Sei sulla pagina 1di 100

Additional Resources for Life Scientists

Ultrafiltration Application and Product Guide


Cell Biology at Millipore. Filter with Millipore for
Visit Millipores new Fast, High-Quality Results.
home for Cell Biology Millipore offers
research. You can hundreds of mem
easily access educa brane-based devices
tional content, labora for sterile filtration, antibodies forensics
tory protocols, literature chromatography,
and troubleshooting tips sample preparation,
along with many other and almost any other
scientific resources. Our application in the life proteins
intuitive cell biology site
lets you navigate, review
sciences laboratory.
To request a copy of the
serum
and purchase directly online thousands of Millipore Analytical Sample Preparation
products across many cell biology workflows. and General Filtration catalogue, visit
Visit www.millipore.com/cellbiology. www.millipore.com/source4filters.
enzymes viruses
Discover Our New Range of Laboratory Water Systems
Immunodetection Tools. Millipore provides total solutions from bench-top
Easily access
educational content,
systems to custom-engineered purification chains
for laboratory buildings. Youll find Millipore water
nucleic acids
laboratory protocols, systems installed in over 70,000 laboratories world-
literature and trouble wide supplying pure water for electrophoresis, PCR,
shooting tips along chromatography and other life science applications.
with many other To learn more, visit www.millipore.com
scientific resources.
You can also browse
and purchase online
Technical Support
thousands of immuno
Millipore Technical Service Specialists support
detection products, including more than
ultrafiltration and many other life science
10,000 antibodies formerly sold by Chemicon

Ultrafiltration
applications, including blotting, DNA
and Upstate, which are now part of Millipore.
sequencing sample preparation, sterile
Visit www.millipore.com/immunodetection.
filtration, and MS sample prep. To contact a
Specialist, call your local office or submit a
question at www.millipore.com/techservice.
To access our library of frequently asked
questions, go to www.millipore.com/faqs.
Application and Product Guide

Lit. No. TP0040EN00 Rev. B 10/07 Printed in U.S.A. 07-493


www.millipore.com
2007 Millipore Corporation, Billerica, MA 01821 U.S.A. All rights reserved.
For over 50 years, Millipore has helped As part of our customer-focused approach to
improve laboratory productivity and efficiency support and service, Millipore sales and in-field
for researchers worldwide by delivering technical teams receive ongoing training in our
innovative products |and services backed by customers research areas. This training allows
the highest levels of quality and expertise. them to provide technical assistance, validation
services, equipment installation, and maintenance
With the recent acquisitions of Upstate,
programs for your specific applications.
Chemicon and Linco, we now offer a wide
range of antibodies; stem cell-related products; We are also developing exciting new web
protein immunodetection kits; and drug profiling tools to facilitate product selection and on-line
products and services. ordering. To experience all that Millipore has
to offer, visit www.millipore.com.
The addition of these core products and
capabilities to our existing sample preparation
and laboratory water offering creates an
extensive portfolio of comprehensive solutions
for immunodetection, cell biology, and
drug discovery.
Table of Contents
Overview of Membrane Filtration Removal of Unincorporated Label
from Labeled Protein . . . . . . . . . . . . . . . . . . 50
Membrane Processes . . . . . . . . . . . . . . . . . 4 Rapid Purification of Monoclonal Antibodies . . 52
Recovery . . . . . . . . . . . . . . . . . . . . . . . . . . 6 A Simple Strategy for Protein Fractionation . . . . 54
Mode of Operation . . . . . . . . . . . . . . . . . . 9 Urine Concentration . . . . . . . . . . . . . . . . . . 56
Diafiltration . . . . . . . . . . . . . . . . . . . . . . . . 10 Use of Centrifugal Filter Devices as an
Fractionation . . . . . . . . . . . . . . . . . . . . . . . 11 Alternative to Stirred Cells . . . . . . . . . . . . . . . 58

Protocols for Nucleic Acids


Membranes and Devices for Ultrafiltration
Purification of DNA Sequencing Reactions . . . 60
Membrane Selection . . . . . . . . . . . . . . . . . . 14
Concentrating and Desalting DNA or RNA . . . 63
Device Selection . . . . . . . . . . . . . . . . . . . . 16
Preparing Samples for Forensics
Microcon Centrifugal Filters . . . . . . . . . . . . . 17 Identification Analysis . . . . . . . . . . . . . . . . . 66
Amicon Ultra-4 Centrifugal Filters . . . . . . . . . 18 Concentration of Genomic DNA for
Amicon Ultra-15 Centrifugal Filters . . . . . . . . . 20 Forensic Analysis . . . . . . . . . . . . . . . . . . . . 67

Centriprep Centrifugal Filters . . . . . . . . . . . . 22 Purification of PCR Products . . . . . . . . . . . . . . 68

Centricon Plus-70 Centrifugal Filters . . . . . . . . 23 Quantitative Recoveries of Nanogram


Amounts of Nucleic Acids . . . . . . . . . . . . . . 70
MultiScreen Filter Plate . . . . . . . . . . . . . . . . 24
RNA Purification and Preparation of
Ultrafree Centrifugal Filters . . . . . . . . . . . . . . 25 Fluorescent cDNA Probe from Human mRNA . . 72
Pellicon XL Cassettes and Purification of In Vitro Synthesized mRNA . . . . . 76
Labscale TFF System . . . . . . . . . . . . . . . . . 26
Effect of Centrifugal Ultrafiltration on
Pellicon 2 Mini Cassettes . . . . . . . . . . . . . . . 28 Large Fragment DNA Integrity . . . . . . . . . . . . 78
Prep/Scale Spiral Wound Filter Cartridges . . . 29 DNA Extraction from Agarose Gels . . . . . . . . 80
Stirred Cells . . . . . . . . . . . . . . . . . . . . . . . . 30 PCR Purification . . . . . . . . . . . . . . . . . . . . . 82

Ultrafiltration Discs . . . . . . . . . . . . . . . . . . . . 31 Enzyme Removal . . . . . . . . . . . . . . . . . . . . 84

Protocols for Virus concentration


Protocols for Proteins
Concentration of Bacteriophage Using
Ta bl e o f C o n t e n t s

