGATTCGATGCCCTTAAGTAATAACGGGTGTTCCGATGAGATGCGAACGACAATGAGGCTAT
CCGGTTCGTCCGGGTGGCTGATGCATCTCTTCGACGCTCTCCATGGTGTCGGTCTCACTCTCAGTGAGTG
TGATTCGATGCCCTTAAGTAATAACGGGCGGGCGTTACGAGGAATTGCG~CGACAATGTGGCTACCTGGTTCT
FIGURE 2 Sequences in FASTA format.Each sequence is composed of two parts.The first part
includes the first line only,which always starts with a greater thansign (>) and is a more or less
complex description of the sequence.To avoid any downstream problems,we recommend using a
nine-characterdescriptor with spaces represented by an underscore sign (e.g.,the last sequence).
The second part is the actual string of nucleotides or amino acids.By default this part will start after
the descriptorparagraph mark and will end just before the next greater thansign.

multiples of three between two codons). This task can be Hence,to be sure that the optimal tree is recovered for 10
performed relatively easily by using CodonAlign (see com- taxa,more than 2 million trees have to be inspected,and
ment in reference 28 p. 146-149)or ARB.These softwares for 20 taxa more than 2 X 10 trees will have to he in-
use a protein alignment as a guide to generate an alignment spected.
of the corresponding DNA sequence.
36.2.2.1. What Type of Tree Can Be Expected from
36.2.2. Tree Building Options the Data?
W h e n starting a phylogenetic analysis, numerous choices The first question one has to answer is not strictly phyloge-
have to he made. Ofthese choices,none is straightforward netic but one that regards the amount of gene flow between
and many still divide the phylogenetic community. the OTUs present in the data set.The tree reconstruction
Therefore,the following section should he read critically methods usually used in phylogenetic analyses assume that
and with the understanding that the perfect analysis, if the data are dichotomous (i.e., that the gene flow has
such is possible,has yet to he developed.The users of phy- stopped). In the remaining part of this chapter, we will
logenetic analysis programs should remember that com- focus on this type of reconstruction. However,if the data
promises are often made among at least three parameters: have been sampled from a population,or if one can expect
(i) power (the amount of data that will produce a result very high levels of lateral gene transfer (LGT) and /or re-
similar to an infinite data set), (ii) consistency (assurance combination, the gene flow should be considered in the
that the correct answer will he found given that enough analysis,and readerscan turn themselves toward population
data are provided), and (iii) efficiency (the amount of genetic methods (if the flow is likely to he very high) or to-
time used to deliver a tree). Efficiency might seem to he an ward network analysis. Network analyses do not assume
irrelevant parameter to enter into this equation, hut at that the data are dichotomous and therefore allow for
first sight only. T h e number of solutions (tree topologies) a taxon to he connected with more than another.Programs
grows factorially with the number of OTUs considered. that can be used to do such analyses include TCS
36. Interpreting Evolutionary Relationships 859

TABLE 1 Useful softwarea


Software Where to get it Platform Price
ARB http://www2.mikro.bio1ogie.t~-muenchen.de/arb/documentation.html Mac, Unix, Linux Free
Bioedit http://www.mbio.ncsu.edu/BioEdit/bioedit.html PC Free
Clustal X http://bips.u-strasbg.fr/fr/Documentation/clustalX PC, Mac, Unix, Linux Free
CodonAlign http://homepage.mac.com/barryghall/CodonAlign.html PC, Mac Free
Consel p/-shimo/prog/consel/
http://www.is.titech.ac.j PC, Unix, Linux Free
CoVARES http://hades.biochem.dal.ca/Rogerlab/Software/software.html Unix Free
Gblocks http://molevol.ibmb.csic.es/Gblocks.html PC, Mac, Unix, Linux Free
GDE ftp://ftp.bio.indiana.edu/molbio/unix/GDE Mac, Unix, Linux, Free
MacClade http://macclade.org/ Mac $125
Mega http://www.megasoftware.net/ PC Free
Modeltest http://darwin.uvigo.es/software/modeltest.html PC, Mac, Unix Free
Molphy http://www.ism.ac.jp/software/ismlib/softother.e.html~olphy Unix Free
MrBayes http://mrbayes.csit.fsu.edu/ PC, Mac, Unix Free
Must herve.philippe@umontreal.ca PC, Linux Free
PAML http://abacus.gene.ucl.ac.uk/software/paml.html PC, Mac, Unix, Linux Free
PAUP* http://paup.csit.fsu.edu/ PC, Mac, Unix, Linux <$150
PHYLIP http://evolution.genetics.washington.edu/phylip.html PC, Mac, Unix, Linux Free
Se-A1 http://evolve.zoo.ox.ac.uk/software.html!id=seal Mac Free
SOAP http://ueg.ulb.ac.be/SOAP/ PC, Mac, Unix, Linux Free
Tree-puzzle http://www.tree-puzzle.de/ PC, Mac, Unix Free
Treeview http://taxonomy.zoology.gla.ac.uk/rod/treeview.html PC, Mac, Unix, Linux Free
aThis list is far from being exhaustive, but presents a few of the most widely used software packages. A more complete list can be found at Joe Felsensteins web page
(http://evolution.genetics.washington.edu/phylip/software.html).

(http://darwin.uvigo.es/software/tcs.html)and Splitstree procedures can be found in reference 61, but more ade-
(http://www.splitstree.org). quately an NJ (neighbor-joining) tree can be used (see
below; 5 1). The search will move using topological pertur-
36.2.2.2. Tree Search: Exhaustive versus Heuristic bation. These rearrangements can affect any part of the tree
Searches and operate in various processes presented as choices to the
As a direct consequence of the size problem (with 20 taxa, user of PAUP or PHYLIP, for instance: nearest-neighbor in-
more than 2 X 10 trees will have to be inspected; see terchanges (1, local rearrangements in PHYLIP), sub-
above), users will have to choose a way to search the tree tree pruning and regrafting (SPR, global rearrangements in
space, comprising all possible topologies (the haystack) in PHYLIP), and tree bisection and reconnection (TBR).
which the optimal topology (the needle) is found. There are Unless time becomes a limitation, the operator should favor
two approaches: (i) picking and inspecting each piece of TBR to SPR and SPR to NNI, as NNI represent only a sub-
straw, one after another (exhaustive search), and (ii) set of the SPR results that are themselves only a subset of
using a magnet to find the needle (heuristic searches). the TBR rearrangements (60, 61). It is important to recall
Unfortunately, phylogeneticists cannot rely on precise that these searches are imperfect. Thus, even if one is sure
heuristic searches to solve their problem, as the needle of having found a high topology, one cannot be sure of hav-
has many of the same properties as all the straws. In fact, ing found the highest one(s), a notion known as local op-
this problem belongs to a class of mathematical problem, timum entrapment. The relative height of topologies will
called NP-complete, for which no efficient algorithms are be discussed in section 36.2.2.4.
known that provide a solution (26). Exhaustive searches are Heuristic searches are not the only alternative to ex-
usually not an option, and thus imperfect heuristic searches haustive searches. Hendy and Penny (29) applied a
have been developed. branch-and-bound method to phylogenetic problems. In
The most typical heuristics are generally called hill simple terms, this method offers a unique way to find the
climbing. They resemble the process of a hiker lost in the best (highest) tree without actually evaluating each tree
fog who is seeking the highest peak in a landscape. Like a (29, 61). However, for large data sets the time required for
hiker who moves forward, avoiding downhill and choosing this type of search may still force one to choose a heuristic
uphill paths, hill-climbing heuristics start from any given approach.
tree and move toward a different and higher topology (in
a tree landscape, each location represents a tree). The anal- 36.2.2.3. Type of Data
ogy is quite easy to visualize, but there is still a need to de- Another crucial choice that the user must make is the form
fine what the heuristic starting point will be, how it will of the data to be analyzed. One must first choose between
move, and what defines a higher topology. The starting DNA and amino acid sequences. It is often argued that to
point of a search can be chosen at random by using stepwise avoid saturation, slowly evolving characters are typically fa-
addition, or star decomposition. A description of these vored to investigate deep phylogenies and faster-evolving
860 H COMMUNITY AND GENOMIC ANALYSIS

characters are used at more shallow levels. This translates known as goodness-of-fit measures, which includes cri-
into using amino acid and nucleotide alignments for deep teria such as Farris's f statistic, least-squares, or Fitch-
and shallow levels, respectively. The second set of choices Margoliash methods, while the second is called minimum
having to do with type of data concerns the utilization of evolution.
raw data or its reduction into a distance matrix. Again this Maximum parsimony is a site-based (raw data) method
choice is far from trivial, since it might greatly affect the that searches for the tree that requires the fewest changes or
outcome and efficiency of the analyses. The use of dis- mutations (i.e., that is the most parsimonious solution). In
tance matrices results in information loss, since the se- its simplest form, changes of all types and at all sites are re-
quence data are reduced to a single distance measure for garded as equally parsimonious (equally weighted parsi-
each pairwise comparison, and will prevent using parsi- mony). However, different positions or types of changes can
mony or maximum-likelihood methods. Therefore this be considered as having different cost (generalized parsi-
conversion is not advisable per se. However, the gain in ef- mony; see, e.g., reference 8).
ficiency from distance matrices can allow analyses of data The maximum-likelihood method is another approach
that would not otherwise be feasible, or permit more real- that utilizes raw data. In this case, the criterion to be opti-
istic assumptions when selecting the optimal (highest) mized is the likelihood of observing the data given the con-
topology. sidered topology and a model of evolution (see below).
Finally, a newcomer in phylogeny is the Bayesian category
36.2.2.4. Tree Selection: Optimality Criterion of methods (33,50).These approaches seek the most prob-
In section 36.2.2.2, we stated that heuristic searches help able tree given the data. Without entering into any mathe-
select higher topologies, but what defines a higher or matical considerations, the main difference between maxi-
more optimal topology? This tree selection process relies mum likelihood and Bayesian approaches is the way they
on the concept of optimality criteria. In choosing a crite- deal with the parameters of the model of evolution.
rion, the user chooses what should be optimized. For dis- Maximum likelihood will seek parameter values that maxi-
tance data, the optimality criterion can seek the tree that mize the likelihood function, whereas a Bayesian inference
minimizes the difference between the observed distance approach integrates over all possible values of those param-
(the distance matrix) and the tree distance for each pair eters (see MrBayes documentation).
of taxa. Another option would be to look for a tree that is In Table 2 we have listed some of the most commonly
the shortest (minimizes the sum of tree distances). The used phylogenetic computer software and the types of
first criterion belongs to a family of optimality criteria analyses that are available in each of the packages.

TABLE 2 Function implemented in some of the most common phylogenetic softwarea


Tree building
Alignment Maximum Model Statistical Tree
Software
Building Editing Distance Parsimony likelihood Bayesian selection test viewing
nt aa nt aa nt aa nt aa
ARB (4) (J)
Bioedit J* J* J*
Clustal X J
CodonAlign
Consel J
Gblocks J
GDE J J J* J* J* J* J* (J") J*
I
MacClade (J) d
Mega J J J J J J
Modeltest J
Molphy J J
MrBayes J J
Must J J J J
PAML J J J
PAUP* J J (J) J
PHYLIP J J J
Se-A1 (J) J
SOAP J J
Tree-puzzle J J (J)
Treeview J
'Absence of a check mark does not mean that the given software cannot perform the task, but that we do not find it advisable to carry out the task. When between
parentheses, a check mark indicates a weak possibility. *, via other software. nt, nucleotides; aa, amino acids.
36. Interpreting Evolutionary Relationships w 861

36.2.2.5. Models of Evolution ences 39 and 41). To correct for this bias, covarion or het-
erotachous models (i.e., that do not assume stationarity)
36.2.5.1. Models of Substitution and State have been proposed (e.g., references 19, 21,39, 40, and 48;
Frequency also see section 36.4). However, they are seldom used.
Models of substitution are ways to define the probability Other standard simplifications include the assumption of
of a change or mutation. They can be seen as the distance, reversibility (the rate of change from A to B equals that
likelihood, and Bayesian equivalent of a parsimony-weight- from B to A), Markovian mutation (no memory is kept of
ing scheme. For nucleic acid sequences, by allowing six in- the former states), and the independence of the sites
dependent rates of change for the six types of nucleotide (change in site X does not affect changes at any other posi-
substitutions without assuming that bases are present in tion). These simplifications are clearly n o more sound than
equal frequency, the General Time Reversible (GTR) assuming a stationary model, but they are often necessary
model (38) is the most complex stationary (see section compromises, since adding a parameter to a model is not
36.2.2.5.3) model commonly implemented. Other classical without cost (see section 36.2.3.5).
models such as Tamura-Nei, HKY85, Kimura-2-Parameter,
or Jukes-and-Cantor are simpler versions of this general 36.2.3. How To Decide?
model (see Fig. 11 in reference 61). All these models are
available in most of the commonly used software for esti- 36.2.3.1. Need for Compromise
mating DNA phylogenies (PAUP, Table 2). In the previous sections we saw that there are at least four
Most models for protein analysis are based on a compila- choices that have to be made before estimating a phyloge-
tion of empirical data and estimation of probability of amino netic tree (type of search, data, optimality criterion, and
acid change. Dayhoff and JIT are the two most classical model of evolution). When considered individually, some
models and were both inferred from nuclear encoded proteins of these choices seem rather trivial. For example, exhaus-
(10, 36). The BLOSUM 62 is also a nuclear matrix but was tive searches are superior to the branch-and-bound method
deduced from very divergent proteins (30). The mtREV24 in describing the complete universe of possible trees, and
was estimated, as its name indicates, from mitochondria1 data both of these are superior to heuristic searches in always
(1). Simpler but analytical models include Poisson and pro- finding the best tree (global optimum). However, all
portional model (respectively, Jukes-and-Cantor and F81 choices have an influence on the time required to complete
counterpart for protein). The commonly used software for the tree search, often forcing the investigator to make diffi-
building protein phylogenies, PHYLIP and TREE-PUZZLE, cult decisions. Depending on the size of the data set, an ex-
together cover most of these models (Table 2). haustive search may only be performed for overly simplistic
models of evolution, while an imperfect heuristic search
36.2.2.5.2. Across-Site Rate Variation would allow usage of more realistic models. Unfortunately,
Models of evolution leave the user free to determine the no objective reason for favoring one set of choices over an-
probabilities of various types of change in the data. other exists. Often analysis methods are chosen because
However, it is very unlikely that any rate of change will re- they are implemented in the users favorite phylogenetic
main constant across the complete length of the sequences. software package. These sorts of choices should be avoided,
Consider an rRNA data set: who would assume that both however, and Table 2 should guide the less experienced
loops and stems evolve at the same rate? This heterogeneity readers toward sound decisions. We also provide a brief
in rate of evolution can be taken into account by introduc- recap of relevant factors below.
ing a new parameter in the model of evolution. This pa-
rameter, known as ct, will determine the shape of a r distri- 36.2.3.2. Type of Search
bution of the rate (68). When a is close to infinity, the As mentioned in the previous section, exhaustive searches
substitution rates are equal for all sites, and when ct is near are preferable to branch-and-bound searches, which are, in
0, then most of the sites do not evolve and a few evolve ex- turn, superior to heuristic searches. However, experienced
tremely rapidly. In addition, a proportion of sites that are in- phylogeneticists often sacrifice exhaustiveness first. In fact,.
variable can often be implemented. These invariable sites congruence between two or three independent heuristic
correspond to only a subset of the constant sites, since some searches (starting from different trees) is often regarded as a
can change but have not done so yet. Several software pack- strong indicator that the heuristic was not trapped in local
ages can be used to estimate these parameters; PAUP and optima and is usually obtained in a fraction of the time re-
TREE-PUZZLE are commonly employed for DNA align- quired by a full search.
ments and TREE-PUZZLE is used for amino acid align-
ments (Table 1). 36.2.3.3. Type of Data
The site-specific option is another way to take rate vari- The choice of data is much more controversial within the
ation into account. Unlike the r-distributed rates, users phylogenetic community. As we have already pointed out,
specify which sites belong to which category. This approach amino acid and nucleotide alignments are generally used for
is useful, for instance, for giving different rates to the three deep and shallow levels, respectively. However, concerns
positions of the codon or for stems versus the loops of RNA. have been raised recently, indicating that DNA alignments
might also be useful at deeper levels (57). Therefore, the
36.2.2.5.3. Over-Time Rate Variation and Further most pragmatic position one can take is to analyze both
Refinements data sets (as long as the proteins show some divergence and
Most of the commonly used algorithms assume that the the DNA sequences can be aligned) and compare the re-
substitution rate, regardless of its variation across sites, has sults. With few exceptions, distance methods are regarded
been constant over time (i.e., that the model is stationary). as less desirable than maximum-likelihood and Bayesian
This assumption, however mathematically convenient, has ones, since distance matrices imply a reduction of the in-
often been shown to be violated, especially for data sets in- formation. Nonetheless, distance matrices often permit a
volving long diverged organisms (see, for instance, refer- much better modeling of sequence evolution and can end
862 COMMUNITY AND GENOMIC ANALYSIS

up being more reliable and/or more robust than a data-based formed. The topology that displays the shortest sum of
analysis. Relative performance of distance-based over branch lengths is then selected, and the two OTUs are
maximum-parsimony methods is far less clear and often seems paired. A new distance matrix is then calculated and the
to depend on the data (e.g., distance methods often under- process is reiterated. The procedure stops when the starlike
perform for sequence collections with much missing data). component is resolved.
This method is extremely popular due to its extreme ef-
36.2.3.4. Type of Optimality Criterion ficiency and the fact that it produces one tree, whereas par-
Choice between the two families of distance optimality cri- simony can support hundreds of equally optimal trees.
teria (goodness of fit or minimum evolution) is usually of However, it is less accurate than maximum-likelihood-
modest importance. Similarly, Bayesian and maximum- based methods (22) and is therefore often seen only as a
likelihood searches usually produce very similar phyloge- good starting point for topological rearrangement per-
nies. Parsimony, on the other hand, can sometimes be at formed by the optimality criterion-based approach.
odds with other methods, particularly when data contain Many software packages include the option to construct
highly divergent homologous sequences. In this context, NJ trees, for example, ClustalX, PHYLIP, PAUP*, and
parsimony is more likely to artificially group such divergent Mega (Table 1).
sequences (a phenomenon better known as long-branch at-
traction [17]). This is not to say that parsimony always will
be erroneous when differing from likelihood, but that this 36.3. INTERPRETATION OF PHYLOGENETIC
known bias should be kept in mind when analyzing the re- TREES
sults. As a rule of thumb, it is always better to run more than
one type of analysis. In the best-case scenario, all methods 36.3.1. Reading of the Tree
will agree (be congruent in phylogenetic jargon) and the
operator will have confidence in the results. O n the other 36.3.1 . I . Rooting and Polarizing the Tree
hand, when some analyses differ, it may be possible to iden- Polarization of phylogenetic trees is an extremely important
tify the artifact. step and is often perceived as the most difficult one (61). At
this stage the investigator defines the directionality of evo-
36.2.3.5. Model of Evolution lution and the topology will pass from an unrooted (Fig.
When choosing between models of evolution, the investi- 3A) to a rooted one (Fig. 3B, C). To root a tree, external in-
gator needs to search for the most realistic model allowed by formation needs to be included. For example, if the se-
the data. From a statistical point of view, adding parameters quences under investigation are known to fall into two
to the model (e.g., increase the number of different rates of monophyletic (see below) groups (e.g., the Crenarchaeota
substitution, take into account base frequencies, or allow and the Euryarchaeota in Fig. 3), then it is possible to use
across-site rate variation) has a cost, and the cost is paid for this information to place the root between these two inde-
by the data. When the data do not contain enough infor- pendent entities. However, most of the time such a priori
mation, adding parameters will increase stochastic error and information is not known and the investigator will have to
therefore reduce the chance of finding the correct topology rely on outgroups. Outgroups are sequences that are as-
(61). sumed not to belong to the group investigated but to be rel-
Posada and Crandall (49) have developed software to atively close (e.g., other taxonomic units, paralogous
help determine which model fits nucleotide data best by genes). Given this assumption, one can use these sequences
maximizing model realism without overparameterizing the (usually at least two) to a posteriori polarize the tree, by
analysis (Modeltest; see Table 1). For proteins, where mod- placing the root between them and those from the group
els are mostly empirically based, it seems sensible to prefer under study. For example, if we are interested in the rela-
the model that was defined on similar data. For example, if tionships within Euryarchaeota, we can add three cren-
the data are of mitochondria1 origin, using the mtREV24 archaeal sequences so that we will be able to root the tree.
model is a good idea, while one would use the PMB matrix Importantly, if the rooting aspect of a tree is crucial to re-
(65) for a set of distant nuclear proteins and the JTT model solve a question, it is also likely to be the most unreliable
in most other cases. part of the analysis. By not being part of the studied group,
outgroups often display the longest branches and are most
36.2.4. Clustering Algorithms: a Shortcut likely to attract long branches within the studied group.
In their war against time, phylogeneticists have developed Therefore, extra care should be taken when discussing in-
clustering algorithms that can find a reasonable topology in ferences based on outgroup position. Note that removing an
only a fraction of the time required by criterion optimiza- outgroup can often reveal weak but true linkages that are
tion methods. What these clustering methods have in com- masked by the phylogenetic noise inherent to distant se-
mon is that the data sets are first reduced to a distance ma- quence usage.
trix and that trees are constructed by sequentially grouping
the sequences that are more similar until a tree is com- 36.3.1.2. Phylogenetic Terminology
pletely resolved. Unlike previous methods, no rearrange- Once polarization has been addressed, it is possible to fur-
ment of the full tree is performed, and the tree recovered is ther define taxonomic relationships. A group of taxa that
not compared with other possible topologies. include all the descendants of a common ancestor, as well as
Depending on the technique, different rules will govern the ancestor, is said to be monophyletic. Two such examples
the clustering process. We will limit ourselves to the most are highlighted by ovals in Fig. 3B. These monophyletic
commonly used: the NJ (neighbor-joining) methods of groups, or clades, are the only units recognized by cladist
Saitou and Nei (51). Using these procedures, all sequences classification, since they are the only ones that are defined
(OTUs) are first clustered in a single starlike tree. Then, a in terms of sharing of derived (evolved) characters: synapo-
first step is initiated in which all topologies having a pair in morphies. When a monophyletic group is composed of two
one side and the remaining starlike tree on the other are entities, generally these entities are referred to as sister
36. Interpreting Evolutionary Relationships H 863

B C
4
Methanosatrim barken
4
-41
Ar~haeoglobusfilgidus Atrhaeoglobus filgida
4 Thermopasma acidophilum

t Fmplasma acidamnus

r Methanobacteriumthermoautvirophicum Methanobacteriumthemutvtmphicum
Methanococcusjamschii
Pyrobaculum aerophilum
Aempymmpemk
Suhblobus solfatancus
Pyrobaculum aemphilum Pymocca f i r i ~ u s

H
20
Aempwm pemix
suhwobus solfatancus
f H
Pymoccus abpsi
Pymoccus honkoshii

20
FIGURE 3 Unrooted (A) versus rooted (B and C) trees. Ovals enclose monophyletic groups and
rectangles enclose paraphyletic groups. Underlined taxa are members of a polyphyletic group. The
black dots on the tree represent each groups ancestor. Note that when the outgroup is changed,
some monophyletic groups may become paraphyletic (e.g., the Euryurchaeom are monophyletic in
tree B but not in C). The scale bar represents substitutions per 100 sites.
864 COMMUNITY AND GENOMlC ANALYSIS

groups. A paraphyletic group, on the other hand, would unite better than the other (e.g., test the best tree versus the next
some but not all of the descendents of a common ancestor best one). Other tests such as the SOWH (61) or the SH
as well as the ancestor. Such a group is represented in Fig. (54) have been proposed to be valid in such cases, but often
3B by the dotted rectangles. Paraphyletic groups were rec- appear too liberal and too conservative, respectively (25).
ognized, in addition to monophyletic groups, by other More recently, Shimodaira (53) introduced a new
schools of classification (e.g., Linnaean and evolutionary Approximately Unbiased (AU) test that seems to be gain-
systematics) and are generally supported by synplesiornor- ing acceptance within the community. The AU test and nu-
phies (shared ancestral characters). Fishes or Reptilia are merous others are implemented in the CONSEL software
classical examples of paraphyletic groups, as they would package (55).
only turn monophyletic if tetrapods and birds were to be in-
cluded, respectively. In Fig. 3B, the third Pyrococcus se- 36.3.2.1.3. Posterior Probabilities
quence would have to be included. Finally, a polyphyletic The major difference between Bayesian inferences and
group is a group of taxa that share similarities that were pre- maximum likelihood lies in the way they deal with param-
sumably not present in their last common ancestor. For ex- eters. Likelihood parameter optimizations are very com-
ample, in Fig. 3B the three underlined taxa share a meth- puter intensive, while Bayesian integrations are analytically
anogenic ability, but it seems unlikely that the last common infeasible (34). Procedures that are able to approximate
ancestor of these three orders, plus the Halobacteriales, posterior probability not only of the tree but also of each
Archaeglobales, and Therrnoplasrnatales, had this function. In node have been implemented (e.g., MrBayes [32]). These
other words, polyphyletic groups have not inherited nor re- local posteriors have been proposed to replace bootstrap
tained their similarity, but obtained the similarity through values (see below) (34), but this early optimism needs to be
convergent evolution. revised as posterior probabilities are more likely to represent
All these definitions imply that the tree has been rooted. reliabilitys upper bound and bootstrap its lower bound (14).
However, the term monophyletic is commonly used if the
investigator feels that it is unlikely that the root will fall 36.3.2.2. Resampling
within the group of interest. Fig. 3C, for example, shows Resampling-based methods attempt to estimate the vari-
that if the real root of our tree lay along the Pyrococcus lin- ance of sampling (variance associated with taking, at best,
eage, instead of between Crenarchaeota and Euryarchaeota, only a few genes from few species into consideration) by
then Halobacteria would remain monophyletic (oval) but generating new data sets by resampling from the original
the Euryarchaeota would become paraphyletic. data set. The number of resamplings should ideally be
greater than 1,000, but one or a few hundred are often all
36.3.2. Robustness that can be reasonably computed.
When phylogenetic relationships have been identified, it is
important to critically evaluate them. In other words, is the 36.3.2.2.1. Bootstrap Percentages (15, 18)
observed relationship the result of a strong phylogenetic sig- Bootstrapping generates new data sets by performing
nal (a sharp peak in the phylogenetic landscape), or is it multiple drawings with replacement. If the initial data con-
only very slightly better than a multitude of different tain 150 sites, then the resampled data set (pseudoreplicate)
arrangements (a rock on a plateau)? To address this very will also be 150 sites long, but the first site could be present
important question, phylogeneticists have developed a large three times while the second may not be included at all. In
number of tools. These tools can be classified as methods subsequent steps, optimal trees for each pseudoreplicate
that do or do not resample the data. are computed and a consensus tree is drawn. For example,
when any given clade is recovered by two thirds of the
36.3.2.1. Initial Matrix without Resampling pseudoreplicates, then this node will have a bootstrap sup-
port of 66.6%. At or above 75%, the nodes are often re-
36.3.2.1.1. Decrease in Value of the Optimality garded as supported, and those above 95% are strongly
Criterion supported. However, evidence seems to suggest that boot-
This approach is highly intuitive and basically states strap values are overly conservative when in the high range,
that the cost, in terms of optimality criterion value, of tear- meaning that a node receiving 75% of bootstrap would be
ing apart a tree or breaking a given clade will depend on the accurate more than 75% of the time (conversely, low boot-
robustness of the tree or clade. This technique is computa- strap support would be overestimates [31]).
tionally efficient and can be used for any criterion-based
methods (4, 44). Software such as PAUP, PAML, Molphy, 36.3.2.2.2. Jackknife (46)
and MacClade (see Table 1) implement possibilities to as- The jackknife procedure shares many features with boot-
sess the optimality criterion for a tree that is chosen a pri- strapping, but instead of generating full-length pseudorepli-
ori. However, this method does not allow the determina- cates, jackknife produces truncated ones. Second, the re-
tion, statistically or objectively, of what is a reliable sampling is done without replacement so a site can only be
hypothesis. present one or zero times. It is unclear how many sites
should be dropped, but somewhere between Felsensteins
36.3.2.1.2. Nonparametric and Parametric 50% and Farriss e- is likely to be reasonable.
Statistical Tests
Statistical tests have been developed to circumvent the
previously mentioned problem. The Templeton test (62) is 36.4. SUMMARY AND CONCLUSION: WHY
classically used with parsimony-based approaches, whereas DO WE CONSTRUCT PHYLOCENETICTREES?
the KH test (37) has long been regarded as the reference for The classical and principal reason for constructing phyloge-
likelihood-based studies. However, the KH test is not valid netic trees is to determine how different sequences are re-
when there is a priori reason to believe that one topology is lated. This can be then extrapolated as reflecting the evo-
36. Interpreting Evolutionary Relationships rn 865

lutionary history of the organisms from which the sequences tion 36.5), and methods have been developed to detect pos-
were sampled. In molecular microbiology, trees estimated itive selection along branches on a phylogeny as well as
from 16s rRNA genes have classically been used for this detecting amino acid sites under positive selection (see ref-
purpose (11). However, with the increasing amount of se- erence 69 for a review, as well as the PAML package in
quences available, mainly from the genome-sequencing Table 1). Since these methods are based on DNA align-
projects, it has become increasingly evident that many (if ments and synonymous mutations reach saturation rapidly,
not most) genes from prokaryotic genomes have different they are best suited for relatively closely related sequences,
evolutionary relationships due to transfer of genes between whereas the methods described above can be used for more
lineages or species, a phenomenon termed lateral or hori- divergent sequence data.
zontal gene transfer (LGT) (12,24). The phylogenetic trees Phylogenetic trees can also be used to reconstruct ances-
estimated from all the genes in a genome will, in most cases, tral sequences. These ancestral sequences can provide struc-
represent gene trees and not organismal trees. However, tural and functional information on protein families, both
phylogenetic analysis can be used to identify the laterally when used in computational studies (45) and when used to
transferred genes (some examples are given in Table 1 in reconstruct ancestral sequences through recombinant DNA
reference 13). For instance, if a bacterial gene is found technology (58). This approach has also been explored to
nested within a eukaryotic clade, it is likely that the gene detect covariation signals in proteins (20). Such signals are
was transferred from a eukaryote to the bacterium. For dis- detected as patterns of compensatory amino acid replace-
cussions on how to detect LGT from phylogenetic analyses, ments. For example, the replacement of an amino acid at a
see, for example, references 2 and 59. given position with a residue that reverses the change may
Phylogenies are not only used for inferring the evolu- consistently be associated with the replacement of an
tionary relationship between sequences per se; phylogenet- amino acid at a second position with a residue that restores
ics can also be used to help in the annotation of genomes the charge. Fukami-Kobayashi et al. (20)showed that re-
(16, 56). Most genes from genome-sequencing projects are constructing ancestral sequences gives a stronger covaria-
annotated based on results from pairwise comparisons from tion signal than comparisons of contemporary sequences.
database searches. However, paralogous genes often do not It should be apparent from this chapter that the range of
have the same function, and such genes cannot be distin- phylogenetic techniques is considerable, as are the ques-
guished by simple pairwise comparisons. Hence, tools for tions that can be addressed using these techniques. It should
automated phylogenetic analyses of complete genome data also be clear that phylogenetic analysis cannot be regarded
have recently been developed which can help facilitate as a black box and that each analysis will be custom
gene annotation as well as flag interesting genes for more made. We hope that this chapter has provided some guide-
detailed phylogenetic analyses (see, e.g., reference 56). lines to what methods and choices are available.
Phylogenetic analyses can also be used to identify func-
tionally and structurally constrained residues in proteins or
RNA (see, e.g., references 5,9, and 35). For instance, using 36.5. SOME DEFINITIONS
maximum-likelihood phylogeny, it is possible to assign rela-
Homology Two genes are homologous if they descend
tive rates to each site and use this quantity as a measure of
from a common ancestral gene. When working with se-
constraints (3). This type of analysis relies on the hypothe-
quence alignments, it is important to remember that
sis that functionally important sites usually evolve more
even if two sequences are homologous, insertions and
slowly than nonfunctional sites. Conversely, if a protein
deletions (indels) may mix up the positional homology
family has been well studied, phylogenetic analyses can also
of the nucleotide or amino acid sites. Note that homol-
be used to investigate whether new sequences also possess
ogy is commonly misused to mean similarity.
functionally important residues. This can provide valuable
information on how proteins evolve new substrate specifici- Orthology Two homologous sequences are orthologs if
ties or how generally applicable previously identified func- their common history can be traced back to a speciation
tional residues may be across taxonomic diversity (see refer- event.
ences 9 and 43 for further details). Paralogy Two homologous sequences are paralogs if their
Another related type of comparative analysis is predic- common history can be traced back to a duplication
tion of functional divergence by identifying site-specific event.
rate shifts (see reference 23 for a review). These analyses Synonymous and nonsynonymous substitutions Syno-
map the evolutionary rates of amino acid replacement at nymous mutations are mutations that do not result in an
sites along the protein sequence, using the gamma distribu- amino acid replacement, whereas nonsynonymous sub-
tion described in section 36.2.2.5.2. If a functional shift has stitutions do result in an amino acid replacement.
occurred in the evolution of a protein family, then the dis- Xenology Two homologous sequences are xenologs if
tribution of evolutionary rates along the protein sequence their common history can be traced back to a lateral
can be expected to vary in different subtrees. When data gene transfer event.
from structural analyses are included, these comparative ap-
proaches can be extremely powerful. Recently, methods for
characterizing rate-independent functional divergence 36.6. FREQUENTLY ASKED QUESTIONS
have been described (3) and can be found in the software
package covARES. One advantage of this package is that it Why is there no single recipe?
provides both evolutionary and structural parameter map- Unlike DNA or RNA extractions, phylogenetic analyses
ping and analysis tools. are custom made and therefore cannot be accurately sum-
Prediction of functionally important sites and detection marized in a single protocol. The analyses will need to be
of molecular adaptation can also be done by measuring the adapted depending on the number of taxa (OTUs), the
ratio of synonymous to nonsynonymous mutations (see sec- length of the sequences analyzed, the degree of divergence,
866 COMMUNITY AND GENOMIC ANALYSIS

the percentage of missing data, and the computing facilities other are formed. The topology that displays the shortest
at hand. sum of branch lengths is then selected and the two OTUs
are paired. A new distance matrix is then calculated and the
Is there an easy way to make a tree? process is reiterated. The procedure stops when the starlike
No, but if you really need a tree quickly and do not want to component is resolved.
put too much time into the analysis:
What method is the best?
1. Create a file in FASTA format (Fig. 2) that includes This question is the most frequent and yet the most debated
all sequences of interest. in the field. Based on simulated data, maximum likelihood is
2. Download and install the latest version of ClustalX often regarded as the best tool (32). Bayesian methods, if we
on your system (http://www.bips.u-strasbg.fr/fr/ consider only the most probable topology (and not the still
Documentation/ClustalX) . highly questionable posterior probabilities of the nodes), are
3. Start ClustalX. likely to be just as useful as maximum likelihood. Depending
4. Click on File, Load sequences. on the data, parsimony- or distance-based methods will then
5. Select your FASTA file. be the next best choice. For example, if a data set contains
6. Click open. much missing data, a parsimony-based reconstruction will be
7. Click on Alignment, DOComplete Alignment, more likely to provide a sensible topology, but this method
Align. will become highly variable when extremely diverged taxa
8. Wait until completion (look for a CLUSTAL- are under study. However, given that likelihood and
Alignment file created [I message at the bottom left of the Bayesian inference can be significantly more time consum-
window). ing and that bias can interfere with simulation results, we
9. Click on Trees. recommend one more time that each phylogenetic analysis
10. Check Exclude Positions with Gap, Correct for should be treated as a unique case and that custom-made de-
Multiple Substitution. cisions should be made before each study.
11. Draw NJ tree.
12. Download (http://taxonomy.zoology.gla.ac.uk/rod/ We are grateful to Andrew Roger, Rebecca Case, W. Ford Doolittle,
treeview.htm1) and install the latest version of Treeview on and two anonymous referees for cornmenu and suggestions. This work
your system to visualize, present, and print your tree. wus supported by Genome Atlantic (an affiliate of Genome Canada),
the Killam Foundation, and the Canadian Institutes for Health
What is maximum parsimony? Research.
The most parsimonious topology is the tree, or set of trees,
that requires the smallest number of evolutionary changes. 36.7. REFERENCES
When doing an exhaustive search (see section 36.2.2.2.),
the minimum number of changes for all sites in the align- 36.7.1. Further Reading
ment is computed for all possible topologies, and the tree re- 1. Felsenstein, F. 2004. Inferring Phylogenies. Sinauer
quiring the fewest changes (or steps, in parsimony jargon) Associates, Sunderland, MA.
is selected. Under a heuristic search, the minimum number The most complete. THE reference.
of steps is determined for the two topologies to be compared 2. Graur, D., and W.-H. Li. 2000. Fundamentals ofMolecular
(the best topology so far in the search and the rearranged Evolution. Sinauer Associates, Sunderland, MA.
one; see section 36.2.2.2) and the more parsimonious of the As indicated by its title, the primary focus of this book is molec-
two is selected and itself rearranged for the next step. Over ular evolution. However, numerous notions introduced mostly
the years, several variants of parsimony have been developed in chapters 3 to 5 will be ofgreat help to any apprentice phylo-
that, for example, forbid convergence (Do110 parsimony), geneticists. The help will nevertheless remain on the theoretical
take into account that some changes are easier than oth- side, as few if any practical considerations are discussed.
ers (generalized parsimony), or assume some sites are better 3. Hall, B. G. 2001. Phylogenetic Trees Made Easy: A HOW-TO
Manual for Molecular Biologists. Sinauer Associates,
phylogenetic markers than others (weighted parsimony). Sunderland, MA.
What is maximum likelihood? By authors own admission, this book should be taken us a
Not unlike maximum parsimony, a maximum-likelihood cookbook and not as a textbook. It will prove most useful for in-
procedure will look for the tree that maximizes the likeli- experienced Mac users who are interested in generating neighbor-
joining and Bayesian trees. This book also includes a very use-
hood of observing the data given the topology, its branch
ful section on generating and improving alignments.
lengths, the model of evolution, and parameters, such as the 4. Page, R. D. M., and E. C. Holmes. 1998. Molecular Euo-
shape of the gamma distribution, that account for the lution: a Phylogenetic Approach. Blackwell Science, Oxford.
among-site rate variation. In this way, searches will compare This book can definitively be regarded as the textbook in phy-
topologies by multiplying the likelihood of each individual logenetic reconstruction. With the exception of Bayesian infer-
site and selecting the tree having the highest product. ence, which wus being developed at the time, most of the major
topics in the field are nicely presented. This work is the most ad-
What is a neighbor joining?
equate introduction to the Swofford et al. (1996) publication.
Neighbor joining (NJ) is an extremely efficient, distance- 5. Page, R. D. M., 0. Gascuel, J. C. Wilgenbush, D.
based algorithm (see section 36.2.4). Unlike maximum Swofford, D. Posada, H. A. Schmidt, A. von Haeseler,
likelihood or parsimony, this method is not based on the and D. H. Huson. 2003. Inferring evolutionary relation-
maximization of any criterion and does not do any compar- ships. In A. D. Baxevanis, D. B. Davison, R. D. M. Page,
ison of trees. O n the contrary, this algorithmic procedure G. A. Petsko, L. D. Stein, and G. D. Stormo (ed.), Current
will carry out an iterative hierarchization of the OTUs. All Protocols in Bioinfomtics, vol. 1. John Wiley &a Sons, New
sequences (OTUs) are first clustered in a single starlike tree. York, NY.
Then, a first step is initiated in which all topologies having This section of the Current Protocols in Bioinformatics pro-
a pair in one side and the remaining starlike tree on the vides classic and yet accessible protocols for choosing models of
36. Interpreting Evolutionary Relationships 867

evolution and finding most parsimonious and neighbor-joining 15. Efron, B. 1979. Bootstrap methods: another look at the
trees as well as visualizing them. The section is completed by an jackknife. Ann. Stat. 7:l-26.
introduction to phylogenetic inference, usage of quartet puzzling 16. Eisen, J. A., and M. Wu. 2002. Phylogenetic analysis and
to approximate the most likely tree, and an alternative approach gene functional predictions: phylogenomics in action.
fur non-treelike data. Theor. Popul. Biol. 61:481487.
6. Swofford, D. L., G. P. Olsen, P. J. Waddell, and D. M. 17. Felsenstein, J. 1978. Cases in which parsimony or com-
Hillis. 1996. Phylogenetic inference, p. 407-514. In D. M. patibility methods will be positively misleading. Syst. Zool.
Hillis, C. Moritz, and B. K. Mable (ed.), Molecular Sys- 27:401-410.
tematics. Sinauer Associates, Sunderland, MA. 18. Felsenstein, J. 1985. Confidence limits on phylogenies: an
This book chupter is essential fur any tree builder who wants to approach using the bootstrap. Evolution 39:783-791.
know more. Afew ofthe newer techniques are missing, but it still 19. Fitch, W. M. 1971. Rate of change of concomitantly vari-
remains valuable. It is likely to be a bit cryptic to any newcomer, able codons. J. Mol. Evol. 1:84-96.
but reading Page and Holmess book first will make things clearer. 20. Fukami-Kobayashi, K., D. R. Schreiber, and S. A.
Benner. 2002. Detecting compensatory covariation signals
36.7.2. General References in protein evolution using reconstructed ancestral se-
1. Adachi, J., and M. Hasegawa. 1996. Model of amino acid quences. J. Mol. Biol. 319:729-743.
substitution in proteins encoded by mitochondria1 DNA. 21. Galtier, N. 2001. Maximum-likelihood phylogenetic
J. Mol. Evol. 42459-468. analysis under a covarion-like model. Mol. Biol. Evol. 18:
2. Andersson, J. O., W. F. Doolittle, and C. L. Nesbo. 866-873.
2001. Genomics. Are there bugs in our genome? Science 22. Gascuel, 0. 2003. Getting a tree fast: neighbor joining
292:1848-1850. and distance based methods, p. 6.3.1-6.3.18. In G. D.
3. Blouin, C., Y. Boucher, and A. J. Roger. 2003. Inferring Stormo (ed.), Current Protocols in Bioinformatics, vol. I.
functional constraints and divergence in protein families John Wiley & Sons, Inc., Hoboken, NJ.
using 3D mapping of phylogenetic information. Nucleic 23. Gaucher, E. A., X. Gu, M. M. Miyamoto, and S. A.
Acids Res. 31:790-797. Benner. 2002. Predicting functional divergence in protein
4. Bremer, K. 1994. Branch support and tree stability. evolution by site-specific rate shifts. Trends Biochem. Sci.
Cladistics 10:295-304. 27:3 15-3 2 1.
5. Cannone, J. J., S. Subramanian, M. N. Schnare, J. R. 24. Gogarten, J. P., W. F. Doolittle, and J. G. Lawrence.
Collett, L. M. DSouza, Y. Du, B. Feng, N. Lin, L. V. 2002. Prokaryotic evolution in light of gene transfer. Mol.
Madabusi, K. M. Muller, N. Pande, Z. Shang, N. Yu, Biol. Euol. 19:2226-2238.
and R. R. Gutell. 31 May 2002. The Comparative RNA 25. Goldman, N., J. P. Anderson, and A. G. Rodrigo. 2000.
Web (CRW) Site: an online database of comparative se- Likelihood-based tests of topologies in phylogenetics. Syst.
quence and structure information for ribosomal, intron, Biol. 49:652-670.
and other RNAs. BMC Bioinfomtics 3:2. [online.] 26. Graham, R. L., and L. R. Foulds. 1982. Unlikelihood
http://www.rna.ccbb.utexas.edu/. that minimal phylogenies for a realistic biological study
6. Castresana, J. 2000. Selection of conserved blocks from can be constructed in a reasonable computational time.
multiple alignments for their use in phylogenetic analysis. Math. Biosci. 60:133-142.
Mol. Biol. Evol. 17540-552. 27. Graur, D., and W.-H. L. Li. 2000. Fundamentals of Molec-
7. Cole, J. R., B. Chai, T. L. Marsh, R. J. Farris, Q. Wang, ular Evolution, 2nd ed. Sinauer Associates, Sunderland, MA.
S. A. Kulam, S. Chandra, D. M. McGarrell, T. M. 28. Hall, B. G. 2001. Phylogenetic Trees Made Easy: A How-
Schmidt, G. M. Garrity, and J. M. Tiedje. 2003. The to Manual for Molecular Biologists. Sinauer Associates,
Ribosomal Database Project (RDP-11): previewing a new Sunderland, MA.
autoaligner that allows regular updates and the new 29. Hendy, M. D., and D. Penny. 1982. Branch and bound al-
prokaryotic taxonomy. Nucleic Acids Res. 31442-443. gorithms to determine minimal evolutionary trees. Math.
8. Cunningham, C. W. 1997. Is congruence between data Biosci. 59:277-290.
partitions a reliable predictor of phylogenetic accuracy? 30. Henikoff, S., and J. G. Henikoff. 1992. Amino acid sub-
Empirically testing an iterative procedure for choosing stitution matrices from protein blocks. Proc. Natl. Acad.
among phylogenetic methods. Syst. Biol. 4 6 4 6 4 4 7 8 . Sci. USA 89:10915-10919.
9. Dacks, J. B., and W. F. Doolittle. 2002. Novel syntaxin 31. Hillis, D. M., and J. J. Bull. 1993. A n empirical test of
gene sequences from Giardia, Trypanosoma and algae: im- bootstrapping as a method for assessing confidence in phy-
plications for the ancient evolution of the eukaryotic en- logenetic analysis. Syst. Biol. 42:182-192.
domembrane system. J Cell Sci. 115:1635-1642. 32. Huelsenbeck, J. P. 1995. The robustness of two phyloge-
10. Dayhoff, M. O., R. M. Schwartz, and B. C. Orcutt. netic methods: four#taxon simulations reveal a slight supe-
1978. A model of evolutionary change in proteins, p. 345- riority of maximum likelihood over neighbor joining. Mol.
352. In M. 0. Dayhoff (ed.), Atlas of Protein Sequence Biol. Evol. 12:843-849.
Structure, vol. 5. National Biomedical Research Founda- 33. Huelsenbeck, J. P., and F. Ronquist. 2001. MRBAYES:
tion, Washington, DC. Bayesian inference of phylogenetic trees. Bioinfomtics
11. DeLong, E. F. 2002. Microbial population genomics and 17: 754-755.
ecology. Curr. Opin. Microbiol. 5520-524. 34. Huelsenbeck, J. P., F. Ronquist, R. Nielsen, and J. P.
12. Doolittle, W. F. 1999. Phylogenetic classification and the Bollback. 2001. Evolution: Bayesian inference of phylo-
universal tree. Science 284:2124-2129. geny and its impact on evolutionary biology. Science 294:
13. Doolittle, W. F., Y. Boucher, C. L. Nesbqi, C. J. Douady, 2310-2314.
J. 0. Andersson, and A. J. Roger. 2003. How big is the 35. Inagaki, Y., C. Blouin, W. F. Doolittle, and A. J. Roger.
iceberg of which organellar genes in nuclear genomes are 2002. Convergence and constraint in eukaryotic release
but the tip? Philos. Trans. Biol. Sci. 358:39-58. factor 1 (eRF1) domain 1: the evolution of stop codon
14. Douady, C. J., F. Delsuc, Y. Boucher, W. F. Doolittle, specificity. Nucleic Acids Res. 30532-544.
and E. J. P. Douzery. 2003. Comparison of Bayesian and 36. Jones, D. T., W. R. Taylor, and J. M. Thornton. 1992.
maximum likelihood bootstrap measures of phylogenetic The rapid generation of mutation data matrices from pro-
reliability. Mol. Biol. Evol. 20:248-254. tein sequences. CABIOS 8:275-282.
868 w COMMUNITY AND GENOMIC ANALYSIS

37. Kishino, H., and M. Hasegawa. 1989. Evaluation of the Phylogeny. Cambridge University Press, Cambridge,
maximum likelihood estimate of the evolutionary tree United Kingdom.
topologies from DNA sequence data, and the branching 53. Shimodaira, H. 2002. An approximately unbiased test of
order in hominoidea. J. Mol. Euol. 29:170-179. phylogenetic tree selection. Syst. Biol. 5 1:492-508.
38. Lanave, C., G. Preparata, C. Saccone, and G. Serio. 54. Shimodaira, H., and M. Hasegawa. 1999. Multiple com-
1984. A new method for calculating evolutionary substitu- parisons of log-likelihoods with applications to phyloge-
tion rates. J. Mol. Euol. 20:86-93. netic inference. Mol. Biol. Euol. 16:1114-1 116.
39. Lockhart, P. J., M. A. Steel, A. C. Barbrook , D. H. 55. Shimodaira, H., and M. Hasegawa. 2001. CONSEL: for
Huson, M. A. Charleston, and C. J. Howe. 1998. A co- assessing the confidence of phylogenetic tree selection.
variotide model explains apparent phylogenetic structure Bioinfomatics 17: 1246-1247.
of oxygenic photosynthetic lineages. Mol. Biol. Euol. 15: 56. Sicheritz-Ponten, T., and S. G. Anderson. 2001. A phy-
1183-1 188. logenomic approach to microbial evolution. Nucleic Acids
40. Lopez, P., D. Casane, and H. Philippe. 2002. Hete- Res. 29545-552.
rotachy, an important process of protein evolution. Mol. 57. Simmons, M. P., H. Ochoterena, and J. V. Freudenstein.
Biol. E d 19:l-7. 2002. Amino acid vs. nucleotide characters: challenging
41. Lopez, P., P. Forterre, and H. Philippe. 1999. The root of preconceived notions. Mol. Phylogenet. Euol. 24:78-90.
the tree of life in the light of the covarion model. J. Mol. 58. Stackhouse, J., S. R. Presnell, G. M. McGeehan, K. P.
E d . 49~496-508. Nambiar, and S. A. Benner. 1990. The ribonuclease from
42. Loytynoja, A., and M. C. Milinkovitch. 2001. SOAP, an extinct bovid ruminant. FEBS Lett. 262:104-106.
cleaning multiple alignments from unstable blocks. 59. Stanhope, M. J., A. Lupas, M. J. Italia, K. K. Koretke,
Bioinfomatics 1 7 573-574. C. Volker, and J. R. Brown. 2001. Phylogenetic analyses
43. Lucas, P., C. Otis, J. P. Mercier, M. Turmel, and C. do not support horizontal gene transfers from bacteria to
Lemieux. 2001. Rapid evolution of the DNA-binding site vertebrates. Nature 41 1:940-944.
in LAGLIDADG homing endonucleases. Nucleic Acids 60. Swofford, D. L. (ed.). 2001. PAUP". PhylogeneticAnalysis
Res. 29:960-969. Using Parsimony ("and Other Methods), Version 4. Sinauer
44. Meireles, C. M., J. Czelusniak, M. P. Schneider, J. A. Associates, Sunderland, MA.
Muniz, M. C. Brigido, H. S. Ferreira, and M. Goodman. 61. Swofford, D. L., G. P. Olsen, P. J. Waddell, and D. M.
1999. Molecular phylogeny of ateline new world monkeys Hillis. 1996. Phylogenetic inference, p. 407-514. In B. K.
(Platyrrhini, atelinae) based on gamma-globin gene se- Mable (ed.), Molecular Systematics. Sinauer Associates,
quences: evidence that brachyteles is the sister group of Sunderland, MA.
lagothrix. Mol. Phylogenet. Euol. 12:lO-30. 62. Templeton, A. R. 1983. Phylogenetic inference from re-
45. Messier, W., and C. B. Stewart. 1997. Episodic adaptive striction endonuclease cleavage site maps with particular
evolution of primate lysozymes. Nature 385:151-154. reference to the evolution of humans and the apes.
46. Mueller, L. D., and F. J. Ayala. 1982. Estimation and in- Evolution 37:22 1-244.
terpretation of genetic distance in empirical studies. 63. Thompson, J. D., D. G. Higgins, and T. J. Gibson. 1994.
Genet. Res. 40:127-137. CLUSTAL W: improving the sensitivity of progressive
47. Page, R. D. M., 0. Gascuel, J. C. Wilgenbush, D. multiple sequence alignment through sequence weighting,
Swofford, D. Posada, H. A. Schmidt, A. von Haeseler, position-specific gap penalties and weight matrix choice.
and D. H. Huson. 2003. Inferring evolutionary relationships. Nucleic Acids Res. 22:4673-4680.
In L. D. Stein and G. D. Stormo (ed.), Current Protocols in 64. Thompson, J. D., F. Plewniak, and 0. Poch. 1999. A
Bioinfomatics, vol. 1. John Wiley & Sons, Hoboken, NJ. comprehensive comparison of multiple sequence align-
48. Penny, D., B. J. McComish, M. A. Charleston, and ment programs. Nucleic Acids Res. 27:2682-2690.
M. D. Hendy. 2001. Mathematical elegance with bio- 65. Veerassamy, S., A. Smith, and E. R. M. Tillier. 2003. A
chemical realism: the covarion model of molecular evolu- transition probability model for amino acid substitutions
tion. J. Mol. Euol. 53:711-723. from BLOCKS. J. Comput. Biol. 10:997-1010.
49. Posada, D., and K. A. Crandall. 1998. MODELTEST: 66. Wuyts, J., P. De Rijk, Y. Van de Peer, T. Winkelmans,
testing the model of DNA substitution. Bioinfomatics 14: and R. De Wachter. 2001. The European Large Subunit
8 17-8 18. Ribosomal RNA database. Nucleic Acids Res. 29:175-
50. Rannala, B., and Z. H. Yang. 1996. Probability distribu- 177.
tion of molecular evolutionary trees: a new method of phy- 67. Wuyts, J., Y. Van de Peer, T. Winkelmans, and R. De
logenetic inference. J. Mol. Euol. 43:304-311. Wachter. 2002. The European database on small subunit
51. Saitou, N., and M. Nei. 1987. The neighbor-joining ribosomal RNA. Nucleic Acids Res. 30:183-185.
method: a new method for reconstructing phylogenetic 68. Yang, Z. 1996. Among-site rate variation and its impact
trees. Mol. Biol. Euol. 4:406-425. on phylogenetic analyses. Trends Ecol. Euol. 11:367-372.
52. Salemi, M., and A.-M. Vandamme. 2003. The Phylogenetic 69. Yang, Z. 1996. Phylogenetic analysis using parsimony and
Handbook. A Practical Approach to DNA and Protein likelihood methods. J. Mol. Euol. 42:294-307.
37
Microbial Nucleotide Fingerprints in Nature
DAVID M. KARL

37.1. INTRODUCTION ......................................... 869


.2. SAMPLING A N D SUBSAMPLING THE ENVIRONMENT ......... 87 1
.3. BIOMASS DETERMINATION BY ATP ......................... 871
.4. MATERIALS REQUIRED ................................... 87 1
37.4.1. Equipment .......................................... 871
.2. Supplies ........................................... 872
................................. 872
.3. Solutions and Reagents
37.5. PROCEDURES ........................................... 872
......... 872
37.5.1. Sampling, Subsampling, Extraction, and Sample Storage
37.5.1.1. Water Column.. .............................. 872
.2. Sediment Column ............................. 872
37.5.2. Detection of ATP by Firefly Bioluminescence ................ 873
.3. ATP Standards ...................................... 874
.......... 874
.4. Data Reduction and Extrapolations of ATP to Biomass
37.6. NOTES AND COMMENTS .................................. 874
3 7.6.1. Sampling and Subsampling .............................. 874
.2. Extraction .......................................... 874
....................... 875
.3. Sensitivity, Precision, and Accuracy
.......... 875
.4. Analytical Interferences and Use of Internal Standards
37.7. ENHANCING THE MICROBIAL NUCLEOTIDE FINGERPRINT .... 875
.8. CONCLUSION ........................................... 876
.9. REFERENCES ............................................ 876

37.1. INTRODUCTION ecological assessment. Despite the recognized importance of


Microorganisms are the key to Earths habitability. They total microbial community assessments, however, few meth-
harvest light energy, produce organic matter, and facilitate ods currently exist, and those that have been developed
the turnover of key bioelements like nitrogen (N), phos- have unique limitations that derive from the fact that the
phorus (P), and sulfur (S). During the process of microbial assay targets are generally diverse, partially uncharacterized,
cell metabolism and growth, inorganic nutrients are assim- and in variable states of metabolism and growth. A forward-
ilated into new biomass that fuels a complex series of in- looking review article by Madsen (40), Epistemologyof en-
teractions between various groups of microorganisms and vironmental microbiology, presents an up-to-date progress
among microbes and multicellular organisms. In summary, report of the constraints imposed by the complexity of nat-
microbes make things happen. ural ecosystems and by limited methodology.
Microbial communities in nature are diverse and com- In pure culture studies, the measurement of biomass is
plex. With the advent of modern techniques in molecular fairly easily satisfied. However, as stated by Koch (36) in his
biology, one can now identify and track even the virtual authoritative review on growth measurements, the meth-
microbes-those species that have successfully evaded pure ods for measuring biomass seem obvious and straight-
culture isolation. It is also possible to interrogate ecosys- forward, but in fact they are complicated if accuracy is
tems for the presence of specific genes and gene products, sought. He goes on to review the methods that are most
and even determine in situ expression of target genes. The often applied to laboratory studies of pure culture isolates,
present level of analytical sophistication and the continued namely wet and dry weight, light scattering and turbidime-
improvement of techniques are impressive and hold great try, and various enumeration techniques. These last in-
promise to yield new, fundamental information regarding clude light and electron microscopy and flow cytometry for
the phylogenetic and physiological structure of microbial direct counts, as well as spread plates, most probable num-
assemblages in nature. By comparison with the develop- ber, and other culture methods for viable cell counts. If the
ment of novel autecological approaches, progress in the de- average dimensions (or average mass or carbon content) of
velopment and refinement of methods that target the en- the cells in culture are known, or measured, then the direct
tire microbial community, specifically estimation of total or viable colony count can be extrapolated to biomass.
living biomass, metabolic activity, growth, and reproduc- However, even with pure culture isolates, cell size, mass,
tion, has been less dramatic. and elemental composition may vary depending on com-
For many ecological studies, accurate estimates of total position of the growth medium, culture conditions (temper-
microbial biomass, metabolic activity, and productivity are ature, light, pressure), and growth rate. These laboratory-
essential; they are the key master variables for a complete based methods for direct and indirect estimation of biomass

869
870 w COMMUNITY AND GENOMIC ANALYSIS

are not easily adapted to the complex mixed cultures of mi- analyses. Furthermore, it now appears that certain ubiqui-
croorganisms that one generally finds in nature. tous marine microorganisms, e.g., Synechococcus and
The direct measurement of carbon, nucleic acids, pro- Prochlorococcus, may have reduced cell quotas of membrane
tein, or other similar cell-derived compounds is generally phospholipids (3,9), so they would not be accurately repre-
unacceptable due to the relatively long residence times sented in the environmental microbial biomass assessment.
in the environment after cell death and lysis. For example, in The second and, perhaps, more broadly applicable class
most open-ocean habitats there is more particulate carbon of cellular biomarkers is nucleotides and their derivatives.
(PC) in nonliving than in living materials, and the propor- All living cells studied to date have an identical suite of
tion varies considerably with water depth (approximately more than 100 different nucleotides that are essential for
50% of total carbon is living near the surface, decreasing to viability, energy transduction, metabolic regulation, and
about 5% at 1,000 m) (29). There is even more nonliving biosynthesis. Although it is impossible to be absolutely cer-
particulate DNA in most natural ecosystems than there is tain on this point, it is highly unlikely that any microor-
inside all of the cells that are present. Nevertheless, mea- ganisms will be discovered with alternative pathways; the
surements of total PC or DNA can be used to set upper universal role of nucleotides and their derivatives appears to
bounds on total biomass and, as such, they have been used be well established (21).
extensively in ecological studies. The central role of the adenine nucleotides, and espe-
A n effective biochemical or molecular biomarker of mi- cially ATP, as intermediate carriers of chemical energy link-
crobial biomass should satisfy the following criteria (22): (i) ing catabolism and biosynthesis has been known since the
the biomarker selected must be present in all living cells or publication of Lipmanns classic paper (39). Nucleotides
in all organisms of a specific population; (ii) it must be readily have at least three other crucial cellular functions: (i) syn-
metabolized, hydrolyzed, or otherwise decomposed follow- thesis of DNA and RNA; (ii) activation and transfer of pre-
ing cell death; (iii) it must exist as a uniform and constant cursors for cellular biosynthesis; and (iii) control and regula-
percentage of total biomass regardless of environmental or tion of cellular metabolism. By measuring the intracellular
physiological conditions; (iv) a convenient method must concentrations of key nucleotides and ratios thereof, the
exist to extract and purify (if necessary) the compound from intracellular pool turnover rates, and nucleic acid (DNA,
environmental samples; and (v) a sensitive, quantitative RNA) synthesis rates, an environmental nucleotide finger-
assay procedure must be available. Microbial biomass in print can be compiled that provides valuable information
many natural ecosystems is often low, so assay sensitivity is concerning in situ biomass and metabolic activity of the
an important criterion. total microbial community.
Only a few common features are shared by all of the di- Over 40 years ago, methods were developed for quanti-
verse members of natural microbial assemblages that might tative extraction of ATP from cells and for its detection in
serve as the basis for an environmental biomass assay sys- cell extracts. Initially, the ATP detection method was pro-
tem, and for this reason, progress in these important areas of moted as a life detection system for possible use on the
microbial ecology has been slow. Two promising candidate Martian spacecraft Viking in 1974 (38), but a few years later
classes of molecular biomarkers for ecological investigation the method was expanded into a total microbial biomass
are membrane lipids and cellular nucleotides. Other com- assay by Holm-Hansen and Booth (15). The steady-state in-
pounds have been used in environmental studies to detect tracellular concentration of ATP (referred to as the ATP
the biomass of a portion of the total microbial assemblage. pool) in metabolically active microorganisms (prokaryotes
These include chlorophyll a for the sum of all photosyn- and eukaryotes) appears to be well regulated at a value of 2
thetically active bacteria and eukarya, lipopolysaccharide to 6 nmol of ATP mg- dry weight (1 to 3 mM) regardless
for gram-negative bacteria, and muramic acid for gram- of growth rate, culture condition, or mode of nutrition (21).
positive plus gram-negative bacteria (29). This served as the theoretical basis for its use in total mi-
Determination of total lipid phosphate (LP) as a quanti- crobial biomass estimation.
tative measure of microbial biomass was originally described Subsequent investigation led to the development of the
by White et al. (51) in their application to marine sedi- adenylate energy charge (ECA) concept and its application
ments. The general method was subsequently expanded to microbial assemblages in nature (53). The E C A is equal
to include (i) LP production rate, using radioactive phos- to one-half of the number of anhydride-bound phosphate
phate (32Por 33P) as a precursor, as a measure of total mi- groups per adenine moiety, or E C A = ([ATP] + [ADPI)/
crobial production (50); (ii) the pattern of signature ester- +
([ATP] + [ADP] [AMP]), and is equivalent to a linear
linked phospholipids fatty acids (PLFA) and hydroxyl fatty measure of the total amount of chemical potential energy
acids in the lipopolysaccharide of gram-negative bacteria as momentarily stored in the adenine nucleotide pool (1). In
measures of microbial community structure (46, 52); and vitro rate responses of several key enzymes in cellular me-
(iii) changes in the concentrations of poly-P-hydroxyalkanoic tabolism to variations in ECA have provided the background
acid (PHA) in bacteria or triglyceride in microeukaryotes as data for this control hypothesis. The well-established corre-
measures of nutritional-physiological status (13). These di- lation between cellular ECA and metabolic or growth po-
agnostic lipid-based profiles, or environmental microbial tentials of individual organisms, in theory, provides a frame-
lipid fingerprints, have proven invaluable in a variety of work for estimating metabolic potentials of naturally
ecological studies (1 1). Major limitations, according to occurring microbial populations (6).
White et al. (52), are the nontraditional units that result In addition to ATP, the measurement of non-adenine
from these analyses (e.g., picomoles of PLFA per sample, NTPs may provide relevant ecological information. Many
rather than total cell mass or carbon), the relative insensi- important cellular reactions, notably those associated with
tivity (detection limit of approximately 10 bacteria per microbial biosynthesis and growth, are coupled to energy
sample for biomass by LP analysis; for oligotrophic ocean derived from NTPs other than ATP (20). For example,
seawaters that would equate to -10 liters), and the require- guanosine-5-triphosphate(GTP) and uridine 5 -triphosphate
ment for specialized analytical equipment (gas chromato- (UTP) are both required for the activation and intercon-
graph-mass spectrometer [GC-MS]) for the lipid profile version of carbohydrate precursors for polysaccharide
37. Microbial Nucleotide Fingerprints 871

biosynthesis; cytidine 5 -triphosphate (CTP), GTP, and ample, when is a sterile sampler absolutely necessary, and
UTP are required for RNA transcription; and the deoxy- when can that requirement be relaxed?
ribose derivatives of CTP, GTP, ATP, and thymidine 5- The number of samples that are necessary to accurately
triphosphate are required for DNA replication. In addition, describe microbial processes in a given ecosystem depends to
GTP is an obligate requirement for the initiation, the a certain extent on the structure of the habitat. Because most
aminoacyl-tRNA binding, and the translocation processes natural habitats are actually mosaics of many microhabitats
of protein synthesis. Unlike the ATP pool, which is main- with a heterogeneous distribution of microorganisms, vari-
tained at a relatively constant level independent of growth ance between multiple samples generally exceeds variance
rate, the intracellular concentrations of non-adenine NTPs between subsamples of a single collection. Therefore, replica-
fluctuate in direct proportion to their requirements for tion for microbial biomass and activity estimation is most
biosynthesis (12, 20, 47). Consequently, quantitative mea- meaningful when performed at the highest level (35).
surements of GTP and the estimation of total GTP:ATP ra- Unfortunately, this is rarely achieved. Automation in sample
tios of microbial communities in nature constitute a key collection and, especially, in sample analysis-including re-
component of the environmental nucleotide fingerprint. mote sensing-should greatly improve the current situation
Some previous studies of ATP in microorganisms and in with respect to the overall accuracy of ecosystem mea-
environmental samples have emphasized the futility of ex- surement.
trapolating these static pool measurements to estimates of
metabolic energy flux. It is the turnover rate of the ATP
pool, rather than the steady-state concentration, that varies 37.3. BIOMASS DETERMINATION BY ATP
in proportion to cellular metabolic energy requirements. ATP has several unique characteristics that make it a reli-
It follows, then, that direct measurements of ATP pool able indicator of microbial biomass in aquatic environ-
turnover, when coupled with independent estimates of ATP ments. It is ubiquitous in all living cells, has a relatively
pool size, should provide useful information on biological short half-life following cell death and autolysis, and is pres-
energy flux in cells, populations, or natural microbial as- ent at a fairly constant intracellular concentration regard-
semblages (26). Finally, the measurement of rates of RNA less of nutritional mode (e.g., photoautotroph, chemo-
and DNA synthesis, using the uptake and incorporation heterotroph, chemolithoautotroph, phagotroph) or growth
of specific radiolabeled precursors ( e.g.,3H-adenine, 3H- rate. Furthermore, particulate ATP (P-ATP) can be rapidly
thymidine, 32P/33P-phosphate)provides invaluable infor- and efficiently extracted from cells and stabilized in solu-
mation to the environmental nucleotide fingerprint (24, tion using boiling buffers, cold mineral or organic acids, or
27, 43). The correlations between nucleic acid synthesis, a variety of organic solvents (21). The preferred method of
protein synthesis, and cell growth are so universally ac- ATP quantification is the firefly bioluminescence reaction,
cepted that they lend themselves well to the study of com- but a variety of analytical techniques are available for either
plex microbial assemblages in nature. discrete sample or continuous flow analyses. Data on the P-
This chapter will focus on the most basic and most ATP content of a water, sediment, or soil sample can be ex-
widely used aspect of the environmental microbial nu- trapolated to total microbial biomass using C:ATP relation-
cleotide fingerprint, namely, the measurement of cellular ships derived from either laboratory or field studies. Other
ATP as a biomass indicator. The stepwise methodology that advantages include the low detection limit (less than lo-
is presented will focus on aquatic habitats. However, ATP -
mol; roughly equivalent to lo3 Escherichia coli cells) and
measurements have also been used for the analysis of soils acceptable level of precision for field replicates (typically 1
and in wastewater treatment and clinical applications (21). to lo%, depending on ATP concentration and operator),
The ATP extraction and analysis protocols will vary con- the high degree of objectivity compared with methods re-
siderably with sample type and expected microbial biomass. quiring operator recognition of live cells and estimation
Other features, including the ECA ratio, GTP:ATP ratio, of cell dimensions, and the potential for near real time
total energy flux determination by cellular ATP turnover, analyses and continuous flow applications. Finally, com-
and measurement of rates of RNA and DNA synthesis, will pared with most other biomass assays, there is also an ex-
be mentioned, but the detailed analytical procedures pub- tensive laboratory and field database for comparisons, con-
lished elsewhere (16, 23-25, 31, 32) will not be repeated clusions, and ecological interpretations.
here.
37.4. MATERIALS REQUIRED
37.2. SAMPLING A N D SUBSAMPLING THE
ENVIRONMENT 37.4.1. Equipment
Sampling is one of the most important aspects of quantita- Sampling gear (water column): Niskin bottles (or equiv-
tive microbial ecology, but sampling design, including fre- alent), Kevlar line or equivalent, subsampling bottles,
quency of sampling, replication, and other relevant issues, is 202-pm Nitex mesh
sometimes overlooked. All naturally occurring microbial Sampling gear (sediment column): corer device, core
communities are variable in space and in time, and the tubes, syringe samplers, glove box (optional)
number of samples typically collected in any single study is
usually small and, therefore inadequate for a complete de- Filtration gear: polyvinyl chloride (PVC) or stainless
scription of the habitat under investigation. To the extent steel manifold (3- or 6-place), equipped with glass filter
possible, multiple samples should be collected, covering as bases with stainless steel screens for 25-mm-diameter fil-
many microenvironments of the bulk habitat as possible to ters and large-volume funnels (100- to 200-ml capacity)
obtain a true representation of the ecosystem under investi- Vacuum pump with gauge (compressed Nzor air can be
gation. For relatively homogeneous aquatic environments used as an alternative to vacuum filtration)
this is straightforward, but for sediments and soils it is not. Block heater (Sybron Thermolyne Type (16500 Dri-bath,
The nature of the sampler itself is also important. For ex- or equivalent), capable of heating extraction menstruum
872 w COMMUNITY AND CENOMIC ANALYSIS

in test tubes to 100 2 1C (1 atm) and maintaining boil- evacuated bottles, syringe samplers, or any other effective
ing temperatures throughout the extraction period means. Selected habitats such as the sea-surface microlayer
Storage freezer ( - ZOOC) or high-temperature hydrothermal vents require the use of
pH meter and reference buffers specialized samplers.
Magnetic stirrer and stir bars 1. Prior to use, the samplers are cleaned with dilute HC1
Light-detection instrument: Any one of many commer- (0.5 to 1 M) or ethyl alcohol (95%) and rinsed thoroughly
cially available general instruments, such as fluorometers, with distilled water; sterilization of the samplers is neither
spectrophotometers, or liquid scintillation counters, is required nor practical for most field studies.
suitable for measuring light emission. However, to obtain 2. To measure microbial ATP, it is necessary to remove
efficient and reliable data with maximum sensitivity, a metazoans and other nonmicrobial ATP prior to sample ex-
specially designed ATP photometer is required. traction. This is done most conveniently by passing water
Instruments specifically designed for ATP analyses are samples through a 202-pm Nitex mesh as part of the sub-
marketed by several manufacturers. Whatever light- sampling procedures (e.g., during subsampling from Niskin
measuring device is selected, it is imperative to have either bottles). It is conceivable that in certain aquatic environ-
a strip-chart recorder, integrator, or suitable computer- ments this procedure may also remove large detrital par-
assisted data station to quantify light emission. ticles to which microorganisms are attached. In these
extreme cases, metazoans can be hand sorted from the re-
37.4.2. Supplies spective water samples before analysis.
3 . Following collection and subsampling, the particulate
Filters (24/25-mm-diameter Whatman GF/F, Millipore matter is concentrated and extracted as soon as possible.
HA/GS, or equivalent) Although numerous methods have been described for the
Filter forceps extraction of ATP from microorganisms (21), the most
Extraction and assay glassware: test tubes for ATP ex- commonly used method for aqueous samples involves P-
traction (approximately 15-mm diameter by 125- to ATP concentration by vacuum or pressure filtration and ex-
160-mm length); assay vials (18-mm diameter by 40- traction into boiling Tris (0.02 M, pH 7.4 to 7.7) or boiling
mm height). Size requirements may vary depending on phosphate (60 mM, pH 7.4) buffers. The latter is recom-
equipment/instruments used. mended for samples suspected of containing alkaline phos-
phatase, as would be expected in phosphorus-depleted habi-
* Adjustable automatic pipettes (0.1- to 1-ml and 1- to tats (28). After concentration of microbes onto a filter, the
5-ml capacities) filter is immersed into the boiling buffer as quickly as possi-
ble after the final portion of liquid passes through the filter.
37.4.3. Solutions and Reagents If left on the filtration manifold for an extended period,
(Unless otherwise indicated, catalog numbers or informa- measured in seconds, the cells desiccate, causing a loss of
tion refer to items sold by Sigma Chemical Company.) cellular ATP. It is imperative that the extraction buffer be
Firefly lantern extract (FLE-50) at boiling point at the time of filter insertion. At tempera-
tures below approximately 90C, inefficient extraction oc-
Potassium arsenate buffer, 100 mM, pH 7.4 curs due to enzyme-catalyzed ATP hydrolysis.
Tris(hydroxy)aminomethane-HC1 (TRIS) buffer, 20 4. After the filter is placed into the boiling buffer, the
mM, pH 7.75 sample is heated for an additional 5 min, during which time
ATP, sodium salt, 2 pM solution in Tris buffer, in 1- to the test tubes are partially covered to minimize evaporation
5-ml aliquots, and stored frozen and resultant volume changes.
Magnesium sulfate solution, 40 mM 5. Following extraction, the samples are removed from
Potassium phosphate buffer, 60 mM, pH 7.4 the heating block or temperature-controlled bath, cooled,
then stored frozen (-20OC) until assayed. At this point the
D-Luciferin, optional (L-9504) sample extracts are extremely stable, with ATP losses of less
H3P04 (1 to 1.5 M) than 1% per year in properly buffered solutions.
NaOH (1 and 0.1 M)
37.5.1.2. Sediment Column
37.5. PROCEDURES Many aquatic sediments are well stratified and character-
ized by steep depth gradients in microbial biomass.
37.5.1. Sampling, Subsampling, Extraction, and Consequently, it is imperative that the sampling and sub-
Sample Storage sampling methods used to collect sediment for microbial
ATP analysis preserve the unique depth distribution. For in-
37.5.1 . I . Water Column tertidal or shallow subtidal habitats, sediment cores can be
Typically, oceanic or lake samples are collected at prede- collected manually by inserting PVC or acrylic tubes (10-
termined depths throughout the water column using stan- to 15-cm diameter, 30- to 50-cm length) into the sediment
dard commercially available PVC bottles (e.g., General and placing stoppers on both ends prior to retrieval. Deeper
Oceanics Niskin or Go-Flo bottles, or equivalent) mounted samples require the use of a spade box corer (or equivalent
on a CTD-rosette sampler. In addition to obtaining com- device), which is designed to minimize both sediment dis-
plementary information on physical and chemical charac- ruption during sampling and winnowing during sample re-
teristics of the water column, this sampling protocol allows covery. Unfortunately, most gravity corers, which are easier
interactive, directed sampling at specific regions of interest to operate than box corers, create an unacceptable bow
(e.g., particle, fluorescence, or oxygen maxima or minima, wave prior to penetration into the sediment. This results in
density discontinuities, etc.). Alternatively, water can be a disruption of the microbial biomass gradients that are of
obtained using submersible pumps, manually operated greatest interest to the microbial ecologist. As for water
37. Microbial Nucleotide Fingerprints H 873

column samples, the greatest variability is expected to occur mixtures (i.e., cell extracts) into individual components
at the level of sample (i.e., sediment core) replication. This (ATP, ADP, AMP, etc.) that can be quantified during a sin-
variability is also of greatest relevance to the microbial ecol- gle sample run. This additional information on the concen-
ogist. trations of non-ATP nucleotides can provide useful data on
Once collected, the core samples should be processed as the metabolic states and in situ growth rates of microbial
quickly as possible and, as discussed above, with care taken communities in nature (21, 22). The more commonly used
to minimize changes in environmental conditions. For most bioluminescence assay, however, has a much lower ATP de-
anoxic sediments, this requires the use of a Nz-filled glove tection limit, is straightforward and inexpensive to perform,
box to prevent sample oxidation and subsequent transitions has a high level of precision, and requires less-specialized in-
in intracellular ATP pools or potential loss of obligate strumentation. Furthermore, non-ATP nucleotides (e.g.,
anaerobe viability. ADP, AMP, GTP, etc.) can also be measured by the firefly
A variety of procedures have been described for core sub- bioluminescence reaction following stoichiometric genera-
sampling, including techniques for the collection of the tion of ATP from other nucleotide triphosphates via spe-
water-sediment interface, where microbial biomass and ac- cific transphosphorylation reactions, as discussed later in
tivity are expected to be the highest (45), and for the this chapter.
preservation of millimeter-scale habitat variability (8). If re- Several reviews have been published concerning the
quired, the sediment samples can be screened to remove specificity, kinetics, and mechanism of the firefly biolumi-
ATP-containing metazoa, macroalgae, or higher plant rhi- nescence reaction. The postulated steps are (10):
zomes prior to analysis. The replicated subsamples from the
Mg+-luciferase
different depth strata are then mixed thoroughly with a LH2 (luciferin) + ATP E-LHZ-AMP
spatula in preparation for final subsampling for ATP extrac-
tion. + PPi (1)
neutral pH
1. Triplicate subsamples (1 to 2 cm3) of each sediment E-LH2-AMP + O2A oxyluciferin + E + COz
fraction are collected using a 3-ml plastic syringe barrel
( h e r lock end removed). Additional replicates are also
+ AMP + light (2)
taken for the determination of wet-volume-to-dry-weight When all necessary reactants are present in excess, the in
conversion and for other bulk chemical parameters (e.g., vitro light emission is directly proportional to the concen-
total carbon). tration of ATP in solution. Reaction kinetics and speci-
2. The plugs of sediment are immediately discharged ficity depend on the purity of the enzyme preparation; sen-
into test tubes containing 10 ml of cold H3P04 (0.5 M, sitivity is controlled by luciferin concentration (30). The
4C), capped, and thoroughly mixed. Additional subsam- measurement of either the initial rise of the luminescence
ples should be prepared with a known amount of ATP curve (0 to 2 s), the peak height of luminescence (0 to 5 s),
added as an internal standard to assess and correct for ATP or a predetermined integrated portion (e.g., 15 to 75 s) of
losses by the combined effects of adsorption, hydrolysis, and the light emission decay curve can be used to relate ATP
various potential sources of chemical interference. Rep- concentrations in reference standards to those in the un-
resentative sediment porewaters should also be collected known sample extracts. The major advantages of the inte-
(by centrifugation or pressure filtration through a 0.2-km- grated mode are increased sensitivity, ease and reliability of
pore-size filter) to assess the potential interference due to mixing, and nonreliance on the peak-height response,
dissolved ATP (D-ATP). If the sediment samples contain which is difficult or impossible to measure with certain in-
CaC03, occasional venting to release accumulated C02gas struments. However, a major disadvantage of the inte-
may be required. grated mode is the nonspecificity of light emission with
3. After an extraction period of 15 to 20 min at 4C, the certain crude enzyme preparations (20). Reliability of peak-
extracted nucleotides are separated from the solid phase by height analyses depends on a very rapid and complete mix-
centrifugation or vacuum filtration. ing of all reactants. This is best accomplished using an
4. The pH of the acid extracts is adjusted to 7.4 by titra- automatic injection system, which ensures consistent mix-
tion with NaOH (1.0 and 0.1 M). At this point, the ATP is ing velocities for all samples. The peak-height mode of
stable in samples stored at -20C. If the ATP concentra- analysis offers the advantages of speed of assay and mini-
tion in the acid extract is > 100 nM, the sample can be di- mum interference from other enzymes or substrates (e.g.,
luted with 60 mM PO4buffer as an alternative to base titra- non-ATP nucleotides) that may affect the rate of the
tion. luciferase-catalyzed reaction.
5. If the ATP concentration is 5 1 nM, the acid extract 1. Firely lantern extract (catalog FLE-50 or FLE-250;
must be concentrated by either the activated charcoal pro-
Sigma Chemical Co., St. Louis, MO) is stored with desic-
cedure (14) or brucite coprecipitation (4,34) prior to analy-
cant at -20C. To activate lyophilized FLE, a 50-mg vial is
sis. At this point, the ATP is relatively stable and the sam-
hydrated in 5 ml of distilled water. This enzyme preparation
ples may either be stored at 4C for up to 2 to 3 weeks or
is allowed to age at room temperature for a minimum of 4
processed immediately.
to 6 h to a maximum of 24 h, depending on the desired sen-
sitivity. During the aging process, endogenous ATP initially
37.5.2. Detection of ATP by Firefly present in the crude extract is consumed, thereby decreas-
Bioluminescence ing the background light emission. Because of variations
Although ATP (and other adenine and nonadenine nu- among individual enzyme preparations, it is imperative that
cleotides) can be measured using any one of a variety of an- only a single batch of enzyme be used for the analysis of a
alytical detection systems, the firefly bioluminescence assay given set of samples and ATP standards.
and high-performance liquid chromatography (HPLC) are 2. Next, the hydrated, aged FLE is further diluted using
most commonly used in ecological studies. A major advan- equal volumes (generally 10 ml each for a single 50-mg vial
tage of HPLC is the ability to separate complex nucleotide of FLE) of MgSO, (0.04 M) and KHAs04 buffer (0.1 M,
874 COMMUNITY AND CENOMIC ANALYSIS

pH 7.4), and the mixture is incubated at room temperature C:ATP ratio when microorganisms are starved for phospho-
for 1 h. rus (21, 22). However, under most conditions found in na-
3. If desired, the FLE preparation can be diluted to a ture, the C:ATP ratio of the microbial community is about
greater final volume depending on the required sensitivity 250:l. Although originally developed to estimate total mi-
of the assay. However, if a single vial of FLE-50 is diluted to crobial biomass, ATP concentrations are, in theory, more
a working volume of >50 ml, the bioluminescence reaction closely coupled to protoplasm biomass and, more specifi-
can become limiting for luciferin, which must be added cally, total biovolume. Because of the obligate role of ATP
back to maintain first-order reaction kinetics with respect in cellular bioenergetics, intracellular ATP levels are care-
to ATP (30). fully maintained at a concentration of approximately 1 to 2
4. Immediately before use, the insoluble residue is re- mM (6). Unfortunately, it is difficult to use biovolume esti-
moved by vacuum filtration (Whatman GF/F filter) or cen- mates directly, however accurate, in most studies of micro-
trifugation (1500 X g, 5 min). bial ecology. Consequently, one must rely on empirically
determined C:ATP ratios to extrapolate P-ATP determina-
37.5.3. ATP Standards tions to estimates of total microbial biomass. In so doing,
For each enzyme preparation, an ATP standard curve is pre- both the level of precision and the accuracy of the initial
pared and analyzed with the sample extracts. ATP determination are decreased. Furthermore, in habitats
where copious amounts of capsular materials, extracellular
1. A stock ATP solution containing 1 to 2 p M ATP is secretions, or slimes occur ( 7 ) , ATP-based values of total
prepared in TRIS buffer (0.02 M, pH 7.4 to 7.7) and stored biomass probably provide only minimum estimates.
at - 20C in 1-ml aliquots until needed. Under these con-
ditions, ATP hydrolysis is <1% per year. The exact con-
centration of the stock solution is determined by absorption 37.6. NOTES AND COMMENTS
spectrophotometry at 259 nm, using the relationship
37.6.1. Sampling
- - and Subsampling. -
A = cle (3)
Exposure of metabolically active microorganisms to envi-
where c = ATP (M); 1 = absorption pathlength (cm); and ronmental conditions that are substantially different from
e = 15.4 X lo3 (ATP molar extinction coefficient at pH the collection site should be avoided, to minimize short-
7.4). term transitions in intracellular ATP pools. However, this
2. A working ATP standard solution is prepared on the becomes nearly impossible during the collection of many
day of the assay by diluting the stock ATP preparation with samples, for example, abyssal water samples from the equa-
the appropriate extraction menstruum (TRIS or phosphate torial ocean. Decreases in pressure and increases in temper-
buffer depending on sample type, and preferably the same ature, even during the time required for the samples to
batch as used for sample extraction). reach the oceans surface, are almost certain to alter micro-
3. Between seven and eight ATP standards (including a bial ATP concentrations and perhaps even cell viability.
buffer blank) covering the expected range of the sample However, at the present time these effects have not been
concentrations (approximately 1 to 100 nM ATP) are pre- systematically evaluated. In the future, a technique that
pared. If 2 1 0 0 samples are analyzed or if the analysis time provides for the in situ extraction of ATP from microorgan-
exceeds approximately 1 h, it is desirable to measure a set of isms needs to be developed and compared with conven-
standards at the beginning and at the end of the analysis to tional sampling procedures in order to provide quantitative
monitor temporal changes in the response of the FLE prepa- constraints on the potential changes in P-ATP during rou-
ration. Otherwise, a single standard curve measured midway tine sample recovery.
through the experiment is sufficient. In general, no signifi-
cant hydrolysis of the diluted ATP standard solutions occurs 37.6.2. Extraction
during a typical 3- to 4-h working period. The concentration step, which is necessary for most
4. Following a single use, all thawed ATP stock standards low-biomass environments (i.e., < I g of C m-3), must be
and serial dilutions thereof should be discarded. performed with extreme care; sample size is especially im-
portant. It has been shown that the recovery of microbial
37.5.4. Data Reduction and Extrapolations of ATP ATP from certain high-particulate-load samples is volume
to Biomass dependent (49). Initially, the observed P-ATP losses were
The relationship between ATP concentration and light thought to be the result of cell lysis during prolonged filtra-
emission is linear provided substrate (0, and luciferin) lim- tion. However, it is now known to be caused by a filtration-
itation does not occur. Peak heights or integrated areas are induced metabolic stress that results in the hydrolysis of
regressed on ATP standard concentrations. From a model I ATP to ADP and AMP (31). Consequently, if the total
linear regression analysis of these data, the ATP concentra- +
adenine nucleotide pool (the sum of ATP ADP AMP) +
tions in sample extracts can be calculated. By correcting for rather than ATP is measured, filtration volume becomes less
the proportion of the sample actually assayed and the vol- critical.
ume of medium originally extracted, the ATP per liter of In certain eutrophic habitats where total microbial bio-
the original water or sediment sample can be determined. mass exceeds 1 g of C mp3, water samples can be extracted
The C:ATP ratio in microorganisms varies considerably, directly, thereby eliminating the preextraction concentra-
although somewhat predictably, among taxa and even for a tion step (17). It is imperative, however, that the sample
given species when grown under different nutritional con- volume injected does not exceed 5% (by volume) of the
straints (data summarized by Karl [21]). Among the most extractant volumes; otherwise, temperature changes may af-
conspicuous differences in C:ATP ratios are those observed fect the efficiency of ATP extraction. If desired, multiple in-
between unicellular microorganisms (i.e., bacteria and mi- jections can be made, allowing time between single injec-
croalgae: C:ATP = 200 to 350, by weight) and micrometa- tions for the extraction menstruum to return to its boiling
zoa (C:ATP = 50 to 150) and the large increases in the temperature. Furthermore, if direct injection is employed
37. Microbial Nucleotide Fingerprints 875