Concentration, Desalting and Buffer Exchange . 34


Ultrafiltration . . . . . . . . . . . . . . . . . . . . . . . 88
Detergent Removal . . . . . . . . . . . . . . . . . . . 38
Concentration of Animal Viruses Using
Two-Dimensional Electrophoresis Ultrafiltration . . . . . . . . . . . . . . . . . . . . . . . 91
Sample Preparation . . . . . . . . . . . . . . . . . . . 40

Purification of Serum Peptides for Biomarker . . . 42 Appendix

Rapid Antibody Concentration . . . . . . . . . . . . 45 High Throughput Applications . . . . . . . . . . . . 94

Affinity Purification . . . . . . . . . . . . . . . . . . . . 46 Glossary . . . . . . . . . . . . . . . . . . . . . . . . . . 95

1
2
Ta bl e o f C o n t e n t s
Overview of Membrane Processes . . . . . . . . . . . . . . . . . . . 4

Recovery . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6

Membrane Filtration Mode of Operation . . . . . . . . . . . . . . . . . . . . 9

Diafiltration . . . . . . . . . . . . . . . . . . . . . . . . . . 10

Fractionation . . . . . . . . . . . . . . . . . . . . . . . . . 11

O v e r v ie w o f M e m br a ne F ilt r at i o n

3
Membrane Processes
Ultrafiltration Ultrafiltration is typically used to separate proteins
from buffer components for buffer exchange,
Ultrafiltration (UF) is the process of separating
desalting, or concentration. Ultrafilters are also ideal
extremely small particles and dissolved molecules
for removal or exchange of sugars, non-aqueous
from fluids. The primary basis for separation is
solvents, the separation of free from protein-bound
molecular size, although in all filtration applications,
ligands, the removal of materials of low molecular
the permeability of a filter medium can be affected
weight, or the rapid change of ionic and/or pH
by the chemical, molecular or electrostatic proper-
environment (see Figure 5, page 10). Depending on
ties of the sample. Ultrafiltration can only separate
the protein to be retained, the most frequently used
molecules which differ by at least an order of
membranes have a nominal molecular weight limit
magnitude in size. Molecules of similar size can
(NMWL) of 3 kDa to 100 kDa.
not be separated by ultrafiltration (see Figure 1).
Ultrafiltration is far gentler to solutes than
Materials ranging in size from 1K to 1000K
processes such as precipitation. UF is more efficient
molecular weight (MW) are retained by certain
because it can simultaneously concentrate and
ultrafiltration membranes, while salts and water will
desalt solutes. It does not require a phase change,
pass through. Colloidal and particulate matter can
which often denatures labile species, and UF can
also be retained. Ultrafiltration membranes can be
be performed either at room temperature or in a
used both to purify material passing through the
cold room.
filter and also to collect material retained by the
filter. Materials significantly smaller than the pore
Microfiltration
size rating pass through the filter and can be
Microfiltration (MF) is the process of removing
depyrogenated, clarified and separated from high
particles or biological entities in the 0.025 m to
O v e r v ie w o f M e m br a ne F ilt r at i o n M e m b r a n e P ro ce s s e s

molecular weight contaminants. Materials larger


10.0 m range from fluids by passage through a
than the pore size rating are retained by the filter
microporous medium such as a membrane filter.
and can be concentrated or separated from low
Although micron-sized particles can be removed
molecular weight contaminants.

Figure 1. Comparison of ultrafiltration with other commonly used membrane separation techniques

Reverse Osmosis Ultrafiltration Microfiltration Clarification


0.0001 m 0.001 m 0.01 m 0.1 m 1 m 10 m 100 m

0.2 kDa 200 kDa 20,000 kDa

Red Blood Smallest


Sugars Proteins B. diminut a
Cell Visible Particle

Amino Acids
Carbon Black Yeast Pollens
Nucleotides

Oligo- Polio
Salts Mycoplasma E. coli Clouds Fog
nucleotides Virus

Human
Antibiotics Mammalian Virus Bacteria
Hair

4
by use of non-membrane or depth materials such as Reverse Osmosis
those found in fibrous media, only a membrane filter
Reverse osmosis (RO) separates salts and small
having a precisely defined pore size can ensure
molecules from low molecular weight solutes
quantitative retention. Membrane filters can be used
(typically less than 100 daltons) at relatively
for final filtration or prefiltration, whereas a depth
high pressures using membranes with NMWLs of
filter is generally used in clarifying applications
1 kDa or lower. RO membranes are normally rated
where quantitative retention is not required or as
by their retention of sodium chloride while ultrafiltra-
a prefilter to prolong the life of a downstream
tion membranes are characterized according to
membrane. Membrane and depth filters offer
the molecular weight of retained solutes. Millipore
certain advantages and limitations. They can
water purification systems employ both reverse
complement each other when used together in a
osmosis membranes as well as ultrafiltration
microfiltration process system or fabricated device.
membranes. Reverse osmosis systems are primarily
The retention boundary defined by a membrane
used to purify tap water to purities that exceed
filter can also be used as an analytical tool to
distilled water quality. Ultrafiltration systems ensure
validate the integrity and efficiency of a system.
that ultrapure water is free from endotoxins as well
For example, in addition to clarifying or sterilizing
as nucleases for critical biological research.
filtration, fluids containing bacteria can be filtered to
trap the microorganisms on the membrane surface
for subsequent culture and analysis. Microfiltration
can also be used in sample preparation to remove
intact cells and some cell debris from the lysate.
Membrane pore size cut-offs used for this type of
separation are typically in the range of 0.05 m
to 1.0 m.

O v e r v ie w o f M e m br a ne F ilt r at i o n M e m b r a n e P ro ce s s e s
Figure 2. Ultrafiltration membranes vs. traditional microporous membranes
Ultrafiltration membranes Microporous membranes are
generally have two distinct generally rigid, continuous
layers: a thin (0.11.5 m), meshes of polymeric material
dense skin with a pore diameter with defined pore sizes. They
of 10 400 and a more are used to retain bacteria,
porous substructure. Any species colloids and particulates.
capable of passing through the Species are either retained
pores of the skin (whose size is on the membrane surface or
precisely controlled in manufac- trapped in its substructure.
ture) can therefore freely pass
Cross-section of ultrafiltration Cross-section of traditional micro-
the membrane.
membrane with skin and porous porous membrane with uniform
substructure. pore structure from top to bottom.

5
Recovery
The ultimate aim of ultrafiltration is to maximize When using membrane ultrafiltration for sample
recovery of solutes of interest, but there are many concentration or desalting, care must be taken
membrane characteristics that affect that goal. to select a membrane (or device) with a NMWL
Factors affecting recovery include: appropriate for the application. Because there
Nominal molecular weight limit (NMWL)/ are several considerations in determining whether
nucleotide cut-off (NCO) a given solute will or will not be retained by a
membrane of a specific cut-off, it is best to choose
Retention
a device with cut-off at about one half of the
Concentration polarization molecular weight of the protein to be concentrated.
Flux This maximizes protein recovery and minimizes
filtration time.
Nominal Molecular Weight Limit
A microfiltration membranes pore size rating,
Nucleotide Cut-off (NCO)
typically given as a micron value, indicates that For most membranes, the NMWL is determined
particles larger than the rating will be retained. experimentally under a standard set of operating
Ultrafiltration membranes are rated according to conditions. These analyses typically employ purified
the nominal molecular weight limit (NMWL), also globular proteins to serve as markers or indicators
sometimes referred to as molecular weight cut-off of the retention characteristics of an ultrafiltration
(MWCO). The NMWL indicates that most dissolved membrane. Although this approach is useful for
macromolecules with molecular weights higher than choosing the appropriate NMWL for most protein
the NMWL will be retained. research applications, selection of a membrane
An ultrafiltration membrane with a stated NMWL with an appropriate NMWL membrane for nucleic
should retain (reject) at least 90% of a globular acid or polysaccharide purification is considerably
solute of that molecular weight in daltons. However, more complex. By virtue of the rod-like three-
for a wider safety margin, the selected cut-off dimensional structures of these molecules, these
O v e r v ie w o f M e m br a ne F ilt r at i o n Re cove r y

should be well below the molecular weight of the types of molecules require a tighter membrane
solute to be retained. When solutes are to be (with a smaller cut-off) than do globular proteins of
exchanged, the cut-off should be substantially the same molecular weight. It is therefore convenient
above that of the passing solute. A lower NMWL to consider the membrane retention characteristics
increases rejection but decreases the filtration rate of nucleic acids as being related to their length
for the same membrane material. (in nucleotides) rather than their molecular weight.
Retention and product recovery are a function Complicating matters even further are several
of a variety of other factors, including the molecular additional factors that affect the recovery of nucleic
shape and size of the molecule; electrical charac- acid fragments from a membrane of a given
teristics; sample concentration and composition; NMWL. These factors include: the strandedness
operating conditions; and device or system of the DNA or RNA molecule; whether the DNA is
configuration. Two membranes may have the linear, relaxed or supercoiled (for plasmid); the ionic
same NMWL but will exhibit different retention of strength of the solvent; the velocity of the process
molecules within a relatively narrow range of sizes. stream over the membrane; and the nature of the
In addition, slender, linear molecules (e.g., nucleic driving force. The overall effect is that optimal
acids) may find their way through pores that will nucleic acid recovery is achieved in low salt buffers
retain a globular species of the same weight. run under conditions of relatively low velocity
Retention can also be affected by hydration with (e.g., low vacuum pressure or low g-force).
counter-ions. Nevertheless, NMWL has proven The membrane and protocol developed for the
to be an effective general indicator of membrane Montage PCR centrifugal filter device takes into
performance for globular proteins. account these conditions in order to provide for high

6
recovery of small PCR products (e.g., ~150 base of the NMWL and the size of the molecule but
pairs [bp]) as quickly as possible. For purifications Millipores recommendation is designed to provide
that are driven by vacuum, significantly tighter maximum recovery. Please see additional information
membranes are typically required to obtain regarding membrane NMWL selection on page 14.
optimal recovery.
If the DNA sample is in the presence of high salt Concentration Polarization
(or the device is run at a higher-than-recommended Another factor affecting the retention characteristics
g-force), a significantly reduced DNA recovery is the potential for membrane fouling, or concentra-
may be observed. Under these conditions, higher tion polarization. This occurs when there is an
DNA or RNA recovery can be achieved by using a accumulation of the retained solute on the surface
tighter membrane. However, it will take significantly of the membrane. At high concentrations, a gel
longer to complete the purification. For applications layer forms that can act as a secondary membrane
such as PCR where removal of unincorporated (Figure 3). This may interfere with passage of the
single-stranded primers from double-stranded DNA molecules through the membrane and can adversely
fragments is required, the molecular weights of affect the flow rate. In addition, pH, buffer compo-
the primer and DNA fragment should differ by at nents, and concentration can result in a protein
least an order of magnitude for efficient separation. behaving in an anomalous manner in terms of its
Millipore offers devices that are specifically retention or passage by UF membranes.
designed for separating and concentrating During concentration polarization, the gel layer
genomic DNA and PCR products by ultrafiltration. on the membrane surface superimposes its own
rejection characteristics on those of the membrane.
Retention Usually, concentration polarization increases
Retention, also sometimes called rejection, is a retention of lower-molecular weight species.
function of molecular size and shape. Nominal A membrane with a 100K NMWL may reject
cut-off levels, defined with model solutes, are 1020% of albumin in a 0.1% solution of pure
convenient indicators. Degree of hydration, counter albumin. However, in the presence of larger solutes
ions, and steric effects can cause molecules with such as IgG, it may reject 90% of the albumin.
similar molecular weights to exhibit very different Concentration polarization makes it very difficult
retention behavior. Many biological macromolecules to use UF for solute fractionation unless the solutes

O v e r v ie w o f M e m br a ne F ilt r at i o n Re cove r y
tend to aggregate, or change conformation under to be separated differ in size by at least an order
varying conditions of pH and ionic strength, so that of magnitude.
effective size may be much larger than the native
molecule, causing increased rejection. Solute/
solvent and solute/solute interactions in the sample
Figure 3. High concentrations
can also change effective molecular size. For
example, some proteins will polymerize under
certain concentration and buffer conditions while
others (e.g., heme proteins) may break into
corresponding subunits. Ionic interactions or
stacking can cause small molecules to behave
similarly to molecules of greater molecular weight.
When this occurs, as in the case of phosphate ions
with a 500 NMWL membrane, the small molecules
may not effectively permeate the membrane.
Millipore recommends the selection of a mem-
brane filter NMWL that is one half the size of the
molecule of interest. Other manufacturers may
Ultrafiltration separates proteins from soluble salts.
recommend a smaller differential between the size Concentration polarization slows down filtration.
The proteins form a gel layer on the membrane surface.