for P-ATP extraction, it is necessary to measure, and correct the rates of protein and nucleic acid biosyntheses, nu-
for, dissolved ATP (operationally defined as passing through cleotide metabolism, metabolic activity, and growth.
a 0.2-Fm-pore-size filter), which is also present in most ma- Furthermore, Karl and Bossard (26) developed a method to
rine (2,44) and freshwater (41) ecosystems. estimate the turnover rates of ATP to quantify energy flux
in natural populations of marine microorganisms. This ex-
37.6.3. Sensitivity, Precision, and Accuracy panded approach of nucleotide fingerprinting is strongly
The sensitivity of the ATP assay is determined by the in- recommended for field investigations to obtain corrobora-
strumentation used to detect light emission and by the pu- tive data relating to the in situ physiological and growth
rity of luciferase and luciferin concentration in the enzyme states of microbial assemblages in nature.
preparation. Using the crude FLE-50 luciferase mixture pre- To calculate the mean cellular ECAratio, measurements
pared as described in this chapter and commercially avail- of ADP and AMP must be made in addition to ATP deter-
able ATP photometers, the lower limit of ATP detection mination, as described above. ADP and AMP are both
(i.e., twice the background light emission) is about 0.2 nM quantitatively coextracted with ATP, so the methods de-
ATP. For greater sensitivity, exogenous luciferin is added to scribed above for sample collection and initial processing
the enzyme preparations, enabling the detection of 1 pM are identical to those for ATP determination. After the ex-
ATP (30). The precision of the peak height assay procedure traction of cellular nucleotides, ADP and AMP are quanti-
as routinely performed is + I to 2% of the mean (n = 8) tatively converted to ATP by stepwise enzymatic reactions
throughout the entire range of ATP standards. Accuracy is involving pyruvate kinase and adenylate kinase plus pyru-
estimated by analyzing diluted ATP standards and treating vate kinase, respectively. ATP is measured again in each
them as unknown samples. At the present time, no com- sample, and ADP and AMP concentrations are calculated
mercially available certified reference materials are avail- by difference (16).
able for independent determination of accuracy. For GTP concentration measurements, the ATP and
UTP present in the nucleotide extracts are first destroyed
37.6.4. Analytical Interferences and Use of Internal by specific enzymatic reactions involving the addition of
Standards hexokinase, glucose-6-phosphate dehydrogenase, and
In addition to the potential problems discussed in the pre- UDP-glucose pyrophosphorylase. The remaining NTPs,
vious sections, several sources of analytical interference are mainly GTP and CTP, are separated kinetically in the light
possible. These include (i) the presence of inorganic and or- emission reaction involving firefly luciferase ( 18-20).
ganic ions in the sample extracts, resulting in loss of ATP in For the determination of ATP and total adenine nu-
solution (i.e., through chelation) or in decreased luciferase cleotide (TAN) pool turnover rates as estimates of energy
activity; (ii) the presence of humic acid-like substances in flux and specific growth rate of microbial populations in na-
the sample extracts that impart a yellow color to the solu- ture, an environmental sample is incubated with radioactive
tion, thereby resulting in attenuation of the emitted light; phosphate (32P- or 33P-P04), as a tracer for P-flux through
(iii) turbidity of the final extracts, resulting in light scatter- the acid anhydride-bound P groups of ATP (P-P and y-P)
ing and absorption; (iv) the presence of a high concentra- and into the a-P position as adenine nucleotide molecules
tion of inorganic particulate material in the final extracts, are removed for net growth (27). With increasing incuba-
resulting in loss of ATP through adsorption; and (v) the tion time, the three phosphate moieties of the community
presence of contaminating enzymes, in either the sample ATP pool (or isolated fraction thereof) become labeled, al-
extracts or the luciferase preparation, that compete with lu- beit at different rates. Experimental work has demonstrated
ciferase for the ATP in solution (e.g., ATPase or adenylate that the kinetic constants of y-P and p-P labeling are indis-
kinase) or that result in the production of ATP through tinguishable, both in laboratory cultures and in field sam-
transphosphorylase reactions (e.g., nucleoside diphosphate ples, because of the rapid intracellular interconversions of
kinase or pyruvate kinase) . ADP and ATP. Because both anhydride bonds have identi-
Most of the preceding sources of error are detected, and cal free energies of hydrolysis, this simplifies the measure-
corrected for, through the use of an ATP internal standard, ment of total metabolic energy flux (5). By comparison, the
as discussed by Strehler (48). The internal standard may a-P labeling lags behind the y-P and p-P moieties because
be added in the form of an ATP salt solution, as live or the a-P position of ATP becomes labeled only as a result of
lyophilized bacterial cells, or as radiolabeled ATP. To mini- the net removal of adenine nucleotides for biosynthesis, the
mize the effects of ionic interference, it is imperative that sum of salvage and de novo synthesis processes. The exact
the standard ATP solutions be prepared in an ionic medium turnover time of the ATP or the TAN pool is determined by
identical with that of the samples. Peak-height mea- an analysis of the time rate of change in the specific activity
surements significantly decrease the analytical interference of either the y-P, P-P, or a-Ppositions of ATP. Labeling of
due to the presence of nonadenine nucleotide triphosphates the individual positions is determined by selective hydroly-
and therefore are strongly recommended. A review of ana- +
sis of ATP to ADP Pi. Since the y-P radioactivity is equal
lytical issues concerned with ATP extraction efficiency &
to th; p-P radioactiyity, a-P can calculated by difference:
from soils has recently appeared (42). (a-P = [total ATP ] - [2 x y-P I).
As predicted by radiotracer theory, the change in the
ATP or TAN pool-specific radioactivity (SA) (nanocuries
37.7. ENHANCING THE MICROBIAL per picomole) following the addition of an appropriate ra-
NUCLEOTIDE FINGERPRINT dioactive precursor is an exponential function of incubation
The basic method of ATP measurement as an indicator of time (t). The initial S A (at t = 0) is zero, and the relative
total microbial biomass has given rise to the more compre- SA (at isotopic equilibrium with precursor pool) is 1.000.
hensive protocol of environmental nucleotide fingerprint- Under these conditions, the decimal equivalent of SA at
ing (21). The development of specific and sensitive tech- any time (SA,) can be described by the equation
niques for quantitative measurements of selected non-ATP
intracellular nucleotides has enabled researchers to estimate
876 COMMUNITY AND GENOMIC ANALYSIS

where N is equal to the number of turnover cycles (N is di- about the phylogenetic and metabolic diversity of microor-
mensionless) observed during the incubation period. ATP ganisms in nature and the probable presence of novel, as yet
or TAN pool turnover time (T) can then be calculated from uncultivatable microbes, there is a renewed interest in mi-
the expression crobial ecology and in whole-community assays. Despite re-
cent and significant progress, the field of microbial ecology
T = t/N (2) is still methods-limited with regard to the most funda-
This relationship is applicable for all incubation periods ( t ) mental properties of natural microbial assemblages, namely,
less than or equal to 5 N , at which time the pools are ex- biomass and metabolic activity estimation of the total pop-
pected to be near isotopic equilibrium and, in theory, would ulation. One of the most significant challenges for the fu-
not change until the exogenous radioactive precursor pool ture is to link microbial phylogeny to biogeochemistry and
is exhausted. For oligotrophic surface seawaters, a 3- to 4-h ecology to gain a better understanding of material and en-
incubation period is sufficient to reach isotopic equilibrium ergy flow in the biosphere.
(5).
Once the turnover time (T) of the ATP or TAN pool
has been determined, it is straightforward to extrapolate 37.9. REFERENCES
these data to the more meaningful ecological parameters of 1. Atkinson, D. E. 1969. Regulation of enzyme function.
energy flux and specific growth rate. Assuming a value of Annu. Rew. Microbiol. 23;47-68.
- 11 ( 2 1) kcal mol- for the free energy of ATP hydrolysis 2. Azam, F., and R. E. Hodson. 1977. Dissolved ATP in the
for either the p-P and y-P positions under in vivo condi- sea and its utilization by marine bacteria. Nature 267:696-
tions (54), then the total microbiological energy flux (EF, 698.
expressed in units of kilocalories per liter of sample per 3. Bertilsson, S., 0. Berglund, D. M. Karl, and S. W. Chis-
hour) can be calculated as holm. 2003. Elemental composition of marine Prochloro-
coccus and Synechococcus: implications for the ecological
stoichiometry of the sea. Limnol. Oceanogr. 48:1721-1731.
where [ATP] is equal to the total particulate ATP pool in 4. Bjorkman, K., and D. M. Karl. 2001. A novel method for
moles per liter- of sample and TATp is ATP pool turnover the measurement of dissolved adenosine and guanosine
time (h). This value for total available free energy can be triphosphate in aquatic habitats: applications to marine
compared directly to the solar energy flux, to the energy microbial ecology. J. Microbiol. Meth. 47:159-167.
5. Bossard, P., and D. M. Karl. 1986. The direct mea-
stored by photosynthesis, or to respiration. In this way, car- surement of ATP and adenine nucleotide pool turnover in
bon and energy fluxes can be directly compared. microorganisms: a new method for environmental assess-
Extrapolation of the measured TAN turnover time ment of metabolism, energy flux and phosphorus dynam-
(TTAN) to specific growth rate ( p ) is based on the theoreti- ics. J. Plankton Res. 8:l-13.
cal predictions (6) and empirical observations (33) that the 6. Chapman, A. G., and D. E. Atkinson. 1977. Adenine
TTAN is equivalent to a value that is 2 to 3% of the genera- nucleotide concentrations and turnover rates. Their corre-
tion time (i.e., TAN pool turns over, on average, 40 times lation with biological activity in bacteria and yeast. Adw.
per generation). Consequently, the doubling time (Td, ex- Microb. Physiol. 15:253-306.
pressed in hours) of the substrate-responsive population is 7. Costerton, J. W., R. T. Irvin, and K. J. Cheng. 1981. The
bacterial glycocalyx in nature and disease. Annu. Rew.
Microbiol. 35:299-3 24.
and specific growth rate (p,expressed in units per hour) is 8. Craven, D. B., R. A. Jahnke, and A. F. Carlucci. 1986.
Fine-scale vertical distributions of microbial biomass and
activity in California Borderland sediments. Mar. Biol. 83:
These kinetic model formulations will provide ideal re- 129- 139.
sults when all of the radiotracer-responsive microorganisms 9. Cuhel, R. L., and J. B. Waterbury. 1984. Biochemical
in a given habitat are growing at identical rates. A novel, composition and short term nutrient incorporation pat-
statistical treatment of field data, described in detail by terns in a unicellular marine cyanobacterium, Synecho-
Laws et al. (37), provides a method for determining the co- coccus (WH7803). Limnol. Oceanogr. 29:370-374.
10. DeLuca, M., and W. D. McElroy. 1978. Purification and
efficient of variation among the individual estimates of
properties of firefly luciferase. Methods Enzymol. 57:3-15.
TATp and TTAN, Consequently, one can quantify the vari-
11. Dobbs, F. C., and R. H. Findlay. 1993. Analysis of mi-
ability among individual subcomponents of the microbial crobial lipids to determine biomass and detect the response
community. Although applied here for the turnover of ATP of sedimentary microorganisms to disturbance, p. 347-358.
and TAN pools, this mathematical formulation can be used In P. F. Kemp, B. E Sherr, E. B. Sherr, and J. 1. Cole (ed.),
to assess heterogeneity in the turnover rate of any intracel- Handbook of Methods in Aquatic Microbial Ecology. Lewis
lular pool for which a suitable radioactive or stable isotopic Publishers, Boca Raton, FL.
precursor exists. 12. Franzen, J. S., and S. B. Binkley. 1961. Comparison of
the acid-soluble nucleotides in Escherichia coli at different
37.8. CONCLUSION growth rates. J. Biol. Chem. 236515-519.
13. Guckert, J. B., C. P. Antworth, P. D. Nichols, and D. C.
No single approach to the study of microorganisms in their White. 1985. Phospholipid, ester-linked fatty acid profiles
natural environments is universally accepted, or acceptable as reproducible assays for changes in prokaryotic commu-
(22). Early in the development of microbiology as a scien- nity structure of estuarine sediments. FEMS Microbiol.
tific discipline, specific groups of microorganisms were iso- Ecol. 31:147-158.
lated from nature and studied in the laboratory. This led to 14. Hodson, R. E., 0. Holm-Hansen, and F. Azam. 1976.
major advances in microbiology, especially physiology, ge- Improved methodology for ATP determination in marine
netics, and metabolism. Now, armed with new information environments. Mar. Biol. 34:143-149.
37. Microbial Nucleotide Fingerprints w 877

15. Holm-Hansen, O., and C. R. Booth. 1966. The mea- Handbook of Phycological Methods, Vol. 111. Physiological and
surement of adenosine triphosphate in the ocean and its Biochemical Methods. Cambridge University Press, New
ecological significance. Limnol. Oceanogr. 11510-519. York, NY.
16. Holm-Hansen, O., and D. M. Karl. 1978. Biomass and 32. Karl, D. M., and 0. Holm-Hansen. 1978. Methodology
adenylate energy charge determination in microbial cell and measurement of adenylate energy charge ratios in en-
extracts and environmental samples. Methods Enzymol. 57: vironmental samples. Mar. Biol. 48:185-197.
73-85. 33. Karl, D. M., D. R. Jones, J. A. Novitsky, C. D. Winn,
17. Jones, J. G., and B. M. Simon. 1977. Increased sensitiv- and P. Bossard. 1987. Specific growth rates of natural mi-
ity in the measurement of ATP in freshwater samples with crobial communities measured by adenine nucleotide pool
a comment on the adverse effect of membrane filtration. turnover. J. Microbiol. Meth. 6:221-235.
Freshwater Biol. 7:253- 260. 34. Karl, D. M., and G. Tien. 1992. MAGIC: a sensitive and
18. Karl, D. M. 1978. A rapid sensitive method for the mea- precise method for measuring dissolved phosphorus in
surement of guanine ribonucleotides in bacterial and envi- aquatic environments. Limnol. Oceanogr. 37:105-116.
ronmental extracts. Anal. Biochem. 89581-595. 35. Kirchman, D., J. Sigda, R. Kapuscinski, and R. Mitchell.
19. Karl, D. M. 1978. Determination of GTP, GDP and GMP 1982. Statistical analysis of the direct count method for
in cell and tissue extracts. Methods Enzymol. 57:85-94. enumerating bacteria. Appl. Environ. Microbiol. 44:376-
20. Karl, D. M. 1978. Occurrence and ecological significance 382.
of GTP in the ocean and in microbial cells. Appl. Environ. 36. Koch, A. 1994. Growth measurement, p. 248-277. In P.
Microbiol. 36:349-355. Gerhardt, R. G. E. Murray, W. A. Wood, and N. R. Krieg
21. Karl, D. M. 1980. Cellular nucleotide measurements and (ed.), Methods for General and Molecular Bacteriology. ASM
applications in microbial ecology. Microbiol. Rev. 44:739- Press, Washington, DC.
796. 37. Laws, E. A., D. Jones, and D. M. Karl. 1986. Method for
22. Karl, D. M. 1986. Determination of in situ microbial bio- assessing heterogeneity in turnover rates within microbial
mass, viability, metabolism and growth, p. 85-176. In J. S. communities. Appl. Enuiron. Microbiol. 52:866-874.
Poindexter and E. R. Leadbetter (ed.), Bacteria in Nature, 38. Levin, G. V., J. R. Clendenning, E. W. Chappelle, A. H.
vol. 2. Methods and Special Applications in Bacterial Ecology. Heim, and E. Rocek. 1964. A rapid method for detection
Plenum Press, New York, NY. of microorganisms of ATP assay. BioScience 14:37-38.
23. Karl, D. M. 1993. Adenosine triphosphate (ATP) and 39. Lipmann, F. 1941. Metabolic generation and utilization of
total adenine nucleotide (TAN) pool turnover rates as phosphate bond energy. Adv. Enzymol. Relat. Areas Mol.
measures of energy flux and specific growth rate in natural Biol. 1:99-162.
populations of microorganisms, p. 483-494. In P. F. Kemp, 40. Madsen, E. L. 1998. Epistemology of environmental mi-
B. F. Sherr, E. B. Sherr, and J. J. Cole (ed.), Current crobiology. Environ. Sci. Tech. 32429439.
Methods in Aquatic Microbial Ecology. Lewis Publishers, 41. Maki, J. S., M. E. Sierszen, and C. C. Remsen. 1983.
Boca Raton, FL. Measurements of dissolved adenosine triphosphate in Lake
24. Karl, D. M. 1993. Microbial RNA and DNA synthesis de- Michigan. Can. J. Fish. Aquat. Sci. 40542-547.
rived from the assimilation of [2-3H]adenine, p. 471-481. 42. Martens, R. 2001. Estimation of ATP in soil: extraction
In P. F. Kemp, B. E Sherr, E. B. Sherr, and J. J. Cole (ed.), methods and calculation of extraction efficiency. Soil Biol.
Current Methods in Aquatic Microbial Ecology. Lewis Pub- Biochem. 33:973-982.
lishers, Boca Raton, FL. 43. Moriarty, D. J. W. 1986. Measurement of bacterial growth
25. Karl, D. M. 1993. Total microbial biomass estimation de- rates in aquatic systems using rates of nucleic acid synthe-
rived from the measurement of particulate adenosine-5- sis. Adw. Microbiol. Ecol. 9:245-264.
triphosphate, p. 359-368. In P. F. Kemp, B. F. Sherr, E. B. 44. Nawrocki, M. P., and D. M. Karl. 1989. Dissolved ATP
Sherr, and J. J. Cole (ed.), Current Methods in Aquatic turnover in the Bransfield Strait, Antarctica, during a
Microbial Ecology. Lewis Publishers, Boca Raton, FL. spring bloom. Mar. Ecol. Prog. Ser. 57:3544.
26. Karl, D. M., and P. Bossard. 1985. Measurement and sig- 45. Novitsky, J. A. 1983. Heterotrophic activity throughout
nificance of ATP and adenine nucleotide pool turnover a vertical profile of seawater and sediment in Halifax
in microbial cells and environmental samples. J. Microbiol. Harbor, Canada. Appl. Environ. Microbiol. 45:1761-
Meth. 3:125-139. 1766.
27. Karl, D. M., and P. Bossard. 1985. Measurement of mi- 46. Pinkart, H. C., D. B. Ringelberg, Y. M. Piceno, S. J.
crobial nucleic acid synthesis and specific growth rate by Macnaughton, and D. C. White. 2002. Biochemical ap-
32P04and [3H]adenine: field comparison. Appl. Environ. proaches to biomass measurements and community struc-
Microbiol. 50:706-709. ture analysis,p. 101-1 13. In C. J. Hurst, R. L. Crawford, G.
28. Karl, D. M., and D. B. Craven. 1980. Effects of alkaline R. Knudsen, M. J. McInemey, and L. D. Stetzenbach (ed.),
phosphatase activity on nucleotide measurements in Manual of Environmental Microbiology, 2nd ed. ASM Press,
aquatic microbial communities. Appl. Environ. Microbiol. Washington, DC.
40549-561. 47. Smith, R. J. 1979. Increasing guanosine 3-diphosphate
29. Karl, D. M., and F. C. Dobbs. 1998. Molecular ap- 5-diphosphate concentration with decreasing growth rate
proaches to microbial biomass estimation in the sea, p. 29- in Anacystis nidulans. J. Gen. Microbiol. 113:403-405.
89. In K. E. Cooksey (ed.), Molecular Approaches to the 48. Strehler, B. L. 1968. Bioluminescence assay: principles
Study of the Ocean. Chapman & Hall, London, United and practice. Meth. Biochem. Anal. 16:99-181.
Kingdom. 49. Sutcliffe, W. H., Jr., E. A. Orr, and 0. Holm-Hansen.
30. Karl, D. M., and 0. Holm-Hansen. 1976. Effects of lu- 1976. Difficulties with ATP measurements in inshore wa-
ciferin concentration on the quantitative assay of ATP ters. Limnol. Oceanogr. 21:145-149.
using crude luciferase preparations. Anal. Biochem. 75: 50. White, D. C., R. J. Bobbie, S. J. Morrison, D. K.
100-112. Oosterhof, C. W. Taylor, and D. A. Meeter. 1977.
31. Karl, D. M., and 0. Holm-Hansen. 1978. ATP, ADP and Determination of microbial activity of estuarine detritus
AMP determinations in water samples and algal cultures, by relative rates of lipid biosynehtsis. Limnol. Oceanogr.
p. 197-206. In J. A. Hellebust and J. S. Craigie (ed.), 22: 1089-1099.
878 COMMUNITY AND GENOMIC ANALYSIS

51. White, D. C., W. M. Davis, J. S. Nickels, J. D. King, and 53. Wiebe, W. J., and K. Bancroft. 1975. Use of adenylate en-
R. J. Bobbie. 1979. Determination of sedimentary micro- ergy charge ratio to measure growth rate of natural micro-
bial biomass by extractible lipid phosphate. Oecologia bial communities. Proc. Natl. Acud. Sci. USA 72:2112-
(Berlin) 4.051-62. 2115.
52. White, D. C., H. C. Pinkart, and D. B. Ringelberg. 54. Wilson, D. F., M. Stubbs, R. L. Veech, M. Erecinska,
1997. Biomass measurements: Biochemical approaches, and H. A. Krebs. 1974. Equilibrium relations between the
p. 91-101. In C. J. Hurst, G. R. Knudsen, M. J. McInemey, oxidation-reduction reactions and the adenosine triphos-
L. D. Stetzenbach, and M. V. Walter (ed.), Manual of phate synthesis in suspensions of isolated liver cells.
Environmental Microbiology. ASM Press, Washington, DC. Biochem. 1. 14057-64.
Construction of BAC and Fosmid libraries from
Naturally Occurring Microbial Populations
EDWARD E DELONG

38.1. CULTIVATION-INDEPENDENT APPROACHES I N MICROBIAL


ECOLOGY A N D POPULATION BIOLOGY ..................... 879
.2. CONSTRUCTION OF LARGE INSERT GENOMIC LIBRARIES FROM
ENVIRONMENTAL SAMPLES: GENERAL CONSIDERATIONS ..... 879
............. 879
38.2.1. Microbial Sampling, Still a Challenging Enterprise
......... 881
.2. Large-Insert D N A Libraries: Contemporary Approaches
......................... 882
.3. Library Screening and Sequencing
38.3. PROTOCOLS ............................................ 882
...... 882
38.3.1. BAC Libraries from Mixed Microbial Populations: Protocol
..... 883
.2. Fosmid Library Construction by Blunt-End Cloning: Protocol
38.4. CONCLUSION AND FUTURE PROSPECTS .................... 884
.5. REFERENCES ............................................ 884

38.1. CULTIVATION-INDEPENDENT sequenced (about 75%) originate from clinically important


APPROACHES IN MICROBIAL ECOLOGY microbes (4). Of the total prokaryotic sequences, about 90%
AND POPULATION BIOLOGY are from the domain Bacteria. To date, most full microbial
genome sequences have originated from pure cultures, al-
Most definitive microbiological studies have been con- though a few are derived from obligately symbiotic or para-
ducted in laboratories using pure cultures. This approach
sitic bacteria.
has been essential for the development of the science of mi- A powerful extension of the tremendous genome-
crobiology and remains a methodological mainstay. No sequencing capacity available today aims to describe ge-
methodology is perfect, however, and it has long been rec- nomically the natural world of indigenous microorganisms.
ognized that many microbial species still resist isolation in
Using the same logic as was applied in rRNA gene surveys
pure culture, for many and varied reasons. One interest of
contemporary microbiologists has therefore been to devise
(9), it has now become theoretically possible, using ge-
nomics, to access large portions of the genomes of all natu-
better methods to identify, characterize, and understand
rally occurring microbes, cultivated or not. This really is
those naturally occurring microbes that are difficult or im- just an extension of Norm Paces approach, but with a
practical to cultivate and study by traditional methods. twist. Instead of analyzing just a single genetic locus like
This chapter discusses one potential approach involving
rRNA, however, it has now become possible to recover and
genomic characterization of the naturally occurring micro-
analyze large portions of the genomes of naturally occurring
bial genomes, without the requirement of isolating individ-
microbes, cultivated or not (Fig. 1) (3, 11, 13, 14, 18).
ual microbes one by one in pure culture.
There are a variety of potential strategies for developing
Norman Pace and colleagues invented molecular phylo-
and applying the environmental genomics (14, 18) or
genetic cultivation-independent approaches to study natu-
metagenomics ( 12) approach. This chapter will describe
ral microbial populations in the mid-1980s (8-10). Paces some of the methodological options and considerations and
strategy exploits nucleic acid-based cloning and sequencing
present a few of the protocols that have been used success-
to access naturally occurring microbial diversity. The de-
fully for preparing genomic libraries from natural microbial
velopment of these approaches has led to the now-common
assemblages.
practice of cloning rRNA genes from mixed microbial as-
semblages. These cultivation-independent rRNA gene sur-
veys have revealed a significant number of new phyloge-
netic lineages, including abundant types of bacteria and 38.2. CONSTRUCTION OF LARGE
archaea previously unknown to science. Our current un- INSERT GENOMIC LIBRARIES FROM
derstanding of the extent and nature of natural microbial ENVIRONMENTAL SAMPLES: GENERAL
diversity has improved dramatically, in part because of the CONSI DERATI0NS
development and application of the cultivation-independ-
ent rRNA approach. 38.2.1. Microbial Sampling, Still a Challenging
The past decade also witnessed an amazing acceleration Enterprise
in genome-sequencing capabilities. Just seven years after the Sampling strategies are always context dependent and in-
first report of a complete microbial genome sequence, more fluenced by the nature of the microbial community in ques-
than 100 full microbial genome sequences have now be- tion, the environment that it occupies, spatial constraints,
come available for study. A large percentage of the genomes population numbers, and other contaminating organisms

879
880 COMMUNITY A N D C E N O M I C ANALYSIS

BAC Cloning Tradjtional Fosrnid Cloning


1A -agarose-embedded DNA IB r9v- *vo agarose-embedded DNA

&- partial Hind fll digestion


(in agarose)

PFGE site selection 2 100kbp - PFGE size selection -40 kby

$. - ligation of DNA fragments to .$. -


oo~o~~~r electroporation into f.colr 0
sticky-end ligation of
fragments to fosmid
vector, packaging,
transfection into E.co/i
- array library in 0
microtiter plates, 50 - - array library in
200 kbp inserts microtiter plates,
40 kbp inserts
-identification of rRNA
- identificationof rRNA
containing clones
containing clones
$. - screen for genomic contigs
(6.*1
$. w- screen for genomic contigs
-
Biunt End Fosrnid Cloning
IC 2 s (Im -iysis of ceflsl
$. standard DNA purlficatior
Ev/v
- blunt-end repair sheared DNA
- PFGE size selection -40 kbp
- blunt-end ligation of
oo)).o0 fragments to fosmid vector,
packaging, transfection
- array library in
microtiter plates,
40 kbp inserts

- identificationof rRNA
containing clones

&- screenina for aenomic contias FIGURE 1 Comparison of BAC (A), traditional fosmid (B),
and blunt-end fosmid (C) cloning of DNA fragments for envi-
ronmental genomic studies.

and substances. Several up-front questions need to be ad- the types of communities amenable to the approach. Other
dressed when designing sampling strategies. Will the cells important questions are: What size of DNA fragments are re-
need to be purified away from a soil, sediment, or rock ma- quired? What cloning strategy will be used? The answers to
trix? The interference of contaminating materials in DNA these questions will dictate whether the cells need to be em-
purification and downstream steps requiring enzymatic bedded in agarose to maintain high-molecular-weight, intact
modifications are major considerations. Do the cells need to chromosomes during DNA preparation. (For inserts of 40 kb
be concentrated in some fashion before DNA extraction? or less, embedding cells in agarose is not an absolute re-
For some procedures, like bacterial artificial chromosome quirement.) What are the complexity, richness and even-
(BAC) libraries, a minimum of lo9 cells or more may be re- ness, and specific composition of the population/sample in
quired, which can sometimes impart severe limitations on question? For example, it would take only a small propor-
38. Construction of BAC and Fosmid Libraries m 881

tion of eukaryotic genomes in a sample, each having 10 to during downstream lysis and purification steps. Although
50 times greater genome size, to dramatically affect the rep- this method is very efficient, any other material copurifying
resentation of prokaryotic genome sequences in any given with cells will also be trapped in the agarose and may in-
library. All these questions and considerations are impor- hibit downstream enzymatic modification (e.g., restriction
tant to keep in mind before embarking on studies in envi- digestion or ligation). Therefore, cells to be embedded in
ronmental genomics. Prior knowledge of the nature, com- agarose need to be relatively free of contaminating materi-
plexity, and composition of the original sample, both matrix als. Cells concentrated from relatively clean, dilute aqueous
and microbes, will largely influence experimental design environments may be more amenable to embedding than
and the final outcome of library construction and down- those derived from more challenging environments like
stream analyses. The possibilities and permutations of sam- soils or sediments. After proteinase K/detergent digestion of
ple preparation from diverse microbial communities are too the agarose-embedded cells, the liberated DNA is subjected
numerous to cover adequately here. But some of these con- to partial digestion by restriction endonucleases (BamHI
siderations can help guide in sampling design and prepara- and HindIII are commonly used). Partial digestion is ad-
tion strategies. justed to generate a maximum amount of DNA in the ap-
propriate size range, approximately 100 to 300 kb. Often
38.2.2. Large-Insert DNA Libraries: Contemporary there is sheared DNA in the original preparation, so partial
Approaches digestion conditions need to be carefully monitored above
Some of the earliest large-fragment genome libraries from and below the size regions of interest. After purification via
natural microbial populations made use of bacteriophage PFGE, the DNA fragments are recovered from the agarose,
lambda cloning protocols. Today, advanced vector systems ligated into the BAC vector, and electroporated into E . coli.
such as fosmids and bacterial artificial chromosomes (BACs) Improvements in blue-white selection and optimization of
are now in wide use. BACs and fosmids are plasmid vectors electroporation conditions have elevated the efficiency of
whose multiplication is regulated by the F1 origin of replica- the entire process. BAC libraries with average insert sizes
tion (6, 15). The development of these new vectors, which approaching 100 kb and maximum insert sizes near 200 kb
stably maintain and propagate extremely large DNA inserts, have been successfully recovered from natural microbial
was an important milestone in the whole-genome-sequenc- populations ( 1). Importantly, these general procedures need
ing revolution. BAC vectors, developed in the early 1990s for to be tuned to optimize them for any given sample exam-
use in the Human Genome Project, have been particularly ined. For standard BAC libraries, it is particularly important
useful. By virtue of their lowcopy number, BACs can be used to ensure that there is sufficient material to work with, in
to stably propagate very large DNA fragments that are other- general a minimum of 10 cells/ml in the original agarose
wise unstable (15). One version of the BAC vector (origi- Plug.
nally termed fosmid, for F1 origin-based cosmid-sized vector Although insert size is smaller on average (ca. 40 kb),
[6])is introduced into Escherichia coli via transfection, after fosmid cloning approaches have several potential advantages
the DNA has been packaged as phage particles using bacte- compared with standard BACs for constructing high-quality
riophage lambdapackagingextracts. Transfection of the pack- libraries from environmental samples. In general, less bio-
aged fosmid DNA is extremely efficient, facilitating con- mass, lower amounts, and smaller-sized DNA are required
struction of BAC libraries with uniform insert sizes closely for successful fosmid library construction in comparison
bracketing 40 kbp from very small amounts of DNA (6). with BACs. This alleviates the stringent requirement in
Another approach to construct BAC libraries relies on the BAC cloning for isolation of largely intact chromosomes
embedding of cells in an agarose plug, followed by in-gel lysis from many (10) cells (see above). The original fosmid
using detergent and proteinase K. The intact, agarose-em- cloning procedure (6) is outlined in Fig. 1B. In the original
bedded DNA is then subjected to partial restriction enzyme protocol, the vector arms are prepared, and Sau3A partial
digestion and size fractionated via pulsed-field gel elec- digests of the DNA are prepared, generally with a size se-
trophoresis (PFGE). The size-separated fragments are excised lection step. After ligation, lambda extract packaging, and
from the gel and ligated into a BAC vector. Recombinant transfection, the libraries are arrayed in microtiter dishes,
BAC clones are then introduced into E. coli cells via electro- and standard plasmid purification and screening procedures
poration and archived in an ordered library. Depending on are used to evaluate the clones.
the source DNA, this approach can yield cloned inserts ex- Newer approaches have greatly improved the efficien-
ceeding 300 kbp (15), representing as much as one quarter of cies in fosmid library preparation. These new techniques
an entire bacterial genome. incorporate random physical shearing of DNA inserts, fol-
Fosmids (for F-factor cosmids) are essentially identical to lowed by end repair and blunt-end ligation into the circu-
BACs in sequence and their other properties, once pack- larized fosmid (Fig. lC), in contrast to the partial restriction
aged via lambda extracts and transfected into E . coli (6). endonuclease digestion and sticky end cloning required
The main difference between BACs and fosmids is the with standard BAC and fosmid cloning approaches. In gen-
method used to introduce them into E . coli. BACs are in- eral, DNA prepared by standard purification procedures
troduced into host cells via electoporation, whereas fosmids produces DNA of the required size (40 kb), eliminating the
are packaged in vitro in phage lambda capsids and trans- agarose embedding of cells, partial restriction endonuclease
fected into E. coli host cells. These differences in method- digestion, and PFGE steps. In addition, more rigorous DNA
ology influence the maximum size of recombinant DNA in- purification protocols (for instance, CsCl equilibrium den-
serts recovered. BACs recovered from natural microbial sity gradient centrifugation) can be used successfully with
populations can be as large as 200 kb ( l ) ,whereas fosmid the blunt-end fosmid cloning approach. These advantages
sizes range from 32 to 45 kb (6, 18). allow the preparation of high-quality fosmid libraries from
The general strategy for BAC cloning used today is es- environmental samples (11). Such blunt-end fosmid librar-
sentially identical in strategy to original protocols intro- ies may even be more representative than standard BAC
duced in the early 1990s (6, 15, 16) (Fig. 1A). In brief, cells libraries (H. Shizuya, personal communication). This is be-
are first embedded in agarose to prevent physical shearing cause regions with few HindIII or BamHI sites are expected
882 w COMMUNITY AND GENOMIC ANALYSIS

to be less well represented in BAC or fosmids prepared 38.3. PROTOCOLS


using partial digests that are not required in the blunt-end
fosmid cloning approach. With this streamlined procedure, 38.3.1. BAC libraries from Mixed Microbial
it is now possible to successfully prepare high-quality fosmid Populations: Protocol
libraries using as little as 1 pg or less of natural population Supplementary protocols for BAC library preparation and re-
DNA (unpublished). lated procedures can be found on the web at http://informa
.bio.caltech.edu/ and http://www.genome.clemson.edu. The
38.2.3. library Screening and Sequencing following protocols are based on several previously published
One of the advantages of BACs and fosmids is that standard protocols (1-3) and the advice and suggestions of Hiroaki
screening approaches developed for conventional plasmids Shizuya. The success in library construction depends on
are all applicable. High-density colony blots (macroarrays) many variables, in particular, cell concentration, absence of
that allow screening of tens of thousands of clones on a contaminating inhibitory materials in the agarose-embedded
20-cm-by-20-cm blot are one typical approach (18). sample, and the quality of the vector preparation. The fol-
Multiplex screening by PCR is another rapid and very use- lowing protocol has been used to create BAC libraries with
ful way to screen for specific recombinant clones. Both average insert sizes approaching 100 kb from naturally oc-
approaches have been used successfully to identify clones curring marine picoplankton populations concentrated by
of interest in environmental BAC and fosmid libraries (1, tangential flow filtration.
18).For screening of rRNA genes, other rapid multiplexing
approaches that exploit automated capillary electrophoresis BAC library Construction Protocol
include length heterogeneity polymorphism (19) or ter-
minal restriction fragment PCR (7) screening. Addi- Cell embedding and lysis
tionally, functional gene screening can be very useful for 1. Resuspend concentrated bacterioplankton cells in 0.5
gaining information about specific metabolic pathways and ml of sterile seawater to a final concentration of loioto 10
processes (2). Another screening method that has proven cellslml. Mix the cell suspension with an equal volume of
very useful is random end sequencing, which can be used for melted and prewarmed (42C) 1% SeaPlaque agarose. Draw
library quality assessment, gene surveys, contig identifica- the cells in agarose into a lml syringe, seal with parafilm,
tion, and comparative community analyses (see, for exam- and chill on ice to solidify the agarose.
ple, http://www.tigr.org/tdb/MBMO/BAC-end-ann-info 2. Place the agarose plug in 15 ml of STE (500 mM
shtml). Although end sequencing of single-copy BACs and NaC1, 0.1 M EDTA [pH 81, 10 mM Tris-HC1 [pH 8]), and
fosmids was once challenging, improvements in sequencing place the plug on a rotating incubator (room temperature)
technologies have now removed technical barriers to high- for 1 h.
throughput BAC end sequencing. This is probably the first 3. Exchange the buffer with 15 ml of lysis buffer (50 mM
pass method of choice for gaining an in-depth qualitative NaC1, 0.1 M EDTA [pH 81, 10 mM Tris-HC1 [pH 8.0],0.2%
and quantitative assessment of any given environmental [wt/vol] sodium deoxycholate, 1% [wt/vol] Sarkosyl, 1
library. mg/ml lysozyme). Place the agarose plug in a rotating incu-
A significant point to consider in library assessments is bator and incubate at 37C for 0.5 h.
the fact that even in low-copy-number BAC vectors (17) 4. Transfer the agarose plug to a 50-ml Falcon tube and
there may be differential recovery of different genomic se- add 45 ml of ESP buffer (0.5 M EDTA [pH 8.01, 1%
quences. These artifacts can arise from the presence of Sarkosyl, 1 mg/ml proteinase-K). Incubate in a rotating in-
highly repetitive, tandem sequence motifs (17), or due to cubator overnight at 50C. After overnight incubation, re-
gene expression of recombinant BAC DNA in E. coli (es- suspend the agarose plug in freshly prepared ESP buffer and
pecially if it is of bacterial origin) (13), which may some- incubate at 50C for another 16 to 24 h.
times prove lethal. Assessing the extent and fidelity of 5. Following ESP buffer incubations, transfer the agarose
genome recovery from complex assemblages is important in plug into 50 mM EDTA (pH 8.0) and store at 4C.
many contexts, but remains difficult to quantify accurately. 6. Before restriction endonuclease digestion, incubate the
Screening for phylogenetic representation within the li- agarose plug in 15 ml of PMSF solution (10 mM Tris-HCl
brary (via rRNA genes) is one useful approach to assess the [pH 7.51, 0.1 mM EDTA, 1 mM phenylmethylsulfonyl fluo-
diversity of genomes recovered in libraries (3, 12) relative ride) for 2 h at 37C. Replace the PMSF solution with fresh
to the representation in the starting DNA. Statistical meth- PMSF solution, and incubate overnight at 37C. Finally,
ods to estimate total genome recovery in populations may wash the plug twice for 2 h each wash in 10 mM Tris-HC1
also prove useful (5), but may require a fairly extensive sam- +
(pH 7.5) 0.1 mM EDTA.
pling regime. The type of coverage desired in recombinant
libraries also largely depends on the purpose of the inves-
Pilot-scale partial digestion of chromosomal DNA
tigation. For genome-oriented population biology studies,
sample normalization is not necessarily desirable, since 1. Incubate a 1-mm slice of the DNA-containing agarose
quantitative information about genome representation is a plug with 100 p l of a mixture of 1x HindIII reaction buffer,
major goal. Bioprospectors, on the other hand, may wish to 0.1 mg/ml acetylated bovine serum albumin, and 4 mM
maximize the diversity of recovered types and therefore seek spermidine on ice for 30 min.
methods to amplify low-abundance genome types in the li- 2. Replace the solution with fresh solution plus HindIII
braries. In either case, both the richness (species number) and allow to diffuse into the plug for 60 min on ice.
and evenness (relative representation of each species) of Typically, HindIII concentrations for pilot titrations range
microbial genomes in the original sample will greatly influ- from 0.5 to 100 U per 100-p1 reaction.
ence the end result. Prior knowledge of this representation 3. Transfer the reaction mixture to a 37C water bath for
may guide the strategies used for any given environmental partial digestion and incubate for 20 min.
sample or microbial assemblage from which libraries are 4. Stop the reaction by adding 1/10 volume of 0.5 M
constructed. EDTA (pH 8.0) and placing the tubes on ice.
38. Construction of BAC and Fosmid Libraries 883

5. Load the partially digested DNA on a 1% agarose gel using the Bio-Rad Gene Pulser I1 (Bio-Rad) using settings
in 0.5X TBE with a wide-bore tip and seal the wells with of 100 Ohms, 13 kV/cm, and 25 pFa.
the same molten agarose as the gel. 2. Transfer the electroporated cells to 15-ml culture
6. Perform PFGE on a CHEF Mapper (Bio-Rad) under tubes with 0.4 to 1 ml of SOC and shake at 220 rpm for 55
the conditions of 6.0 V/cm, 90-s pulse, 0.5X TBE buffer, min at 37C.
12"C, 18 h. 3. Spread the SOC medium from step 2 on one or two
7. After checking the ethidium bromide-stained gel from LB plates containing 12.5 pg/ml chloramphenicol, 50 pg/
step 6, choose the enzyme concentration giving a majority ml X-Gal (stock, 20 mg/ml in dimethylformamide), and 25
of DNA fragments ranging from about 300 to 600 kb. Due pg/ml isopropyl-P-D-thiogalactopyramoside (IPTG) and in-
to DNA trapping during the step 6 running conditions, we cubate at 37C for 20 to 36 h.
expect the actual size of the DNA fragments obtained from
the 300-to-600-kb fraction to be between 50 and 300 kb 38.3.2. Fosmid library Construction by Blunt-End
in size. Environmental samples often contain considerable Cloning: Protocol
amounts of low-modecular-weight DNA, which masks the The following protocol is a modified version of a blunt-end
partially digested DNA of interest. It is sometimes neces- repair and cloning strategy for preparing fosmid libraries
sary, therefore, to "guesstimate" the appropriate concentra- that is conveniently available as a commercial kit from
tion range of restriction enzyme to use in partial digests. EpiCentre. There are several advantages to the general ap-
The results are then evaluated by ligation, electroporation, proach of blunt-end fosmid cloning, as discussed previously
and assessment of actual BAC clone sizes. in this chapter. The method has been used successfully
to prepare libraries from soil (11) as well as from seawater
Preparing partially digested DNA for ligation and hydrothermal vent sulfide samples (unpublished),
1. Select the amount of enzyme giving the optimum di- using less than 1 pg of DNA. Standard detergent lysis and
gest and perform the digestion reaction in a large scale by phenol/chloroform extraction procedures can be used to
carrying out several reactions (10 to 20 reactions) under ex- prepare the DNA initially, which yields DNA in the ap-
actly the same conditions as above (same volumes, same propriate size range (40 kb) for blunt-end fosmid cloning.
tubes, same dilution of enzyme, same enzyme tube, etc.). Small-scale cesium chloride purification can then be used
2. Load the partially digested DNA on a 1% LMP in a final purification step before size fractionation by gel
agarose gel (Seaplaque GTG, FMC) in 1X TAE and seal electrophoresis. These steps greatly facilitate the removal
with the same molten agarose. of contaminants and potentially inhibitory substances that
3. Perform PFGE on a CHEF Mapper under the condi- often contribute to cloning failures. Additionally, potential
tions of 6.0 V/cm, 20 to 40 s pulse, l x TAE buffer, 12"C, biases introduced by partial restriction endonuclease diges-
18 h. tion of the genomic DNA template are entirely avoided,
4. Cut DNA fragments ranging from about 300 to 600 kb further enhancing cloning efficiency and library represen-
from the gel and use for ligation. tation.
Fosmid library Construction Protocol
ligation
Small-scale CsCl DNA purification
1. Melt the dialyzed LMP DNA piece from the size se-
lection at 65C for 5 min and transfer to a 45C water 1. Purify DNA by standard detergent lysis and phenol
bath. chloroform extraction procedures. This generally produces
2. Add 1 unit of GELase (Epicentre) for every 100 mg of DNA in the appropriate size range of 2 4 0 kb.
excised, melted agarose gel slice and incubate at 45C for 1 h. 2. In a 1 . 5 ~epitube,
1 combine 160 mg of CsC1, 178 pl
3. Check the concentration of the DNA solution by of DNA, and 10 pl of 10 pg/pl ethidium bromide.
loading 10 pl on a 1% agarose minigel with ethidium bro- 3. After mixing and dissolving the CsC1, transfer to a
mide and running a quick electrophoresis at 60 V for 1 h in 200-pl polycarbonate ultracentrifugation tube (Beckman
0.5X TBE with uncut lambda DNA standard solutions catalog no. 342775).
(e.g., lambda 5 ng, 10 ng, 20 ng, 40 ng, and 80 ng/well). 4. Place the tubes in a Beckman TLA 100 rotor, ensur-
Usually the concentration of the size-selected DNA is be- ing that the rotor is balanced.
tween 0.5 and 2 ng/p1. 5. Centrifuge the samples at 100,000 rpm, 2O"C, over-
4. Combine 50 to 200 ng of the size-selected DNA with night.
the HindIII-digested, dephosphorylated BAC vector in a 6. Remove the tubes and visualize the DNA bands with
molar ratio of 1 to 10 and incubate at 55C for 10 min. Cool a longwavelength UV lamp.
to room temperature for 15 min and only then add the lig- 7. Using a sterile 1-ml syringe and needle, remove the
ase and buffer in a total volume of 100 pl. Use 4 to 6 units DNA band and place in an epitube.
of T4 DNA ligase and incubate at 14C for 16 h. 8. Extract the DNA with an equal volume of water-
5. Drop-dialyze the ligation solution by pipetting into a saturated butanol. Remove the upper, ethidium bromide-
Millipore filter (filter type VS, 0.025 pm) floating on Tris- and butanol-containing layer. Repeat the extraction three
EDTA buffer (TE). Incubate for 1 h at room temperature to times.
remove the salts present in the ligation buffer. Carefully re- 9. Bring the volume of the DNA to 2 ml with sterile TE,
move the ligation from the top of the floating filter by pipet- and place the sample in an Amicon Centricon 100 spin
ting. If necessary, store at 4"C, or proceed to the next step. dialysis unit. Centrifuge at 500 x g at room temperature
until the DNA volume is reduced to approximately 50 p1.
Resuspend the DNA to 2 ml again with sterile TE. Repeat
Transformation by electroporation
this washing step two more times to remove residual CsC1.
1. Transform 1 to 2.5 pl of the ligation material into 20 In the final centrifugation, concentrate the DNA to about
to 25 pl of E. coli DHlOB competent cells (Gibco BRL) by 50 pl final volume.
884 COMMUNITY AND GENOMIC ANALYSIS

End repair and PFGE gel purification 2. In the morning, inoculate 50 ml of LB/10 mM MgSO,
with 3 ml of the overnight culture of E. coli EPIlOO plating
1. Place 52 pl of the CsC1-purified DNA ( 2 4 0 kb strain.
DNA) in a 0.5-ml sterile epitube. 3. Grow to an OD600of 0.8 to 1.0 (do not allow culture
2. Add 8 p l of lox end-repair buffer, 8 p l of 2.5 mM to exceed OD600= 1.0).
dNTP mix, and 8 p l of 10 mM ATP. Add 4 p l of Epicentre 4. Cells may be stored at 4C until needed, for up to 3
End-Repair Enzyme Mix. days.
3. Incubate at room temperature for 45 min. 5. For typical titers in environmental libraries, mix 25 p l
4. Heat inactivate at 70"C, 10 min. of packaged library with 500 p l of E. coli EPIlOO plating
5. Add 10 p l of lox gel loading buffer (50% glycerol, strain (from above). This dilution can be bracketed with se-
xylene cyanol, bromphenol blue in TE). rial dilutions to more accurately determine the titer.
6. Prepare a 0.8% SeaPlaque agarose gel in I X TAE 6. Incubate at room temperature 20 min. To titer, plate
buffer. 25 pl and 250 pl onto LB plates (150-mm petri dish) con-
7. Load sample and also 100 ng of T7 DNA size marker taining 12.5 pg/ml chloramphenicol. Incubate plates for 24
in adjacent lanes. Run gel overnight on a PFGE apparatus h at 37C and count colonies to determine titer in the
(CHEF-DR I1 (Bio-Rad), or the equivalent, in 1X TAE packaging mix.
Buffer, under the following conditions: 6 V/cm, 5 to 15 s
pulse switch time, 13 h run time, 12C buffer temperature.
8. The next morning, remove the gel and stain with 1X 38.4. CONCLUSION AND FUTURE
SybrGold (10,OOOx stock SybrGold diluted 1/10,000 in ster- PROSPECTS
ile water ) for 30 min, per the manufacturer's instructions.
9. Visualize DNA on DarkReader Transilluminator The applications of BAC libraries for environmental mi-
(EpiCentre). Excise DNA in the 40- kb size range, and crobiology are many and varied. Bioprospecting for antibi-
place in a 1.5-ml epitube. otics and other natural products has been one of the earlier
10. Melt gel slice at 70"C, 10 min. and more obvious applications to exploit their novel ge-
11. Place at 45"C, 10 min. netic content. Tremendous potential exists for genomic ap-
12. Add an appropriate volume of 50X Gelase Buffer to proaches using these libraries to answer many questions
the melted gel slice to achieve a 1X final Gelase Buffer con- about evolution, ecology, and population biology in the
centration. Add 1 p1 of Epicentre GELase for every 100 pl context of natural microbial ecosystems. BAC and fosmid
of melted gel slice. Mix gently by inverting the tube several libraries are useful tools that can be used to sidestep many
times, and incubate for 1 to 2 h at 45C. thorny problems that still inhibit the accurate characteriza-
13. Inactivate Gelase by incubation at 70C for 10 min. tion of naturally occurring microbial communities. They
14. Bring the volume of DNA to 500 p l with sterile represent useful tools for conducting sequence surveys to
HzO. Wash the DNA by spin dialysis by centrifuging at determine the phylogenetic and functional content and
4,000 X g in an Amicon Microcon-100 unit. Resuspend to properties inherent in native microbial populations. They
500 pl with sterile H2O. Repeat this washing step two more can also serve as reagents for gene discovery, gene expres-
times. Bring the final volume of the DNA down to about 50 sion, and functional genomic characterization of naturally
p l final volume. The DNA is now ready for ligation. occurring microbes, some still resistant to cultivation tech-
niquess. BAC and fosmid libraries can also serve as useful
ligation and packaging reagents in microarray construction and analyses of natu-
1. Place 15 p l of end-repaired, gel-purified (now 40-kb- rally occurring microbial dynamics and variability.
sized) DNA in a 0.5-ml epitube. Environmental genomic approaches are cultivation in-
2. Add 2 p1 of lox Fast-link Ligase buffer, 1 pl of 10 dependent and theoretically can provide equal access to the
mM ATP, 1 pl of pEPIFOS plasmid, and 1 p1 of Fast-link collective genomes contained in natural microbial assem-
ligase. Mix by gentle pipetting, avoiding bubbles. blages. Applied microbial population genomics can now
3. Incubate at room temperature for 2 h. facilitate predictions about the relationships between geno-
4. Inactivate ligase at 70C for 10 min. (Can now store type and ecological distribution and function. These ap-
the ligation at 4C before proceeding to the packaging proaches can broaden our knowledge of natural environ-
step.) mental processes and extend our abilities to exploit natural
5. Thaw MaxPlax packaging mix on ice. Remove 25 p1 microbial products and processes. Perhaps most impor-
into prechilled epitube. Place this aliquot at -80C for tantly, application of environmental genomic approaches
later use in step 8. has the potential to foster a better understanding of micro-
6. To remaining packaging mix, add 10 pl of ligation bial genome evolution, ecology, and population biology and
(avoid bubble formation and mix by gentle pipetting). to contribute significantly to theoretical models and predic-
7. Incubate 90 min at 30C. tive capabilities in environmental microbiology and micro-
8. Remove remaining 25 p l of packaging mix from bial ecology.
-80C. Add the entire 25 pl to the packing reaction at
30C. Incubate another 90 min at 30C. 38.5. REFERENCES
9. Add 100 p1 of phage dilution buffer (10 mM Tris [pH 1. BhjA, O., L. Aravind, E. V. Koonin, M. T. Suzuki, A.
8.31, 100 mM NaC1, 10 mM MgC12), then add 5 pl of Hadd, L. P. Nguyen, S. B. Jovanovich, C. M. Gates,
CHC13.Mix gently. Centrifuge briefly. The packaged library R. A. Feldman, J. L. Spudich, E. N. Spudich, and E. F.
may be stored at 4C for up to several weeks before trans- DeLong. 2000. Bacterial rhodopsin: evidence for a new
duction and plating. type of phototrophy in the sea. Science 289:1902-1906.
Transduction 2. Bhji, O., M. T. Suzuki, J. F. Heidelberg, W. C. Nelson,
C. M. Preston, T. Hamada, J. A. Eisen, C. M. Fraser,
1. Inoculate 50 ml of LB/10 mM MgSO, with E. coli and E. F. DeLong. 2002. Unsuspected diversity among
EPIlOO plating strain. Grow overnight with shaking at marine aerobic anoxygenic phototrophs. Nature 415:630-
37C. 633.
38. Construction of BAC and Fosrnid Libraries 885

3. Bejh, O., M. T. Suzuki, E. V. Koonin, L. Aravind, A. Gilman, M. S. Osburne, J. Clardy, J. Handelsman, and
Hadd, L. P. Nguyen, R. Villacorta, M. Amjadi, C. R. M. Goodman. 2000. Cloning the soil metagenome: a
Garrigues, S. B. Jovanovich, R. A. Feldman, and E. F. strategy for accessing the genetic and functional diversity
DeLong. 2000. Construction and analysis of bacterial ar- of uncultured microorganisms. Appl. Environ. Microbiol.
tificial chromosome libraries from a marine microbial as- 66~2541-2547.
semblage. Environ. Microbiol. 2:5 16-529. 13. Rondon, M. R., S. J. Raffel, R. M. Goodman, and J.
4. Doolittle, R. F. 2002. Microbial genomes opened up. Handelsman. 1999. Toward functional genomics in bacte-
Nature 392:339-342. ria: analysis of gene expression in Escherichia coli from a
5. Hughes, J. B., J. J. Hellmann, T. H. Ricketts, and B. J. bacterial artificial chromosome library of Bacillus cereus.
Bohannan. 2001. Counting the uncountable: statistical Proc. Nutl. Acad. Sci. USA 96:6451-6455.
approaches to estimating microbial diversity. Appl. En- 14. Schleper, C., E. F. DeLong, C. M. Preston, R. A.
viron. Microbiol. 67:4399-4406. Feldman, K. Y. Wu, and R. V. Swanson. 1998. Genomic
6. Kim, U.-J., H. Shizuya, P. Dejong, B. Birren, and M. analysis reveals chromosomal variation in natural popula-
Simon. 1992. Stable propagation of cosmid sized human tions of the uncultured psychrophilic archaeon Cen-
DNA inserts in an F-factor based vector. Nucleic Acids Res. archaeum symbiosum. J. Bucteriol. 1805003-5009.
20~1083-1185. 15. Shizuya, H., B. Birren, U. J. Kim, V. Mancino, T.
7. Marsh, T. L., P. Saxman, J. Cole, and J. Tiedje. 2000. Slepak, Y. Tachiiri, and M. Simon. 1992. Cloning and
Terminal restriction fragment length polymorphism analy- stable maintenance of 300-kilobase-pair fragments of
sis program, a web-based research tool for microbial com- human DNA in Escherichia coli using an F-factor-based
muni ty analysis. Appl . Environ. Microbiol. 66:3 6 16-3 620. vector. Proc. Nutl. Acad. Sci. USA 89:8794-8797.
8. Olsen, G. J., D. J. Lane, S. J. Giovannoni, N. R. Pace, 16. Shizuya, H., and H. Kouros-Mehr. 2001. The develop-
and D. A. Stahl. 1986. Microbial ecology and evolution: ment and applications of the bacterial artificial chromo-
a ribosomal RNA approach. Annu. Rew. Microbiol. 40: some cloning system. Keio J. Med. 50:26-30.
337-365. 17. Song, J., F. Dong, J. W. Lilly, R. M. Stupar, and J. Jiang.
9. Pace, N. R. 1997. A molecular view of microbial diversity 2001. Instability of bacterial artificial chromosome (BAC)
and the biosphere. Science 276:734-740. clones containing tandemly repeated DNA sequences.
10. Pace, N. R., D. A. Stahl, G. J. Olsen, and D. J. Lane. Genome 44:463-469.
1985. Analyzing natural microbial populations by rRNA 18. Stein, J. L., T. L. Marsh, K. Y. Wu, H. Shizuya, and
sequences. ASM News 51:4-12. E. F. DeLong. 1996. Characterization of uncultivated
11. Quaiser, A., T. Ochsenreiter, H. P. Klenk, A. Kletzin, prokaryotes: isolation and analysis of a 40-kilobase-pair
A. H. Treusch, G. Meurer, J. Eck, C. W. Sensen, and C. genome fragment from a planktonic marine archaeon.
Schleper. 2002. First insight into the genome of an uncul- J. Bacteriol. 178:591-599.
tivated crenarchaeote from soil. Environ. Microbiol. 4: 19. Suzuki, M., M. S. Rappe, and S. J. Giovannoni. 1998.
603-6 11. Kinetic bias in estimates of coastal picoplankton commu-
12. Rondon, M. R., P. R. August, A. D. Bettermann, S. F. nity structure obtained by measurements of small-subunit
Brady, T. H. Grossman, M. R. Liles, K. A. Loiacono, rRNA gene PCR amplicon length heterogeneity. Appl.
B. A. Lynch, I. A. MacNeil, C. Minor, C. L. Tiong, M. Environ. Microbiol. 64:4522-4529.
Single Cell Identification by Fluorescence In Situ
Hybridization
BERNHARD M. FUCHS, JAKOB PERNTHALER, AND RUDOLF AMANN

39.1. INTRODUCTION ........................................ 886


.2. rRNA DATABASE ........................................ 886
.3. PROBEDESIGN ......................................... 887
.4. SELECTION OF FLUORESCENT LABEL ...................... 887
.5. QUALITY CHECK OF PROBES ............................. 888
.6. CELLFIXATION...... ................................... 888
.7. PROBE TESTING; OPTIMIZATION OF HYBRIDIZATION
CONDITIONS ........................................... 889
.8. HYBRIDIZATION OF CULTURED ORGANISMS ON GLASS
SLIDES ................................................ 890
.9. HYBRIDIZATION OF FIXED CELLS ON MEMBRANE FILTERS .... 891
.lo. HYBRIDIZATION IN LIQUID FOR FLOW CYTOMETRY ......... 892
.11. HYBRIDIZATION FOLLOWED BY TYRAMIDE SIGNAL
AMPLIFICATION (CARD-FISH) ............................ 892
.12. TROUBLESHOOTING .................................... 893
.13. APPLICATIONS AND FURTHER READING ................... 893
39.13.1. Methodological Aspects .............................. 893
.2. Marine Environments ................................ 893
.3. Sediment ......................................... 893
.4. Activated Sludge ................................... 894
.5. Limnology ........................................ 894
39.14. REFERENCES ........................................... 894

39.1. INTRODUCTION called probes, with a typical length of about 15 to 30 nu-


This chapter presents an updated collection of protocols for cleotides. For standard FISH applications, oligonucleotides
the identification of individual microbial cells by fluores- are typically monolabeled with a fluorochrome.
cence in situ hybridization (FISH) with rRNA-targeted FISH of whole cells starts with a fixation of the sample
oligonucleotide probes. This phylogenetic staining (14) containing the target cell types (Color Plate 14). Fixation
method allows the enumeration, biomass determination, stabilizes macromolecules and cytoskeletal structures, thus
and localization of microorganisms in their environmental preventing lysis of the cells during hybridization, and at the
context. FISH is an integral part of the rRNA approach to same time permeabilizes the cell walls for fluorescently la-
microbial ecology and evolution (38). beled oligonucleotide probes. The fixed cells are incubated
Several reviews are available on the principles of hy- in a buffer containing the specific probe at a temperature
bridization (5, 58) and on methodological aspects and ap- near but below the melting point of the probe-rRNA hybrid.
plications of FISH with rRNA-targeted oligonucleotides (1, The subsequent washing step will remove unbound probe
4). Therefore, we will first outline the background in brief and leave only those probe-rRNA pairs intact that have no
and then focus on protocols. mismatches in the hybrid. Consequently, only target cells
The rRNAs are among the most conserved macromole- that contain the full signature sequence on their rRNA will
cules. The three prokaryotic rRNAs, 5S, 16S, and 23s be stained. Finally, hybridized cells can be enumerated by
rRNA, are found in abundance in every cell. Therefore, the epifluorescence microscopy or by flow cytometry.
following protocols are not readily applied to the detection
of molecules that have low cellular abundance, such as
mRNA, plasmids, or chromosomal genes. Comparative 16s 39.2. rRNA DATABASE
rRNA sequence analysis has become a standard method in The reliability of FISH for the identification of cells in com-
microbial taxonomy. Consequently a 16s rRNA database plex environments strongly depends on the quality of the
has been compiled that comprises to date more than 400,000 16s rRNA database. A comprehensive and well-maintained
(2007) sequences covering not only most pure cultures but database is vital for rational probe design. Powerful software
also many 16s rRNA genes of so far uncultured bacteria (9, tools are required to manage such a database. We recom-
30,59). The comparison of 16s rRNA sequences allows for mend the highly integrated software package ARB released
the reconstruction of microbial phylogeny (65) and the by the Technical University of Munich (60). This program
identification of short signature sequences that are unique runs on different UNIX-based operating systems (including
for different groups of microorganisms. These signatures are Linux), it is capable of maintaining several hundred thou-
used as targets for short complementary oligonucleotides, sand aligned 16s rRNA sequences, and it allows the easy

886
39. Single Cell Identification by FISH 887

import of additional sequences from various sources. In an the diagnostic probe, thereby opening the target region
ideal world, probe design would be solely based on high- for the probe. Helpers should be a few nucleotides longer
quality, full-length sequences. In practice, the public do- than the diagnostic probe; that is, if the probe is an 18-mer,
main databases are filled with partial sequences and se- the helper should be a 21-mer to ensure a tight binding be-
quences that contain sequencing errors. The sequence yond the melting point of the diagnostic probe.
gaps in partial sequences can limit probe design by reduc- Another strategy of probe selection is searching for al-
ing the number of potential target regions and by failing to ready published probes. Loy and coworkers established a
provide information about the specificity of existing probe database with currently more than 890 entries, not all
probes (Fig. 1). developed for FISH (www.microbia1-ecology.net/probebase)
(29).
39.3. PROBE DESIGN
For FISH, a probe length of 15 to 25 nucleotides is most 39.4. SELECTION OF FLUORESCENT LABEL
commonly used. Sequence signatures serving as suitable tar- A range of fluorochromes is available for the labeling of nu-
get sites for nucleic acid probing can be convenient and cleic acids, but not all fluorochromes are equally suited as
automatically searched with the PROBE-DESIGN tool labels for oligonucleotides. Newly developed dyes, in par-
within the ARB software package. First, a target group of ticular, should be checked for nonspecific staining.
organisms must be specified, e.g., in a phylogenetic tree Standard labels for in situ hybridization are the green fluo-
contained in ARB. The PROBE-DESIGN tool searches for rescein and the red tetramethylrhodamine derivatives
possible signature sequences that are diagnostic for the se- (Table 1). These dyes are well suited for standard applica-
lected species. The G + C content of probe sequences influ- tions when the ribosome content of target cells is high. In
ences their melting behavior. By default this parameter is addition, these dyes can be used in conjunction to label dif-
set in PROBE-DESIGN between 50 and 100% to ensure a ferent probes in double-staining experiments. CY3 and CY5
tight binding, but usually it ranges between 50 and 70%. are members of the indocarbocyanine family. The high sig-
Sometimes the PROBE-DESIGN tool cannot find a suit- nal intensity of these two dyes makes them the fluo-
able probe. However, the program provides options for mod- rochromes of choice when trying to detect small cells with
ifying the search parameters to look for signatures in subsets low ribosome content, such as bacterioplankton cells.
of the group originally selected, or by choosing to allow for These fluorochromes exhibit high fluorescence because of
the signature to be found in a defined number of species their high quantum yield and high molar extinction coeffi-
outside the target group. By the combinatorial use of probes cient (Table 1). It is important to note that the CY5 deriv-
with overlapping specificity, the selected group of organisms ative that fluoresces in the near infrared is detectable only
may be fully targeted. with a CCD camera or on a confocal laser scanning micro-
A problem that should be considered during the design scope with red laser excitation. Concomitant to the selec-
of FISH probes is target site accessibility. The higher-order tion of the fluorescent label, care should be taken that the
structure of the ribosome may hinder the binding of the right optical filters are chosen for the detection of the re-
probe to its target site. The 16s rRNA in situ accessibility spective dyes. Well-adapted optical filters with a high trans-
for oligonucleotide probes has recently been studied for two mission in the emission spectrum of the dye and a strong
members of the domain Bacteria (Escherichia coli, Pirellula and sharp blocking of the excitation light are essential for a
sp. strain l ) , one eukaryote (Saccharomyces cereuisiae), and confident detection of weak signals.
one archaeon (Metallosphaera sedula) (8, 18) (Color Plate In environmental samples, single oligonucleotides carry-
15). In 2001 a complete accessibility map for the 23s rRNA ing only one fluorochrome may not provide enough fluores-
of E. coli was published (17) (Color Plate 16). These color- cence signal to detect cells with low ribosome content (43).
coded maps clearly demonstrate dramatic differences in the Polynucleotide probes with a length of more than 100 nu-
binding of different fully complementary probes of approxi- cleotides, carrying several fluorochromes, are an alternative
mately identical length to one batch of fixed target cells. (13, 62). However, these probes lack specificity for narrow
Even though the secondary structure of the ribosome is target groups that are at the level of species and genera. A n
highly conserved, and even though a consensus map could alternative labeling technique that increases fluorescence
be developed from the 16s rRNA accessibility studies (8), signal intensity uses horseradish peroxidase (HRP)-labeled
each probe should be checked on its respective target group oligonucleotides. When using HRP-labeled probes, fluores-
of organisms to ensure high probe signals. cent staining results from a secondary incubation with fluo-
It has recently been shown that inaccessible target re- rescently labeled tyramide. The specifically bound peroxi-
gions can be made accessible by the use of unlabeled dase molecules catalyze the deposition of these labeled
oligonucleotides called helpers (16). These bind adjacent to reporter compounds within cells targeted by the HRP-tagged

Probe 3 l - 5 GGAAGCCCGGAGAACGGT

UCGCAAGACCAAAGAGGG CCUUCGTGCCUCUUGCCA CGGAUGUG


Non-target? . . . . . . . . . . . . . . . . . . . . . . . . . . .CCUCUUGCCA CGGAUGUG
UCGCAAGACCNNNGAGGG CCUUCGGSCCUCUUGYCA CGGAUGUG
888 COMMUNITY AND CENOMIC ANALYSIS

TABLE 1 Dye labels frequently used for oligonucleotide probes and their characteristics"
Label Excitation ( ? 10 nm) Emission ( ? 10 nm) Mol mass (Da) Eb(mol-' cm-')
CY3' 5121552 5651615 766 150,000
CY5' 625-650 670 792 250,000
Carboxyfluorescein (FAM)d 492 518 376 79,000
Fluoresceind 490 520 389 77,000
Tetramethylrhodamined 543 571 444 99,000
Carboxytetramethylrhodamine (TAMRA)d 542 568 430 91,000
"Fluorescein and derivatives are pH sensitive and exhibit maximum fluorescence at pH 2 9.
"e, molar extinction coefficient.
'Data compiled from Amersham Biosciences, San Francisco, CA.
dData compiled from reference 50 and Invitrogen, Carlsbad, CA.

probe. FISH signals are up to 20-fold brighter with HRP- solution can be stored in the freezer, but once an aliquot is
labeled probes than with conventional single-labeled thawed to prepare working solutions, keep it at 4C.
probes (52). However, cell permeabilization protocols need Lyophilized HRP-labeled probes are suspended in sterile
to be adjusted to enable the larger enzyme-labeled oligonu- H 2 0 , too. For calculating the concentration, it has to be
cleotides to penetrate into cells (42). taken into account that the enzyme itself contributes to the
measured absorbance at 260 nm. Therefore the measured
Aoligohas to be lowered by a correction factor (cf) of 0.276.
39.5. QUALITY CHECK OF PROBES Presuming optimal labeling, the peak ratio (A260/AHRP)
Oligonucleotide probes are custom made by solid-phase should be about 3.
synthesis. In the last step of synthesis a fluorochrome is
added to the 5' end of the oligonucleotide. Purified probe 39.6. CELL FIXATION
stocks are frequently delivered lyophilized. Upon reconsti-
tution with 100 p1 of sterile water, a probe synthesis at FISH of whole fixed microbial cells with rRNA-targeted
0.02-pmol scale yields approximately a 1,500 ng p1-I stock oligonucleotide probes is based on the binding of a short,
solution. To determine the exact probe concentration, the fluorescently labeled oligonucleotide to the intracellular

-
absorbance of the 1:100-diluted stock solution at 260 nm
should be measured, giving 1 OD260nm 20 ng pl-' DNA.
Furthermore, the labeling of the oligonucleotide should be
target nucleic acids (Color Plate 14). Therefore the fixation
of the sample is one of the most critical steps in the proto-
col. A good fixative should preserve the cell morphology
while concomitantly permeabilizing all cells for the labeled
checked. For a pure monolabeled oligonucleotide, the ratio
of absorption of the dye (Adye)versus the absorption of the oligonucleotide. Standard fixatives are aldehydes and alco-
nucleic acids at 260 nm (A260)of a monolabeled oligonu- hols. For many microorganisms, good results are achieved
cleotide should match the ratio of the extinction coeffi- by fixation at a final concentration of 1% formaldehyde
cients ( E ) of the dye and oligonucleotide. The extinction overnight at 4C. The formaldehyde solution should be
coefficient E at 260 nm ( E ~of~an~oligonucleotide
) can be freshly prepared from paraformaldehyde.
estimated from its nucleotide composition as the sum of the Preparation of a 4% Paraformaldehyde Fixative
extinction coefficients of the individual nucleotides (dATP
= 15.4 cm3 pmol-', dCTP = 7.3 cm3 pmol-', dGTP = 1. Pour 2 g of paraformaldehyde (PFA) powder in 50 ml
11.7 cm3 pmol-I, dTTP = 8.8 cm3 pmol-', from reference of phosphate-buffered saline (PBS; 130 mM NaC1, 10 mM
50). Taking into account the extinction coefficient of the Na2HP04/NaH2P04[pH 7.41) or a similar buffer (use mask
dye (dye;see also Table 2), the quality of labeled oligonu- for weighing PFA; irritant if inhaled).
cleotide can be estimated by calculating a ratio k according 2. Heat to ca. 60C (must not boil!) until suspension is
to the following formula: clear (ca. 0.5 h); if not, add some drops of 1N NaOH.
3. Check pH and adjust to pH 7.0.
k= '260 I 'dye 4. Filter through 0.2-pm-pore-size filter and place on ice.
A260/Adye
5. Usable for up to 1 week, and up to 6 months if stored
in the dark under nitrogen atmosphere at room temperature
Values of k < 1 indicate an incomplete labeling of a probe, (RT).
whereas values >1 point to the presence of additional, po-
tentially unbound dye. Considering inaccuracies in the es- Commercially available 35 to 37% formalin solution is
timation of the extinction coefficients of oligonucleotides, often stabilized with methanol, which decreases FISH sig-
k values between 0.7 and 1.3 are acceptable. nal intensity and tends to precipitate upon longer storage.
Working solutions are prepared at concentrations of 50 After the fixation, a dehydration series of 50, 80, and 96%
ng pl-' and stored in the dark at -20C. Prepare only ethanol may help to permeabilize cells for FISH.
small portions of probe working solutions (50 to 100 pl).
Fixation of Pure Cultures with Cram-Negative Cell
Repeated freeze-thawing may damage the probe and might
Wall
result in a precipitation of the probe, which in turn would
lead to weak hybridization signals and high background. 1. Harvest cells during logarithmic growth by centrifuga-
Do not freeze working solutions of HRP-labeled probes, tion of an aliquot (ca. 2 ml) in microcentrifuge (10 min at
but store them in the refrigerator at 4C. Portions of stock 4,000 X g).
39. Single Cell Identification by FISH rn 889

2. Discharge supernatant and resuspend cells in 750 pl of Germany); if required, the sonicated sample can be further
PBS (145 mM NaC1, 1.4 mM NaH2P04,8 mM Na2HP04 diluted.
[PH 7.41). NOTE: Depending on the type of sediment or soil, it
3. Fix by adding 250 pl of a 4% PFA fixative (1% final might be necessary to also adapt the aliquot size prior to
concentration). sonication. If too much sediment is suspended, sonication
4. Incubate for 1 h (for thick-walled cells) to 24 h (for will lead to incomplete detachment of cells from particles.
fragile cells) at 4C. 9. Place cellulose nitrate support filters beneath the
5. Pellet cells by centrifugation (10 min at 4,000 X g); membrane filters to improve the distribution of cells; add 15
discharge supernatant. to 20 pl of aliquot from the sonicated sample to 2 ml of dis-
6. Thoroughly resuspend fixed cells in 500 pl of PBS. tilled water, and filter this volume onto the membrane fil-
7. Repeat steps 5 and 6. ters.
8. Add 500 pl of absolute ethanol and resuspend cells 10. Air-dry filtered preparations and store in petri dishes
thoroughly. at -20C until hybridization.
9. At this stage samples can be stored at -20C for sev-
eral months.
39.7. PROBE TESTING; OPTIMIZATION OF
Fixation of Pure Cultures with Cram-Positive Cell Wall HY BR IDIZAT I0N C O N DITI0NS
1. Harvest cells during logarithmic growth by centrifuga- When using probes that have been previously described, it
tion of an aliquot (ca. 2 ml) in microcentrifuge (10 min at is important to always check the probe against a recently
4,000 X g). updated 16s rRNA database (e.g., by a NCBI BLAST search;
2. Discharge supernatant and wash cells in PBS. www.ncbi.nlm.nih.gov/BLAST/). Ideally, the probe of inter-
3. Pellet cells by centrifugation (10 min at 4,000 X g); est still has more than one base mismatch with all nontarget
discharge supernatant. microorganisms. For the design of new probes, it is important
4. Add 500 pl of PBS and resuspend cells thoroughly. to keep discriminatory positions central since a mismatch
5. Add 500 pl of cold, absolute ethanol and mix. between probe and nontarget rRNA at the 3' or 5' end of
6. A t this stage samples can be stored at -20C for sev- the oligonucleotide is only weakly destabilizing. Competitor
eral months. oligonucleotides (32) have been shown to strongly enhance
single-mismatch discrimination. Competitors are unlabeled
Fixation of Planktonic Samples
oligonucleotides, which are fully complementary to the
1. Add freshly prepared PFA fixative to water sample to mismatch-containing nontarget sequence. They suppress un-
a final concentration of 1 to 3% and fix for 1 to 24 h at 4C. specific probe binding to a defined one-mismatch sequence.
2. Place a moistened support filter (0.45 p m pore size, If possible, new probes should be tested by FISH of iso-
cellulose nitrate, 47 mm diameter; Sartorius, Germany) and lates that have zero, one, and more mismatches with the
a membrane filter (0.2 p m pore size, white polycarbonate, oligonucleotide. A series of hybridizations is performed at
47 mm diameter; Millipore, Eschborn, Germany) into a fil- increasing stringency either by increasing the temperature
tration tower. of hybridization or by increasing the concentration of a de-
3. Filter an appropriate volume of the fixed sample by naturing agent like formamide in the hybridization buffer.
applying gentle vacuum. Support filters may be utilized for The changes in the fluorescence intensities of individual
several samples. For cell numbers of approximately lo6 cells can be quantified by computer-assisted image analysis
ml-', 10 ml of sample is generally sufficient. (37) or by flow cytometry (18). The most desirable hy-
4. After complete sample filtration, wash with 10 to 20 bridization stringency often occurs at the point immediately
ml of sterile HzO; remove H 2 0by filtration. before the target cell fluorescence begins to decrease (Fig.
5. Put membrane filter in a plastic petri dish, cover, and 2). At this formamide concentration, hybridization to the
allow air drying. nontarget organism should be low or absent. As a rule of
6. Store at -20C until processing; filters can be stored thumb, an 18-mer oligonucleotide with a G+C content be-
frozen for several months without apparent loss of hy- tween 50 and 60% will start to dissociate from its fully com-
bridization signal. plementary rRNA target at a formamide concentration of
approximately 30 to 40% in our standard buffer at 46C.
Fixation and Preparation of Sediment and Soil
Alternatively, to determine their temperature of dissocia-
Samples
tion (TJ, probes can be radiolabeled and hybridized to ex-
1. Suspend 0.5 ml of freshly collected sediment or soil tracted rRNAs that have been blotted onto nylon mem-
in 1.5 ml of 4% PFA fixative in a 2-ml screw-top microcen- branes (58).
trifuge tube; fix for 1 to 24 h. Frequently, new probes are designed to target yet-uncul-
2. Centrifuge at 10,000 rpm for 5 min; pour off super- tured microorganisms that are only known from their
natant. rDNA sequences. In this case it is not possible to test these
3. Add 1.5 ml of PBS and resuspend sample. probes on isolates. Two strategies are available to optimize
4. Repeat steps 2 and 3. the hybridization conditions. (i) The cloned 16s rRNA
5. Centrifuge at 10,000 rpm for 5 min; pour off super- gene of interest can be transcribed in vitro into RNA,
natant. which is then blotted on a nylon membrane and hybridized
6. Add 1.5 ml of a 1:l mix of PBS/ethanol and store with a radiolabeled oligonucleotide at increasing levels of
sample at -20C until further processing. formamide (e.g., references 45 and 47). This quite laborious
7. Resuspend sample and transfer 20 to 100 pl of method is based on the assumption that the temperature of
aliquot to 500 pl of a 1:l mix of PBS/ethanol in a 2-ml mi- dissociation from isolated rRNA is the same as from rRNA
crocentrifuge tube. in fixed cells. (ii) Recently a new method was published
8. Sonicate aliquot for 20 to 30 s at low intensity using using clones for adjusting the hybridization conditions.
1-s sonication pulses (Sonopuls HD70, Bandelin, Berlin, Clones carrying the target sequence of the new probe are
890 COMMUNITY AND GENOMIC ANALYSIS

specificity low high low

temperature, formamide
[stringency]

FIGURE 2 Theoretical dissociation profiles of a nucleic acid probe from a perfectly matched
(bold line) and an imperfectly matched immobilized target nucleic acid (dashed line). The probe-
conferred signal (Y axis), which is directly proportional to the sensitivity, is shown over tempera-
ture and formamide concentration (x axis), which represents the hybridization stringency. Note
that the temperature of dissociation of the perfect hybrid is higher than that of the imperfect hy-
brid. The primary goal of the probe design is to maximize the difference between the temperature
of dissociation of a probe from target and nontarget nucleic acid. The bar above the dissociation
profiles indicates hybridization stringencies with high and low specific discrimination of target and
nontarget nucleic acid, respectively.

grown with chloramphenicol and isopropyl-P-D-thiogalac- 4. For the hybridization mixtures, add 1 volume of
topyranoside (IPTG). If a vector with an inducible pro- probe working solution (50ng p1-l) to 9 volumes of hy-
moter upstream of the multiple cloning site is chosen, then bridization buffer in a 0.5-ml microcentrifuge tube; keep
this treatment leads to an in vivo transcription of the probe solutions dark and on ice.
cloned 16s rDNA and accumulation of 16s rRNA of the 5. Prepare hybridization vessels from 50-ml polyethyl-
uncultured organism inside the E. coli cell. After standard ene tubes: insert a piece of blotting paper into a polyethyl-
fixation of these clones, they could be used as analogs to ene tube and soak it with the remaining hybridization
cultured organisms for determining the melting point of buffer; use separate tubes for each concentration of for-
probes (54). mamide.
6. Add 10 pl of hybridization mix to the samples in
each well and place the slide into the polyethylene tube (in
39.8. HYBRIDIZATION OF CULTURED a horizontal position).
ORGANISMS ON GLASS SLIDES 7. Incubate at 46C for at least 90 min (maximum, 3 h).
In a quick but nonquantitative protocol, fixed cells are 8. Prepare 50 ml of washing buffer in a polyethylene
transferred onto gelatin-coated slides and incubated in a tube and preheat in a 48C water bath (see Tables 4 and 5).
moisture chamber with a buffer containing an oligonu-
cleotide probe. After a brief washing, the cells are embed-
ded in antifading reagent for microscopic visualization.
1. Heat a 0.01% CrK(SO& (prevents fouling of gela- TABLE 2 Standard hybridization buffer
tin)-0.1 % gelatin solution to 65C; dip precleaned multi-
well slides in this solution; let air dry. Final concn in
Stock reagent Volume
2. Spot 2 to 20 pl of fixed cell suspension (depending hybridization buffer
on cell density) in the wells of the gelatin-coated slide; let
air dry, then dehydrate for 3 min each in 50,80, and 100% 5 M NaCl 360 p1 900 mM
ethanol. 1 M Tris-HC1 40 p1 20 mM
3. Prepare hybridization buffer (Tables 2 and 3). Formamide % depending
NOTE: The formamide concentration depends on the on probe (see
probe used and determines the stringency of the hybridiza- Table 3)
tion. Hybridization stringency may also be adjusted by tem- Distilled HzO Add to 2 ml
perature rather than by the chemical compositon of buffers.
10% SDS (added 2 pl 0.01%
We find that it is more convenient to keep the incubator last to avoid
and water bath at one set temperature and to modulate the precipitation)
stringency by adding formamide.
39. Single Cell Identification by FISH 891

TABLE 3 Formamide concentration of some frequently applied probes"