7
Flux (UF Flow Rate) Many antifoams exhibit a phenomenon called
cloud point. As temperatures increase, antifoam
During ultrafiltration, it is important to balance speed
comes out of solution, forming a second phase.
with retention to obtain optimal performance. A
Increasing temperature above the cloud point
membranes flux is defined as the flow rate divided
causes flux to decrease.
by the membrane area. Using membranes with
higher NMWL ratings will increase the flow, but pH
at the same time lower the retention. A membrane Changing solution pH often changes molecular
should be selected for required rejection, consistent structure. This is especially true for proteins. At its
with desired flow rate. This is determined by surface isoelectric point, a protein begins to precipitate,
area, macrosolute type, solubility, concentration causing a flux decrease.
and diffusivity, membrane type, temperature effects
on viscosity and, to some extent, pressure. When Fouling
concentration polarization is rate-controlling, flux Flux decrease due to concentration polarization
is affected by solute concentration, fluid velocity, should not be confused with the effect of membrane
flow channel dimensions, and temperature. fouling. Fouling is usually the deposition and
accumulation of submicron particles and solute on
Effects of Operating Parameters on Flux the membrane surface and/or crystallization and
precipitation of smaller solutes on or within the
Pressure
pores of the membrane. There may be a chemical
When ultrafiltering dilute protein solutions or colloid
interaction with the membrane.
suspensions, flux will increase with increasing
transmembrane pressure (TMP). These effects are Importance of Recovery
most apparent when operating under controlled
While rejection is used to characterize membrane
positive pressure, such as when using a stirred
performance, it does not always directly correlate
cell. When the process is membrane-controlled
with solute recovery from a sample or volume.
(i.e., when the resistance of the gel layer is much
Actual solute recoverythe amount of material
smaller than that of the membrane), the flux-pressure
recovered after ultrafiltrationis generally based
relationship is linear. When the process is controlled
on mass balance calculations.
by polarization (e.g., when the resistance of the
In many cases, especially when working with
O v e r v ie w o f M e m br a ne F ilt r at i o n Re cove r y

gel layer is much larger than that of the membrane),


small samples of dilute, valuable solutions, the
flux will reach a plateau and may actually decrease
degree of recovery of a target solute is vitally
with increases in pressure.
important. In such cases, potential loss by
Concentration non-specific adsorption must be considered.
When concentration of the retained species is very Different membrane materials adsorb biomol-
low, flux is independent of concentration. As solute ecules to varying degrees. Where maximum
concentration rises during operation, increased recovery is desired, the choice of a membrane
viscosity and the polarization effect cause flux with the least non-specific adsorbtivity is essential.
to decrease. Millipores Ultracel regenerated cellulose mem-
branes were specifically developed to minimize
Temperature non-specific adsorption.
Increasing the operating temperature normally Since adsorption is a direct function of membrane
increases UF rates. A higher temperature increases and device surface area, device size must be
solute diffusivity (typically 33.5% per degree considered when recovery is important. Small, dilute
Celsius for proteins) and decreases solution samples should be concentrated with membranes of
viscosity. Common practice is to operate at the minimal surface area, commensurate with achieve-
highest temperature tolerated by the solutes and ment of reasonable flow rates. Millipore offers a
the equipment. wide range of centrifugal devices, stirred cells, and
An exception to the rule is fermentation broth tangential flow systems with an extensive choice of
concentration in the presence of some antifoams. membrane areas and NMWLs.

8
Mode of Operation
The pressure required for ultrafiltration can be Normal vs. Tangential Flow Filtration
supplied in a number of different ways depending
Filtration can be broken down into two different
on the product in use. For example, Millipores
operational modes: normal flow filtration (NFF)
small volume ultrafiltration products generally use
and tangential flow filtration (TFF). The difference
centrifugal force. Pump pressure is used with the
in fluid flow between these two modes is shown
tangential-flow filtration (TFF) products and com-
in Figure 4.
pressed gas is utilized with the stirred cell products.
In addition, Millipore provides multiwell ultrafiltration
products that utilize vacuum and centrifugation.

Figure 4. Normal flow filtration (NFF) vs. tangential flow filtration (TFF)
In normal flow filtration (NFF), fluid is convected directly toward the the fluid flow occurs in the direction normal to the membrane surface,
membrane under an applied pressure. Particulates that are too large to so NFF is a more descriptive and preferred name. NFF can be used
pass through the pores of the membrane accumulate at the membrane for sterile filtration of clean streams, clarifying prefiltration, and virus/
surface or in the depth of the filtration media, while smaller molecules protein separations.
pass through to the downstream side. This type of process is often In tangential flow filtration (TFF), the fluid is pumped tangentially
called dead-end filtration. However, the term normal indicates that along the surface of the membrane. An applied pressure serves to
force a portion of the fluid through the membrane to the filtrate side.
Normal Flow Filtration Tangential Flow Filtration As in NFF, particulates and macromolecules that are too large to
pass through the membrane pores are retained on the upstream side.
Feed Flow Pressure Pressure
However, in this case the retained components do not build up at the
surface of the membrane. Instead, they are swept along by the tangen-
tial flow. This feature of TFF makes it an ideal process for finer sized-

O v e r v ie w o f M e m br a ne F ilt r at i o n M o d e of O p e r a t io n
based separations. Although TFF is more commonly associated with
Feed Flow
large scale processing, centrifugal UF devices with vertical membrane
panels, such as Amicon Ultra devices, also benefit from a TFF-like
mode of separation, particularly in a swinging bucket rotor. TFF is also
Membrane Membrane commonly called cross-flow filtration. However, the term tangential
is descriptive of the direction of fluid flow relative to the membrane,
so it is the preferred name.
Filtrate Filtrate

9
Diafiltration
Millipore membranes provide an inexpensive means With very small sample volumes, dilution of the
of separating macromolecular mixtures into size- sample before the initial concentration spin can
graded classes either by direct ultrafiltration or by often decrease salt concentration to an acceptable
diafiltration. Diafiltration removes microsolutes by level. For example, if a 200 L sample containing
adding solvent to the solution being ultrafiltered 100 mM salt is diluted to 4,000 L before concen-
at a rate equal to the UF rate, independent of tration in an Amicon Ultra centrifugal filter unit, the
microspecies concentration. This rapid, efficient salt concentration in the 4,000 L sample will be
process washes microspecies from the solution at 5 mM. The concentrate will still contain 5 mM
constant volume, thereby purifying the retained salt. If more complete salt removal is desired, a
species. This process is most effective if the passing re-dilution/spin cycle should be added. In this
molecules are at least 10 times smaller than the example, if the original spin ended with 50 L of
molecules to be retained and concentrated by the retentate, redilution to 4,000 L results in 0.06 mM
membrane. Diafiltration is useful for sample desalt- salt concentration. The sample can then be recon-
ing and buffer exchange. centrated to 50 L in an Amicon Ultra centrifugal
When diafiltration is used for sample desalting filter device.
or buffer exchange, there is no resulting change in Diafiltration can be a continuous or a discontinu-
buffer composition. A solution volume with 100 mM ous process. In continuous diafiltration, such
salt still contains 100 mM salt after the initial as in a stirred cell or a TFF device, the solution is
concentration spin. Rediluting the retentate with water maintained at a fixed volume while solvent flows
and spinning again effectively decreases the salt continuously through the mixture. Salts and other
concentration of the sample by the concentration microsolutes are steadily removed by convective
factor of the ultrafiltration. For example, if a 4,000 L transport. Microsolute exchange can be accom-
sample containing 100 mM salt is concentrated to plished using the same principle. Constant operator
50 L (80X) in an Amicon Ultra centrifugal filter unit, attention is not required and the possibility of solute
O v e r v ie w o f M e m br a ne F ilt r at i o n D i af i l t r a t io n

rediluted with water to 4,000 L, and reconcen- denaturation by overconcentration is eliminated.


trated, the salt concentration will be reduced 80X In discontinuous diafiltration, such as in a centrifugal
to 1.25 mM. ultrafiltration device, salts and microsolutes are
To achieve more complete salt removal, multiple removed by repeated concentration and dilution
concentration and redilution spins are required. For (Figure 5).
most samples, two concentration/reconstitution cycles
will remove about 99% of the initial salt content.

Figure 5. Removing salts from retained solutes using diafiltration

100 L Add 90 L 100 L


10 L 10 L
UF membrane Milli-Q water

10X 10-fold 10X


concentration 90 L dilution concentration 90 L
of protein of protein

100 mM 100 mM 10 mM 10 mM
NaCl NaCl NaCl NaCl

The sodium chloride concentration is reduced by dilution.

10
Dialysis vs. Diafiltration Table 1. Comparison of diafiltration and dialysis
Dialysis is a traditional method for removing Diafiltration Dialysis
microsolutes or exchanging solvents. It is a slow Transport convective with solvent, Transport diffusion-controlled,
diffusive process generally employing regenerated- independent of microsolute dependent on type of microsolute.
cellulose tubing as the barrier membrane. In dialysis, composition.
the process solution and exchange solvent are Rapid rate. Fractional removal Slow transport. Lower efficiency
on opposite sides of the semi-permeable barrier independent of content. with decreased microsolute
membrane through which permeating microsolutes concentration.

diffuse. The permeation rate of solutes from sample Ultrafiltration rate reduced Marked temperature dependence
with decreased temperature (reduced transport at lower
to dialysate is a direct ratio to the solute concentra-
(net effect not as marked). temperature).
tion and inversely proportional to the solutes
At elevated macrosolute content, Microsolute transport relatively
molecular weight. Desalting by dialysis is time-
ultrafiltration rate reduced. unaffected by macrosolute content.
consuming and relatively inefficient at low
Minimal exchange solvent Frequent dialysate change.
concentrations. required; easily contained in Recirculation about bags to
Millipores Amicon centrifugal concentrators reservoir. maximize transport.
provide a fast, convenient, high-recovery alternative Simple automation with endpoint Automation possible with complex
to dialysis or precipitation without diluting samples. control apparatus. equipment.
The relative merits of diafiltration and dialysis are
summarized in the Table 1.

Fractionation

O v e r v ie w o f M e m br a ne F ilt r at i o n F r a c t io n a t io n
Fractionation is the process of separating a mixture Factors to consider for efficient fractionation:
into its components using a combination of physico- 1. Selection of the appropriate molecular weight
chemical properties of the solute. Ultrafiltration cutoff: Selecting the appropriate MWCO of the
membranes have been used for fractionation of membrane is critical to ensure efficient fraction-
protein solutions on the basis of size1. This tech- ation. The size of the retained and the passing
nique is also called membrane partitioning chroma- species affect the selection criteria for the
tography1,2. Here, proteins larger than the pore size membrane pore size5.
are retained and those smaller than the pore size

pass through into the permeate. Since fractionation 1. Molecular weight of retained MWCO x 0.9
is rarely absolute, acceptability of the results will 2. MWCO Molecular weight of passing x 3
depend on whether the passing solute, retained
3. Molecular weight of retained
solute or both are of interest to the researcher and Molecular weight of passing x 3
their levels of purity and yield. Hence fractionation
efficiency is defined as a function of yield and purity Example separation: Separating a 10 kDa
relative to the starting solution3. species from a 40 kDa protein is very likely to
succeed using a 30K MWCO membrane. This
is due to the fact that the retained solute is >0.9X
of the MWCO. Secondly the size difference
between the solutes is 4X and the ratio of the
MWCO to the passing solute is also 4X.