Probe Specificity Sequence Formamide % Target Competitor Reference
ALF968 Alphaproteobacteria GGTAAGGTTCTGCGCGTT 35 16s 35
ARCH9 15 Archaea GTGCTCCCCCGCCAATTCCT 0 16s 58
BET42a Betaproteobacteria GCCTTCCCACTTCGTTT 35 23s Gam42a 32
CF319a Cy tophagalFluvobacterium TGGTCCGTGTCTCAGTAC 35 16s 31
cluster
EUB338 Most Bacteria, no GCTGCCTCCCGTAGGAGT 0 16s 3
Planctomycetales
EUK5 16 Eukarya ACCAGACTTGCCCTCC 0 16s 2
GAM42a Gammaproteobacteria GCCTTCCCACATCGTTT 35 23s Bet42a 32
HGC69a Many Actinobacteria TATAGTTACCACCGCCGT 20 23s 49
LGCa354 Some Firmicutes TGGAAGATTCCCTACTGC 20 16s 34
Non338 Nonsense probe, for detection ACTCCTACGGGAGGCAGC 0 16s 58
of nonspecific binding
Pla886 Planctomycetales GCCTTGCGACCATACTCCC 35 16s cPla 36
'For horseradish-labeled probes, use 20% more formamide in hybridization buffer. See text for details

NOTE: The stringency in the washing buffer is UV excitation will also bleach the probe signal; for count-
achieved by adjusting the NaCl concentration. This avoids ing, it is therefore safer to first quantify probe-stained cells
the use of excess amounts of formamide. and subsequently all cells from the same field of vision in
9. Quickly rinse the slide carefully with a bit of wash- UV excitation.
ing buffer, transfer slide into preheated washing buffer, and
incubate for 25 min at 48C (water bath).
10. Rinse slide with distilled HzOand let air dry. 39.9. H Y B R I D I Z A T I O N OF FIXED CELLS ON
11. For counterstaining, cover each well with 10 pl of a MEMBRANE FILTERS
1 pg ml-' 4', 6'-diamidino-Z-phenylindole(DAPI) solu- 1. Cut sections from membrane filters with a razor
tion and incubate for 3 min; rinse slide with distilled HzO
blade. A 47-mm-diameter filter should allow the prepara-
and let air dry.
tion of 16 to 20 individual hybridizations; a 25-mm-diameter
12. Samples are mounted in a 4:l mix of Citifluor filter yields eight sections. Label filter sections with a pen-
(Citifluor Ltd, London, United Kingdom) and Vecta Shield
cil, e.g., by numbering them.
(Vector Laboratories, Inc., Burlingame, CA). Vecta Shield
2. Put filter sections on glass slides (facing up). Several
contains a superior antibleaching reagent, but quenches
filter sections can be placed on one slide and hybridized si-
DAPI fluorescence. The wells have to be completely dry be- multaneously with the same probe.
fore embedding; otherwise a fraction of cells will detach
3. Prepare 2 ml of hybridization buffer in a microcen-
during inspection.
trifuge tube (see above).
13. Double-stained and air-dried preparations as well as 4. Per filter section, mix 20 p1 of hybridization buffer
mounted slides can be stored in the dark at -20C for sev-
with 2 p1of probe working solution.
eral days without substantial loss of probe fluorescence.
14. Probe-conferred fluorescence fades much more rap- 5. Put a piece of blotting paper into a polyethylene tube
and soak it with the remaining hybridization buffer.
idly than DAPI fluorescence in the microscopic image, and
6. Carefully cover the filter section with the hybridiza-
tion mix and place the slide with filter sections into the
polyethylene tube (in a horizontal position).
TABLE 4 Standard washing buffer 7. Incubate at 46C for at least 90 min (maximum, 3 h).
8. Prepare 50 ml of washing buffer in a polyethylene
Final concn in tube (see above).
Stock reagent Volume
hybridization buffer 9. Quickly transfer filter sections into preheated wash-
ing buffer and incubate for 15 min at 48C (water bath).
5 M NaCl Concentration 10. Pour washing buffer with filter sections into a petri
depending dish. Pick filter sections and rinse them by placing them
on probe (see into a petri dish with distilled H 2 0for several seconds, then
Table 5) let them air dry on blotting paper.
1 M Tris-HC1 1 ml 20 mM 11. For counterstaining, put filter sections on a glass
0.5 M EDTA 500 p1 5 mM plate, cover with 50 pl of DAPI solution, and incubate for
Distilled HzO Add to 50 ml 3 min; afterward, wash filter sections for several seconds in
80% ethanol to remove unspecific staining, followed by
10% SDS (added 50 ~1 0.01%
rinsing in distilled H 2 0 and air drying.
last to avoid
precipitation)
12. Samples are mounted in a 4:l mix of Citifluor and
Vecta Shield. The filter sections have to be completely dry
892 w COMMUNITY A N D GENOMIC ANALYSIS

TABLE 5 Corresponding NaCl concentration in SO ml of washing buffer'


Washing at 35C Washing at 48C
% Formamide in hybridization
[NaCl] in mol pl S M NaCl [NaCl] in mol ~ 1 M5NaCl
0 - - 0.900 8,900
5 - - 0.636 6,260
10 - - 0.450 4,400
15 - - 0.318 3,080
20 0.145 1,350 0.225 2,150
25 0.105 950 0.159 1,490
30 0.074 640 0.112 1,020
35 0.052 420 0.080 700
40 0.037 270 0.056 460
45 0.026 160 0.040 300
50 0.019 90 0.028 180
55 0.013 30 0.020 100
60 0.009 0 0.014 40
65 0.007 0 - -
70 0.005 0 - -
'Calculated using the formula from Lathe (26). Note that the addition of EDTA contributes to the Na' concentration. Therefore the required volume of 5 M NaCl
solution in the washing buffer is reduced by 100 ~ 1 .

before embedding; otherwise part of the cells might detach 39.1 1. HYBRIDIZATION FOLLOWED BY
during inspection. TY R A MID E SIGNAL AMPLl Fl C A T l 0N
13. Double-stained and air-dried preparations as well as (CARD-FISH)
filters mounted on slides can be stored in the dark at -20C
for several days without substantial loss of probe fluores- Fixation
cence.
1. Sample is concentrated on a membrane filter and air
NOTE: At least 500 DAPI-stained cells should be dried.
counted in plankton samples to obtain a counting error of 2. Dip filter in 0.2% low-gelling-point agarose, place
less than *5%. Do not attempt to determine absolute cell filters face-up onto glass slides, and let air dry.
counts from filters after hybridization, but only the percent- 3. Dehydrate in 96% ethanol (1 min, RT) and let air
age of hybridized cells. The distribution of cells on sections dry.
of a 47-mm-diameter membrane filter is never as even as on
NOTE: The embedding in agarose prevents the lysis of
a small filter, resulting in a higher error of the total DAPI
cells during subsequent permeabilization.
counts. We find that, following our procedure, 80 to 90% of
the initial bacterial cell numbers are recovered after hy- Inactivation of Endogenous Peroxidases
bridizations of bacterioplankton on membrane filters. This
fraction, however, may depend on the type of sample and 4. Incubate membrane filters with 0.01 M HC1 for 10
should be verified experimentally. min at RT.
5. Wash twice in distilled HzO.
6. Let air dry.
39.10. HYBRIDIZATION IN LIQUID FOR
F L O W CYTOMETRY Permeabilization
1. Prepare hybridization buffer (Table 2). 7. Incubate in lysozyme (10 mg ml-', dissolved in 0.05
2. Distribute subsamples of 100 p1 of hybridization buffer M EDTA, 0.1 M Tris-HC1 [pH 7.51) at 37C for 60 min.
in a 1.5-ml centrifuge tube. 8. Wash twice in distilled H 2 0for 1 min at RT.
3. Add approximately lo6 to lo7 cells. 9. Wash in 96% ethanol (1 min, KT).
4. Add 4 pl of probe working solution (2.5 ng p1-I) and 10. Let air dry.
mix well.
5. Incubate at 46C for 2 to 3 h. Hybridization
6. Cells are pelleted by centrifugation for 2 min at 4,000 11. Cut filters in sections.
x g and resuspended in 100 pl of prewarmed hybridization 12. Place sections in reaction vial (0.5 ml, 10 to 20 sec-
buffer containing no probe for washing.
tions per vial).
7. After washing for 25 min at 46"C, cells are pelleted by
centrifugation for 2 min at 4,000 X g. NOTE: The hybridization and washing buffers used for
8. Cells are resuspended with 300 pl of 1 x PBS (pH 7.2 HRP-oligos are different from the standard hybridization
for CY3 and pH 9.0 for FLUOS), immediately placed on mixtures described above (see Table 6). First, because of the
ice, and analyzed within 3 h in the flow cytometer. instability of the enzyme at higher temperatures, the tem-
39. Single Cell Identification by FISH 893

TABLE 6 Hybridization buffer for CARD-FISH 19. Prepare substrate mix: 1 part of tyramide plus 100 to
300 parts of PBS (pH 7.6; important for proper functioning
Final concn in of HRP) containing 0.0015% H202.
Stock reagent Volume
hybridization buffer 20. Dab filter on blotting paper to remove excess solu-
5 M NaCl 360 p1 900 mM tion, but do not blot to dryness; incubate in substrate mix at
37"C, 10 min, in the dark.
1 M Tris-HCI 40 p1 20 mM 21. Dab filter on blotting paper to remove excess solu-
Dextran sulfate 0.2 g 10% (wtlvol) tion, but do not blot to dryness.
20% SDS 2d 0.02% (wt/vol) 22. Wash in 10 ml of l x PBS amended with 0.05% of
Formamide % depending on Triton X-100 (RT, 10 min, in the dark, mild agitation).
probe (see Table 23. Wash in 10 ml of distilled H 2 0 (RT, 1 min).
3, plus 20%) 24. Wash in 10 ml of 96% ethanol (RT, 1 min).
10% blocking 400 p1 2% (wtlvol) 25. Let air dry.
reagent 26. Cells can be counterstained with DAPI as described
above or directly embedded in antifading mountant.
Distilled HzO Add to 2 ml
NOTE: At this point filters can be stored at -20C.

perature for hybridization is lowered to 35C and that for 39.1 2. TROUBLESHOOTING
washing is lowered to 37C. The amount of formamide is
Some common failure symptoms during FISH, their possi-
therefore 20% higher than for the standard hybridization
ble causes, and suggestions for improvements are listed in
conditions to compensate for the lower incubation temper-
Table 8.
atures. Second, the buffer contains dextran sulfate, which
enhances the binding of the probe to the target, and block-
ing reagent to block potential unspecific binding sites of the 39.1 3. APPLICATIONS AND FURTHER
HRP. Therefore the amount of water is adjusted accord- READING
ingly. If the concentration of formamide should exceed
60%, the volume of blocking reagent is reduced. 39.1 3.1. Methodological
- Aspects
NOTE: The hybridization buffer is prepared as follows: Behrens and coworkers compare the native three-dimen-
distilled HzO,dextran sulfate, NaC1, sodium dodecyl sulfate sional structure of the ribosome (6) with the data from
(SDS), and Tris-HC1 are mixed, brought into solution at FISH on fixed ribosomes within cells (7). Apparently the
60"C, and subsequently cooled on ice. Then add formamide FISH procedure strongly denatures the three-dimensional
and blocking reagent. The hybridization buffer can be structure of the ribosome (7).
stored at -20C for up to 3 months without any apparent
effects on FISH detection rates. 39.1 3.2. Marine Environments
13. Mix 400 p,l of hybridization buffer and 4 p,l of probe In marine planktonic research, several manuscripts deal
working solution (50 ng p1-l) and add to filter sections. with FISH alone or in combination with a complementary
14. Incubate at 35C for 2 h. technique. A comprehensive FISH study on coastal waters
15. Wash filters in prewarmed washing buffer (5 min, was done by Eilers et al. in 2000 (15). This study explored
37C) (Table 7). the distribution of different phylogenetic groups in the
16. Do not let filter run dry. North Sea. Fuchs and coworkers describe the combination
of flow cytometric sorting and FISH for a dilution culture
Tyramide Signal Amplification and point out the potential of such an approach for the
open ocean (19). Karner and coworkers found a surprising
17. Dab filter on blotting paper to remove excess solu-
dominance of Archaea in deep ocean water masses (24).
tion, but do not blot to dryness.
Amino acid uptake by Archaea was shown by Ouverney
18. To equilibrate the probe-delivered HRP, filter sec-
and Fuhrman in 2000 (41). The ubiquitously found SARl1
tions are placed in 10 ml of 1X PBS amended with 0.05%
clade was successfully brought into pure culture by a FISH-
of Triton X-100 for 15 min at room temperature.
guided cultivation approach (46). A multidisciplinary ap-
proach including FISH was used to show that anaerobic am-
TABLE 7 Washing buffer for CARD-FISH monium oxidation (51) occurs also in anoxic sea basins like
the Black Sea (25). Fuller and coworkers developed probes
Final concn in for the Synechococcus clade, a dominant prokaryotic primary
Stock reagent Volume producer in the marine environment (20). Cottrell and
hybridization buffer
Kirchman demonstrated with their study the necessity of
5 M NaCl Concentration FISH for proving the abundance of bacterioplankton groups
depending on in the environment (11).
probe (see
Table 5) 39.1 3.3. Sediment
1 M Tris-HCI 1 ml 20 mM In a pioneering work, the composition of the microbial
0.5 M EDTA 500 pl 5 mM community in the sediments of tidal mud flats (Wadden
Distilled H 2 0 Add to 50 ml Sea) was analyzed by Llobet-Brossa et al. (28). FISH was
also used to study permanently cold marine sediments (48).
10% SDS (added 50 p1 0.01%
FISH played an important role in the discovery that struc-
last to avoid
tured microbial consortia perform the anaerobic oxidation
precipitation)
of methane (10,39).
894 COMMUNITY AND GENOMIC ANALYSIS

TABLE 8 Common failure symptoms during FISH, their possible causes, and suggestions for improvements
Svmutom Cause

No cells stained 1. Probe is damaged; check probe in photometer, whether oligonucleotide and label are present
in equimolar amounts; check probe on a polyacrylamide gel: only a single band representing
intact probe should be visible.
2. Cells are overfixed:excess fixing in PFA cross-links cell wall components until cells are
completely impermeable to probes; this may occur when cells are stored for a long period in
PFA; try an increasing enzymatic digestion of the cells with, e.g., lysozyme, proteinase K, or
achromopeptidase or incubate in 0.1 M Hcl.
3. No SDS in hybridization buffer; SDS denatures the native ribosome structure by removing
the ribosomal proteins; it helps the probe penetrating into the cell.
4. CARD-FISH: check age of reagents, especially dextran sulfate, HRP probes, tyramides.
Unspecific staining of cells . . . . . . . 1. Probe is damaged; check probe in photometer, whether oligonucleotide and label are present
in equimolar amounts; check probe on a polyacrylamide gel: only a single band representing
intact probe should be visible.
2. Check sequence of the probe ordered.
3. Use a pencil (not a marker) to label slides and filters; marker dyes are often fluorescent.
4. Prepare fresh formamide stock.
FISH signal low . .. . .. . .. . .. .. . 1. Probe target site is inaccessible; try to use helper probes (16).
2. For probes labeled with fluorescein derivatives: check if pH of mountant/resuspensionbuffer
exceeds pH 9.0 (fluorescence maximum of fluorescein is pH > 9.0).
3 . Try to use CARD-FISH.
4. Use alternative mounting reagent.
No cells visible . . . . . . .. . .. .. . .. 1. Fixation was too short and cells lysed; try to fix longer or fix the remaining sample again.
2. Lysing procedure was too harsh; reduce lysozyme concentration and incubation time.
3. Slides were not covered with gelatin; cells were washed off.

39.1 3.4. Activated Sludge acknowledge Annelie Pernthaler and Sebastian Behrens for valuable
comments on earlier versions of the manuscript.
FISH has been widely applied t o activated sludge samples
( 3 3 , 6 3 , 6 4 ) ; in recent years, Crocetti e t al. (12) used FISH Note in proof. The protocols and references herein represent the
t o study polyphosphate-accumulating bacteria in a phos- state of the art in 2003. For updates and further information, the
phate removal reactor. Filamentous activated sludge bacte- reader is referred to publications by the authors and others on
ria Eikelboom type 021N were further phylogenetically af- FISH, and to www.arb-silva.de.
filiated and characterized by FISH from t h e Kanagawa
group (23). Schramm and coworkers coupled microelec- 39.14. REFERENCES
trodes and FISH for structure-function analysis in biofilms 1. Amann, R., B. M. Fuchs, and S. Behrens. 2001. The
(53, 55). A direct link of activity and identity could be identification of microorganisms by fluorescence in situ
achieved by the combination of microautoradiography and hybridisation. Cum. Opin. Biotechnol. 12:23 1-236.
FISH (27,40). 2. Amann, R. I., B. J. Binder, R. J. Olson, S. W. Chisholm,
R. Devereux, and D. A. Stahl. 1990. Combination of 1 6 s
39.1 3.5. Limnology rRNA-targeted oligonucleotide probes with flow cytome-
try for analyzing mixed microbial populations. Appl.
Some of the earliest community studies with FISH were Environ . Microbiol. 56: 1919-1925.
conducted by Pernthaler and coworkers on Lake 3. Amann, R. I., L. Krumholz, and D. A. Stahl. 1990.
Gossenkolle (44,45). T h e complicated trophic interactions Fluorescent-oligonucleotide probing of whole cells for de-
i n a eutrophic reservoir were studied by a group around terminative, phylogenetic, and environmental studies in
Simek in 1997 (57). A FISH-based comparison of domi- microbiology. J. Bacteriol. 172:762-770.
nant groups of bacterioplankton in oceans and lakes was 4. Amann, R. I., W. Ludwig, and K. H. Schleifer. 1995.
done by Glockner et al. (21). Grossart studied river snow by Phylogenetic identification and in situ detection of indi-
a combined approach of microsensor measurements, radio- vidual microbial cells without cultivation. Microbiol. Rew.
tracer experiments, and FISH (22). Community analysis of 59: 143-169.
sulfate reducers in a high mountain lake were done by 5. Amann, R. I., and K.-H. Schleifer. 2001. Nucleic acid
Tonolla and coworkers (61). Recently, the protocol for t h e probes and their application in environmental microbiol-
abundant limnic Actinobacteria cluster was improved by ogy, p. 67-82. In G. Garrity, D. R. Boone, and R. W.
Sekar and coworkers (56). Castenholz (ed.), Bergeys Manual of Systematic Bacteriology,
2nd ed, vol. 1. Springer-Verlag, New York, NY.
The original method development by our group reviewed in this man- 6. Ban, N., P. Nissen, J. Hansen, P. B. Moore, and T. A.
uscript was supported by the Max Phnck Society, the Germun Science Steitz. 2000. The complete atomic structure of the large
foundation, the Federal Ministry of Education and Research, and the ribosomal subunit at 2.4 angstrom resolution. Science 289:
European Community (BASICS, EVK3-CT-200240078), We 905-920.
39. Single Cell Identification by FISH 895

7. Behrens, S., B. M. Fuchs, F. Mueller, and R. Amann. 22. Grossart, H. P., and H. Ploug. 2000. Bacterial production
2003. Is the in ditu accessibility of the 16s rRNA of Esch- and growth efficiencies: direct measurements on riverine
erichia colt for Cy3-labeled oligonucleotide probes pre- aggregates. Lirnnol. Oceunogr. 45:436-445.
dicted by a three-dimensional structure model of the 30s 23. Kanagawa, T., Y. Kamagata, S. Aruga, T. Kohno, M.
ribosomal subunit? Appl. Enuiron. Microbiol. 69:4935- Horn, and M. Wagner. 2000. Phylogenetic analysis of and
4941. oligonucleotide probe development for Eikelboom type
8. Behrens, S., C. Ruhland, J. Inacio, H. Huber, A. 021N filamentous bacteria isolated from bulking activated
Fonseca, I. Spencer-Martins, B. M. Fuchs, and R. sludge. Appl. Enuiron. Microbiol. 665043-5052.
Amann. 2003. In situ accessibility of small-subunit rRNA 24. Karner, M. B., E. F. DeLong, and D. M. Karl. 2001.
of members of the domains bacteria, Archaea, and Eucarya Archaeal dominance in the mesopelagic zone of the
to Cy3-labeled oligonucleotide probes. Appl. Enuiron. Pacific Ocean. Nature 409507-5 10.
Microbiol. 69:1748-1758. 25. Kuypers, M. M. M., A. 0. Sliekers, G. Lavik, M.
9. Benson, D. A., I. Karsch-Mizrachi, D. J. Lipman, J. Schmid, B. B. Joergensen, J. G. Kuenen, J. S. S. Damste,
Ostell, B. A. Rapp, and D. L. Wheeler. 2002. GenBank. M. Strous, and M. S. M. Jetten. 2003. Anaerobic ammo-
Nucleic Acids Res. 30:17-20. nium oxidation by anammox bacteria in the Black Sea.
10. Boetius, A., K. Ravenschlag, C. J. Schubert, D. Rickert, Nature 422:608-611.
F. Widdel, A. Gieseke, R. Amann, B. B. Jorgensen, U. 26. Lathe, R. 1985. Synthetic oligonucleotide probes deduced
Witte, and 0. Pfannkuche. 2000. A marine microbial from amino acid sequence data: theoretical and practical
consortium apparently mediating anaerobic oxidation of considerations. J. Mol. Biol. 183:l-12.
methane. Nature 407:623-626. 27. Lee, N., P. Nielsen, K. Andreasen, S. Juretschko, J.
11. Cottrell, M. T., and D. L. Kirchman. 2000. Community Nielsen, K. Schleifer, and M. Wagner. 1999. Combina-
composition of marine bacterioplankton determined by tion of fluorescent in situ hybridization and microautoradi-
16s rRNA gene clone libraries and fluorescence in situ ography-a new tool for structure-function analyses in mi-
hybridization. Appl. Enuiron. Microbiol. 6 6 5 1 16-5122. crobial ecology. Appl. Enuiron. Microbiol. 65: 1289-1297.
12. Crocetti, G. R., P. Hugenholtz, P. L. Bond, A. Schuler, 28. Llobet-Brossa, E., R. Rossell6-Mora, and R. Amann.
J. Keller, D. Jenkins, and L. L. Blackall. 2000. Iden, 1998. Microbial community composition of Wadden Sea
tification of polyphosphate-accumulating organisms and sediments as revealed by fluorescence in situ hybridization.
design of 16s rRNA-directed probes for their detection Appl. Enuiron. Microbiol. 64:2691-2696.
and quantitation. Appl. Enuiron. Microbiol. 3:1175-1182. 29. Loy, A., M. Horn, and M. Wagner. 2003. probeBase-an
13. DeLong, E., L. Taylor, T. Marsh, and C. Preston. 1999. online resource for rRNA-targeted oligonucleotide probes.
Visualization and enumeration of marine planktonic ar- Nucleic Acids Res. 31514-516.
chaea and bacteria by using polyribonucleotide probes and 30. Maidak, B. L., J. R. Cole, T. G. Lilburn, C. T. J. Parker,
fluorescent in situ hybridization. Appl. Enuiron. Microbiol. P. R. Saxmann, R. J. Farris, G. M. Garrity, G. J. Olsen,
65 :5554-5563. T. M. Schmidt, and J. M. Tiedje. 2001. The RDP I1
14. DeLong, E. F., G. S. Wickham, and N. R. Pace. 1989. (Ribosomal Database Project). Nucleic Acids Res. 29: 173-
Phylogenetic stains: ribosomal RNA-based probes for the 174.
identification of single cells. Science 243:1360-1363. 31. Manz, W., R. Amann, W. Ludwig, M. Vancanneyt, and
15. Eilers, H., J. Pernthaler, F. 0. Glockner, and R. Amann. K.-H. Schleifer. 1996. Application of a suite of 16s
2000. Culturability and in situ abundance of pelagic bac- rRNA-specific oligonucleotide probes designed to investi-
teria from the North Sea. Appl. Enuiron. Microbiol. 66: gate bacteria of the phylum Cytophaga-Flauobacter-
3044-305 1. Bacteroides in the natural environment. Microbiology
16. Fuchs, B. M., F. 0. Glockner, J. Wulf, and R. Amann. 142:1097-1106.
2000. Unlabeled helper oligonucleotides increase the in situ 32. Manz, W., R. Amann, W. Ludwig, M. Wagner, and
accessibility to 16s rRNA of fluorescently labeled oligonu- K.-H. Schleifer. 1992. Phylogenetic oligodeoxynucleotide
cleotide probes. Appl. Enuiron. Microbiol. 66:3603-3607. probes for the major subclasses of proteobacteria: problems
17. Fuchs, B. M., K. Syutsubo, W. Ludwig, and R. Amann. and solutions. Syst. Appl. Microbiol. 15593-600.
2001. In situ accessibility of Escherichia coli 23s rRNA 33. Manz, W., M. Wagner, R. Amann, and K.-H. Schleifer.
to fluorescently labeled oligonucleotide probes. Appl. 1994. In situ characterization of the microbial consortia
Enuiron. Microbiol, 67 :96 1-968. active in two wastewater treatment plants. Water Res.
18. Fuchs, B. M., G. Wallner, W. Beisker, I. Schwippl, W. 28: 1715-1723.
Ludwig, and R. Amann. 1998. Flow cytometric analysis of 34. Meier, H., R. Amann, W. Ludwig, and K. Schleifer.
the in situ accessibility of Escherichia coli 16s rRNA for flu- 1999. Specific oligonucleotide probes for in situ detection
orescently labeled oligonucleotide probes. Appl. Enuiron. of a major group of Gram-positive bacteria with low DNA
Microbiol. 6449734982. G+C content. Syst. AppL Microbiol. 22:186-196.
19. Fuchs, B. M., M. V. Zubkov, K. Sahm, P. H. Burkill, and 35. Neef, A. 1997. Application of in situ identification of bac-
R. Amann. 2000. Changes in community composition teria to population analysis in complex microbial commu-
during dilution cultures of marine bacterioplankton as as- nities. Ph.D. dissertation. Technical University of Munich,
sessed by flow cytometric and molecular biological tech- Munich, Germany.
niques. Enuiron. Microbiol. 2:191-202. 36. Neef, A., R. Amann, H. Schlesner, and K.-H. Schleifer.
20. Fuller, N. J., D. Marie, F. Partensky, D. Vaulot, A. F. 1998. Monitoring a widespread bacterial group: in situ
Post, and D. J. Scanlan. 2003. Clade-specific 16s riboso- detection of planctomycetes with 16s rRNA-targeted
mal DNA oligonucleotides reveal the predominance of a probes. Microbiology 144:3257-3266.
single marine Synechococcus clade throughout a stratified 37. Neef, A., A. Zaglauer, H. Meier, R. Amann, H.
water column in the Red Sea. Appl. Enuiron. Microbiol. Lemmer, and K. H. Schleifer. 1996. Population analysis
69:2430-2443. in a denitrifying sand filter: conventional and in situ iden-
21. Glockner, F. O., B. M. Fuchs, and R. Amann. 1999. tification of Paracoccus spp. in methanol-fed biofilms.
Bacterioplankton composition in lakes and oceans: a first Appl . Enuiron . Microbiol. 6 2 4 329-4339.
comparison based on fluorescence in situ hybridization. 38. Olsen, G. J., D. J. Lane, S. J. Giovannoni, N. R. Pace,
Appl. Enuiron. Microbiol. 6 5 3 72 1-3 726. and D. A. Stahl. 1986. Microbial ecology and evolution:
896 COMMUNITY A N D GENOMIC ANALYSIS

a ribosomal rRNA approach. Annu. Rev. Microbiol. 40: 53. Schramm, A., D.de Beer, J. C. van den Heuvel, S.
337-3 65. Ottengraf, and R. Amann. 1999. Microscale distribution
39. Orphan, V. J., C. H. House, K. U. Hinrichs, K. D. of populations and activities of Nitrosospiru and Niaospiru
McKeegan, and D. F. Delong. 2001. Methane-consuming spp. along a macroscale gradient in a nitrifying bioreactor:
archaea revealed by directly coupled isotopic and phyloge- quantification by in situ hybridization and the use of mi-
netic analysis. Science 293:484-487. crosensors. Appl. Environ. Microbiol. 65:3690-3696.
40. Ouverney, C. C., and J. A. Fuhrman. 1999. Combined 54. Schramm, A., B. M. Fuchs, J. L. Nielsen, M. Tonolla,
microautoradiography-16srRNA probe technique for de- and D. A. Stahl. 2002. Fluorescence in situ hybridization
termination of radioisotope uptake by specific microbial of 16s rRNA gene clones (Clone-FISH) for probe valida-
cell types in situ. Appl. Environ. Microbiol. 65:1746-1752. tion and screening of clone libraries. Environ. Microbiol.
41. Ouverney, C. C.,and J. A. Fuhrman. 2000. Marine 4:713-720.
planktonic Archaea take up amino acids. Appl. Enuiron. 55. Schramm, A., L. H. Larsen, N. P. Revsbech, N. B.
Microbiol. 66:48294833. Ramsing, R. Amann, and K.-H. Schleifer. 1996.
42. Pernthaler, A., J. Pernthaler, and R. Amann. 2002. Structure and function of a nitrifying biofilm as deter-
Fluorescence in situ hybridization and catalyzed reporter mined by in situ hybridization and the use of microelec-
deposition for the identification of marine bacteria. Appl. trodes. Appl. Environ. Microbiol. 62:46414647.
Environ. Microbiol. 68:3094-3 101. 56. Sekar, R., A. Pernthaler, J. Pernthaler, F. Warnecke, T.
43. Pernthaler, A., C. M. Preston, J. Pernthaler, E. F. Posch, and R. Amann. 2003. An improved protocol for
DeLong, and R. Amann. 2002. Comparison of fluores- quantification of freshwater actinobacteria by fluorescence
cently labeled oligonucleotide and polynucleotide probes in situ hybridization. Appl. Environ. Microbiol. 69:2928-
for the detection of pelagic marine Bacteria and Archaea. 2935.
Appl. Enuiron. Microbiol. 68:661-667. 57. Simek, K., P. Hartman, J. Nedoma, J. Pernthaler, D.
44. Pernthaler, J., A. Alfreider, T. Posch, S. Andreatta, and Springmann, J. Vrba, and R. Psenner. 1997. Community
R. Psenner. 1997. In situ classification and image cytom- structure, picoplankton grazing and zooplankton control of
etry of pelagic bacteria from a high mountain lake heterotrophic nanoflagellates in a eutrophic reservoir dur-
(Gossenkollesee, Austria). Appl. Environ. Microbiol. 63: ing the summer phytoplankton maximum. Aquat. Microb.
4778-4783. Ecol. 12:49-63.
45. Pernthaler, J., F. 0. Glockner, S. Unterholzner, A. 58. Stahl, D.A., and R. Amann. 1991. Development and ap-
Alfreider, R. Psenner, and R. Amann. 1998. Seasonal plication of nucleic acid probes, p. 205-248. In E.
community and population dynamics of pelagic bacteria Stackebrandt and M. Goodfellow (ed.), Nucleic Acid
and archaea in a high mountain lake. Appl. Enuiron. Techniques in Bacterial Systematics. John Wiley & Sons
Microbiol. 64:42994306. Ltd., Chichester, United Kingdom.
46. Rappe, M. S., S. A. Connon, K. L. Vergin, and S. J. 59. Stoesser, G., W. Baker, A. van den Broek, E. Camon,
Giovannoni. 2002. Cultivation of the ubiquitous SARl1 M. Garcia-Pastor, C. Kanz, T. Kulikova, R. Leinonen,
marine bacterioplankton clade. Nature 6898:630-632. Q. Lin, V. Lombard, R. Lopez, N. Redaschi, P. Stoehr,
47. Raskin, L., B. E. Rittmann, and D. A. Stahl. 1996. M. A. Tuli, K. Tzouvara, and R. Vaughan. 2002. The
Competition and coexistence of sulfate-reducing and EMBL Nucleotide Sequence Database. Nucleic Acids Res.
methanogenic populations in anaerobic biofilms. Appl. 30:21-26.
Environ. Microbiol. 62:3847-3857. 60. Strunk, O., R. Westram, H. Meier, G. Jobb, L. Richter,
48. Ravenschlag, K.,K.Sahm, C. Knoblauch, B. B. Jorgensen, M. May, S. Hermann, N. Stuckmann, 0. Gross, B.
and R. Amann. 2000. Community structure, cellular rRNA Nonhoff, M. Lenke, R. Jost, B. Reichel, W. Foerster, T.
content, and activity of sulfate-reducing bacteria in ma- Ginhart, A. Vilbig, S. Gerbers, and W. Ludwig. 2002.
rine Arctic sediments. Appl. Environ. Microbiol. 66:3592- ARB: a software environment for sequence data. [Online.]
3602. www.arb-home.de.
49. Roller, C.,M. Wagner, R. Amann, W. Ludwig, and 61. Tonolla, M., A. Demarta, S. Peduzzi, D. Hahn, and R.
K.-H. Schleifer. 1994. In situ probing of Gram-positive Peduzzi. 2000. In situ analysis of sulfate-reducing bacteria
bacteria with high DNA G + C content using 23s rRNA- related to Desulfocapsa thiozymogenes in the chemocline of
targeted oligonucleotides. Microbiology 140:2849-2858. meromictic Lake Cadagno (Switzerland). Appl. Enuiron.
50. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Mo- Microbiol. 66:820-824.
lecular Cloning: u Laboratory Manual, 2nd ed, vol. 3, p. 62. Trebesius, K., R. Amann, W. Ludwig, K. Miihlegger, and
11.21. Cold Spring Harbor Laboratory Press, Cold Spring K.-H. Schleifer. 1994. Identification of whole fixed bac-
Harbor, NY. terial cells with nonradioactive 23s rRNA-targeted polynu-
51. Schmid, M., U. Twachtmann, M. Klein, M. Strous, S. cleotide probes. Appl. Environ. Microbiol. 60:3228-3235.
Juretschko, M. Jetten, J. W. Metzger, K. H. Schleifer, 63. Wagner, M., R. Amann, H. Lemmer, and K.-H.
and M. Wagner. 2000. Molecular evidence for genus level Schleifer. 1993. Probing activated sludge with oligonu-
diversity of bacteria capable of catalyzing anaerobic cleotides specificfor proteobacteria: inadequacy of culture-
ammonium oxidation. Syst. Appl. Microbiol. 23:93- dependent methods for describing microbial community
106. structure. Appl. Environ. Microbiol. 59: 1520-1 525.
52. Schonhuber, W., B. Fuchs, S. Juretschko, and R. 64. Wallner, G., R. Erhart, and R. Amann. 1995. Flow cyto-
Amann. 1997. Improved sensitivity of whole-cell hy- metric analysis of activated sludge with rRNA-targeted
bridization by the combination of horseradish peroxidase- probes. Appl. Environ. Microbiol. 61:1859-1866.
labeled oligonucleotides and tyramide signal amplifica- 65. Woese, C. R. 1987. Bacterial evolution. Microbiol. Reu.
tion. Appl. Environ. Microbiol. 63:3268-3273. 51:221-271.
Measurement of rRNA Abundance by
Hybridization with Oligodeoxynucleotide Probes
DANIEL H . BUCKLEY AND THOMAS M. SCHMIDT

40.1. BACKGROUND INFORMATION ............................. 897


40.1.1. Introduction ........................................ 897
.2. Factors That Influence rRNA Abundance ................... 898
40.1.2.1. rRNA and Growth Rate ......................... 898
. 2. Measurement of rRNA versus rRNA ............... 898
40.1.3. Basic Approach to Measuring rRNA Abundance .............. 898
40.1.3.1. Overview ................................... 898
. 2. Quantity of RNA Needed for Probe Hybridization
Assays..................................... 898
. .............................. 899
3. Potential Pitfalls
40.1.4. RNA Handling Considerations ........................... 899
40.2. RNA EXTRACTION ....................................... 899
40.2.1. Basic Considerations .................................. 899
40.2.1.1. Comparison between RNA and DNA Extraction ....... 899
. ................................... 899
2. Cell Lysis
. 3. RNA Yield and Purity.......................... 900
40.2.2. RNA Extraction from Cultures ........................... 900
.3. Extraction of RNA from Soil ............................ 900
40.3. OLIGONUCLEOTIDE PROBES .............................. 901
40.3.1. Probe Selection ...................................... 901
.2. Probe Design ........................................ 901
.3. Probe 5 End Labeling ................................. 902
.4. Td Determination ..................................... 902
40.4. RNA HYBRIDIZATION .................................... 903
40.4.1. Basic Considerations .................................. 903
.2. Hybridization Methodology .............................. 904
40.5. DETERMINATION OF rRNA ABUNDANCE .................... 905
40.5.1. Calculation of rRNA Relative Abundance ................... 905
.2. Data Analysis ....................................... 906
40.6. REFERENCES ............................................ 906

40.1. BACKGROUND INFORMATION tivities may have far-reaching impacts on ecosystems at


local. regional. or even global scales. As a result. numerous
.
40.1 1 . Introduction techniques have evolved that allow for the study of mi .
The past 20 years have been witness to a phenomenal rev- croorganisms as they occur in their natural habitats and
olution in our understanding of microbial diversity. During without the need for cultivation.
this time. the number of recognized bacterial groups has Two fundamental questions that need to be answered
more than quadrupled and newly discovered genera have when examining a natural microbial community are: Who
become legion (15). Two primary driving forces have been is there? and What is the relative abundance of microbial
behind this revolution. The first was the discovery that the populations? More formally. these questions refer to two
sequences of rRNA genes could be used to infer the evolu- components of community diversity: richness and even-
tionary history of microorganisms. and the second was the ness. where richness is the total number of different species
invention of PCR. With these tools. it has been possible to present and evenness represents the relative proportions of
recover rRNA genes directly from natural environments those species (16). Molecular techniques for studying mi-
and determine the phylogenetic diversity of the microor- crobial communities may be more or less well suited to an-
ganisms present . Numerous studies have used this approach swering these two questions. For example. techniques that
to discover novel groups of microorganisms that had previ- utilize PCR provide qualitative data on the richness of mi-
ously escaped detection because they had not been culti- crobial communities. but because of several types of PCR
vated in the laboratory (for review see references 15 and artifacts that can cause the relative proportion of rRNA
29) . Many of these novel microbial groups are widespread genes to change during PCR amplification (10. 13.48.51).
and numerically abundant in our biosphere. and their ac- these techniques may not be suitable for quantitative

897
898 w COMMUNITY AND CENOMIC ANALYSIS

analyses of the evenness of communities. Quantitative mea- used to directly estimate cell number unless the number of
sures of microbial abundance can be obtained by designing rRNA genes has previously been determined for the target
DNA probes that target either rRNA or the rRNA-encoding organisms. The rRNA operon copy number for an organism
gene (rDNA) sequences from particular microorganisms or can be determined by performing a Southern hybridization
microbial groups. These probes can be hybridized to nucleic while using a probe specific for the rRNA genes; alterna-
acids in intact cells by fluorescent in situ hybridization tively, a compilation of rRNA operons previously deter-
(FISH) as described in chapter 39, or they can be hybridized mined is provided by Klappenbach et al. (20).
to nucleic acids extracted from a microbial community as rRNA rather than rDNA is the target of choice for most
described in this chapter. In both approaches, hybridization hybridization studies primarily because there are thousands
of probes generates quantitative data on microbial abun- or tens of thousands more copies of a given rRNA molecule
dance that can provide valuable insight on the ecology of per cell than there are copies of the corresponding rDNA.
microorganisms and microbial communities. As a practical concern, this increased number of target mol-
This chapter provides background information and basic ecules greatly enhances the ability to detect a particular
methodologies required to carry out the measurement of molecular signature in an environmental sample. The use of
rRNA abundance by hybridization with oligonucleotide rDNA as a target for hybridization in microbial ecology oc-
probes. Much of the discussion in this chapter focuses on curs most frequently in attempts to determine the in situ
hybridization to RNA, but many of the methods presented growth rate of specific microorganisms (21, 26,30). As the
are equally applicable to DNA. This chapter addresses the cellular concentration of rRNA is correlated with growth
extraction of RNA from environmental samples, the design rate, there is a direct relationship between the rRNA/rDNA
of oligonucleotide probes, probe labeling and hybridization, ratio in a cell and cellular growth rate that is characteristic
and methods for calculating rRNA abundance from hy- for the growth of an organism over a wide range of growth
bridization data. Several methodological studies have eval- rates. Provided that it is possible to determine the
uated the use of RNA hybridization in microbial ecology rRNA/rDNA ratio for a particular species over a range of
and have contributed to many of the protocols and method- growth rates in the laboratory, it should be possible to esti-
ological caveats presented in this chapter (1, 7, 9, 21, mate the in situ growth rate of the species by extracting
32-34,46). Wherever possible, we have tried to provide in- community nucleic acids and using hybridization with
formation on the assumptions and potential biases that species-specific probes to determine the amount of rRNA
exist in the methods, to allow the reader to evaluate the and rDNA from the given species that is present in the en-
suitability of these methods toward their individual research vironment.
objectives.
40.1.3. Basic Approach to Measuring rRNA
Abundance
40.1.2. Factors That Influence rRNA
Abundance 40.1.3.1. Overview
40.1.2.1. rRNA and Growth Rate rRNA hybridization is a method that is useful for determin-
ing the relative physiological activity of microorganisms in
It is important to note that the measurement of rRNA the environment (3,38, 50). The sequence conservation of
abundance does not provide a measure of microbial cell rRNA genes makes it possible to design oligonucleotide
numbers in the same manner as would direct cell counts probes that target the rRNA molecules from specific groups
made by FISH. rRNA abundance does not correspond to of organisms. These probes can be used to quantify the
cell numbers, because the number of ribosomes per cell is abundance of specific rRNAs in a microbial community.
regulated in response to cellular growth rate (18,39,43).As Once an oligonucleotide probe for a specific microbial
a result, rapidly growing cells tend to have higher numbers group has been designed and tested empirically, rRNA
of ribosomes than the same cells when growing at a slower abundance can be determined by radioactively labeling the
rate. In addition, different species tend to carry different probe, allowing the probe to hybridize to an RNA sample,
numbers of ribosomes at any given growth rate depending and then measuring the amount of radioactivity associated
on their particular needs for protein synthesis. The amount with the RNA sample.
of rRNA present in an environment is therefore a function
of both the number of cells present in the environment and 40.1.3.2. Quantity of RNA Needed for Probe
their mean growth rate at the time of sampling. rRNA Hybridization Assays
abundance of each sequence type therefore provides a mea-
Probe hybridization assays require considerably more nu-
sure of the relative contribution of each actively growing
cleic acids than PCR-based assays. While under ideal cir-
community member to the protein synthetic capacity of the
cumstances the detection limit of hybridization should be
entire community (50).
less than 1 pg of target nucleic acid, environmental nucleic
acid extracts are rarely ideal samples. For nucleic acids ex-
40.1.2.2. Measurement of rRNA versus rDNA tracted from environmental samples, actual detection limits
Although the number of rRNA molecules can vary from will vary depending on sample characteristics. The presence
lo3 to lo5 per cell depending on species and growth rate, of inhibitors and nonhomologous nucleic acids can raise the
the number of rRNA genes (rDNA) present per genome actual detection limit of end-labeled oligonucleotide probes
varies little for a given species. The genes for the 5s rRNA, to approximately 100 pg of 1 6 s rRNA. As a result, typical
the 16s rRNA, and the 23s rRNA tend to be organized amounts of community RNA added to individual slots in
into operons. Different microbial species have different slot-blot hybridizations range from 10 ng to 1 to 3 p,g. The
characteristic numbers of rRNA operons. Although many total amount of RNA needed to perform hybridization
microbial species have only one or a few rRNA operons per analyses on a given environmental sample will ultimately
genome, others have as many as 15 (19). As a result, the depend on the number of different probes to be tested and
amount of rDNA present in an environment cannot be the number of replications per probe.
40. Measurement of rRNA Abundance by Hybridization H 899

40.1.3.3. Potential Pitfalls considerations: cell lysis, RNA stabilization, yield, and pu-
The two most important considerations when measuring rity. Relative to techniques depending on DNA and RNA
rRNA abundance by hybridization are the quality of RNA amplification, RNA hybridization requires a large amount
extracted from environmental samples and the hybridiza- of nucleic acid (up to 1 to 3 pg of community RNA per hy-
tion conditions used to ensure that probes achieve their de- bridization), and so extraction protocols need to be de-
sired specificity. Issues associated with probe specificity are signed to provide large quantities of RNA. In addition,
covered in depth in section 40.3, so only issues related to when extracting RNA from an environment such as soil,
the quality of rRNA will be discussed in this section. humic acids in the soil tend to copurify with nucleic acids.
Several problems can arise during the extraction of These contaminating humic acids can inhibit nucleic acid
RNA from environmental samples. The first problem that hybridization (2, 49), so extraction protocols must be de-
may be encountered is the inability to obtain sufficient signed to eliminate or at least minimize the presence of
quantities of RNA to perform the desired analyses. In- humic acids. The environmental RNA extraction protocol
creasing the size or the number of samples used to generate described in this section was designed to yield RNA of suf-
RNA best solves this problem. A second problem associated ficient quantity and quality to allow multiple RNA hy-
with RNA extracts results from RNA degradation. bridization experiments. Though the protocol was designed
Degradation of RNA (as described in sections 40.1.4 and for soil, it should be easily adaptable to provide RNA from
40.2.1.3) can reduce the availability of targets for hybridiza. other environmental samples.
tion, but a more insidious problem can result if selective 40.2.1 . I . Comparison between RNA and DNA
degradation of certain portions of rRNA molecules occurs.
As described in section 40.4, it is usually advisable to relate Extraction
rRNA abundance measurements made with a probe that Fewer methods for RNA extraction from environmental
targets a specific microbial group to measurements made samples have been published than for DNA extraction. The
with a probe that targets all rRNA molecules in a commu- differences between RNA extraction protocols and DNA
nity. The two probes will usually target different regions of extraction protocols are necessitated by the need for steps
the rRNA molecule. Thus, if selective degradation of these to both inactivate and prevent contamination from RNA-
probe target regions occurs, then measurements of rRNA degrading RNases. RNases are both ubiquitous and notori-
relative abundance can be either increased or decreased ar- ously hardy enzymes, and special precautions need to be
tificially. It is therefore necessary to take precautions to pre- taken to ensure that laboratory equipment and solutions are
vent RNA degradation (as described in sections 40.1.4 and free of RNase contamination (see section 40.1.4). Solutions
40.2.1.3). The last problem associated with RNA extracts used for RNA extraction frequently contain powerful
is caused by the presence of inhibitory compounds that co- chaotropic agents such as guanidine isothiocyanate or
purify with RNA. Discussion of the potential problems as- guanidine hydrochloride to inactivate RNases either pres-
sociated with inhibitors of hybridization and methods for ent in the environmental matrix or originating from lysed
overcoming these problems are presented in section 40.4.1. bacteria (17). Another difference between RNA and DNA
extraction protocols occurs when an organic extraction
40.1.4. RNA Handling Considerations (with phenol or phenol/CHC13) is required, as a neutral or
Very stable RNase enzymes that rapidly degrade RNA are alkaline pH will cause both RNA and DNA to partition
everywhere, on hands, lab counters, and other contact sur- into the aqueous phase during an organic extraction, while
faces. Care must be taken to ensure that all materials that a more acidic pH (as used in most RNA extractions) will
might come in contact with extracted RNA are free of cause DNA to be localized to the interface between the
RNases. All glassware and heat-resistant materials (e.g., aqueous and organic phase ( 17). When DNA-free RNA or
glass wool, spatulas) should be baked in an oven at greater RNA-free DNA is needed, it is common practice to include
than 150C for 5 h before use. Disposable plasticware either a DNase or an RNase step, respectively. Methods for
should be used wherever possible, and all pipette tips and simultaneous extraction of DNA and RNA from soils have
microcentrifuge tubes should be handled with gloved hands also been described (12, 31, 41). The choice of a particular
prior to autoclaving. During sample handling, care should method will depend on the experimental objectives, the
be taken to prevent contamination of any materials with amount and type of nucleic acid needed for analysis, the
bare hands and untreated laboratory surfaces. Water used chemical and physical properties of the samples, and char-
during extraction and purification can be treated by adding acteristics of the microorganisms under study.
0.2% diethylpyrocarbonate (DEPC), stirring for at least 2 h
to disperse DEPC droplets, and autoclaving. All solutions 40.2.1.2. Cell Lysis
and buffers should be made up with DEPC-treated water Most extraction methods combine chemical, enzymatic,
(36). and/or physical treatments to achieve efficient cell lysis. A n
ideal extraction buffer causes all microorganisms in the sam-
ple to become detached from soil colloids, renders cells sus-
40.2. RNA EXTRACTION ceptible to breakage, maintains the integrity of nucleic acids
released from cells by inactivating all nuclease enzymes, and
40.2.1. Basic Considerations prevents adsorption of nucleic acids to the environmental
The techniques used to isolate RNA from environmental matrix. Typically, soils or sediments are suspended in alka-
samples share many of the concerns associated with the ex- line buffers (pH 8) containing high concentrations of Na
traction of DNA from environmental samples, as described salts to promote osmotic disruption and disperse soil col-
in chapter 41. The primary difference between RNA and loids. Phosphate buffers (100 to 200 mM) are commonly
DNA extractions stems from the fact that RNA is much used for direct extraction, as the excess phosphate is thought
more labile than DNA and so RNA extraction protocols to compete with nucleic acid binding to clays in soil (28),
must be designed to prevent RNA degradation. The extrac- though higher phosphate concentrations may lead to the
tion of RNA from environmental samples has four primary formation of insoluble phosphate precipitates. Detergents
900 H COMMUNITY AND GENOMIC ANALYSIS

such as sodium dodecyl sulfate (SDS) or sarcosine are used 1. Pellet cells from 2 to 5 ml of culture by spinning at
to destabilize cell membranes and render cells more suscep- 5,000x g for 15 min at 4C (50 to 100 mg wet weight of
tible to breakage, while chaotropic agents such as guanidine cells is required).
isothiocyanate or guanidine hydrochloride are used to inac- 2. Resuspend cell pellet in 0.7 ml of acetate buffer.
tivate the enzymes that degrade RNA. Once the soil or sed- 3. In a 2.2-ml conical tube on ice, combine the 0.7 ml
iment particles are suspended and dispersed, they are sub- of cells suspension from above with 0.7 ml of phenol, (pH
jected to one or more physical treatments to achieve further 5.1), 50 ~1 of 20% SDS, and 0.7 ml of silica-zirconium
cell lysis. Physical treatments include heating at 80C, re- beads (0.1 mm ) (beads should be baked as glassware, to de-
peated freeze-thawing, sonication, and bead-beating, either stroy RNase enzymes).
with glass or silica-zirconium beads. Methods that rely on 4. Place in a bead beater (Mini-Beadbeaterl [no.
mechanical lysis through bead mill homogenization provide 31lOBX] or Mini-Beadbeater8 [no. 6931 from BioSpec
higher rates of cell disruption than most other methods (25). Products [www.biospec.com], or FastPrep Instrument
Cell lysis efficiency affects RNA yield, but more impor- [FP12OA] from QBiogene [previously BiolOl; www.qbio-
tantly, the cell lysis procedure must be designed to minimize gene.com] and shake for 2 min at the highestspeed setting
the possibility of differential cell lysis. If certain microbial (it may be necessary to repeat this step for organisms that
groups are less susceptible to lysis than others and the lysis are difficult to lyse).
technique used is not sufficiently rigorous, certain microbial 5. Spin tube at 500 X g in a centrifuge for 10 min
groups may be consistently underrepresented in extracted (CAUTION: spinning at higher speeds may cause beads to
RNA. To ensure that the RNA present in extracts is repre- rupture tube).
sentative of the indigenous microbial community, high cell 6. Transfer the aqueous phase to a fresh 1.5-ml Eppen-
disruption efficiencies must be achieved. The most effective dorf tube containing 0.7 ml of phenol/chloroform/isoamyl
way to assess cell lysis efficiency is by comparing direct mi- alcohol (pH 5.1), mix well, and then spin at 14,000 X g for
croscopic counts of soil smears before lysis treatments to di- 5 min. Perform this step a total of two times.
rect counts of the extracted pellets after treatment. In 7. Transfer the aqueous phase to a fresh 1.5-ml
preparing soil smears, do not apply more than 0.25 mg of dry Eppendorf tube containing 0.7 ml of chloroform, mix well,
soil per cm2 of slide area, to prevent masking of cells by soil and then spin at 14,000 X g for 5 min. Perform this step for
particles (5). Suitable cell stains are acridine orange, 4,6- a total of two times.
diamidino-2-phenyindole (DAPI), or DTAF 5-[4,6- 8. Transfer the aqueous phase to a fresh 1.5-ml
dichlorotriazin-2-yl]amino fluorescein (DTAF) (5). Eppendorf tube and precipitate RNA by adding 0.1 volume
The extraction protocol described in section 40.2.3 re- of 3 M sodium acetate and 2 volumes of 100% ethanol, mix
lies on bead mill homogenization in the presence of a strong well, and then spin at 14,000 X g for 20 to 30 min in a cen-
chaotropic agent. Microscopic cell counts of DTAF-stained trifuge at 4C.
cells made before and after homogenization revealed that 9. Decant carefully and resuspend in 1 ml of 70%
97.3% 2 1.6% (standard error) of cells in selected soil sam- ethanol, mix well, and then spin at 14,000 X g for 5 to 10
ples were disrupted (7). The observed lysis efficiency is con- min.
sistent with previous measurements of lysis efficiency ob- 10. Decant carefully and dry briefly under a vacuum.
tained for bead mill homogenization of soil samples (25). RNA will not form a pellet like DNA, but rather will be dis-
Thus, while the lysis protocol employed does not provide tributed on the lower wall of the tube. Resuspend RNA in
complete cell lysis, the disruption rate is high enough to en- 250 ~1 of RNase-free water.
sure that any bias caused by differential cell lysis will be kept
Solutions
below a few percent of the total rRNA abundance.
Acetate Buffer. 50 mM sodium acetate, 10 mM EDTA, pH
40.2.1.3. RNA Yield and Purity to 5.1.
The removal of humic acids from RNA extracts requires ex- Phenol pH 5.1. Add acetate buffer to an equal volume of
tensive purification steps including nucleic acid precipita- phenol and shake well. Allow phases to separate and
tion with polyethylene glycol and purification on both hy- check the pH of the buffer. Remove the buffer and re-
droxyapatite and Sephadex G-75 columns. Assessment of peat until the buffer pH after mixing is 5.1.
humic acid contamination of RNA extracts requires mea- Phenol/chloroform/isoamyl alcohol (pH 5.1). Mix vol-
suring sample light absorbance at 230 nm and 260 nm. As umes of phenol, chloroform, and isoamyl alcohol at a
humics absorb light strongly at 230 nm and nucleic acid ab- 25:24:1 ratio. Equilibrate at pH 5.1 with acetate buffer as
sorbs light at 260 nm, increasing A260/AZ30 ratios tend to in- for phenol.
dicate increasing sample purity (52). Total RNA concen-
trations in extracts can be estimated by using the orcinol
reaction to determine ribose concentrations (1 1). Sizes of 40.2.3. Extraction of RNA from Soil
extracted RNA can also be evaluated by denaturing agarose Using the protocol below, RNA can be extracted from eight
gel electrophoresis (37). If total RNA has not been sheared, 10-g samples in approximately 8 to 10 h (7). All solutions
distinct bands for 23s and 16s subunit RNAs can be visu- should be prepared using RNase-free water, and standard
alized. precautions should be taken to prevent RNase contamina-
tion of samples at all steps.
40.2.2. RNA Extraction from Cultures
Extraction of RNA
The following is a fairly rapid protocol suitable for the ex-
traction of total RNA from pure cultures. To maximize the 1. Add the following to the chamber of a beadbeater
yield of rRNA, cultures should be harvested late in the ex- (Beadbeater [1107900] with small chamber kit [110803]
ponential phase of growth. All solutions should be prepared from BioSpec Products [www.biospec.com]): 10 g of soil, 20
with RNase-free water, and standard precautions should be g of 0.1-mm silica/zirconium beads (BioSpec Products), and
taken at all steps to prevent RNase contamination of sam- 20 ml of soil homogenization buffer (SHB).
ples (see section 40.1.4). 2. Bead beat for two 1-min cycles on ice.
40. Measurement of rRNA Abundance by Hybridization H 901

3. Decant soil slurry to a sterile 50-ml tube on ice. min. Plug 3-ml syringe barrels with baked glass wool, and
4. Centrifuge at 3,000 X g for 5 min. slowly add Sephadex until the syringe is completely filled.
5. Decant supernatant fluid into another sterile 50-ml Wash the column three times with 1 ml of DEPC-treated
tube on ice. water by centrifugation at 1,400 X g for 4 min. Columns are
6. Resuspend soil pellet in 10 ml of SHB. ready for use and may be stored hydrated (do not spin after
7. Repeat steps 4 and 5, pooling the supernatant frac- adding water the last time) at 4C for several days.
tions on ice.
8. Add 0.1 volume of 5 M NaCl and 0.5 volumes of Solutions
50% polyethylene glycol (PEG 8000). Soil homogenization buffer (SHB).4 M guanidine isothio-
9. Mix well and incubate on ice for 2 h. cyanate, 200 mM sodium phosphate (pH 8), 25 mM
sodium citrate, and 0.5% N-lauryl sarcosine.
Sample Concentration and Phenol Extraction
10. Centrifuge sample for 30 min, 4"C, at 12,000 X g. 40.3. OLIGONUCLEOTIDE PROBES
11. Discard supernatant fluid and carefully rinse pellet
with cold 70% ethanol. 40.3.1. Probe Selection
12. Discard ethanol and resuspend pellet in 2 ml of 120 rRNA molecules consist of a mosaic of conserved and vari-
mM sodium phosphate buffer (pH 7.2) containing 0.7 M able domains whose nucleotide sequences evolve at differ-
NaC1. ent rates. As a result, it is possible to design rRNA-based
13. To each of two 2-ml microcentrifuge tubes, add 1 ml probes that have different levels of phylogenetic specificity.
of extract and 1/10 volume of 10% hexadecyltrimethylam- For example, probes can be designed to target a single
monium bromide (CTAB) solution containing 0.7 M NaC1. species, a particular microbial group, or an entire domain.
14. Heat at 60C for 5 min. In general, rRNA molecules have a very slow rate of evolu-
15. Add 0.7 ml of phenol/chloroform/isoamyl alcohol tion and so different strains of the same species commonly
(see section 40.2.2 for details on preparing solution) and have few if any differences in their rRNA sequences. Thus,
mix well. rRNA-targeted probes cannot distinguish organisms below
16. Centrifuge for 5 min at 15,000 x g, and collect the the species level. A large selection of phylogenetic probes
aqueous phase. can be found in the Oligonucleotide Probe Database (1) or
in a review by Amann et al. (3).
Hydroxyapatite Column Purification There are some general considerations to keep in mind
17. Add pooled sample gently to the top of a hydroxy- when preparing to use oligonucleotide probes to measure
apatite (HPT) column. rRNA abundance. A n oligonucleotide probe is designed
18. Centrifuge for 2 min at 100 X g, or until sample com- using the rRNA sequence database that is available at a
pletely passes through the column. given point in time. At any given time in the foreseeable fu-
19. Wash the column three times with 1 ml of 120 mM ture, it is safe to imagine that there will be more species in
sodium phosphate (pH 7.2). the world than there are represented in our species data-
20. Place a 1.5-ml tube with cap removed under the bases. As a result, a possibility remains that a probe target-
syringe barrel. Add 1 ml of 300 mM K2HP04 and elute at ing a particular group may also target other organisms pres-
100 x g for 4 min. ent in an environment that are unknown in sequence
databases. Whenever possible, it is advisable to use multiple
probes that target a particular group or to use nested probes
Sephadex Desalting and Purification that will allow independent corroboration of hybridization
21. Gently add eluent from HPT column to the top of data (3,45). In addition, when choosing a probe reported in
the packed Sephadex column. the literature, it is advisable to check the specificity of that
22. Place a 1.5-ml tube under the column and spin at probe against the sequences available in the current riboso-
1,400 X g for 6 min at room temperature. mal database (22).
23. Precipitate the RNA overnight at -20C with 1/10
volume of 3 M sodium acetate and 0.6 volume of isopropyl 40.3.2. Probe Design
alcohol. There are two critical stages in the process of designing a
24. Resuspend the RNA in 200 pl of DEPC-treated new oligonucleotide probe. The first stage involves deter-
water. mining the actual probe DNA sequence that will provide
selectivity for the organisms that you desire to target. The
Preparing Hydroxyapatite (HTP) Columns second stage requires an empirical determination of the hy-
Prepare a slurry of HTP (BioZRad HTP BioGel, not DNA- bridization conditions required to provide probe specificity
grade) in 10 mM sodium phosphate (pH 7.2). Add the and validation that the probe actually functions as de-
slurry to a 3-ml syringe barrel that has been plugged with signed. Many of these issues have been reviewed elsewhere
baked glass wool until the bed volume is 2.5 ml. Centrifuge (3,44,45,50).
the syringe in a 15-ml Corex glass tube at 100 X gfor 2 min The basic idea behind designing an oligonucleotide for a
to pack the column, which should have a final volume of species or a group of species is to find a stretch of sequence
1.5 to 2 ml. Wash the column three times with 1 of ml 120 from 15 to 25 bases long that is conserved in all of the tar-
mM sodium phosphate (pH 7.2). Columns are ready to use get organisms but is not present in any other sequence. The
and may be stored at 4C for several days, if needed. difficulty of this enterprise will vary considerably depending
on the particular group of organisms being targeted, in a
Preparing Sephadex G-75 Columns manner that is difficult to predict a priori. Perhaps the best
Prepare a slurry of Sephadex G-75 by mixing 4 g of way to design phylogenetic probes is to use the ARB phylo-
Sephadex with 50 ml of DEPC-treated water. Allow the genetic software package (47). This software package has
Sephadex to swell Overnight or by autoclaving for 15 to 30 sophisticated but simple-to-use tools that streamline the
902 COMMUNITY AND GENOMIC ANALYSIS

probe design process. The ARB package, installation in- purified following labeling to remove unincorporated
structions, and user manual are available free over the [y-32P]ATP.There are several different protocols for sepa-
Internet (www.mpi-bremen.de/molecol/arb). Without rating unincorporated nucleotides from oligonucleotides.
ARB, the most basic way to design a probe is to bring all of Most of these methods require some sort of size exclusion or
the target sequences into a DNA sequence editor and to ion exchange resin. Methods for end labeling and probe pu-
make a consensus sequence from the group of sequences. rification are provided. Noted that 32P-labeledprobes have
This consensus sequence will reveal the regions of conser- a limited shelf life, as the half-life of 32Pis approximately 14
vation between the target organisms. These conserved re- days. To achieve maximum sensitivity, hybridization exper-
gions should then be compared with homologous sequences iments should be carried out soon after probe labeling.
from closely related nontarget organisms to identify puta-
tive probe regions that are specific to the targeted organ- 5 End Labeling of Oligonucleotides
isms. A single base pair mismatch with nontarget organisms 1. Mix together 25 pmol of oligonucleotide, 50 pmol of
can be sufficient to provide specificity; however, it should [Y-~~P]ATP (3,000 Cilmmol), 5 p l of 1OX T4 polynu-
be noted that the character and location of the mismatch in cleotide kinase buffer (supplied with enzyme), and 20 units
the probe sequence will have an effect on the ability of the of T4 polynucleotide kinase (New England Biolabs) and
probe to discriminate between target and nontarget se- add RNase-free water to a final volume of 50 pl.
quences. For example, a mismatch located in the middle of 2. Incubate reaction at 37C for 30 min.
a probe sequence will tend to be more destabilizing than a 3. Stop reaction by heat inactivation at 65C for 20 min,
mismatch on the termini of the probe and will thus make or by adding EDTA to a final concentration of 10 mM.
for a better probe. It is also important to keep in mind that
for RNA-RNA or RNA-DNA hybrids, guanine is able to Oligonucleotide Purification
base-pair to uracil to form G-U base pairs. In addition, mis-
pairing between adenine and guanine is only weakly desta- 4. Combine probe-labeling reaction with 12 p1 of 250
bilizing relative to all other base pair mismatches. Putative mM ammonium acetate (pH 8.5 with ammonium hydrox-
probe targets should be checked by performing a BLAST ide) and add this mixture to the top of a TSK-DEAE col-
search of GenBank to determine whether the probes com- umn (see preparation of TSK-DEAE columns below).
plement sequence is present in organisms outside your tar- 5. Wash off unincorporated [y-32P]ATPby adding 1 ml of
get group. (As many regions of rRNA molecules are highly 50 mM ammonium acetate (pH 8.5) followed by 1 ml of 250
conserved, it may be necessary to screen many putative se- mM ammonium acetate (pH 8.5) and then 1 ml of 500 mM
quence targets until a successful probe sequence is found.) ammonium acetate (pH 8.5). Allow solutions to pass through
Once a probe sequence has been identified, it is neces- column by the force of gravity. Do not allow the meniscus to
sary to test the performance of the probe empirically. The reach the level of the resin in the column at any time.
objective of this exercise is to determine the hybridization 6. Elute labeled probe from the column with 1 ml of 1 M
conditions that provide the greatest amount of hybridiza- ammonium acetate (pH 8.5).
tion between probe and target sequences while minimizing 7. Dry down the probe in a SpeedVac (alternatively, the
hybridization between probe and nontarget sequences. probe can be precipitated with 0.1 volume of sodium ac-
When using oligonucleotide probes, it should be possible to etate and 2.5 volumes of 100% ethanol).
achieve discrimination between sequences that differ in a 8. Resuspend the probe in 50 pl of 50% methanol, then
single nucleotide position. The affinity with which nucleic repeat step 7 (this step is not needed if ethanol precipita-
acids bind to one another is influenced by the degree of se- tion is used).
quence complementarity, buffer composition, and tempera- 9. Resuspend the probe in 100 pl of RNase-free water
ture. Thus, the hybridization conditions required to achieve and determine the activity of 1 p1 by using a liquid scintil-
probe specificity are determined by hybridizing a probe to lation counter.
both target nucleic acids and nontarget nucleic acids and
then performing wash steps of increasing stringency by rais- PreparingTSK-DEAE Columns
ing either the temperature or changing the composition of Use a small quantity of glass wool to plug the bottom of a 1-
the wash buffer. For example, formamide can be added at ml plastic pipette tip. Slide a 4-cm-long piece of silicon tub-
various concentrations to both hybridization and wash ing (0.1 to 0.2 mm diameter) over the narrow end of the
buffers to increase the specificity of hybridization in lieu of pipette tip. Clamp the tubing and suspend the pipette tip on
altering temperature (this alternative is commonly used in a ring stand. Add 250-pl of water to the pipette tip and mark
FISH, reviewed in chapter 39). The temperature of dissoci- the location of the meniscus. Remove the clamp and allow
ation (Td)is a standard measure of the affinity between nu- the water to drain from the column. Add TSK-DEAE resin
cleic acid molecules in solution and those on a membrane (Supelco, DEAE-650M or equivalent) as a slurry in water to
(as opposed to the T,, which is a measure of the affinity be- the column until the bed volume reaches the 250-p1 mark.
tween different nucleic acid molecules in solution). The T d Wash the column two times with 1 ml of 50mM ammonium
of a probe for a given nucleic acid immobilized on a mem- acetate (pH 8.5). To ensure the settled integrity of the col-
brane is defined as the temperature at which one half of the umn, do not allow the meniscus to go below the top of the
probe becomes dissociated from that nucleic acid. Note that gel bed; use the clamp to stop the flow through the column.
formulas used to estimate the Td of a probe very rarely agree
with empirical measurements of T& 40.3.4. Td Determination
Before determining the Td of an oligonucleotide probe, it is
40.3.3. Probe 5 End Labeling necessary to isolate RNA from an organism that has a nu-
End labeling of oligonucleotide probes is accomplished cleic acid sequence complementary to the probe. The RNA
by using T4 polynucleotide kinase to transfer and exchange from this organism is then immobilized on a nylon mem-
toPthe 5 hy-
the 32Pifrom the y position of [ Y - ~ ~ P ] A T brane, and hybridization between 32P-labeledprobe and tar-
droxyl terminus of an oligonucleotide. Probes need to be get is carried out as described in section 40.4.2. To deter-
40. Measurement of rRNA Abundance by Hybridization m 903

mine the T d , it will be necessary to have at least four slots tification. Compounds such as humic acids have been ob-
containing 1 pg of RNA on the membrane. served to inhibit hybridization (2,49). The quantity of humic
acids present, and hence the degree of inhibition, can vary
1. After hybridization, wash the membrane twice in 50 depending on the characteristics of the parent sample, pre-
ml of wash buffer (see section 40.4.2for details) for 30 min
senting the possibility for a systematic bias in hybridization
at room temperature.
experiments with environmental nucleic acid extracts.
2. Cut out each of the slots that contain target RNA on Fortunately, it is possible to design RNA hybridization exper-
the membrane and place into separate vials.
iments to account for the presence of inhibitory compounds.
3. Add scintillation fluid to one of the membrane pieces Inhibitors of hybridization, such as humic acids, most
and use a liquid scintillation counter to determine the total
commonly act by competing with nucleic acids for mem-
counts per min (cpm) of the probe on the membrane.
brane occupancy during the immobilization of nucleic acid
4. Add 10 ml of wash buffer to each of the other mem- samples (2,6). Prior to cross-linking, positively charged
brane pieces and place in a shaking incubator at 30C and
nylon membranes bind and retain negatively charged nu-
100 rpm. cleic acids. Negatively charged compounds like humic acids
5. After the samples have reached the desired tempera- can saturate the positive charge on the membrane and pre-
ture, incubate for exactly 10 min.
vent nucleic acids from associating with membranes (Fig. 2).
6. Remove 500 pl from each vial and place in scintilla- It is possible to detect the presence of hybridization in-
tion fluid for liquid scintillation counting. Replace the re-
hibitors by blotting a sample at a range of concentrations
moved buffer by adding 500 p1 from a fresh supply of wash
and observing the amount of probe bound following hy-
buffer kept at temperature in the incubator.
bridization as a function of sample dilution. A nonlinear re-
7. Raise the temperature in 2C increments and repeat lationship characterized by a logarithmic decline in signal
steps 5 and 6 at each interval.
response is characteristic of inhibition (6). If humic acids or
8. At the end of the experiment, place each membrane other inhibitors are present in any degree, then any attempt
piece in liquid scintillation fluid to determine the amount
at absolute quantitation of the number of rRNA molecules
of probe remaining on the membrane.
present by hybridization results will result in an underesti-
9. Plot the percentage of cpm released as a function of mate. In addition, since the amount of humic acids present
the wash temperature (Fig. 1).The T d is the temperature at
in a sample may vary, it would be difficult to distinguish
which one half of the probe has been released from the
changes in rRNA absolute abundance that result from
membrane.
changes in the microbial community from apparent changes
that result from changes in the humic acid content of sam-
40.4. RNA HYBRIDIZATION ples. A simple way to compensate for the presence of

40.4.1. Basic Considerations


In hybridization experiments it is important to account for
the presence of contaminants that may affect rRNA quan-
60

50

\
40

E 30 Soil RNA Extract + 60 ngTarget RNA


8
20

10

Soil RNA Extract


I , 1 1
0 U u ' 1.1 . Is

0 200 400 600 800 1000


Soil RNA Extract (ng)
40 50 60 70 I
Temperature (OC) FIGURE 2 Effect of humic acid contamination on rRNA
hybridization. A constant amount of target RNA (60 ng) was
FIGURE 1 The temperature of dissociation (Td)for an added to increasing amounts of a soil RNA extract that was
oligonucleotide probe calculated from three replicate experi- heavily contaminated by humic acids. Hybridization with a ra-
ments. Data are obtained by hybridizing the probe to 1 pg of diolabeled oligonucleotide probe that bound only to the target
complementary RNA on a nylon membrane. The probe is then RNA was performed on the target RNA spiked with soil RNA
removed from the RNA by gradually increasing the wash tem- extract, soil RNA extract alone, and target RNA alone.
perature. The probe removed is expressed as a percentage of the Hybridization of probe to target RNA decreased with increas-
probe bound to the RNA prior to washing. ing amounts of the contaminated soil RNA extract.
904 COMMUNITY AND GENOMIC ANALYSIS

inhibitors when quantifying rRNA abundance is to normal- conditions used, it is advisable to select several negative
ize hybridization results to the total amount of rRNA pres- controls that vary in the number and type of mismatches to
ent in a sample as measured by a universal probe that tar- the probe.
gets the rRNA of all known organisms (as described in
section 40.5.1). Provided that the same amount of sample is Preparation of RNA
immobilized on the membranes used for hybridization with 1. Mix each RNA sample with 3 volumes of glutaralde-
both group-specific and universal probes, then calculating hyde solution and let nucleic acids denature for 15 min at
the ratio of group-specific probe signal to universal probe sig- room temperature. (RNA samples should contain between
nal will cancel out the effects of hybridization inhibitors. 300 ng and 3,000 ng of RNA; the lower value is recom-
mended for controls while the upper value is recommended
40.4.2. Hybridization Methodology for mixed community samples. Note that exact quantifica-
The general procedure for performing RNA hybridization tion of RNA is not crucial for measurement of rRNA rela-
requires that RNA be denatured and subsequently immobi- tive abundance.)
lized on a nylon membrane (46). The membrane is bathed 2. Add poly(A) water to a final volume of 1,300 r~.land
in hybridization solution containing a labeled probe and place samples on ice.
then a series of wash steps is used to remove probe that is
nonspecifically bound to the membrane. The temperature Blotting Denatured RNA on a Membrane
of the final wash step is set to ensure that probe is washed
3. Wet Magna Charge nylon membrane (Micron Sep-
off noncomplementary nucleic acid sequences. The temper-
aration, Inc.) and several pieces of absorbent filter paper
ature of this stringent wash step needs to be determined em-
with RNase-free water. (Note: Membranes other than Magna
pirically for each probe (see section 40.3). The stringent
Charge can be used as described in reference 32).
wash temperatures for a variety of different probes are given
in Table 1.
4. Place membrane and filter paper into a clean blot-
ting apparatus so that the membrane is facing up. Use
It is important to include both positive and negative
enough pieces of filter paper so that the wells of the blotting
controls on all membranes used in hybridization experi-
apparatus do not leak when assembled (this can be checked
ments. Controls consist of RNA extracted from pure cul-
before by running a dye through the wells of the blotting ap-
tures of organisms that have either a nucleic acid sequence
paratus).
that is complementary to the probe being used (in the case
of a positive control) or a nucleic acid sequence that differs
5. Add 400 ~ 1 320, ~ 1 240, ~ 1 160, ~ 1 and , 80 ~1 of
each sample to five adjadent welis on the membrane: Re-
from the probe target in only a few base positions (in the
peat for all samples and controls.
case of negative controls). In the case that rRNA from a
pure culture is not available to use as a control, as is fre-
6. Apply a slight vacuum to the blotting apparatus to
pull the samples onto the nylon membrane.
quently the case with noncultivated organisms, it is possible
to use rRNA that has been transcribed in vitro from a
7. Disassemble blotting apparatus and immobilize RNA
on the membrane by UV cross-linking with a total energy
cloned sequence (7). Methods for in vitro transcription are
of 1,200 X 100 ~J.Jcm-. Alternatively, RNA can be im-
described elsewhere (37). The best negative controls will be
mobilized on the membrane by baking for 30 min at 80C
those organisms that have nucleic acid sequences that are as
in a vacuum oven, or heating for 2 to 3 min at full power in
similar as possible to the probe target sequence. To verify
a microwave oven (750 to 900 W ) as described elsewhere
that the probe provides specificity under the hybridization
(37).
Hybridization with 32P-labeledOligonucleotide
Probes
TABLE 1 Final stringent wash temperatures for a variety of
oligonucleotide probes 8. Place the membrane in a hybridization bottle (35 by
150 mm) with the RNA side facing the center of the bot-
Probe Final wash temp (C) Reference tle. Add 10 ml of hybridization buffer and incubate in a
rolling incubator at 45C for at least 30 min.
Univ 1390 45 46 9. Pour out buffer and replace with 10 ml of fresh hy-
Eub338 45 45 bridization buffer. Add 32P-labeled oligonucleotide probe
Arc915 45 45 (see section 40.3.3 for labeling instructions) so that the
Euk1195 45 14 final hybridization buffer contains approximately lo6
Acd3 1 53 4 cpm/ml hybridization buffer.
10. Hybridize the membrane in a rolling incubator at
Alflb 55 23 45C overnight.
Bet42a 62 23
CF319 55 23 Washing the Membrane
Cren745 61 7 11. Pour hybridization buffer into a radioactive waste
Gam42a 62 23 container and replace with 50 ml of wash buffer prewarmed
HGC69a 50 35 to 45C; incubate for 30 min in the rolling incubator at
45C.
Nso1225 47 24 12. Pour wash buffer into a radioactive water container
Pla46 55 27 and replace with 50 ml of fresh wash buffer prewarmed to
Ver49 45 8 45C; incubate for 30 min in the rolling incubator at 45C.
Stringent wash temperatures for a selection of commonly used rRNA-
13. Pour wash buffer into a radioactive water container,
targeted oligonucleotide probes in buffer lacking formamide as described in sec- replace with 50 ml of fresh wash buffer prewarmed to the
tion 40.4.2. stringent temperature specified for the probe, and incubate
40. Measurement of rRNA Abundance by Hybridization rn 905

in the rolling incubator for 30 min at the desired tempera- 40.5. DETERMINATION OF rRNA
ture. (Wash temperatures for many probes are listed in ABUNDANCE
Table 1; otherwise it may be necessary to determine this
temperature empirically as described in section 40.3.4.) 40.5.1. Calculation of rRNA Relative Abundance
14. Pour wash buffer into a radioactive water container, RNA molecules extracted from pure cultures of microor-
remove membrane from hybridization bottle using forceps, ganisms are placed on every hybridization membrane to
and rinse membrane briefly with Milli-Q water in a shallow provide the two levels of control. These RNA samples are
dish. both from organisms that are targeted by the rRNA probe
15. Allow membrane to air dry and then wrap in Mylar being used and from several microorganisms that are closely
or Saran Wrap. related but outside of the target group. The positive controls
16. Determine the amount of probe bound to each spot are used to account for variations in the labeling efficiency
on the membrane by using a system that is capable of de- of different oligonucleotide probes, while the negative con-
tecting the emission of beta particles, either directly or trols are used to account for any hybridization signal that re-
through the exposure of phosphor plates. sults from nonspecific interactions. Calculations of relative
abundance are made by taking the ratio of signal intensities
Solutions obtained for specific and universal probe binding to an
Glutaraldehyde solution. 2% glutaraldehyde, 50 mM sodium RNA sample as R = C"i=l[Gi(Ui)-']n-', where Gi and Ui
phosphate, pH 7.0. Filter sterilize and store at -20C. represent, respectively, the corresponding signal intensities
obtained for group-specific and universal probe binding to
Poly(A) water. 1 pg ml-' polyadenylic acid, 0.06 pg ml-'
bromphenol blue, 0.5% glutaraldehyde. Filter sterilize each aliquot representing the sample, and n equals the total
and store at - 20C. number of aliquots representing the RNA sample. The
value R is then calculated for each soil RNA sample (RJ,
Hybridization buffer. 900 mM NaC1, 120 mM Tris-HC1 and a mean value of R is determined for all positive (R,) and
(pH 8.0),6 mM disodium EDTP, 0.5% SDS, 1 X negative (R,) controls present on each membrane. The rel-
Denhardt's solution, 100 pg ml- polyadenylic acid. ative abundance (expressed as a percentage) of rRNA from
Make fresh daily. a specific microbial group is then defined as (R, - R,)(R, -
Wash buffer. 300 mM NaC1, 40 mM Tris-HC1 (pH 8.0),2 RJ' X 100. A n example calculation of rRNA relative
mM disodium EDTA, 0.5% SDS. abundance is shown in Table 2.
Denhardt's solution. 1% Ficoll, 1% polyvinylpyrrolidone, The calculation of rRNA relative abundance from RNA
1% bovine serum albumin (fraction V). Filter sterilize hybridization data involves multiple levels of control that
and store at -20C. account completely for any possible problems caused by

TABLE 2 Example calculation of rRNA relative abundance for a single RNA samplea
Universal Group-specific
LJJ - '
Gi( Equation
Name Volume (pl) probe signal probe signal
Positive control 400 12080000 3450000 0.2856 C",=,[G,(U,)-'] = 1.4065;n = 5
320 9800000 2800000 0.2857
240 7360000 2 100000 0.2853 RP = C",=,[G,(U,)-'ln-'
160 5000000 1410000 0.2820 RP = (1.4065) (5)-'
80 2650000 7 10000 0.2679 Rp = 0.2813

Negative control 400 25590000 100000 0.0039 X',=1[G,(UZ)-']= 0.0214; n = 5


320 22 130000 80000 0.0036
240 17270000 60000 0.0035 RN = Cn,=l[G,(U,)-'lC1
160 10420000 50000 0.0048 RN = (0.0257)(5)-'
80 5360000 30000 0.0056 RN = 0.0043

Sample 400 63670000 930000 0.0146 S",=,[G,(U,)-'] = 0.0807; n = 5


320 55200000 790000 0.0143
240 36450000 700000 0.0192 Rs = C",,1[Gt(U,)-']n-'
160 25050000 400000 0.0160 Rs = (0.0807)(5)-'
80 11470000 190000 0.0166 Rs = 0.0161

( R , - RN)(Rp - RN)-' X 100 = percent rRNA abundance


(0.0161 - 0.0043)(0.2813 - 0.0043)-' X 100 = 4.3%
"Data are from one hybridization experiment with a universal probe and a second hybridization experiment with a group-specific probe. The hybridization experi-
ments were carried out on duplicate membranes that contained RNA from a positive control organism, a negative control organism, and a soil sample. Five different
volumes of each RNA sample were blotted on each membrane. With a 96-well blotting apparatus it is possible to create a single membrane that contains 19 different
samples and controls.
906 COMMUNITY AND GENOMIC ANALYSIS

impurities in soil RNA extracts and variability in soil sam- The particular type of hypothesis being addressed will
ple RNA abundance. To control for differences in RNA inform the choice of analytical method. Decisions about
quantity and quality, all determinations of rRNA relative the type of analysis to be performed should be made prior to
abundance are based on the ratio of the results from two the design of experiments. A regression analysis is appropri-
hybridization experiments carried out with two distinct ate for determining the likelihood of a linear relationship
probes. In such hybridization experiments, it is important between a continuous variable and the rRNA abundance of
that the same amount of RNA (and humic acids as the case a particular microorganism. When it is necessary to exam-
may be) is present in each corresponding well on the blots ine the degree to which multiple continuous variables cor-
used for specific probe hybridization and on the blots used relate with rRNA abundance, then either multiple regres-
for universal probe hybridization. Thus, the amount of in- sion analysis or canonical correspondence analysis will be
hibition for a given sample will be the same on both blots, necessary. In many cases, an investigator manipulates one or
and dividing the hybridization signal for the sample on a more factors to determine their effect on the rRNA abun-
specific blot by its signal on a universal blot will cancel out dance of a given organism. A n example would be an exper-
any inhibition effects, giving an accurate measure of rRNA iment in which the rRNA abundance of the autotrophic
relative abundance. ammonia oxidizers is examined in response to fertilizer ad-
There are two caveats to keep in mind when using the dition in a set of fields that received different amounts of
above technique to calculate rRNA relative abundance. fertilizer. In these cases, an analysis of variance (ANOVA)
First, a change in rRNA relative abundance can result from is the analytical method of choice. ANOVA is an extremely
a change in the amount of a target rRNA in a community powerful analytical technique for examining the relation-
(specific probe signal) or from a change in the total number ship between one or more factors and a single dependent
of rRNA molecules present in a community (as detected by variable, such as rRNA abundance. It is also possible to
a universal rRNA probe). Thus, it is important to consider examine variation in the rRNA abundance of multiple mi-
the hypotheses that are being tested to determine whether crobial groups simultaneously in response to one or more
relative abundance measures are appropriate. Comparisons experimentally manipulated factors through the use of mul-
of universal probe signal intensity can be used to determine tiple analysis of variance (MANOVA) or correspondence
the degree to which changes in community rRNA abun- analysis. MANOVA is useful for determining whether there
dance influence measurements of group-specific rRNA rel- is an overall change in the structure of a microbial commu-
ative abundance, though inhibitors such as humic acids, if nity in response to experimental manipulation. All of these
present, may have a significant impact on total signal in- analytical techniques can be carried out on computer pack-
tensity. If inhibitory substances are verified to be absent, ages such as StatView v 5.0 (SAS Institute, Inc.) and SAS
then it is possible to calculate the absolute abundance of v 7.0 (SAS Institute, Inc.).
rRNA present from a specific group by relating hybridiza-
tion signal intensity directly to known amounts of positive
control rRNA included on hybridization membranes. A 40.6. REFERENCES
second caveat to using this method of calculating rRNA 1. Alm, E. W., D. B. Oerther, N. Larsen, D. A. Stahl, and
relative abundance is that the negative controls used to cor- L. Raskin. 1996. The oligonucleotide probe database.
rect for nonspecific hybridization tend to be overly conser- Appl. Environ. Microbiol. 62: 355 7-3 559.
vative. For example, a typical negative control may contain 2. Alm, E. W., D. D. Zheng, and L. Raskin. 2000. The pres-
only a single mismatch in the probe target region, while a ence of humic substances and DNA in RNA extracts af-
microbial community will contain an array of sequences in fects hybridization results. Appl. Environ. Microbiol. 66:
the probe target region, most of which will have more than 454711554.
a single mismatch. As a result of this conservative use of 3. Amann, R. I., W. Ludwig, and K.-H. Schleifer. 1995.
negative controls, it is difficult to detect signal from target Phylogenetic identification and in situ detection of indi-
groups that are present at low relative abundance (generally vidual microbial cells without cultivation. Microbiol. Rew.
<0.5%). 59:143-169.
4. Barns, S. M., S. L. Takala, and C. R. Kuske. 1999. Wide
distribution and diversity of members of the bacterial
40.5.2. Data Analysis Kingdom Acidobacterium in the environment. Appl. En-
Measurements of rRNA relative abundance are commonly viron. Microbiol. 65 :1731-1 73 7.
made to examine whether some component of a microbial 5. Bloem, J. 1995. Fluorescent staining of microbes for total
community is affected by a particular factor or group of fac- direct counts, p. 1-12. In A. D. L. Akkermans, J. D.
tors. In the simplest case, it might be necessary to test VanElsas, and E J. deBruijn (ed.), Molecular Microbial
whether the activity of a particular microbial group is influ- Ecology Manual. Kluwer Academic Publishers, Dordrecht,
enced by a single experimental treatment. In a more com- The Netherlands.
plicated analysis, it may be necessary to examine whether 6. Buckley, D. H. 2000. The diversity and dynamics of mi-
microbial community structure varies in relation to a range crobial groups in soils from agroecosystems. Ph.D. disserta-
tion. Michigan State University, East Lansing, MI.
of experimental treatments administered over time. These 7. Buckley, D. H., J. R. Graber, and T. M. Schmidt. 1998.
are ultimately ecological questions about microbial commu- Phylogenetic analysis of nonthermophilic members of the
nities. A suite of analytical tools has been developed for the kingdom Crenurchaeota and their diversity and abundance
analysis of ecological experiments that are useful for the in soils. Appl. Environ. Microbiol. 644333-4339.
analysis of rRNA relative abundance data. A n in-depth re- 8. Buckley, D. H., and T. M. Schmidt. 2001. Environ-
view of these techniques is out of the scope of this chapter, mental factors influencing the distribution of Verru-
but brief descriptions of some techniques that may be of in- comicrobia in soil. FEMS Microbiol. Ecol. 35105-1 12.
terest are provided below. More information on the design 9. Buckley, D. H., and T. M. Schmidt. 2001. The structure
and analysis of ecological experiments can be found else- of microbial communities in soil and the lasting impacts of
where (40, 42). cultivation. Microb. Ecol. 42:ll-21.
40. Measurement of rRNA Abundance by Hybridization w 907

10. Chandler, D. P., J. K. Fredrickson, and F. J. Brockman. tection of Planctomycetes with 16s rRNA-targeted
1997. Effect of PCR template concentration on the com- probes. Microbiology 144:3 257-3 266.
position and distribution of total community 16s rDNA 28. Ogram, A,, G. S. Sayler, and T. Barkay. 1987. The ex-
clone libraries. Mol. Ecol. 6:475482. traction and purification of microbial DNA from sedi-
11. Daniels, L., R. S. Hanson, and J. A. Phillips. 1994. ments. 1.Microbiol. Meth. 7:57-66.
Chemical analysis, p. 536. In P. Gerhardt, R. G. E. Murray, 29. Pace, N. R. 1997. A molecular view of microbial diversity
W. A. Wood, and N. R. Krieg (ed.), Methods for General and the biosphere. Science 276:734-740.
and Molecular Bacteriology. American Society for Micro- 30. Poulsen, L. K., G. Ballard, and D. A. Stahl. 1993. Use
biology, Washington, DC. of ribosomal-RNA fluorescence in situ hybridization for
12. Duarte, G. F., A. S. Rosado, L. Seldin, A. C. Keijzer- measuring the activity of single cells in young and es-
Wolters, and J. D. van Elsas. 1998. Extraction of riboso- tablished biofilms. Appl. Enuiron. Microbiol. 59:1354-
mal RNA and genomic DNA from soil for studying the di- 1360.
versity of the indigenous bacterial community.J. Microbiol. 31. Purdy, K. J., T. M. Embley, S. Takii, and D. B. Nedwell.
Meth. 32:21-29. 1996. Rapid extraction of DNA and rRNA from sediments
13. Farrelly, V., F. A. Rainey, and E. Stakebrandt. 1995. by a novel hydroxyapatite spin-column method. Appl. En-
Effect of genome size and m gene copy number on PCR uiron. Microbiol. 62:3905-3907.
amplification of 16s rRNA genes from a mixture of bacte- 32. Raskin, L., W. C. Capman, M. D. Kane, B. E. Rittmann,
rial species. Appl. Enuiron. Microbiol. 61:2798-2801. and D. A. Stahl. 1996. Critical evaluation of membrane
14. Giovannoni, S. J., E. F. DeLong, G. J. Olsen, and N. R. supports for use in quantitative hybridizations. Appl. En-
Pace. 1988. Phylogenetic group-specific oligonucleotide uiron. Microbiol. 62:300-303.
probes for identification of single microbial cells. J. Bac- 33. Raskin, L., W. C. Capman, R. Sharp, and D. A. Stahl.
teriol. 170:720-726. 1997. Molecular ecology of gastrointestinal ecosystems,
15. Hugenholtz, P., B. M. Goebel, and N. R. Pace. 1998. p. 243-298. In R. I. Mackie, B. A. White, and R. E.
Impact of culture-independent studies on the emerging Isaacson (ed.), Ecology and Physiology of Gastrointestinal
phylogenetic view of bacterial diversity. J. Bacteriol. 180: Microbes, vol. 2. Chapman and Hall, New York, NY.
47654774. 34. Raskin, L., L. K. Poulsen, D. R. Noguera, B. E.
16. Huston, M. A. 1994. Biological Diuersity. Cambridge Rittmann, and D. A. Stahl. 1994. Quantification of
University Press, Cambridge, United Kingdom. methanogenic groups in anaerobic biological reactors by
17. Johnson, J. 1994. Similarity analysis of rRNAs, p. 685. In oligonucleotide probe hybridization. Appl. Enuiron.
P. Gerhardt, R. G. E. Murray, W. A. Wood, andN. R. Krieg Microbiol. 60:1241-1248.
(ed.), Methods for General and Molecular Bacteriology. 35. Roller, C., M. Wagner, R. Amann, W. Ludwig, and K.-H.
American Society for Microbiology, Washington, DC. Schleifer. 1994. In situ probing of Gram-positive bacteria
18. Kemp, P. F., S. Lee, and J. LaRoche. 1993. Estimating the with high DNA G + C content using rRNA-targeted
growth rate of slowly growing marine bacteria from RNA oligonucleotides. Microbiology 140:2849-2858.
content. Appl. Enwiron. Microbiol. 59:2594-2601. 36. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Mo-
19. Klappenbach, J. A., J. M. Dunbar, and T. M. Schmidt. lecular Cloning: a Laboratory Manual, 2nd ed. Cold Spring
2000. rRNA operon copy number reflects ecological stra- Harbor Laboratory Press, Cold Spring Harbor, NY.
tegies of bacteria. Appl. Enuiron. Microbiol. 66:1328-1333. 37. Sambrook, J., and D. W. Russell (ed.). 2001. Molecular
20. Klappenbach, J. A., P. R. Saxman, J. R. Cole, and T. M. Cloning: a Laboratory Manual, 3rd ed. Cold Spring Harbor
Schmidt. 2001. rmdb: the ribosomal RNA operon copy Laboratory Press, Cold Spring Harbor, NY.
number database. Nucleic Acids Res. 29:181-184. 38. Sayler, G. S., and A. C. Clayton. 1990. Environmental
21. Lepp, P. W., and T. M. Schmidt. 1998. Nucleic acid con- application of nucleic acid hybridization. Annu. Rev. Mi-
tent of Synechococcus spp. during growth in continuous crobiol. 44:625-648.
light and light/dark cycles. Arch. Microbiol. 170:201-207. 39. Schaechter, M. O., 0. Maaloe, and N. 0. Kjeldgaard.
22. Maidak, B. L., J. R. Cole, T. G. Lilburn, C. T. Parker, 1958. Dependency on medium and temperature of cell
P. R. Saxman, R. J. Farris, G. M. Garrity, G. J. Olsen, size and chemical composition during balanced growth
T. M. Schmidt, and J. M. Tiedje. 2001. The RDP-I1 of Salmonella typhimurium. J. Gen. Microbiol. 19592-
(Ribosomal Database Project). Nucleic Acids Res. 29:173- 606.
174. 40. Scheiner, S. M., and J. Gurevitch (ed.). 1993. Design and
23. Manz, W., R. Amann, W. Ludwig, M. Wagner, and K.-H. Analysis of Ecological Experiments. Chapman and Hall, New
Schleifer. 1992. Phylogenetic oligonucleotide probes for York, NY.
the major subclasses of proteobacteria: problems and solu- 41. Selenska-Pobell, S. 1995. Direct and simultaneousextrac-
tions. Syst. Appl. Microbiol. 15593-600. tion of DNA and RNA from soil, p. 1-17. In J. D. v. E. D.
24. Mobarry, B. K., M. Wagner, V. Urbain, B. E. Rittman, L. Akkermanr, and E. M. H. Wellington (ed.), Molecular
and D. S. Stahl. 1996. Phylogenetic probes for analyzing Microbial Ecology Manual. Kluwer Academic Publishers,
abundance and spatial organization of nitrifying bacteria. Dordrecht, The Netherlands.
Appl. Enuiron. Microbiol. 62:2156-2 162. 42. Sokal, R. R., and F. J. Rohlf. 1995. Biometry: the Principles
25. More, M. J., J. B. Herrick, M. C. Silva, W. C. Ghiorse, and Practice of Statistics in Biological Research. W. H.
and E. L. Madsen. 1994. Quantitative cell lysis of indige- Freeman & Co., New York, NY.
nous microorganisms and rapid extraction of microbial 43. Srivastava, A. K., and D. Schlessinger. 1990. Mechanism
DNA from sediment. Appl. Enwiron. Microbiol. 60: 1572- and regulation of bacterial ribosomal RNA processing.
1580. Annu. Rew. Microbiol. 44:105-129.
26. Muttray, A. F., and W. W. Mohn. 1999. Quantitation of 44. Stahl, D. A. 1997. Molecular approaches for the mea-
the population size and metabolic activity of a resin acid surement of density, diversity, and phylogeny, p. 102-1 14.
degrading bacterium in activated sludge using slot-blot hy- In C. J. Hurst, G. R. Knudsen, M. J. McInemey, L. D.
bridization to measure the rRNArDNA ratio. Microb. Stetzenbach, and M. V. Walter (ed.), Manual ofMethods in
Ecol. 38:348-357. Environmental Microbiology. ASM Press, Washington, DC.
27. Neef, A., R. Amann, H. Schlesner, and K. H. Schleifer. 45. Stahl, D. A., and R. Amann. 1991. Development and ap-
1998. Monitoring a widespread bacterial group: in situ de- plication of nucleic acid probes in bacterial systematics,
908 COMMUNITY AND GENOMIC ANALYSIS

p. 205-248. In E. Stackebrandt and M. Goodfellow (ed.), 49. Tebbe, C. C., and W. Vahjen. 1993. Interference of humic
Nucleic Acid Techniques in Bacterial Systematics.John Wiley acids and DNA extracted directly from soil in detection
& Sons Ltd., Chichester, United Kingdom. and transformation of recombinant DNA from bacteria
46. Stahl, D. A., B. Flesher, H. R. Mansfield, and L. and yeast. Appl. Environ. Microbiol. 59:2657-2665.
Montgomery. 1988. Use of phylogenetically based probes 50. Ward, D. M., M. M. Bateson, R. Weller, and A. L. Ruff-
for studies of ruminal microbial ecology. Appl. Environ. Roberts. 1992. Ribosomal RNA analysis of microorgan-
Microbiol. 54:1079-1084. isms as they occur in nature. Adv. Microb. Ecol. 12:219-
47. Strunk, O., and W. Ludwig. 1997. ARB: A Software En- 268.
vironment for Sequence Data, version 2.5. Department of 51. Wintzingerode, F. V., U. B. Gobel, and E. Stackebrandt.
Microbiology, Technical University of Munich, Munich, 1997. Determination of microbial diversity in environ-
Germany. mental samples: pitfalls of PCR-based rRNA analysis.
48. Suzuki, M. T., and S. J. Giovannoni. 1996. Bias caused by FEMS Microbiol. Rev. 21:213-229.
template annealing in the amplification of mixtures of 16s 52. Zhou, J., M. A. Bruns, and J. M. Tiedje. 1996. DNA re-
rRNA genes by PCR. Appl. Enuiron. Microbiol. 62:625- covery from soils of diverse composition. Appl. Environ.
630. Microbiol. 62:3 16-322.
41
Analysis of Microbial Communities with Denaturing
Gradient Gel Electrophoresis and Terminal
Restriction Fragment Length Polymorphism
CINDY H . NAKATSU AND TERENCE L. MARSH

41.1. INTRODUCTION ......................................... 909


4 1.1.1. Microbial Communities ................................ 909
.2. Microbial Community Analysis Methods .................... 910
41.2. STRATEGIES AND TECHNIQUES SHARED BY DGGE AND T-RFLP 910
41.2.1. General Design Features of an Experiment ................... 910
.2. Community Nucleic Acid Extraction ....................... 911
.3. PCR and DNA Quantification ........................... 911
.4. Data Analysis ....................................... 911
41.3. DGGE .................................................. 912
41.3.1. DGGE PCR ........................................ 912
.2. Denaturing Gradient Gel Preparation ...................... 913
.3. Running the Gel ..................................... 913
.4. Detailed DGGE Protocol ............................... 914
41.3.4.1. PCR Amplification for DGGE .................... 914
.2. Preparing Gels Using the BiopRad DCode Mutation
Detection System .............................. 914
.3. Running the Gel .............................. 914
.4. DGGE Band Sequence Determination ............... 915
41.4. T-RFLP ................................................. 915
41.4.1. The Target ......................................... 915
.2. Primer Selection ..................................... 916
.3. Primer Specificity .................................... 916
41.4.3.1. In Vitro Specificity ............................ 916
.2 .In Silico Specificity ............................ 916
41.4.4. PCR .............................................. 916
41.4.4.1. The Reaction ................................ 916
.2. Specificity of the PCR .......................... 917
.3. PCR Components ............................. 917
.4. Purification of PCR Products ..................... 917
.5. Quantitation of PCR Products .................... 917
41.4.5. Restriction Digestion .................................. 917
41.4.5.1. Selection of Enzymes ........................... 917
.2. The Digestion Reaction ......................... 919
.3. Incomplete Digestions. Nonspecific Cuts. and
Conformational Twists .......................... 919
4 1.4.6. Loading the Gels or Capillaries-Size Selection of Restriction
Fragments .......................................... 919
41.4.6.1. Electrophoresis Conditions ....................... 920
.2. Size Markers and Internal Controls ................. 920
41.4.7. Viewing and Collecting the Data .......................... 920
.8. Detailed Protocol for T-RFLP ............................ 920
41.5. SUMMARY .............................................. 922
.6. REFERENCES ............................................ 922

41 .1. INTRODUCTION three domains of life (11. 18.42.47) . In order to better un-
derstand these communities. microbial ecologists seek a de-
..
41 1 1. Microbial Communities tailed descrbtion of the communitv that would include (i) ..
Microbial communities can exhibit an enormous range of a phylogenetic census of all populations as a function of
complexity. from those with a mere handful of populations time. (ii) a description of all food webs and the participat-
(38) to those with thousands of species derived from all ing populations. (iii) the identification of all sexual cohorts.

909
910 COMMUNITY AND CENOMIC ANALYSIS

(iv) the nature of all inter- and intraspecies interactions


(quorum sensing and chemical warfareldefense), and (v)
the life cycles that influence population dynamics (e.g.,
sporulation and encystment). Such a complete description
of a community is indeed a challenging task. As a first step,
many of us labor simply to identify the collection of species Extraction of community DNA
present. This in itself is difficult inasmuch as the majority of
the microbes present in most communities have not yet
been cultivated (1, 46). This so-called plating paradox has
1
led to the development of culture-independent approaches PCR amplification
for describing diversity within a community. Critical to this
approach is the use of phylogenetic markers (16s-23s
rRNA, rpoC, and gyrases) that are present in most, if not
DGGE A T-RFLP

all, populations. Within these markers, there are both con- Gradient gel Purify
served and variable regions of sequence. The former permit electrophoresis PCR product
PCR amplification directly from community DNA that has
been extracted from environmental samples, while the lat-
ter provide a view of diversity. Thus, an early strategy of
community analysis included the comparative sequence
/ Stain and

-
Digest with
analysis of clone libraries that established the single-gene document gel restriction endonuclease
phylogenetic diversity of the community (21, 35). While
this approach can provide a richly detailed view of diversity,
such detail comes at an expense that renders ecological
questions addressing temporal or spatial changes in ecosys-
f 1
Fingerprint
1
Denaturing gel or
tems much too dear. In recent years, the solution to this analysis capillary electrophoresis
dilemma has been to sacrifice detail for high-throughput ca-
pabilities. For community analysis, genetic fingerprinting
techniques allow the comparative profiling of many envi-
ronmental samples and thus facilitate the spatial and tem-
poral analysis of microbial communities in ecosystems. Two
of these approaches, denaturing gradient gel electrophoresis
(DGGE) and terminal restriction fragment length polymor-
phism (T-RFLP), are described herein.
G 1
Re-amplify band
and sequence
1
Database o f terminal
fragment sizes

FIGURE 1 Flow diagram of the steps for microbial commu-


nity analysis using either DGGE or T-RFLP.

41 .I .2. Microbial Community Analysis Methods


Microbial community analysis methods fall into two broad
categories, those that interrogate the community without printing technology with DGGE and T-RFLP, the investi-
PCR amplification and those that are dependent on PCR. gator plays a game of chance regarding the possibility that
Examples of PCR-independent methods include fluores- the critical population in a changing community will be de-
cence in situ hybridization (see chapter 39), reverse genome tected with the experimental technique. Targeting a com-
probing, DNA:DNA reassociation kinetics (chapter 26), munity with multiple primer pairs of different specificities is
nucleic acid hybridization (chapter 30), phospholipid fatty one approach to increasing the number of detectable popu-
acid analysis (chapter 15), and community level meta- lations and hence the odds of identifying a relevant com-
genome analysis (chapter 38). PCR-dependent approaches munity shift.
include DGGE, T-RFLP, single-strand conformational poly- The general protocols for DGGE and T-RFLP are pre-
morphism, and quantitative PCR. One should note that sented in Fig. 1. Note that the first two steps, extraction of
PCR-dependent analytical techniques are subject to all of nucleic acid and PCR amplification, are common to both
the potential pitfalls of PCR, including inhibition from protocols. These steps, along with data analysis approaches
compounds that copurify with nucleic acids, the formation common to both, are discussed in section 41.2 below.
of chimeras, preferential priming of select targets, nonspe- Details specific to DGGE and T-RFLP follow in sections
cific priming of unexpected targets (49), and the produc- 41.3 and 41.4, respectively. The intent is to provide the in-
tion of single-stranded products (10) and formation of het- vestigator with sufficient information such that he/she can
erologous duplexes. The investigator is forewarned that avoid the potential pitfalls, regardless of the habitat being
many of these pitfalls are likely to arise in any experiment, investigated.
underscoring the need for controls.
Analytical approaches like DGGE and T-RFLP have
been referred to as genetic fingerprinting. By targeting a ro- 41.2. STRATEGIES A N D TECHNIQUES
bust phylogenetic marker such as 16s rRNA, the investiga- SHARED BY DCCE A N D T-RFLP
tor hopes that all populations in a community will be re-
vealed in the fingerprint. In reality, complex microbial 41.2.1. General Design Features of an Experiment
communities with population richness exceeding several As mentioned above, the power of DGGE and T-RFLP lies
hundred species are beyond single-primer pair PCR analy- in their high-throughput features. This high throughput
sis. The best gels and capillaries resolve 20 to 30 operational makes robust comparative analysis possible at the commu-
taxonomic units in DGGE and 75 to 125 in T-RFLP. nity level. A n essential prerequisite for all comparative
Clearly, this is a relatively small fraction of a complex com- analyses is the standardization of protocols. For both DGGE
munity. Thus, at the level of current community finger- and T-RFLP, this means that all steps of the protocol, in-
41. Analysis of Microbial Communities 911

cluding DNA extraction, PCR amplification, primer design most obvious data from these techniques is a simple count
and construction, electrophoresis systems, size standards, of OTUs (or phylotypes), which is an estimate of species
and data analysis approaches, must be identical for each richness. The evenness of detectable sequence phylotypes is
sample or community in the experiment. Because of the obtained from the measurement of band intensities. In
sensitivity and highly technical nature of the protocols, DGGE this requires a densitometric scan of the gel image,
both experimental and technical replicates are required. while in T-RFLP the amplitude of each fragment signal is
The investigator should assure himselfherself that all steps automatically recorded. While this can be a highly quanti-
in the protocol are reproducible. tative measurement of species evenness within the PCR
product pool, because the techniques are PCR dependent
41.2.2. Community Nucleic Acid Extraction and hence one uncontrolled step removed from the original
The extraction of community DNA from environmental community structure, the investigator cannot extrapolate
samples has been intensively investigated, and many tech- back to the original community and assert that peak ampli-
nical variations exist in the literature (see chapter 38). In tudes reflect population sizes. For this reason, the applica-
general, nucleic acids can be extracted directly from the tion of biodiversity indices (13) to the species richness and
substratum (e.g., soil and sediment) or from cells that have evenness estimates from DGGE and T-RFLP are not in-
been stripped off of the substratum. The latter approach de- formative regarding community structure.
creases contamination with substratum-bound components More statistically robust comparisons of communities
but may also be biased towards cells that are separated more can be made by treating the DGGE and T-RFLP profiles as
easily from substratum. Most researchers isolate DNA be- collections of characters that can be aligned using elec-
cause it is more stable than RNA; however, protocols have trophoretic mobility values associated with each band. The
been developed for the isolation of RNA from various en- resulting multiple alignments can be used to produce a ma-
vironments (see chapter 40). Commercial kits that provide trix for statistical comparisons. For example, using common
a rapid and standardized approach are also available (see, for similarity indices, such as that of Dice (5) [C, = 2j/(a + b),
example, MoBio and Q-BIOgene). A n important factor where a is the number of bands in sample A, b is the num-
when extracting nucleic acids from environmental samples ber of bands in sample B, and j is the number of bands com-
is to ensure that the approach achieves equivalent lysis of mon to A and B], a large number of communities can easily
all cell types so that the analysis is representative of the be compared. These data can be represented by cluster
community. To this end, robust physical techniques such as analysis, e.g., unweighted-pair group method with arith-
bead-beating and freeze-thaw have been coupled to harsh metic means (UPGMA) (44), in which a dendrogram is
chemical treatment to provide efficient, reasonably unbi- generated illustrating the relationship between communi-
ased cell lysis. The reader is encouraged to read reviews in ties (32). A n example of such an analysis is presented in
this collection and elsewhere to identify a lysis protocol Color Plate 17. Color Plate 17A shows T-RFLP data taken
suitable for hisher specific needs. into a spreadsheet program and aligned based on terminal
fragment size. The complete data set includes four replicates
41.2.3. PCR and DNA Quantification of three different soils (labeled H, N, and T). Each profile
Although it is emphasized in other chapters, it is important consists of fragment lengths for each detectable phylotype
to note here that PCR conditions must be optimized each (only a fraction of each profile is shown). The next panel
time DNA extracts from a new community are being exam- shows the same data set after being converted to binary
ined. DNA extraction methods have advanced consider- form (1's or O's, representing presence or absence). The sim-
ably, but some chemical contamination may still occur and ilarity of these profiles can be determined using similarity
interfere with PCR. Also, DNA concentrations must be de- indices such as that of Dice (described above) or the T-
termined before and after PCR. Nucleic acid concentra- RFLP analysis function available at the Ribosomal Database
tions in a sample can be estimated using any of the common Project (RDP) (4). The results from the latter are shown in
approaches: spectroscopy, fluorimetry, and agarose gel elec- Color Plate 17C. The data are also amenable to cluster
trophoresis (see chapters 26 and 30). Use of suboptimal analysis with, for example, UPGMA. Color Plate 17D pre-
PCR conditions and DNA concentrations can lead to PCR sents a dendrogram of the UPGMA analysis of the data in
artifacts that can contribute to the over- or underestimation Color Plate 17B from the three soil data sets. In Color
of community diversity. Equivalent concentrations of PCR Plates 17C and D, the three soils are easily distinguished
products need to be used for DGGE or T-RFLP analyses if and the replicate samples form a statistical cluster.
profile comparisons are going to be made. Alternatively, multidimensional scaling ( 15) can be used
to obtain a graphical representation of these (dis)similari-
41.2.4. Data Analysis ties. As data sets increase in size, sources of variation in-
In general, microbial ecologists are interested in the struc- creases and multivariate analysis methods, such as principal-
ture and function of specific communities, as in the gas- component analysis (37), become more appropriate. This
trointestinal tract, or large-scale trends in the structure and method also provides a visual representation of the rela-
function of communities across spatial or temporal zones. tionship between the profiles. Principal-component analy-
The distribution of phylogenetic markers across communi- sis calculates and ranks the contribution of each variable in
ties, as determined by DGGE and T-RFLP, provides a rapid a profile, and the approach can be used to identify the main
assessment of microbial diversity. Phylogenetic markers de- sources of variation observed between profiles (52). For ex-
rived from different populations are separated by sequence- ample, in DGGE profiles, the source (band) contributing
determined helix stability (DGGE) or terminal restriction the greatest variability can be statistically determined, and
fragment length (T-RFLP). Each unique band or terminal then the bands can be extracted from the gel and its nu-
fragment represents an operational taxonomic unit (OTU), cleotide sequence can be determined to identify the popu-
and the distribution of these OTUs constitutes the profile. lation. A limitation of DGGE is that a complex community
By comparing profiles derived from communities, one can (e.g., soil) may be comprised of numerous populations
quantify the degree of relatedness across communities. The (>100) in relatively equivalent proportions, thus resulting
912 m COMMUNITY A N D GENOMIC ANALYSIS

in a smear of bands which makes it difficult to identify in- G + C content will migrate through the gel to higher con-
dividual populations (34). It is still possible to qualitatively centrations of denaturant. Separation of a mixture of PCR
state that two communities are different if the smear of products of ribosomal DNA (rDNA) amplified from a com-
bands looks different; however, the converse may not be munity produces a profile, which represents the different
true. Even better resolution of very complex data sets may populations present in that community.
be obtained using methods such as self-organizing map neu- Typically, polyacrylamide gels are used for DGGE, but
ral networks (6). Many would consider the current analyti- denaturing high-performance liquid chromatography
cal methods more exploratory in nature, just the beginning (dHPLC) is being examined as an alternative (50,53). This
to quantitatively address the complex relationships between approach may overcome some of the resolution and repro-
microbial communities. ducibility challenges faced when using polyacrylamide gra-
There are commercial software programs available dient gels. Specialized dHPLC instruments (WAVE System;
specifically for community fingerprint analysis, for example, Transgenomics Inc., Omaha, NE) have been tested for mi-
Bionumerics (Applied Maths, Belgian) and Fingerprinting crobial analysis (7, 12, 14).
I1 Informatix (Bio-Rad). Many other statistical packages are
available that can be adapted to DGGE and T-RFLP data 41.3.1. DCCE PCR
analysis, and some, like Phylip (http://evolution.genetics Typically, PCR products of specific gene sequences are used
.washington.edu/phylip.html), are available at no cost. for DGGE and TGGE analysis. For microbial community
analysis, the most commonly used target has been the 16s
rRNA gene, but it has also been used to determine genetic
41.3. DCCE polymorphisms in other genes, e.g., (Ni/Fe) hydrogenase
The premise of DGGE is that DNA strands of the same genes (5 1). Some commonly used primers for small-subunit
length will migrate differentially when electrophoresed (SSU) rDNA DGGE analysis are listed in Table 1. The
through a linear gradient of denaturant if their sequence primers chosen for PCR can be used to determine the level
composition differs. Denaturation occurs when hydrogen of discrimination between groups in the community. For ex-
bonds holding together the double strands of DNA are dis- ample, primers can be used to amplify all Bacteria, or only
rupted by exposure to denaturants. The strands partially the p subdivision ammonia oxidizer (17). As the number of
separate when the lowest melting temperature within the se- submissions to genetic databases increases, all primers
quence is reached, at which point the separation of the strands should be reassessed to ensure that the desired specificity is
retards migration in the gel. To ensure that the strands do conserved. Primers for DGGE and TGGE that yield a rela-
not entirely separate, a high-GC sequence (called the GC tively short PCR product length are preferred in order to
clamp) is added to one primer (43). There are two types of minimize the number of melting domains and to reduce
denaturants used: chemicals such as urea and formamide in chances of producing PCR artifacts. Some researchers also
DGGE and temperature in thermal gradient gel elec- use touchdown PCR to improve specificity (33). In this ap-
trophoresis (TGGE). The concentration of denaturant de- proach the first few cycles are performed at temperatures
termines the distance the DNA molecules migrate through above the standard annealing temperature and then the
the gel. Higher denaturant concentrations are needed to temperature is lowered with subsequent cycles until the an-
disrupt the three hydrogen bonds formed by guanine and nealing temperature is reached. Nested PCR has been used
cytosine base pairing. Therefore, sequences with higher to amplify target DNA that constitutes a very small fraction

TABLE 1 Examples of PCR primers for DGGE and T-RFLP


Primer Position" Target rRNA Synthesized primer sequenceb Reference
BA338F 338-35 8 Bacteria, 16s 5" ACT CCT ACG GGA GGC AGC AG 3' 20
UN5 18R 534-5 18 Universal, 16s 5' ATT ACC GCG GCT GCT GG 3' 33
ARC344 344-363 Archaea, 16s 5" ACG GGG IGC AGC AGG CGC GA 3' 40
ARC915 915-934 Archaea, 16s 5' GTG CTC CCC CGC CAA TTC CT-3' 45
F1427 1427-1452 Eukarya, 18s 5'd TC TGT GAT GCC CTT AGA TGT TCT GGG 3' 48
R1616 1616-1595 Eukarya, 18s 5' GCG GTG TGT ACA AAG GGC AGG G 3' 48
1511R 1526-15 11 Universal 5' YGC AGG TTC ACC TAC 30
1492R 1506-1492 Universal 5' ACC TTG TTA CGA CTT 1
1392R 1406-1392 Universal 5' ACG GGC GGT GTG TRC 1
21F 2-2 1 Archaea, 16s 5' TTC CGG TTG ATC CYG CCG GA 1
25F 9-25 Eukarya, 18s 5' CTG GTT GAT CCT GCC AG 30
27F 8-27 Bacteria, 16s 5' AGA GTT TGA TCM TGG CTC AG 1
63F 42-63 Bacteria, 16s 5' CAG GCC TAA YAC ATG CAA GTC 26
1387R 1404-1 387 Bacteria, 16s 5',GGG CGG WGT GTA CAA GGC 26
927R 942-927 Bacteria, 16s 5' ACC GCT TGT GCG GGC CC 1
'For prokaryotes relative to 165 rRNA gene sequence of Escherichia coli and for eukaryotes 18s rRNA gene sequence of Saccharomyces cereuisiae.
*A GC clamp must be added to the 5' end of one of the primer pairs used for PCR.
%C clamp used 5' CGC CCG CCG CGC GCG GCG GGC GGG GCG GGG GCA CGG GGG G 3'.
'GC clamp used 5' CGC CCG CCG CGC CCC GCG CCC GGC CCG CCG CCC CCG CCC C 3'.
41. Analysis of Microbial Communities 91 3

of the community (34). In this method, PCR is performed


twice, first with a set of primers that may also amplify DNA
from populations outside of the target group. Then the sec-
ond PCR is performed using primers within the first ampli-
con and specific for the target group. As mentioned earlier,
one of the primers used for PCR amplification for DGGE
and TGGE analysis must include a G C clamp (43) to stabi-
lize strand melting.
41.3.2. Denaturing Gradient Gel Preparation
A vertical polyacrylamide gel electrophoresis apparatus and
gradient former are needed to run the chemical type of de-
naturing gradient. Bio-Rad Laboratories Inc. (DCode muta-
tion detection system) and C.B.S. Scientific Company Inc.
(Del Mar, CA) market equipment for DGGE analysis.
Alternatively, a standard vertical protein gel apparatus can
be adapted for DGGE. The system is adapted by using a I gradient range to use for anabsis
constant-temperature bath to maintain the buffer tempera- 0% 1I
ture at 60C and a magnetic stirrer to keep the buffer well b
mixed during electrophoresis. A specialized cam-operated Increasing Denaturants
gradient former is supplied with the Bio-Rad system; other-
wise a standard two-cylinder gradient former can be used. In FIGURE 2 Diagram of a melting profile of a single PCR prod-
the latter case, as the different denaturant gel solutions are uct separated using perpendicular DGGE. The approximate
being mixed, they can be delivered into the gel plates using denaturing gradient to use would be 25 to 50% denaturant.
gravity flow or, for better reproducibility, using a peristaltic
pump-
The appropriate denaturing gradient for the analysis of a
community can be determined using either perpendicular or mixture of PCR products is loaded into the gel at 20-min
parallel gels. These gel types are differentiated by orienta- intervals. At the end of the experiment, the minimum time
tion of the gradient with respect to the direction of elec- for maximum separation of the PCR products can be ob-
trophoresis; the electrical current is applied either parallel served on the gel (Fig. 3).
or perpendicular to the denaturing gradient. In parallel gels Standard markers are used to monitor gel-to-gel variabil-
the higher denaturant concentration is towards the bottom ity. The marker should be composed of DGGE PCR prod-
of the gel, whereas in perpendicular gels it is towards the ucts, and under no circumstances should size markers be
right side. Perpendicular gels are poured with the plates on used. Enough organisms with different sequence composi-
their side and then rotated to the correct orientation for tions should be chosen for the markers to span the denatur-
electrophoresis. Using parallel gels to determine the appro- ing gradients typically used in ones laboratory In addition,
priate gradient, communities are first examined using a the most commonly studied organisms in your laboratory
broad gradient range (e.g., 20 to 80% denaturant). The de- should be included to readily detect potential contamina-
naturant concentrations are then adjusted to include all the tion in your PCR. To minimize intensity differences between
bands while maximizing their separation. The advantage of
this approach is that optimal gradients for many different
communities can be assessed simultaneously, whereas the
denaturing gradient for only one community at a time can Sample Loading Time (min.)
be determined using perpendicular gels. In this method, a 40 100 160 220 280 340 400
well spanning the width of the gel is loaded with the com- I I I I I I I

munity PCR product. After electrophoresis, the gel is ex-


amined to determine the approximate denaturant concen- c
3
(

trations at which the DNA strands melted (Fig. 2). These


denaturant concentrations are then used for subsequent 4
OI
analysis using parallel gels. The advantage of this method is vr.
3
that the melting behavior of the DNA can be observed, ro
which is particularly useful if previously untested primers P
ID
are being used. Regardless of the method chosen to deter- 3
mine the appropriate gradient, community analysis is per- no segamtbn
Iu,
C
formed using parallel gels. In some cases, it may be neces-
sary to characterize communities using two or more
J
different gradients to resolve bands with similar melting 2
characteristics.
41.3.3. Running the Gel
0%
The other important variable that must be determined is FIGURE 3 Diagram of the separation of a two-member com-
the time required for electrophoresis to maximize resolution munity inoculated over 6.6 h at 20-min intervals using parallel
between bands. Once the denaturant gradient to be used is DGGE. This example uses a 25 to 60% denaturing gradient
established, the optimal electrophoresis time can be deter- and electrophoresis at 200 V and 60C. The optimal separation
mined using a time interval experiment. For example, a time would be 4.5 to 5 h.
914 COMMUNITY AND GENOMIC ANALYSIS

strains, PCR should be performed independently on each gether using the 1.5-mm-thick spacer. The spacers can be
strain chosen and then the products should be mixed in lightly coated with silicone grease to reduce smiling of
equal concentrations. Standard markers must be run on all bands in the outer lanes of the gel.
gels to assess gel variations and to make comparisons be- 3. Before locking the plates into the pouring stand, put a
tween gels. drop of melted 1.5% agarose at each end of the gasket under
the position where the plate spacers will fall to prevent leak-
41.3.4. Detailed DGGE Protocol ing. Clamp in the plates, and then seal around the bottom
edges of the plates if leaking has been a problem. If leaking
41.3.4.1. PCR Amplification for DGGE becomes excessive, it may be necessary to replace the gasket.
A basic protocol for amplification of the V3 region (bases 4. For each gel, make 15 ml of desired low-denaturing
338 to 534) of the 1 6 s rRNA gene from community DNA and 15 ml of high-denaturing solutions. For example, make
extract is as follows. a low denaturant of 25% (mix 11.25 ml of 8% bis-polyacry-
1. Make a PCR master mix for the total number of reac- lamide gel solution containing 0% denaturant and 3.75 ml
tions (final volume, 50 pl per reaction) to be performed. of gel solution containing 100% denaturant) and a high de-
When calculating the required volume, include additional naturant of 60% (mix 6.0 ml of 0% denaturant gel solution
reactions for negative (no DNA) control, positive control, and 9.0 ml of 100% denaturant gel solution). If gels with
and liquid loss during handling. The final concentrations in the same denaturant concentrations are to be repeatedly
the PCR mixture are: used, make stock solutions of 8% bis-polyacrylamide gel so-
lution with those denaturant concentrations. The volumes
1X PCR buffer for different gel systems may vary and can be determined by
2.0 p M MgClz measuring the volume of water needed to fill the gel plates.
0.8 mM deoxynucleoside triphosphates (dNTPs) 5. The cam-operated gradient former uses disposable
a 0.5 p M concentration of each primer (BA338F and 30-ml syringes to deliver the denaturant gel solutions. The
UN5 18R) levels of the syringes on the cam must be adjusted for the vol-
ume of the gel. Before filling syringes, make sure they move
0.1% bovine serum albumin (BSA) and 5% dimethyl smoothly when the cam is turned and that the syringes will
sulfoxide completely extrude the solutions. A Y tubing system mixes
1 U of Tq polymerase the two denaturant solutions and delivers it between the gel
2. Dispense 49 pl (less if more than 1 pl of DNA tem- plates using a syringe needle.
plate is needed) into each PCR tube. 6. Once everything is properly assembled, add 120 pl of
3. Add 1 p1of DNA (5 to 50 ng) template. The optimal APS (ammonium persulfate; 10% fresh or frozen stock) and 5
template DNA concentrations to maximize PCR products p1 of N,N, N, N-tetramethylenediamine(TEMED) to the
and minimize PCR artifacts may vary with the source and low- and high-denaturant solutions and swirl gently to mix.
should be determined empirically. 7. Take up denaturing and acrylamide solutions into
4. Place tubes into a thermal cycler and run the follow- the syringes. For parallel gels, the syringe with the low de-
ing PCR program. naturant is placed in front and the high denaturant is
placed on the backside of the gradient maker. Make sure all
Denaturation at 94C for 5 min the air is removed from the syringes before placing them
25 cycles of: into the gradient maker. Place the delivery syringe needle
w Denaturation at 94C for 30 s
between the two plates.
8. Slowly turn the wheel until the syringes are empty
w Annealing at 55C for 30 s (this temperature will and the gel is completely poured. For reproducibility, each
vary depending on the primer pair chosen) operator must pour gels at the same rate each time.
w Extension at 72C for 90 s 9. Let the gel polymerize for about 10 to 15 min and
Final extension at 72C for 10 min then place comb at a slight angle between the plates.
10. To form wells, add 40 yl of APS and 2 p1of TEMED
Hold at 4C until retrieved to 5 ml of 8% bis-polyacrylamide containing 0% denaturant
5. Check PCR products for size and quantity on an and mix by swirling. Using a pipette, slowly add solution to
agarose gel. Load 1 to 5 pl onto a 1 to 1.5% agarose gel the top of the gel and then gently push in comb, avoiding
(agarose percentage is dependent on PCR product size; making bubbles. Let gel polymerize for at least 2 h to
buffer is 1X TAE where a liter stock of 5 0 X TAE contains overnight.
242 g of Tris base, 57.1 ml of glacial acetic acid, and 100 ml
of 0.5 M EDTA of pH 8.0 (41). 41.3.4.3. Running the Gel
41.3.4.2. Preparing Gels Using the Bio-Rad DCode 1. To run the gel, make approximately 7 liters of 0.5X
Mutation Detection System TAE (must be the same concentration as the gel) and fill
the buffer chamber. Reserve about 0.5liter for use later.
A basic protocol using the DCode mutation detection sys-
2. Preheat buffer until the temperature is about 50C,
tem (Bio-Rad Laboratories Inc.) is given below. See the
interrupt heating, and attach the gel plates to the core ap-
Bio-Rad manual for more details.
paratus. Two sets of plates must be attached. If only one gel
1. Prepare two stocks of 8% bis-polyacrylamide (from a is required, then two plates with no spacers or gel should be
40% stock made with 37.5: 1 acrylamide/bisacrylamide) in used as the other set.
0.5X TAE gel solution with one containing 0% and the 3. Place plates into the buffer chamber, fill top reservoir
other 100% (7 M urea, 40% formamide) denaturants. with remaining buffer, and continue heating until 65C is
2. Wash glass plates, spacers, and combs thoroughly reached (about 2 h). The buffer will cool to the desired
with warm water and detergent. Assemble the plates to- 60C while loading.
41. Analysis of Microbial Communities 915

4. Flush wells with a syringe to expel dissolved acryl- 41.3.4.4. DGGE Band Sequence Determination
amide. Load 5 to 50 p l of PCR product with loading dye One of the advantages of this method is that bands from the
into the wells. The volume loaded will depend on the num- gel can be sampled and the nucleotide sequence determined
ber of expected bands; as the number of bands increases, the (36). However, only a fraction of the rRNA gene is deter-
volume loaded must increase. mined from DGGE bands; therefore, identification of the
5. Reset the temperature to 60"C, run at 20 V for 10 population represented by the band is tentative.
min and then at 200 V for 10 min, and then turn on the re-
circulation pump (this minimizes sample washout from 1. Locate the band(s) of interest after staining and then
wells). Continue running at 200 V for the predetermined collect a sample by touching a sterile 1-ml pipette tip to the
optimal time (typical time is 5 h). It is possible to vary the center of the band.
voltage and electrophoresis time to accommodate your 2. Remove core and place into 20 p l of sterile PCR-
workday. However, comparisons should be done to ensure grade H 2 0 in a 1.5-ml microcentrifuge tube. Use a new
that bands remain sharp for easier analysis. pipette tip for each band.
6. When the run is complete, take apart the gel appa- 3. Incubate tube at 4C overnight to allow the PCR
ratus and remove the gel onto plastic wrap for easier ma- product to diffuse from the gel into the water.
nipulation. 4. Confirm that the correct band has been extracted by
7. Leave the gel on the plastic wrap and place into a no. amplifying 5 to 10 p l of the water extract by PCR and com-
5 plastic container that just fits the gel. Glass trays should pare the product by DGGE with the original sample.
not be used because glass binds the SYBR Green dye. 5. If a mixture of bands is found, the band picking
8. Stain the gel for 10 to 15 min with SYBR Green I process can be repeated until a single band is achieved.
(FMC) diluted 5,000 times (4 pl in 20 ml of 0.5 X TAE for Alternatively, the mixture can be PCR amplified using the
a full gel) with slight shaking and minimal light exposure. same primers but without the GC clamp. The PCR products
9. Pour off the stain and rinse gel briefly with 0.5X can then be cloned and the band of interest identified by
TAE to remove excess dye. DGGE after amplification of each clone using the primers
10. Visualize on a transilluminator and photograph. with GC clamp.
Alternatively, bands can be visualized after silver staining or 6. Either the PCR product or clone can then be used for
ethidium bromide staining. Silver staining is as sensitive as nucleotide sequence determination. The primer used for
SYBR Green I but is more time-consuming, whereas ethi- amplification can also be used for sequencing.
dium bromide takes a similar amount of time but is less sen- 7. Since it is possible that a band represents more than
sitive. Figure 4 illustrates different community profiles ob- one population, multiple clones or PCR products must be
served using DGGE. sequenced.

41.4. T-RFLP
2 3 4 5 T-RFLP has been used as an effective tool in the dissection
of microbial communities ( 3 , 16, 19, 23-25, 27, 28, 31).
The overall protocol is depicted in Fig. 1 and includes (i)
the isolation of community DNA (or RNA), (ii) the PCR
amplification of the target, (iii) restriction digestion, and
(iv) the size separation of the restriction fragments. The es-
sential technical features of the procedure include the use of
fluorescently tagged primers in the amplification step and
the size separation of fragments on a high-resolution auto-
mated sequencing system. This combination of technical
protocols yields a system with high throughput capabilities
and sensitivity, making it ideal for microbial ecological
studies where extensive samplings and replicates are neces-
sary.
41.4.1. The Target
Similar to the case with DGGE, the 16s rDNA has been
the most frequently used marker for T-RFLP analysis. The
tremendous phylogenetic utility of this marker and the ro-
bust sequence database have contributed to this. Moreover,
when dissecting or comparing communities, one of the first
attributes of interest is the biodiversity, or what species are
present? T-RFLP and DGGE of the SSU rRNA genes pro-
vide a reasonable first draft estimate of biodiversity, al-
though as pointed out previously, neither DGGE nor T-
FIGURE 4 DGGE profiles of PCR products of 16s rDNA RFLP of SSU rRNA genes can describe the enormous
communities from bioreactors (lanes l ) , lakes (lanes 2), corn diversity of the microbial world.
rhizosphere (lanes 3), bulk agricultural soils (lanes 4), and soils Other markers have been targeted using the T-RFLP ap-
contaminated with heavy metals and organic solvents (lanes proach. These include mercury resistance genes ( 3 ) , nitrate
5). With the exception of the bulk agricultural soils, intense reductase (39), nijH genes, and methyl-coenzyme M reduc-
bands are observed in each profile. Shown is an inverted image tase, alpha-subunit (rnuA/mrtA), in methanogenic archaea.
of DGGE gels stained with SYBR Green I. In the case of translated genes, codon degeneracy and the
916 COMMUNITY AND GENOMIC ANALYSIS

wobble base can make primer design considerably more plification across a temperature range for these isolates will
challenging. However, judicious degeneracy within the establish the level of stringency required for the advertised
primer is tolerated in T-RFLP, where the final separation is specificity.
based on fragment length and not thermal stability, as with
DGGE or TGGE. Care and rigor in primer design must be 41.4.3.2. In Silico Specificity
exercised nonetheless, particularly in the case of markers In addition to in vitro tests of specificity, it is also worth-
with known paralogs. Paralogs that are amplified along with while to check the specificity in silico, realizing that the
the intended target will confound estimates of diversity. In specificity of primers is related to the robustness of the data-
this regard, the robustness of ones results resides in the base at the time of design. With sequences being added to
specificity of ones primers. databases at an increasing rate each year, phylogenetic di-
versity is a changing entity, and a primer perceived as highly
41.4.2. Primer Selection specific 5 years ago may be only moderately specific with
In the following discussion, rDNA is used as an example for todays database. The most familiar method is undoubtedly
primer selection. In the case of 16s rDNA, there are cur- a BLAST search against the nonredundant sequence data-
rently over 78,000 rRNA sequences in the RDP (4). base (BLAST at http://www.ncbi.nlm.nih.gov/BLAST/).
Considerable effort has been invested in identifying univer- The RDP (4) also provides a Probe Match function that
sal, domain-specific, group-specific, and even species- searches the updated database for matches. In addition, it is
specific probes and primers (1,22). There are several highly worth mentioning ARB, a freely distributed software writ-
conserved regions in 1 6 s rRNA that have served as targets ten and maintained by the Technical Institute of Munich
for probes. Table 1 presents the more commonly employed (http://www.arb-home.de/). This software provides not only
primers for the bacteria, archaea, and eukaryotes. With gel a systematic probe/primer analysis function that reports on
and capillary electrophoresis, the limits of resolution for the number and phylogenetic distribution of hits (matching
fragment size are approximately 700 and 1,000 bases, re- sequences) in an rRNA database but also alignment and
spectively. Optimally, the primers selected for T-RFLP phylogenetic functions and a variety of features that make
should amplify nearly the entire gene so that all possible re- maintaining a curated database within the laboratory possi-
striction sites within the target are included. However, with ble. ARB runs on Unixbinux and is a powerful and sophis-
the stated resolution of the electrophoresis systems, not all ticated sequence analysis environment with a commensu-
fragments will be detectable if only one labeled primer is rate learning curve.
employed. This can lead to losing entire phylogenetic
groups from the analysis. Thus, complete coverage of the 41.4.4. PCR
molecule through the use of both forward and reverse la- PCR is an exquisitely sensitive technology, and for this rea-
beled primers is recommended. It should be noted that the son, great care must be taken to avoid contamination. PCR
collection of fragments derived from both forward and re- amplification from community DNA is frequently more dif-
verse labeled primers are a partially overlapping set; hence, ficult than amplification from isolates. This is certainly due,
diversity cannot be estimated by a simple addition of data in part, to the copurifying contaminants that can accom-
sets. The type of attached fluorescent label is usually dic- pany DNA through the extraction protocols (as discussed
tated by the detection technology employed in the auto- above). PCR adjuvants such as BSA and T4 gene 32 pro-
mated sequencers. All automated sequencers with multiple tein can suppress the actions of inhibitors. Thus, a combi-
fluor detection capabilities allow for the option of running nation of effective DNA isolation procedures and PCR ad-
three differently labeled PCR products as well as labeled size ditives will give the most consistent results.
markers in the same lane or capillary. This increases the
cost-effectiveness of the procedure. For the ABI systems, 41.4.4.1. The Reaction
carboxyhexachlorofluorescein (HEX), 6-carboxyfluorescein After isolating community DNA, pilot PCRs are run to es-
(FAM), 6-carboxy-2,4,7,7-tetrachlorofluorescein (TET), 6- tablish that the DNA is of sufficient purity to serve as a pro-
carboxytetramethylrhodamine (TAMRA), and carboxyrho- ductive template. Pilot reactions as small as 12.5 pl can be
damine (ROX) have been routinely employed. Table 1 lists performed with thermal cyclers designed to handle 200- to
primers commonly used in T-RFLP of 16s rRNA genes. 250-pl tubes. Several template concentrations are used to
test a 10-fold concentration range (5 to 50 ng for a 25-pl re-
41.4.3. Primer Specificity action), and unlabeled primers are used to reduce costs. The
Primer design has been a particularly productive experi- resulting PCR products are visualized on an agarose gel (0.8
mental field. The use of a phylogenetic marker in the analy- to 1.0%) for product abundance (PCR efficiency) and prim-
sis means that the sequence differences that distinguish ing specificity. If needed, PCR efficiency can be improved
phylogenetic groups can be utilized in the construction of through a systematic optimization of component concen-
primers with phylogenetic specificity. Well-characterized trations. Focusing on the template and Mg2+ concentra-
primers and probes have been collected in several reviews tions will bring most amplifiable templates to an acceptable
( 1, 22) and databases (http://www.microbial-ecology.de/ level of productivity in PCRs. After optimizing the PCRs,
probebase/). fluor-derivatized primers are substituted in the reaction for
unlabeled primers and the reaction volume is increased to
41.4.3.1. In Vitro Specificity 100 p1. Depending on amplification efficiency, from one to
Once primers have been selected and synthesized, it is im- three 100-p1 reactions may be required to provide enough
portant to test for the expected specificity. A thermocycler labeled product. The detection system used in the sequenc-
that can form a temperature gradient across the block is in- ing apparatus dictates the fluors that can be used. In the
valuable in this step. PCR amplifications are set up with ge- ABI systems, the detectable dyes are 6-FAM, HEX, TET,
nomic DNA from representative isolates from inside and TAMRA, and ROX. Because these fluors are subject to
outside of the specifically targeted group. Testing PCR am- photobleaching, the primer stocks and the reactions should
41. Analysis of Microbial Communities 917

be protected from direct and prolonged exposure to light. If extension. A final extension at 72C for 5 min completes
two labeled primers are to be used for full coverage of the the amplification.
target as mentioned above, different fluors should be se-
lected for the forward and reverse primers so that both sig- 41.4.4.4. Purification of PCR Products
nals can be detected in the same lanelcapillary. PCR products should be purified away from residual Taq ac-
tivity and ionic conditions that may be suboptimal for re-
41.4.4.2. Specificity of the PCR striction digestion. Commercially available PCR cleanup
Specificity in PCR amplification is a critical issue for com- kits have proved effective for this step (e.g., Promega and
parative analysis. The fluors used in T-RFLP provide high Qiagen).
sensitivity in the gel and capillary systems; hence, any spu-
rious PCR products will be detected and possibly scored as 41.4.4.5. Quantitation of PCR Products
a restriction fragment. If PCR products of unanticipated As mentioned above, comparative analysis requires that all
sizes are detected in the analytical agarose gel, the PCR samples be equivalent in concentration. Thus, PCR prod-
must be optimized, usually through manipulation of the an- ucts are quantitated after purification by A260 or fluorome-
nealing temperature or concentrations of MgC12, primer, or try (see above). Because sequencing machines vary in sen-
template. If secondary products remain problematic after sitivity, determination of the optimal concentration loaded
optimization attempts, the PCR product of correct size can onto the system is derived empirically.
be gel purified. This is an unsavory option because of the
additional preparative step and the highly variable recovery 41.4.5. Restriction Digestion
of DNA from the gel. Digestion with restriction endonucleases is conducted ac-
cording to the manufacturer's recommendations. While this
41.4.4.3. PCR Components would seem to be a trivial aspect of the procedure, given the
Standard PCR conditions are presented in Table 2. Ob- extensive use made of restriction enzymes, a number of pit-
serving standard aseptic techniques throughout the setup of falls face the investigator at this step.
PCRs will prevent contaminating templates from con-
tributing to one's experiments. Many commercially avail- 41.4.5.1. Selection of Enzymes
able T q polymerases are suitable for T-RFLP. In most cases, The selection of an enzyme for T-RFLP analysis is not triv-
the vendor supplies lox buffer and a separate MgCl solu- ial. For many investigators, the objective is to reveal as
tion. In practice, the PCR is usually set up in a two-step many phylotypes in the community as possible. In this way,
process that serves to minimize chances of contamination. community diversity can be estimated and/or followed as a
A master mix is assembled in a clean UV-irradiated area. function of time or amendment. If 16s or 23s rDNA is the
The master mix contains, in order of addition, water, buffer, target, then a sufficiently large sequence database exists for
MgC12, dNTPs, primer, and Taq. The templates are then identifying enzymes appropriate to the task. In general, four-
added to the reaction tubes and a uniform volume of master base cutters have been used in order to maximize the likeli-
mix is pipetted into each reaction tube. Reproducibility will hood of a restriction target within the boundaries of the am-
be increased if the stock concentrations are set up such that plified product. However, some four-base target sequences
the volumes pipetted are not smaller than 2 to 3 ~ 1It. is im- are more conserved within the 16s rRNA sequence than
perative to include both a negative and positive PCR in others. Figure 5 displays the frequency distribution of 1 6 s
every set of PCR amplifications. rDNA terminal fragments generated from the in silico di-
Either a hot-start or touchdown PCR technique (8) is gestion of 20,070 16s rDNA sequences (1,200 bases mini-
recommended to diminish nonspecific priming events at mum in length) with two restriction enzymes, HaeIII and
the outset of PCR. A n initial soak for 3 to 5 min at 94C is RsaI. The digestions were carried out assuming that PCR
followed by 25 to 30 cycles of 1 min of denaturation at products were generated with a labeled 63F bacterial do-
94"C, 45 s of annealing at 45 to 60C (temperature deter- main-specific primer. Hence, all fragments are derived from
mined by primer melting temperature), and 45 s at 72C for the 5' terminus of bacterial rRNA genes. In the top panel,
showing the HaeIII digestion, there is a skewed distribution
with two highly conserved positions at approximately posi-
tions 32 and 171 (Escherichia colz numbering) and 503
unique terminal fragment sizes. In contrast, digestion with
TABLE 2 Standard PCR conditions RsaI revealed a broader distribution of terminal fragments,
Component Stock concn Final concn though still clustered at some level, with a greater number
of unique fragments (730) derived from the same database.
Buffer lox 1x Thus, the phylotype resolving power of RsaI was substan-
dNTPs 2.0 mM 200 p,M tially better than that of HaeIII. While HaeIII is clearly
suboptimal for detecting the greatest diversity, under cir-
M a 2 25 mM 1.5-2.5 mM
cumstances where a targeted phylotype has a HaeIII restric-
Forward primer -50-100 pmol/pl -5-10 pmo1/25-p,l tion site different from the highly conserved site, it may
reaction prove useful by separating most of the community from the
Reverse primer -50-100 pmol/pl -5-10 pmo1/25-pl target of interest. Using restriction enzymes in this way can
reaction only be achieved with reference to the database. Figure 6
BSA 1% 0.05% displays the number of unique fragments that are generated
Template 10-500 ng/p,l 5-50 ng/25-pl with 19 different restriction enzymes in an in silico restric-
reaction tion site analysis of 27,060 rRNA sequences. The data were
derived using PatScan (9) and the 63F primer with a single
H70 To volume
restriction site as the queried sequence motifs. Note that
918 COMMUNITY AND GENOMIC ANALYSIS

1800 -
1600
Frequency Distribution of Hae 111
target sites on 16s rRNA genes
1400

uh 1200 -'

51 1000
B
............. ~

A. . . . .- -
1 100 199 298 397 496 595 694 793 892 991 1090 1189 1288 1387 1486

Fragment Length
800

....... Frequence Distribution of Rsa I


I I
700 ..... .......
target sites on 16s rRNA genes
600

500
Ea 400
%
2 300

200

100

0
1 201 401 601 801 1001 1201 1401

Fragment Length

FIGURE 5 Terminal fragment size distribution. An rRNA database of 20,070 sequences was di-
gested in silico with HaeIII (upper panel) and RsaI (lower panel). The frequency (ordinate) of ter-
minal fragment sizes (abscissa)appearing in the database is presented.

two restriction enzymes result in the absence of unique frag restriction sites across the 16s gene. T h e table is sorted in
ments because there is a conserved restriction site within order of increasing number of unique fragments derived
the primer (CviRI and NlaIII). Based on this analysis, the from the listed enzymes. While NciI and Mae11 look attrac-
enzymes most suited for estimating phylotypes diversity are tive based on the number of unique fragments generated,
AvaII, DpnI, HhaI, NciI, RsaI, and TaqI. Table 3 summa- note that there are a significant number of sequences with
rizes the same digestions but also shows the distribution of target sites more than 1,000 bp from the 5' terminus.

I nnn

Enzyme

FIGURE 6 Number of unique fragments generated with commonly used restriction endonucle-
ases. A 20,070-sequence rRNA database was digested in silico with 19 restriction enzymes (ab-
scissa). The number of unique terminal fragment sizes is reported for each enzyme (ordinate).
41. Analysis of Microbial Communities H 919

TABLE 3 Number of 5 proximal restriction target sequences on 16s rDNA

Restriction Recognition No. of sequences Distribution of terminal fragments


enzyme sequence primed (63F) and digestedb 6 0 0bp 500-1,000 bp 1,000-1,500 bp
NlaIII CATG 18,929 0 0 0
CviRI TGCA 18,928 0 0 0
Sau96I GGNCC 18,923 18,828 87 8
AccI CGCG 18,929 18,864 65 0
AciI CCGC 18,929 18,886 43 0
AluI AGCT 18,928 18,463 411 54
HaeIII GGCC 18,917 18,249 483 185
MspI CCGG 18,914 18,205 699 10
TspEl AATT 18,926 10,989 7,935 2
BstNI CCWGG 18,913 6,149 12,752 12
MseI TTAA 18,928 9,515 9,387 26
Mae11 ACGT 18,923 16,588 796 1,539
RsaI GTAC 18,927 14,305 4,569 53
HhaI GCGC 18,913 14,854 2,934 1,125
Mae1 CTAG 18,888 13,903 4,332 653
DpnI GATC 18,795 16,358 1,219 1,218
TaqI TCGA 18,865 7,086 11,655 124
AvaII GGACC 15,365 10,695 4,297 373
NciI CCCGG 18,478 7,462 6,932 4,084
Based on an RDP library of 27,060 sequences, each of which is at least 1,200 bases long (REF). The database was screened with PatScan (REF) for primer and re-
striction sites.
bAtotal of 18,932 out of 27,060 sequences were recognized bv the 63F primer when the criteria required a perfect match in the four bases at the 3 terminus and al-
lowed up to three mismatches in the 5 proximal 17 bases.

Hence, if only the forward primer is labeled, neither gel nor as potential problems ( 10) because kinetically rather than
capillary electrophoresis will resolve these large terminal thermodynamically formed secondary structures may block
fragments. Thus, in selecting restriction enzymes for T- proximal restriction sites and reconstitute distal restric-
RFLP, both the number of unique targets and the distribu- tion sites, leading to pseudoterminal fragments. Hence,
tion of target sites should be considered. running controls is imperative. Included should be undi-
gested samples for each PCR and a digested amplified prod-
41.4.5.2. The Digestion Reaction uct derived from an isolate or clone to test for spurious
The community itself is a variable, and some empirical as- PCR products and partial or nonspecific digestions, respec-
sessment of enzyme effectiveness will inevitably be required tively. In addition, pretreating the PCR products with
for each community. In our experience, digesting from 200 mung bean nuclease may be necessary (10). Any inexpli-
to 600 ng of PCR product in a 10- to 15-pl reaction pro- cable band detected in these controls serves as an indicator
vides sufficient fluorescent material for subsequent detec- of undesirable events.
tion on an ABI gel or capillary, assuming that approxi-
mately 1 to 3 p l of the digest is loaded onto gels. If the 41.4.6. loading the Gels or Capillaries-Size
resulting profiles appear to be underloaded, the amount of Selection of Restriction Fragments
material loaded should be increased. In a complex commu- After digestion with restriction endonucleases and inacti-
nity, we have found that a total fluorescence of 5,000 to vation of the enzyme, the products are mixed with labeled
10,000U is optimal (2). size markers and denaturant (formamide), loaded, and elec-
trophoresed under denaturing conditions in an automated
41.4.5.3. Incomplete Digestions, Nonspecific Cuts, sequencing apparatus. The general format among the differ-
and Conformational Twists ent manufacturers of this equipment is roughly the same. In
Restriction enzymes are usually well behaved if used as rec- general, a slab gel format will provide reliable reads to 600
ommended by the vendor. However, because the fluorescent to 700 bases, while capillary setups extend the reliability of
detection is quite sensitive, controls should be employed to runs out to 800 to 1,000 bases. The former are relatively un-
ensure that the digestions are complete. Clearly, partial di- affected by the ionic conditions of digestion. However, cap-
gestions will produce fragments that will be misinterpreted illaries are loaded by electrophoretically injecting the DNA
as terminal. In the same spirit, overly long digestions (>4h) into the capillary; therefore, the ionic conditions of the re-
should be eschewed so as to avoid the formation of illegiti- striction digest may, if possessing high ionic strength, re-
mate digestion products. Recently, single-stranded PCR duce the efficiency of DNA loading. In this case a desalting
products and/or malformed duplexes have been implicated step is recommended after digestion. This can be as simple
920 a COMMUNITY AND GENOMIC ANALYSIS

as performing a quick ethanol precipitation on the digestion eral extractions of data may be required to collect all of the
products. In many cases, excellent results can be obtained tabulated data from the Genotyper project file. T-RLFP has
without desalting. another advantage in that the sequence database can be
used to determine the possible genus and species of the de-
41.4.6.1. Electrophoresis Conditions tected populations (29). It should be clear that one cannot
The conditions employed for electrophoresis vary from one identify the genus and species present with the size of a sin-
automated system to another. We provide two examples gle terminal fragment. Hence, the information derived from
here, derived from experience with ABI gel and capillary comparison of detected terminal fragment sizes with the
systems. For acrylamide gels, restriction digests of PCR- database supplies guidance for confirmatory experiments,
amplified community DNA are denatured at 94"C, chilled, not identity.
and loaded onto a 36-cm, 6% denaturing polyacrylamide
gel. Size standards (e.g., ABI Tamara 2500) are included in 41.4.8. Detailed Protocol for T-RFLP
each lane. Electrophoretic runs were 16 to 20 h in an ABI The following specific protocols have been productive for
automated sequencer (model 373A; Applied Biosystems broad surveys of phylogenetic domains using domain-spe-
Instruments, Foster City, CA) run in GeneScan mode with cific primers.
limits of 2,500 V and 40 mA. For capillary systems (e.g., 1. PCR amplification for T-RFLP. After extracting
ABI 3100 Genetic Analyzer) the denatured sample con- community DNA samples, small-volume pilot PCRs are
taining size standards (e.g., MM1000; Bioventures) is performed to determine optimal conditions followed by la-
loaded with injection times of 10 to 60 s into a 36-cm cap- beling reactions with increased volumes. As with DGGE, a
illary containing POP4 polymer. Electrophoresis is for ap- master mix containing all components except for the tem-
proximately 2 h at 50 kV. plate is prepared under aseptic conditions. The template is
41.4.6.2. Size Markers and Internal Controls aliquoted to reaction tubes, and the reaction is initiated
with the addition of master mix to the tubes. The following
Of paramount importance for fragment size determination PCR conditions are for 25-p1 pilot reactions. The volume is
is the inclusion of size standards in each electrophoretic scaled up to 100 p1 for the labeling reactions.
run. Because multiple detection channels can be operated
on many systems, size markers can be included in every Sterile deionized water . . . . . . . . . . . . . . . . . . 15.25 p l
lane, making size comparisons across lanes highly repro- lox PCR buffer ....................... 2.5 p l
ducible. In general, each automated electrophoresis manu- dNTP mix (2 mM each) . . . . . . . . . . . . . . . . . 2.5 p l
facturer includes software for determining fragment sizes MgC12 (50 mM) ....................... 0.75 pl
based on the electrophoretic mobility of size standards in- BSA (10 mg/ml) ....................... 0.25 pl
cluded in the run. In most cases, this provides sizing accu- 27F primer (10 pM) .................... 0.5 pl
racy of less than one base. However, in both gels and capil- 1492R or 1392R primer (10 pM) . . . . . . . . . . 0.5 p1
laries we have seen anomalies with respect to fragment size Tq DNA polymerase (10 U/pl) . . . . . . . . . . . 0.25 pl
estimate errors that are larger and range from 1 to 4 bases. Template DNA ........................ 2.5 pl
Such anomalies underscore the need for technical repli-
cates. 2. When employing the general domain-specific
primers described in Table 1, the following thermal cycling
41.4.7. Viewing and Collecting the Data parameters have proven successful.
Viewing and retrieving the data are largely dependent on
vendor-supplied software. In the case of ABI, Genescan, 94C for 5 min
the software used to run the gel, is used to view and collect 25 to 30 cycles of:
tabulated data as well. Figure 7 shows a T-RFLP profile de- 94C for 40 s for denaturation
rived from a soil sample in full range (Fig. 7A, 50 to 600
bases) and expanded range (Fig. 7B, 100 to 200 bases). Note 55 to 57C for 40 s for annealing
that by expanding the horizontal scale (fragment size), one w 72C for 1.5 min for polymerization
can easily discern peaks at a resolution of less than a base in 72C for 10 min (final extension)
length. From a GeneScan view of the profiles, the tabulated
4C until retrieval
data are extractable in an ASCI formatted file. The viewing
screen of GeneScan is limited to eight panels of data; thus, 3. Check PCR uroducts (2 to 5 d)on a 1% agarose gel
comparisons within a large data set are problematic. (1X TAE buffer; see section 41.3.4.2) and optimize PER
Genotyper software (ABI) overcomes this limitation by conditions if necessary.
permitting the user to import as many GeneScan files (a 4. Scale up volume to two 100-pl PCRs/sample for la-
single GeneScan file corresponds to one lane or capillary) beling and check reaction products on an agarose gel as de-
as is desired. Moreover, it also facilitates the alignment of T- scribed above.
RFLP profiles by controlling the extraction of the tabulated 5. Combine sample duplicates and purify the PCR
data from all selected scans in a way that allows the user to products with a commercially available kit (e.g., Promega or
bin all fragments within user-specified ranges. Thus, all frag- Qiagen). We have noted that elution of the DNA from
ments that are within a window of, for example, 20.5 base some commercial matrices is influenced by the source of the
are grouped together in the tabulated data. The data are for- DNA (presumably by copurifying salts).
matted with community samples (lanes or capillaries) in 6. Quantitate DNA concentration spectrophotometri-
rows and fragments (size and peak height) in columns. The cally (A260)or by fluorometry with PicoGreen (Molecular
shortcoming of this software is the column limitation of Probes, Inc.).
122. Thus, if your data set is derived from a community with 7. Incubate 200 to 400 pg of PCR product in a restric-
high diversity and therefore many terminal fragments, sev- tion digestion according to the manufacturer's recommen-
41. Analysis of Microbial Communities 921

3500 i Expanded in

3000
2500
i- Panel B

2000 -
1500 -
1000 -
500

0,
1
$ I I I I I I I I I I I
50 100 150 200 250 300 350 400 450 500 550

2400
2200 :
2000 :
1800 I
1600 -
1400 I
1200 --
1000 I
800
600
400 :
200 :
0 -
I
I I I I I I I
120 130 140 150 160 170 180

FIGURE 7 T-RFLP profile of a soil community. Samples were amplified with HEX-labeled 27F
primer matched with unlabeled 1492R targeting the 16s rRNA genes. PCR products were digested
with HhaI and run on an ABI 273A gel system. T-RFLP profiles were viewed with Genescan. (A)
A 50- to 600-base scale on abscissa; (B) expanded base scale of data from panel A.

dation. Generally this is performed in 10- to 2 0 - 4 reactions 10. Terminal fragment sizes are estimated with several al-
as described below. gorithms that are usually part of software for the automated
gel system. For example in the ABI systems, the GeneScan
200 to 400 ng of labeled PCR product software provides five algorithms for interpolating fragment
5 to 10 U of restriction endonuclease sizes from known standards (second- and third-order least
1.0 p1 of lox buffer squares, cubic spline, and local and global southern).
HzO to 10 p1 11. The fragment data, including fragment length and
peak amplitude, are binned or grouped by fragment length
8. Terminate digestion by heating to 75C for 10 min, across samples or communities. These data are extracted
and freeze samples until electrophoresis. from the sequencer software and imported into a spread-
9. Load 1 to 2 ~1 of each sample onto a sequencing gel sheet (e.g., ExcelTM)for comparative community analysis
or inject for 10 to 30 s onto a capillary for electrophoretic (see above). Fragment sizes of apparent interest, as judged
separation of fragments. Each lane or capillary also contains by distribution across communities or peak amplitudes, are
size markers for determination of fragment sizes. Electro- compared to a terminal restriction fragment database to de-
phoresis is performed as described above. termine possible phylogenetic affiliation.
922 COMMUNITY AND GENOMIC ANALYSIS

41.5. SUMMARY 13. Hill, T. C. J., K. A. Walsh, J. A. Harris, and B. F.


It has been only through the window of molecular tech- Moffett. 2003. Using ecological diversity measures with
bacterial communities. FEMS Microbiol. Ecol. 43: 1-1 1.
niques that we have come to appreciate the great diversity 14. Hurtle, W., D. Shoemaker, E. Henchal, and D.
of the microbial world. DGGE and T-RFLP are imperfect Norwood. 2002. Denaturing HPLC for identifying bacte-
methods for assessing this microbial diversity because at ria. BioTechniques 33:386-391.
present they are incapable of distinguishing every popula- 15. Johnson, A. R., and D. W. Wichern. 2003. Applied
tion within a community and every genomic variant within Multivariate Statistical Analysis, 5th ed. Prentice Hall,
a population. Nonetheless, these techniques provide the Upper Saddle River, NJ.
most rigorously tested and cost-effective approaches for the 16. Kitts, C. L. 2001. Terminal restriction fragment patterns:
ecological assessment of a large number of communities. a tool for comparing microbial communities and assessing
Used judiciously, comparative appraisals of complex com- community dynamics. Cum. Issues Intest. Microbiol. 2:17-25.
munities with DGGE and T - R E P can reveal even subtle 17. Kowalchuk, G. A., J. R. Stephen, W. Deboer, J. I.
changes in community structure. Prosser, T. M. Embley, and J. W. Woldendorp. 1997.
Analysis of ammonia-oxidizing bacteria of the subdivi-
sion of the class Proteobucteria in coastal sand dunes by de-
4 1.6. REFERENCES naturing gradient gel electrophoresis and sequencing of
1. Amann, R. I., W. Ludwig, and K.-H. Schleifer. 1995. PCR-amplified 16s ribosomal DNA fragments. Appl. En-
Phylogenetic identification and in situ detection of indi- viron. Microbiol. 63:1489-1497.
vidual microbial cells without cultivation. Microbiol. Rev. 18. Kudo, T., M. Ohkuma, S. Moriya, S. Noda, and K.
59: 143-1 69. Ohtoko. 1998. Molecular phylogenetic identification of
2. Blackwood, C. B., T. Marsh, S. H. Kim, and E. A. Paul. the intestinal anaerobic microbial community in the
2003. Terminal restriction fragment length polymorphism hindgut of the termite, Reticuliternes speratus, without cul-
data analysis for quantitative comparison of microbial tivation. Extremophiles 2:155-161.
communities. Appl . Enuiron. Microbiol. 69:926-93 2. 19. Kuske, C. R., L. 0. Ticknor, M. E. Miller, J. M. Dunbar,
3. Bruce, K. D., and M. R. Hughes. 2000. Terminal restric- J. A. Davis, S. M. Barns, and J. Belnap. 2002.
tion fragment length polymorphism monitoring of genes Comparison of soil bacterial communities in rhizospheres
amplified directly from bacterial communities in soils and of three plant species and the interspaces in an arid grass-
sediments. Mol. Biotechnol. 16:261-269. land. Appl. Environ. Microbiol. 68:1854-1863.
4. Cole, J. R., B. Chai, T. L. Marsh, R. J. Farris, Q. Wang, 20. Lane, D. J. 1991. 16S/23S rRNA sequencing,p. 115-175.
S. A. Kulam, S. Chandra, D. M. McGarrell, T. M. In M. Goodfellow (ed.),Nucleic Acid Techniques in Bacterial
Schmidt, G. M. Garrity, and J. M. Tiedje. 2003. The Systematics. John Wiley &a Sons, New York, NY.
Ribosomal Database Project (RDP-11): previewing a new 21. Lane, D. J., B. Pace, G. J. Olsen, D. A. Stahl, M. L.
autoaligner that allows regular updates and the new Sogin, and N. R. Pace. 1985. Rapid determination of 16s
prokaryotic taxonomy. Nucleic Acids Res. 3 1:442443. ribosomal RNA sequences for phylogenetic analyses. Proc.
5. Dice, L. R. 1945. Measures of the amount of ecologic as- Natl. Acad. Sci. USA 82:6955-6959.
sociation between species. Ecology 26:297-302. 22. Lipski, A., U. Friedrich, and K. Altendorf. 2001.
6. Dollhopf, S. L., S. A. Hashsham, and J. M. Tiedje. 2001. Application of rRNA-targeted oligonucleotide probes in
Interpreting 16s rDNA T-RFLP data: application of self- biotechnology. Appl. Microbiol. Biotechnol. 56:40-5 7.
organizing maps and principal component analysis to de- 23. Liu, W. T., T. L. Marsh, H. Cheng, and L. J. Forney.
scribe community dynamics and convergence. Microb. 1997. Characterization of microbial diversity by determin-
Ecol. 42:495-505. ing terminal restriction fragment length polymorphisms of
7. Domann, E., G. Hong, C. Imirzalioglu, S. Turschner, J. genes encoding 16s rRNA. Appl. Environ. Microbiol. 63:
Kuhle, C. Watzel, T. Hain, H. Hossain, and T. 45 16-45 22.
Chakraborty. 2003. Culture-independent identification of 24. Liu, W. T., T. L. Marsh, and L. J. Forney. 1998. De-
pathogenic bacteria and polymicrobial infections in the termination of the microbial diversity of anaerobic-aerobic
genitourinary tract of renal transplant recipients. J. Clin. activated sludge by a novel molecular biological tech-
Microbiol. 4 1:5500-55 10. nique. Water Sci. Technol. 37:417422.
8. Don, R. H., P. T. Cox, B. J. Wainwright, K. Baker, and 25. Lukow, T., P. F. Dunfield, and W. Liesack. 2000. Use of
J. S. Mattick. 1991. TouchdownPCR to circumvent spu- the T-RFLP technique to assess spatial and temporal
rious priming during gene amplification. Nucleic Acids Res. changes in the bacterial community structure within an
19:4008. agricultural soil planted with transgenic and non-trans-
9. Dsouza, M., N. Larsen, and R. Overbeek. 1997. genic potato plants. FEMS Microbiol. Ecol. 32:241-247.
Searching for patterns in genomic data. Trends Genet. 26. Marchesi, J., T. Sato, A. Weightman, T. Martin, J. Fry,
13:497498. S. Hiom, and W. Wade. 1998. Design and evaluation of
10. Egert, M., and M. W. Friedrich. 2003. Formation of useful bacterium-specific PCR primers that amplify genes
pseudo-terminal restriction fragments, a PCR-related bias coding for bacterial 16s rRNA. Appl. Environ. Microbiol.
affecting terminal restriction fragment length polymor- 64:795-799.
phism analysis of microbial community structure. Appl. 27. Marsh, T. L. 1999. Terminal restriction fragment length
Environ. Microbiol. 69:2555-2562. polymorphism (T-RFLP): an emerging method for charac-
11. Friedrich, M. W., D. Schmitt-Wagner, T. Lueders, and terizing diversity among homologous populations of ampli-
A. Brune. 2001. Axial differences in community Structure fication products. Curr. Opin. Microbiol. 2:323-327.
of Crenarchaeota and Euryarchueota in the highly compart- 28. Marsh, T. L., W.-T. Liu, L. J. Forney, and H. Cheng.
mentalized gut of the soil-feeding termite Cubiternes or- 1998. Beginning a molecular analysis of the eukaryal com-
thognarhw. Appl. Environ. Microbiol. 67:48804890. munity in activated sludge. Water Sci. Technol. 37:455-
12. Goldenberg, O., S. Herrmann, G . Marjoram, M. Noyer- 460.
Weidner, G. Hong, S. Bereswill, and U. B. Gobel. 2007. 29. Marsh, T. L., P. Saxman, J. Cole, and J. Tiedje. 2000.
Molecular monitoring of the intestinal flora by denaturing Terminal restriction fragment length polymorphism analy-
high performance liquid chromatography. J. Microbiol. sis program, a web-based research tool for microbial com-
Methods 68:94-105. munity analysis. Appl. Enuiron. Microbiol. 66:3616-3620.
41. Analysis of Microbial Communities rn 923

30. Medlin, L., H. J. Elwood, S. Stickel, and M. L. Sogin. 42. Schmitt-Wagner, D., M. W. Friedrich, B. Wagner, and
1988. The characterization of enzymatically amplified eu- A. Brune. 2003. Axial dynamics, stability, and interspecies
karyotic 16s-like rRNA-coding regions. Gene 71:491- similarity of bacterial community structure in the highly
499. compartmentalized gut of soil-feeding termites ( C u b i t m s
31. Moeseneder, M. M., J. M. Arrieta, G. Muyzer, C. spp.). Appl. Enwiron. Microbiol. 69:60184024.
Winter, and G. J. Herndl. 1999. Optimization of termi- 43. Sheffield, V. C., D. R. Cox, L. S. Lerman, and R. M.
nal-restriction fragment length polymorphism analysis for Myers. 1989. Attachment of a 40-base-pair G + C-rich
complex marine bacterioplankton communities and com- sequence (GC-clamp) to genomic DNA fragments by the
parison with denaturing gradient gel electrophoresis. Appl. polymerase chain reaction results in improved detection of
Environ. Microbiol. 65:35 18-3525. single-base changes. Proc. Natl. Acad. Sci. USA 86:232-
32. Morgan, C. A,, A. Hudson, A. Konopka, and C. H. 236.
Nakatsu. 2002. Analyses of microbial activity in biomass- 44. Sokal, R., and P. H. A. Sneath. 1963. Principles of
recycle reactors using denaturing gradient gel elec- Numerical Taxonomy. Freeman Publishers, San Francisco,
trophoresis of 16s rDNA and 16s rRNA PCR products. CA.
Can. J. Microbiol. 48:333-341. 45. Stahl, D. A., and R. I. Amann. 1991. Development and
33. Muyzer, G., E. C. de Waal, and A. G. Uitterlinden. amplification of nucleic acid probes, p. 205-248. In M.
1993. Profiling of complex microbial populations by dena- Goodfellow (ed.), Nucleic Acid Techniques in Bacterial
turing gradient gel electrophoresis analysis of polymerase Systematics. Wiley, New York, NY.
chain reaction-amplified genes coding for 16s rRNA. 46. Staley, J. T., and A. Konopka. 1985. Measurement of in
Appl. Environ. Microbiol. 59:695-700. situ activities of nonphotosynthetic microorganisms in
34. Nakatsu, C. H., V. Torsvik, and L. Ovreias. 2000. Soil aquatic and terrestrial habitats. Annu. Rev. Microbiol. 39:
community analysis using DGGE of 16s rDNA poly- 321-346.
merase chain reaction products. Soil Sci. SOC.Am. J. 64: 47. Torsvik, V., J. Goksoyr, and F. L. Daae. 1990. High
1382-1388. diversity in DNA of soil bacteria. Appl. Environ. Microbiol.
35. Olsen, G. J., N. R. Pace, M. Nuell, B. P. Kaine, R. 56:782-787.
Gupta, and C. R. Woese. 1985. Sequence of the 16s 48. van Hannen, E. J., M. P. van Agterveld, H. J. Gons, and
rRNA gene from the thermoacidophilic archaebacterium H. J. Laanbroek. 1998. Revealing genetic diversity of eu-
Sulfolobus solfataricus and its evolutionary implications. karyotic microorganismsin aquatic environments by dena-
J. Mol. Eool. 22:301-307. turing gradient gel electrophoresis.J. Phycol. 34:206-213.
36. Ovreis, L., L. Forney, F. L. Daae, and V. Torsvik. 1997. 49. von Wintzingerode, F., U. B. Gobel, and E.
Distribution of bacterioplankton in meromictic Lake Stackebrandt. 1997. Determination of microbial diversity
Saelenvannet, as determined by denaturing gradient gel in environmental samples: pitfalls of PCR-based rRNA
electrophoresis of PCR-amplified gene fragments coding analysis. FEMS Microbiol. Rev. 21:213-229.
for 16s rRNA. Appl. Enwiron. Microbiol. 63:3367-3373. 50. Wagner, T., D. Stoppa-Lyonnet, E. Fleischmann, D.
37. Pielou, E. C. 1969. An Introduction to Mathematical Muhr, S. Pages, T. Sandberg, V. Caux, R. Moeslinger, G.
Ecology. Wiley-Interscience,New York, NY. Langbauer, A. Borg, and P. Oefner. 1999. Denaturing
38. Preston, C. M., K. Y. Wu, T. F. Molinski, and E. F. high-performance liquid chromatography detects reliably
DeLong. 1996. A psychrophilic crenarchaeon inhabits a BRCAl and BRCA2 mutations. Genomics 62:369-376.
marine sponge: Cenarchaeum symbiosum gen. nov., sp. nov. 51. Wawer, C., and G. Muyzer. 1995. Genetic diversity of
Proc. Natl. Acad. Sci. USA 93:6241-6246. Desulfowibrio spp. in environmental samples analyzed by
39. Priemt, A., G. Braker, and J. M. Tiedje. 2002. Diversity denaturing gradient gel electrophoresis of [NiFe] hydroge-
of nitrite reductase (nirK and nirS) gene fragments in nase gene fragments. Appl. Enuiron. Microbiol. 61:2203-
forested upland and wetland soils. Appl. Environ. Microbiol. 2210.
68: 1893-1900. 52. Wilbur, J. D., J. K. Ghosh, C. H. Nakatsu, S. M.
40. Raskin, L., J. M. Stromley, B. E. Rittmann, and D. A. Brouder, and R. W. Doerge. 2002. Variable selection in
Stahl. 1994. Group-specific 16s rRNA hybridization high-dimensional multivariate binary data with applica-
probes to describe natural communities of methanogens. tion to the analysis of microbial community DNA finger-
Appl. Enuiron. Microbiol. 60:1232-1240. prints. Biomecrics 58:378-386.
41. Sambrook, J., and D. W. Russell. 2001. Molecular 53. Xiao, W. Z., and P. J. Oefner. 2001. Denaturing high-
Cloning: a Laboratory Manual, 3rd ed. Cold Spring Harbor performance liquid chromatography: a review. Hum.
Laboratory Press, Cold Spring Harbor, NY. Mutat. 17:439474.
introduction to Mycology 927 44. Microbiological and Genetic Methods
GEORGE A. MARZLUF, Editor for Filamentous Fungi 965
ROWLAND H. DAVIS AND
42. Methods for Studying Terrestrial Fungal A. JOHN CLUTTERBUCK
Ecology and Diversity 929
R. G. THORN, J. SCOTT, 45. Principles and Practice of DNA
AND M. A. LACHANCE Microarray Technology 978
KRISHNAMURTHY NATARAJAN,
43. Physiology, Metabolism, and Molecular MATTHEW J. MARTON, AND
Aspects of Filamentous Fungi 952 ALAN G. HINNEBUSCH
GEORGE A. MARZLUF
Introduction to Mycology
GEORGE A. MARZLUF

It is fitting that fungi have been included in this new edi- ronmental settings, including sampling methods, micro-
tion of Methods for General and Molecular Microbiology. scopic examination, and culturing. Complete formulations
Filamentous fungi and yeasts represent a huge and diverse to prepare eight different common media for fungal isola-
group of organisms that impact human life in many ways, tion are provided. The chapter by Davis and Clutterbuck
including their use in the production of various foods and describes practical microbial and genetic techniques for fil-
of life-saving antibiotics. Many fungal plant pathogens amentous fungi, including basic minimal and complete
cause enormous loss of food production, and others con- media as well as a special synthetic crossing medium. They
tribute to spoilage. Pathogenic fungi which attack animals, present several methods for measuring growth rate and de-
including humans, cause severe diseases and are difficult to scribe procedures for extraction of low-molecular-weight
control. In another vein, both filamentous fungi and yeasts molecules and macromolecules, followed by a description
have been employed as model organisms in research that of methods to isolate mutants and to analyze them by vari-
has provided major new scientific insights. The first aux- ous genetic approaches. The next chapter describes the
otrophic mutants isolated in any organism were obtained physiology and special molecular aspects of filamentous
in Neurospora by Beadle and Tatum in their ground-breaking fungi. This includes a description of the biochemistry and
work that represented the first step in the molecular revo- the genetic regulation of nitrogen, carbon, sulfur, and phos-
lution of genetics. Neurospora, yeasts, Aspergillus, Sordaria, phorus metabolism, and assays for representative enzymes
Coprinus, Ascobolus, and other fungi have made many in each of these areas. After a description of pH control of
fundamental contributions, e.g., to understanding the enzyme synthesis and iron acquisition, various special mo-
mechanism of recombination, chromosome behavior, lecular techniques for filamentous fungi are presented, in-
complementation, and eukaryotic gene regulation. With cluding nucleic acid isolation, cloning, transformation pro-
the advent of recombinant DNA approaches, interest in tocols, and methods to prepare knockout strains. In the
the fungi has increased dramatically in recent years, and chapter, the strategies and power of microarray analysis are
major developments in understanding complex phenom- presented in sufficient detail to allow new investigators to
ena such as circadian rhythms and light responses have re- adopt it in their work.
sulted. DNA-mediated transformation is now routine with To gain more insight into the topics presented in these
yeasts and multiple species of filamentous fungi; indeed, it chapters and to explore many other important areas relat-
appears that most fungal species can be transformed. The ing to filamentous fungi, one can consult The Mycota, a se-
Fungal Genetics Stock Center located at the University of ries of eight volumes that represent a comprehensive trea-
Kansas maintains and distributes thousands of wild-type tise on fungi as experimental systems for basic and applied
and mutant strains of Aspergillus, Neurospora, Fusarium, research (2). A number of diverse and exciting subjects are
Podospora, Sordaria, Ascobolus, and other fungal species, as covered in an earlier work, Molecular Biology of Filamentous
well as vectors and genomic libraries. The Fungal Genetics Fungi ( 6 ) , and also in a very recent volume, Molecular and
Meeting held biannually has grown in attendance so Cellular Biology of Filamentous Fungi (7). A superb book en-
greatly that space at the Asilomar Conference grounds in titled Neurospora, Contributions of a Model Organism, by
Monterey, CA, is limited. A new, rigorous American Rowland Davis (coauthor of the second chapter in the sec-
Society for Microbiology-sponsored journal, Eukaryotic tion devoted to fungi), although focused on Neurospora,
Cell, is devoted to eukaryotic microorganisms and high- presents a wealth of information for anyone interested in
lights many of the most exciting new developments with filamentous fungi (1). A recently published volume deals
fungi. The entire genomes of Neurospora crassa, Aspergillus with fungal genomics and addresses issues that arise with
nidulans, Saccharomyces cerevisiae, and other fungi have complete genomic sequences (5). Finally, three extensive
completely revolutionized molecular approaches and volumes by C. Guthrie and G. R. Fink in the Methods in
allow meaningful comparisons of entire genomes and pro- Enzymology series present a comprehensive set of sophisti-
teomes. cated genetic and molecular biology methods for yeasts ( 3 ,
The chapters devoted to the fungi are designed to pro- 4).
vide information to allow new investigators to initiate
work with these fascinating organisms but also to be of as- REFERENCES
sistance to workers already in the field. The ecology of 1. Davis, R. H. 2000. Neurospora, Contributions of a Model
fungi and the methods used to study this challenging area Organism. Oxford University Press, Oxford, United
are addressed in an authoritative chapter by Thorn et al. in Kingdom.
which they describe the various groups of fungi and other 2. Esser, K., and P. A. Lemke (ed.). 1995. The Mycota, vol.
fungus-like organisms and point out that no single method 1 to 8. Springer-Verlag,Berlin, Germany.
will allow identification and isolation of fungi from vastly 3. Guthrie, C., and G. R. Fink. 1991. Methods in
different situations. A series of detailed methods are then Enzymology, vol. 194. Guide to Yeast Genetics and Molecu-
described to study fungi and yeasts from a diversity of envi- lar Biology. Academic Press, San Diego, CA.