11
In the case of separating an 8 kDa from a i.e. 87.5%. This relation holds true for the most
24 kDa using a 30K MWCO, fractionation will part since the total amount of the retained solute
not be successful because the 24 kDa species is is unchanged. With multiple spins, more of the
only 80% of the pore size of the 30K MWCO passing solute goes through into the permeate,
and it may leak into the permeate. Thus, careful leaving the retained fraction purer and increasing
attention needs to be paid to the size of the the yield of the passing solute.
solutes and the available MWCOs. 4. A serial fractionation strategy for compartmen-
If the application requires the retained species, talizing unknown or complex mixtures:
then the rules are reversed. For maximal purity, Our recommendation for fractionation of
the MWCO chosen should be very open, and for unknown mixtures is to start by separating the
maximal yield, the MWCO should be very tight. proteins using the highest MWCO available.
2. Starting concentration of proteins: Starting The permeate fraction from this separation is
concentrations of protein solutions affect fractionated on the next highest MWCO and
fractionation efficiency4,5. When fractionating so on, serially. Thus, the permeate fraction from
with an Amicon Ultra device, the starting a device with a 100K MWCO membrane
concentration of the passing solute, to 10 mg/ contains proteins <100 kDa and has undetect-
mL does not affect yields or purity of the solute in able amounts of proteins with higher molecular
the permeate fraction. At the other end, for dilute weights. Similarly, the permeate from the 50K
proteins, where the concentrations are below MWCO contains proteins <50 kDa and so on.
0.5 mg/mL, the polarizing gel-like layer does A point to note is that the efficiency of removing
not foul the membrane. In this case we recom- proteins higher than the cutoff depends on the
mend using the lowest MWCO possible to size difference between the solute and the
prevent any trace amounts of retained solute MWCO, and the starting concentration of that
appearing in the filtrate. solute. Smaller solutes will pass through preferen-
Example separation: Consider the separation of tially compared to the larger ones.*
a 17 kDa protein from a 66 kDa protein. If a
MWCO of 50K is chosen (ratio of MWCO to References
O v e r v ie w o f M e m br a ne F ilt r at i o n F r a c t io n a t io n

passing ~3X), yields and purity of the passing 1. Blatt WF, Hudson BG, Robinson SM, Zipilivan
solute are not affected even when the starting EM. Fractionation of protein solutions by
concentration of the retained is greater than membrane partition chromatography.
5 mg/mL. However, if the 30K MWCO is Nature 1967;(216)511-513.
chosen to maximize purity of the passing solute, 2. Blatt WF. Membrane partition chromatography:
then yields are significantly affected as concen- a tool for fractionation of protein mixtures.
tration of the retained increases to 5 mg/mL and J Agric Food Chem 1971;19:589-594.
beyond. This is due to the fact that the ratio of
3. Ngiam SH, Bracewell DG, Zhou Y, Titchener-
the MWCO to the passing solute is ~1.8X.
Hooker NJ. Quantifying process tradeoffs in
3. Multiple diafiltration steps to increase yields: the operation of chromatographic sequences.
In order to increase yields of the passing solute Biotechnol Prog 2003;19:1315-1322.
and to increase purity of the retained solute,
4. Robertson BC, Zydney AL. Polarization and
multiple spins are necessary. This approach is
adsorption effects on sieving in membrane
also called as diafiltration, where the retentate
protein filtration. ASAIO Trans 1987;33:
fraction is diluted back to the starting volume and
118-122.
the sample is centrifuged again. For e.g., if 50%
5. Nel RG, Oppenheim SF, Rodgers VG. Effects of
of the passing solute was recovered in the filtrate
solution properties on solute and permeate flux
in the first spin, 50% of the remaining, i.e., 25%
in bovine serum albumin-IgG ultrafiltration.
would be recovered in a subsequent spin. The
Biotechnol Prog 1994;10:539-542.
total for 3 spins would be 50 + 25 + 12.5,

*Refer to A Simple Strategy for Protein Fractionation Using


Ultrafiltration for a protocol.

12
Membranes and Devices Membrane Selection . . . . . . . . . . . . . . . . . . . . 14

Device Selection . . . . . . . . . . . . . . . . . . . . . . . 16

for Ultrafiltration Microcon Centrifugal Filters . . . . . . . . . . . . . . . 17

Amicon Ultra-4 Centrifugal Filters . . . . . . . . . . 18

Amicon Ultra-15 Centrifugal Filters . . . . . . . . . . 20

Centriprep Centrifugal Filters . . . . . . . . . . . . . . 22

Centricon Plus-70 Centrifugal Filters . . . . . . . . . 23

MultiScreen Filter Plate . . . . . . . . . . . . . . . . . . 24

Ultrafree Centrifugal Filters . . . . . . . . . . . . . . . 25

Pellicon XL Cassettes and


Labscale TFF System . . . . . . . . . . . . . . . . . . . . 26

Pellicon 2 Mini Cassettes . . . . . . . . . . . . . . . . . 28

Prep/Scale Spiral Wound Filter Cartridges . . . . 29

Stirred Cells . . . . . . . . . . . . . . . . . . . . . . . . . . 30

Ultrafiltration Discs . . . . . . . . . . . . . . . . . . . . . 31

e m br
Guid
M ct inD ge vMiceems br
e taoneSse laend a f ilt
s arnd
f oarneUlt ic ei os n M e m b r a n e P ro ce s s e s
D e rv at

13
Membrane Selection
Millipore offers a complete range of centrifugal Biomax Ultrafiltration Membrane
devices used for sample concentration, purification, To concentrate or desalt higher volumes of more
and desalting or buffer exchange of soluble concentrated samples (recommended for protein
macromolecules. Millipore products are also concentrations greater than 1.0 mg/mL), use
available for applications such as cleaning up Biomax polyethersulfone (PES) ultrafiltration mem-
PCR reactions, separating protein-bound from free branes. Biomax membranes are recommended for
ligands, removing restriction enzymes, and recover- samples such as serum, plasma, or conditioned
ing oligonucleotides from agarose gels. This section tissue culture media.
provides information to aid in choosing the correct
product for a particular application. Durapore Microporous Membrane
Millipore offers three distinct types of membranes To clarify biological samples, recover DNA from
to choose from. This section will describe these agarose gels, retain chromatography resins or
three types of membranes and then provide suspended solid media, use Durapore hydrophilic
information about choosing the correct membrane PVDF microporous membranes. Durapore mem-
based on typical recoveries or application. branes allow all soluble protein and nucleic acids
to pass, retaining sub-cellular fragments, whole cell
Types of Membranes and particulate materials. Durapore membranes are
extremely hydrophilic, and they provide the lowest
Ultracel Ultrafiltration Membrane
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n M e m b r a n e S e l e c t io n

binding of proteins and other biologicals of all


To concentrate or desalt dilute solutions, use commercially available microporous membranes.
Ultracel series regenerated cellulose ultrafiltration
membranes. The hydrophilic, tight microstructure Membranes Organized by Typical Recoveries
of Ultracel membranes assures the highest possible For globular proteins, there is a good correlation
retention with the lowest possible adsorption of between molecular weight and Stokes radius.
protein, DNA or other macromolecules. This usually allows one to predict the recovery of a
protein based on its molecular weight if a membrane
with the same nominal molecular weight rating is
used (see typical recovery of a panel of protein
Table 1. Membrane selection by recovery solutes in Table 1). However, in order to accommo-
Cytochrome c BSA IgG date the wide range of potential protein solutes with
NMWL (12.4 kDa) (67 kDa) (156 kDa) different tertiary structures, we suggest initially using
3K Q Q Q the rule of two to ensure optimal recovery.
10K P Q Q
Rule of Two
30K Q Q
For Ultracel (regenerated cellulosic) membranes,
50K P Q
Millipore recommends using a membrane with
100K P a NMWL at least two times smaller than the
molecular weight of the protein solute that one
intends to concentrate.