927
928 w MYCOLOGY

4. Guthrie, C., and G. R. Fink. 2002. Methods in En- 6. Stahl, U., and P. Tudzynski (ed.). 1992. Molecular Biology
zymology, vol. 350 and 351. Guide to Yeast Genetics andMo- of Filamentous Fungi. VCH, Weinheim, Germany.
lecuhr and Cell Biology. Academic Press, San Diego, CA. 7. Talbot, N. (ed.). 2001. Molecular and Cellular Biology
5. Prade, R. A,, and H. J. Bohnert (ed.). 2003. Genomics of of Filamentous Fung-a Practical Approach. Oxford
Plants and Fungi. Marcel Dekker, Inc., New York,NY. University Press, Oxford, United Kingdom.
Methods for Studying Terrestrial Fungal Ecology
and Diversity
R . G . THORN. J . SCOTT. AND M . A. LACHANCE

42.1. INTRODUCTION ......................................... 929


...........................
42.1.1. Importance of Terrestrial Fungi 929
. .................. 930
2. Groups of Fungi and Funguslike Organisms
42.1.2.1. Nonfungi: Actinomycetes. Oomycota.
.............. 930
Hyphochytriomycota. and Slime Molds
. 2. Kingdom Fungi: Chytridiomycota. Zygomycota.
....... 930
C&nneromycota. Ascomycota. and Basidiomycota
42.2. METHODS .............................................. 930
............................ 930
42.2.1. Terrestrial Filamentous Fungi
............... 930
42.2.1.1. Collecting and Culturing Macrofungi
. .............. 934
2. Detection and Isolation of Microfungi
. ........... 934
3. Methods for Mycorrhizae and Endophytes
.......... 935
42.2.2. Sampling and Analysis of Indoor Environmental Fungi
42.2.2.1. Sampling Methods............................. 935
.......... 945
42.2.3. Principles and Methods of Yeast Community Analysis
42.2.3.1. ................................ 945
Introduction
. ...........................
2. Collection Methods 946
. 3. Sampling and Data Management. Sample Size. and
................................... 947
Numbers
. ................................ 947
4. Identification
. ........................ 947
5. Ecological Interpretation
42.2.4. Common Media and Reagents ........................... 947
42.2.4.1. Rose Bengal Agar ............................ 947
. 2. DG18 Agar................................. 948
. 3. ME Agar................................... 948
. 4. MMN Medium ............................... 948
.
.5 BAF Agar .................................. 948
. 6. YMAgar ................................... 948
. ........................... 948
7. Yeast Nitrogen Base
. ............................ 948
8. Yeast Carbon Base
. .......................... 948
9. Antibacterial Agents
............................ 948
.10. Antifungal Agents
... 948
.11. 2X CTAB for Preserving Samples for DNA Studies
42.3. REFERENCES ............................................ 949

42.1. INTRODUCTION they may also protect plant roots from attack by pathogens.
including other fungi (4. 102). Fungi living within above-
.
42.1 1 . Importance of Terrestrial Fungi ground plant parts. the endophytic fungi. may protect these
Fungi are of fundamental importance in terrestrial ecosys- plants from herbivory or attack by pathogens. sometimes at
tems. and their roles and importance are usually overlooked a cost of flowering and sexual reproduction (33.90.92.96).
or underestimated by ecologists who study plants or ani- The filamentous growth form of many fungi and their abil-
.
mals Fungi drive terrestrial nutrient cycles through their ity to simultaneously or successively perform more than
abilities to decompose the complex carbohydrates-cellu- one nutritional role (decomposer. symbiont. or predator)
lose. hemicelluloses. and lignin-that make up the major- allow them to act as bridges across time. space. and trophic
ity of plant biomass. which in turn accounts for approxi- levels in ecosystems (5. 76. 109. 111). In contrast. predom-
mately 90% of the total biomass in most terrestrial inantly unicellular. nonfilamentous yeasts are adapted for
ecosystems (34.37.89). Other fungi. the mycorrhizal fungi. rapid response to rich. moist environments and may con-
.
form symbiotic associations with the roots of living plants vert sugar solutions such as nectar or plant sap into more
These mutualistic symbioses are vital to the survival of nutritious-or alcoholic-food for associated insects or
most green plants in natural ecosystems. Mycorrhizal fungi other animals. including humans (70. 115). Filamentous
help plants obtain nutrients and water in environments fungi with a habit of exuding adhesive extracellular poly-
where these are chronically or periodically lacking. and saccharides or mucopolysaccharides are important in soil

929
930 MYCOLOGY

stabilization through the formation of microaggregates and The Glomeromycota (Fig. Id) are the fungi that form mu-
the binding of aggregates and particles (29, 122). In the tualistic symbioses called arbuscular mycorrhizae with the
human environment, fungi are important in many food and roots of approximately 90% of the vascular plants in the
industrial fermentations, but they also cause food spoilage world; these symbioses enable plants in natural environ-
and grow unwanted in our living and working spaces, creat- ments to survive drought and nutrient stress (102).
ing problems of environmental health (7,41,87, 100). This Glomeromycota are coenocytic, with tubular cells contain-
chapter can only touch on the fundamentals of discovering ing hundreds of nuclei. Although they form large spores, of
and identifying fungi in these diverse environments. 100 to 1,000 p,m in diameter, and networks of hyphae that
may extend a meter or more, they are otherwise only mi-
42.1.2. Groups of Fungi and Funguslike Organisms croscopically visible. Recognition of the monophyletic phy-
lum Glomeromycota resolves the paraphyly of the formerly
42.1.2.1. Nonfungi: Actinomycetes, Oomycota and broad Zygomycota including Glomales with respect to the
Hyphochytriomycota, and SI ime Molds Chytridiomycotu (98). The remaining Zygomycota (Fig. l e )
Several groups of organisms that are not true fungi have tra- are a diverse group of fungi in terms of morphology and
ditionally been studied by mycologists or thought of as ecology and include soil saprobes, parasites of soil fungi or
fungi. Actinomycetes are prokaryotic and are members of the algae, and parasitoids of insects and soil invertebrates, but
G+C-rich gram-positive Bacteria (85, 119). They resemble few significant plant pathogens (20).
fungi because of their filamentous growth, reproduction by The Ascomycotu (Fig. l a ) are the largest group of fungi
spores, and production of extracellular enzymes (46, 62, 88; and include most of the asexual or mitosporic fungi that
http://www.nih.go.jp/saj/DigitalAtlas/index.htm). Although were formerly classified in the Deuteromycetes or Fungi
mycologists and soil biologists will encounter actinomycetes Imperfecti (60,93,99). The Ascomycota include soil-borne
in their studies, they are not treated here. The Oomycota pathogens of crop plants, endophytes that live within plant
and Hyphochytriomycotu are water molds with motile, flag- tissues, usually without causing disease symptoms, most
ellated spores. The anterior flagella in both groups are fungi that cause human and animal diseases, plus groups
called tinsel flagella and have two rows of fine, tubular hairs that are mycorrhizal, parasitic on other soil fungi, or preda-
resembling those of the golden-brown algae. The natural re- tory or parasitic on insects or other invertebrates (16, 19,
lationships of the water molds, as determined by DNA se- 30, 33, 45, 58, 86, 92, 94, 96). In addition, the majority of
quence analyses and other lines of evidence, are with the lichens are in the Ascomycota (60). Lichens are symbiotic
chrysophyte algae in the kingdom Stramenopila (113, 114) associations between fungi and green algae or cyanobacte-
(also called Chromistu or Heterokonta [13]). Slime molds ria. The fungal partner (mycobiont) forms the characteris-
(the Mycetozoa or fungus animals) are relatives of amoe- tic structure that we call a lichen, which encloses and
bae and have an ameboid feeding stage and a spore-bearing protects the alga or cyanobacterium (photobiont) (2, 50).
reproductive stage (11-13, 56). None of these groups be- Finally, most yeasts (Fig. If), including the most economi-
longs in the kingdom Fungi. cally important ones such as Succhuromyces cerewisiue, be-
long in the Ascomycota (65). Yeasts are fungi adapted to life
42.1.2.2. Kingdom Fungi: Chytridiomycota, in aqueous environments by growth as separate, usually
Zygomycota, Glomeromycota, Ascomycota, and elliptical cells that divide by budding or fission (14,63) and
Basidiomycota are discussed further in section 42.2.3 below.
Most current classifications of fungi recognize four phyla: The Busidiomycotu (Fig. lb) are the second largest group
Chytridiomycota, Zygomycotu, Ascomycota, and Busidiomycotu of true fungi and may be divided into four major groups, the
(3, 60). There is good evidence from molecular phyloge- Ustilaginomycetes (smuts), Urediniomycetes (rusts), Hetero-
netic studies, fossil record, and ecology for a fifth fungal basidiomycetes (jelly fungi), and Homobasidiomycetes (mush-
phylum, Glomeromycota (98). The Assembling the Fungal rooms and relatives) (60). The thousands of species of
Tree of Life group (http://aftol.org/) has spearheaded recent Homobasidiomycetes whose mycelial phase occurs in soil
phylogenetic analyses of the fungi based on sequences of ri- have mostly been overlooked in surveys of soil fungi (55,
bosomal and other genes. These have led to substantial 110).The Homobasidiomycetes include important crop path-
changes to classification and naming of fungal groups (52a) ogens, including Rhizoctonia (sexual state Thunatephorus [6,
compared to the conservative approach taken here. In ad- 103, 1051) and timber pathogens Armillaria, Phellinus, and
dition, sequencing of ribosomal DNAs (rDNAs) isolated G a n o d e m ( 108), saprotrophic leaf- and wood-decomposing
from soils has led to the recent discovery of several un- fungi (53, 121), and the majority of fungi that form ecto-
known and uncultured lineages with very deep roots in the mycorrhizal symbioses with woody vascular plants in 30
fungal kingdom (112). The truly innovative investigator families (47, 54, 102).
may develop techniques to see, cultivate, and describe these The fundamentally different biologies of different groups
fungi and discover their role in nature, but our discussion is of fungi mean that no single method will work to discover or
limited to groups known as whole organisms. isolate all fungi in any material or area. For this reason, some
Like the Oomycota, chytrids are aquatic fungi with flag- general methods are presented here, which users may need to
ellated zoospores that require free water for motility (Fig. modify to study their fungi of particular interest. Biodiversity
lc). Zoospores of most chytrids have one posterior flagel- of Fungi (81a) presents an extensive review of this topic.
lum, but members of one group found in the rumen of her-
bivorous mammals are multiflagellate. Soil-inhabiting
chytrids include decomposers of cellulose, chitin, and ker- 42.2. METHODS
atin, as well as parasites of soil algae, invertebrates, and vas-
cular plants (15). The chytrids in rumen contribute to the 42.2.1. Terrestrial Filamentous Fungi
digestion of cellulosic plant cell walls in the animals food
(81). The four other phyla consist of predominantly fila- 42.2.1 . I . Collecting and Culturing Macrofungi
mentous forms, plus secondarily unicellular yeasts belong- Macrofungi are those fungi with fruiting bodies large
ing to both Ascomycotu and Basidiomycota. enough to see with the naked eye, usually defined as being
FIGURE 1 The major types of fungi. (a) Ascomyotu: Morchella esculentu, showing the correct method of collecting macrofungi;
inset, eight ascospores are produced in each sac-like ascus in this Succobolus (photo by G. L. Barron, University of Guelph). (b)
Busidiomycotu: Agaricus, associated with a fairy ring in Wyoming. (c) Chytridiomycotu: Cutenuria unguillulae attacking nematodes
(drawing by G. L. Barron, University of Guelph, from reference 18). (d) Glomeromycotu: a spore and hyphae of an endomyorrhizal
fungus washed from soil. (e) Zygomycotu: Cunninghmella echinulata, a striking and common fungus from soil. The inset shows a
close-up of the sporangiospores (both photos by G. L. Barron, University of Guelph). (f) Vegetative cells and ascus of Metschnikowiu
hwuiiensis, an ascomycetous yeast.

931
932 MYCOLOGY

at least 1 cm in one dimension. These fungi are relatively from drying by covering with a cup or small collecting jar
easy to collect, but a systematic approach and sampling de- (Fig. 3c). Keep in a cool place overnight, and a pattern of
sign are required for studies intended to yield results that are thousands or millions of spores will be deposited (Fig. 3d),
comparable between areas or years. Sampling plots should enabling the detection of spore color en masse and micro-
be based on a stratified random or regular transect design for scopic measurement of mature spores.
comparability between studies (Fig. 2), but for discovery of Once a spore print is set up and notes are taken, includ-
maximum diversity they should be combined with oppor- ing color annotations with reference to some color standard
tunistic sampling of known or suspected good habitats such as in the work of Kornerup and Wanscher (61) , the de-
(91). Plots should be repeatedly sampled during the fruiting cision is made whether to culture the specimen, preserve a
season or seasons, once a week being desirable but once per small piece of tissue for future DNA studies, or dry it for the
month probably more practical in terms of collection time herbarium. A good field preservative for tissues intended for
and effort. DNA studies is 2X CTAB buffer (see section 42.2.4.1 1 and
Care should be taken in collecting fruiting bodies of reference 43). A piece of tissue approximately 3 by 3 by 3
macrofungi to obtain the entire fruiting body (Fig. la). The mm, not including any contaminated external surface, can
base of the stem, if one is present, may be buried in the soil be cut out with a flamed and cooled scalpel and preserved
or other substratum. Collections should immediately be as- in 0.5 to 1.0 ml of 2 X CTAB in a 1.5- to 2.0-ml screw-cap
signed a collection number and recorded in a field note- vial and then kept at ambient temperatures until it is prac-
book. Collections should be packaged in the field to pre- tical to refrigerate. Freezing is unnecessary and may not
vent damage during transit back to the lab, including drying preserve the DNA as well as temperatures of 4 to 5C.
or, in rainy areas, saturation. Waxed paper packets or Kraft Specimens, and their field packets if Kraft paper bags were
paper bags are suitable for relatively robust specimens and used, should be dried at 40 to 50C with ample air flow. In
dry collecting areas, aluminum foil for rainy areas, and hard many situations, a portable food dehydrator with an electric
plastic fishing tackle boxes for minute or fragile specimens. heating element and fan is ideal. Most specimens will dry
The collection number should be written on the packet, or overnight, and very large specimens may be cut into slices
a slip of paper with the collection number should be packed to speed drying. In humid areas, dried specimens should
with the specimens. be packed immediately into airtight plastic bags such as
Many macrofungi can only be reliably identified if de- ZiplocTMbags to prevent rehydration and attack by molds or
tailed notes are made of characters detectable only when insects.
fresh (Fig. 3a). A good photograph, in situ or in the lab, can Culturing from specimens of macrofungi is often very
reduce the need for description of colors, textures, and size, simple and adds a whole new biological dimension to the
but notes on taste, odor, staining reactions, and the color of collection beyond the preserved, dead herbarium specimen.
a fresh and dried spore print are often vital for final identi- The spores of many saprotrophic macrofungi germinate
fication. Too often a mycologist is asked to identify the readily on simple agar media, such as ME agar (see section
voucher specimens from a detailed ecological study in some 42.2.4.3). The isolation medium may be supplemented with
remote locale, but finds that the specimens are without antibacterial antibiotics and, for isolation of basidio-
notes and, because they were poorly dried, are useless for mycetes, with benomyl. To obtain a polyspore culture, affix
study of microscopic features as well. For details and termi- a small portion of fruiting body, approximately 2 by 2 mm,
nology of macroscopic features of macrofungi, see reference hymenium side down, to the lid of a petri dish using a dot
72. To obtain a spore print, lay a fresh, moist portion of the of petroleum jelly. When spores on the agar surface are vis-
fruiting body, hymenium (spore-bearing surface) down on a ible by eye or using a dissecting or compound microscope,
piece of white paper or a microscope slide, and protect it the piece of tissue may be removed and a small piece of agar
bearing spores may be transferred aseptically to a fresh plate
of isolation medium. To obtain monospore, usually haploid,
cultures for mating studies, make a dilution series of spores
in sterile distilled water (sdH20).Spores deposited on agar
should be resuspended and plated immediately after deposit,
whereas spores collected on sterile microscope slides may be
resuspended and plated up to several weeks after deposit.
The correct dilution must be chosen so that germinating
spores are well separated on the agar surface, and germi-
nated spores should immediately be transferred to separate
plates or tubes. For robust specimens, and for fungi such as
ectomycorrhizal species whose spores do not readily germi-
nate, it is easier or better to make a tissue culture, which
will usually be dikaryotic. Break open the fruiting body and
extract a small tissue piece from this freshly exposed surface
using flamed and cooled forceps or scalpel, and transfer it
FIGURE 2 Sampling designs: stratified random (left side) directly to a plate of isolation medium (Fig. 3b). To be cer-
and regular grid (right). The heavy horizontal line represents tain that you are growing what you intended, you should
the baseline, filled circles represent 10-m intervals from which follow the germination and growth of spores or tissue pieces
sampling transects are struck at 90 to the baseline, and open at frequent intervals using a dissecting and compound mi-
circles represent sampling plots 4 m2 in area, with a radius of croscope, and immediately transfer the target fungus out of
1.14 m (91). In stratified random sampling, sample plots are lo- any plate that becomes contaminated with molds. Once
cated on the transects at distances taken from a random num- cultures are growing cleanly, they should be transferred to
bers table, e.g., in the left line, with their centers at 6.33 m plates (short-term working cultures) or slants (stock cul-
from the baseline, and then 2.17, 5.94, 4.67, and 8.07 m from tures) of medium without antibiotics. Slant cultures may be
the center of the previous plot. preserved by aseptically filling the vials with sterile, heavy
42. Methods for Fungi 933

FIGURE 3 Collecting and culturing macrofungi (a to d) and microfungi (e to f). (a) Taking notes on a fresh collection of
Amanita, Basidiomycota, using a color guide (61); (b) making a tissue culture from a fruiting body of Aguricus, Basidiomycotu; (c) the
procedure for obtaining a spore print; (d) a spore print; (e) a soil sprinkle plate for isolation of nematophagous fungi; (f) particle
washing to remove spores of abundantly sporulating molds to selectively recover more recalcitrant fungi from soil, including
Basidiomycotu. In this example, 5 g of soil was first dispersed by shaking for 1 h at 4C in 0.1 M sodium pyrophosphate decahydrate
and then poured through sieves of 250- and 53-pm mesh. Particles remaining o n the 53-pm mesh sieve were washed with a stream
of tap water. T h e remaining organic materials may be picked up in a l-ml, broad-bore pipette and inoculated onto agar or in l i p
uid medium or used in DNA extractions that are enriched for filamentous fungi.
Next Page

934 MYCOLOGY

mineral (paraffin) oil and storing at room temperature or soil fungal community, but it generally misses most basid-
with cryoprotectant such as 15% (vol/vol) glycerol and iomycetes and nutritional specialists such as the nematode-
storing in the vapor phase of liquid nitrogen or in a freezer destroying fungi, mycoparasites, and mycorrhizal fungi (32,
at -80C (59). 110). Comparability between studies requires a systematic
approach to picking colonies to be identified: either all or
42.2.1.2. Detection and Isolation of Microfungi the first 25 to 30 encountered randomly per plate should be
Many of the most interesting and important fungi cannot chosen (32). Particle plating starts by dispersing the mate-
be seen with the naked eye and collected. Instead, one must rial, or grinding it to fine particles if necessary, and washing
collect the material on which or in which they occur and particles (Fig. 3f) through sieves of a chosen size range, such
either examine this microscopically to observe the fungi as 50 and 250 pm, with tap water or sdHzO to remove the
or process it in some way to isolate the fungi into culture. spores of heavily sporulating molds such as Penicillium and
What follows is a discussion of tnethods suitable for recov- Aspergillus (10, 22). Washed particles are plated on standard
ery of many filamentous microfungi, particularly those be- or selective isolation medium, depending on the group of
longing to the Ascomycota and Zygomycota. Although many fungi desired (21, 110). Although a few principal species
Chytridiomycota are members of terrestrial soil ecosystems, may be recovered by such techniques, there may also be an
methods for their isolation into culture are essentially those enormously long tail to the species accumulation curve,
for aquatic fungi and are not included here (23, 42). with 8 to 15 previously unseen species added for every 100
However, amplifications of genomic DNA from soil using incremental isolates past 1,100 (31,32).
fungus-specific primers yield diverse sequences in the
Chymidiomycota and related basal lineages of fungi (112). 42.2.1.3. Methods for Mycorrhizae and Endophytes
One of the best and easiest methods to detect microfungi Fungi growing symbiotically within plant tissues require
is periodic observation of organic materials kept in moist specific techniques for isolation into culture. These include
chambers, using a dissecting microscope at X 5 to X50 mag- fungi involved in a variety of mycorrhizal associations and
nification. Fresh or moistened herbivore dung, leaf litter, also those growing endophytically within aerial tissues of
tree bark, living or dead roots, and decorticated dead wood plants, usually without causing disease symptoms. Among
are all excellent materials for study by the moist-chamber the mycorrhizal fungi are Glorneromycota forming arbuscular
method. A moist chamber may be as small as a petri dish or mycorrhizae (also called vesicular-arbuscular mycorrhizae or
as large as a 10-liter plastic crisper. Depending on the mate- endomycorrhizae), Ascomycota and Basidiomycotu forming
rial, a layer of moistened sand, perlite, or vermiculite cov- ectomycorrhizae, and several other forms. As mentioned
ered with filter paper or paper towels may be required in the above, the basidiospores of most ectomycorrhizal basid-
bottom of the chamber to maintain humidity. The soil iomycetes do not germinate readily on standard media, al-
sprinkle plate method for isolation of nematode-destroying though they may be stimulated to do so using activated
fungi (18) is a variation on the moist-chamber technique, charcoal or filter-sterilized root extracts (25, 84). Many of
in which a small amount of soil or organic matter is sprin- these fungi form large fruiting bodies from which tissue cul-
kled onto a plate of water agar (15 g of agar per liter of dis- tures may be derived, using more complex media than
tilled water) and approximately 200 nematodes from a stock might be used for saprotrophic fungi. Two agar media com-
culture are added as bait to stimulate germination, trap for- monly used for isolation of ectomycorrhizal basidiomycetes
mation, and development of nematode-destroying fungi are MMN medium and BAF agar (sections 42.2.4.4 and
(Fig. 3e). Astute fungal biologists may develop their own 42.2.4.5), usually supplemented with antibacterial antibi-
particular moist-chamber technique to detect or discover otics and with benomyl to suppress molds (57). These
their fungi of interest. Observations are made periodically media may also be used for isolation of ectomycorrhizal ba-
to note the appearance of novel fungi, and these may often sidiomycetes from mycorrhizal root tips that have been
be obtained in culture by picking single spores with a scrupulously washed and surface sterilized, although many
Pasteur pipette that has been drawn to a hair tip in a small attempts will still yield nonmycorrhizal ascomycetous molds
flame and transferring these to plates of isolation medium. that were on or in the root tip (107). MMN medium and
In addition to the moist-chamber technique, two basic BAF agar without benomyl may be used to isolate asco-
methods for culturing fungi from soil or organic materials are mycetous mycorrhizal fungi from root tips or fruiting bodies.
dispersion plating (also called dilution plating) and particle Ectomycorrhizal root tips may be characterized and identi-
plating (with soil, also called soil washing). For dispersion fied to some extent using morphological features (1, 49),
plating of soil, a small amount such as 5 g (fresh weight) of and the fungal associate may be identified by selective am-
soil is dispersed in a series of volumes of sdHzO to obtain a plification followed by sequencing of fungal ribosomal genes
dispersion that yields 25 to 30 colonies on a petri plate of iso- (27, 43, 44). Glomalean mycorrhizal fungi (GMF) have
lation medium. The series of 5 g of soil in 125 ml of sdH20, generally resisted attempts at axenic culture but can be cul-
1 ml dispersion 1 in 39 ml of sdH20, and 1 ml of dispersion tivated from infected root pieces or spores recovered from
2 in 9 ml of sdHzO yields dispersion ratios of 1:25, 1:1,000, soil in coculture with a host plant such as leeks (Allium por-
and 1:10,000. The last two are useful for many soils, with 0.5 rum 1251). Selective primers are also available to amplify
to 1.0 ml spread evenly over the surface of a 100-mm petri rDNA of GMF from roots or soil (101). Sequence-based
dish. It is important that the soil be well dispersed and identification of GMF promises to open the field of research
evenly suspended when taking the aliquot for the subse- into their ecology, previously inhibited by the difficult and
quent dispersion or for plating. Unused portions of soil sam- imprecise identification using spore shape and wall struc-
ples are weighed, dried, and reweighed to back-calculate the ture (20). One difficulty with molecular identification of
dry weight of soil used in the dispersion. A low-nutrient GMF, however, is that they are coenocytic. Their spores
medium supplemented with antibacterial antibiotics is gen- may each contain over 1,000 genetically distinct nuclei,
erally used, and we recommend Martins soil extract medium with several different alleles for rDNA (95), and frequently
with rose bengal and chloramphenicol (see section also contain sequences of unrelated contaminant or en-
42.2.4.1). Dispersion plating yields a diverse subset of the dosymbiotic fungi as well (97).
43
Physiology. Metabolism. and Molecular Aspects of
Filamentous Fungi
GEORGE A . MARZLUF

43.1. INTRODUCTION ........................................ 952


.
2. NITROGEN METABOLISM ................................ 953
43.2.1. Nitrogen Regulation ................................. 953
.
2. Nitrate Reductase .................................. 953
.
3. Other Nitrogen Pathways ............................. 954
43.3. CARBON METABOLISM .................................. 955
43.3.1. Carbon Sources and Catabolite Repression ................. 955
.
2. Complex Regulatory Systems .......................... 956
.
3. Enzyme Assays .................................... 956
43.4. SULFUR METABOLISM ................................... 956
43.4.1. The Sulfur Regulatory Circuit ......................... 956
.
2. Enzymes of Sulfur Metabolism ......................... 957
43.5. PHOSPHORUS METABOLISM .............................. 957
43.5.1. Genetic Control of Phosphorus Metabolism ................ 957
.
2. Phosphorus-Related Permeases and Enzymes ...............957
43.6. pH CONTROL OF EXTRACELLULAR ENZYMES AND
PERMEASES ............................................ 957
.
7. ACQUISITION OF IRON .................................. 958
43.7.1. Mechanisms for Iron Acquisition ........................ 958
.
2. Assays ........................................... 958
43.8. RNA AND PROTEIN SYNTHESIS ........................... 958
.
9. ISOLATION OF NUCLEIC ACIDS ........................... 959
.1 0. IDENTIFICATION AND CLONING OF FUNGAL GENES ........ 959
.1 1. TRANSFORMATION WITH EXOGENOUS DNA ............... 960
............................ 960
43.1 1.1. Transformation Procedures
. .................................. 960
2. Selectable Markers
. .................... 960
3. Nature of Transformed Fungal Strains
. .................. 961
4. Constructing Gene Disruptions in Fungi
43.12. OTHER METABOLIC PATHWAYS ........................... 961
.13. REFERENCES ........................................... 961

43.1. INTRODUCTION several fungal genomic libraries. The Stock Center oversees
The filamentous fungi display remarkable and diverse meta- the annual publication of the Fungal Genetics Newsletter.
bolic pathways and show great versatility in the utilization The genome sequences of N . crassa, A . nidulans. and
of sources of carbon. nitrogen. phosphorus. sulfur. and other Magnaporthe @sea have been completed. and sequencing of
metabolites and in acquiring essential elements. e.g., iron the genomes of several additional filamentous fungi will be
and potassium . Many of the filamentous fungi. particularly finished soon. All of this sequence information is available
the well-studied Aspergillus and Neurospora species. can via online links with the Fungal Genetics Stock Center
grow on defined media. Other than inorganic salts. e.g., website .
sodium nitrate and ammonium sulfate. Neurospora crussa Three entire volumes of Methods in Enzymology (41-43)
requires only a carbon source and one vitamin. biotin . The are devoted to the methods of yeast genetics and molecular
ability to grow on defined media coupled with a haploid and cell biology. with articles ranging from Getting Started
genome has facilitated the isolation of mutants which have with Yeast to detailed descriptions of genomics and proto-
been instrumental in studying fungal metabolism (see chap- cols specialized for yeasts.Researchers who wish to carry out
ter 44) . Mutants which lack a specific enzyme are very use- any work with the yeast Saccharomyces cereoisiae should
ful in metabolic studies. Many mutant strains of Aspergillus consult these authoritative volumes .
nidulans and other Aspergillus species. Neurospora crussa and The filamentous fungi secrete many enzymes into the ex-
additional Neurospora species. Fusarium. and Gekzsinospora tracellular environment. which allows them to hydrolyze
are available at the Fungal Genetics Stock Center. polysaccharides. nucleic acids. and proteins and to utilize the
University of Kansas Medical Center (http://www.fgsc.net). small-molecular-weight products for growth. Many fungi
The Stock Center also maintains and distributes an excel- also carry out a pattern of secondary metabolism. following
lent selection of cloned genes from Neurospora and the exponential growth phase. which results in the synthesis
Aspergillus as well as useful cloning vectors for fungi and of unusual compounds. e.g., penicillin (7. 14. 91). Entire

952
43. Physiology and Molecular Aspects of Fungi 1 953

areas of metabolism appear to be regulated in an integrated and NIT2 in Neurospora, turn on the expression of multiple
fashion, most often at the level of transcription, and involve sets of structural genes which encode enzymes of various ni-
the action of regulatory DNA binding proteins, metabolic trogen catabolic pathways (11, 17, 33,35, 70, 77, 95, 114).
inducers, and repressors. Feedback inhibition, compartmen- AREA and NIT2 are members of the GATA family of tran-
tation, and protein turnover also contribute significantly to scription factors, which have a Cys2lCysZ-type of zinc fin-
metabolic controls. Extensive and authoritative reviews of ger DNA binding motif. Interestingly, filamentous fungi
many aspects of fungal metabolism are available (5, 17, 28, contain multiple GATA factors; e.g., five GATA factors
50, 70, 71, 74, 96). Two recent books, The Neurospora have been characterized in Neurospora crussa, each of
Compendium by Perkins et al. (92) and Neurospora by Davis which is responsible for controlling a specific set of down-
(22), are invaluable for Neurospora workers. Neurospora con- stream genes (Table 1); the Neurospora genome sequence
tains a wealth of information concerning fungal cell biology suggests the presence of a sixth GATA factor which has not
and genetics and describes experimental approaches and yet been characterized. Nitrogen metabolite signaling in-
techniques which should be generally applicable to various volves an interaction of a negative-acting protein, NMR,
fungi. A n article by Goldman and Morris (39) describes with both the C terminus and GATA domain of the AREA
methods for cell and molecular biology studies of Aspergillus or NIT2 global activator (90, 93). Expression of each spe-
nidulans, many of which can provide insight for work with cific set of nitrogen catabolic genes also usually requires in-
any fungal species. Consult chapter 44 in this volume for a duction by a substrate or intermediate of the pathway and is
list of websites useful for fungal researchers. This chapter em- mediated by a pathway-specific regulatory protein. The pro-
phasizes practical aspects, methods, and simple assays for in- moters of the candidate structural genes contain binding
vestigating fungal physiology and metabolism and intro- sites for the global activator (AREA or NIT2) and for the
duces some of the specialized molecular techniques that pathway-specific activator, most of which have a single bi-
have proved invaluable in studying this fascinating group of nuclear Cys6/Zn2 DNA binding motif (Table 2).
eukaryotic microorganisms.
43.2.2. Nitrate Reductase
Investigators often focus on a key enzyme within a pathway
43.2. NITROGEN METABOLISM of interest, although several additional enzymes should also
be examined to ensure that correct conclusions are ob-
43.2.1. Nitrogen Regulation tained. Most filamentous fungi can utilize nitrate as an ex-
Nitrogen metabolism and its genetic regulation are impor- cellent nitrogen source, and its use requires a nitrate trans-
tant topics since most of the macromolecules of living cells porter and the enzymes nitrate reductase and nitrite
are rich in nitrogen, most notably nucleic acids and pro- reductase. Synthesis of these activities is highly regulated by
teins. The filamentous fungi, particularly Aspergillus nidu- nitrogen catabolite repression, induction, and major and
lam and Neurospora crassa, have contributed much to the minor control genes. Nitrate reductase is regarded as the
field of nitrogen metabolism (17, 70, 93). These fungi must key activity for the pathway of nitrate assimilation and has
assimilate nitrogen from a wide range of potential nitroge- been studied extensively (12, 17, 34). Nitrate reductase is a
nous compounds and utilize it to derive all the precursors very large homodimeric enzyme and possesses three separate
required for DNA, RNA, and protein synthesis. Both domains, an amino-terminal domain that contains an un-
Aspergillus and Neurospora preferentially utilize certain ni- usual molybdopterin cofactor (Mo-Co), a smaller central
trogen sources, e.g., ammonium and glutamine, a phenom- heme-containing domain, and a carboxy-terminal domain
enon known as nitrogen catabolite repression. Under dere- which contains a flavin (FAD). Electrons derived from
pression conditions, when such primary nitrogen sources NADPH are transferred stepwise to the flavin domain, to
are lacking or available only in limited amounts, global the heme domain, and to the molybdopterin-containing
positive-acting regulatory proteins, AREA in Aspergillus domain and, finally, used to reduce nitrate to nitrite.

TABLE 1 GATA factors of filamentous fungi


GATA factor Regulatory function" No. of zinc fingers Organism
AFAREA Nitrogen + 1 Aspergillus fumigatus
AREA Nitrogen + 1 Aspergillus nidulans
AREA Nitrogen + 1 Gibberella fujikuroi
AREA Nitrogen + 1 Metarhizium anisopliae
NRE Nitrogen + 1 Penicillium chrysogenum
NIT2 Nitrogen + 1 Neurospora crassa
NUT1 Nitrogen + 1 Magnaporthe pisea
SRE Siderophore - 2 Neurospora crassa
SREP Siderophore - 2 Penicillium chrysogenum
URBSl Siderophore - 2 Ustilago maydis
wc-1 Light + 1 Neurospora crassa
wc-2 Light + 1 Neurospora crassa
ASD4 Ascospore formation + 1 Neurospora crassa
'+, positive regulation; -, negative regulation.
954 MYCOLOGY

TABLE 2 Pathway-specific regulatory factors which mediate induction of catabolic genes in filamentous fungi
Pathway-specific factor Inducer Pathway DNA binding domain Species
ALCR Alcohol Alcohol cYs6 lzn2 Aspergillus nidulans
AMDA Acetate Acetate Cys2/His2 Aspergillus nidulans
AMDR T-amino acid Acetamide cYs6 En2 Aspergillus nidulans
FACB Acetate Acetate C Y SIzn2
~ Aspergillus nidulans
N IRA Nitrate Nitrate cYs6 IZn2 Aspergillus nidulans
NIT4 Nitrate Nitrate CYs61Zn2 Neurospora crassa
PRNA Proline Proline cYs6 IZn2 Aspergillus nidulans
UAY Purine Purine cYs6 IZn2 Aspergillus nidulans
PCOl Purine Purine CYSG IZn2 Neurospora crassa

Nitrate reductase activity can be readily determined xanthine dehydrogenase (Table 3). Moreover, mutations
using a convenient colorimetric assay. The candidate fungus that define regulatory genes which control nitrate reductase
should be grown on contrasting media containing as nitro- are also obtained in the same procedure (Table 3). When
gen sources 25 mM ammonium or glutamine (repression), 5 plated on solid medium containing a derepressing nitrogen
mM xanthine or proline (derepression), 5 mM nitrate (in- source plus chlorate, wild-type cells express nitrate reduc-
duction), and 25 mM ammonium or glutamine plus 5 mM tase and are killed, presumably due to conversion of chlo-
nitrate (repression and induction). Alternatively, the or- rate to chlorite by the enzyme. Mutants lacking nitrate re-
ganism can be grown on a single medium and then divided ductase are chlorate resistant and survive and grow in this
into aliquots which are incubated for 2 to 4 h on the vari- medium, thus providing a powerful selective system for ni-
ous contrasting media before the cells are harvested for trate reductase-negative mutants. Since such mutants can-
assay. The fungal cells are collected and homogenized in a not grow with nitrate as a sole nitrogen source, revertants
small volume of buffer using any of several convenient and suppressors can easily be obtained by plating the mu-
methods, one of the simplest being to grind the mycelia to tant cells on nitrate medium. This system has been utilized
a milky-like paste in an ice-cold mortar with acid-washed to provide selectable markers for introduction of recombi-
sand and a small amount of buffer (see chapter 44). The ho- nant DNAs into various fungal species (110).
mogenate is centrifuged to remove cell debris, and the su- The expression of the structural gene for nitrate reduc-
pernatant fluid is used for assay. Another popular method tase can also be examined by Northern blot analysis of
for making protein extracts utilizes a Mini-BeadBeater RNA isolated from wild-type and mutant cells grown under
(Biospec Products, Bartlesville, OK). This instrument holds contrasting conditions. In Neurosporu, upon induction ni-
2-ml tubes, each of which is filled one-third to one-half full trate reductase mRNA is synthesized very rapidly and
with zirconia/silica 0.5-mm beads, to which is added ap- reaches a steady-state level within 15 min (84). Turnover of
proximately 0.3 g of a fungal cell pad plus sufficient extrac- nitrate reductase message also occurs quickly upon repres-
tion buffer to fill the tube. The tubes are pulsed at maxi- sion, with a half-life of approximately 5 min (84).
mum speed for 1 min and then put on ice for 4 min; this is
repeated four times. The tubes are then centrifuged at 43.2.3. Other Nitrogen Pathways
14,000 rpm in a microfuge, and the supernatant is used for The filamentous fungi are extremely versatile in their abil-
enzyme assays. This procedure allows rapid preparation of ity to use a wide range of metabolites as nitrogen sources.
multiple samples. These include nitrate, purines, urea, amino acids, peptides,
Nitrate reductase activity is assayed in a mixture con- amides, and extracellular proteins (45,49, 50, 102, 115). In
taining 20 mM phosphate buffer (pH 7.75), 20 mM the area of amino acid metabolism, assay of transport activ-
NaNO?, 5 mM sodium sulfite, 100 pM FAD, and 0 to 0.2 ity with radioactively labeled basic, acidic, neutral, and aro-
ml of the enzyme extract (38). The reaction is carried out matic amino acids is easily carried out (115). L-Amino acid
in a total volume of 0.5ml and is started by adding NADPH oxidase is an enzyme involved in amino acid metabolism
to a final concentration of 200 kM. The samples are incu- that is readily assayed. The candidate fungus should be
bated at 25C for 0 to 30 min. Zero-time blanks should be grown or incubated in a medium to induce the enzyme, e.g.,
included. The reaction is stopped by addition of 1% sul- using phenylalanine as the sole nitrogen source. A useful
fanilamide in 20% (vol/vol) HC1. After mixing well, add control to include is a parallel culture grown or incubated
0.5 ml of 0.129% (wt/vol) naphthylethylene-diamine- with ammonium, which would be expected to repress the
diHC1, then add 1.5 ml of distilled water, mix well, and de- activity. A cell-free homogenate is prepared in phosphate
termine the absorbance of samples at 540 nm. Subtract buffer, pH 6, as described above and dialyzed overnight
background for each sample from an appropriate zero-time against the extraction buffer. The assay mixture contains
blank. The samples can be centrifuged before their ab- 0.5 ml of 0.2 M phosphate buffer, pH 7.5, with 2 mM L-
sorbance is taken if they appear cloudy. A standard curve leucine plus 10 ~1 of catalase to which is added 0.0 to 0.5
can be constructed with freshly prepared sodium nitrite. ml of the crude enzyme plus sufficient water to obtain a
One of the reasons that nitrate reductase is an attractive total volume of 1 ml. Individual samples can be incubated
enzyme in the study of nitrogen metabolism is that a two- for 0, 30, and 60 min at 37C; the reaction is stopped by
way selection system is highly efficient to obtain mutants adding 1 ml of 0.1% 2,4-dinitrophenylhydrazinein 2 M
and revertants in its structural gene and in multiple cofac- HC1. After mixing and standing for 5 min, 2 ml of absolute
tor genes required for the synthesis and assembly of Mo-Co, ethanol and 5 ml of 2.5 M NaOH are added. The tubes are
the molybdopterin cofactor, which is also a component of mixed well and allowed to stand for 5 min, centrifuging if
43. Physiology and Molecular Aspects of Fungi 955

TABLE 3 A. nidulans and N.crassa genes which function in nitrate assimilation


Genetic locus Growth of mutant upona
Activity
Aspergillus Neurospora NH4+ Nitrate Nitrite Xanthine
Encodes nitrate transporter crnA nit- 10 + +I-
Encodes nitrate reductase niaD nit-3 + +
Encodes nitrite reductase niiA nit-6 + -

Gene for synthesis or assembly cnxABC nit-1 + +


of a molybdenum cofactor
cnxE nit-7 + +
CnXF nit-8 + +
cnxG nit-9 + +
CnXH + +
Pathway-specificcontrol gene nirA nit4 + +
(mediates induction)
Global nitrogen regulatory gene areA nit-2 + -

(activator during N derepression)


Negative-acting regulatory gene nmrA nmr + +
(mediates nitrogen repression)
Wild-type strains grown on all of these nitrogen sources. Wild-type and all mutant strains grow using glutamine as the sole nitrogen source. Nitrate transporter mu-
tants (crnA and nit-10 mutants) cannot grow with low concentrations of nitrate.

necessary to clarify, when the absorbance at 550 nm is de- expressed at a very low abundance in a constitutive fashion.
termined. A n alternative procedure involves grinding cells Assays are available for various purine catabolic enzymes,
in approximately 20 volumes of buffer in an omnimixer, e.g., uricase, allantoinase, and allantoicase, but a favorite
precipitating the enzyme by adding ammonium sulfate to enzyme to follow in this pathway appears to be xanthine
85% saturation, and then dialysis. The dialyzed sample is dehydrogenase, whose activity is easily determined using a
used for assay. A standard curve can be derived using pyru- fluorometric assay (40). The reaction is carried out in a cu-
vic acid. vette with 1 ml of 0.1 M phosphate buffer, pH 8, contain-
Fungi secrete proteases into the extracellular medium ing 10 nmol of 2-amino-4-hydroxypteridine, 200 nmol of
under conditions when their growth depends upon the use NAD+, and 50 p l of a fungal crude extract. The excitation
of an exogenous protein as a source of nitrogen, sulfur, or wavelength is set at 326 nm, and the emission wavelength
carbon (45,48).A mixture of proteases may be present and is set at 410 nm. Xanthine dehydrogenase reduces the sub-
their relative abundance may depend upon the pH of the strate, 2-amino-4-hydroxypteridineto isoxanthopterin,
growth medium; e.g., acidic proteases are expected to pre- which is fluorescent (40), and enzyme activity is readily fol-
dominate when fungi are grown in medium at pH 5, lowed as an increase in fluorescence. Expression of each of
whereas alkaline proteases will be the major species when the genes encoding purine catabolic enzymes can also be
the growth medium is buffered at a more alkaline pH (4). A directly determined using Northern blot analysis of total
simple sensitive protease assay is achieved by incubating a RNA isolated from fungal cells grown under contrasting
1-ml sample of the cell-free growth medium with 1-ml of repressing versus inducing conditions. Additional enzyme
2% casein for various times, after which 2 ml of 5% activities are of interest, several important ones being glu-
trichloroacetic acid is added; this stops the reaction and tamate dehydrogenase (22), glutamine synthetase, and var-
precipitates any undigested protein, which is removed by ious transaminases, as well as specific activities such as pro-
centrifugation. The soluble products released by protease line oxidase and phenylalanine hydroxylase, but none of
activity are determined with 1 ml of the supernatant using these can be described here.
the Folin phenol reagent as described by Lowry et al. (68),
recording the absorbance at 750 nm. By the judicious use of
specific inhibitors, e.g., phenylmethylsulfonyl fluoride to in- 43.3. CARBON METABOLISM
hibit alkaline serine protease activity, and determining the
activity at different pH values, the contribution of distinct 43.3.1. Carbon Sources and Catabolite Repression
proteases can be ascertained. The filamentous fungi are capable of utilizing a variety of
Purines serve as relatively good nitrogen sources for the compounds as sources of carbon and energy. These include
filamentous fungi. In Aspergillus nidulans, synthesis of the monosaccharides such as arabinose, glucose, fructose, galac-
various purine catabolic enzymes requires nitrogen dere- tose, and mannose; disaccharides such as sucrose, trehalose,
pression and induction and is subject to precise genetic con- lactose, and maltose; and polysaccharides, sorbitol, mannitol,
trol. A pathway-specific regulatory gene, uaY, mediates in- ethanol, and quinic acid, to name some of the more com-
duction of at least eight unlinked structural genes which monly used carbon sources. Carbon catabolite repression
encode permeases and enzymes involve in purine transport refers to a mechanism that allows certain carbon sources,
and degradation. The UAY protein contains a GAL4-like e.g., glucose, to be used preferentially to less readily utilized
Cys6/Zn2 binuclear DNA binding domain and recognizes compounds. It is well known that in Escherichia coli, when
promoter elements with the sequence TCGG-N6-CCGA glucose is limiting, cyclic AMP serves to signal a lifting of
(102). The uaY gene itself is not subject to regulation but is catabolite repression, but its involvement, if any, in fungal
956 rn MYCOLOGY

regulation is unclear. Pioneering research in carbon metab- sources, including glucose, which should strongly repress in-
olism with filamentous fungi has been carried out primarily vertase synthesis. In Neurospora, sucrose is not transported
with Aspergillus nidulans (28). A hierarchy among carbon into the cells but must be hydrolyzed extracellularly, after
sources for Aspergillus has been determined based upon their which the fungus can import the monosaccharide products.
strength of repressors (6). Detailed studies of the genetic Thus, in Neurospora invertase is largely an extracellular en-
regulation of enzymes subject to specific induction in addi- zyme and can be assayed using a suspension of conidia or
tion to carbon catabolite repression have been quite in- young mycelia without a need to prepare a homogenate.
structive. These studies have been conducted at several lev- Although a number of assays for invertase are available, a
els, including growth, enzyme activity, and RNA content. simple colorimeteric assay which depends upon the release
Two of the most noteworthy systems are the alc genes, of reducing sugars is one of the most convenient. The cells
which control alcohol dehydrogenase activity (ADH) and or enzyme extract is incubated at 37C in 0.05 mM acetate
ethanol utilization in A. nidulans, and the quinate (qa) uti- or succinate buffer, pH 5.5, with 0.5 mM sucrose in a total
lization cluster of genes in N. crussa and in A. nidulans (28). volume of 1.5 ml for various times, and then the reaction is
Carbon catabolite repression in Aspergillus is mediated by stopped by placing in a boiling water bath for 1 min. After
CREA, a DNA binding protein with two CysZ/HisZ type cooling and centrifugation, if necessary, to clarify, to 1 ml of
zinc fingers that acts negatively to prevent gene expression the mixture add 2 ml of DNS reagent, mix, and heat the
(28,57). During conditions of repression, CREA apparently capped tubes in a boiling water bath for 5 min. The tubes
competes with positive activators by binding at elements are cooled quickly in an ice bucket and then allowed to
that overlap the elements for the positive factors, thereby reach room temperature, and then the absorbance at 560
preventing the latter's function. nm is determined. In some cases it will be more convenient
ADH and its regulation have been extensively studied in to stop the reaction by adding 3 ml of the DNS reagent, fol-
Aspergillus. A cluster of closely linked genes includes, in lowed by the heat step. The DNS reagent is prepared as fol-
order, alcR, alcX, &A, alcM, and aldA (28, 30, 57). lows: 10 g of dinitrosalicylic acid, 400 ml of 1 M NaOH, and
Expression of &A, which encodes ADH, and of aldA, 300 g of KNaC4H4064H20are mixed and diluted to I
which encodes aldehyde dehydrogenase, requires induction liter. Trehalose is also a nonreducing sugar, and thus treha-
by ethanol and is mediated by ALCR, a positive-acting lase activity can be assayed in a similar fashion, using tre-
transactivator protein that contains a Cys6/Zn2 DNA bind- halose as the substrate. Chromogenic substrates such as 0-
ing motif (31,56,57).Induction by ALCR of both alcA and nitrophenyl-a-glucoside, 0-nitrophenyl-P-galactoside, and
aldA is inhibited by CREA in the presence of repressing 0-nitrophenyl-P-N-acetylglucosamineare useful to follow
amounts of glucose. specific hydrolase activities of enzymes, e.g., P-galactosi-
dase, that are subject to carbon catabolite repression. ADH
43.3.2. Complex Regulatory Systems activity is determined by following the increase in ab-
Certain compounds can serve as both a carbon source and a sorbance at 340 nm, which reflects the reduction of NAD+
nitrogen source for Aspergillus, and their utilization is highly to NADH. Cell extracts derived from fungal cells that have
regulated by both carbon and nitrogen catabolite repression been induced with ethanol should be compared with ex-
in addition to specific induction (28). Proline serves as both tracts from cells grown on repressing medium with glucose
a nitrogen and carbon source for Aspergillus, and its utiliza- as the carbon source. The reaction is carried out in a total
tion involves a cluster of five closely linked genes, arranged volume of 3 ml in a cuvette at 25"C, and the reaction mix-
in the order p r A , -X,-D, -B, and -C(100, 101). The prnA ture contains 15 mM pyrophosphate buffer (pH 8.8), 0.3 M
gene codes for a positive-acting regulatory protein which ethanol, and 7.8 mM NAD+. The enzyme extract (0 to 0.2
mediates proline induction; pmD, -B, and -C encode the ml) is added at zero time and the absorbance at 340 nm is
proline catabolic enzymes and permeases, proline oxidase, recorded at 15-s intervals.
proline permease, and pyrroline-5-carboxylate dehydroge-
nase, respectively. Expression of the pm genes requires a
functional PRNA protein, induction with proline, and a lift- 43.4. SULFUR METABOLISM
ing of either carbon or nitrogen catabolite repression. Nitro-
gen derepression is signaled by the AREA protein, whereas 43.4.1. The Sulfur Regulatory Circuit
the CREA protein mediates carbon catabolite repression, as Sulfur is an essential nutrient and is required for the syn-
described above. In Neurospora and Aspergillus, an extracel- thesis of cysteine and methionine plus many other sulfur-
Mar protein can serve as the sole source of carbon, nitrogen, containing compounds. Thus, it is not surprising that the
or sulfur and a single alkaline protease is synthesized and se- fungi possess a sophisticated genetic system to ensure that a
creted in response to a limitation for any of these required steady supply of sulfur can be assimilated from a wide vari-
elements (45). Acetamide also serves as both a carbon and ety of possible environmental sources. Methionine and in-
nitrogen source for Aspergillus. The genetic regulation of ac- organic sulfate are preferred sulfur sources, and both
etamidase and several related enzymes has been the subject strongly repress the expression of multiple genes that en-
of elegant research by Hynes and Davis (50). Control of ex- code enzymes necessary for use of various secondary sulfur
pression of the a d s gene, which encodes acetamidase, is sources, e.g., tyrosine-0-sulfate and choline-0-sulfate (7 1).
very complex and involves the positive action of AREA and During sulfur derepression conditions, which occur when fa-
the negative signaling of CREA plus interactions with a vored sulfur sources are missing or present in small amounts,
number of pathway-specific effectors (50, 78). the genes encoding an entire set of sulfur catabolic enzymes
are activated (71). Unlike the situation with the nitrogen
43.3.3. Enzyme Assays and carbon metabolic circuits, induction by substrates is not
The utilization of sucrose requires its hydrolysis by inver- required and derepression alone leads to expression of the
tase, whose synthesis is subject to catabolite repression ex- sulfur-related structural genes. Activation of this set of
erted by glucose. Invertase activity is determined with fun- structural genes in Neurospora crussa is mediated by CYS3,
gal cells that have been grown with different carbon a positive-acting DNA binding protein with a basic leucine
43. Physiology and Molecular Aspects of Fungi I 957

zipper (bzip) motif (36, 37, 89). The CYS3 protein binds an excellent source of phosphorus, but when it is not avail-
specifically at elements in the promoter region serving the able the fungi can utilize a variety of secondary phosphorus
various downstream structural genes (63). The cys-3 gene sources, including nucleic acids and various phosphate
itself is controlled by positive autogenous regulation, and esters. In Neurosporu, in which regulation of phosphorus
both the cys-3 mRNA and the CYS3 protein are subject to metabolism has been extensively studied, the synthesis of a
relatively rapid turnover under sulfur-repressing conditions number of enzymes involved in phosphorus metabolism is
(104). Additional sulfur regulatory factors, SCONl and repressed as a group by high levels of inorganic phosphate.
SCON2, appear to act by controlling the synthesis, activity, These activities include a high-affinity phosphate perme-
or stability of CYS3 (58, 88). Recently, Sizemore and ase, 0-phosphorylethanolamine permease, an alkaline
Paietta (99) have characterized a new sulfur regulatory fac- phosphatase, an acid phosphatase, nucleotidases, and one
tor, SCON3, a Skp-1 homolog which occurs in a complex or more nucleases (47, 62, 67, 81). Three distinct, interre-
with SCON2. Skp-1 is an important component of ubiqui- lated regulatory genes, nuc-1 , nuc-2, and preg, control the
tin conjugating systems in S. cerevisiue, suggesting an im- synthesis of this entire family of phosphorus catabolic en-
portant role for SCON3 in controlling turnover of CYS3. zymes (61, 66). The regulatory factors act sequentially,
such that when phosphorus is limiting, NUC2 protein in-
43.4.2. Enzymes of Sulfur Metabolism hibits the expression of preg, whose product in turn inhibits
In Neurospora, the uptake of inorganic sulfate is highly reg- the expression of nuc-1 . The N U C l protein, which is ex-
ulated (51, 54, 71). The unlinked genes which encode two pressed only during phosphorus limitation conditions, is a
distinct sulfate permease species, cys-13 and cys-14, are both positive-acting DNA binding protein that activates the
controlled as members of the sulfur regulatory circuit, and expression of the entire set of phosphorus-related genes
their expression requires activation by CYS3 and only oc- (53).
curs during conditions of sulfur derepression. Sulfate trans-
port assays are easily conducted with 35S-labeled sulfate 43.5.2. Phosphorus-Related Permeases and
using cells that have been grown with derepressing condi- Enzymes
tions, e.g., with 0.25 mM methionine as the sole sulfur Permeases for phosphate transport can be assayed using 32P*
source, versus cells growing with 5 mM methionine, which labeled phosphate with cells which have been grown with
completely represses the uptake systems (69). Assays can be high repressing levels of inorganic phosphate in comparison
carried out with young mycelia or with conidia. Other fungi with cells grown with derepressing levels of phosphate or a
are expected to have active transport systems for the uptake poorly utilized phosphorus source (10, 67). Alkaline phos-
of sulfate. The cells are incubated in medium or buffer with phatase and acid phosphatase are convenient enzymes to
35S-labeledsulfate for various times, and then the cells are examine with respect to phosphorus regulation, and their
collected on a filter and washed several times with a solu- activities can be determined in cells grown under repressing
tion of nonradioactive sulfate prior to determining the and derepressing conditions, as well as with cells with mu-
amount of radioactivity by scintillation counting. Nitrate tations in regulatory or structural genes. Convenient assays
strongly inhibits sulfate transport and should not be present for phosphatases utilize the release of p-nitrophenol from p-
in the assay mixture. nitrophenyl phosphate (61, 81). To assay alkaline phos-
Aryl sulfatase, an enzyme which hydrolyzes aromatic sul- phatase, various amounts (0 to 0.2 ml) of a cell extract are
fate esters, is subject to a high degree of regulation and is incubated with 3 ml of 1 M Tris buffer, pH 8, containing 1
simple to assay, and thus, it is a valuable activity to exam- mM p-nitrophenyl phosphate. The enzyme reaction can be
ine and can be used to monitor repression/derepression con- followed directly by determining the absorbance at 410 nm
ditions of fungal cells (72, 76). Fungal cells which have at various time intervals. Alternatively, the assay can be
been grown under contrasting conditions of repression and conducted in a 1-ml total volume; individual reaction mix-
derepression are homogenized using acid-washed sand in tures are incubated for various times (0 to 30 min), and
0.05 M Tris-HC1 buffer, and the clear supernatant obtained then 2 ml of 0.5 M NaOH in 90% ethanol is added and,
after centrifugation is used as the crude enzyme. Various after centrifugation if necessary to clarify, the absorbance at
amounts (0.05 to 0.2 ml) of a cell extract are incubated with 410 nm is determined. Acid phosphatase activity can be as-
0.3 M Tris-HC1 buffer, pH 8.1, and 6 mM p-nitrophenyl- sayed similarly in 0.1 M sodium acetate buffer, pH 5, and 1
sulfate in a total volume of 1 ml at 37C for various times mM p-nitrophenyl phosphate. In this case, it is necessary to
(0 to 60 min), after which 2 ml of 0.5 M NaOH in 90% stop the reaction with NaOH in ethanol before determin-
ethanol is added and, after centrifugation if necessary to ing the absorbance at 410 nm. Some fungi may secrete
clarify, the absorbance at 405 nm is determined. If the sub- phosphatases into the extracellular growth medium under
strate has a significant yellow color due to the presence of phosphorus derepression conditions, and these activities
free p-nitrophenol, it can be treated with norite and filtered can be assayed in the spent growth medium after the cells
before use. Paietta isolated the Neurospuru structural gene are removed by filtration or centrifugation.
encoding arylsulfatase and demonstrated that it is strongly
regulated at transcription (87).
43.6. pH CONTROL OF EXTRACELLULAR
ENZYMES AND PERMEASES
43.5. PHOSPHORUS METABOLISM Pioneering research carried out by Arst and his colleagues
with Aspergillus nidulans has revealed that fungi contain a
43.5.1. Genetic Control of Phosphorus Metabolism remarkable regulatory system that controls the synthesis of
Phosphorus is another element that is an important com- specific extracellular enzymes and permeases depending
ponent of nucleic acids, coenzymes, and many other bio- upon the pH of the growth medium (4). Thus, under acidic
logical molecules. The fungi possess complex regulatory conditions, an extracellular acid phosphatase and other en-
mechanisms to maintain a sufficient intracellular pool of zymes and permeases with acidic pH optima are synthesized
phosphate for continuous growth. Inorganic phosphate is and secreted. In sharp contrast, an alkaline phosphatase and
958 MYCOLOGY

other activities with alkaline pH optima are synthesized and concentrations, an equal volume of a solution of 5 mM
secreted when the external pH is basic (4). Expression of a Fe(C104)3-0.1 M HClO, is added to the cell-free super-
fungal penicillin biosynthetic gene is strongly influenced by natant. After mixing, the absorbance at 495 nm is deter-
pH (26). The regulatory mechanism involves a series of six mined, using fresh medium for a background reading.
pal genes which appear to sense the ambient pH and con- ~-ornithine-N~-oxygenase activity is assayed with fungal
trol the activity of PacC, a DNA binding protein (4, 25, cell homogenates prepared in potassium phosphate buffer,
79). The full-length PacC protein, which predominates pH 8, after centrifugation to remove cell debris. A total of
during acid pH conditions, is not functional, and genes ex- 0.4 ml of a cell extract is added to an assay mixture consist-
pressed under acidic conditions are actively expressed, but ing of 0.1 mM phosphate buffer (pH 8), 1.5 mM L-omithine,
those expressed under alkaline conditions are silent (106). 0.5 mM NADPH, and 5 pM FAD in a total volume of 1 ml,
Under alkaline pH conditions, the PacC protein is specifi- and the samples are incubated for various times from 0 to
cally proteolyzed to yield its active form (86). A t alkaline 2 h. The reaction is stopped by adding 0.5 ml of 0.2 M per-
ambient pH, PacC activates expression of genes expressed chloric acid. After centrifugation, 1 ml of the supernatant
under alkaline conditions and also inhibits transcription of fluid is subjected to the iodine oxidation test (108) by adding
those expressed under acidic conditions (25, 106). It can be 4 ml of 5% sodium acetate and 0.5 ml of 1% sulfanilic acid in
expected that most fungi have a similar pH control mecha- 25% acetic acid and 0.2 ml of 1.3% iodine in glacial acetic
nism, which implies that the expression of various extracel- acid. After 5 min at room temperature, 0.2 ml of 0.1 M
lular enzymes may be strongly dependent upon the ambient sodium thiosulfate and 0.1 ml of 0.6% a-naphthylamine in
pH. A PacC homolog appears in the annotated Neurospora 30% HC1 are added and the samples are held at room tem-
genome sequence, suggesting the presence of a similar sys- perature for 30 min. After an additional centrifugation, if
tem for pH control. needed to remove any denatured protein, the absorbance at
520 nm is recorded.

43.7. ACQUISITION OF IRON


43.8. RNA AND PROTEIN SYNTHESIS
43.7.1. Mechanisms for Iron Acquisition In studies of metabolism and gene expression, it is often nec-
In addition to the obvious requirement for substantial essary to examine RNA and protein synthesis. Moreover,
amounts of carbon, nitrogen, sulfur, and phosphorus, the the ability to specifically inhibit either RNA synthesis or
fungi and other organisms need smaller amounts of numer- protein synthesis helps to determine whether the appear-
ous other metabolites, including MgZ+,K+, CaZ+,Na+, ance of a new activity or a significant increase in an activ-
ZnZ+,Cuz+,Moo4-, and Fe2+.Fungi possess specific mech- ity requires transcription and/or translation. Neither acti-
anisms to acquire sufficient levels of such ions for optimal nomycin D nor oc-amanitin inhibits RNA synthesis in
growth. Iron is needed to complete synthesis of a number Aspergillus or in Neurospora, and both probably are ineffec-
of enzymes and coenzymes, most notably the cytochromes. tive in most fungi. Proflavin has been used with some suc-
Pioneering work by Leong and her colleagues with Ustilago cess to block transcription in fungi (19, 105). Proflavin at a
has contributed much to our understanding of iron home- final concentration of 100 pg/ml completely inhibited the
ostasis in fungi (2, 3 , 73, 111).Fe2+ is present in the envi- induction of nitrate reductase in Neurospora (105). To en-
ronment at extremely low concentrations due to its very sure that the block imposed occurs at the intended level, it
limited solubility. Bacteria and fungi secrete low-molecular- is desirable to show that proflavin, or any potential in-
weight compounds known as siderophores which chelate hibitor, actually inhibits RNA synthesis and does not act at
Fez+ for transport into the cells. O n the other hand, too an unexpected level, e.g., protein synthesis. In Aspergillus
great an intracellular level of Fe2+ is extremely toxic due and Neurospora, RNA synthesis can be examined with the
to its role in the oxidation of macromolecules, membrane use of 3H- or 14C-labeleduridine, preferably with uridine la-
lipids, proteins, and DNA. Thus, the fungi have evolved a beled with tritium in the 6 position. Uridine will be incor-
system for genetic control of siderophore synthesis to ensure porated primarily into RNA, although a small amount may
that intracellular Fez+ is maintained within a safe concen- label DNA due to metabolic conversions of uridine to other
tration range. When the level of Fe2+available is very low, pyrimidines. PHIuridine is added to a final concentration of
the enzymes in the pathway responsible for siderophore s n- 0.1 mM to a fungal culture, either one newly inoculated
thesis are expressed, but under conditions of high Fe ,
I+ with conidia or one with young rapidly growing mycelia.
siderophore synthesis is strongly repressed, thereby preclud- After various times, homogeneous samples are removed,
ing additional uptake of iron. When iron is present at re- collected on filter papers, and washed with at least five
pressing levels, a negative-acting GATA factor-URBS 1 in aliquots of ice-cold 10% trichloroacetic acid, with a final
Ustilago and its homologs, SRE in Neurospora and SREP in wash with distilled water. The filters are dried and radioac-
Penicillium-which contains two zinc finger motifs represses tivity is determined by scintillation counting.
the expression of ~-ornithine-N'-oxygenase,the first en- The increase in enzyme activity which occurs upon ad-
zyme unique to siderophore formation(44, 111, 116). When dition of an inducer or lifting of catabolic repression often
Fez+ is available only in limited amounts, this enzyme and requires de novo protein synthesis, rather than simply acti-
others of the synthetic pathway are expressed and vating a preexisting, inactive protein. Cycloheximide at a
siderophores are made and secreted into the extracellular final concentration of 0.1 mM is very effective at inhibiting
medium. cytoplasmic protein synthesis in at least the majority of
fungi and can be used to support the concept that expres-
43.7.2. Assays sion of a specific enzyme is dependent upon de novo protein
After the candidate fungus has been grown in an iron- synthesis. To ensure that cycloheximide is inhibiting ro-
depleted liquid medium (low iron) and a parallel culture tein synthesis, incorporation of [3H] leucine or of [ S]- R
with a repressing level (0.01 mM FeS04) of iron, the cells methionine can be examined. After a culture has been in-
are removed by centrifugation. To determine siderophore cubated in the presence of the radioactive amino acid for
43. Physiology and Molecular Aspects of Fungi 959

various times, homogeneous samples are collected on filter lysis buffer (50mM sodium acetate buffer [pH 5.31, 10 mM
paper, washed with trichloracetic acid, and processed as de- EDTA, 1% SDS) using a ratio of 1 g/5 ml. A n equal volume
scribed above. In the case of Neurosporu, 0.1 mM cyclohex- of acidic phenol-chloroform-equilibrated with the lysis
imide nearly completely inhibits any incorporation of buffer (pH 5.3), omitting SDS, at 65C-is added to the
amino acids. Use of a mutant strain that requires leucine (or lysate, and the mixture is incubated at 65C for 30 min with
methionine) can improve the incorporation of the radioac- shaking. After centrifugation, the aqueous phase is recov-
tive precursor. ered, and this extraction process is repeated several addi-
tional times until no protein interface is visible. KNA is
then precipitated by adding 0.6 vol of isopropyl alcohol and
43.9. ISOLATION OF NUCLEIC ACIDS recovered after centrifuging. The RNA pellet is dissolved in
A number of different methods have been described to iso- a small volume of DEP-treated water or buffer and can be
late DNA from filamentous fungi, all of which are useful used for Northern blot analysis, reverse transcription-PCR,
(59, 75, 82, 83, 97). A method devised by Metzenberg and in vitro translation, or preparation of cDNA libraries.
Baisch works very well and uses the chaotropic agent Various alternative methods for RNA isolation are avail-
ethanolic perchlorate, which is easier (and safer) to utilize able, including one which employs guanidinium isothio-
than phenol (75). Schechtman (97) combined steps from cyanate and was originally devised to obtain RNA from
several different procedures to develop a simple protocol mammalian tissues rich in RNase (16) and another which
that yields 200 to 300 pg of pure DNA of greater than 50 uses aurintricarboxylic acid, which irreversibly and tightly
kb in length from a single 40-ml culture. A t least a dozen binds KNA (65), have proved successful for isolation of fun-
samples can be processed in 1 day, and the procedure can gal RNA. Poly(A) RNA can be isolated from total RNA by
readily be scaled up. The mycelial pad harvested from a 40- use of oligo(dT) cellulose chromatography.
ml culture is washed with water and ground to a fine pow-
der in a prechilled mortar with liquid nitrogen. Transfer the
powder to a 50-ml polypropylene tube, add 15 ml of 50 mM 43.10. IDENTIFICATION AND CLONING
EDTA (pH 8.5)-0.2% sodium dodecyl sulfate (SDS)-15 p l OF FUNGAL GENES
of diethylpyrocarbonate (DEP), and after extracting by vig- Many hundreds of fungal genes, including structural, regu-
orous shaking, incubate the sample at 70C for 15 min, latory, rRNA, and tRNA genes, have now been isolated and
then chill on ice for at least 10 min. Add 0.95 ml of potas- characterized, leading to a revolution in our understanding
sium acetate (pH 4.3) and incubate on ice for 1 h, then cen- of molecular, genetic, and cellular processes. A number of
trifuge at 4C for 15 min. Then add 15 ml of isopropanol to fungal genes have been isolated by approaches in wide-
the supernatant with gentle mixing, which will immedi- spread use, e.g., differential screening of cDNA libraries
ately precipitate DNA. Rinse the DNA pellet with 70% (27), using heterologous probes, PCR approaches, or chro-
ethanol, drain, and air dry, then resuspend in 4 ml of 1 mM mosome walking. The urn gene of Neurosporu CTUSSU was iso-
EDTA (pH 8.0). Add 2 ml of high-salt buffer plus 15 p1of lated using a synthetic DNA probe based upon the amino
10 mg/ml RNase A (pre-boiled), and incubate the sample at acid sequence of the encoded product, glutamate dehydro-
37C for 30 min. Add 180 p l of 0.1 M spermidine-HC1, genase (55). Paietta employed chromosome walking to iso-
mix, incubate on ice for 20 min, and pipette off the liquid late the urs gene, which encodes arylsulfatase (87).
from the clotted DNA. Rinse the pellet three times for 30 However, in many cases, fungal genes can be isolated using
min with 2 ml of 75% ethanol-10 mM Mg acetate-0.3 M more specialized techniques that are applicable to fungi.
sodium acetate (pH 6.0), pipetting off the rinse each time, The wealth of mutants of A. nidulans, N. crassu, and
then rinse with 70% ethanol and drain and air dry the other fungi makes possible gene isolation by complementa-
pellet. Resuspend the pellet overnight in 1 ml of 10 mM tion. However, the recovery of a cloned gene following
Tris-HC1 (pH 8.0)-1 mM EDTA-0.1 M NaCl. After this, complementation of a given mutant by simply recovering a
reprecipitate the DNA with 2 ml of ethanol at room tem- recombinant plasmid is usually not possible because of the
perature. Pipette off the supernatant, rinse the pellet again lack of suitable autonomously replicating vectors. Trans-
with 70% ethanol, air dry, and dissolve in 1 ml of TE (10 forming DNA usually integrates into random chromosomal
mM Tris-HC1, 1 mM EDTA [pH 8.01). The DNA obtained locations in filamentous fungi, making recovery of the com-
is of high molecular weight and is readily cut by all restric- plementing DNA problematic. One approach which has
tion enzymes tested. It can be used for cloning, Southern been employed is marker rescue, in which genomic DNA of
blotting, DNA footprinting, gel shift assays, in vitro tran- the transformed strain is cut with a restriction enzyme and
scription, and other procedures. the fragments are ligated to form circular molecules; this
Methods are available to make minipreps of DNAs from complex mixture is then used to transform E. coli to recover
multiple fungal samples (59, 83). the vector which carries the desired gene. Marker rescue
The isolation of high-quality RNA from any organism, was used to obtain the p y G gene of Aspergillus nidulans
and particularly from fungi, requires vigilance to prevent its (82). Akins and Lambowitz (1) devised a more generally ap-
degradation by ever-present nucleases. Reagents and glass- plicable technique, known as sib selection, to isolate genes
ware for RNA preparation should be kept separate from without a requirement for any knowledge of the nature of
general laboratory supplies, and glassware should be baked the gene product. In sib selection, a gene is isolated from a
overnight at 250C prior to each use. Buffers should be genomic library by successive rounds of transformation of a
treated with 0.1% DEP overnight and then autoclaved. A mutant fungal strain with pools of plasmid or cosmid DNAs
number of useful methods for RNA isolation from filamen- which are progressively reduced in complexity until a single
tous fungi have been devised, only one of which will be candidate clone is obtained. In the case of Neurosporu, a
described here. Total RNA can be isolated with a modifi- cosmid with a dominant selectable marker for benomyl re-
cation of the procedure described by Weaver et al. (113). sistance was used to generate libraries which contain large
Mycelia are ground to a fine powder with a mortar and pes- DNA inserts. Individual cosmid clones are ordered in the
tle in the presence of liquid nitrogen and then suspended in wells of microtiter dishes, allowing rapid and efficient
960 MYCOLOGY

screening of libraries. This approach has been responsible agar (2% sorbose, 0.05% glucose, 0.05% fructose, Vogel's
for the isolation of many different genes which were identi- salts, and 1.5% agar plus an appropriate selective agent, e.g.,
fied by complementation. A n important virtue of sib selec- benomyl [5 pg/ml] or lacking an essential metabolite, e.g.,
tion is that it allows the isolation of genes identified only histidine). The plates are incubated at the desired tempera-
by mutation, including those that affect complex cellular ture until transformed colonies appear.
processes for which nothing is known of the nature of the Transformation can also be carried out by electropora-
gene product. Cosmid-based genomic libraries for Asper- tion, using a protocol devised by K. Borkovich. Conidia
gillus nidulans as well as lambda-based and YAC libraries are from 8- to 10-day-old cultures are harvested, washed, resus-
available from the Fungal Genetics Stock Center. The pended in 25 ml of ice-cold 1 M sorbitol, and pelleted in a
Aspergillus and Neurosporu genome sequences have revolu- clinical centrifuge. This procedure is repeated twice, after
tionized gene cloning in these fungi. Primers can be used to which the final pellet is resuspended in 0.5 ml of 1 M sor-
clone any gene of interest using PCR. In the case of bitol. The concentration of conidia is determined with a
Neurospora, the ends of all clones in the Orbach/Sachs cos- hemacytometer and adjusted to 2.5 X lo9 spores/ml; 40-pl
mid library have been sequenced, which has allowed the aliquots are added to microtubes on ice. One tube serves as
Whitehead annotation group to place genes within individ- a negative control with no added DNA; 1 to 2 kg of DNA
ual cosmids. It is far easier to clone from a cosmid DNA, is added to the experimental tube. After electroporation
available from the Fungal Genetics Stock Center, than from (e.g., with an Eppendorf model 25 10 electroporator), sam-
genomic DNA. Work is progressing to allow extensive tran- ples are plated on selective media as described for the
scriptional profiling using microarrays with both Aspergillus spheroplast transformation protocol.
and Neurosporu.
43.1 1.2. Selectable Markers
The structural gene which encodes nitrate reductase (niuD'
43.1 1. TRANSFORMATION WITH in Aspergillus; nit-3 in Neurosporu) can be used as a selective
EXOGENOUS DNA marker for many different filamentous fungi. Mutants lack-
ing this enzyme are readily selected via chlorate resistance.
43.1 1 .I. Transformation Procedures The Neurospora pyr-4' gene, which codes for orotidine-5'-
The ability to readily transform filamentous fungi with ex- phosphate decarboxylase, is frequently used to select for
ogenous DNA has been revolutionary and provided the transformants of Aspergillus nidulans using pyrG mutant
essential step required in cloning, characterization, and so- strains as the recipient (82). The his-3+ gene of Neurospora
phisticated manipulation of individual genes. Trans- has been used to target exogenous DNA to a precise ge-
formation now is a standard procedure with Aspergillus and nomic site as described below. Orbach et al. (85) developed
Neurosporu (85, 107, 112) and can be accomplished with a very useful dominant selectable marker, a mutant N.
many other fungi, e.g., Gibberella fujikuroi (109), Penicillium crassa P-tubulin gene that confers resistance to benomyl,
chrysogenum (13), and the rice pathogen Mugnuporthe griseu which makes it possible to transform virtually any wild-type
(23). A typical protocol used with Neurosporu involves or mutant Neurosporu strain. The bacterial hygromycin re-
treating germinated conidia suspended in 1 M sorbitol with sistance gene, hph, and the bleomycin resistance gene, ble,
filter-sterilized Novozyme (2.5-mg/ml final concentration) from transposon Tn5 both work well as selectable markers
for 30 to 60 min at 30C with gentle shaking in order to in Aspergillus and Neurosporu, and in other fungi.
produce spheroplasts. The extent of spheroplasting is It is frequently desired to introduce a manipulated gene
checked by placing a 5-pl sample onto a slide, adding a cov- for which no selective condition is available but a lack of
erslip, and then adding water. When a sufficient amount of suitable restriction sites may make it difficult to move it
the cell wall has been digested, the spheroplasts will swell into a vector with a proven selectable marker. In these
and burst. The Novozyme digestion should not be pro- cases, cotransformation with two vectors, one carrying the
longed because the viability of the spheroplasts will be gene being studied and the other with a convenient selec-
significantly reduced. The resulting spheroplasts can be used table marker, works quite well because a high percentage of
immediately or suspended in Tris buffer containing sorbitol, the transformants selected will contain both vectors. Fungal
calcium chloride, polyethylene glycol, and dimethyl sulfox- transformations can be carried out with linear DNA as well
ide and dispensed in aliquots into microtubes and frozen at as closed circular plasmid DNAs and even very large cosmid
-80C; they will remain competent for transformation for DNAs, 30 to 40kb in size.
at least several months (112). Preparation of competent
spheroplasts from some mutant strains may prove consider- 43.1 1.3. Nature of Transformed Fungal Strains
ably more difficult than with wild-type ones. Each batch of Most transformants of N. crassu, A. nidulans, and probably
spheroplasts must be tested for competence; in the case of most filamentous fungi have one or more copies of the
Neurospora, this is easily done using plasmid pBT6, which transforming DNA integrated into ectopic genomic sites,
carries the benomyl resistance gene (85). and only rarely will the exogenous DNA be incorporated at
For transformation, after thawing on ice, 100 p1 of com- the homologous site unless special procedures are employed
petent spheroplasts is incubated with 1 pg of DNA plus 5 (107). N. c r a m conidia and thus spheroplasts prepared
p l of heparin (5 mg/ml) in STC ( 1 M sorbitol, 50 mM Tris- from them are multinucleate, with an average of two or
HC1 [pH 8.01, 50 mM CaC12) on ice for 30 min. Then 1 ml three nuclei, but usually only one of the nuclei is actually
of PTC (40% PEG 4000,50 mM Tris-HC1 [pH 8.0],50 mM transformed by integration of one or more copies of the ex-
CaC12) is added and incubated for 20 min at room temper- ogenously supplied DNA. Thus, the transformed strains are
ature, after which samples are placed in 50-ml screw-cap heterokaryotic, which makes them unsuitable for analysis of
tubes to which 15 ml of molten but cooled top agar (2% sor- the function or regulation of the inserted gene of interest.
bose, 1 M sorbitol, 0.05% glucose, 0.05% fructose, Vogel's Homokaryons can be obtained by plating microconidia,
salts, and 2.8% agar) is added. After gentle mixing, the sam- which have a single nucleus, obtained from the hetero-
ple is poured onto petri plates containing 25 ml of bottom karyon. It should be noted that genetic crosses cannot be
43. Physiology and Molecular Aspects of Fungi 961

used to obtain homokaryons in Neurosporu because of re- (98). This occurs whether the duplicated copies are closely
peat-induced point mutations (RIP), as described briefly linked or even are situated on different chromosomes. The
below. multiple point mutations introduced by RIP can be so ex-
In some cases, although an exogenous gene has been suc- tensive that the DNA fragment cannot be recognized by
cessfully inserted into a fungal host, it may not be expressed, Southern blot hybridization when probed with the original
possibly because of DNA methylation or its incorporation wild-type sequence (29, 98). RIP can be used to obtain mu-
in a region of the genome which is not accessible for tran- tant strains in which the desired gene has been completely
scription. inactivated (29, 52, 98). A special technique, sheltered
In examining various manipulated forms of a cloned RIP, can be utilized when disruption of the target gene
gene for function via transformation of a fungal host strain, might be lethal (46). See chapter 44 for additional infor-
a serious consideration is the possibility of significant vari- mation concerning RIP.
ability in expression due to incorporation of the transform-
ing DNA at different genomic sites. To examine a series of
transformants, each of which possesses a different manipu- 43.12. OTHER METABOLIC PATHWAYS
lated version of a gene of interest, but all present at the Additional metabolic pathways and closely related subjects
same genomic location, the constructs can each be coupled beyond those discussed above represent important topics
with a truncated selectable marker which can function only and research areas, some of which are especially relevant to
upon homologous recombination with the hosts endoge- fungi. These include polyamine biosynthesis (21 ), pathways
nous mutated gene. Such a strategy has proved invaluable and regulatory mechanisms of amino acid biosynthesis (22,
with Neurosporu, in which selection for hist colonies iden- 96), compartmentation of metabolic reactions (8, 20, 22),
tifies transformants in which a truncated his-3 gene has light-regulated gene expression (15, 64, 103), mating type
been targeted to the his-3 locus, and the vector carrying this determination and sporulation (9, BO), heat shock proteins
truncated selectable marker includes the gene of interest, and stress responses (94), the biological clock and circadian
whose presence can be verified by Southern blotting. A rhythms (18, 24, 32, 60), and the virulence factors of plant
similar approach should be applicable to any fungus which and animal fungal pathogens (23). The fungi have many re-
can be transformed with exogenous DNA. markable characteristics and promise a wealth of exciting
new phenomena and discoveries as their biology and genet-
43.1 1.4. Constructing Gene Disruptions in Fungi ics are further explored.
Gene disruption, a strategy in which a specific gene is par-
tially or totally deleted or otherwise rendered completely
inactive, is one extremely valuable use of transformation.
43.13. REFERENCES
Gene disruptions, or knockouts, allow determination of 1. Akins, R. A,, and A. M. Lambowitz. 1985. General
the phenotype resulting from a complete loss of function of method for cloning Neurosporu crussu nuclear genes by
any cloned gene and also provide ideal host strains for complementation of mutants. Mol. Cell. Biol. 5:2272-
analysis of various manipulated forms of the gene being 2278.
investigated. The most direct procedure for disruption in- 2. An, Z. Q., B. G. Mei, W. M. Yuan, and S. A. Leong.
volves replacing all or a central region of the cloned gene 1997. The distal GATA sequences of the sidl promoter of
Ustilago maydis mediate iron repression of siderophore
with a dominant selectable marker, e.g., hygromycin resis- production and interact directly with Urbsl, a GATA fam-
tance, leaving long regions of 5 and 3 flanking DNA for ily transcription factor. EMBO J. 16:1742-1750.
homologous recombination. The construct is transformed 3. An, Z. Q., Q. Shao, J. McEvoy, W. M. Yuan, J. L.
into a wild-type host, selecting for the dominant marker Markley, and S. A. Leong. 1997. The second finger of
and against the function of the gene being disrupted, if pos- Urbsl is required for iron-mediated repression of sidl in
sible. Transformants with the desired properties must be Ustilago maydis. Proc. Nutl. A c d . Sci. USA 945882-5887.
screened via PCR or Southern blotting to identify those 4. Arst, H. N. 1996. Regulation of gene expression by pH,
with the desired disruption. A more powerful version of this p. 235-240. In R. Brambl and G. A. Marzluf (ed.), The
approach involves the use of two dominant selectable mark- Mycotu: Biochemistry and Molecular Biology. Springer,
ers in the construct, one of which is selected for as described Berlin, Germany.
above and the other selected against because it will be ab- 5. Arst, H. N., and C. Scazzocchio. 1985. Formal genetics
sent in the desired transformants where a double crossover and molecular biology of the control of gene expression in
has occurred and disrupted the target gene. The second Aspergillus nidulans, p. 309-343. In J. Bennett and L. Lasure
dominant marker will usually be present if the DNA has (ed.), Gene Manipulations in Fungi. Academic Press,
integrated at ectopic sites; thus, such transformants can Orlando, FL.
quickly be eliminated from consideration, allowing the in- 6. Bailey, C. R., and H. N. Arst. 1975. Carbon catabolite
vestigator to focus only on those which may have the de- repression in Aspergillus nidulam. Eur. J. Biochem. 5 1573-
sired knockout. If the function of a cloned gene is uncer- 577.
tain, it is quite possible that disrupting it will be lethal and 7. Barredo, J. L., P. van Solingen, B. Diez, E. Alvarez, J. M.
Cantoral, A. Kattevilder, E. B. Smaal, M. A. Groenen,
thus no simple knockouts can be obtained. When the na- A. E. Venstra, and J. F. Martin. 1989. Cloning and char-
ture of the fungus being studied makes it possible, one can acterization of the acyl-coenzyme A:6-amino-penicillanic
use a diploid host strain for the disruption technique, al- acid acyltransferase gene of Penicillium chrysogenum. Gene
lowing one copy of the gene of interest to be disrupted with 83:291-300.
the other copy remaining functional to ensure viability. 8. Bernhardt, S. A., and R. H. Davis. 1972. Carbamoyl
RIP is an intriguing phenomenon which occurs in phosphate compartmentation in Neurosporu: histochemi-
Neurosporu crassu. When a second copy of a cloned gene is cal localization of aspartate and omithine transcarbamoy-
introduced into a wild-type strain by transformation, both lases. Proc. Nutl. Acud. Sci. USA 69:1868-1872.
copies undergo multiple GC-to-AT mutations throughout 9. Bolker, M., R. Dahl, R. Schlesinger, B. Bergemann, B.
the length of the duplicated segments during a genetic cross Gillissen, R. Schauwecker, M. Urban, B. Schroeer, and
962 MYCOLOGY