Rule of Three
For Biomax (polyethersulfone) membranes for
stirred cells and TFF, Millipore recommends using
a membrane with a NMWL at least three times
smaller than the molecular weight of the protein
solute that one intends to concentrate.

14
Membranes Organized by Application
See Table 2 to determine which centrifugal product
to use based on application and membrane.

Microporous
Table 2. Membrane selection by application Ultrafiltration Membranes Membranes Specialty
Molecular Weight Devices
Type* Pore Size (m)
(NMWL)

Montage Gel Extraction


Micropure-EZ
Montage PCR
Ultracel
Biomax

100K

0.45
0.65
30K
50K
10K

0.2

5.0
0.1
3K
5K
Protein concentration
Protein purification/desalting/buffer exchange

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n M e m b r a n e S e l e c t io n
Desalting of column fractions
Protein isolation from cell lysates
Peptide concentration/desalting/buffer exchange
Antibody concentration
Virus concentration or removal
Nucleic acid concentration/desalting/buffer exchange
Oligonucleotide concentration/desalting/buffer exchange
PCR cleanup
Remove linkers prior to cloning
Remove labeled nucleotides
Antibody purification from hybridoma cells
Rapid restriction mapping
Clarify samples of particulate prior to HPLC
Clarification of cell lysates and tissue homogenates
Cell harvesting
Natural product screening
Restriction enzyme removal
Bound vs. free drugs from serum/plasma (protein removal)
DNA/RNA recovery from polyacrylamide gel
DNA recovery from agarose gel
Oligonucleotide recovery from polyacrylamide gel
Removal of unincorporated label (e.g., fluorescein) from protein
Removal of imidazole from His-tag fusion protein
*Selection of ultrafiltration membrane: Ultracel regenerated cellulose membrane is ideal for protein samples and nucleic acids. Biomax polysulfone
membrane is ideal for complex samples (e.g., serum).

15
Device Selection
See Table 3 to determine which centrifugal product
to use for protein concentration based
on initial sample molecular weight and volume.

Table 3. Protein concentration devices by filtration capacity


Filtration Capacity
Membrane
0.5 2 4 15 20 70 1 2 10 10
Type
Millipore Device mL mL mL mL mL mL L L L L
Small Volume Filtration Devices
Microcon Centrifugal Filters* U
Ultrafree-MC Centrifugal Filters M
MultiScreen Filter Plate with Ultracel Membrane** U
Medium Volume Filtration Devices
Ultrafree-CL Centrifugal Filters M
Amicon Ultra-4 Centrifugal Filters U
Amicon Ultra-15 Centrifugal Filters U
Centriprep Centrifugal Filters U
Large Volume Filtration Devices
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n D ev ice S e l e c t io n

Centricon Plus-70 Centrifugal Filters U


Amicon Stirred Cells
Series 8000, High Output, Solvent-Resistant Stirred Cells U
Tangential Flow Filtration (TFF)
Pellicon XL 50 Ultrafiltration Devices U
Prep/Scale Spiral Wound UF Modules

U
Pellicon 2 Ultrafiltration Modules U
U = Ultrafiltration M = Microporous
*Bold type indicates recommended devices.
**96-well plate. Volume per well.

16
Microcon Centrifugal Filters

mL
0.5
Microcon centrifugal filters are the lab standard
for small volume concentration.
They allow you to process macromolecular Highest
solutions up to 500 L using any centrifuge that can Recovery
accept 1.5 mL tubes. The devices low-adsorption
Ultracel YM membrane and patented invert
recovery spin combine to yield unusually high
recovery ratestypically >95% of the sample,
with concentration factors as high as 100X.
Maximum starting volume: 500 L
Typical sample concentration volume: 515 L
Low-binding Ultracel-YM regenerated
cellulose membrane
Solute recoveries typically >95%

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n M icro co n C e n t rif u g a l F i l te r s


Typical Protein Recovery
Retentate Recovery (%) by Nominal MW
Solute (Concentration) MW 3,000 10,000 30,000 50,000 100,000
Cytochrome c (0.25 mg/mL) 12,400 94 95
-Chymotrypsinogen (1 mg/mL) 25,000 95
Ovalbumin (1 mg/mL) 45,000 95
Bovine Serum Albumin (1 mg/mL) 67,000 95 95
Bovine IgG Fraction II (1 mg/mL) 156,000 95 95

Protocol
Ordering Information
Description NMWL Qty/Pk* Catalogue No.
Microcon Filter Units 3,000 24 42403
Add
Sample 100 42404
10,000 24 42406
100 42407
30,000 24 42409
100 42410
Invert into 50,000 24 42415
Receiver 100 42416
100,000 24 42412
100 42413

*Additional package sizes available. Contact Millipore.


Recover
Sample

For additional information, visit www.millipore.com/microcon

17
Amicon Ultra-4 Centrifugal Filters
1 4 m L

Amicon Ultra-4 Centrifugal Filters set the perfor-


mance standard for concentrating small-volume
samples. Ultracel regenerated cellulose low-binding
ultrafiltration membrane combines with a vertical
housing for fast sample processing with high
recovery. Ultra
Processes 24 mL in as few as 10 minutes Recovery,
Ultra
>90% typical sample recoveries Speed

Compatible with most rotor types


Double membrane panels increase surface
area and reduce filtration times
Deadstop prevents sample from spinning
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n A m ico n U l t r a - 4 C e n t rif u g a l F i l te r s

to dryness and eliminates the need for an


inverse spin
-marked for in vitro diagnostic use
(10K NMWL)

Typical Spin Times Comparative Performance


Spin conditions: 4000 x g, swinging bucket rotor at 100
25 C, 4 mL sample. 3K and 10K: Cytochrome c,
0.25 mg/mL; 30K and 50K: BSA, 1 mg/mL; 80
Recovery (%)

100K: lgG, 1 mg/mL.