R. Kahman. 1991. Mating type genes of Ustilago maydis, specific DNA binding and functions in ascus and as-
p. 231-240. In U. Stahl and P. Tudzynski (ed.), Molecular cospore development. Biochemistry 39:11065-11073.
Biology of Filamentous Fungi, VCH, Weinheim, Germany. 30. Fillinger, S., and B. Felenbok. 1996. A newly identified
10. Burns, D. J., and R. E. Beever. 1979. Mechanisms con- gene cluster in Aspergillus nidulans comprises five novel
trolling the two phosphate uptake systems in Neurospora genes localized in the alc region that are controlled both
crassa. J. Bacteriol. 139:195-204. by the specific transactivator AlcR and the carbon-catabo-
11. Caddick, M. X., H. N. Arst, L. H. Taylor, R. I. Johnson, lite repressor CreA. Mol. Microbiol. 20:475-488.
and A. G. Brownlee. 1986. Cloning of the regulatory gene 31. Fillinger, S., C. Panozza, M. Mathieu, and B. Felenbok.
areA mediating nitrogen metabolite repression in As- 1995. The basal level of transcription of the alc genes in
pergillus nidulans. EMBO J. 5:1087-1090. the ethanol regulon in Aspergillus nidulans is controlled by
12. Campbell, W. H., and J. R. Kinghorn. 1990. Functional the specific transactivator AlcR and the general carbon
domains of assimilatory nitrate reductases and nitrite re- catabolite repressor CreA. FEBS Lett. 368647-550.
ductases. Trends Biochem. Sci. 15:315-319. 32. Froehlich, A. C., Y. Liu, J. J. Loros, and J. C. Dunlap.
13. Cantorai, I. M., B. Diez, J. L. Barredo, E. Avarez, and 2002. White Collar- 1, a circadian blue light photorecep-
J. F. Martin. 1987. High frequency transformation of tor, binding to the frequency promoter. Science 297:815-
Penicillium chrysogenum. Biotech 5:494-497. 819.
14. Car, L. G., P. L. Skatrud, M. E. Scheetzii, S. W. 33. Fu, Y.+H., and G. A. Marzluf. 1987. Characterization of
Queener, and T. D. Ingolia. 1986. Cloning and expression nit-2, the major nitrogen regulatory gene of Neurosporu
of the isopenicillin N synthetase gene from Penicillium crassa. Mol. Cell. Biol. 7:1691-1696.
chrysogenum. Gene 48:257-266. 34. Fu, Y. H., and G. A. Marzluf. 1987. Molecular cloning
15. Chamberlain, N. L., E. D. Driver, and R. L. Miesfeld. and analysis of the regulation of nit-3, the structural gene
1994. The length and location of CAG trinucleotide re- for nitrate reductase in Neurospora crassa. Proc. Natl.
peats in the androgen receptor N-terminal domain affect A c d . Sci. USA 84:8243-8247.
transactivation function. Nucleic Acids Res. 22:3 181-3 186. 35. Fu, Y. H., and G. A. Marzluf. 1990. nit-2, the major pos-
16. Chirgwin, J. M., A. E. Przybyla, R. J. MacDonald, and itive-acting nitrogen regulatory gene of Neurospora crassa,
W. J.Rutter. 1979. Isolation of biologically active ribonu- encodes a sequence-specific DNA-binding protein. Proc.
cleic acid from sources enriched in ribonuclease. Bio- Natl. Acad. Sci. USA 875331-5335.
chemistry 185294-5299. 36. Fu, Y. H., and G. A. Marzluf. 1990. cys-3, the positive-
17. Crawford, N. M., and H. N. Arst. 1993. The molecular acting sulfur regulatory gene of Neurospora crussa, encodes
genetics of nitrate assimilation in fungi and plants. Annu. a sequence-specific DNA binding protein. J. Biol. Chem.
Rew. Genet. 27:115-146. 2 6 5 1 1942-1 1947.
18. Crosthwaite, S. K., J. C. Dunlap, and J. J.Loros. 1997. 37. Fu, Y.-H., J. V. Paietta, D. G. Mannix, and G. A.
Neurospora wc-1 and wc-2: transcription, photoresponses, Marzluf. 1989. cys-3, the positive-acting sulfur regulatory
and the origins of circadian rhythmicity. Science 276:763- gene of Neurospora crassa, encodes a protein with a puta-
769. tive leucine zipper DNA-binding element. Mol. Cell. Biol.
19. Cybis, J., and P. Weglenski. 1972. Arginase induction in 9:1120-1127.
Aspergillus nidulans. Eur. J. Biochem. 30:262-268. 38. Garrett, R. H., and A. Nason. 1969. Further purification
20. Davis, R. H. 1986. Compartmental and regulatory mech- and properties of Neurospora nitrate reductase. J. Biol.
anisms in the arginine pathways of Neurospora c r a m and Chem. 244:2870-2882.
Saccharomyces cerewisiae. Microbiol. Reu. 50:280-3 13. 39. Goldman, G. H., and N. R. Morris. 1995. Aspergillus
21. Davis, R. H. 1996. Polyamines in fungi p. 347-356. In nidulans as a model system for cell and molecular biology
R. Brambl and G. A. Marzluf (ed.), The Mycotu, vol. 111. studies, p. 48-65. In K. W. Adolph (ed.), Microbial Gene
Biochemistry and Molecular Biology. Springer, Berlin, Techniques. Academic Press, New York, NY.
Germany. 40. Griffith, A. N., and R. H. Garrett. 1987. Xanthine dehy-
22. Davis, R. H. 2000. Neurospora. Oxford University Press, drogenase expression in Neurospora crassa does not require
New York, NY. a functional nit-2 regulatory gene. Biochem. Genet. 26:
23. Dobinson, K. F., and J. E. Hamer. 1991. Magnaporthe 37-52.
grisea, p.67-86. In U. Stahl and P. Tudzynski (ed.), Mo- 41. Guthrie, C., and G. R. Fink (ed.). 1991. Methods in
lecular Biology of Filamentous Fungi. VCH, Weinheim, Enzymology, vol. 194. Guide to Yeast Genetics and Molecular
Germany. Biology. Academic Press, San Diego, CA.
24. Dunlap, J. C. 1999. Molecular bases for circadian clocks. 42. Guthrie, C., and G. R. Fink (ed.). 2002. Methods in
Cell 96:271-290. Enzymology, vol. 350. Guide to Yeast Genetics and Molecular
25. Espeso, E. A., and H. N. Arst. 2000. On the mechanism and Cell Biology, Part B. Academic Press, San Diego, CA.
by which alkaline pH prevents expression of an acid- 43. Guthrie, C., and G. R. Fink (ed.). 2002. Methods in
expressed gene. Mol. Cell. Biol. 20:3355-3363. Enzymology, vol. 351. Guide to Yeast Genetics and Molecular
26. Espeso, E. A., J. Tilburn, H. N. Arst, and M. A. and Cell Biology, Part C. Academic Press, San Diego, CA.
Penalva. 1993. pH regulation of a major determinant in 44. Haas, H., K. Angermayr, and G. Stoffler. 1997. Molecu-
expression of a fungal penicillin biosynthetic gene. EMBO lar analysis of a Penicillium chrysogenum GATA factor en-
J . 12 :3 94 1-3 958. coding gene (sreP) exhibiting significant homology to the
27. Exley, G. E., J. D. Colandene, and R. H. Garrett. 1993. Ustilago maydis urbsl gene. Gene 184:33-37.
Molecular cloning, characterization, and nucleotide se- 45. Hanson, M. A., and G. A. Marzluf. 1975. Control of the
quence of nit-6, the structural gene for nitrite reductase in synthesis of a single enzyme by multiple regulatory circuits
Neurospora crussu. J. Bucteriol. 175:2379-2392. in Neurospora crussa. Proc. Natl. Acad. Sci. USA 72:1240-
28. Felenbok, B., and J. M. Kelly. 1996. Regulation of carbon 1244.
metabolism in mycelial fungi, p. 357-368. In R. Brambl 46. Harkness, T. A., R. L. Metzenberg, R. Schneider, R.
and G. A. Marzluf (ed.), The Mycotu, vol. 3, Biochemistry Lill, W. Neupert, and F. E. Nargang. 1994. Sheltered Rip
and Molecular Biology. Springer, Berlin, Germany. in Neurospora crussa. Genetics 136:107-118.
29. Feng, B., H. Haas, and G. A. Marzluf. 2000. ASD4, a 47. Hasunuma, K., and T. Ishikawa. 1977. Control of the
new GATA factor of Neurospora crassa, displays sequence- production and partial characterization of repressible ex-
43. Physiology and Molecular Aspects of Fungi w 963

tracellular 5-nucleotidase and alkaline phosphatase in 65. Lindgren, K. M., A. Lichens-Park, J. L. Loros, and J. C.
Neurosporu crussu. Biochim. Biophys. Actu 480:178-193. Dunlap. 1990. A quick RNA mini-prep for Neurosporu
48. Hensel, M., C. M. Tang, H. N. Arst, and D. W. Holden. mycelial cultures. Fungal Genet. Newsl. 37:21-22.
1995. Regulation of fungal extracellular proteases and 66. Littlewood, B. S., W. Chia, and R. L. Metzenberg. 1975.
their role in mammalian pathogenesis. Can. J. Bot. 73 Genetic control of phosphate-metabolizing enzymes in
(Suppl. l):S1065-S1070. Neurosporu crussu: relationships among regulatory mutants.
49. Hull, E. P., P. M. Green, H. N. Arst, and C. Genetics 79:4 19434.
Scazzocchio. 1989. Cloning and characterization of the 67. Lowendorf, H. D., G. F. Bazinet, and C. W. Slayman.
L-proline catabolism gene cluster of Aspergillus nidulans. 1975. Phosphate transport in Neurosporu. Derepression of
Mol . Microbiol. 3:553-5 60. a high-affinity transport system during phosphorus starva-
50. Hynes, M. J., and M. A. Davis. 1996. Regulation of tion. Biochim. Biophys. Actu 389541-549.
acetamide catabolism, p. 381-393. In R. Brambl and G. A. 68. Lowry, 0. H., N. J. Rosebrough, A. L. Farr, and R. J.
Marzluf (ed.), The Mycotu, vol. 111. Biochemistry and Molec- Randall. 1951. Protein measurement with the Folin phe-
ular Biology, Springer, Berlin, Germany. nol reagent. J. Biol. Chem. 193:265-275.
51. Jarai, G., and G. A. Marzluf. 1991. Sulfate transport in 69. Marzluf, G. A. 1970. Genetic and biochemical studies of
Neurosporu crussu: regulation, turnover, and cellular local- distinct sulfate permease species in different developmen-
ization of the CYS14 protein. Biochemistry 30:47684773. tal stages of Neurosporu crussu. Arch. Biochem. Biophys.
52. Jarai, G., and G. A. Marzluf. 1991. Generation of new 138:254-263.
mutants of nmr, the negative-acting nitrogen regulatory 70. Marzluf, G. A. 1997. Genetic regulation of nitrogen me-
gene of Neurosporu crussu, by repeat induced mutation. tabolism in the fungi. Microbiol. Mol. Biol. Rev. 61:17-32.
Cum. Genet. 20:283-288. 71. Marzluf, G. A. 1997. Molecular genetics of sulfur assimi-
53. Kang, S., and R. L. Metzenberg. 1990. Molecular analy- lation in filamentous fungi and yeast. Annu. Rev. Micro-
sis of nuc-l+, a gene controlling phosphorus acquisition in biol. 5 1:73-96.
Neurosporu crussu. Mol. Cell. Biol. 105839-5848. 72. Marzluf, G. A., and R. L. Metzenberg. 1968. Positive
54. Ketter, J. S., and G. A. Marzluf. 1988. Molecular cloning control by the cys-3 locus in regulation of sulfur metabo-
and analysis of the regulation of cys-f4+, a structural gene lism in Neurosporu. J. Mol. Biol. 33:423-437.
of the sulfur regulatory circuit of Neurosporu crussu. Mol. 73. Mei, B., A. D. Budde, and S. A. Leong. 1993. sidl , a gene
Cell. Biol. 8:1504-1508. initiating siderophore biosynthesis in Ustilago maydis: Molec-
55. Kinnaird, J. H., M. A. Keighren, J. A. Kinsey, M. Eaton, ular characterization,regulation by iron, and role in phyto-
and J. R. S. Fincham. 1982. Cloning of the am (glutamate pathogenicity. Proc. Nutl. Acad. Sci. USA 90:903-907.
dehydrogenase) gene of Neurosporu crussu through the use 74. Metzenberg, R. L. 1979. Implications of some genetic
of a synthetic DNA probe. Gene 20:387-396. control mechanisms in Neurosporu. Microbiol. Rew. 43:
56. Kulmburg, P., N. Judewicz, M. Mathieu, F. Lenouvel, D. 36 1-383.
Sequeval, and B. Felenbok. 1992. Specific binding sites 75. Metzenberg, R. L., and T. J. Baisch. 1981. An easy
for the activator protin, ALCR, in the ulcA promoter of method for preparing Neurosporu DNA. Neurosporu Newsl.
the ethanol regulon of Aspergillus nidulans. J. Biol. Chem. 28:20-21.
267:21146-21153. 76. Metzenberg, R. L., and J. W. Parson. 1966. Altered re-
57. Kulmburg, P., M. Mathieu, C. Dowzer, J. Kelly, and B. pression of some enzymes of sulfur utilization in a temper-
Felenbok. 1993. Specific binding sites in the ulcR and ulcA ature-conditional lethal mutant of Neurosporu. Proc. Natl.
promoters of the ethanol regulon for the CREA repressor Acud. Sci. USA 55:629-635.
mediating carbon catabolite repression in Aspergillus nidu- 77. Muro-Pastor, M., R. Gonzalez, J. Strauss, F. Narendja,
fans. Mol. Microbiol. 7:847-857. and C. Scazzocchio. 1999. The GATA factor AreA is es-
58. Kumar, A., and J. V. Paietta. 1995. The sulfur controller- sential for chromatin remodelling in a eukaryotic bidirec-
2 negative regulatory gene of Neurosporu crussu induces a tional promoter. EMBO J. 18:1584-1597.
protein with 6-transducin repeats. Proc. Nutl. Acud. Sci. 78. Narendja, F. M., M. A. Davis, and M. J. Hynes. 1999.
USA 92:3343-3347. AnCF, the CCAAT binding complex of Aspergillus nidu-
59. Leach, J., B. Finkelstein, and J. A. Rambosek. 1986. lans, is essential for the formation of a DNase I-hypersen-
Rapid miniprep of DNA from filamentous fungi. Neuro- sitive site in the 5 region of the amdS gene. Mol. Cell.
sporu Newsl. 33:32-33. Biol. 19:6523-6531.
60. Lee, K., J. J. Loros, and J. C. Dunlap. 2000. Inter- 79. Negrete-Urtasun, S., W. Reiter, E. Diez, S. H. Denison,
connected feedback loops in the Neurosporu circadian sys- J. Tilburn, E. A. Espeso, M. A. Penalva, and H. N. Arst.
tem. Science 289:107-110. 1999. Ambient pH signal transduction in Aspergillus: com-
61. Lehman, J. F., M. K. Gleason, S. K. Ahlgren, and R. L. pletion of gene characterization. Mol. Microbiol. 33:994-
Metzenberg. 1973. Regulation of phosphate metabolism in 1003.
Neurosporu crussu. Characterization of regulatory mutants. 80. Nelson, M. A., and R. L. Metzenberg. 1992. Sexual de-
Genetics 7561-73. velopment genes of Neurosporu crussu. Genetics 132: 149-
62. Lehman, J. F., and R. L. Metzenberg. 1976. Regulation of 162.
phosphate metabolism in Neurosporu crassu: identification 81. Nelson, R. E., J. F. Lehman, and R. L. Metzenberg.
of the structural gene for repressible alkaline phosphatase. 1976. Regulation of phosphate metabolism in Neurosporu
Genetics 84: 175-182. crussu: identification of the structural gene for repressible
63. Li, Q., and G. A. Marzluf. 1996. Determination of the acid phosphatase. Genetics 84:183-192.
Neurosporu crussu CYS3 sulfur regulatory protein consen- 82. Oakley, B. R., J. E. Rinehart, B. L. Mitchell, C. E.
sus DNA-binding site: amino-acid substitutions in the Oakley, C. Carmona, G. L. Gray, and G. S. May. 1987.
CYS3 bZip domain that alter DNA-binding specificity. Cloning, mapping and molecular analysis of the pyrG
Cum. Genet. 30:298-304. (orotidine-5-phosphate decarboxylase)gene of Aspergillus
64. Linden, H., and G. Macino. 1997. White collar 2, a part- nidulans. Gene 61:385-399.
ner in blue light signal transduction, controlling expres- 83. Oakley, C. E., C. F. Weil, P. L. Kretz, and B. R. Oakley.
sion of light-regulated genes in Neurosporu crussu. EMBO 1987. Cloning of the riboB locus of Aspergillus nidulans.
J. 16:98-107. Gene 53:293-298.
964 MYCOLOGY

84. Okamoto, P. M., Y. H. Fu, and G. A. Marzluf. 1991. nit- 101. Sophianopoulou, V., T. Suirez, G. Diallinas, and C.
3, the structural gene of nitrate reductase in Neurospora Scazzocchio. 1992. Operator derepressed mutations in
crussa: nucleotide sequence and regulation of mRNA syn- the proline utilisation gene cluster of Aspergillus nidulans.
thesis and turnover. Mol. Gen. Genet. 227:213-223. Mol. Gen. Genet. 236:209-213.
85. Orbach, M. J., E. B. Porro, and C. Yanofsky. 1986. 102. Suarez, T., M. V. de Queiroz, N. Oestreicher, and C.
Cloning and characterization of the gene for p-tubulin Scazzocchio. 1995. The sequence and binding specificity
from a benomyl-resistantmutant of Neurospora C T ~ S andU of UaY, the specific regulator of the purine utilization
its use as a dominant selectable marker. Mol. Cell. Biol. pathway in Aspergillus nidulans, suggest an evolutionary
6:2452-2461. relationship with the PPRl protein of Saccharomyescere-
86. Orejas, M., E. A. Espeso, J. Tilburn, S. Sarkar, and visiae. EMBO]. 14:1453-1467.
H. N. Arst. 1995. Activation of the Aspergillus PacC 103. Talora, C., L. Franchi, H. Linden, P. Ballario, and G.
transcription factor in response to alkaline ambient pH Macino. 1999. Role of a white collar-1-white collar-2
requires proteolysis of the carboxy-terminalmoiety. Genes complex in blue-light signal transduction. EMBO ]. 18:
Dev. 9:1622-1632. 4961-4968.
87. Paietta, J. V. 1989. Molecular cloning and regulatory 104. Tao, Y., and G. A. Marzluf. 1998. Synthesis and differ-
analysis of the arylsulfatase structural gene of Neurospora ential turnover of the CYS3 regulatory protein of
crassa. Mol. Cell. Biol. 9:3630-3637. Neurospora crussa are subject to sulfur control. J. Bacteriol.
88. Paietta, J. V. 1990. Molecular cloning and analysis of the 180:478-482.
scon-2 negative regulatory gene of Neurosporu crassa. Mol. 105. Tao, Y., and G. A. Marzluf. 1999. The NIT2 nitrogen
Cell. Biol. 105207-5214. regulatory protein of Neurospora: expression and stabil-
89. Paietta, J. V., R. A. Akins, A. M. Lambowitz, and ity of nit-2 mRNA and protein. Curr. Genet. 36:153-
G. A. Marzluf. 1987. Molecular cloning and characteri- 158.
zation of the cys-3 regulatory gene of Neurosporu c~ussu. 106. Tilburn, J., S. Sarkar, D. A. Widdick, E. A. Espeso, M.
Mol. Cell. Biol. 7:2506-2511. Orejas, J. Mungroo, M. A. Penalva, and H. N. Arst.
90. Pan, H. G., B. Feng, and G. A. Marzluf. 1997. Two dis- 1995. The Aspergillus PacC zinc finger transcription
tinct protein-protein interactions between the NIT2 and factor mediates regulation of both acid- and alkaline-
NMR regulatory proteins are required to establish nitro- expressed genes by ambient pH. EMBO ]. 14:779-
gen metabolite repression in Neurospora crussa. Mol. 790.
Microbiol. 26:721-729. 107. Tilburn, J., C. Scazzocchio, G. G. Taylor, J. J. Zabicky-
91. Perez-Esteban, B., M. Orejas, E. Gomez-Pardo, and Zissman, R. A. Lockingham, and R. W. Davies. 1983.
M. A. Penalva. 1993. Molecular characterization of a Transformation by integration in Aspergillus nidulans.
fungal secondary metabolism promoter: transcription of Gene 26:205-221.
the Aspergillus nidulans isopenicillin N synthetase gene is 108. Tomlinson, G., W. H. Cruickshank, and T.
modulated by upstream negative elements. Mol. Micrc.. Viswanatha. 1971. Sensitivity of substituted hydroxy-
biol. 9:881-895. lamines to determination by iodine oxidation. Anal.
92. Perkins, D. D., A. Radford, and M. S. Sachs. 1981. The Biochem. 44:670479.
Neurosporu Compendium. Academic Press, New York, NY. 109. Tudzynski, B., V. Homann, B. Feng, and G. A.
93. Platt, A., T. Langdon, H. N. Arst, D. Kirk, D. Marzluf. 1999. Isolation, characterization and disruption
Tollervey, J. M. Sanchez, and M. X. Caddick. 1996. of the ureA nitrogen regulatory gene of Gibberella fu-
Nitrogen metabolite signaling involves the C-terminus jikuroi. Mol. Gen. Genet. 261:106-114.
and the GATA domain of the Aspergillus transcription 110. Unkles, S. E., E. I. Campbell, D. Carrez, C. Grieve, R.
factor AREA and the 3 untranslated region of its Contreras, W. Fiers, and C. A. M. van den Hondel.
mRNA. EMBO]. 15:2791-2801. 1989. Transformation of Aspergillus niger with the homol-
94. Plesofsky-Vig, N. 1996. The heat shock proteins and the ogous nitrate reductase gene. Gene 78:157-166.
stress response, p. 171-190. In R. Brambl and G. A. 111. Voisard, C., J. Wang, J. L. McEvoy, P. Xu, and S. A.
Marzluf (ed.). The Mycota, vol. 111. Biochemistry and Mo- Leong. 1993. urbsl , a gene regulating siderophore biosyn-
lecular Biology. Springer, Berlin, Germany. thesis in Ustilago maydis, encodes a protein similar to the
95. Punt, P. J., J. Strauss, R. Smit, J. R. Kinghorn, C. A. erythroid transcription factor GATA-1. Mol. Cell. Biol.
M. J. J. van den Hondel, and C. Scazzocchio. 1995. The 13:7091-7100.
intergenic region between the divergently transcribed 112. Vollmer, S. J., and C. Yanofsky. 1986. Efficient cloning
niiA and niaD genes of Aspergillus nidulans contains mul- of genes of Neurospora crussa. Proc. Natl. Acud. Sci. USA
tiple NirA binding sites which act bidirectionally. Mol. 83:4869-4873.
Cell. Biol. 155688-5699. 113. Weaver, P. L., C. Sun, and T.-H. Chang. 1997. DbpSp,
96. Sachs, M. S. 1996. General and cross-pathway controls a putative RNA helicase in Saccharomyces cerevisiae, is re-
of amino acid biosynthesis, p. 315-345. In R. Brambl and quired for efficient pre-RNA processing predominantly at
G. A. Marzluf (ed.). The Mycota, vol. 3, Biochemistry and site A3. Mol. Cell. Biol. 17:1354-1365.
Molecular Biology. Springer, Berlin, Germany. 114. Wilson, R. A., and H. N. Arst, Jr. 1998. Mutational
97. Schechtman, M. 1986. A moderate-scale DNA prep for analysis of AREA, a transcriptional activator mediating
Neurosporu. Fungal Genet. Newsl. 33:45-46. nitrogen metabolite repression in Aspergillus nidulans
98. Selker, E. U., and P. W. Garrett. 1988. DNA sequence and a member of the streetwise GATA family of tran-
duplications trigger gene inactivation in Neurospora scription factors. Microbiol. Mol. Biol. Rev. 62586-
crussu. Proc. Natl. Acud. Sci. USA 85:6870-6874. 596.
99. Sizemore, S. T., and J. V. Paietta. 2002. Cloning and 115. Wolfinbarger, L., Jr. 1976. Mutations in Neurosporu
characterization of scon-3+, a new member of the Neuro- CTUSSU which affect multiple amino acid transport systems.
sporu crussa sulfur regulatory system. Eukuryot. Cell 1: Biochim. Biophys. Actu 436:774-788.
875-883. 116. Zhou, L. W., H. Haas, and G. A. Marzluf. 1998.
100. Sophianopoulou, V., and C. Scazzocchio. 1989. The Isolation and characterization of a new gene, sre, which
proline transport protein of Aspergillus nidulans is very encodes a GATA-type regulatory protein that controls
similar to amino acid transporters of Succharomyces cere- iron transport in Neurospora crussa. Mol. Gen. Genet.
visiae. Mol. Microbiol. 3:705-714. 259532-540.
Microbiological and Genetic Methods for
Filamentous Fungi
ROWLAND H . DAVIS AND A . JOHN CLUTTERBUCK

44.1. INTRODUCTION ......................................... 965


.
2. MEDIA ................................................. 966
44.2.1. Aspergillus nidulans ................................... 966
44.2.1.1. Minimal Medium .............................. 966
.
2. Complete Media ............................... 966
44.2.2. Neurosporu crmsa .................................... 966
44.2.2.1. Minimal Medium .............................. 966
.
2. Synthetic Crossing Medium ...................... 968
.3. Complete Media ............................... 968
44.2.3. Podospora anserina ................................... 968
44.2.3.1. Minimal Medium .............................. 968
44.2.4. Sordariu spp......................................... 968
44.2.4.1. Minimal Medium .............................. 968
.2. Crossing Medium .............................. 968
44.2.5. Coprinus spp. and Schizophyllum commune .................. 968
44.2.5.1. Minimal Medium .............................. 968
.2. Complete Medium (Supports Fruiting) .............. 968
44.2.6. Nutritional Supplements ................................ 968
44.3. GROWTH ............................................... 968
44.3.1. Measurement of Growth ................................ 968
44.3.1.1. Mycelial Yield ................................ 968
.2. Growth Rate ................................. 968
.3. Mass Culture ................................. 968
.4. Induction of Development ....................... 969
44.4. COLLECTION AND EXTRACTION .......................... 969
44.4.1. Dry-Weight Measurement ............................... 969
.2. Extraction Methods ................................... 969
44.4.2.1. Small Molecules............................... 969
.
2. Macromolecules ............................... 969
.3. Permeabilized Cells............................ 969
44.5. GENETIC METHODS ...................................... 970
44.5.1. Mutagenesis and Mutant Selection ......................... 970
.2. Heterokaryons and Diploids: Complementation Tests ........... 970
.3. Parasexual Analysis ................................... 971
.4. Sexual Crosses and Genetic Analysis ....................... 971
..................................... 971
44.5.4.1. General
. ................... 974
2. Procedures for Particular Fungi
44.5.5. Stock Management .................................... 975
44.6. REFERENCES ............................................ 975

44.1. INTRODUCTION Conference in 1961. Investigators working on Aspergillus


The study of filamentous fungi has a long history. covering nidulans soon joined this group. which expanded to all
investigations of their taxonomy. life cycles. physiology. workers on filamentous fungi in the mid-1980s under the
and nutrition . These studies led to the genetic. biochemi- heading of the newly named biennial Fungal Genetics
cal. and molecular investigations initiated by Dodge. Conferences. The current field is united by discourse based
Beadle and Tatum. and Pontecorvo between 1927 and on genetics. cell biology. development. and the many areas
1953 (20.36). The area loosely called fungal genetics and of molecular biology.
biology is now a formal scientific field and includes stud- The filamentous growth habit of the fungi and their con-
ies of all major groups of filamentous fungi. The field orig- spicuous. differentiated sexual structures set them apart from
inated. in a sense. in the first Neurospora Information other microbial taxa. Methods for cultivating filamentous

965
966 MYCOLOGY

fungi and for study of their classical molecular genetic mech- To these are added, per liter: NaN03, 2 g; trace ele-
anisms have also developed in distinct ways from those used ments, 1 ml (e.g., the solution described by Cove [17]: 40
for bacteria and yeast. This article describes methods drawn mg of Na2B4O7. 10H20,400 mg of C u S 0 4 . 5HZ0,800 mg
largely from the study of the ascomycetes Neurospora crmsa of FeP04 . 2H20, 800 mg of MnS04 * 2Hz0, 800 mg of
and A. nidulans. We add, where appropriate, comments and Na2Mo04. 2H20, 8 g of ZnS04 * 7H20), 10 g of D-glucose,
references to other species now used in similar research. and 15 g of agar (if required).
Certain features of filamentous fungi require special technical A slight, but readily dispersed, precipitate may develop
attention in any methodological treatment. These are (i) a on autoclaving for 10 min at 15 lb. Longer autoclaving
filamentous habit, restricting studies of steady-state liquid may also cause caramelization. This medium supports both
cultures; (ii) a tough cell wall, requiring harsh cell disruption vegetative growth and cleistothecium production. Other
methods for biochemical and molecular work; (iii) the use of nitrogen sources (generally 10 mM) may replace sodium
heterokaryons, mycelia having genetically different nuclei in nitrate and alternatives to glucose as carbon source are
the same cell; and (iv) in many cases, the lack of a natural used at 1%.
diploid vegetative phase, for which heterokaryons, partial
diploids, or artificial diploids may often substitute. 44.2.1.2. Complete Media
The life cycles of two model ascomycetes, N. crussu and A simple yeast extract medium (31) consists of 5 g of yeast
A. nidulans, are shown in Fig. 1, with some of the structural extract and 25 g of glucose per liter. For a more complex
terminology used in this article. The biology and life cycles medium, add to minimal medium 2 g of Difco peptone, 1 g
of other fungi, in particular the basidiomycetes such as of yeast extract, 1.5 g of casein hydrolysate (if acid-
Coprinus cinereus and Schizophyllum commune, may be found hydrolyzed, this lacks tryptophan), and 1 ml of vitamin so-
in the treatise of Fincham et al. (25). Many other general lution (containing 100 mg of riboflavin, 100 mg of nicoti-
references may be consulted (7, 8, 15, 16, 20, 21, 36, 42, namide, 10 mg of p-aminobenzoic acid, 5 mg of pyridoxine,
43). A recent, comprehensive analysis of the genome of N. 5 mg of thiamine, and 1 mg of biotin per liter).
crussu (9) contains much relevant information of use to
modern workers. 44.2.2. Neurospora crassa (20, 21)
A World Wide Web site of use to fungal geneticists, with
links to many other fungal websites, includes the Fungal 44.2.2.1. Minimal Medium
Genetics Stock Center (FGSC) (http://www.fgsc.net). This
site, initially serving only the Neurospora community, is now Vogels Medium N Formula for 5 0 X Concentrate
a clearinghouse for general information on filamentous fungi. To 750 ml of HZOadd, with stirring in the following order:
Other URLs include, for A. nidulans, http://www.genetics 150 g of Na3 citrate * 5.5 H20, 250 g of KHzP04, 100 g of
.unimelb.edu.au/hdlab/ (Hynes), http://www.ibls.gla.ac.uk/ NH4N03, 10 g of MgSO,. 7Hz0, 5 g of CaC12 . 5HZ0,5
aspergillus/index.html (linkage maps), http://www.genome ml of trace elements (see below), 5 ml of D-biotin stock (see
.broad.mit.edu/annotation/fungi/aspergillus(Broad Insti- below), and a few milliliters of chloroform as preservative.
tute; genome), and http://www.aspergillus.man.ac.uk/index Predissolving the CaC12 in ca. 20 ml of HzO aids greatly in
.html. For N.crassa, http://www.fgsc.net (FGSC) and http:// introducing it into the solution. Store at room temperature.
www.genome.broad.mit.edu/annotation/fungi/neurospora For use, dilute 50X and add 15 to 20 g of sucrose and 15 g
(Broad Institute; genome) are available. A general site for of agar per liter if required. All ingredients may be auto-
fungi, having many other useful links, is http://mycology claved together.
.cornell.edu/fgenetic.html. Trace Elements
This article concentrates most heavily on A. niduluns For truce elements, dissolve successively, with stirring, in 95
and N. crussa as model organisms, allowing us to give spe- ml of water: citric acid * H20, 5 g; ZnS04 . 7Hz0,5 g;
cific information on culture and techniques rather than ex- Fe(NH4)z(S04)z. 6H20, 1 g; CuS04 . 5Hz0, 0.25 g;
haustive variations for all filamentous fungi that have con- MnS04 . lHzO, 0.05g; HB03, 0.05g; Na2Mo04. 2Hz0,
tributed to the growth of the field. The reader is referred to 0.05g; and chloroform as preservative, 1 ml. Store at room
the references given and to the websites above for up-to- temperature. Any other formulation that assures similar
date information about other fungi. amounts of the metal ions is allowable.
Biotin Solution
44.2. MEDIA For biotin solution, dissolve 5 mg of D-biotin in 100 ml of
Ingredients are given per liter final volume unless otherwise 50% ethanol. Store at 4C.
indicated.
44.2.2.1.1. Plating and Spotting Media
44.2.1. Aspergillus nidulans (1 5 ) If colonial growth on agar is required, replace the sucrose
by 20 g of sorbose, 0.5 g of glucose, and 0.5g of fructose.
44.2.1 . I . Minimal Medium (Platings are better if the synthetic crossing salts, below, are
This medium is a better-buffered version of the minimal used; 10 g rather than 20 g of sorbose per liter is sufficient
medium described by Pontecorvo et al. (44) and Cove (17). in that medium.) For spot tests of growth on agar, use 8 g of
Salts: KHZPO4,1.4 g; K2HP04 . 3H20, 0.9 g; MgS04 . sorbose and 4 g of sucrose per liter. All ingredients may be
7Hz0,O.l g; KC1,O.l g. (These salts can be kept as a lOOx autoclaved together; sorbose will darken the medium with
concentrate.) autoclaving but remains effective.

FIGURE 1 Life cycles. (Left) Neurospora crassa; (right) Aspergillus nidulans. For scale, note that the ascus of N. crussu is about
150 p m long, and ascospores are about 20 pm long. By contrast, A. nidulans asci are only 15 k m in diameter, and the ascospores
are correspondingly smaller.
N. crassa

aerial hypha,
conidium 0
-A. nidulans

A7
conidia

ASEXUAL CYCLE

e
P

mycelium

ascospore
ascospore germling

protoperithecium
conidium, opposite
mating type
germling immature
cleistothecium
(with hulk cells)
0
I / SEXUAL CYCLE ascospore i
J,
SEXUAL CYCLE

I -.
-
w

ascospore
22
VI
n
c
3
ascus with mature cleistothecium,
%.
with cutaway showing asci
ascospores

ascus with ascospores ro


QI
U
968 MYCOLOGY

44.2.2.2. Synthetic Crossing Medium ommended above be inhibitory or insufficient. For Asper-
Per liter, use KNO3, 1 g; K2HP04, 0.7 g; KH2P04, 0.5 g; gillus, add vitamins, 0.1 to 100 pg/ml; purines and pyrim-
MgSO, . 7H20,0.5 g; NaC1, 0.1 g; CaC12, 0.1 g; D-biotin, idines, 50 to 100 pg/ml; amino acids, 4 mM for the most de-
5 pg; and trace elements, as for Vogels at the same final manding mutants. Better uptake may be obtained if an
concentration. Can be kept as 2X strength stock. Add for amino acid (e.g., arginine or proline), rather than nitrate or
use: nutritional supplements, carbon source (see below), ammonium, is supplied as sole nitrogen source. Alternative
and agar, 15 g. Heat to melt agar, distribute to culture tubes, nitrogen sources are supplied at 5 to 10 mM, alternative car-
and insert Whatman no. 1 filter strips as carbon source; au- bon sources at 1% (wtlvol).
toclave, and slant. Grow one mating type at 25C for 6 days
to form protoperithecia before fertilizing by barely wetting
the surface of the slant or plate with a suspension of conidia
44.3. GROWTH
of the other mating type. A few crosses proceed better with 44.3.1. Measurement of Growth
10 g of sucrose per liter in place of filter paper. An alterna-
Fungal strains may be grown to test for the amount of
tive to synthetic medium is corn meal agar (without glu-
growth or the rate of growth in given conditions. Strains
cose; Difco). For plating of conidia, the salts above may be
may be grown for isolation of small molecules, macromole-
used with 0.5 g of fructose, 0.5 g of glucose, and 10 g of D-
sorbose per liter. cules, organelles, or permeabilized cells. Finally, some
species may be grown in exponential cultures for kinetic
44.2.2.3. Complete Media measurements of enzyme regulation, short-term isotope in-
Addition of 0.5% yeast extract and 0.5% casein hydrolysate corporation, and the like.
is sometimes used for N.crussu. Other complex additives 44.3.1 . I . Mycelial Yield
may be made (20,21) but are now rarely used. Many addi-
tives, particularly those that serve as nitrogen sources, may Small inocula are introduced into media in small flasks and
inhibit crossing. harvested when growth is complete, or at a specified time if
rates of growth are quite different. Such tests are often done
44.2.3. Podospora anserina (24) to test for the efficiency with which strains use a supple-
ment to the medium or display a response to an inhibitor.
44.2.3.1. Minimal Medium
44.3.1.2. Growth Rate
Use N. crmsa synthetic crossing medium salts, trace ele-
ments, and biotin, to which 20 g of fructose and 200 pg of Multiple cultures such as those just described can be har-
thiamine are added. However, modifications are required vested at different times to gauge roughly the rate of growth.
for some purposes, such as mating, microconidial formation, In many fungi, growth rate can be ascertained by the rate of
and spore germination. These may be found in Esser (24). increase of colony radius in petri dish cultures. For A. nidu-
lam, which grows naturally as a dense colony, the degree of
44.2.4. Sordaria spp. (32) conidiation and the visual diameter and bulk of the colony
are good qualitative indicators of the efficiency of use of
44.2.4.1. Minimal Medium various carbon and nitrogen sources. In rapidly spreading
Vogels minimal medium (see above for Neurosporu). For as- species like Neurospora, one may measure daily progress
cospore germination, add 7 g of sodium acetate per liter. down a race tube, a 300- to 500-cm glass tube (bent up-
ward at the ends) half-filled with solid medium and inocu-
44.2.4.2. Crossing Medium lated at one end (Fig. 2). A simpler arrangement is the use
Cornmeal agar (17%) with 0.2% glucose; alternatively, 10 g of disposable 25-ml plastic pipettes partially filled with
of sucrose, 7 g of glucose, 1 g of yeast extract, 0.1 g of molten medium, allowed to solidify in a horizontal position.
KH2P04,and 17 g of Difco cornmeal agar. and later broken and capped for use (53).

44.2.5. Coprinus spp. and Schizophyllum 44.3.1.3. Mass Culture


commune (46) Heavy inocula must be used for large liquid cultures. For
Neurosporu, these are prepared by growing crops of conidia
44.2.5.1. Minimal Medium for inocula for 7 days (the first two at 32C,the last at room
L-Asparagine (1.5 g) or 1.5 g of (NH4)2HP04,0.46 g of temperature) in 25 or 50 ml of solidified medium in 125-ml
KH2P04, 1 g of K2HP04,0.5 g of MgSO, * 7H20,trace el- or 250-ml flasks, respectively, followed by suspension of the
ements (as for N. crussa, above), 120 pg of thiamine, 20 g resulting conidia in sterile water introduced into the growth
of glucose, and 15 g of agar. flask. Conidia are then filtered through two layers of
cheesecloth, held by a rubber band as a bag in the mouth of
44.2.5.2. Complete Medium (Supports Fruiting) a sterile, dry flask. The conidia are centrifuged and resus-
Two grams of yeast extract, 2 g of peptone, 0.46 g of
KHZPO,, 1 g of KlHPO,, 0.5 g of MgSO4 * 7H20, 20 g of
glucose, and 15 g of agar. Coprinus fruits much better on
sterilized horse dung. Direction of growth
*
44.2.6. Nutritional Supplements
In general, work with auxotrophic strains requires additions
of vitamins (5 pglml), amino acids (50 to 200 pg/ml), and FIGURE 2 Neurosporu race tube. The tube (300 to 500
pyrimidines and purines or their nucleosides (100 pg/ml) as mm long) is supported on a rack in the position shown and is
appropriate. The concentrations of each required nutrient inoculated at one end. The position of the mycelial frontier is
should be established specifically in each case lest those rec- marked at intervals and distances are recorded thereafter.
44. Microbiological and Genetic Methods for Filamentous Fungi 969

pended in a small volume of water for use. Collection of A. mycelium is coherent, small cultures may be collected with
nidulans conidia in water may be accomplished by flooding a simple spatula. Dry-weight measurements can be made on
a mature plate culture or slant with 0.1% Tween 80 and material dried on towels or filters at 80C or dried with ace-
scraping the surface. Large cultures (200 to 2,000 ml) of liq- tone while the material is collected on a suction filter.
uid culture will yield large amounts of mycelium if they are These weights may be slightly different owing to acetone
well aerated by shaking or by bubbling filtered air through leaching of lipids and some small molecules. Therefore, a
them. In such cultures, a large inoculum (ca. lo6 conidia per consistent method should be decided on at the outset.
ml for Neurospora) will begin to grow rapidly, and growth
will remain relatively homogeneous and well dispersed for 44.4.2. Extraction Methods
several doublings. Many fungi clump or grow in tight but-
tons, leading to poor aeration in the centers of growth and 44.4.2.1. Small Molecules
a consequent heterogeneity of cells. Therefore, if experi- The small molecules of mycelia may be extracted in 5%
ments permit it, large inocula are better to maintain a trichloroacetic acid or 5% perchloric acid, and more selec-
young, rapidly growing culture. In A. nidulans, growth at tively with acetone, chloroform, hot alcohol, and the like,
37C in 100 ml of medium in 250-ml flasks or 300 ml in depending on the experiment. The methods differ little
1,000-ml flasks with vigorous shaking assures a well- from those used in animal and plant cell cultures.
dispersed culture, which can be harvested through nylon
net muslin. In general, methods for mass growth are best for 44.4.2.2. Macromolecules
short-term work, owing to the dangers of contamination. For extraction of macromolecules and organelles, the tough
Subcultures of such cultures, if they must be made, should cell wall of fungi presents a problem. Three methods gener-
be checked carefully for contamination before further use. ally get around this. The first is to wash the mycelium, as it
Continuous culture is important in industrial mycology and is collected, in the buffer to be used, draw off excess liquid
is therefore the subject of considerable experiment, but can or press the mycelium dry, and grind the pad in a cold mor-
only be touched on here. Typical of experimental studies tar with sand or glass beads. One should add very little
with a genetical basis are those considering the effect of reg- buffer until the macerate is collected. The second is to sus-
ulatory mutants on secondary metabolite production (e.g., pend the washed mycelium in buffer and subject it to 10- to
reference 1) and studies of the stability of fungi transformed 20-s periods of agitation with glass beads in a Bead-Beatera
for overexpression of native or exogenous genes (22). (Biospec Products, Bartlesville, OK). This instrument, with
a selection of chambers, has been used widely to disrupt
44.3.1.4. Induction of Development cells in a large range of volumes (18). The chamber is half-
In A.nidulans, conidial and sexual development occur spon- filled with buffer-wetted beads (usually 0.3 to 0.5 mm). The
taneously in surface cultures, but submerged cultures are usu- buffer (e.g., 10 mM TES [pH 7.21 with 1 mM EDTA) is os-
ally sterile. Submerged conidiation is, however, possible motically stabilized with 1 M sorbitol if the goal is to isolate
under certain conditions (35). For developmental studies, organelles. For proteins, the outcome is a rather dilute sus-
conidiation can be induced by exposing liquid-grown pension, inappropriate for many studies. The third method
mycelium to air by plating onto agar or filter paper. Cultures is to collect cells in cold acetone and grind them to a pow-
must reach a certain age (-16 h at 37C) before they are der in a spark-free blendor (Omnimixer). The powder is air
competent to conidiate (4), the first signs of conidiophore dried and then used, at high density ( > l o 0 mg/ml buffer),
development becoming evident ca. 8 h after induction. to extract proteins. Many enzymes and other proteins sur-
Wild-type strains do not conidiate well in the dark, but most vive this very well and may therefore be extracted with lit-
laboratory strains carry the weAl (velvet) mutation, which tle difficulty in a relatively lipid-free solution.
overcomes the requirement for light. Sexual development is Lyophilization of pressed-dry mycelium is often used to
encouraged by partial anaerobiosis (see section 44.5.4.2) and prepare materials for extraction. Powders can be made by
occurs more slowly than conidiation, starting at about 50 h vortexing the pad in a plastic tube with several large glass
at 37C (14). The first ascospores are formed at 90 h, and beads. Finally, for DNA and RNA extractions, many inves-
cleistothecia are fully mature in 3 weeks, by which time each tigators use mycelium that is pressed dry and frozen. The
contains up to 5 x lo5 ascospores. pad is ground to a fine powder in liquid nitrogen with a
In N.crassa, formation of macroconidia occurs on the mortar and pestle, and the macerate is mixed with an ap-
surface of cultures and is much more profuse when cultures propriate extraction mixture while still frozen (e.g., guani-
are exposed to light. Starvation for carbon and nitrogen also dine thiocyanate buffer for RNA). See chapter 43 by
elicits rapid conidiation. Macroconidial development can Marzluf, this volume, for handling nucleic acids.
be synchronized by transferring mycelial mats to filter paper
and exposing them to light. Macroconidia do not form in 44.4.2.3. Permeabilized Cel Is
submerged culture of wild-type strains. N. crassa also forms Permeabilized cells are extremely useful for rapid enzyme
uninucleate microconidia, a quite different form of propag- assay (6). Most enzymes can be monitored in this way, as-
ule. They are used occasionally to isolate pure strains of suming that specificity can be assured. The following method
transformants, which are usually heterokaryotic when first is based on Neurospora. Five-milliliter samples are withdrawn
isolated (23). Conditions for induction of protoperithecia from growing, dispersed mass cultures and placed in chilled
of N. c r a m are given above (section 44.2.2.2). 12-ml centrifuge tubes. The tubes are centrifuged, the super-
natant medium is poured off, and tubes are drained well by
inversion on a paper towel. The cells are resuspended in 3 ml
44.4. COLLECTION AND EXTRACTION of 10 mM EDTA in 50 mM K+P04 (pH 7.2). A toluene-
ethanol mixture (0.2 ml; 1:4 [vol/vol]) is added, and the sus-
44.4.1. Dry-Weight Measurement pension is mixed. The suspension is centrifuged after 1 or 2
Mycelia can be collected on paper or membrane filters, min, the supernatant is poured off, and the cells are frozen at
using suction flasks, or by filtration through muslin. If the -80C. Freezing of cells is essential for full permeabilization,
970 w MYCOLOGY

even if they are to be used immediately after collection. plates and tested by spot testing or replication onto selec-
Small molecules are largely removed by the toluene treat- tive media to identify mutants.
ment and centrifugation prior to freezing. They therefore do Two hundred or more distinct Aspergillus microcolonies
not complicate the ingredients of enzyme reaction mixtures can be obtained on a 9-cm petri dish by adding 0.01%
when they are added. After thawing, the cells are brought to Triton X-100 to the medium before pouring. Mutants can
1 ml with approximately 0.85 ml of a buffer compatible with be identified after replica plating such colonies from com-
the enzyme assay. Once suspended, they may be sampled for plete, or other supplemented medium, to minimal medium
enzyme assay and for protein determination. The latter may using damp velvet or filter paper (Color Plate 18). In
require extraction overnight in 1 N NaOH to solubilize all Neurospora, a similar density of colonies is obtained on plat-
proteins. ing medium (see above), but they must be picked to indi-
Treatment of Aspergillus mycelium with toluene vapor is vidual tubes and spot tested when mature.
sufficient for X-Gal detection or assay of P-galactosidase fu- Gene disruption by random integration of plasmids or
sion proteins, while protoplasting enzymes are used to make transposable elements has the advantage that the target
cells permeable to antibodies (29). DNA is tagged for subsequent gene isolation, e.g., in
Mugnuporthe grisea (5 1). Plasmid integration can be facili-
tated by restriction enzyme-mediated integration (REMI),
44.5.GENETIC METHODS in which a restriction enzyme is included in the transfor-
mation mix, e.g., in Aspergillus fumigatus (13) and Co-
44.5.1. Mutagenesis and Mutant Selection prinus cinerea ( 19), while Agrobacterium-mediated transfor-
Suspend single-celled units (conidia, oidia, etc.) in water mation has been used for insertional mutagenesis in
and irradiate with UV light at 260 nm to 50% survival. Fusarium (39). In A. nidulans, transposon-induced muta-
Chemical mutagens such as ethylmethane sulfonate, hy- genesis is less effective than had been hoped, possibly
droxylamine, ICR- 170, and nitrosoguanidine may also be because disruption of functional genes seems to be infre-
used, although their mutant/survivor ratios may be differ- quent (33).
ent, depending on the dose, than with UV. Nitroquinoline Targeted mutagenesis-that is, the mutation of particu-
oxide (5) is recommended as an effective but readily inacti- lar genes-takes advantage of cloned DNA. Two general
vated mutagen. The 50% survival level with UV light is suf- methods are available in N. crassa (20) and A. nidulans
ficient to induce many mutations, but not so heavy as to (38), each dependent on a different mode of stable integra-
yield a large fraction of multiple mutants. Nevertheless, tion of DNA during transformation. In one mode, the DNA
outcrossing the initial mutants to wild type should be done integrates by recombination with the homolog in the ge-
to remove cryptic, additional mutations from the back- nome. In the other, the DNA integrates quasirandomly in a
ground and to establish that all phenotypic attributes of the nonhomologous point or points in the genome. A linear, in-
primary mutant are associated with a single gene. UV light complete fragment of the gene to be disrupted may be
is also suspected of inducing more chromosomal mutations introduced into cells. Cells that integrate it (by a double
(translocations and inversions) than chemical mutagens. crossover) into the homologous gene will suffer a disruption
In direct selection, mutants are plated at high density of the gene in question, and the resulting mycelium should
and picked from a selective medium, as one would do for display a mutant phenotype, if known or predicted. The
antibiotic-resistant variants or prototrophic revertants of an transformant, once purified by serial conidial isolation, may
auxotroph. These must be separated from nonmutant cells then be crossed (see below) to verify its normal genetic be-
by successive conidial isolation or by crossing. The latter is havior and to purify the mutant nuclear type of other, non-
necessary in species in which vegetative propagules are homologous integration events. In Neurospora, this tech-
multinucleate and sexual spores are uninucleate. nique has been greatly simplified by the use of strains
In indirect selection, several enrichment schemes may unable to integrate DNA into nonhomologous locations in
be applied. For auxotrophic mutants of Neurosporu, which the genome (4 1).
has a spreading mode of growth, the filtration-concentra- In A. nidulans, recessive lethal gene disruptions can be
tion method is suitable. This is done by suspending muta- identified by analysis of heterokaryons or diploids in which
genized conidia in minimal liquid medium, shaken contin- only one of the two copies in the cell has been disrupted
uously over 48 to 60 h, with filtration of the culture through (e.g., reference 34).
cheesecloth at close enough intervals so that little growth In N.crussu, a peculiar phenomenon named repeat in-
remains in the flask. Wild-type cells grow and are removed; duced point mutation (RIP) has been discovered (48). The
auxotrophs pass through the cheesecloth and survive. The RIP phenomenon takes advantage of the frequent non-
culture is finally concentrated by brief centrifugation and homologous integration of transforming DNA in normal
plated on media appropriate to the mutant phenotype de- strains. Such integration creates a strain with a genetic du-
sired (see references 20 and 21 for details). Other indirect plication. Both copies of the duplicated DNA, resident and
methods include inositol-less (Neurosporu) or biotinless exogenous, are frequently and multiply mutated during the
(Aspergillus) death, in which the starting material bears a sexual cycle. This mutagenic technique is explained in sec-
nonrevertible inositol or biotin mutation. Such strains will tion 44.5.4.
die if allowed to germinate and grow without their supple-
ments. However, if additional mutations are induced that 44.5.2. Heterokaryons and Diploids:
block growth early in minimal medium, these derivatives Complementation Tests
will prevent the suicidal growth and thereby cells with new Heterokaryons and (in Aspergillus) diploids are used for
mutations will become enriched among the survivors after determination of dominance and complementation. In
24 to 36 h of growth. Plating of the final population on Aspergillus, diploid formation is also the first step in para-
media with inositol or biotin and the appropriate supple- sexual analysis (see section 44.5.3), which is routinely used
ment for the desired new mutants follows. Germlings are to locate new mutants to chromosomes and which can be
then picked into individual tubes or onto a grid pattern on used for further mapping.
44. Microbiological and Genetic Methods for Filamentous Fungi m 971

Finally, in any species in which hyphal fusion can be wild strains. In A. nidulans, incompatibility is determined
arranged, extranuclear replicating elements such as mito- by at least eight loci ( 3 ) ,but most laboratory strains are de-
chondrial DNA, viruses, and plasmids may be transferred by rived from a single wild isolate, so incompatibility is not a
making a heterokaryon and then resolving it, after growth, problem. The same considerations apply to N. crassu, but
into the homokaryons. In one of these homokaryons, the some lineages in use require that investigators be sure of
extranuclear element will be associated with another nu- compatibility before embarking on complementation tests.
clear type. A great many extranuclear variants are infective
and may prove hard to remove without resorting to sexual 44.5.3. Parasexual Analysis
crosses, as indicated below. In A. nidulans, extranuclear in- Aspergillus diploids are moderately stable. Diploid colonies
heritance is readily demonstrated using mitochondrially grow like haploids, but give rise to genetically variant sec-
encoded markers (11, 12), e.g., csA (cold sensitivity), oliA, tors by two processes: haploidization and mitotic recombi-
and cumA (resistance to oligomycin and chloramphenicol, nation. Both events are rare and occur independently, but
respectively). Heterokaryons between mutant and wild- the combined result is a messy, stepwise equivalent to meio-
type strains for these markers show vegetative segregation, sis after diploid formation, a series of events that is known
independently of nuclear markers. Recombination can also as the parasexual cycle (30).
occur between mitochondria1 markers, and a simple linkage Haploidization of diploids formed between new mutants
map has been drawn up for four loci (52). Similar work in and a master strain carrying markers for all eight chromo-
N.crassu has been reviewed (20). somes is routinely used to locate new loci to chromosomes
Complementation tests enable one to group mutants into (37). Haploidization can be induced with the mitotic poi-
allelic classes. For Neurosporu, heterokaryons of all possible son benomyl (2 yg/ml in complete medium), haploid sec-
pairs of mutants are made and tested for growth on a medium tors being identified by segregating spore color markers and
rigorously selective for wild-type growth (see references 20 conidial diameter. Since haploidization is independent of
and 21 for more detailed procedures). This is done conve- crossing over, classification of a small number of haploids is
niently for Neurosporu in half-grids of tubes (13 x 100 mm) usually sufficient to show complete linkage of the new mu-
with 1 ml of liquid medium, introducing suspensions of coni- tation with one of the chromosomal markers, and inde-
dia of each mutant into rows and columns of tubes. Even pendent assortment with the remainder. Linkage to markers
in minimal medium, conidia of auxotrophs will germinate on two different chromosomes is indicative of a transloca-
enough to fuse at the bottom of tubes. Smaller-scale tests are tion in the mutant strain.
made on doubly inoculated spots on agar medium. All such Mitotic crossing over can be used to order markers on
tests should be accompanied by tests of the homokaryons so one chromosome with respect to the centromere and to
that positive responses, where they occur in a heterokaryon, each other. During or after duplication of two homologous
can be compared with possible leaky growth of one or the chromosomes in mitosis, a crossover may occasionally occur
other mutant. Note: the mating-type locus (mtA/mtu, or A between nonsister chromatids of a homologous pair, result-
and a) of N. cra~sublocks the formation of heterokaryons, so ing in exchange of distal chromosomal segments. Because
mutants derived from different mating types cannot be as- thereafter the two chromatids of each homolog go to oppo-
sessed directly for complementation. site poles at nuclear division, a recombinant chromatid may
A. nidulans heterokaryons are unstable unless the het- end up in the same daughter nucleus as a nonrecombinant
erokaryotic condition is forced by the use of additional, chromatid of the other homolog. The result will be homo-
complementing auxotrophies. Therefore, heterokaryons are zygosity for all markers distal to the crossover. The crossover
made by germinating mixed conidia of the parent strains on may take place at any point, and therefore selection for such
liquid or solid enriched medium and transferring the result- events, facilitated by selection for homozygosity of a distal,
ing growth (i) to medium permissive for the mutants to be visible, or drug-resistance marker, allows ordering of all
tested, but selective for complementation between the forc- more proximal markers with respect to the centromere.
ing markers, then (ii) to the test medium, allowing growth This technique has great value in Aspergillus, since meiotic
only for pairs showing complementation between the mu- chromosomal linkage maps are long (some more than 400
tants to be tested. map units) and loci on the same chromosome may therefore
Since Aspergillus spp. heterokaryons vary in the propor- not show meiotic linkage. However, it is not a routinely em-
tions of the two constituent nuclei, the use of diploids will ployed procedure because it requires test strains carrying all
give clearer results in both complementation and domi- the markers to be mapped in coupling with a distal selec-
nance tests. Aspergillus conidia are uninucleate, so rare table marker.
diploids can be selected by plating conidia from a het- Haploidization is believed to be precipitated by mitotic
erokaryon on media selective for complementation between nondisjunction of single chromosomes (30). Rarely, how-
the forcing markers. Heterozygous diploid conidia form by ever, some nondisjunctions are followed by loss or gain of
fusion of unlike nuclei in heterokaryons at a frequency of chromosomes to restore the diploid state. If in this process
approximately 1 X lop7. They are best selected by dispers- one homolog is lost and replaced by a copy of the other, the
ing conidia of a heterokaryon (at various concentrations) result will be homozygosity for whole chromosomes, to be
into selective agar medium cooled to 40C and still molten. distinguished from homozygosity of chromosome arms re-
A valuable aid to the identification of diploids is the use of sulting from mitotic crossing over.
different spore color markers in the two parents, e.g., yA Neurosporu spp. do not form diploids suitable for para-
(yellow) and WA(white), which complement in individual, sexual analysis.
uninucleate diploid spores to give the wild-type green color.
(Heterokaryotic colonies will yield a variegated spore 44.5.4. Sexual Crosses and Genetic Analysis
mass.) Another visible characteristic of diploid conidia is
their volume, which is twice that of haploid conidia. 44.5.4.1. General
Heterokaryon incompatibility in many fungal species Genetic analysis can be performed by rationales described
limits formation of heterokaryons and diploids between in a variety of chapters, books, and manuals (10, 12, 16, 20,
972 rn MYCOLOGY

21, 2 5 , 3 2 , 3 7 , 4 4 ) focused on various species. In general, al- It is good practice to cross new mutants, after isolating
most all genetic analysis can be done with random meiotic them, to wild-type or standard strains to ascertain whether
products if the latter derive from a single nucleus and are the phenotypic features associated with the new mutant are
easily separable in suspension. Platings of sexual spores on determined by one or more genes, and also to obtain a mu-
nonselective medium, followed by picking the germlings to tant stock that is derived from a single nucleus. In haploid
individual tubes and testing them thereafter, will allow the fungi, such crosses are expected to yield a 1:l ratio of mu-
ratio of phenotypic classes to be ascertained. Germination tant and wild-type progeny if all features of the mutant are
percentages should be recorded in the event that certain determined by a single gene, but the results will be more
classes of progeny are inviable and might be accounted for complex if the mutant phenotype is due to mutation of
in the nongerminated fraction. more than one gene. In determining progeny ratios, it is im-
Segregation, independent assortment, and linkage can portant to be aware that deleterious mutants often reduce
be recognized crosses by characteristic ratios among random spore viability, so that mutant phenotypes may be under-
meiotic products (ascospores or basidiospores). If parental represented.
strains differ in one characteristic (e.g., having the pheno- In classical genetic practice, the next step is to deter-
types Arg+ and Arg-), a cross can reveal whether they bear mine the position of the new mutant locus on a linkage
alternative alleles at a single gene. Thus the cross Arg+ x map. This procedure has several functions. First, it relates
Arg- may yield 65 Arg' and 70 Arg-. The approximate 1:l the mutant locus to other genes, sometimes revealing close
ratio among the progeny signifies a single gene difference linkage or possible allelism to mutants of the same pheno-
between alleles arg+ and urg-. Notice that the notation dif- typic class. In other cases, close linkage may indicate a clus-
fers for phenotype (Arg-) and genotype (urg-). In what fol- ter of genes of related function and possibly coordinate con-
lows, the nomenclatural conventions of N. crassu are used trol. Other features of map position, e.g., proximity to
(20). centromeres or telomeres, where heterochromatin is often
If the parental strains have two phenotypic differences located, may also be of interest. Finally, an ascertained map
(e.g., Pyr+ Arg- and Pyr- Argf), the genes responsible may position can facilitate cloning that requires chromosome
be unlinked and show independent assortment. Thus the walking.
cross of Pyr+ Arg- x Pyr- Arg' might yield 103 Pyr+ Recombination frequencies are subject to both genetic
Arg-, 99 Pyr- Arg', 89 Pyr' Arg+, and 110 Pyr- Arg-, and environmental variation, so linkage map distances may
that is, about 25% of each possible class. By the convention vary considerably from one cross to another or among dif-
of the previous cross, we would assign gene and allele sym- ferent laboratories where standard conditions and the ge-
bols pyr'lpyr- and arg'larg- to the two genes. netic background of fungal strains might differ. Because of
If the two differences are due to linked genes, linkage is this variability, reliable linkage maps require data drawn
detected by observing more parental phenotypes than re- from three-point crosses, in which the position of any new
combinant phenotypes among the meiotic products. Thus a gene is related to two other markers in the same cross.
cross of strains having genotypes argt inl- X arg- inl+ may These crosses at least yield unambiguous gene order, what-
yield 345 argc inl-, 320 arg- i d + , 23 arg' inl' and 30 arg- ever the source of the information. Methods for efficient lo-
inl-. Such a ratio, with the recombinant genotypes in the calization and mapping of fungal genes with three-point
minority, is the criterion for linkage of the genes urg and inl. crosses may be found in several accessible sources (e.g., ref-
In this example, the map distance between the two genes is erence 25). In N. crussu, rapid localization of new mutations
simply the percentage of recombinants among the total to linkage groups may be made by crossing the mutants to
meiotic products: 531718 = 7.4%, or 7.4 map units. special translocation strains (20, 21) and assessing the out-
If two mutants have the same phenotype (e.g., Arg-), come on the basis of random-spore analysis, whereas in A.
the outcome of crosses can be interpreted with these rules nidulans initial localization of mutants to linkage groups is
in mind. First, if two strains carry mutations for different arg done via the parasexual cycle (see section 44.5.3).
loci, arpl and arg-2, the cross can best be understood with In work with cloned DNA, detailed linkage maps can be
the following notation: arg-1- arg-2+ X arg-1' arg-2-. obtained without a collection of already mapped mutants.
Among the progeny of this cross, we will usually see an aux- Such maps are based on polymorphism for restriction frag-
otrophic class, Arg-, comprising three genotypes (arg-1- ment length (RFLP) or other DNA markers. In N. crassa, a
arg-2+, arg-Ic arg-Z-, arg-1- arg-2-) and a prototrophic species in which considerable polymorphism is known,
class, Arg+ (arg-l+ a ~ g - 2 ~If) .the genes are unlinked, the crosses of a mutant-marked standard strain with an exotic
percentage of Arg' phenotypes among the progeny will be strain have provided the community with sets of progeny
approximately 25% (assuming viability among the four (maintained at the Fungal Genetics Stock Center) recom-
genotypes is equal). This will also be true of a cross between binant for numerous polymorphic sites, determined with
an Arg- strain of the genotype arg-1- arg-2- and a pro- DNA probes. These sites have been mapped with respect to
totroph (arg-1' arg-2+), the result differing quantitatively one another and with respect to standard genetic markers.
and diagnostically from the case of segregation of two alle- A DNA probe that recognizes a polymorphism between the
les of a single gene, above. If linkage prevails, the pro- parents can then be used to screen the progeny set. The
totroph percentage will be even lower, allowing one to cal- progeny will each display one or the other parent's variant
culate the linkage distance. In both of these crosses, one when probed; distribution among the progeny will be
must double the percentage of prototrophs, because it must unique to that probe and other probes used to make the
be assumed that there are an equal number of double- RFLP map for that chromosomal region. The identity (or
mutant progeny among the Arg- auxotrophs. In the case of near identity) of the pattern revealed by the new probe is
independent assortment of arg-l and arg-2, 25% pro- thus determined by reference to the RFLP map.
totrophs is a maximum, because in either cross (mutant X Detailed RFLP maps are also available for other fungi,
mutant or double mutant x wild type) it indicates 50% re- e.g., Cochliobolus heterostrophus (49). Surprisingly little
combinant genotypes. DNA variation has been detected between wild A. nidulans
44. Microbiological and Genetic Methods for Filamentous Fungi 973

strains, but hybrids between related species can be used to pairs of allelic mutants in strains carrying outside markers
provide molecular markers (50). that are used to orient crossover events, effectively giving a
In fungi, centromeres can be mapped genetically without four-point map. Such crosses invariably demonstrate the
reference to cytology. For species forming vegetative phenomenon of gene conversion. This term was given to
diploids, this can be achieved by analysis of mitotic recom- nonreciprocal recombination events first detected depend-
bination (see section 44.5.3). In species such as Neurospora, ably in fungi (see reference 25). The phenomenon can be
meiosis distributes the first-division products cleanly to the most clearly demonstrated by means of tetrad analysis, in
upper and lower halves of the ascus. Examination of the which one isolates an entire ascus and observes or dissects
resulting eight-spored ascus demonstrates this as a first- out the ascospores. For instance, in Sordaria spp. segregation
division segregation (Fig. 3). However, a crossover be- patterns of viable ascospore color mutants can be analyzed
tween the marker gene and the centromere is detected by by visual inspection (Fig. 3). Gene conversion in its sim-
the formation of a second division segregation ascus. In plest form is seen as a 6:2 or a 5:3 ratio of alleles, rather than
such asci, both spore phenotypes appear in both the top and the orthodox 4:4expected in normal segregation. The term
bottom halves of the ascus (Fig. 3). Since each ascus con- gene conversion derives from the impression that one of the
tains the products of a single meiosis, the frequency of cross- alleles had converted the other one during pairing at
ing over in the interval between a scorable marker and its meiosis.
centromere can be measured readily. Podospora differs here, Intragenic recombination is normally a rare event, so re-
owing to the overlap of spindles at the second meiotic divi- combinants are usually obtained by selective plating for
sion, and this must be taken into account (25). wild types. In such an experiment, conversion is detected as
Crosses may also be used in allele testing: if two muta- an event that replaces a mutant site with the wild-type ver-
tions are unlinked, they must be at different loci, while if sion, but without crossing over of outside markers (Fig. 4).
they are alleles of the same gene, they will be completely In most crosses between two strains with different muta-
linked except in cases where intragenic crossing over might tions in the same gene, conversion at one site is usually
take place. Because lack of observable recombination is not more common than the other, and if enough mutational
a secure criterion of allelism, complementation tests should sites are studied, a gradient of conversion frequency will be
be done to decide the matter. Intragenic crosses, i.e., those found along the gene; this is termed conversion polarity. In
used to map sites within a gene, are only practicable in mi- A. nidulans, intragenic crossing over is sufficiently frequent
croorganisms, including fungi. Such crosses usually employ to allow ordering of the sites by examination of outside

t
A = Black
-a a = White
-a

First-division Second-division Gene conversion


segregation segregation (6:2or 3:5)
FIGURE 3 Meiotic tetrads of various types. In first-division segregation, no crossover occurs be-
tween the gene A/u and the centromere of the chromosome. Segregation of centromeres (arrows)
places all copies of the A allele in one half and all copies of the a allele in the other half. In second-
division segregation, crossing over exchanges the distal parts of the two central chromatids; segre-
gation of A and a alleles is delayed until the chromatids (lines) separate at the second meiotic di-
vision. The patterns shown are all equally probable, but in all cases, both alleles of the A/a gene are
found in both halves of the ascus. In gene conversion, molecular interaction of chromatids leads to
replacement of information of one chromatid by transfer from the other. One or both DNA strands
of the recipient chromosome may be replaced, leading to 3:5 and 6:2 segregations, respectively.
Conversions may occur in either direction, from A to a (as shown in the 6:3 example) or from a to
A (as shown in the 3:5 example).
974 1 MYCOLOGY

arg- 1

ad-5+ a & csp-l+

ad-5 a + b csp-1

Orthodox crossing over: ad-5 arg-P csp-l+

Gene conversion a to a+: ad-5+ arg-P csp-I+

Gene conversion b to b+:ad-5arg-P csp-1


FIGURE 4 Intragenic crossing over. Rare cases of intragenic crossing over must be detected by
selective means, such as plating ascospores of a cross between two auxotrophs on minimal medium.
Here, two different sites of mutation, a and b in the arg-1 gene, are shown. Prototrophic progeny
(arg-1) recovered in plating may arise in three ways. In the first, orthodox crossing over leads to
reconstruction of the wild-type allele. (In such a process, a double mutant will form as the recipro-
cal product.) The crossover leads to the recombination of the markers on either side, as shown. urg-
I prototrophs may, however, frequently form by gene conversion of either the u mutational site or
+

the b mutational site without any recombination of the flanking markers.

markers (45). For instance, if in the equivalent of the cross many other fungi (e.g., references 2, 28, and 40). So far, the
shown in Fig. 4, the most common crossover genotype was RIP process has been used as a mutagenic system only in N.
ad-5 arg csp, it would be concluded that the b site was to crassa.
the right of the a site. However, in many fungi, including N. Extranuclear variants, such as the respiratory deficiency
crassa, almost all intragenic recombinants arise by conver- caused by the mitochondria1 boky] mutation, have a dis-
sion, and sites can only be ordered in terms of the conver- tinct pattern of inheritance. First, they are rarely or never
sion gradient, e.g., if in a cross of a x b, site a is converted transmitted by the male (conidial) parent in sexual crosses.
more frequently than site b, then a is at the higher end of Second, the ascospore progeny display a lack of segregation
the polarity gradient. Descriptions of the molecular events for the variant; they all have the phenotype of the maternal
underlying gene conversion and its association with meiotic parent. In A. nidulans, heterokaryon-incompatible strains
recombination may be found in most sophisticated genetics have been used to show that mitochondria are inherited, as
texts. in other fungi, from the parent taking the female role in the
N. crussa offers a novel mutagenic technique based on cross, the latter being identified by maternally determined
the RIP phenomenon (48). As mentioned above (see sec- cleistothecial pigment markers (47).
tion 44.5.1), N. crussa DNA fragments or fragments carried
by plasmids integrate most frequently into nonhomologous 44.5.4.2. Procedures for Particular Fungi
(ectopic) locations of the genome, creating a duplication
(or higher multiple) of the resident gene(s). Once purified, 44.5.4.2.1. N. crmsa (20, 21)
the behavior of the transformant in sexual crosses is irregu- Neurospora spp. shoot their spores from perithecia as
lar. During the several divisions of the transformant nucleus they develop, and these collect in culture tubes on the wall
just prior to nuclear fusion, both the exogenous (if more opposite the agar slant. They are collected no less than 21
than about 1 kb) and the resident copies of the gene or days after fertilization to ensure their maturity. This is done
DNA segment are mutated, often massively, by GC-to-AT by scraping a sample from the wall of the tube with a wet in-
transitions. The segments are often heavily methylated as oculation loop and suspending the spores in 0.5 ml of ster-
well. Therefore, the ascospore products of a cross of an ec- ile water in a culture tube. The tubes are heated to 60C for
topic transformant X wild type will include many that dis- 30 min and cooled. The heating activates the ascospores
play the mutant phenotype for the gene in question. If vi- and kills any conidia that may have been collected with
able, the mutants may then be studied physiologically them. The activated spores are plated on nonselective plat-
thereafter, although screening the mutant ascospore prog- ing medium (see section 44.2.2.1.1) if the ratios of all prog-
eny is desirable to recognize those in which the ectopic eny types are to be determined. (Selective platings may be
copy has been lost by recombination or independent assort- appropriate, particularly if percentages of rare recombinants
ment. If the mutant phenotype is inviable or unpredictable, are of interest.) Colonies are picked at a very early germling
more sophisticated means of recovering RIPped nuclei in stage (using a dissection microscope with substage illumina-
heterokaryons may have to be used. The sheltered RIP tion and a condenser) with a spear-point needle or similar
method devised by Harkness et al. (27) may be consulted device to individual tubes. This avoids colony overlap with
for details. The active mutagenic RIP process has been further growth and ensures that only one germling is being
demonstrated so far only in Neurospora and Podosporu (26), isolated each time. (The dark spore wall is a good indicator
although evidence of RIPped sequences have been found in of whether the germling is a single meiotic product.) When
44. Microbiological and Genetic Methods for Filamentous Fungi 975

mature, the cultures are spot tested in 5 X 5 arrays on petri after growth and anastomosis, to the formation of fruiting
dishes of spotting medium (see section 44.2.2.1.1). In spot bodies, which form basidiospores, the haploid products of
testing, a straight inoculation needle is used to stab very meiosis. Plating of the spores on suitable media is followed
small inocula to spots premarked on a dish. The inocula- by germination; the germlings can be picked and analyzed
tions are done as the dish remains inverted, to minimize genetically as a random population of meiotic products.
contamination. The results are scored after overnight With respect to analysis of vegetative cultures, Coprinus
growth at 32"C, with confirmation later in the day if nec- forms uninucleate oidia from monokaryons and chlamy-
essary. Different temperatures may be used if necessary or dospores from dikaryons; in both cases these are suitable for
convenient. plating and, in the former case, for resolving heterokaryons.
Schizophyllum does not form single-celled spores except via
44.5.4.2.2. A. nidulans meiosis. Therefore, macerates must be used to resolve het-
Sexual interaction is achieved by mixing conidia in a erokaryons and for work involving mutagenesis. More de-
small area on thick (40 ml per 9-cm dish) minimal agar and, tailed methods may be found in the articles in the volume
after germination, sealing the plates with cellophane tape by King (32).
to produce partial anaerobiosis (44). Crosses are normally
made between strains bearing complementing auxotrophies 44.5.5. Stock Management
and spore colors. Germination and fusion of the parent Any use of fungi requires good management of stocks, in
strains on minimal medium is encouraged by addition of a particular in genetic studies. Most stocks should be stored in
few drops of complete medium at the point of inoculation. a manner that minimizes growth or aging of cultures. Such
Cleistothecia develop within 10 days and are fully mature methods include freezing ( - 20"C), lyophilization, and sil-
in 3 weeks. By this stage the asci, which are very fragile, ica gel storage (for N. crassa, see reference 20). In this way,
have broken down, leaving up to 5 x lo5 free ascospores in the accumulation of spontaneous mutants in heterokaryotic
each cleistothecium. Ascospores germinate without activa- association with the wild type, or selective survival of mu-
tion, but high germination rates are only achieved with tants over wild type, during prolonged storage in the refrig-
spores from fully mature fruiting bodies. erator can be avoided. Stocks should have a formal designa-
A. nidulans is homothallic, so cleistothecia developing tion and recorded source, and this designation should be
from a mixed inoculum may result from a self-cross or from distinct from the name of any mutant loci in the strains.
a cross between the parents (hybrid cleistothecium). For The names of loci, in turn, should be distinct from the par-
this reason, strains used as parents should be marked with ticular allele of the locus used. Standard rules of nomencla-
conidial color mutants so that hybrid cleistothecia and their ture may be found at many of the websites (for links, see
contents can be recognized as such. Where analysis is by se- www.fgsc.net) and books cited and may be inferred from re-
lective plating, massed, uncleaned hybrid cleistothecia cent publications.
taken from the surface of the mycelium (Color Plate 18A)
can be crushed and the resulting ascospore suspension is Note: Although minor changes in this chapter have been made more
plated as desired. Alternatively, single cleistothecia are recently, no attempt has been made to incorporate the many rationales
cleaned of adhering conidia and Hulle cells by rolling on and techniques that have appeared since 2003. Readers should consult
relevant websites, reviews, and technical literature as they embark on
sterile agar (Color Plate 18B), then each is crushed in 10 ml the procedures described here.
of sterile water and the suspension is test-plated on one
dish, usually containing Triton X-100 (see section 44.5.1).
Hybrid cleistothecia can be identified by the mix of spore 44.6. REFERENCES
colors among the progeny, and suspensions from selected 1. Agger, T., A. B. Spohr, and J. Nielsen. 2001. [alpha]-
cleistothecia are then replated on a larger scale, usually Amylase production in high cell density submerged culti-
omitting the Triton X-100, which may select against some vation of Aspergillus oryzae and A. nidulans. Appl. Micro-
phenotypes. Occasionally, "twinned" cleistothecia are biol. Biotechnol. 55:81-84.
found; these contain a mixture of ascospores of hybrid and 2. Aleksenko, A., and A. J. Clutterbuck. 1996. The AMAl
selfed origin. plasmid replicator in Aspergillus nidulans is an inverted du-
Ascospore-derived colonies are classified by transferring plication of a low-copy-numberdispersed genomic repeat.
them individually to a master plate containing a 5 x 5 Mol. Microbiol. 19:565-5 74.
colony array, which is then replicated to test media using an 3. Anwar, M. M., J. H. Croft, and R. G. B. Dales. 1993.
appropriately spaced multipin replicator on a translucent Analysis of heterokaryon incompatibility between het-
plastic base (Color Plate 18). Master plates should be erokaryon-compatibilty (h-c) groups R and GL provides
poured some hours before inoculation, or else the open, in- evidence that at least eight het loci control somatic in-
verted plates may be briefly dried at 37C. Inoculation of compatibility in Aspergillus nidulans.j . Gen. Microbiol. 139:
master plates should be done in still air, holding the recipi- 1599-1 603.
ent plate inverted so that stray spores fall away from it. 4. Axelrod, D. E. 1972. Kinetics of differentiation of conid-
Similarly, when replicating from a master plate, place it, iophores and conidia by colonies of Aspergillus nidulans.
j. Gen. Microbiol. 73:181-184.
open, on the bench, stab the multipin replicator down onto 5. Bal J., E. M. Kajtaniak, and N. J.Pieniazek. 1977. 4-
it, then invert the replicator and bring recipient plates, in Nitroquinoline-1-oxide: a good mutagen for Aspergillus
turn, down onto its pins. nidulans. Mutat. Res. 56:153-156.
6. Basabe, J. R., C. A. Lee, and R. L. Weiss. 1979. Enzyme
44.5.4.2.3. Coprinus and Schizophyllum assays using permeabilized cells of Neurospora. Anal. Bio-
The tetrapolar basidiomycetes Coprinus and Schizo- chem . 92:356-3 60.
phyllum require the recognition and use of monokaryotic 7. Bennett, J. W., and L. L. Lasure (ed.). 1985. Gene
mycelia of compatible mating type (differences at the un- Manipulations in Fungi. Academic Press, Inc., Orlando, FL.
linked A and B loci) before mating. Confrontation of 8. Bennett, J. W., and L. L. Lasure (ed.). 1991. More Gene
monokaryons on complete medium in petri dishes leads, Manipulations in Fungi. Academic Press, Inc., San Diego, CA.
976 MYCOLOGY

9. Borkovich, K. A., L. A. Alex, 0. Yarden, M. Freitag, 27. Harkness, T. A. A,, R. L. Metzenberg, H. Schneider, R.
G. E. Turner, N. D. Read, S. Seiler, D. Bell-Pedersen, J. Lill, W. Neupert, and F. E. Nargang. 1994. Inactivation
Paietta, N. Plesofsky, M. Plamann, M. Goodrich- of the Neurospora crassa gene encoding the mitochondria1
Tanrikulu, U. Schulte, G. Mannhaupt, F. E. Nargang, protein import receptor MOM19 by the technique of
A. Radford, C. Selitrennikoff, J. E. Galagan, J. C. sheltered RIP. Genetics 136:107-1 18.
Dunlap, J. J. Loros, D. Catcheside, H. Inoue, R. 28. Hua-Van, A., F. Hericourt, P. Capy, M. J. Daboussie,
Aramayo, M. Polymenis, E. U. Selker, M. S. Sachs, and T. Langin. 1998. Three highly divergent subfamilies
G. A. Marzluf, I. Paulsen, R. Davis, D. J. Ebbole, A. of the impala transposable element coexist in the genome
Zelter, E. R. Kalkman, R. ORourke, F. Bowring, J. of the fungus Fusarium oxysporium. Mol. Gen. Genet.
Yeadon, C. Ishii, K. Suzuki, W. Sakai, and R. Pratt. 259:354-362.
2004. Lessons from the genome sequence of Neurospora 29. Jung, M. K., N. Ovechkina, C . E. Oakley, and B.
crassa: tracing the path from genomic blueprint to multi- Oakley. 2000. The use of beta-D-ghcanase as a substitute
cellular organism. Microbiol. Mol. Biol. Reu. 68:l-108. for Novozyme 234 in immunofluorescenceand protoplast-
10. Bos, C. J. (ed.). 1996. Fungal Genetics, Principles and ing. Fungal Genet. Newsl. 47:65-66.
Practice. Marcel Dekker, New York, NY. 30. Kafer, E. 1961. The processes of spontaneous recombina-
11. Brown, T. A. 1993. Mitochondria1 genome of Aspergillus tion in vegetative nuclei of Aspergillus nidulans. Genetics 46:
nidulans, p. 3.85-3.86. In S. J. OBrien (ed.). Genetic Maps, 1581-1609.
6th ed. Cold Spring Harbor Press, New York, NY. 31. Kafer, E. 1977. Meiotic and mitotic recombination in
12. Brown, T. A. 1994. Mitochondria, p. 505-524. In S. D. Aspergillus and its chromosomal aberrations. Adw. Genet.
Martinelli and J. R. Kinghorn (ed.), Aspergillus 50 Years 19:33-131.
On. Elsevier, Amsterdam, The Netherlands. 32. King, R. C. (ed.). 1974. Handbook of Genetics, vol. 1.
13. Brown, J. S., A. Aufauvre-Brown, and D. W. Holden. Bacteria, Bacteriophages and Fungi. Plenum Press, New
1998. Insertional mutagenesis of Aspergillus fumigatus. Mol. York, NY.
Gen. Genet. 259:327-335. 33. Li Destri Nicosia, M. G., C. Brocard-Masson, S.
14. Champe, S. P., D. L. Nagle, and L. N. Yager. 1994. Demais, A. H. Van, M.-J. Daboussi, and C. Scazzocchio.
Sexual sporulation, p. 429-454. In S. D. Martinelli and 2001. Heterologous transposition in Aspergillus nidulans.
J. R. Kinghorn (ed.), Aspergillus SO Years On. Elsevier, Mol. Microbiol. 39:1330-1344.
Amsterdam, The Netherlands. 34. Marhoul, J. F., and T. H. Adams. 1996. AspergillusfabM
15. Clutterbuck, A. J. 1974. Aspergillus nidulans, p. 447-510. encodes an essential product that is related to poly(A)-
In R. C. King (ed.), Handbook of Genetics, 1. Bacteria, binding proteins and activates development when over.
Bacteriophages and Fungi. Plenum Press, New York, NY. expressed. Genetics 144:1463-1470.
16. Clutterbuck, A. J. 1995. Genetics of fungi, p. 239-253. In 35. Martinelli, S. D. 1976. Conidiation of Aspergillus nidulans
N. A. R. Gow and G. M. Gadd, (ed.),The Growing Fungus. in submerged culture. Trans. Br. Mycol. SOC.67:121-128.
Chapman and Hall, London, United Kingdom. 36. Martinelli, S. D., and J. R. Kinghorn (ed.). 1994. Asper-
17. Cove, D. J. 1966. The induction and repression of nitrate gillus: 50 Years On. Elsevier,Amsterdam, The Netherlands.
reductase in the fungus Aspergillus nidulans. Biochim. 37. McCully, K. S., and E. Forbes. 1965. The use of master
Biophys. Acta 11361-56. strains of Aspergillus nidulans for assigning genes to linkage
18. Cramer, C. L., J. L. Ristow, T. J. Paulus, and R. H. groups. Genet. Res. 6:352-359.
Davis. 1983. Methods for mycelial breakeage and isolation 38. Miller, B. L., K. Y. Miller, and W. E. Timberlake. 1985.
of mitochondria and vacuoles of Neurospora. Anal. Bio- Direct and indirect gene replacements in Aspergillus nidu-
chem. 128:384-392. lans. Mol. Cell. Biol. 5:1714-1721.
19. Cummings, W. J., M. Celerin, J. Crodian, L. K. Brunick, 39. Mullins, E. D., X. Chen, P. Romaine, R. Raina, D. M.
and M. E. Zolan. 1999. Insertional mutagenesis in Co- Geiser, and S. Kang. 2001. Agrobacterium-mediated trans-
prinus cinereus: use of a dominant selectable marker to gen- formation of Fusarium oxysporium: an efficient tool for in-
erate tagged, sporulation-defective mutants. Cum. Genet. sertional mutagenesis and gene transfer. Phytopathology 9 1:
39:371-382. 173-180.
20. Davis, R. H. 2000. Neurospora: Contributions ofa Model Or- 40. Nakayashiki, H., N. Nishimoto, K. Ikeda, Y. Tosa, and
ganism. Oxford University Press, Oxford, United Kingdom. S. Mayama. 1999. Degenerate MAGGY elements in a
21. Davis, R. H., and F. J. de Serres. 1970. Genetic and mi- subgroup of Pyricularia grisea: a possible example of suc-
crobiological research techniques for Neurospora. Methods cessful capture of a genetic invader by a fungal genome.
Enzymol. 17A:79-143. Mol. Gen. Genet. 261:958-966.
22. Dunn-Coleman, N. S., E. A. Brodie, G. L. Carter, and 41. Ninomiya, Y., K. Suzuki, C. Ishii, and H. Inoue. 2004.
G. L. Armstrong. 1992. Stability of recombinant strains Highly efficient gene replacements in Neurospora strains
under fermentation conditions, p. 152-174. In J. R. deficient for nonhomologous end-joining. Proc. Natl.
Kinghorn and G. Turner (ed.), Applied Molecular Genetics Acud. Sci. USA 101:12248-12253.
of Filamentous Fungi. Chapman & Hall, London, United 42. Perkins, D. D., and R. H. Davis. 2000. Neurospora at the
Kingdom. millennium. Fungal Genet. Biol. 31:153-167.
23. Ebbole, D., and M. S. Sachs. 1990. A rapid and simple 43. Perkins, D. D., A. Radford, and M. S. Sachs. 2001. The
method for isolation of Neurospora CI(ISSU homokaryons Neurosporu Compendium. Chromosomal Loci. Academic
using microconidia. Fungal Genet. Newsl. 37:17-18. Press, New York, NY.
24. Esser, K. 1974. Podospora anserina, p. 531-551. In R. C. 44. Pontecorvo, G., J. A. Roper, L. M. Hemmons, K. D.
King (ed.), Handbook of Genetics, vol. 1. Bacteria, Bacter- MacDonald, and A. W. J. Bufton. 1953. The genetics of
iophages and Fungi. Plenum Press, New York, NY. Aspergillus nidulans. Adw. Genet. 5:141-238.
25. Fincham, J. R. S., P. R. Day, and A. Radford. 1979. 45. Pritchard, R. H. 1955. the linear arrangement of a series
Fungal Genetics. Univ. of California Press, Berkeley, CA. of alleles in Aspergillus nidulans. Heredity 9:343-371.
26. Graia, F., 0. Lesprinet, B. Rimbault, M. Deqard- 46. Raper, J. R., and R. M. Hoffman. 1974. Schizophyllum
Chablat, E. Coppin, and M. Picard. 2001. Genome qual- commune, p. 597-626. In R. C. King (ed.), Handbook of
ity control: RIP (repeat-induced point mutation) comes to Genetics, vol. 1. Bacteria, Bacteriophages and Fungi. Plenum
Podospora. Mol. Microbiol. 40586-595. Press, New York, NY.
44. Microbiological and Genetic Methods for Filamentous Fungi m 977

47. Rowlands, R. T., and G. Turner. 1976. Maternal inheri- mosomes, using chromosome-suhstituted segregints of hy-
tance of extranuclear markers in Aspergillus niduluns. brid A. nidulans and A. qwdrilineutus. Cum.Genet. 25:
Genet. Res. 285:281-290. 3 11-3 17.
48. Singer, M. J., and E. U. Selker. 1995. Genetic and epige- 51. Villalba, F., M.-H. Lebrun, A. Hua-Van, M.-J. Daboussi,
netic inactivation of repetitive sequences in Neurosporu and M.-C. Grosjean-Cournoyer. 2001. Transposon impala,
crassa: RIP, D N A methylation and Quelling. Curr. Top. a novel tool for gene tagging in the rice blast fungus Mag-
Microbiol. Immunol. 197: 165-177. nuporthe grisea. Mol. Plant-Microbe Interact. 14:308-3 15.
49. Tzeng, T.-H., L. K. Lyngholm, C. F. Ford, and C. R. 52. Waring, R. B., and C. Scazzocchio. 1983. Mitochondria1
Bronson. 1992. A restriction fragment length polymor- four-point crosses in Aspergillus niduluns: mapping of a sup-
phism map and electrophoretic karyotype of the fungal pressor of a mitochondrially inherited cold-sensitive muta-
maize pathogen Cochliobolus heterostrophus. Genetics 130: tion. Genetics 103:297-311.
81-96. 53. White, B., and D. 0. Woodward. 1995. A simple method
50. Varga, J., and J. H. Croft. 1994. Assignment of RFLP, for making disposable race tubes. Fungal Genet. Neeuslet.
KAPD and isozyme markers to Aspergillus nidulans chro- 42:79.
45
Principles and Practice of DNA Microarray
TechnoIogy
KRISHNAMURTHY NATARAJAN. MATTHEW J . MARTON.
AND ALAN G . HINNEBUSCH

45.1. INTRODUCTION ......................................... 978


.
2. MICROARRAY TECHNOLOGY .............................. 979
........................ 979
45.2.1. Microarray Design and Fabrication
. ................................. 980
2. Growth of Yeast Cells
. ...................................... 981
3. Cell Harvesting
. .......... 981
4. Preparation of Total RNA by the Hot-Phenol Method
............. 982
45.2.4.1. Purification and Concentration of RNA
45.2.5. Isolation of Poly(A) ' RNA............................. 982
. ................... 982
6. Synthesis of Fluorescently Labeled cDNA
45.2.6.1. Direct Fluorescent Dye Labeling during Reverse
.................................
Transcription 983
. .................. 983
2. Purification of the Labeled cDNA
. 3. Two-step Labeling of cDNA Using Aminoallyl-dUTP
........................ 983
and Coupling to Cy Dyes
................. 984
45.2.7. Hybridization and Processing of Microarrays
. ....................... 984
8. Image Processing and Data Analysis
............... 984
45.2.8.1. Feature Extraction and Quantification
. ............................ 985
2. Data Normalization
. 3. Statistical Tools Accelerate the Identification of True
............................ 986
Expression Changes
. 4. Microarray Data Reproducibility. Standards. and
................................... 987
Databases
45.3. APPLICATIONS .......................................... 987
..... 987
45.3.1. Uncovering Genetic Circuits Governing Metabolic Pathways
.2. Unsupervised Identification of Gene Function: a
Compendium Approach ................................ 988
. .................................. 988
3. Translational Arrays
.4. mRNA Splicing Arrays ................................ 988
.
.5 Fitness Testing in Competitive Growth Assays To Identify Gene
.............................. 989
Function and Drug Targets
.6. Use of Intergenic DNA Microarrays To Detect Protein-DNA
......................................... 989
Interactions
.7. Comparative Genome Hybridization....................... 989
.8. Integration of Technologies for Comprehensive Assessment
of Genome Function .................................. 990
45.4. REFERENCES ............................................ 991

45.1. INTRODUCTION no known homologs in other organisms. The postgenome


The availability of whole-genome sequences has made it era has heralded new technologies that alleviate these diffi-
possible to identify the full complement of genes and begin culties and accelerate gene discovery and functional analy-
a comprehensive assignment of gene functions . However. sis.Valuable clues about the function of the uncharacterized
there are serious limitations to functional analysis of se- genes can be obtained using microarray technology to assess
quenced genomes. Several organisms are not amenable to expression of the entire genome under numerous experi-
genetic analysis. and mutant construction and phenotype mental conditions in parallel . Microarray technology has
analysis cannot be carried out routinely. Even for geneti- evolved rapidly. and indeed this powerful technology is not
cally tractable organisms. such as budding yeasts. reliable intimidating and can now be established in most laborato-
and sensitive assays are not available for numerous biologi- ries. This has been possible in large part due to the ready
cal processes and pathways. Thus. one-third of the yeast availability of reagents in the form of kits. prearrayed
genome has not been characterized experimentally and no whole-genome microarray slides. data analysis software. and
function can be ascribed to this large gene set. as there are instrumentation from several vendors .

978
45. DNA Microarray Technology 979

In this chapter we describe the principles and detailed limitation of the arrays made from expressed sequence tag
methodology for carrying out microarray experiments using and genomic DNA libraries is probe redundancy, such that
the yeast Succharomyces cereuisiae. These protocols can he complete genome coverage requires a severalfold increase in
adapted to other microorganisms, including different fungi, the number of clones that must he spotted to the microarray.
after suitable modifications to the protocols for growing, Two approaches are used for essing differential gene
harvesting, and breaking cells. Although microarray tech- expression using the microarrays. The short-oligonucleotide
nology is widely used to study the transcriptome and its dif- platform employs hybridization of a biotin-labeled cDNA or
ferential expression, this technology has also been applied cRNA derived from a single test RNA sample to a single
in novel ways to study various aspects of cell biology, and glass microarray. The hybridized target is then stained with
these innovations are reviewed briefly in the latter part of phycoerythrin (43), referred to as single-color hybridiza-
this chapter. tion, and the intensity of the signal provides a direct esti-
mate of the concentration of the transcript. The reference
RNA sample is treated similarly and hybridized to a dupli-
45.2. MIC ROARRAY TECH N 0LOGY cate microarray. The ratio between the fluorescence inten-
Central to the microarray technology is probe-target hy- sities emanating from experimental and the reference target
bridization as practiced in Southern and Northern blot hy- hybridizations is calculated for each feature on the micro-
bridization experiments. A microarray or a chip contains array.
thousands of cDNAs or oligonucleotides printed on a sub- In this chapter, we provide detailed methodology for ex-
strate glass slide in a high-density array with defined x-y co- pression profiling using the cDNA platform and dual-color
ordinates. I t is important to note that in the microarray hybridization. The two RNA samples, one derived from ex-
nomenclature, immobilized DNA on a glass slide is the perimental RNA and the other from the control RNA sam-
probe on the microarray, and fluorescently labeled cDNA or ple, are converted to cDNA and labeled individually using
cRNA synthesized from total or poly(A)+ RNA is the tar- fluorescent dyes such as cyanine 5 (Cy5) and cyanine 3
get nucleic acid. (Cy3). Next, the two labeled samples (targets) are com-
Several microarray platforms are available, including the bined in equal proportions and hybridized to a single micro-
short oligonuecleotide chips from the company Affymetrix, array slide (Fig. l ) , therefore referred to as dual-color hy-
spotted cDNA, spotted long oligonucleotides (50-to 60-mer bridization, involving competitive hybridization, i.e.,
sequences), and in situ-synthesized long oligonucleotides simultaneous hybridization of the reference and experimen-
using either an ink-jet technology (26) from the company tal targets to a single microarray (12, 60). The fluorescent
Agilent o r a maskless photodeprotection technology (65) signal emanating from each feature on the microarray slide
from the company Nimblegen. In this chapter, we discuss is quantified by scanning the slide in a microarray scanner.
some of the most commonly used microarray platforms. The The ratio of the fluorescence intensities emanating from
Affymetrix microarray platform is made of 25-mer oligonu- the experimental versus control samples is then subjected to
cleotides (43) and contains perfect-match probes and corre- extensive statistical analyses to obtain the relative expres-
sponding mismatched probes for each transcript. Several sion level of each gene on the microarray (Fig. 1).
such pairs of oligonucleotides spanning each transcript are
used, with the perfect-match probe being used as a reporter 45.2.1. Microarray Design and Fabrication
and the mismatch probe used as a control for hybridization Microarray technology is exquisitely sensitive such that
specificity (43). The cDNA microarray platform contains often changes in gene expression as small as 1.2-fold can be
PCR-amplified DNA representing either the complete open reliably detected. Therefore, it is very important to devote
reading frame (ORF) or relatively large portions of the ORF, careful attention to the experimental design in order to
e.g., 500 to 600 bp from the 3' end of each ORE Microarrays minimize systematic noise einanating from cultures and
containing 50-to 70-mer oligonucleotides representing each handling of samples. For instance, replicate hybridizations
ORF are increasingly favored because cumbersome PCR am- exhibited smaller-magnitude fluctuation than the biological
plification steps are not required and ready-to-spot oligonu- variability of separate cultures (see reference 48, Fig. 1). I t
cleotides can he custom-synthesized or purchased, albeit at a has also been observed that two seemingly identical wild-
high cost. The PCR-amplified ORF probes suffer from de- type cultures with no treatments and subjected to identical
fects due to cross-hybridization between homologous or or- medium and growth conditions displayed small hut signifi-
thologous genes, whereas the long oligonucleotide probes cant differences in transcript levcls of certain genes (see ref-
can he designed to minimize such cross-hybridization and erence 27, Fig. 2). In addition, IleRisi et al. showed that rel-
therefore have higher selectivity (26). More recently, high- atively small differences in cell densities can have dramatic
density long-oligonucleotide microarrays have been devel- consequences, such as in expression of genes involved in
oped with up to 385,000 oligonucleotide features synthesized sugar metabolism (12). Therefore, experiments have to be
in situ on glass slides. High-density genomic tiling micro- devised carefully when comparing strains that have differ-
arrays cover a complete genome or a large fraction of it with ent growth rates or when comparing different culture con-
densely tiled oligonucleotide probes. These arrays are used ditions. Although the procedures below may seem excessive
for interrogation of transcriptome profiles, DNA-protein to those unfamiliar with the extreme sensitivity of microar-
binding (section 45.3.6), and comparative genome hy- rays, they are designed to minimize variability of results ob-
bridization (section 45.3.7). Of course, the use of oligonu- tained in nominally identical replicate experiments. There
cleotide arrays is limited to sequenced genomes. In cases are several other important considerations for optimal de-
where genome sequence is not available, DNA amplified sign of microarray experiments, including adequate techni-
from expressed sequence tag libraries (5) can be spotted to cal and biological replicition of experiments and correct
generate microarrays. Moreover, for smaller fungal genomes choice of pair of samples for hybridization, and they are dis-
such as that of Aspergillus terreus, it has been possible to use cussed in detail elsewhere (9, 78).
a short restriction fragment library (average size, -2 kb) as A critical issue with the microarray technology is the
probes deposited on glass microarrays (3).However, a major choice of experimental controls. Both positive and negative
980 MYCOLOGY

RNA/poly(A)+ RNA

Fabrication of Micromay
c
Experimental
i
Control
on glass slides
c c
I
Hybiidimtion of CyS-Ky3-cDNA
Cy5-labeled

10
cDNA

\ J
Cy3-labeled
cDNA

probes on microarray slide

Rcpcat hybridi7ation to rcproduce protilcs


FIGURE 1 Overview of DNA microarray scheme

hybridization controls have to be carefully chosen ahead of rayer are described a t Pat Brown's lab website (http:l/
time and printed alongside the probe spots during fahrica- cmgm.stanford.edu/pbrown/mguide/index.htinl). For our
tion of the microarrays. Positive controls on the microarray studies, a custom-built spotter with 16 quill pins was iised to
represent housekeeping genes and often serve as normaliza- deposit PCR products corresponding to >6,000complete
L
tion controls (see also section 45.2.8.2). Typically, negative ORFs from the S. cerevisiae genome within a 2- by 2-cm
controls are either synthetic artificial sequences or se- area o n standard laboratory glass slides treated with poly-L-
quences derived from heterologous organisms. Thus, a yeast lysine. T h e spotted slides were postprocessed using standard
microarray could contain control spots representing plant protocols (reference 5 and http://cmgm.stanford.edu/
or vertebrate genes with no sequence homology to any yeast pbrown/protocols/index.html). However, to accelerate re-
genes. Randomized versions of oligonucleotide sequences search using microarrays, ready-to-use prearrayed microar-
derived from the same organism may also serve a s negative ray slides containing PCR-amplified DNA or synthetic long
controls in the oligonucleotide microarrays. oligonucleotides can be purchased from several companies
T h e PCR-amplified DNA or oligonucleotides can be de- or from nonprofit microarray facilities worldwide.
posited on glass slides by robotic spotter instruments. These
can be fabricated in-house, as pioneered i n the Pat Brown 45.2.2. Growth ,of Yeast Cells
laboratory, or purchased from commercial sources. A de- If the microarray experiment involves comparison of two
tailed comparison of the features of the commercial spotter different strains, both experimental and control strains
instruments can be found elsewhere (25). Details pertaining should be revived in parallel from a -80C freezer stock.
to various aspects of the fabrication of a robotic microar- T h e strains can be stored on plates for up to 1 week a t 4C.
45. DNA Microarray Technology 981

For strains harboring growth defects, it is advisable to de- apply vacuum for one additional minute, and then place the
termine the generation time prior to actual experiments. It membrane filter in a polypropylene conical centrifuge tube,
is important to also note that the apparent optical density flash-freeze it briefly in liquid nitrogen, and store at -80C
at 600 nm (OD,,) and cell density (cells per milliliter) can until RNA isolation.
be different for strains of different ploidy. In our laboratory, To harvest cultures by Centrifugation, transfer small-
1 OD6oounit corresponds to 2 X lo7 cells/ml for haploid scale cultures to 50-1n1conical tubes and immediately spin
wild-type S. cerevisiae BY4741. For each strain or condition at 3,000 to 4,000 x g in a tabletop swinging-bucket cen-
under examination, a control strain should be assigned in trifuge for 2 min or the minimum time required to pellet
advance that is processed strictly in parallel with the exper- cells. When the centrifuge has stopped, remove up to six
imental sample, from cell growth and harvesting to the hy- tubes at a time from the centrifuge and process them to
bridization steps. completion (three experimental-control pairs). For each
tube, decant the supernatant and invert on Parafilm or
1. Streak out strain(s) to obtain single colonies on a YPD paper towels and proceed to the next tube. After the first six
plate (1% yeast extract, 2% peptone, 2% glucose, 2% Bacto tubes have been drained, and while maintaining the in-
agar) or the most nutrient-rich tnedium permissible, and
verted orientation, flick each tube twice to remove any re-
grow for 2 days at 30C. maining liquid and re-cap the tube. Freeze six tubes at a
2. Resuspend a single colony of about a 3-mm diameter time by immersing the bottoms of the tubes in liquid nitro-
in 1 ml of synthetic complete (SC) medium (48).
gen for about 5 s. Immediately transfer the tubes to a rack
3. Inoculate 5 ml of SC medium with 50 pl of the resus- in the -80C freezer. Repeat for the next six tubes.
pended colony, and grow overnight for 10 to 14 h such that For large-scale cultures, harvest the cells by centrifuga-
this culture is still in logarithmic phase of growth. However,
tion in 250-ml bottles in the GSA rotor (in a Sorvall RCSC
for strains with growth defects, 250 pl of the resuspended
centrifuge) for 2 min at 7,000 rpm at ambient temperature.
colony suspension may be used to achieve higher initial cell
Decant the supernatant, place the bottle at a slight angle to
density such that the final cell densities of wild-type and
facilitate drainage of liquid from the pellet, and aspirate
mutants are comparable. residual liquid with a micropipette. Resuspend cells in 4 ml
4. Measure the ODhooof the overnight culture on the of AE buffer (see section 45.2.4), transfer to a 50-ml coni-
following day, and use only those cultures with an ODhooof
cal centrifuge tube and pellet cells as described above. The
less than 1.0 to inoculate cultures for microarray analysis. cell pellets can be stored frozen as described above or can be
The overnight cultures are diluted in 50 ml (for sinall scale) used immediately for RNA isolation.
or 250 ml (for large scale) of prewarmed SC medium to a
starting OD600 of 0.05to 0.1 and incubated in an air shaker
at 30C for 6 to 7 h at 220 rpm. Cells are harvested when 45.2.4. Preparation of Total RNA by the Hot-Phenol
cultures reach a final cell density of about 0.6 ODhoounit Method
(1.2 x lo7 cells/ml), as outlined in the next section. The following protocol is essentially derived from the
method described previously (35), and it relies on the prin-
45.2.3. Cell Harvesting ciple of selective fractionation of DNA into the phenol
phase and interphase at acidic pH while retaining RNA in
When cells are ready to be harvested, try to work as the aqueous phase. For isolation of RNA from actively bud-
quickly as possible to minimize the time during which any ding yeast cells, glass beads are not required in this protocol.
changes in gene expression may occur. Therefore, before However, for ascospore or mycelial cultures, 3.5 to 4 volumes
the cultures reach the required cell density, label the req-
of glass beads are added along with phenol. The following
uisite number of 50-ml conical tubes to collect cells and protocol is for 50-ml cultures. For each sample, combine
obtain liquid nitrogen for freezing the cell pellets. It is also
400 pl of AE buffer (50mM sodium acetate [pH 5.31, 10
critical to maintain the temperature for cell harvest as
mM EDTA, 1% sodium dodecyl sulfate [SDS])with 400 pl
close to the culture conditions as possible in order to min- of phenol equilibrated with sodium acetate buffer (50 mM
imize gene expression changes. Remove flasks from the
sodium acetate and 10 mM EDTA, adjusted to pH 5.3) in
shaker in pairs that will end up later in the same hy-
a set of microcentrifuge tubes. Prepare also a second set of
bridizations. The number of cultures to be processed in tubes with 400 pl of phenol-chloroform (1:l) mixture.
parallel will also depend on the type and capacity of the
Hold all tubes in a water bath at 65C.
centrifuge or the harvesting method to be adopted. We
provide two methods for cell harvesting. For experiments 1. To the cell pellets (in 50-ml tubes, either fresh or
involving time course studies, such as cell cycle analysis, frozen) immediately add 800 p1 of hot phenol-AE buffer
drug treatments, or temperature shifts, we highly recom- mixture, vortex briefly, and incubate in a 65C water bath.
mend the membrane filtration method for harvesting If cells were harvested by membrane filtration, wash the
cells. For a11 of our single-time-point experiments, we cells from the membrane with the 800 pl of hot phenol-AE
have harvested cells by centrifugation. buffer, rinsing three or four times, to completely strip the
To harvest cells by filtration, we use the Millipore membrane filter of all cells. Remove the filter from the tube
polyvinylidene difluoride membrane (0.45-pm pore size) in and discard. Incubate resuspended cells for 10 min at 65C.
a membrane filtration device (Millipore). Both the mem- Vortex for 30 s several times (typically three or four times)
brane filter and the filtration device are to be presterilized during the incubation period, and return tubes to 65C.
by autoclaving for 15 min. If multiple strains are to be han- Transfer contents from large tubes to new microcentrifuge
dled in parallel, it is helpful to use a multiple-filtration man- tubes. Vortex briefly to keep the cell-phenol mixture resus-
ifold, also available from Millipore. pended.
After the cultures reach the appropriate cell density or at 2 . Cool down tubes to room temperature by placing in
defined time intervals, cells are collected by quickly passing an ice-water bath for 2 min. Avoid cooling the tubes below
each culture through a membrane filter connected to a suit- 15"C, as this will lead to precipitation of SDS. Spin in a mi-
able vacuum source. When all of the liquid is removed, crocentrifuge (16,000 X g) for 10 min.
982 MYCOLOGY

3. Transfer aqueous supernatant to the microcentrifuge of total RNA may be saved at this stage to examine by gel
tube containing 400 pl of phenol-chloroform mixture (pre- electrophoresis.
warmed to 65C). Vortex for 10 s, and spin in the micro- 3. Add an equal volume (40 11.) of 2 X column loading
centrifuge for 10 min. Collect aqueous supernatant and buffer to the RNA sample prepared as described in section
process as described in section 45.2.4.1. 45.2.4.1, and apply to the column by gravity flow. When all
of the liquid has entered the column, wash the column with
45.2.4.1 . Purification and Concentration of RNA 1 column volume of I X loading buffer by gravity flow.
Collect flowthrough and reload on the column two addi-
The following procedure is employed to remove residual
tional times. Prior to each reloading, reheat the samples at
phenol-chloroform by repeated exchange with 10 mM Tris-
65C for 5 min, followed by cooling for 1 min on ice, to im-
HCI-1 mM EDTA (TE) buffer (pH 7.5) and to concentrate
prove the yield.
the RNA without the need for an ethanol precipitation
step.
4. Wash with 8 ml of l x column loading buffer.
Discard flowthrough.
1. Transfer aqueous supernatant directly to a prewetted 5. Wash column with 2 column volumes of middle-
Microcon YM-30 concentrator. Spin in a microcentrifuge wash buffer (20 mM Tris-HC1 [pH 7.61, 150 mM NaCl, 1
(16,000 X g) for 20 min, or until the volume is reduced to mM EDTA, 0.1% sodium lauryl sarcosine) and allow buffer
< l o 0 pl. The volume remaining in the filter tube can be to drain from the column completely.
estimated by monitoring the level of the liquid above the 6. Elute poly(A)+ RNA with 1.2 ml of elution buffer
white section of the Microcon filter. For reference, use a fil- (10 mM Tris-HC1 [pH 7.61, 0.1 mM EDTA, 0.005% sodium
ter tube containing 100 pl of water. If the volume is not re- lauryl sarcosine), preheated to 65"C, by adding 400 pl of
duced to <lo0 pl after 20 min, the filter is probably elution buffer to the column in three aliquots and collect-
clogged. In that event, transfer the contents to a new ing in the same tube. The resulting poly(A)+ RNA prepa-
Microcon filter. Because most of the RNA will be in a ge- ration is then purified on the same oligo(dT) column for a
latinous clump, be sure to resuspend it by repeated pipetting second time as follows to nrovide a substantial enrichment
before transferring it to a new filter tube. for mRNA.
2. Add 400 pl of TE buffer to the Microcon concentra- 7. Wash the column with 4 ml of DEPC-treated water
tor and spin for 15 min as described above. The volume and with 2 to 4 ml of l x loading buffer. Meanwhile, the
should now be -20 to 50 pl. eluate from the first purification is heat denatured as in step
3. Repeat step 2 twice and concentrate the sample to 2. Mix with an equal volume of 2X loading buffer and load
-20 to 30 pl. At this point the samples should be devoid of as in step 3. Collect flowthrough and reload a s in step 3.
phenol. Remember to heat samples as in step 2 except for cooling on
4. Invert the Microcon concentrator into a microcen- ice for 2 min. Wash with 5 ml of 1x loading buffer, followed
trifuge tube and spin for 10 s at full speed to collect the by washing with middle-wash buffer as in step 5.
RNA. If samples were divided into different Microcon 8. Elute mRNA with two 400-pl aliquots of elution
tubes, collect them all in the same tube. Estimate the vol- buffer in separate microcentrifuge tubes. To each tube of
ume recovered and adjust it to about 40 p1 with TE buffer eluate, add 44 pl of 3 M sodium acetate (pH 5.2), 4 pl of
and mix well. 2 . 5 - p g / ~linear
l acrylamide, and 1.1 ml of ethanol and mix
5. The concentration of RNA may be estimated by mea- well. Store at -80C for at least 30 min. Note: Linear acryl-
suring the ODL60(58). The sample will be viscous but can amide is used as a neutral coprecipitating agent (18) and
be pipetted accurately after brief heating to 65C. At this can be prepared in the laboratory (5) or purchased
stage, the RNA can be immediately carried to the labeling (Ambion).
step or stored at -80C. The RNA yield should be 400 to 9. Spin for 60 min in a cold microcentrifuge at top
500 pg from 50 ml of cell culture. speed, and aspirate the supernatant carefully.
10. Wash pellet with 75% ethanol as follows. Add 500
45.2.5. Isolation of Poly(A)+ RNA pl of cold (-20C) 75% ethanol, being careful not to dis-
lodge the pellet. Aspirate the supernatant and discard.
For studies with S. cerevisiue, total RNA can be used directly
Remove the residual liquid with a micropipette tip and
as a template for cDNA synthesis. However, additional pu-
allow to air dry for 5 to 10 min. Resuspend in 10 pl of
-'
rification steps during p l y ( A) RNA isolation have the
DEPC-treated water or 2 X TE buffer (20 mM Tris [pH 7.61,
benefit of eliminating contaminants that inhibit cDNA
0.2 mM EDTA). To quantify by spectrophotometry, dilute 1
synthesis. In addition, random primers can be used for
cDNA synthesis from poly(A)+ RNA.
pl in 60 pl of TE buffer, and read in a low-volume (e.g., 50-
11.) cuvette.
1. Preparation of oligo(dT) columns. Weigh out 0.7 g of 11. Regeneration and storage of columns. Wash column
oligo(dT)-cellulose (New England Biolabs) (sufficient for with 4 to 6 ml of DEPC-treated water and store in 0.5ml of
12 to 14 0.25-in1 columns) and resuspend in 0.1 N NaOH. water at 4C. Columns can be used up to three times after
Mix on a rotary mixer such as the Nutator (Clay Adams) for being regenerated with 2 ml of 0.1 N NaOH, followed by
a few minutes. To a 0.8- by 4-cm sterile polypropylene col- equilibration in column loading buffer, as in step 1 above.
umn (such as from Bio-Rad), pack the slurry by gravity flow The typical yield of poly(A)+ RNA from S. cerevisiae is 1.5
until a 0.25-ml bed volume is achieved. Wash the column to 2.5% of total RNA.
with 4 ml of diethyl pyrocarbonate (DEPC)-treated water
(58),followed by washing with 2 to 4 ml of 1X column
loading buffer (20 mM Tris-HCI [pH 7.61, 0.5M NaC1, 1 45.2.6. Synthesis of Fluorescently Labeled cDNA
mM ELITA, 0.1% N-lauryl sarcosine, sodium salt), or until There are two methods for producing cDNA labeled with
the pH of the eluate is -8.0. fluorescent dyes. In the direct-labeling method, Cy3- or
2. To denature the RNA, heat sample at 65C for 5 Cy5-deoxynucleoside triphosphate (Cy5-dNTP) is incorpo-
min, followed by quick chilling on ice for 3 min. A n aliquot rated into cDNA during reverse transcription using total
45. DNA Microarray Technology 983

RNA or poly(A)+ as a template. If the biological material Typhoon). Successful labeling would yield a smear of la-
is in limiting amounts, one or two RNA amplification steps beled cDNA ranging from about 400 to at least 2,000 nu-
could be introduced as described by Van Gelder et al. (72), cleotides long. The concentration of cDNA and the
and the amplified RNA is reverse transcribed prior to label- amount of incorporated dye can be quantified by spec-
ing. The direct-labeling method is relatively simple and was trophotometry (5).
extensively used in microarray labeling. However, it is now
clear that there are several limitations with this method. 45.2.6.2. Purification of the Labeled cDNA
Because some reverse transcriptase enzymes are sluggish to Add 400 pl of TE buffer to the reservoir of a Microcon YM-
incorporate the bulky fluorescent dyes into cDNA, there is 30 concentrator (Amicon/Millipore). Transfer the Cy3 and
low fluorescence intensity of labeled cDNA molecules. Cy5 reaction products to separate Microcon filter tubes,
Another limitation is that Cy3-dNTP and Cy5-dNTP are and mix briefly by repeated pipetting up and down. Spin the
not incorporated with equal efficiencies by the reverse tran- tube in a microcentrifuge at full speed for 8 min or until the
scriptase. Therefore, dye-swap experiments are routinely volume is reduced to -20 pl. Add 400 pl of TE buffer again
practiced now, wherein the Cy dyes are interchanged be- and spin as described above to reduce the sample volume to
tween experimental and control RNA samples so that the -20 pl. Recover sample by centrifuging the filter tube in an
dye bias can be corrected by averaging the data from the inverted position into a sample collection tube. Store at
two independent hybridizations. Although this increases 4C in the dark, or proceed directly for hybridization.
the cost for microarray experiments, it is an effective way to
perform a replicate measurement while correcting a known 45.2.6.3. Two-step Labeling of cDNA Using
systematic bias. The second labeling method is a two-step Aminoallyl-dUTP and Coupling to Cy Dyes
indirect-labeling procedure in which aminoallyl-dUTP is
first incorporated into cDNA during reverse transcription 45.2.6.3.1. Reverse Transcription
and then the resulting primary amine groups of the amino
1. Prepare 12.2 pl of the reaction premix as follows:
ally1 residues are nonenzymatically conjugated to N-
hydroxysuccinimidyl (NHS)-esterified Cy3 or Cy5 (26). 6 pl of 5 X first-strand buffer (Superscript reverse tran-
This method overcomes both of the limitations associated scriptase kit)
with the direct-labeling method. The reagents for microar- 1.2 p1 of 25X dNTP-aminoallyl-dUTP labeling mix (a
ray target labeling are available in kit formulations from 12.5 mM concentration each of dATP, dCTP, and
several vendors. Indeed, the indirect-labeling method is dGTP; 10 mM aminoallyl-dUTP and 2.5 mM dTTP)
being adopted as the preferred method in several laborato- 3 pl of 0.1 M dithiothreitol
ries, despite being a more demanding technique due to the
additional labeling steps (see section 45.2.6.3.2). 2 p1 of Superscript I1 enzyme
2. Set up one reaction each for sample RNA and a sepa-
45.2.6.1. Direct Fluorescent Dye Labeling during rate reaction for control RNA. To anneal primer, mix l to
Reverse Transcription 4 pg of poly(A)+ RNA with 5 pg of oligo(dT)lo(2 p1 of 2.5
1. Prepare two reaction premixes, one for Cy3 and one pg/pl) in a total volume of 17.8 pl. Heat to 70C for 10
for Cy5. For n number of RNA samples, the size of the pre- min. Cool on ice for 10 min.
+
mix is 11.5 pl X (n 0.5).Each reaction premix of 11.5 p1 3. Combine the reaction premix with the RNA sample
(step l ) , mix well, and incubate the reaction mixture for 2
is prepared as follows:
h at 42C.
6 pl of 5 X first-strand buffer (Superscript I1 reverse tran- 4. To each reaction mixture, add 3 pl of 0.1 M EDTA.
scriptase kit) Mix thoroughly and add 3.5 p1 of 0.1 M NaOH. Incubate
0.5p1 of 50X dNTPs (10 mM dTTP and a 25 mM con- the reaction mixture for 10 min at 65C to hydrolyze the
centration each of dATP, dGTP, and dCTP) RNA.
3 pl of 0.1 M dithiothreitol 5. Neutralize the reaction mixture by the addition of
2 pl of Superscript I1 enzyme HEPES (pH 7.0) to a final concentration of 500 mM.
6. At this stage, the aminoallyl-cDNA is to be purified
2. For cnch RNA to be assayed, dispense 1 to 2 p g of from unincorporated aminoallyl-dUTP, hydrolyzed RNA,
poly(A)' RNA into two microcentrifuge tubes. Add 2 pl and free amines. For this purification, a Microcon YM-30
of 2.5-pg/pl oligo(dT)20and adjust the volume to 15.5 pl device is used essentially a s described above (section
with DEPC-treated water (58) for each tube. Incubate at 45.2.6.2), with a critical difference of using water instead of
70C for 2 min to denature the RNA, and chill on ice for TE in all steps. This is essential because free amines left in
10 min. the sample would interfere with the subsequent coupling
3. To one tube, add 11.5 pl of premix followed by 3 pl of -
step. The samples are concentrated to a volume of 10 pl.
Cy3-clUTP, mix, and incubate for 2 h at 42C. Prepare an
identical reaction mixture in the other tube using Cy5-
dUTP.
45.2.6.3.2. Nonenzymatic Covalent Coupling and
4. Stop the reaction by adding 10 pl of a solution of 0.5 Purification of cDNA
N NaOH-0.25 M EDTA. Mix well. Incubate at 65C for 20 The Cy5-NHS ester and Cy3-NHS ester postlabeling
to 30 min. Neutralize the reaction by adding 20 to 25 p1 of reactive dye packs are available from GE Healthcare (prod-
1 M Tris-CI (pH 7.5). uct no. RPN 5661). These are available in single-use
aliquots and therefore are inore stable and convenient.
5. The quality of labeled cDNA can be judged by resolv-
ing 1 to 2 p1 of the Cy5-labeled sample by electrophoresis 1. To the aminoallyl-UTP-labeled cDNA samples from
in a 2% agarose gel (prepared in l x TAE buffer) followed step 45.2.6.3.1, add 1.5 p1 of freshly made 1 M sodium bi-
by fluorescent imaging on a fluorescence scanner equipped carbonate (pH 9.0). Remove an alicluot each of NHS-Cy3
for 630-nm excitation and 650-nm emission (e.g., GE and NHS-Cy5, add the bicarbonate-buffered cDNA, and
984 w MYCOLOGY

resuspend the pellet vigorously. Gently but thoroughly mix M NaCl plus 0.015 M sodium citrate)-0.75-mg/ml poly(dA)
by repeated pipetting. Incubate for 1 h at ambient tempera- (GE Healthcare)-0.2% SDS for 12 to 16 h at 63C as follows.
ture in the dark and wrapped with foil, with intermittent
1.To dye-labeled cDNA (from sections 45.2.6.2 and
mixing by pipetting every 15 min.
45.2.6.3), add 4 pl of 20X SSC and 2 pl of lO-mg/ml poly
2. Stop the reaction by quenching the unreacted NHS- (dA) and make up volume to about 25 pl. Poly (dA) of 40
esterified Cy dye molecules by addition of 3.6 pI of 4 M hy-
droxylamine (Sigma) for a final concentration of 1.5 M.- to 60 dATP bases in length promotes specific hybridization
between the labeled cDNA and probe DNA on the chip by
Mix well and allow reaction to continue for 15 min in the
annealing to the poly(T) tract in the labeled cDNA. Note
dark.
that SDS is added only after the filtration step.
3. At this point, the efficiency of cDNA synthesis and 2. Remove all particulate material from the hybridization
labeling can be judged by gel electrophoresis as outlined in
buffer, by filtration through a Millipore 0.22-pm-pore-size
section 45.2.6.1, step 5.
centrifugal membrane filter tube as follows. Wet the mem-
4. To remove unincorporated/quenched Cy dye, purify brane with 20 pl of sterile water. Spin briefly in a micro-
the DNA using QIAquick columns available as part of the
centrifuge (up to 60 s) at 8,000 X g, then decant water from
PCR purification kit (Qiagen). The method described the bottom collection tube. Add labeled target to the side
below is as specified by the manufacturer.
of the filter tube (avoid extended contact of probe with
5. To the Cy3 and Cy5 reaction mixtures in separate membrane). Spin briefly (30 to 60 s) at 8,000 X g. Typically
tubes, add 70 p1 of water and mix.
the recovery is approximately 26 pl.
6. Add 500 pl of PB buffer, apply to a QIAquick col- 3. Transfer flowthrough to a fresh PCR tube. Add 0.4 pl
umn, and spin at 12,000 x g in a microcentrifuge for 1 min.
of 10% SDS. (Do not put on ice after adding SDS.) To de-
7. Decant flowthrough. Add 750 pl of PE buffer and nature labeled cDNA, heat to 94C for 2 min, and transfer
spin at 12,000 X g for 30 to 60 s. Decant flowthrough. Spin
to a water bath at room temperature for 3 to 4 min.
as described above to dry the filter unit, and transfer to a
fresh microcentrifuge tube.
4. The hybridization cassette is prepared a s follows. En-
sure that the clamps fit snugly to the frame. While the la-
8. Add 30 pl of EB buffer to the center of the filter and
beled target is cooling, add a 10-pl drop of water to the bot-
incubate for 3 min at room temperature. Spin at 12,000 x
tom well of the hybridization chamber and load the
g for 1 min. Repeat elution step again with another 30 p1 of
microarray slide in the hybridization chamber. Add the en-
EB buffer. tire labeled target to the top of the microarray slide and
9.Pool eluates and add 20 pl (20 pg) of herring sperm with Millipore forceps quickly apply at an angle the 22- by
DNA. Add 420 p1 of TE buffer (pH 8) and apply to a fresh
30-mm coverslip (no. 1 thickness), avoiding bubbles. Close
Microcon YM-30 filter to concentrate the dye-labeled
the hybridization chamber and incubate in a water bath for
cDNA. Spin at 12,000 X g to reduce the volume to 20 p,l.
12 to 16 h at 63C.
10. The labeled cDNA can be used for hybridization to
microarrays as described in section 45.2.7. We note that other Following hybridization, slides are washed essentially as
laboratories use slightly different versions of the indirect- reported previously (46). Briefly, remove the hybridization
labeling protocol. The labeled cDNA and amount of dye in- chamber from the water bath and blot dry. Lay the cassette
corporation can be quantified by spectrophotometry (5). (top of slide facing up) on a clean bench surface and disas-
semble the hybridization cassette. Care is required so that
45.2.7. Hybridization and Processing of the printed spots on the slide are not damaged by the for-
Microarrays ceps. Remove slide from chamber with forceps and immedi-
Microarray hybridization can be performed either in an au- ately immerse in wash solution 1 (1.lX SSC, 0.03% SDS)
tomated hybridization instrument or in a simple manual de- at room temperature until the cover glass comes off. If only
vice. The automated hybridization instruments are available a few slides are being processed, this can be done individu-
from many vendors and seem to offer many advantages, such ally by hand; otherwise, we recommend placing the slides
as good reproducibility, low hybridization volume, the op- (up to 20), with coverslip still attached, into a slide holder.
tion of reusing hybridization solutions, and high-throughput Dip them all simultaneously into the wash solution until all
capability of handling 12 to 24 slides in parallel. Detailed in- the coverslips come off. To minimize carryover of SDS and
structions come with the instruments. Hybridization can salt to the secondary wash, it may be better to transfer slides
also be performed inanually either in a humidified box or one by one into a new rack. Transfer the rack containing
using a hybridization cassette device, which can be pur- slides to a slide-staining dish containing wash solution I1
chased from many vendors, including Telechem Interna- (0.06X SSC). Rinse vigorously by moving the slide holder
tional. Below we describe a procedure for manual hybridiza- up and down several times (up to 50 times). Keeping the
tion carried out in the hybridization cassette. It is of utmost slides submerged in the secondary wish, carry the slides to
importance to prevent dust contamination. The microarray the centrifuge, remove them from the wash solution, and
slide should not be allowed to dry during any of the steps spin dry the slides at 600 rpm for 4 min at room temperature
after initiating the hybridization experiment, since the la- (centrifugation at speeds in excess of 600 rpm could break
beled target may stick irreversibly to the microarray. A con- the slides). Hybridized arrays are extremely sensitive to en-
stant humidity has to be maintained in the chamber since vironmental ozone (17), and once dried, the arrays are sta-
high humidity tends to flood liquid around the coverslip and ble for several weeks if stored protected from light and ozone.
reduces signal strength, whereas low humidity will evaporate
the hybridization fluid. Suppression of bubble formation be- 45.2.8. Image Processing and Data Analysis
tween slide and cover glass is also very critical for the success
of hybridization. For hybridization, equal amounts of Cy3 45.2.8.1 . Feature Extraction and Quantification
and Cy5 incorporated dyes are combined. Typically, about Several types of microarray scanners, along with image ex-
40 pmol of each dye-labeled DNA is used for hybridization. traction software, are available from commercial sources.
The hybridization is performed in 3 x SSC (1 x SSC is 0.15 The type of instrument most commonly used is a confocal
45. DNA Microarray Technology 985

laser scanner with the ability to detect at least two fluo- be plotted on a scatter plot and would reveal the behavior
rophores and with the additional capability to adjust laser of the genes in the two experiments. For two hybridization
power, photomultiplier tube (PMT) voltage, and scan reso- experiments that are highly reproducible, the scatter plot
lution in the software. Optimization of laser power is impor- would yield a correlation coefficient (r') close to -1.0. An
tant, as high laser intensity could induce photobleaching of alternative and very useful approach to visualizing microar-
the hybridized slide and high PMT settings may saturate ray data is to construct a scatter plot of the log (to base 2)
pixel intensity. In practice, with a fixed laser power, the mi- ratio against the average intensity of each feature on the mi-
croarray is scanned using multiple PMT voltage settings in- croarray. These plots are called an R-I plot or M-A plot be-
dependently for each channel. The PMT voltage is set so as cause average intensity is referred to as A and log ratio as M.
to have the brightest pixels just below the level of saturation. If the two channels behave similarly, then all points would
Obtaining high-quality data from microarray experi- be centered around a horizontal line passing through zero.
ments is hampered by several technical limitations, includ-
ing imperfect microarrays (especially the spotted arrays) 45.2.8.2. Data Normalization
such as nonuniform spot sizes, abnormal spot morphology The next step in data analysis (Fig. 1) is data normalization,
(not perfectly round), often less-efficient labeling and hy- which helps in removing systematic variation potentially
bridization steps, and uneven hybridization patterns affecting the measured level of gene expression. For exam-
(doughnut o r crescent moon shapes). Thus, robust feature ple, for a self-versus-self experiment, the spot signals should
extraction methods are needed to minimize noise, extract be equal in both channels across the dynamic range of the
the best-quality data, and identify authentic differentially intensities. However, differences in reverse transcriptase
hybridized array elements. In the past, feature extraction performance during incorporation of Cy dyes into cDNA in
methods relied on manual techniques for flagging improper separate labeling reactions can potentially Cause a differen-
spots, but automated techniques are now available (45). tial change of signals in the two channels. Other sources for
There are several steps for feature extraction that form part such systematic variation between signal intensities include
of microarray data analysis (Fig. 1) and are briefly described inherent chemical differences in dyes with respect to their
here. First, a grid is superimposed over the scanned images. quantum efficiencies, variability in the amount of mRNA
Upon input of vital information about the coordinates of used for labeling, substrate fluorescence, propensity of non-
the microarray spots on a glass slide by the user, all image specific binding to the glass surface, possible biases intro-
extraction software can automatically determine and pro- duced by microarray scanner response, i.e., differences in
vide a tentative grid bearing the spot location on a slide; the the power settings of the two lasers and PMT voltages, gra-
user may only confirm the correct grid location and the dient effects arising from hybridization and washing steps
center of each spot. Prior to quantification, the user can and array printing processes.
edit the grid to exclude spots with slide artifacts such as The normalization process involves an initial probe se-
dust, abnormal spot morphology (i.e., pixel heterogeneity), lection step that is subsequently used for computation and
salt residue, and spots with saturated intensities in either optimization of dye normalization parameters. This is
channel. Once the features have been identified and cor- broadly of two types: a signal-independent, nonlocal linear
rectly griddect, image analysis software measures the inten- regression performed across the entire normalization data
sities in each channel for each of the features on the mi- set, essentially resulting in a global scaling of the data, and
croarray. Typically, we calculate the ratio of fluorescence a signal-dependent, locally weighted linear regression
intensities for each feature in the two channels for the Cy3- method or LOWESS, which essentially normalizes the data,
Cy5 two-color hybridization. Quantification of ratios and taking into account the consequences of any local pertur-
intensities is carried out as follows. First, the background is bation. Normalization using mean signal intensity in each
determined either from the signal emanating from the in- channel was practiced during the early phase of implemen-
terfeature intensity in each of the Cy3 and Cy5 channels or tation of the microarray technology. It is now clear that this
from the signal values derived from a set of negative control approach will not be successful if, for example, hybridizing
spots outlined in section 45.2.1. Second, the intensity of signals from the bulk of the spots changed as a result of a
each spot in the foreground is measured in each of the mutation that reduced expression of the majority of the
channels. Finally, the spot intensity is determined by sub- genome. Therefore, other methods have been developed
tracting the background signal value derived from step 1 that utilize a minimum set of housckceping genes because
above. Image processing and normalization routines have their expression is constant across a range of samples in se-
been described in detail before (7, 66, 67). lected experiments. The selection of normalization probes
In two-color ratio measurements, one would like to cal- is very important and involves use of either preselected
culate Cy5/Cy3 ratio value for each spot such that the ratio housekeeping genes or virtual housekeeping genes that are
is a measure of RNA abundance in the two channels. generated from the body of microarray data itself (59, 69),
Because ratios are not normally distributed, it is necessary and is used to select a set of invariant genes that have Same
that the ratios be log transformed. For example, we would the intensities in both Cy5 and Cy3 channels. However,
see a normal distribution if we take log (to base 2) of ratio the use of a small set of housekeeping genes is not always
2 and 0.5;we would get 1 and -1, respectively. The most ideal due to the unforeseen changes in their differential ex-
common data visualization tool is a scatter plot of the log pression under certain conditions.
ratio data on the y axis and sum of medians on a log x axis Another approach is to spot genomic DNA samples of
that would reveal the performance of the microarray exper- increasing concentrations from the same organism on the
iment. A scatter plot can be easily constructed in a microarray. This has been employed for yeasts but may also
Microsoft Excel spreadsheet or in other dedicated microar- be useful for other organisms with a low proportion of non-
ray data analysis software. For instance, the induced and re- coding sequences in the genome. In cases where this is not
pressed genes and the low-intensity spots would be clearly feasible, a concentration series of probes comprising a pool
revealed graphically in the scatter plot. Also, the log (base of the cDNAs or oligonucleotides can be incorporated as
2) Cy5/Cy3 ratio of two microarray hybridization data can control spots on the same microarray. However, a limitation
986 w MYCOLOGY

of this approach is the possibility of overrepresenting a few study, Hughes et al. (27) carried out 63 control pairs of ex-
highly expressed genes that could confound the signal val- periments performed in epoch with the actual experiments,
ues. The use of external standards, or spike-ins, is efficient wherein expression profiles from two untreated isogenic
and a good option, since they serve as a basis to correct for wild-type yeast cultures grown in parallel were compared to
RNA recovery, dye-labeling efficiency, and other technical each other. Remarkably, at least one gene exhibited 2 2 -
limitations; importantly, this method does not assume a pri- fold change (up- or down-regulated) in 55 of the 63 exper-
ori that the total RNA content is the same in the reference iments (27). Examination of these genes revealed that
and experimental populations (2, 26, 60, 71). For instance, many were known to be regulated by nutritional quality or
spots representing five to seven Bacillus subtilis genes that do stress. These fluctuations were considered a form of biolog-
not cross-hybridize to the yeast genome were spotted on the ical noise, and this information was used to derive an error
microarray as normalization control probes. Then their cog- model so as to reduce the significance of expression changes
nate mRNAs were transcribed in vitro and known amounts for these groups of genes. Indeed, data analysis of the 300
were added to the reference and test RNA populations prior compendium experiments showed that expression of these
to mRNA isolation, cDNA synthesis, and labeling (2, 71). groups of genes fluctuated in most of these experiments, in-
Although a single recommendation cannot be made as to dicating that the same biological noise was inherent in the
the choice of the normalization method, it is often helpful 300 experiments (27). These results emphasize the impor-
and prudent to consider various options and test the most tance of negative control experiments involving compar-
suitable ones before settling with a normalization method. isons of identically treated cells and the need for a gene-
specific error model. The goal in the implementation of an
45.2.8.3. Statistical Tools Accelerate the error model is to determine variance when t r u t h is
Identification of True Expression Changes known, i.e., all expression ratios are equal to 1.0. Deviations
The core DNA microarray technology was established sev- from 1.0 are noise. The error model assigns a statistical sig-
eral years ago, but methods for microarray data analysis are nificance numerically expressed as a P value for the expres-
still evolving. Several new software tools have been devel- sion ratio of each spot on the array, which is applied to all
oped recently that aim to increase the reliability of detect- similarly performed experiments with the assumption that
ing true expression changes based on robust statistical crite- those experiments will experience similar errors. A range of
ria. In this section, we briefly review some of the steps in P value cut is employed, often with a P value of 50.01 (a
microarray data analysis. confidence level of 99% or more). If a given gene produces
A key initial step in handling microarray data is to assess a ratio greater than seen in same-versus-same control ex-
the reproducibility of the data by repeating each experi- periments, the change is assigned a very small P value, in-
ment at least three times (i.e., from start to finish, from the dicating a very low likelihood that the change was due to
same glycerol stock of the yeast strain) and examining the chance and this represented a true expression change. Many
reproducibility of profiles in the multiple experiments using commercially available feature extraction software packages
either scatter plot or clustering algorithms. The minimum apply error models, and a comparative study of the perform-
number of repeat experiments is often determined by the ance of the different error models is also available (55). A
quality of data and the magnitude of expression change. detailed comparison of the features of some of the commer-
Next, the question becomes, what is a statistically mean- cial software suites for microarray data analysis is also avail-
ingful change? That depends on the level of noise in the able (13).
assay. The noise is broadly composed of biological noise re- The above sections described a numerical basis to obtain
flected in RNA preparation, which can be assayed by repli- statistically significant data that minimized systematic er-
cates, and platform noise that emanates from processing of rors and biological variability. But true expression change
RNA samples. The individual sources of noise can be sorted can often be verified by cleverly designed follow-up experi-
out with analysis of variance (ANOVA),a statistical tool to ments. Numerous microarray studies have revealed that
determine variability in multiple data (11). The ANOVA yeast cells, upon exposure to a variety of perturbations,
model is applied to transformed intensity data obtained elicit large changes in the mRNA expression profile (for ex-
from a logarithmic transformation of raw intensity data. It amples, see references 8, 12, 19, 20, 27, 33, and 48). Such
allows one to account for sources of variation in the data expression changes could be a direct effect of the perturba-
that are attributable to factors other than differential ex- tion o r due to secondary effects generated in the experi-
pression of genes; thus, it effectively normalizes the data. A mental situation. It is therefore necessary to identify what
software implementation of ANOVA for microarray data constitutes a true biologically relevant expression change.
analysis is also available (76). Although not infallible, time course studies have the po-
Significance analysis of microarray (SAM) is another tential to minimize secondary effects by focusing on early
statistical approach to determine how likely it is that the events during gene expression reprogramming. In addition,
gene is expressed differently between the two cell popula- use of a genetic strategy such as mutant analysis or regulated
tions (70). For instance, SAM assigns each gene a A score gene expression to validate the results would help to au-
based on both its level of expression and reproducibility. All thenticate the biological relevance of the microarray data.
genes whose scores are higher than a selected threshold The magnitude of expression change is not the sole indica-
are then deemed significant. SAM also calculates a false- tor of a biologically relevant phenomenon. Upon obtaining
discovery rate for each threshold of significance. This value statistically significant data, the next task is to assess the
estimates the percentage of the genes with scores higher expression pattern of known genes and infer the functions
than a given threshold that are likely to be false positives. of unknown genes from their expression patterns. These
SAM is a very popular statistical tool, and the software is analyses were pioneered using cluster analysis ( 16), wherein
available for download in the public domain (70). genes are sorted and arranged on the basis of their expres-
A rigorous way to deal with inherent noise in the micro- sion patterns across a range of experiments. The rationale
array data is by examination of control same-versus-same for this approach is that genes that function in a common
experimental data as reported earlier (27). In a seminal pathway are often co-regulated in different experimental
45. DNA Microarray Technology 987

conditions. Cluster analysis has three steps: (i) applying s i p 45.3. APPLICATIONS
nificance cuts to the expression data matrix, (ii) defining a
similarity (or dissimilarity) measure matrix, and (iii) per- 45.3.1. Uncovering Genetic Circuits Governing
forming clustering based on the similarity matrix. In two- Metabolic Pathways
dimensional cluster analysis, gene clustering and experi- Numerous microarray studies have been carried out to ex-
ment clustering are performed independently, without plore differential gene expression. As an example of how
interference between the two dimensions. It is noted that one can dissect metabolic pathways, we briefly review the
formalisms for gene clustering and experiment clustering experimental setup and salient findings from a few studies
are symmetric between genes and experiments. A detailed (12, 48). DeRisi et al. ( 12) used microarrays to assess global
description of cluster analysis can be found elsewhere (16, changes in gene expression during diauxic shift, a ilatural
54,63). transition of yeast cells from fermentative met boI'ism to
respiration. They demonstrated that genes displaying simi-
45.2.8.4. Microarray Data Reproducibility, lar expression patterns were functionally related and regu-
Standards, and Databases lated by upstream sequence motifs common to the promot-
The reproducibility of gene expression measurements using ers of the genes. Indeed, expression of about one-third of
the different microarray platforms has been a matter of con- the yeast genome was altered as cells encountered glucose
cern. Several attempts were made to compare data obtained limitation during the diauxic shift. Interestingly, expression
using one platform with another using the same sample patterns of many uncharacterized genes provided clues to
RNA. For example, using the 60-mer ink-jet-synthesized their possible functions.
oligonucleotide microarray and spotted cDNA microarray Yeast cells respond to starvation for one of the many
platforms, transcriptome differences were studied in yeast amino acids and other forms of stress by inducing the trans-
cells grown in sporulation medium versus synthetic com- lation of Gcn4p, a bZIP transcriptional activator (23, 24).
plete medium (26). Remarkably, transcript abundance ra- Gcn4p binds to general control response elements
tios correlated exceptionally well between measurements in (UASGCRE)in the promoter regions to activate transcrip-
the two platforms. However, in a different study, same pairs tion of cognate target genes. To determine the genome-wide
of RNA samples were analyzed in three different platforms changes in gene expression elicited by histidine starvation,
involving single-color or dual-color hybridizations. While microarray hybridizations were carried out using fluores-
the results of the two single-color platforms largely corre- cently labeled cDNA from wild-type cells treated with 3-
lated well, little overlap was found between genes that aminotriazole (3AT), an inhibitor of histidine biosynthesis,
showed significant gene expression change between one- and from untreated cells as a control. We employed a I' value
color and the dual-color hybridization platforms (68). criterion (see section 45.2.8.3) of 50.01 or 50.05 to filter
Systematic comparisons were carried out on a monumental genes whose expression was less statistically probable with a
scale by several laboratories both outside and within the confidence level of 95 to 99%. Microarray data analysis re-
framework of the Microarray Quality Control Project. The vealed that 3AT treatment led to up-regulation of about
reproducibility of microarray data emanating from multiple 24% of the yeast genome and down-regulation of about an
microarray platforms and laboratories was evaluated (4, 30, equal fraction (-28%) of the genome (48). Multiple hy-
38, 51, 64). In general, it was concluded that the use of bridization experiments were carried out with a second non-
standardized protocols, RNA reference samples, and careful isogenic strain, and the data revealed that the vast majority
design of experiments analysis methods yielded quite com- of the genes that were induced or repressed in the first ex-
parable data across different microarray platforms and across periment also exhibited similar behavior in the different
different laboratories. strain background. This suggested that true expression
As microarray data are accumulated at a rapid pace, changes are likely to be reproduced in multiple strain back-
archival expression data become important. New insights grounds and, therefore, such comparisons could be adopted
could be obtained by analyses of the new microarray data as a general practice. To identify what fraction of the 3AT-
alongside previous expression data. Moreover, accumula- regulated genes is dependent on Gcn4p for regulation, ex-
tion of expression profiles from a large number of separate pression profiling was conducted in isogenic wild-type and
conditions facilitates the use of clustering algorithms to re- gcn4A mutant strains both exposed to 3AT. Agglomerative
veal coordinately regulated genes (16). It was realized that two-dimensional hierarchical clustering was carried out ;as
for reliable comparison of micrcxarray data, there was a need described previously (27). The results showed that the bulk
for standardized methods to describe and communicate mi- of the genes up- or down-regulated by 3AT treatment of
croarray experiments and to develop centralized databases wild-type strains also showed Gcn4p dependence for their
to facilitate meta-analysis. One published standard and expression in the GCN4/gcn4A experiment, indicating that
guideline was developed by the Microarray Gene Expres- Gcn4p function was required for both up-regulation and
sion Data Group (6). Minimum Information About a Mi- down-regulation of genes in amino acid-starved cells.
croarray Experiment (MIAME) provides a framework for a Furthermore, to comprehensively identify the set of genes
comprehensive specification of the details of the microarray regulated by Gcn4p, we applied Boolean logic, which re-
data, including experimental and array design, sample quired that genes up-regulated upon 3AT treatment in a
preparation, hybridization protocols, actual quantitative re- wild-type strain should also yield a twofold change in the
sults, and normalization controls (6). This initiative has GCN4lgcn4A experiment. Applying these criteria, :i group
been accepted widely, and most major journals now require of 539 genes representing about 9% of the genome were
compliance with MIAME for any new submissions employ- identified to be induced by Gcn4p in response to amino acid
ing microarray data. Details of the micrcxirray experiments starvation (48). Searching the upstream promoter regions of
and raw microarray data may be deposited with one of the the Gcn4p target genes revealed that only 235 genes (44%)
two public repositories: Gene Expression Ominbus (http:l/ had one or more consensus UASC;CREelements in its pro-
www.nchi.nlm.nih.gov/geo), and ArrayExpress (http://www moter, indicating that they are directly regulated by Gcn4p.
.ebi.ac.uk/arrayexpress) . The rest of the Gcn4p target genes may have other cis
988 MYCOLOGY

elements, such as the cyclic AMP response element recog- achieved by coordination of mRNA transcription rates and
nized by Gcn4p (49, 61). Alternatively, they may be regu- stability.
lated by another transcription factor which, in turn, is regu- An additional level of gene regulation is achieved by dif-
lated by Gcn4p. The majority of genes negatively regulated ferential efficiency of mRNA translation. Microarrays have
by Gcn4p lacked UASGCRE,leading us to propose that their been used to assess mRNA translation efficiency on a
repression results from squelching by Gcn4p, wherein gen- genome-wide scale by comparing mRNA to protein levels
eral transcription factors or cofactors are sequestered by (2, 34, 36, 62). The ratio between the proportions of an
Gcn4p at its target genes and less available for other tran- mRNA associated with polyribosomes versus monosomes is
scriptional activators. In any case, these results revealed that indicative of its translation initiation rate. Accordingly,
G c d p is a pervasive transcriptional activator in S. cereuisiae. mRNA was isolated from sucrose gradient fractions con-
Comparative expression profiling is a powerful approach taining polyribosomes or from monoribosome fractions, and
to infer new connections between regulatory pathways. the total unfractionated mRNA, used as a reference, was re-
Central to this approach is the two-dimensional clustering verse transcribed to prepare fluorescently labeled cDNAs.
of expression data from different experimental situations. The cDNAs from polysomes and monosomes were each
Cluster analysis of the above-described data from 3AT ex- combined with the reference cDNA and hybridized to all of
periments with the microarray data obtained by treatment the ORFs in the yeast genome (2). It was found that 43
of yeast cells with methyl methanesulfonate (MMS) (32, mRNAs were not associated with either monosomes or
33) showed that the bulk of genes induced by Gcn4p in polysomes and thus could potentially be regulated at the
3AT-treated cells are also induced in MMS-treated cells. translational level by associating with the translation ma-
Indeed, it was shown that Gcn4p expression was induced by chinery only under certain special conditions. An addi-
MMS in a manner dependent on the upstream regulator tional group of 53 mRNAs, including GCN4 and CPAI,
Gcn2p (48). both known to be regulated by translational control mech-
anisms mediated by upstream ORFs, was predominantly as-
45.3.2. Unsupervised Identification of Gene sociated with the monoribosome fraction. These results
Function: a Compendium Approach provided a first genome scale view of posttranscriptional
Uncovering the function of novel genes is an important as- gene regulatory events.
pect of functional genomics projects. Hughes et al. (27)
proposed that a compendium or a comprehensive database 45.3.4. mRNA Splicing Arrays
of diverse expression profiles could serve Splicing of mRNA has important consequences in synthe-
which a query expression profile could be compared to ob- sis of functional mRNA in different genomes. In higher eu-
tain clues as to the pathway(s) that is affected by a pertur- karyotes, alternative splicing leads to novel forms of mRNA
bation or a mutation in a novel gene. In a landmark study, from a primary transcript and has also been implicated in
a compendium of 300 expression profiles associated with a producing proteins of distinct or even antagonistic func-
multiplicity of diverse mutations, regulated expression of tion. A detailed analysis of the use of splicing arrays for dis-
genes, and chemical treatments was created (27). The fol- covery of alternative splice forms can be found elsewhere
lowing example illustrates the utility of this compendium (39). For most organisms, however, splicing of mRNA leads
for gene discovery. Inhibition of ergosterol biosynthesis to intron removal to produce a functional mRNA. The
leads to transcriptional induction of the genes encoding the functional genomics and genome annotation project largely
pathway enzymes. As expected, microarray analysis of mu- depends on bioinformatics tools to predict mRNAs that are
tants and treatments affecting ergosterol biosynthesis processed by the alternate splicing pathways. Therefore, a
showed similar expression profiles. Profiling of the yer044cA systematic genome-wide analysis is required to accurately
mutant strain and two-dimensional cluster analysis showed assess the role of splicing in genome function.
that the expression profile of this mutant was clustered with Splicing occurs in the nucleus in a large multiprotein
those elicited by the known sterol pathway mutants erg2A complex called the spliceosome, which contains essential
and erg3A, and upon overexpression of ERGI I . These find- and several nonessential subunits. To discriminate between
ings suggested that YER044C (renamed ERG28) functions spliced and unspliced RNAs for each intron-containing
in ergosterol biosynthesis, and indeed follow-up experi- yeast gene, a novel microarray was employed (10).
ments provided confirmation (27). Oligonucleotides were designed to detect the splice junc-
tion (specific to spliced RNA and not found in the ge-
45.3.3. Translational Arrays nome), the intron (present in unspliced RNA), and the sec-
DNA microarrays have also been used to assess genome- ond exon (common to spliced and unspliced RNA) for each
wide profiles of mRNA translation or mRNA stability. For of the 244 known or predicted intron-containing genes.
example, Wang et al. determined that yeast mRNAs have The oligonucleotides were printed on glass slides to create
half-lives between 3 min and 90 min (73). In this study, a splicing-sensitive microarrays for yeasts. As normalization
temperature-sensitive allele of a gene encoding an RNA controls, oligonucleotides corresponding to seven intron-
polymerase I1 subunit (RPBI) was used to rapidly initiate lacking genes were spotted in the Same microarray slide.
transcription at the nonpermissive temperature. Following Fluorescently labeled cDNA was prepared from total RNA
the temperature shift, total RNA was isolated at short time by reverse transcription with a mixture of oligo(dT) and
intenrals and fluorescently labeled cDNA was synthesized random hexamer primers and hybridized to the microarray.
and hybridized to ORF microarrays. Interestingly, it was To validate this approach, cDNA made from a wild-type
found that mRNAs that encode proteins involved in a strain was labeled with Cy3, whereas Cy5-labeled cDNA
common function or that are constituents of the same was made from RNA isolated from temperature-sensitive
macromolecular complex, such as ribosomal protein mutant prp4-I, defective in a subunit of the spliceosome.
mRNAs or the nucleosomal core histone mRNAs, have Hybridization revealed that intron-containing probes accu-
similar half-lives (73). These results add a new dimension to mulated, whereas splice junction probes declined, indicating
gene regulation wherein fine control of gene regulation is a reduction in splicing in the mutant grown at a nonpermis-
45. DNA Microarray Technology 989

sive temperature. To evaluate the contribution of different cell growth, cells are fixed with formaldehyde, which cross-
mRNA processing factors for splicing, strains bearing muta- links protein-protein and protein-DNA intcractions.
tions in 18 different nonessential subunits were employed Chromatin is isolated and sonicated to shear chromatin
(10). Their analysis showed a requirement for a specific fragments to an average size of about 300 to 1,000 bp.
mRNA processing factor(s) vis-8-vis a particular intron- Antibodies to either epitope-tagged proteins or highly spe-
containing gene, rather than a general requirement for all cific antibodies are used to coimmunoprecipitate genomic
or most mRNA processing factors. It was previously thought regions associated with proteins of interest. The immuno-
that Prpl7 and Prpl8 factors were required for splicing of precipitated DNA is amplified and labeled by either of two
mRNA with short distance of branch point to the 3 splice methods. In the first method, a two-step PCR is employed
site using ACT1 in vitro. In contrast, results from the splic- wherein degenerate primers are used to nonspecifically
ing arrays involving almost all intron-containing genes prime two rounds of DNA synthesis using the immunopre-
showed no such strict distance specificity. The splicing array cipitated DNA. In the second step, a specific primer is used
could be employed for genome annotation and validation of to prime exponential amplification (31, 37, 41, 57). In the
alternatively spliced genes, but perfect designing of splicing second method, ligation-mediated PCR was carried out to
array would depend on the strength of the bioinformatic incorporate fluorescent dye into the immunoprecipitated
tools to accurately predict all potential intron-containing and control DNAs samples (40, 56, 77). In both the ap-
genes. proaches, labeled DNAs from experimental and control im-
munoprecipitated samples were mixed and hybridized to in-
45.3.5. Fitness Testing in Competitive Growth tergenic array consisting of spots corresponding ..tothe DNA
Assays To Identify Gene Function and Drug Targets sequences located between two ORFs.
Subtle phenotypes associated with mutations are often dif- In a systematic large-scale study, the ChlP-array tech-
ficult to score by classical techniques, and therefore, mi- nology has been employed to investigate genome-wide pro-
croarray technology has been used to determine phenotypes moter occupancy of 106 transcriptional regulators encoded
of mutants in a mixed population of yeast strains (21, 27). by the yeast genome (40). Based on these studies, Lee et al.
Briefly, a library of yeast deletion mutants corresponding to (40) provided evidence that alteration in gene expression
each of the approximately 4,400 nonessential genes was first profiles is executed by a network of interactions among
constructed by individually replacing every ORF with a transcriptional regulators. Several types of interactions were
(3418 resistance selectable marker gene and short sequence uncovered by their study. In the most common situation,
tags called UP tag and DOWN tag that are unique to each one or more transcription factors bound directly to the pro-
deletion. These two unique sequence tags serve as molecu- moters of a target group of coregulated genes. In mother in-
lar bar codes with which to identify the abundance of each teresting situation, one transcriptional activator bound to
mutant strain. Pools of mutant strains were cocultured the promoter of a second transcriptional activator and both
under specific growth conditions in a competitive growth activators in turn bound to the promoter of the same target
assay, and cells were harvested at zero time and subse- gene. In this way, a given transcriptional activator could
quently at defined time intervals. The rationale of this assay regulate expression of the target gene directly and also indi-
is that if a mutation impaired growth of the mutant cell in rectly by controlling the expression of the second transcrip-
a particular growth condition, then the relative abundance tion factor working at the same target gene. Lee et al. (40)
of such a mutant strain would diminish in the population showed that, depending on the choice of the P value cutoff,
upon extended incubation time. O n the other hand, dele- the number and reliability of identified targets can vary
tion mutations that do not impair growth would he selec- dramatically. Only a few of the expected binding sites for
tively enriched in the population. Thus, the rate of decrease the transcriptional activator Gcn4p could he identified de-
of the mutant cells from a population in a competitive spite using a relaxed P value filter (reference 39 ;ind our un-
growth assay is indicative of the requirement for its cognate published data), indicating that the experimental and sta-
gene for growth. As a proof-of-principle experiment, the tistical methods need to be refined for comprehensive
competitive growth assay was employed to identify all genes identification of genome-wide transcription factor binding
required for growth in galactose media (21). In addition to in vivo. Extending this study, Pokholok et al. (53) em-
genes already known to he involved in galactose utilization, ployed a modified tiling microarray that contained 44,290
about 9 or 10 new genes required for galactose metabolism features consisting of 60-mer oligonulceotide probes that
were identified by this approach. Microarray coupled with covered 12 Mb of the yeast genome (85%),with an average
competitive growth assay has also been used for drug target probe density of 266 bp, barring the highly repeated regions.
identification (44). Here pools of heterozygous mutants By employing further modifications to the experimental
were exposed to different compounds and the fitness of the protocol, Pokholok et al. obtained substantially higher res-
individual mutants was scored essentially as described above olution and accuracy than previous methods. Analysis of
for the galactose regulon. Drug targets for several known Gcn4 binding with the new method revealed a total of 210
compounds were validated, and new targets for several com- genes whose promoters are bound at high confidence values
pounds were discovered. Thus, the competitive growth test within an optimal p value threshold of 6 x (53).
in combination with the microarray technology is a valu-
able tool to identify gene function and to identify gene tar- 45.3.71 Comparative Genome Hybridization
gets for pharmaceutical compounds and drugs. The microarray technology has also been adopted for com-
parative genome hybridization (CGH) studies. Briefly, CGH
45.3.6. Use of Intergenic DNA Microarrays To involves isolation of genomic DNA from a sample strain and
Detect Protein-DNA Interactions the reference strain and then labeling by Cy3 and Cy5, fol-
The microarray technology has been used in conjunction lowed by Cy3/Cy5 hybridization to a standard cDNA micro-
with the chromatin immunoprecipitation assay in so-called array containing whole-genome probes from the reference
ChIP-array experiments to detect genome-wide protein- strain. This strategy would enable detection of the relative
DNA association in vivo. Briefly, at an appropriate stage of abundances of the chromosome(s) or chromosomal segments
990 w MYCOLOGY

in the sample versus the reference strain. In one of the early teomics technologies can be used at the whole-genome
implementations of this methodology, Hughes et al. (28) re- level to assess either the proteome abundancc or relative
ported that several mutant yeast strains used in microarray proteome differences between two conditions or cell types
experiments in various laboratories were aneuploid for (1, 50). Although these technologies have not yet reached
whole chromosomes or chromosomal segments that presum- the robustness and sensitivity achieved in transcriptome
ably emanated from strain construction steps. The rationale analysis, the current estimates show that at least 1,000 pro-
behind this discovery was their observation that a substan- teins can be identified in a single liquid chromatography-
tial fraction of the mutants exhibited chromosome-wide tandem MS experiment (1,42).
aneuploidies. Therefore, it has now become imperative that Several reports have now provided insights into the cor-
each expression profile be judged for artifactual expression relation between mRNA and protein levels (22, 29, 74).
biases by plotting the expression data on a chromosomal For instance, Washburn et al. studied the impact of cultur-
map. More recently, CGH has been applied to phylogenetic ing yeast cells in rich versus minimal media (74). They
studies by comparing the relatedness of all genes between showed that almost the entire amino acid and purine
two organisms (14, 15,47, 75). Once the CGH technology biosynthetic pathways were induced in minimal medium at
is improved to be able to use small amounts of clinical ma- both the transcriptome and proteome levels (74). Increases
terial, CGH could be used routinely for pathogenic strain or of mRNA and protein expression ratios of several amino
species typing in a rapid and robust manner. acid and purine biosynthetic pathways were well correlated
at each locus and at each pathway level, and also largely
45.3.8. Integration of Technologies for correlated well with an earlier transcriptome study carried
Comprehensive Assessment of Genome Function out under histidine starvation conditions (48). This sug-
For comprehensive assessment of the function(s) of novel gests that culturing yeast cells in minimal medium elicits a
genes and that of a newly sequenced genome, technologies physiological response similar to that of inducing Gcn4p
are now available for measurement of mRNA expression expression upon severe histidine starvation. Interestingly,
(transcriptome), the protein expression (proteome), and several proteome level changes did not correlate with the
the metabolites (metabolome), leading to a systems biology transcriptome level change. For example, in rich medium
approach integrating all of this information (Fig. 2). In this the Tupl repressor protein was eightfold overexpressed, but
scheme, the microarray technology may be the first level of this change was not detectable at the mRNA level; the
analysis leading to assessment of gene expression under spe- RNA polymerase I1 holoenzyme subunits Ancl p and Sin4p
cific conditions. Next, leads obtained from the microarray were overexpressed in minimal medium, but these changes
data can be pursued by phenotype testing using appropriate were not detectable at the transcript level. Moreover, the
mutant/genetic strategies (27, 48), to narrow down specific overall correlation between mRNA and protein expression
pathway(s) or regulators responsible for alteration in gene was weak, with a Spearman rank correlation coefficient of
expression as discussed earlier (see section 45.3.1). Given 0.45 for 678 loci. These results highlight the importance of
that proteins are effectors of programmed gene expression, application of both transcriptome and proteome mea-
assessment of their abundance and activity is critical to un- surements to assess gene expression changes (Fig. 2).
derstand genome function. Also, posttranscriptional regula- There are also examples where a transcriptional profile

-
tory mechanisms can offset the measured abundance of the generated from microarrays has been integrated into
transcriptome of a cell. Mass spectrometry (MS)-based pro- metabolite profiles obtained using liquid chromatography-

,
,.-- Microarray Data

Phenotype Analysis Mutants/


CeneticApproaches
-
(e.g.A Compendium approach)
I
Proteorne Analysis

Metabolome Analysis

Protein-Protein
/ Interaction Network
Pathway Assignment

* Biological Function 4

FIGURE 2 Suggested workflow for comprehensive assessment of genome function. Integration


of results from microarray studies into the results of proteome analysis, metabolome analysis, and
protein-protein interaction data would provide a broad platform for assignment of genes to respec-
tive pathways and inference of the biological function of novel genes.
45. DNA Microarray Technology rn 991

MS technology for the lovastatin-producing fungal strain Lasarev, J. Li, Y. J. Li, E. K. Lobenhofer, X. Lu, R. L.
Aspergillus terreus (see reference 3, Fig. 2). Askenazi et al. Malek, S. Milton, S. R. Nagalla, P. OMalley J, V. S.
devised an association approach linking the microarray Palmer, P. Pattee, R. S. Paules, C. M. Perou, K. Phillips,
data, and the metabolite profiling data and several interest- L. X. Qin, Y. Qiu, S. D. Quigley, M. Rodland, I. Rusyn,
ing leads were obtained (3). Lovastatin and (+)-geodin are L. D. Samson, D. A. Schwartz, Y. Shi, J. L. Shin, S. 0.
synthesized from polyketide-derived secondary metabolites. Sieber, S. Slifer, M. C. Speer, P. S. Spencer, D. I.
Genes involved in catabolism of fatty acids leading to the Sproles, J. A. Swenberg, W. A. Suk, R. C. Sullivan, R.
synthesis of polyketide precursors acetyl coenzyme A and Tian, R. W. Tennant, S. A. Todd, C. J. Tucker, B. Van
malonyl coenzyme A, positively correlate with lovastatin Houten, B. K. Weis, S. Xuan, and H. Zarbl. 2005.
Standardizing global gene expression analysis between lab-
and (+ )-geodin production and negatively correlate with
oratories and across platforms. Nut. Methocls 2:35 1-356.
genes involved in anabolic steps of fatty acid metabolism. 5. Bowtell, D., and J. Sambrook. 2003. DNA Microarrays: a
Genes encoding enzymes that catabolize alternative nutri- Molecular Cloning Manual. Cold Spring Harbor Laboratory,
ent sources (such as glycogen phosphorylase, lysophospholi- Cold Spring Harbor, NY.
pase, and alkaline protease) are expressed in late growth 6. Brazma, A., P. Hingamp, J. Quackenbush, G. Sherlock,
phase, and this positively correlated with lovastatin pro- P. Spellman, C. Stoeckert, J. Aach, W. Ansorge, C. A.
duction and negatively correlated with genes involved in Ball, H. C. Causton, T. Gaasterland, P. Glenisson, F. C.
energy generation via the glycolytic and tricarboxylic acid Holstege, I. F. Kim, V. Markowitz, J. C. Matese, H.
pathways active during early growth phase ( 3 ) . Parkinson, A. Robinson, U. Sarkans, S. Schulze-
Several studies have found coregulation of genes encod- Kremer, J. Stewart, R. Taylor, J. Vilo, and M. Vingron.
ing proteins that are integral subunits of multiprotein com- 2001. Minimum Information About a Microarray Experi-
plexes. For example, mRNA levels for 90 genes encoding ment (MIAME)-toward standards for microarray data.
60s and 40s ribosomal subunits were coordinately down- Nut. Genet. 29:365-371.
regulated upon severe amino acid starvation (48). More- 7. Causton, H. C., J. Quackenbush, and A. Brazma. 2003.
over, proteome analysis showed that eight loci encoding Microarray Gene Expression Data Analysis: a Beginners
subunits of the 60s ribosomal particle were overexpressed Guide, p. 40-69. Blackwell Publishing Limited, Malden,
in rich medium (YPD) compared to minimal medium (74). MA.
Indeed, association analysis of microarray data of transcript 8. Causton, H. C., B. Ren, S. S. Koh, C. T. Harbison, E.
levels using two-dimensional clustering approaches re- Kanin, E. G. Jennings, T. I. Lee, H. L. True, E. S.
Lander, and R. A. Young. 2001. Remodeling of yeast
vealed that loci that function as a multisubunit complex are
genome expression in response to environmental changes.
more often clustered together (27,48). Thus, integration of Mol. Biol. Cell 12:323-337.
protein-protein interaction assays (52) with complementary 9. Churchill, G. A. 2002. Fundamentals of experimental
mRNA and protein expression analysis (Fig. 2) would pro- design for cDNA microarrays. Nut.. Genet. 32(Suppl.):
vide vital clues about function of novel genes. In summary, 490-495.
we have provided detailed methodology for carrying out the 10. Clark, T. A., C. W. Sugnet, and M. Ares, Jr. 2002.
DNA microarray technology and briefly reviewed its atten- Genomewide analysis of mRNA processing in yeast using
dant applications. A careful interpretation of the microar- splicing-specificmicroarrays. Science 296:907-910.
ray data is important so that meaningful hypotheses can be 11. Cui, X., and G. A. Churchill. 2003. Statistical tests for
generated for designing follow-up experiments. Further re- differential expression in cDNA microarray experiments.
finement of the statistical tools for microarray data analysis, Genome Biol. 4:210.
including standardization of data normalization methods, 12. DeRisi, J. L., V. R. Iyer, and P. 0. Brown. 1997.
would yield more robust and reliable data and enable com- Exploring the metabolic and genetic control of gene ex-
parison of microarray data generated in different laborato- pression on a genomic scale. Science 278:680-686.
ries and possibly find use in clinical practice. 13. Dresen, I. M., J. Husing, E. Kruse, T. Boes, and K. H.
Jockel. 2003. Software packages for quantitative microar-
ray-based gene expression analysis. Curr. Pharm. Biorech-
45.4. REFERENCES nol. 4:417-437.
Aebersold, R., and M. Mann. 2003. Mass spectrometry- 14. Dunham, M. J., H. Badrane, T. Ferea, J. Adams, P. 0.
hascd proteomics. Nature 422: 198-207. Brown, F. Rosenzweig, and D. Botstein. 2002.
Arava, Y., Y. Wang, J. D. Storey, C. Liu, P. 0. Brown, Characteristic genome rearrangements in experimental
and D. Herschlag. 2003. Genome-wide analysis of mRNA evolution of Succharomycescerevisiue. Proc. Nutl. Acad. Sci.
translation profiles in Saccharomyces cereoisim. Proc. Nutl. USA 99: 16144-16149.
Acud. Sci. USA 100:3889-3894. 15. Edwards-Ingram, L. C., M. E. Gent, D. C. Hoyle, A.
Askenazi, M., E. M. Driggers, D. A. Holtzman, T. C. Hayes, L. I. Stateva, and S. G. Oliver. 2004. Com-
Norman, S. Iverson, D. P. Zimmer, M. E. Boers, P. R. parative genomic hybridization provides new insights into
Blomquist, E. J. Martinez, A. W. Monreal, T. P. the molecular taxonomy of the Saccharomyces sensu stricto
Feibelman, M. E. Mayorga, M. E. Maxon, K. Sykes, J. V. complex. Genome Res. 14:1043-1051.
Tobin, E. Cordero, S. R. Salama, J. Trueheart, J. C. 16. Eisen, M. B., P. T. Spellman, P. 0. Brown, and D.
Royer, and K. T. Madden. 2003. Integrating transcrip- Botstein. 1998. Cluster analysis and display of genome-
tional and metabolite profiles to direct the engineering of wide expression patterns. Proc. Natl. Acad. Sci. USA 95:
lovastatin-producing fungal strains. Nur. Biorechnol. 2 1: 14863- 14868.
150- 156. 17. Fare, T. L., E. M. Coffey, H. Dai, Y. D. He, D. A.
Bammler, T., R. P. Beyer, S. Bhattacharya, G. A. Kessler, K. A. Kilian, J. E. Koch, E. LeProust, M. J.
Boorman, A. Boyles, B. U. Bradford, R. E. Bumgarner, Marton, M. R. Meyer, R. 8. Stoughton, G. Y. Tokiwa,
P. R. Bushel, K. Chaturvedi, D. Choi, M. L.. and Y. Wang. 2003. Effects of atmospheric ozone on mi-
Cunningham, S. Deng, H. K. Dressman, R. D. Fannin, croarray data quality. Anal. Chem. 75:4672-4675.
F. M. Farin, J. H. Freedman, R. C. Fry, A. Harper, 18. Gaillard, C., and F. Strauss. 1990. Ethanol precipitation
M. C. Humble, P. Hurban, T. J. Kavanagh, W. K. of DNA with linear polyncrylamide as carrier. Nucleic
Kaufmann, K. F. Kerr, L. Jing, J. A. Lapidus, M. R. Acids Res. 18:378.
992 MYCOLOGY

19. Gasch, A. P., M. Huang, S. Metzner, D. Botstein, S. J. proteomic analyses of a systematically perturbed metabolic
Elledge, and P. 0. Brown. 2001. Genomic expression re- network. Science 292:929-934.
sponses to DNA-damaging agents and the regulatory role 30. Irizarry, R. A., D. Warren, F. Spencer, I. F. Kim, S.
of the yeast ATR homolog Meclp. Mol. Biol. Cell 12: Biswal, B. C. Frank, E. Gabrielson, J. G. Garcia, J.
2987-3003. Geoghegan, G. Germino, C. Griffin, S. C. Hilmer, E.
20. Gasch, A. P., P. T. Spellman, C. M. Kao, 0. Carmel- Hoffman, A. .E. Jedlicka, E. Kawasaki, F. Martinez-
Harel, M. B. Eisen, G. Storz, D. Botstein, and P. 0. Murillo, L. Morsberger, H. Lee, D. Petersen, J.
Brown. 2000. Genomic expression programs in the re- Quackenbush, A. Scott, M. Wilson, Y. Yang, S. Q. Ye,
sponse of yeast cells to environmental changes. Mol. Biol. and W. Yu. 2005. Multiple-laboratory comparison of mi-
Cell 11:4241-4257. croarray platforms. Nut. Methods 2:345-350.
21. Giaever, G., A. M. Chu, L. Ni, C. Connelly, L. Riles, S. 31. Iyer, V. R., C. E. Horak, C. S. Scafe, D. Botstein, M.
Veronneau, S. Dow, A. Lucau-Dada, K. Anderson, 8. Snyder, and P. 0. Brown. 2001. Genomic binding sites of
Andre, A. P. Arkin, A. Astromoff, M. El-Bakkoury, R. the yeast cell-cycle transcription factors SBF and MBE
Bangham, R. Benito, S. Brachat, S. Campanaro, M. Nature 409:533-538.
Curtiss, K. Davis, A. Deutschbauer, K. D. Entian, P. 32. Jelinsky, S. A., P. Estep, G. M. Church, and L. D.
Flaherty, F. Foury, D. J. Garfinkel, M. Gerstein, D. Samson. 2000. Regulatory networks revealed by transcrip-
Gotte, U. Guldener, J. H. Hegemann, S. Hempel, Z. tional profiling of damaged Saccharomyces cerevisiae cells:
Herman, D. F. Jaramillo, D. E. Kelly, S. L. Kelly, P. Rpn4 links base excision repair with proteasomes. Mol.
Kotter, D. LaBonte, D. C. Lamb, N. Lan, H. Liang, Cell. Biol. 20:s 157-81 67.
H. Liao, L. Liu, C. Luo, M. Lussier, R. Mao, P. Menard, 33. Jelinsky, S. A., and L. D. Samson. 1999. Global response
S. L. Ooi, J. L. Revuelta, C. J. Roberts, M. Rose, P. of Saccharomyces cerevisiae to an alkylating agent. Proc.
Ross-Macdonald, B. Scherens, G. Schimmack, B. Nutl. Acud. Sci. USA 96:1486-1491.
Shafer, D. D. Shoemaker, S. Sookhai-Mahadeo, R. K. 34. Johannes, G., M. S. Carter, M. B. Eisen, P. 0. Brown,
Storms, J. N. Strathern, G. Valle, M. Voet, G. and P. Sarnow. 1999. Identification of eukaryotic mRNAs
Volckaert, C. Y. Wang, T. R. Ward, J. Wilhelmy, E. A. that are translated at reduced cap binding complex eIF4F
Winzeler, Y. Yang, G. Yen, E. Youngman, K. Yu, H. concentrations using a cDNA microarray. Proc. Natl.
Bussey, J. D. Boeke, M. Snyder, P. Philippsen, R. W. Acad. Sci. USA 96:13118-13123.
Davis, and M. Johnston. 2002. Functional profiling of the 35. Kohrer, K., and H. Domdey. 1991. Preparation of high
Saccharomyces cerevisiue genome. Nature 418:387-391. molecular weight RNA. Methods Enumol. 194:398-405.
22. Griffin, T. J., S. P. Gygi, T. Ideker, B. Rist, J. Eng, L. 36. Kuhn, K. M., J. L. DeRisi, P. 0. Brown, and P. Sarnow.
Hood, and R. Aebersold. 2002. Complementary profiling 2001. Global and specific translational regulation in the
of gene expression at the transcriptome and proteome lev- genomic response of Saccharomyces cerevisiue to a rapid
els in Saccharomyces cereoisiae. Mol. Cell. Proteomics transfer from a fermentable to a nonfermentable carbon
1:323-333. source. Mol. Cell. Biol. 21:916-927.
23. Hinnebusch, A. G. 1997. Translational regulation of yeast 37. Kurdistani, S. K., D. Robyr, S. Tavazoie, and M.
GCN4: a window o n factors that control initiator-tRNA Grunstein. 2002. Genome-wide binding map of the his-
binding to the ribosome. J. Biol. Chem. 272:21661-21664. tone deacetylase Rpd3 in yeast. Nut. Genet. 31:248-254.
24. Hinnebusch, A. G., and K. Natarajan. 2002. Gcn4p, a 38. Larkin, J. E., B. C. Frank, H. Gavras, R. Sultana, and
master regulator of gene expression, is controlled at multi- J. Quackenbush. 2005. Independence and reproducibility
ple levels by diverse signals of starvation and stress. Eukar- across microarray platforms. Nut. Methods 2:337-344.
yot. Cell 1:22-32. 39. Lee, C., and M. Roy. 2004. Analysis of alternative splic-
25. Holloway, A. J., R. K. van Laar, R. W. Tothill, and ing with microarrays: successes and challenges. Genome
D. D. Bowtell. 2002. Options available-from start to fin- Biol. 5:231.
ish-for obtaining data from DNA microarrays 11. Nut. 40. Lee, T. I., N. J. Rinaldi, F. Robert, D. T. Odom, Z. Bar-
Genet. 32(Suppl.) :48 1-489. Joseph, G. K. Gerber, N. M. Hannett, C. T. Harbison,
26. Hughes, T. R., M. Mao, A. R. Jones, J. Burchard, M. J. C. M. Thompson, I. Simon, J. Zeitlinger, E. G. Jennings,
Marton, K. W. Shannon, S. M. Lefkowitz, M. Ziman, H. L. Murray, D. B. Gordon, B. Ren, J. J. Wyrick, J. B.
J. M. Schelter, M. R. Meyer, S. Kobayashi, C. Davis, H. Tagne, T. L. Volkert, E. Fraenkel, D. K. Gifford, and
Dai, Y. D. He, S. B. Stephaniants, G. Cavet, W. L. R. A. Young. 2002. Transcriptional regulatory networks in
Walker, A. West, E. Coffey, D. D. Shoemaker, R. Saccharomyces cerevisiue. Science 298:799-804.
Stoughton, A. P. Blanchard, S. H. Friend, and P. S. 41. Lieb, J. D., X. Liu, D. Botstein, and P. 0. Brown. 2001.
Linsley. 2001. Expression profiling using microarrays fab- Promoter-specific binding of Rap1 revealed by genome-
ricated by an ink-jet oligonucleotide synthesizer. Nut. Bio- wide maps of protein-DNA association. Nut. Genet. 28:
technol. 19342-347. 327-334.
27. Hughes, T. R., M. J. Marton, A. R. Jones, C. J. Roberts, 42. Lin, D., D. L. E b b , and J. R. Yates 111. 2003. Large-scale
R. Stoughton, C. D. Armour, H. A. Bennett, E. Coffey, protein identification using mass spectrometry. Biochim.
H. Dai, Y. D. He, M. J. Kidd, A. M. King, M. R. Meyer, Biophys. Acta 1646:l-10.
D. Slade, P. Y. Lum, S. B. Stepaniants, D. D. 43. Lockhart, D. J., H. Dong, M. C. Byrne, M. T. Follettie,
Shoemaker, D. Gachotte, K. Chakraburtty, J. Simon, M. M. V. Gallo, M. S. Chee, M. Mittmann, C. Wang, M.
Bard, and S. H. Friend. 2000. Functional discovery via a Kobayashi, H. Horton, and E. L. Brown. 1996.
compendium of expression profiles. Cell 102: 109-126. Expression monitoring by hybridization to high-density
28. Hughes, T. R., C. J. Roberts, H. Dai, A. R. Jones, M. R. oligonucleotide arrays. Nut. Biotechnol. 14: 1675-1680.
Meyer, D. Slade, J. Burchard, S. DOW,T. R. ward, M. J. 44. Lum, P. Y., C. D. Armour, S. B. Stepaniants, G. Cavet,
Kidd, S. H. Friend, and M. J. Marton. 2000. Widespread M. K. Wolf, J. S. Butler, J. C. Hinshaw, P. Garnier,
aneuploidy revealed by DNA microarray expression profil- G. D. Prestwich, A. Leonardson, P. Garrett-Engele,
ing. Nut. Genet. 25:333-337. C. M. Rush, M. Bard, G. Schimmack, J. W. Phillips, C. J.
29. Ideker, T., V. Thorsson, J. A. Ranish, R. Christmas, J. Roberts, and D. D. Shoemaker. 2004. Discovering modes
Buhler, J. K. Eng, R. Bumgarner, D. R. Goodlett, R. of action for therapeutic compounds using a genome-wide
Aebersold, and L. Hood. 2001. Integrated genomic and screen of yeast heterozygotes. Cell 116:121-137.
45. DNA Microarray Technology n 993

45. Luscombe, N. M., T. E. Royce, P. Bertone, N. Echols, with a complementary DNA microarray. Science 270:467-
C. E. Horak, J. T. Chang, M. Snyder, and M. Gerstein. 470.
2003. Expressyourself: a modular platform for processing 61. Sellers, J. W., A. C. Vincent, and K. Struhl. 1990.
and visualizing microarray data. Nucleic Acids Res. 31: Mutations that define the optimal half-site for binding
3477-3482. yeast GCN4 activator protein and identify an ATF/CREB-
46. Marton, M. J., J. L. DeRisi, H. A. Bennett, V. Iyer, like repressor that recognizes similar DNA sites. Mol. Cell.
M. R. Meyer, C. J. Roberts, R. Stoughton, J. Burchard, Biol. 10:5077-5086.
D. Slade, H. Dai, D. E. Bassett, L. H. Haratwell, P. 0. 62. Serikawa, K. A., X. L. Xu, V. L. MacKay, G. L. Law, Q.
Brown, and S. H. Friend. 1998. Drug target validation Zong, L. P. Zhao, R. Bumgarner, and D. R. Morris.
and identification of secondary drug target effects using 2003. The transcriptome and its translation during recov-
DNA microarrays. Nut. Med. 4:1293-1301. ery from cell cycle arrest in Sacchuromyces cerevisiae. Mol.
47. Murray, A. E., D. Lies, G. Li, K. Nealson, J. Zhou, and Cell. Proteomics 2:191-204.
J. M. Tiedje. 2001. DNA/DNA hybridization to microar- 63. Shannon, W., R. Culverhouse, and J. Duncan. 2003.
rays reveals gene-spe differences between closely re- Analyzing microarray data using cluster analysis.
lated microbial genomes. Proc. Nutl. Acud. Sci. USA Phurmucogenomics 4:4 1-5 2.
98:9853-9858. 64. Shi, L., L. H. Reid, W. D. Jones, R. Shippy, J. A.
48. Natarajan, K., M. R. Meyer, B. M. Jackson, D. Slade, Warrington, S. C. Baker, P. J. Collins, F. de Longueville,
C. J. Roberts, A. G. Hinnebusch, and M. J. Marton. E. s. Kawasaki, K. Y. Lee, Y. Luo, Y. A. Sun, J. c.
2001. Transcriptional profiling shows that Gcn4p is a mas- Willey, R. A. Setterquist, G. M. Fischer, W. Tong, Y. P.
ter regulator of gene expression during amino acid starva- Dragan, D. J. Dix, F. W. Frueh, F. M. Goodsaid, D.
tion in yeast. Mol. Cell. Biol. 21:4347-4368. Herman, R. V. Jensen, C. D. Johnson, E. K. Lobenhofer,
49. Pascual-Ahuir, A., R. Serrano, and M. Proft. 2001. The R. K. Puri, U. Schrf, J. Thierry-Mieg, C. Wang, M.
Skolp repressor and Gcn4p activator antagonistically Wilson, P. K. Wolber, L. Zhang, S. Amur, W. Bao, C. C.
modulate stress-regulated transcription in Succhuromyces Barbacioru, A. B. Lucas, V. Bertholet, C. Boysen, B.
cerevisiue. Mol. Cell. Biol. 21:1625. Bromley, D. Brown, A. Brunner, R. Candles, X. M. Cao,
50. Patterson, S. D., and R. H. Aebersold. 2003. Proteomics: T. A. Cebula, J. J. Chen, J. Cheng, T. M. Chu, E.
the first decade and beyond. Nut. Genet. 33(Suppl):311- Chudin, J. Corson, J. C. Corton, L. J. Croner, C. Davies,
323. T. S. Davison, G. Delenstarr, X. Deng, D. Dorris, A. C.
51. Patterson, T. A., E. K. Lobenhofer, S. B. Fulmer- Eklund, X. H. Fan, H. Fang, S. Fulmer-Smentek, J. C.
Smentek, P. J. Collins, T. M. Chu, W. Bao, H. Fang, Fuscoe, K. Gallagher, W. Ge, L. Guo, X. Guo, J. Hager,
E. S. Kawasaki, J. Hager, I. R. Tikhonova, S. J. Walker, P. K. Haje, J. Han, T. Han, H. C. Harbottle, S. C.
L. Zhang, P. Hurban, F. de Longueville, J. C. Fuscoe, W. Harris, E. Hatchwell, C. A. Hauser, S. Hester, H.
Tong, L. Shi, and R. D. Wolfinger. 2006. Performance Hong, P. Hurban, S. A. Jackson, H. Ji, C. R. Knight,
comparison of one-color and two-color platforms within W. P. Kuo, J. E. LeClerc, S. Levy, Q. Z. Li, C. Liu, Y.
the MicroArray Quality Control (MAQC) project. Nut. Liu, M. J. Lombardi, Y. Ma, S. R. Magnuson, B.
Biotechnol..24:1140-1150. Maqsodi, T. McDaniel, N. Mei, 0. Myklebost, B. Ning,
52. Phizicky, E., P. I. H. Bastidens, H. Zhu, M. Snyder, and N. Novoradovskaya, M. S. Orr, T. W. Osborn, A.
S. Fields. 2003. Protein analysis on a proteomic scale. Papallo, T. A. Patterson, R. G. Perkins, E. H. Peters, R.
Nature 422:208-2 15. Peterson, et al. 2006. The MicroArray Quality Control
53. Pokholok, D. K., C. T. Harbison, S. Levine, M. Cole, (MAQC) project shows inter- and intraplatform repro-
N. M. Hannett, T. I. Lee, G. W. Bell, K. Walker, P. A. ducibility of gene expression measurements. Nut. Biotech-
Rolfe, E. Herbolsheimer, J. Zeitlinger, F. Lewitter, D. K. nol. 24:1151-1161.
Gifford, and R. A. Young. 2005. Genome-wide map of 65. Singh-Gasson, S., R. D. Green, Y. Yue, C. Nelson, F.
nucleosome acetylation and methylation in yeast. Cell 122: Blattner, M. R. Sussman, and F. Cerrina. 1999. Maskless
517-527. fabrication of light-directed oligonucleotide microarrays
54. Quackenbush, J. 2001. Computational analysis of micro- using a digital micromirror array. Nut. Biotechnol. 17:974-
array data. Nut. Rev. Genet. 2:418-427. 978.
55. Rajagopalan, D. 2003. A comparison of statistical meth- 66. Stekel, D. 2003. Microarray Bioinformutics, p. 62-72.
ods for analysis of high density oligonucleotide array data. Cambridge University Press, Cambridge, United Kingdom.
Bioinfonnutics 19: 1469-1476. 67. Stekel, D. 2003. Microarray Bioinformutics, p. 73-99.
56. Ren, B., F. Robert, J. J. Wyrick, 0. Aparicio, E. G. Cambridge University Press, Cambridge, United Kingdom.
Jennings, I. Simon, J. Zeitlinger, J. Schreiber, N. 68. Tan, P. K., T. J. Downey, E. L. Spitznagel, Jr., P. Xu,
Hannett, E. Kanin, T. L. Volkert, C. J. Wilson, S. P. D. Fu, D. S. Dimitrov, R. A. Lempicki, B. M. Raaka,
Bell, and R. A. Young. 2000. Genome-wide location and and M. C. Cam. 2003. Evaluation of gene expression mea-
function of DNA binding proteins. Science 290:2306- surements from commercial microarray platforms. Nucleic
2309. Acids Res. 315676-5684. ,
57. Robyr, D., Y. Suka, 1. Xenarios, S. K. Kurdistani, A. 69. Tseng, G. C., M. K. Oh, L. Rohlin, J. C. Lido, and
Wang, N. Suka, and M. Grunstein. 2002. Microarray W. H. Wong. 2001. Issues in cDNA microarray analysis:
deacetylation maps determine genome-wide functions for quality filtering, channel normalization, models of vnria-
yeast histone deacetylases. Cell 109:437-446. tions and assessment of gene effects. Nucleic Acids Res.
58. Sambrook, J., and D. W. Russel. 2001. Molecular Cloning: 29:2549-2557.
u Laboratory Munual, 3rd ed. Cold Spring Harbor Labor- 70. Tusher, V. G., R. Tibshirani, and G. Chu. 2001. Sig-
atory, Cold Spring Harbor, NY. nificance analysis of microarrays applied to the ionizing ra-
59. Schadt, E. E., C. Li, B. Ellis, and W. H. Wong. 2001. diation response. Proc. Nutl. Acud. Sci. USA 985116-5121.
Feature extraction and normalization algorithms for high- 71. van de Peppel, J., P. Kemmeren, H. van Bakel, M.
density oligonucleotide gene expression array data. J. Cell. Radonjic, D. van Leenen, and F. C. P. Holstege. 2003.
Biochem. Suppl. 37:120-125. Monitoring global messenger RNA changes in externally
60. Schena, M., D. Shalon, R. W. Davis, and P. 0. Brown. controlled microarray experiments. EMBO Rep. 4:387-
1995. Quantitative monitoring of gene expression patterns 393.
994 MYCOLOGY

72. Van Gelder, R. N., M. E. von Zastrow, A. Yool, W. C. strains: DNA microarray-based comparative genomic hy-
Dement, J. D. Barchas, and J. H. Eberwine. 1990. Ampli- bridization using a yeast DNA microarray with 6000 genes.
fied RNA synthesized from limited quantities of heteroge- Yeast 21:351-365.
neous cDNA. Proc. Natl. Acad. Sci. USA 87:1663-1667. 76. Wu, H., K. M. Kerr, X. Cui, and G. A. Churchill. 2003.
73. Wang, Y., C. L. Liu, J. D. Storey, R. J. Xbshirani, D. MAANOVA: a software package for the analysis of spot-
Herschlag, and P. 0. Brown. 2002. Precision and func- ted cDNA microarray experiments, p. 313-341. In
tional specificity in mRNA decay. Proc. Natl. Acad. Sci. G. Parmigiani, E. S. Garett, R. A. Irizarry, and S. L. Zeger
USA 99:5860-5865. (ed.), The Analysis of Gene Expression Data: Methods and
74. Washburn, M. P., A. Koller, G. Oshiro, R. R. Ulaszek, Software. Springer, New York, NY.
D. Plouffe, C. Deciu, E. Winzeler, and J. R. Yates 111. 77. Wyrick, J. J., J. G. Aparicio, T. Chen, J. D. Barnett,
2003. Protein yathway and complex clustering of corre- E. G. Jennings, R. A. Young, S. P. Bell, and 0. M.
lated mRNA and protein expression analyses in Aparicio. 2001. Genome-wide distribution of ORC and
Saccharomyces cereuisiae. Proc. Natl. Acad. Sci. USA 100: MCM proteins in S. cerevisiae: high-resolution mapping of
3 107-3 112. replication origins. Science 294:2357-2360.
75. Watanabe, T., Y. Murata, S. Oka, and H. Iwahashi. 75. Yang, Y. H., and T. Speed. 2002. Design issues for cDNA
2004. A new approach to species determination for yeast microarray experiments. Nat. Reu. Genet. 3579-588.
COLOR PLATE 3 (chapter 2 ) Gram stain of Bacillus subtilis
that has been incubated with egg white lysozyme to digest the
peptidoglycan in the cell wall. Some cells have lysed and there-
fore stain as gram negative (red), whereas others remain intact
and stain as gram positive (purple). Scale bar, 5 pm. (Kindly
prepared and supplied by R. Van Twest.)
COLOR PLATE 5 (chapter 3 ) (A) Three-channel 1P-LSM 2-series projected as a stereo pair
showing the colocalization of binding sites for three lectins, Triticum uulgaris-TRITC (red),
Lycopersicum esculentum-FITC (green), and Tetragonolobus purpureas-Cy5 (blue), in a river biofilm.
A variety of microcolonies and individual cells can be seen to bind single or combinations of the
lectins as indicated by the color coding of the area. (B) High-resolution imaging of a microcolony
stained with the lectin conjugates Solanum tuberosum-FITC (green), Cicer arietinum-Alexa568
(red), and Tetragonolobus purpureas-Cy5 (blue). (C) Wisteria floribundu-FITC (green), Lens culinaris-
TRITC (red), and Canaualia ensifomis-Cy5 (blue). These three-channel images show the details of
binding and structure of the multicomponent polymer surrounding individual bacterial cells in a mi-
crocolony. (A color wheel is included to allow interpretation of stain combinations.)
COLOR PLATE 6 (chapter 3 ) Multiparameter image showing the distribution of bacteria
(green), algae (autofluorescence red), and exopolymeric substances stained with Trticum vulgaris
lectin (blue). The image stack was projected with one version of the stack offset and aligned to form
a stereo pair.
COLOR PLATE 7 (chapter 4) An example of the use of ESI, TEM, and three-dimensional re-
construction (see chapter 5) to determine the location of rRNA (by following the phosphorus line)
on the surface of the small (30s) ribosomal subunit of E. coli. A 150- t9-eV loss (green) and NetP
(orange) are shown in four views which are related to one another by 90" rotations about the long
axis of the subunit. In panel a the reconstructions are represented as solid surfaces, in panel b only
the NetP reconstructions are shown, and in panel c both reconstructions are shown as wire mesh
models. Spheres with numbers indicate positions of proteins mapped by neutron diffraction in stud-
ies by other researchers. In the first column of images, the intersubunit face of the small subunit is
facing the viewer. The bottom of the subunit is shown to be phosphorus rich (i.e., rRNA rich).
(From D. R. Beniac, G. J. Czarnota, B. L. Rutherford, F. P. Ottensmeyer, and G. Harauz,J. Microsc.
188:24-35, 1997.)
COLOR PLATE 8 (chapter 5) As described in reference 23, an atomic model of the S. marinus
phosphoenolpyruvate synthase subunit was constructed by using Insight software (i) and 24 subunits
were computationally matched to the 3-D reconstruction in Fig. 8d. Here, the view down the three,
fold axis of rotational symmetry shows the enzymatically active sites of each subunit clustered
around a putative channel. This figure was provided by F. P. Ottensmeyer, Ontario Cancer Institute,
Toronto, Canada.
COLOR PLATE 9 (chapter 16) Diversity of symbiosis. (A) Anaerobic methane-oxidizing con-
sortium (FISH image; red, archaeal group; green, sulfate-reducing bacteria (credit: V. Orphan,
Caltech). Scale bar, 10 pm. (B) Nematode (Eubostrichus dianae) with epibionts (credit: M. Polz,
MIT, and M. Bright, University of Vienna). Scale bar, 100 pm. (C) Riftiu pachyptila tube worms liv-
ing near a deep-sea hydrothermal vent (credit: S.Goffredi, Monterey Bay Aquarium Research
Institute). (D)Termite, Zootermopsis nevudensis (credit: Amy Vu, Caltech). (E) Hawaiian bobtail
squid, Euprymna scolopes. Vibrio fischeri-containing light organ is located o n the ventral side of man-
tle cavity (not shown) (credit: M. Mcfall-Ngai, University of Wisconsin at Madison). (F) GFP- and
dsRed-labeled Sinorhizobium meliloti inside infection threads of alfalfa. Clonal selection during the
development of the root nodule is evident due to distinct GFP and dsRed-S. meliloti in each infec-
tion thread. Scale bar, 50 p,m. [credit: D. Gage, University of Connecticut (61)].(G) GFP-labeled
Photorhabdus luminescens in the intestine of an infective juvenile nematode, Heterorhabditisbacterio-
phora. The nematode and bacteria cooperate in insect pathogenesis (credit: T. Ciche, Michigan
State University), (H) The gutless marine annelid Inanidrilus kukodermatus (credit: A. Blazejak,
Max Planck Institute).
COLOR PLATE 10 (chapter 16) (A) Transmission electron micrograph of the symbiont-
containing region of Inunidrilus kukodermatus. Smaller and larger arrows indicate smaller and larger
symbiont morphotypes, respectively. Scale bar, 2 pm. cu, cuticle (44). (B) In situ epifluorescence
identification of bacterial symbionts in 0.
mussitmicutus. Cross sections through the entire worm.
Dual hybridization with the GAM42a and DSS658/DSR65 1 probes, showing y-proteobacterial
symbionts (red) and 6-proteobacterial symbionts (green). Scale bar, 20 ( p m (12).

COLOR PLATE 1 1 (chapter 16) mRNA FISH on gill fila-


ment of the mussel Bathymodiolus puteoscrpentis. Blue,
thiotrophic symbiont labeled with a specific 16s rRNA probe;
red, methanotrophic symbiont labeled with a specific 16s
rRNA probe; green, activity of particulate methane monooxy-
genase of the symbiont (subunit A; pmoA), a key enzyme of
aerobic methane oxidation (creclit: Annelie lernthaler, Max
Planck Institute).
A. -
ACAG GAGCGTTTCCTGGGCAGACTCGGCCGTGGTTCTTI E

C A A A T A C C A C T G A G A T C C C TAG T A A C G G C C G C C A G TG T G C T
. 4000, . 4080, . 4160, . 4240, . 4320, 4400,

I C I: TCG G G G G R A AAI: G T C G TCAh G C T G Ah A G C C C C T T T G I


n 343 351 361 3

000, 2080, 21 60, 2240,


- , ' .
2320, 2400,
1 --
2480,
1
1600, 1680, 1760, 1840, 1920, 20
C T A T T A C C G T I I T C C G AC TG G I:T A C C A T C G T T G T G G A T G AG C
C T A A G C C C T T A T C C T A T A C TG T A T A C T T T G C T T C G A A A G
c. =

COLOR PLATE 12 (chapter 27) DNA sequence chromatogram tracings. Screen shots were
taken during the running of Sequencher software (Gene Codes Corp., Ann Arbor, MI). The se-
quence identified by the computer program is given above each tracing. (A) Normal sequence; (B)
double peaks from a mixed clone; (C) failure with dirty template; (D) heterozygote template; (E)
failure with high salt contamination; (F) blockage possibly due to hairpin formation.
COLOR PLATE 13 (chapter 3 3 ) Halobacterium sp. NRC-1.
(A) Colonies of the model haloarchaeal strain Halobacterium
sp. NRC-1. Among pink wild-type (NRC-1) colonies, an or-
ange gas vesicle-minus (Vac-) mutant is visible. (B) Liquid
cultures ofNRC-1 (tube l),Vac- mutant SD109 (tube Z), and
a SD109 transformant containing the gas vesicle gene cluster
on pFL2 (tube 3). Vac+ floating cells at the top of tubes 1 and
3 and Vac- nonfloating cells at the bottom of tube 2 are visi-
ble. The meniscus is visible at the top of tube 2.

sample

epifluorescence
/ microscopy
fiation
J permeabilization

detection 2
00-
0 0 A Ps,
ri o omes

fixed cells,
permeabilized hybridized cells

L hybridization
9- 4 cy
t
fluorescently labeled
--+9
washing
oligonucleotides (probes)
COLOR PLATE 14 (chapter 39) General protocol of whole-cell fluorescence in situ hybridiza-
tion.
Pirellula strain 1 Saccharomyces cerevisiae

class^: 81-100% classW: 2140%


H classn: 61-80% classv: 6-20%
r]
1- class m: 41-60% classVI: 0-5%
COLOR PLATE 15 (chapter 39) In situ accessibility of the 16s rRNA of E. coli
(Enterobacteriaceae),Pirellula sp. strain 1 (Planctomycetales), M. sedula (Archaea), and S. cerevisiue
(Eukarya) for fluorescently labeled oligonucleotide probes. Different colors indicate differences in
accessibility. From reference 8 with permission.
class I: S ~ - I O O %
class I 1 61-80%
class 111 41-60%
class I V 21.40%
class V 6.20%
class VI: 0-5%

COLOR PLATE 16 (chapter 39) In situ accessibility of the 23s rRNA of E. coli. Different col-
ors indicate differences in accessibility for oligonucleotide probes. From reference 17 with permis-
sion.
A.
0 A B C D E F G H I I K L U N O P Q R S
1 Samplelnto A 0 1 1 A 0 2 1 A031 A 0 4 1 A 0 5 1 A071 A091 A l l 1 A121 A 1 4 1 A161 A 1 8 1 A 1 9 1 A 2 0 1 A221 A231 A241 A251

88 109.92
0.78 8
7 NZH 124.28
8 NSH 124.05
9 N4H 57.22 57.22 60.00 64.04 6601 81 8 1 83.60 92.16 9403 95 80 124.0s
10 TlH 57.47 59.02 6235 6 6 0 8 77 0 5 81 8 9 89.75 92 79 94.83 109 8 9
11 T2H 59 07 61 4 4 65 11 8071 8262 9211 94 109.9
12 T3H 5907 6015 51 44 65 32 8071 8252 9211 9422 9422 109.9
13 T4H 58.92 61 07 6 4 95 806 J 91.88 94

B.

6 /N2H 0 1 1 0 1 1 1 0 1 1 1 1 1 1 0 0 0 1
0 1 1 1 1 0 1 0 0 0 1
0 1 1 1 1 0 1 0 0 0 1
9 T1H 0 1 1 0 1 0 1 1 1 1 1 1 0 0 0 1 0 0
10 T2H 0 0 1 0 1 0 1 0 1 1 1 1 0 0 0 0 1 0
11 T3H 0 0 1 1 1 0 1 0 1 1 1 1 1 0 0 0 1 0
12 T4H 0 0 1 0 1 0 1 0 1 1 1 1 0 0 0 0 0 0

C.

N2 0.488 0.56 0.56 0.5 0.979


N3 0.488 0.58 0.54 0.48 0.958 0.96
N4
IT1
T2
0.349
0.5
0.558
0.47
0.48
0.67
0.4
0.43
0.58
0.44
0.45
0.62
0.867
0.475
0.533

0.87 0.84
0.45 0.43 0.58
0.58 0.6 0.47
1
0.8 1
T3 0.558 0.66 0.58 0.59 0.479 0.52 0.5 0.42 0.825 0.91 1
T4 0.558 0.73 0.58 0.61 0.5 0.55 0.55 0.41 0.7 0.86 0.96 1

COLOR PLATE 17 (chapter 41) An example of analysis of T-RFLP data from three soils. Each
soil type (H, N, and T ) is presented with four replicates. (A) Aligned profiles from 12 samples taken
into ExcelTM.Each profile includes the estimated size for each detected fragment. The checks indi-
cate that a fragment was detected by visual inspection of the electropherogram but was not scored
by the software because the peak amplitude value was below the preset limit. These are recorded as
present. (B) The data set from panel A converted into binary format (presence or absence) for
similarity comparisons. (C) Similarity analysis of the 12 samples with the T-RFLP function of the
RDP. (D) UPGMA analysis of the 12 samples with PAUP (http://paup.csit.fsu.edu/).
COLOR PLATE 18 (chapter 44) (Top left) Replica plating block used for replication of A. nidu-
lam colonies. (Top right) A. nidulans cleistothecia appearing on mycelium near the mycelial fron-
tier. (Bottom left) A. nidulans cleistothecia. The needle shaft is about 1 mm wide. The cleistothe-
cium to its immediate left is uncleaned, with adhering Hulle cells and other debris. The
cleistothecium to the far left has been cleaned by rolling on the agar. (Bottom right) Multipin repli-
cator (to the right) used for rapid transfer of arrays of A. nidulans colonies. The colonies on the
upper plate have grown after spreading conidia. Colonies are sampled individually from this plate
and grown in the array shown in the lower plate. This will be the master plate used for transfers
made with the multipin replicator.
AUTHOR INDEX

Index Terms Links

Actis, Luis A. 709


Adney, William S. 596
Amann, Rudolf 886

Baker, John O. 595


Barkley, W. Emmett 997
Baushke, Sam W. 286
Beveridge, Terry J. 3 19 54
Blum, Paul H. 800
Breznak, John A. 171 309
Buckley, Daniel H. 897

Cecchini, Gary 539


Ciche, Todd A. 394
Clutterbuck, A. John 965
Costilow, Ralph N. 309
Crosa, Jorge H. 709
Cypionka, Heribert 215

Daniels, Lacy 462


DasSarma, Shiladitya 800
Davis, Rowland H. 965
de Bruijn, Frans J. 684
Decker, Stephen R. 596
DeLong, Edward E 879
Dosoretz, Carlos G. 611
Douady, Christophe J. 856
Dufrcne, Yves F. 96

This page has been reformatted by Knovel to provide easier navigation.


Index Terms Links

Dunny, Gary M. 756

Emerson, David 200

Foster, Patricia L. 676


Fuchs, Bemhard M. 886

Ghema, Robert L. 1019


Goffredi, Shana K. 394
Gunsalus, Robert P. 539

Hanson, Richard S. 462


Harauz, George 82
Harris, Bob 54
Hashsham, Syed A. 270 286
Hatfull, Graham F. 825
Hausinger, Robert P. 504
Hendrickson, William 653
Himmel, Michael E. 596
Hinnebusch, Alan G. 978

Jacobs, William R., Jr. 825


Jacobs-Sera, Deborah 825
Johnson, John L. 624

Karl, David M. 869


Koch, Arthur L. 172
Koval, Susan F. 108
Krieg, Noel R. 330
Kristich, Christopher J. 756

This page has been reformatted by Knovel to provide easier navigation.


Index Terms Links

Kukor, Jerome J. 586

Lachance, M. A. 929
Lam, Joseph S. 138
Larsen, Michelle H. 825
Lawrence, John R. 19 34

Marquis, Robert E. 527


Marsh, Terence L. 909
Marton, Matthew J. 978
Marzluf, George A. 927 952
Meganathan, R. 558
Mickelson, Claudia A. 997
Moyles, Dianne 54
Mulrooney, Scott B. 424
Murray, R. G. E. 5 19
Mutharia, Lucy M. 138

Nakatsu, Cindy H. 909


Natarajan, Krishnamurthy 978
Nesb, Camilla L. 856
Neu, T. R. 34

Paterek, J. R. 424
Pearson, William R. 842
Pernthaler, Jakob 886
Peters, Joseph E. 735
Phillips, Allen T. 504
Phillips, Jane A. 462

This page has been reformatted by Knovel to provide easier navigation.


Index Terms Links

Ranganathan, Yamini 558


Reddy, C. A. 423 558 611 623
1019
Robinow, Carl F. 5
Rossbach, Silvia 684

Salomon, Christine E. 756


Schmidt, Thomas M. 841 897
Schroder, Imke 539
Scott, J. 929
Sikorski, Johannes 330
Smibert, Robert M. 330
Snyder, L. R. 623
Sowers, Kevin R. 800
Sprott, G. Dennis 108

Tang, Jane 200


Teske, Andreas 215
Thorn, R. G. 929
Tndall, Brian J. 330
Tolmasky, Marcelo E. 709

Walthers, Don 653


Wawrik, Boris 586
Welch, Timothy J. 709
Whitman, William B. 624
Wood, Willis A. 424

Zylstra, Gerben J. 586

This page has been reformatted by Knovel to provide easier navigation.


APPENDICES

46. Laboratory Safety 997 47. Culture Preservation 1019


W. EMMETT BARKLEY AND ROBERT L. GHERNA AND C. A. REDDY
CLAUDIA A. MICKELSON
46
Laboratory Safetyt
W. EMMETT BARKLEY AND CLAUDIA A . MICKELSON

46.1. FEDERAL BIOSAFETY GUIDELINES AND REGULATIONS ....... 998


46.1.1. NIH Recombinant DNA Guidelines ...................... 998
.
2. Biosafety in Microbiological and Biomedical Laboratories ........ 999
.
3. OSHA Occupational Exposure to Bloodborne Pathogens Rule .... 999
.
4. HHS and USDA Rules for the Possession. Use. and Transfer of
Select Agents and Toxins .............................. 999
46.2. PRINCIPLES ............................................ 1000
46.2.1. Directors' Responsibilities .............................. 1000
.
2. Biosafety Principles .................................. 1000
.
3. Facility Features ..................................... 1001
46.3. HISTORICAL PERSPECTIVE ....................... ....... 1001
.
4. BIOSAFETYLEVELS ...................................... 1002
.
5. LEVEL 2 BIOSAFETY PRACTICES ........................... 1002
46.5.1. Personal Hygiene .................................... 1004
.
2. Pipetting .......................................... 1004
.
3. Hypodermic Syringes and Needles ........................ 1004
.
4. Opening Containers .................................. 1005
.
5. Centrifuging ........................................ 1006
.
6. Mixing and Disruption ................................ 1007
.
7. Handling Animals .................................... 1007
.
8. Vacuum Systems .................................... 1008
.
9. Miscellaneous Operations .............................. 1008
46.6. LEVEL 3 BIOSAFETY PRACTICES ........................... 1008
.
7. BIOSAFETY CABINETS ................................... 1008
46.7.1. Class I Cabinets ..................................... 1009
.
2. Class I1 Cabinets .................................... 1009
46.8. CHEMICAL DISINFECTION ................................ 1009
46.8.1. General Guide ...................................... 1010
.
2. Disinfectant Types ................................... 1010
46.8.2.1. Alcohols ................................... 1010
.
2. Formaldehyde ................................ 1010
.
3. Phenolic Compounds .......................... 1011
.
4. Quaternary Ammonium Compounds ............... 1011
.
5. Chlorine ................................... 1011
.
6. Iodophors .................................. 1011
.
7. Heavy Metals ................................ 1011
46.8.3. Disinfectant Characteristics ............................. 1011
.
4. Environmental Surfaces ............................... 1012
.
5. Bacterial Spills ...................................... 1012
.
6. Instruments ........................................ 1013
.
7.Hands ............................................ 1013
46.9. STERILIZATION ......................................... 1013
46.9.1. Moist Heat ......................................... 1014
.
2. Dry Heat .......................................... 1014
.3.Gases ............................................. 1015
46.9.3.1. Ethylene Oxide .............................. 1015
.
2. Formaldehyde ................................ 1015
.
3. Hydrogen Peroxide ............................ 1015
.
4. Chlorine Dioxide ............................. 1015
46.9.4. Filtration .......................................... 1016

'We acknowledge the contributions of the late John H . Richardson. coauthor of the original chapter. whose words. guide
ance. and spirit remain in this revision .

997
998 APPENDICES

46.10. NONBIOLOGICAL SAFETY CONSIDERATIONS ..............1016


.11. SECURITY ............................................. 1016
.12. REFERENCES .......................................... 1016

Laboratory safety requires an awareness of the possible risks cal safety (22), radiation safety (35), physical safety (16),
associated with the handling of hazardous materials, knowl- and disinfection and sterilization (3, 31) are available.
edge of mechanisms by which exposures may occur, use of Technical handbooks on protocols in molecular biology
safeguards and techniques that reduce the potential for (6, 33) and microbiology (13) are an excellent sources of
exposures, and vigilance against compromise and error. laboratory safety information. Also a safety manual (10) is
Microorganisms, radioisotopes, and hazardous chemicals are available that can serve as an excellent example of how
invariably found in bacteriology, microbiology, and biomed- one university met the intent of regulatory standards that
ical laboratories. The risk of occupational infection in these require written safety plans. These references provide de-
laboratories is associated with the use of pathogenic micro- tailed information, which is beyond the basic scope of this
organisms or the handling of materials contaminated with chapter.
them. Biological safety and the prevention of laboratory-
acquired infection is the principal subject addressed in this
chapter. When pathogenic microorganisms are not present 46.1. FEDERAL BIOSAFETY GUIDELINES AND
in the laboratory, the risk of physical injury and accidental REGULATIONS
exposure to hazardous chemicals and radioactive materials
should be the primary concern of the laboratory worker, and 46.1 .l.NIH Recombinant DNA Guidelines
a brief discussion of these hazards is also included. The NIH Recombinant DNA Guidelines (NIH Guide-
Laboratory safety is the subject of several federal health lines) were developed in 1976 as a response to the public
and safety regulations and guidelines. The Nuclear Reg- and scientific concern that gene splicing technology
ulatory Commission has regulated the use of radioisotopes might give rise to new biological agents with unknown and
in laboratories since 1946 (47). The National Institutes of possibly dangerous characteristics. The NIH Guidelines de-
Health (NIH) established federal guidelines for the safe scribed four levels of physical containment-referred to at
conduct of research involving recombinant DNA molecules that time as P1, P2, P3, and P4-each providing a level of
in 1976 (42). In 1984 the Centers for Disease Control and protection greater than the one preceding it. The assess-
Prevention (CDC) and NIH established consensus guide- ment of potential risks used to define classes of experiments
lines for protecting the health of laboratory workers who assigned to each containment level, and the selection of
handle pathogenic microorganisms (44). The Occupational safe laboratory practices, containment equipment, and fa-
Safety and Health Administration (OSHA) established cility safeguards that describe the safety requirements for
rules in 1990 and 1991 to protect the health and safety of each level were based on the scientific knowledge and ex-
laboratory workers from occupational exposures to haz- perience acquired in bacteriological and microbiological
ardous chemicals and blood-borne pathogens, respectively laboratories conducting research and diagnostic studies in-
(48, 49). The CDC and the U S . Department of Agri- volving human pathogens. The NIH Guidelines establish a
culture (USDA) issued two new rules in 2002 as part of the multilevel review and approval process that is designed to
nations priority homeland security initiative to protect an- ensure active and robust oversight of the uses of recombi-
imal, plant, and public health from acts of bioterrorism (39, nant DNA technology. The process involves the participa-
40, 45). These rules contain provisions for controlling the tion of the principal investigator, the Institutional Biosafety
possession, security, and use of select agents and toxins to Committee, the NIH Office of Biotechnology Assessment,
safeguard the public health and to protect the health and the NIH Recombinant DNA Advisory Committee (RAC),
safety of laboratory workers who will handle these agents in and the NIH Director. The NIH Guidelines are mandatory
conducting medical and biodefense research. for institutions that receive any federal funding for research
Biological safety is an important concern for people who involving recombinant DNA technologies. The NIH also
work in bacteriology, microbiology, and biomedical labora- encourages other institutions that do not receive NIH fund-
tories. It is natural to want to protect oneself from harm, ing to adopt the NIH Guidelines voluntarily.
and thoughtful people would never knowingly do some- Recombinant DNA technology provides a powerful set
thing that would increase the risk for a colleague to contract of biological tools for identifying and isolating genes and
an occupational illness. The ability to prevent laboratory- discerning gene function. These tools are well character-
acquired infection, however, requires certain skills and ized hostlvector systems and restriction enzymes and in-
knowledge that can best be acquired through training and clude genetically engineered bacterial hosts and plasmids,
careful guidance provided by experienced colleagues. A debilitated bacteriophages and viruses, and enzymes with
study of the literature is not sufficient to prepare a labora- the capability of cutting DNA at specific sequences. The
tory worker for safely handling pathogenic microorganisms. development and use of host-vector systems that lack viru-
It is necessary to develop proficiency in microbiological lence genes, require enriched laboratory media for growth,
techniques through practice with nonpathogenic microor- and cannot colonize the human gut (9, 17) demonstrated
ganisms before higher-risk microorganisms are introduced that scientists could exploit this new technology to en-
into the laboratory routine (2). hance the safety of infectious disease research by studying
Several books provide comprehensive coverage of the the virulence and pathogenicity of infectious agents at the
practice of biological safety (9, 19,21,44). The consensus molecular level rather than by performing experiments that
guidelines published by the CDC and the NIH (44) involve handling the infectious agent. The capability to de-
should be consulted before pathogenic bacteria are intro- velop debilitated vectors also made it possible to consider
duced into the laboratory. Authoritative books on chemi- applying this technology to the conduct of human gene
46. Laboratory Safety 999

transfer experiments for the purpose of evaluating the safety vestigators to conduct risk assessments in selecting the ap-
and potential therapeutic benefits of the emerging field of propriate practices for use in their laboratories.
molecular medicine.
The NIH Guidelines were frequently revised as knowl- 46.1.3. OSHA Occupational Exposure to
edge and experience from the use of this technology in re- Bloodborne Pathogens Rule
search was acquired. The rapid gain in knowledge quickly The Occupational Safety and Health Administration
allayed the original concerns that led to the development of (OSHA) of the Department of Labor issued voluntary
the initial NIH Guidelines. NIH delegated compliance re- guidelines in 1983 to reduce the risk of occupational expo-
sponsibility to the institutions in 1978. In subsequent revi- sure to hepatitis B virus in the health care industry. In 1987
sions, NIH tasked the RAC to develop detailed and ex- the Department of Labor and the Department of Health
planatory appendices that would aid institutions in carrying and Human Services published a Joint Advisory Notice on
out their responsibilities. For example, Appendix B presents the protection against occupational exposure to hepatitis B
a classification of human etiological agents on the basis of virus (HBV) and human immunodeficiency virus (HIV).
hazard. The classification schema uses prevalence of infec- OSHA initiated rulemaking in 1987 after determining that
tion and severity of disease as the primary indicators of haz- over 2,000,000 health care workers in the United States,
ard. Investigators should review Appendix B when deter- including 500,000 laboratory workers, were at risk for occu-
mining the appropriate biosafety level for experiments pational exposure to hepatitis B virus (46). These agencies
involving the introduction of recombinant DNA into a estimated that 12,000 cases of HBV infection occur in
human pathogen or the cloning of DNA from a human health care workers each year, that an estimated 10% of
pathogen into a nonpathogenic prokaryotic or lower eu- these patients become chronic HBV carriers, and that 200
karyotic host-vector system. The current primary role of the to 300 fatalities occur as a result of HBV-related disease.
RAC is to provide guidance to investigators and IBCs on Other blood-borne pathogens (such as human immunodefi-
the scientific and ethical issues associated with human gene ciency virus and the causative agents of syphilis, malaria,
transfer experimentation and to maintain the relevancy and and hepatitis C) have similar transmission patterns and may
applicability of the NIH Guidelines to modern biological pose an occupational hazard for health care workers. The
research. NIH also changed the P designations for physical OSHA blood-borne pathogens standard was published on
containment to correspond with the biosafety level (BL) December 6, 1991, and became effective on March 6, 1992
nomenclature adopted by CDC and NIH in 1984 for new (49).
consensus guidelines for the safe use of human pathogens in The OSHA blood-borne pathogens standard has had a
microbiological and biomedical laboratories (44). far-reaching effect on most of the biological research labo-
ratories in the United States. Careful consideration was
46.1.2. Biosafety in Microbiological and Biomedical given to the possible risks inherent in a wide range of
laboratories human-derived materials, including primary cells, tissues,
Occupational health and safety, environmental, and com- organs, and established human cell lines. The inclusion of
munity protection must be integral components of the rou- established human cell lines in the regulatory framework in
tine practices and procedures of research laboratories. It is 1994 has extended the requirements of the OSHA
the responsibility of institutions, investigators, and staff to Standard into basic research and molecular biology labora-
encourage safety and promote the safe conduct of research. tories, far from the health care and clinical diagnostic set-
Following the development of the NIH Guidelines, the tings originally considered in the preamble to the regula-
CDC and NIH recognized the value in promoting biosafety tion. The OSHA blood-borne pathogens standard requires
more broadly among diagnostic and research laboratories institutions, investigators, and laboratory safety personnel
that handle human pathogens and zoonotic agents. CDC to work together to assess the risks inherent in the research;
and NIH formed a partnership to develop a voluntary code develop control and containment practices and procedures,
of practice that would provide authoritative guidance for including the offer of a very safe vaccine for individuals at
protecting the health and safety of laboratory workers and risk from their work; and ensure awareness and expertise
the public health from hazards associated with the posses- through training. The concept of universal precautions, the
sion and use of infectious materials in microbiological and safety practices and procedures outlined in the OSHA
biomedical laboratories. CDC and NIH invited scientists blood-borne pathogens standard for work with human ma-
and laboratory directors, experienced laboratory workers, terials, cells, and cell lines are consistent with the CDC and
and health and safety professionals to work collaboratively NIH definition of biosafety level 2 practices and procedures.
in developing consensus practices for the code. CDC and The 4th edition of Biosafety in Microbiological and Biomedical
NIH published the first edition of the code of practice, Laboratories contains a section on containment for working
Biosafety in Microbiological and Biomedical Laboratories with human cells and tissues.
(BMBL) in 1984 (44). They published the fourth edition in
1999 and the fifth edition in 2007. 46.1.4. HHS and USDA Rules for the Possession,
This code of practice is an excellent resource. I t de- Use, and Transfer of Select Agents and Toxins
scribes four biosafety levels that parallel the four contain- The September 11, 2001 terrorist attack on the United
ment levels of the NIH Guidelines. Recommendations for States and the subsequent acts of bioterrorism involving the
the laboratory control and containment of specific agents dissemination of Bacillus anthracis exposed the vulnerability
and their use in small laboratory animals is based on the po- of the United States to terrorism, and the risk to humans,
tential hazard of the agent, the mode of transmission, the animals, and plants from the use of dangerous biological
severity of the disease, and the laboratory activity. The agents in domestic or international terrorism. Two new laws
BMBL provides consensus recommendations on laboratory strengthen the nations ability to protect the public health
practices and techniques, safety equipment and primary and safety from future bioterrorism threats. The USA
barriers, and facility features for each of the four biosafety Patriot Act of 2001 strengthens the criminal laws against
levels. The BMBL encourages laboratory directors and in- terrorism and makes it illegal to possess any biological agent
1000 APPENDICES

or toxin, including any genetically engineered organism for the laboratory is helpful in clearly defining safety expec-
created using recombinant DNA technology, for any pur- tations and setting a positive attitude that will sustain a
pose not reasonably justified by a prophylactic, protective, high level of safety awareness. The references cited above
bona fide research, or other peaceful purpose. The act also emphasize the importance and value of safety manuals that
defines who may transfer and possess dangerous pathogens are relevant to the actual work activities of the laboratory
and toxins. The Public Health Security and Bioterrorism and provide excellent guidance for developing such manu-
and Response Act of 2002 improves the ability of the als.
United States to prevent, prepare for, and respond to bioter- The laboratory director has a legal obligation to prepare
rorism and other public health emergencies. The primary and use a safety manual or plan if the laboratory is involved
goals of this law are to ensure the prompt reporting to the in the handling of hazardous chemicals, human blood,
federal government of entities and individuals who possess blood-borne pathogens, or select agents and toxins.
select agents, to increase the security over such agents, and Hazardous chemicals, as defined by the OSHA standard on
to establish a comprehensive and detailed national database occupational exposure to hazardous chemicals in laborato-
of the location and characterization of such agents and the ries (48), are found in virtually every general or molecular
identities of individuals who have authorization to possess bacteriology laboratory, and therefore the standard will
them. apply to them all. Among the provisions of the standard is
The Secretary of the Department of Heath and Human the requirement to develop and follow a written Chemical
Services (HHS) has the authority and responsibility for reg- Hygiene Plan. The plan must describe the specific measures
ulating activities regarding select agents and toxins to pro- that are to be taken to protect employees from health haz-
tect the public health and safety. The Secretary of the ards associated with hazardous chemicals in the laboratory.
Department of Agriculture (USDA) has the authority and Laboratories that handle human blood and blood-borne
responsibility for regulating activities regarding select pathogens are required by the OSHA blood-borne
agents and toxins to protect animal and plant health and pathogens standard (49) to establish a similar document
animal and plant products. The list of regulated bacterial, called an Exposure Control Plan. The purpose of this plan
rickettsial, and fungal select agents and toxins is accessible is to describe the emergency controls, laboratory practices,
on the HHS web page www.cdc.gov/od/sap/docs/salist.pdf. personal protective equipment, and housekeeping practices
Several select agents come under the jurisdiction of both that will eliminate or minimize employee exposure to hu-
agencies. Interagency coordination assures that implement- man blood and blood-borne pathogens.
ing regulations regarding these overlap agents are not in A healthful and safe laboratory environment cannot be
conflict. Clinical and diagnostic laboratories are exempt sustained without every laboratory worker sharing in the re-
from most provisions of the HHS and USDA select agents sponsibility for safety. All persons in the laboratory must be
and toxins rules provided that all exemption criteria stated vigilant toward their own safety and the safety of their col-
in the rules apply. leagues and neighbors. They must know and follow safety
Implementing regulations were issued in December 2002 guidelines, regulations, and procedures that apply to their
(39, 40, 45). Laboratory directors and principal investiga- work. They should actively seek information and advice to
tors have a moral and legal responsibility to obtain author- improve their safety performance. They should always re-
ization before taking possession of a select agent and to port accidents, injuries, and unsafe conditions to their su-
ensure that they meet all of the requirements of the im- pervisor or safety officer. It is imperative that all laboratory
plementing regulations for the possession and use of select employees know how to respond to emergencies. Directors
agents, such as the provisions addressing safety plans, secu- can promote good work habits among all members of the
rity plans, emergency response plans, training, record keep- laboratory by encouraging them to participate in the selec-
ing, inspections, and notifications. The HHS rule encourages tion of safe practices and corresponding proficiency stan-
entities to consider the consensus guidelines in Biosafety in dards that all should meet to ensure safe working conditions
Microbiological and Biomedical Laboratories in preparing the for everyone (2). Ideally, the methods and attitudes that en-
required select agent safety plan (44). hance laboratory safety will become a conscious part of all
work activity in the laboratory.

46.2. PRINCIPLES 46.2.2. Biosafety Principles


Awareness of the potential risks, infectious and otherwise,
46.2.1. Directors Responsibilities associated with the procedure or activity being conducted is
Laboratory directors have the primary responsibility for set- the safety cornerstone in clinical and research laboratories.
ting the expectations and means for protecting the health To be aware of these risks, the individual worker must be
and safety of all laboratory workers under their supervision. knowledgeable about the biological agents and materials,
This involves ensuring that (i) hazards associated with lab- reagents, chemicals, equipment, and facilities being used.
oratory activities are recognized, (ii) routine and special Knowledge implies appropriate training in risk recognition
practices that are helpful in controlling or eliminating those and prevention, in the proper execution of procedures, and
hazards are selected for use in the laboratory, and (iii) labo- in the use of equipment.
ratory workers are provided with adequate instruction and The concept of universal precautions that is used in the
training so that they understand the relative significance of clinical setting is a prudent standard (41). It is based on the
the hazards that relate to them personally and acquire the realization that all clinical specimens, body fluids, tissues,
proficiency necessary to protect themselves and their col- and cultures are potentially infectious and should be han-
leagues from these hazards. The laboratory director is also dled with common sense practices and barrier precautions.
responsible for deciding who should have access to the lab- Standard and special microbiological practices, contain-
oratory and for establishing additional requirements aimed ment equipment, and facilities recommended in the CDC
at reducing potential exposure of persons who may be at in- and NIH consensus guidelines (44) for biosafety level 2 (see
creased risk of infection. The preparation of a safety manual section 46.5) constitute universal precaution for the labora-
46. Laboratory Safety 1001

tory and should be adopted as a minimal standard for work tions listing 222 viral infections, 21 of which were fatal
with any potentially infectious materials. (37). A second survey, involving 5,000 laboratories and
Containment of infectious agents within laboratory and published in 1951, summarized 1,342 cases, only one-third
research animal work areas is provided by appropriate com- of which had previously been reported (38). Brucellosis was
binations of laboratory practices, safety equipment, and fa- the most common occupational infection; together, brucel-
cility design features. Primary containment protects the losis, tuberculosis, tularemia, typhoid, and streptococcal in-
worker and the immediate work area from infection or con- fections accounted for 72% of all bacterial infections and
tamination. It is provided by consistent use of accepted 319 of the total number of cases caused by all categories of
good laboratory practices and appropriate use of safety infectious agents. The overall case fatality rate was 3%.
equipment, such as the use of biological safety cabinets The survey data were updated in 1965 and again in
(43), for activities involving known or potentially infec- 1976, summarizing a cumulative total of 3,921 reported oc-
tious materials. The use of safe and efficacious vaccines may cupational infections worldwide (27, 28). Brucellosis was
provide additional reductions in infection risks of workers. again the most commonly reported infection; brucellosis,
Secondary containment, i.e., the protection of the envi- typhoid fever, tularemia, tuberculosis, hepatitis, and
ronment external to the laboratory, is provided by a combi- Venezuelan equine encephalitis were the most frequently
nation of facility design features (e.g., directional airflow, recorded occupational infections at that time.
sealed penetrations, double doors), management of wastes Of 703 cases in which the source of infection was iden-
and other infectious materials, and access control. tified, 80% were due to contact with infectious materials
from spills and splashes, accidents involving the use of nee-
46.2.3. Facility Features dles and syringes, mouth pipetting, and injury from con-
Safety features of laboratory facilities provide architectural taminated broken glass or other sharp objects. Bites and
and mechanical barriers that serve to segregate activities scratches by experimentally or naturally infected animals or
performed within a laboratory module from nonlaboratory ectoparasites was the fifth major cause of known accidents
areas such as administrative offices and cafeterias. The ven- resulting in laboratory-acquired infection. The most fre-
tilation system provides for correct operations of safety cab- quently reported cases were 18 cases of ratbite fever (Screp-
inets and, in containment laboratories, can prevent the mi- tobacillus modifomis), 13 cases of leptospirosis (Leptospira
gration of airborne contaminants between occupied spaces. interrogans), and 9 cases of B virus (herpesvirus simiae) in-
The design of entry areas to laboratories can be helpful in fections. Awareness of occupational risks, good practices,
maintaining proper access control. It is also important that and modest barrier precautions might have effectively pre-
an autoclave for decontaminating infectious laboratory vented these occupational infections.
wastes be available within the facility. From these cited reports, as well as others, a consistent
The basic safety features of individual laboratory mod- pattern and trend characterizing occupational infections in
ules within a biosafety level 2 facility are floor and wall sur- laboratory workers have emerged. Pertinent issues include
faces that are easily cleaned, a hand-washing sink, and the following.
bench tops that are impervious to water and resistant to
1. Occupational infections are underreported. The
acids, alkalis, organic solvents, and moderate heat. Spaces
Sulkin and Pike studies (37,38) indicated that, historically,
between and beneath casework and equipment should be
only one-third of the occupational infections that were
accessible for cleaning. A safety shower and eye wash foun-
brought to their attention were reported in the scientific lit-
tain should also be easily accessible to all laboratory occu-
erature. Furthermore, no national focus exists for the re-
pants.
porting or active surveillance of occupational infections.
Additional safety features are required for biosafety level
2. Fewer than 20% of occupational infections are associ-
3 facilities. The laboratory is physically separated from non-
ated with an identified exposure, accident, or incident. The
laboratory areas by entry corridors that require passage
most plausible explanation is that diseases such as brucel-
through two sets of doors. Floor, wall, and ceiling surfaces are
losis, tuberculosis, and tularemia, and those caused by cer-
water resistant. Penetrations through these surfaces should
tain arboviruses occur as a result of exposure to respirable
be sealed to allow space decontamination. Foot-operated,
airborne infectious particles (droplet nuclei). Diseases such
elbow-operated, or automatically operated hand-washing
as typhoid, cholera, and viral hepatitis are most likely to be
sinks are located in each laboratory near the exit door. The
associated with poor laboratory practices, poor personal hy-
ventilation system creates directional airflow by drawing air
giene, and failure to use simple barrier precautions.
into the laboratory through the entry area. Exhaust air is dis-
3. The majority of cases in which the source of infection
charged directly to the outside and dispersed away from oc-
is identified could have been prevented by regular use of
cupied areas and air intakes of buildings.
good laboratory safety practices.
Harding and Byers (12) analyzed data from more than
46.3. HISTORICAL PERSPECTIVE 200 reports on laboratory-associated infections found in the
Occupational infections of laboratory workers have been literature worldwide between 1979 and 1999. Their work
documented for a century. Although infections with spe- finds that the pattern and trend of laboratory-associated in-
cific agents have varied by time and place, occupational in- fections observed in earlier studies in general have not
fections have typically involved agents prevalent in the changed. The top 10 laboratory-acquired diseases in both
community and, consequently, likely to be present in mate- periods included brucellosis, Q fever, and tuberculosis, all
rials received and handled in laboratories involved in diag- of which are diseases that can result from inhalation ex-
nostic, production, reference, and research activities. posures. Although hepatitis remains on this list a decline
In 1941, the first survey of occupational infections in in the number of infections has been noted. There were no
laboratory workers in the United States was published by reports of laboratory-associated infections with HBV in
Meyer and Eddy (18). In 1949, Sulkin and Pike published the United States after 1987, suggesting that the OSHA
the first in a series of surveys of laboratory-associated infec- blood-borne pathogens standard has had a positive impact
1002 APPENDICES

on worker protection. The authors noted that the distri- mass destruction (e.g., B. anthracis), are currently diseases of
bution of laboratory-associated infections among diagnos- low incidence and sporadic distribution. The standard and
tic and research laboratories has changed. In the earlier special practices and containment equipment described for
studies, diagnostic and research laboratories accounted for biosafety level 3 are recommended for the handling of path-
17% and 59%, respectively, of all reported laboratory- ogenic organisms presenting an aerosol risk of infection
associated infections, whereas the distribution observed (44); these are discussed further in section 46.6.
between 1979 and 1999 for diagnostic and research labo- Tuberculosis, historically important as an occupational
ratories was 45% and 51%, respectively. These trends re- infection, continues to represent an especially significant
inforce the importance of the safe practices, good habits, infection hazard to laboratory workers, who may encounter
training, and proficiency testing in contemporary bacteri- this agent in sputum samples, biopsy materials, and cul-
ology, microbiology, and biomedical laboratories for pre- tures. Mycobacterium bovis BCG vaccine, which is licensed
venting laboratory-associated infections. and readily available, is not customarily used in the
United States, so susceptibility to tuberculosis infection is
essentially universal in this country. The adult infectious
46.4. BIOSAFETY LEVELS dose is estimated to be 10 or fewer organisms (51). Al-
Although the number of laboratories in the United States is though the incidence of occupationally acquired tubercu-
unknown, it is reasonable to assume that clinical laborato- losis is presently unknown, reports over several years attest
ries (including physicians office laboratories) constitute the to the continuing risk of infection with this classical and
most common type of laboratory operation. Clinical labora- still important pathogen (G. P. Kubica, personal commu-
tory activities typically involve the handling, manipulation, nication). A survey in 1976 indicated that the incidence
and, sometimes, collection of blood, urine, feces, and other of tuberculosis among medical laboratory workers in
body fluids, excretions, or secretions. These clinical materi- England was five times greater than that for the general
als are all potentially infectious and may contain a variety community (11). Tuberculosis is an appropriate represen-
of bacterial, fungal, viral, and protozoal agents. The concept tative of occupational infections that may be transmitted
of universal blood and body fluid precautions (universal via infectious aerosols.
precaution) should apply to all clinical specimens since it is Biosafety level 1 is not appropriate for work with patho-
not realistically possible to rule out all infectious agents on genic bacteria. This level was described in guidelines devel-
the basis of medical history and signs and symptoms at the oped by CDC and NIH (44) to indicate basic practices ap-
time of medical examination or clinical diagnosis (41). It is propriate for undergraduate and secondary educational
also difficult to determine, with reasonable confidence, training and teaching laboratories for which introductory
whether certain body fluids typically not associated with microbiological protocols would involve only defined and
the transmission of blood-borne diseases may contain visi- characterized strains of viable microorganisms not known to
ble blood. Consequently, it is prudent to handle all body cause disease in healthy adult humans. Emphasis is placed
fluids, tissues, and cultures as potentially infectious. The es- on the use of mechanical pipetting aids; hand-washing; not
timated prevalence of HBV infection in the United States eating, drinking, or smoking in the work area; and daily de-
(approximately 300,000 new cases annually) would suggest contamination of work surfaces. This minimal level of con-
that this viral disease is the occupational infection hazard tainment has been adopted by molecular biology labo-
most likely to be present in clinical materials submitted for ratories involved in research activities for which the
laboratory examination. host-vector systems in use have been exempted from the
HBV shares a common transmission pattern in the labo- NIH Guidelines for handling recombinant DNA molecules
ratory occupational setting with most bacterial pathogens (42).
that are regularly present in a community. Such agents may Most human pathogens encountered in diagnostic and
be transmitted parenterally, by ingestion, via broken skin, clinical laboratories and used in biomedical and microbio-
and following exposure of the mucous membranes of the logical research are safely handled using practices described
mouth, eyes, or anterior nose. Characteristically, these path- for biosafety levels 2 and 3. These practices are the subject
ogens are not transmitted in the laboratory via infectious of this chapter. The safe conduct of research on exotic
aerosols. lethal pathogens like the hemorrhagic viruses requires bio-
Consequently, it is reasonable to use laboratory practices safety level 4 practices and sophisticated biocontainment
and barrier precautions that prevent exposure of laboratory facilities. A discussion of biosafety level 4 practices, con-
workers from the infectious agent most likely to be present tainment equipment, and facilities is beyond the scope of
in specimens handled on a regular basis. It is especially im- this chapter. However, investigators and technicians who
portant that clinical chemists, geneticists, and other scien- will join the nations scientific initiative to conduct re-
tists without specific training or experience in working with search to protect the public health from the adverse conse-
potentially infectious materials are made aware of such haz- quences of bioterrorism will have to gain a thorough under-
ards and informed about appropriate preventive measures. standing of this area of biological safety, acquire proficiency
These practices and precautions are succinctly summarized in the use of safe technique, and develop the habits neces-
in the description of standard and special microbiological sary to sustain a high level of performance.
practices recommended for biosafety level 2 (44) and are
discussed in detail in section 46.5.
A higher level of occupational hazard occurs with the 46.5. LEVEL 2 BIOSAFETY PRACTICES
handling of primary pathogens that can cause disease The consistent use of good practices for the manipulation of
through the inhalation of aerosols as well as routes of direct potentially infectious materials is the single most important
exposure. Diseases such as brucellosis, tularemia, tuberculo- element in preventing occupational infections. It is prudent
sis, anthrax, Q fever, and diseases caused by certain indige- to consider that all bacteria are potential hazards to human
nous arboviruses are transmitted by aerosols. All of these health. Consequently, biosafety level 2 practices should be
diseases, except for tuberculosis and diseases caused by the adopted even in general and molecular bacteriology labora-
deliberate release or use of a select agent as a weapon of tories.
46. Laboratory Safety 1003

Biosafety level 2 practices are recommended for most infectious materials are in use and where special provisions
laboratory activities involving known pathogens and for ex- for entry are required, such as an immunization requirement
periments involving either the introduction of recombinant to handle hepatitis B virus. The OSHA blood-borne
DNA into pathogenic bacteria or the introduction of DNA pathogens standard requires the sign to be orange or orange-
from pathogenic bacteria into nonpathogenic procaryotes red with symbol and lettering in a contrasting color (49).
or lower eucaryotes. Brucella spp., Francisella tularensis, M. Also display the biohazard symbol on freezers, refrigerators,
bovis, and Mycobacterium tuberculosis require level 3 contain- and other units used to store infectious materials.
ment because of the serious hazard of aerosol infection. 3. Decontaminate work surfaces daily and immediately
However, DNA from these bacteria can be cloned in after spills of viable material.
nonpathogenic procaryotic or lower-eucaryotic host-vector 4. Decontaminate infectious liquid or solid wastes be-
systems under level 2 containment (42). The deliberate for- fore disposal. If wastes are decontaminated at a site away
mation of recombinant DNAs containing genes for the from the laboratory, place them in a durable leak-proof con-
biosynthesis of toxic molecules lethal for vertebrates, and tainer and close it before removal from the laboratory.
the deliberate transfer of drug resistance genes to microor- 5. Always use a mechanical pipetting device when
ganisms that are not known to acquire the trait naturally, pipetting.
may require RAC review or NIH approval before experi- 6. Do not permit eating, drinking, smoking, storage or
ments are initiated. preparation of food for human consumption, or cosmetics
Use the following biosafety level 2 practices routinely application in the laboratory.
when handling even potentially pathogenic bacteria. The 7. Wear protective gloves when contact with infectious
bacteria capable of causing serious disease when inhaled in material may be unavoidable. Remove gloves carefully when
infectious aerosols require level 3 containment. they become soiled with contaminated material, and wash
1. Keep laboratory doors closed when work is in hands immediately. Put on a new pair of gloves before con-
progress. tinuing the procedure. Do not wash or reuse soiled gloves.
2. Post a hazard warning sign incorporating the univer- 8. Wash hands after handling infectious materials and
sal biohazard symbol (Fig. 1) on the laboratory door when always before leaving the laboratory.

/BIOHAZARDj
ADMITTANCE TO AUTHORIZED PERSONNEL ONLY
Hazard identity:
Responsible Investigator:
In case of emergency call:
Daytime phone Home phone
Authorization for ontrance must be obtained from
tha Responsible Inverti((ator named above.

FIGURE 1 Biohazard warning sign for biomedical facilities.


1004 APPENDICES

9. Avoid the use of syringes, needles, and other sharp inoculation. Keep personal items such as coats, hats, storm
instruments whenever possible. When they are required, rubbers or overshoes, umbrellas, and purses out of the labo-
handle them with caution to avoid accidental punctures or ratory. Laboratory design that allows personal items to re-
cuts. Use only needle-locking syringes or disposable syringe- main outside the work area plays an important role in over-
needle units for injection or aspiration of infectious fluids. all safety.
10. Discard used needles and disposable cutting instru-
ments into a puncture-resistant container that has a lid. -
46.5.2. Pipetting
11. Wear laboratory coats, gowns, smocks, or uniforms Mouth pipetting and suctioning have been identified as
while in the laboratory. practices that are significant and consistent causes of occu-
12. Exercise care in all procedures and manipulations to pational infections (20, 21). Pipetting aids prevent acci-
minimize aerosol formation. Use biological safety cabinets dental aspiration of cultures or other infectious fluids into
or physical containment devices such as centrifuge safety the mouth and eliminate the hazard of transferring infec-
cups to contain operations that will generate aerosols capa- tious agents from contaminated surfaces to the pipette
ble of causing significant contamination or hazardous expo- mouthpiece by bare or gloved hands. Other hazards are as-
sure. sociated with pipetting infectious materials. Forcibly ex-
13. Wear face protection, such as goggles, mask, or face pelling residual contents from pipettes, bubbling air
shield, when performing outside a biological safety cabinet through liquid cultures, and falling drops of liquid culture
a procedure that may splash or spray infectious materials on from pipette tips can create infectious aerosols in the
the face. breathing zone of the worker. These hazards are eliminated
14. A biological safety manual that is appropriate for the by good technique. Use pipettes that do not require expul-
work of the laboratory should be available in the laboratory sion of the last drop. Take care to avoid dropping cultures
for staff to use for reference as necessary. from the pipette. When pipetting materials containing
pathogenic bacteria, place a disinfectant-soaked towel on
Several routine operations in the laboratory can easily
the work surface. Discharge liquids from pipettes as close as
become the source of laboratory-acquired infections. They
possible to the fluid or agar level of the receiving vessel, or
require vigilance to guard against compromise and error.
allow the contents to run down the wall of the receiving
The following sections emphasize the hazards associated
vessel. Avoid dropping the contents from a height into ves-
with these operations and underscore the precautions nec-
sels. Place contaminated pipettes into a container with
essary to conduct them safely.
enough suitable disinfectant for complete immersion.
A variety of pipetting aids are available, ranging from
46.5.1. Personal Hygiene simple bulb- and piston-actuated devices to sophisticated
Appropriate personal-hygiene practices for laboratory work- devices that contain their own vacuum pumps (19). In se-
ers have the simple objective of reducing personal risks of self- lecting a pipetting aid, consider the procedure, the ease of
infection by avoiding ingestion, skin, or mucous-membrane operation, and the accuracy needed. Use several pipetting
exposure to infectious materials. Wash hands with soap and aids; none is available that is appropriate for all procedures.
water following the completion of bench activities, the re-
moval of protective gloves, and the overt contamination of 46.5.3. Hypodermic Syringes and Needles
skin and when leaving the laboratory. Hand washing is Accidents involving the use of syringes and needles are
among the most important actions that workers can take in among the most common cause of occupational infections
reducing the risk of infection by agents commonly handled in laboratories and health care facilities. They account for
in biomedical laboratories. A 10-second hand wash with an estimated one-fourth of laboratory-acquired infections
soap and water reduces the number of contaminating tran- that are caused by accidents (29). Needle sticks (punc-
sient microorganisms to levels below the infective thres- tures) account for an estimated one-third of work-related
hold. Use powdered soap from a dispenser rather than bar or accidents in health care workers (14). In some health care
liquid soap, which may harbor bacteria (see section 46.8.7). facilities, annual injury rates from being accidentally stuck
Follow hand washing by drying with disposable towels. by a needle may exceed 100 per 1,000 employees, and these
Hand washing may also have the added benefit of reducing injuries may be underreported by 40 to 70% (1, 14).
the level of cross-contamination between specimens or ac- Accidental parenteral inoculation of infectious fluids dur-
tivities. ing phlebotomy procedures is the most obvious and com-
Wash hands promptly after removing protective gloves. mon means of occupational infection associated with nee-
Unrecognized small holes, abrasions, or tears, an opening at dles. Infections have also been associated with sprays or
the wrist, or solvents can enable bacteria to pass the glove aerosols of infectious materials when needles separate from
barrier and contaminate the skin. A disinfectant wash or syringes and with direct or indirect skin contact with infec-
dip may be desirable, but this procedure can cause roughen- tious fluids from leaking syringes or needles.
ing, desiccation, or sensitization of the skin. Of seven categories of phlebotomy devices used in a uni-
Work with bacteria should not be performed by anyone versity hospital, disposable syringes are the devices most
with a new or old cut, an abrasion, a lesion of the skin, or commonly associated with occupational needle sticks (14).
any open wound, including that resulting from a tooth ex- However, when the data comparing the number of injuries
traction. with the number of devices purchased and presumably used
Food, candy, gum, and beverages for human consump- were reviewed, disposable syringes had the lowest rate of
tion should not be stored or consumed in the laboratory. associated injuries among all categories of devices used.
Smoking should not be permitted. A beard is undesirable Injuries associated with the use of disposable syringes most
because it retains particulate contamination and because a commonly occurred during the recapping of used needles.
clean-shaven face is essential to the adequate fit of a face In contrast to health care activities, when needles and
mask or respirator. Develop the habit of keeping hands away syringes or other phlebotomy devices are commonly used to
from the mouth, nose, eyes, face, and hair to prevent self- collect blood and other fluids from patients, there are rela-
Next Page

46. Laboratory Safety w 1005

tively few indications for the use of such devices in most or- droplets, aerosols, or contamination of the skin or the im-
dinary laboratory activities. Restrict the use of needles and mediate work area.
syringes to the collection of blood or fluids from patients or Because the level of energy applied in such activities is
experimental animals, the injection of materials into pa- relatively low, the efficiency for creating aerosols is also rel-
tients or experimental animals, and the aspiration of re- atively low and the droplet is typically large. The potential
agents or other materials from containers with diaphragm for creating respirable droplets (in the 1- to 5-pm range) is
stoppers. Conduct all other manipulations of potentially in- relatively low. However, the potential for creating larger
fectious or hazardous fluids with mechanical pipettes or de- droplets, which may contaminate hands and immediate
vices other than needles and syringes. work surfaces, is relatively high.
In response to the continuing problem of needle sticks, The most common opening activity in most health care
manufacturers of phlebotomy devices have developed a va- laboratories is the removal of stoppers from containers of
riety of safety syringes (e.g., a retractable needle). The use clinical materials (e.g., blood, urine, spinal fluid, feces) sub-
of these devices may provide additional safeguards during mitted for examination. Seventeen percent of blood sam-
phlebotomy procedures. The wearing of gloves may also fur- ples received in a clinical laboratory have blood on the con-
ther reduce the risk of occupational exposures of skin to tainer label, 6% of stool specimens have feces on the
body fluids during phlebotomy and other procedures in- outside of the container, and 4 to 5% of laboratory speci-
volving the collection or handling of potentially infectious men forms are visibly blood-stained (5). Specimens should
body fluids. be received and opened only by personnel who are knowl-
The following practices are recommended for hypoder- edgeable about occupational infection risks and of the con-
mic needles and syringes when used for parenteral injec- cept of Universal Precautions. Open clinical specimen con-
tions. tainers in well-lighted designated areas with impervious and
easily cleanable work surfaces. Wear a laboratory coat and
1. Avoid quick and unnecessary movements of the suitable gloves. The use of plastic-backed absorbent paper
hand holding the syringe. towels on work surfaces facilitates cleanup and minimizes
2. Examine syringes for chips and cracks, and examine the creation of aerosols in the event of overt or unobserved
needles for barbs and plugs. Do this before sterilization and spills of infectious materials. Some laboratories have found
before use. it to be advantageous to open all incoming specimens in the
3. Use needle-locking (Luer-Lok type) syringes only, work chamber of a class I or class I1 biological safety cabi-
and be sure that the needle is locked securely. net, which is especially useful if incoming specimens are
4. Wear surgical or other rubber gloves. broken or leaking.
5. Fill syringes carefully to minimize air bubbles and Appropriate techniques for opening sealed ampoules, dry
frothing. powders, sealed containers under relatively negative pres-
6. Expel excess air, liquid, and bubbles vertically into a sure, embryonated eggs, and the Like are described in detail
cotton piedget moistened with a suitable disinfectant or elsewhere (20). The use of appropriate personal protection
into a small bottle of sterile cotton. (coats, gloves), primary containment (biological safety cab-
7. Do not use a syringe to forcefully expel infectious inet), and physical barriers (e.g., acrylic beta shields) in
fluid into an open vial or tube for the purpose of mixing. general addresses the ubiquitous problems of exposure of
Mixing with a syringe is appropriate only if the tip of the personnel and contamination of work surfaces during the
needle is held below the surface of the fluid in the tube. opening of containers.
8. If syringes are filled from test tubes, take care not to Tubes containing bacterial suspensions should be manip-
contaminate the hub of the needle, since this may result in ulated with care. Simple procedures such as removing a tube
transfer of infectious material to the fingers. cap or transferring an inoculum can create aerosols. Clearly
9. When removing a syringe and needle from a rubber- mark tubes and racks of tubes containing biohazardous ma-
stoppered bottle, wrap the needle and stopper in a cotton terial. Use safety test tube trays in place of conventional test
pledget moistened with a suitable disinfectant. If there is tube racks to minimize spillage from broken tubes. A safety
danger that the disinfectant may contaminate sensitive ex- test tube tray is one that has a solid bottom and sides deep
perimental materials, a sterile dry pledget may be used and enough to hold the liquid should the test tube break.
discarded immediately into a disinfectant solution. Vigorous shaking of liquid cultures creates a heavy
10. Inoculate animals with the hand positioned behind aerosol. In general, swirling action will create a homoge-
the needle to avoid punctures. neous suspension with a minimum of aerosol. When resus-
11. Be sure that the animal is properly restrained prior to pending a liquid culture, allow a few minutes to elapse be-
inoculation, and be alert for any unexpected movements. fore opening the container, to reduce the aerosol.
Consider anesthetizing the animal during injections if ap- The insertion of a sterile hot loop or needle into a liquid
proved by the Institutional Animal Care and Use Com- or slant culture can cause spattering and release of aerosol.
mittee (IACUC). To minimize aerosols, allow the loop to cool in the air or be
12. Before and especially after injecting an animal, swab cooled by touching to the inside of the container or to the
the injection site with a disinfectant. agar surface where no growth is evident prior to contact with
13. Do not bend, shear, recap, or remove the needle from the culture or colony. After use of the inoculating loop or
the syringe by hand. Place used needle-syringe units directly needle, sterilize it in an electric or gas incinerator specifically
into a puncture-resistant container, and decontaminate be- designed for this purpose rather than heating it in an open
fore disassembly, reuse, or disposal. flame. These small incinerators have a shield to contain any
material that may spatter from the loop. Disposable inocu-
46.5.4. Opening Containers lating loops are available commercially; rather than decont-
The opening of vials, flasks, petri dishes, culture tubes, em- aminating them with heat, discard them into a disinfectant.
bryonated eggs, and other containers of potentially infec- Streaking an inoculum on rough agar results in aerosol
tious materials poses potential but subtle risks of creating production, created by the vibrating loop or needle. This

Potrebbero piacerti anche