60
4.0
40
3.5
Filtrate Volume (mL)

3.0 20
2.5
0
2.0 3,000 NMWL Brand V Brand PG Brand P Amicon Ultra-4
1.5 10,000 NMWL
30,000 NMWL Devices with 10 kDa* NMWL using Cytochrome c
1.0 50,000 NMWL
0.5 100,000 NMWL
(0.025 mg/mL).
0
0 5 10 15 20 25 30
Spin Time (min)

Complex sample volumes of 4 mL can be concen-


trated or diafiltered in as few as 15 minutes.

*Brand PG has a 9K membrane.

18
Typical Protein Recovery

1 4 m L
Retentate Recovery (%) by Nominal MW
Solute (Concentration) MW 3,000 10,000 30,000 50,000 100,000
Cytochrome c (0.25 mg/mL) 12,400 94 95
Bovine Serum Albumin (1 mg/mL) 67,000 95 94
IgG (1 mg/mL) 156,000 91
Typical recoveries for 4 mL starting volume in fixed angle rotor at 7500 x g at 25 C. Spin times: 5K (20 minutes); 10 and 30K (10 minutes);
50K (5 minutes); 100K (15 minutes).

Protocol
Ordering Information
Description NMWL Qty/Pk* Catalogue No.
Amicon Ultra-4 Centrifugal Filters 3,000 8 UFC8 003 08
are assembled with 24 UFC8 003 24
centrifuge tubes and caps 96 UFC8 003 96
Add
10,000 8 UFC8 010 08
Sample
24 UFC8 010 24

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n A m ico n U l t r a - 4 C e n t rif u g a l F i l te r s


96 UFC8 010 96
30,000 8 UFC8 030 08
24 UFC8 030 24
96 UFC8 030 96
50,000 8 UFC8 050 08
24 UFC8 050 24
96 UFC8 050 96
100,000 8 UFC8 100 08
24 UFC8 100 24
96 UFC8 100 96

*Additional package sizes available. Contact Millipore.

Recover
Purified
Sample

For additional information, visit www.millipore.com/ultra4

19
Amicon Ultra-15 Centrifugal Filters
mL
15

Amicon Ultra-15 Centrifugal Filters are the premier


devices for concentrating mid-volume samples.
They incorporate low-binding Ultracel regenerated
Ultra
cellulose ultrafiltration membrane and a vertical Recovery,
design for maximum recovery and minimal spin times. Ultra
Speed
Processes up to 15 mL in as few as 10 minutes
>90% typical sample recoveries
Compatible with most rotor types
Double membrane panels increase surface area
and reduce filtration times
Deadstop prevents sample from spinning to dry-
ness and eliminates the need for an inverse spin
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n A m ico n U l t r a -15 C e n t rif u g a l F i l te r s

-marked for in vitro diagnostic use


(10K NMWL)

Typical Spin Times Comparative Performance


Spin conditions: 4000 x g, swinging bucket rotor at 100
25 C, 15 mL sample. 3K and 10K: Cytochrome c,
0.25 mg/mL; 30K and 50K: BSA, 1 mg/mL; 80
Recover (%)

100K: lgG, 1 mg/mL.


60
16
40
14
Filtrate Volume (mL)

12 20
10
0
8 3,000 NMWL Brand V Brand PG Brand P Amicon Ultra-15
6 10,000 NMWL
30,000 NMWL Devices with 10 kDa* NMWL using Cytochrome c
4 50,000 NMWL
2 100,000 NMWL
(0.025 mg/mL).
0
0 5 10 15 20 25 30 35 40 45 50 55 60 65
Spin Time (min)

Complex sample volumes of 15 mL can be concen-


trated or diafiltered in as few as 15 minutes.

*Brand PG has a 9K membrane.

20
Typical Protein Recovery

mL
Retentate Recovery (%) by Nominal MW

15
Solute (Concentration) MW 3,000 10,000 30,000 50,000 100,000
Cytochrome c (0.25 mg/mL) 12,400 91 93
Bovine Serum Albumin (1 mg/mL) 67,000 98 93
IgG (1 mg/mL) 156,000 89
Typical recoveries for 15 mL starting volume in swinging bucket rotor at 400 x g at 25 C. Spin times: 5K (45 minutes);
10 and 100K (20 minutes); 30K (10 minutes); 50K (15 minutes).

Protocol
Ordering Information
Description NMWL Qty/Pk* Catalogue No.
Amicon Ultra-15 Centrifugal 3,000 8 UFC9 003 08
Filters are assembled with 24 UFC9 003 24
Add centrifuge tubes and caps 96 UFC9 003 96
Sample
10,000 8 UFC9 010 08
24 UFC9 010 24

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n A m ico n U l t r a -15 C e n t rif u g a l F i l te r s


96 UFC9 010 96
30,000 8 UFC9 030 08
24 UFC9 030 24
96 UFC9 030 96
50,000 8 UFC9 050 08
24 UFC9 050 24
96 UFC9 050 96
100,000 8 UFC9 100 08
24 UFC9 100 24
96 UFC9 100 96

Recover *Additional package sizes available. Contact Millipore.


Purified
Sample

For additional information, visit www.millipore.com/ultra15

21
Centriprep Centrifugal Filters
5 15 m L

Use Centriprep Centrifugal Filters to concentrate


and desalt high solute biological samples in the
For
515 mL volume range. The devices are compatible High Solute
with most centrifuges that accommodate 50 mL Samples
centrifuge tubes.
Unique inverse flow mode of operation
with large deadstop
Starting volume from 515 mL
Low-binding Ultracel regenerated
cellulose membrane
Fast sample processing with typical
recovery of >90%

Typical Protein Recovery


M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n C e n t ri p re p C e n t rif u g a l F i l te r s

Retentate Recovery (%)


Solute (Concentration) Nominal MW 3,000 10,000 30,000 50,000
Cytochrome c (0.25 mg/mL) 12,400 90 85
-Chymotrypsinogen (1 mg/mL) 25,000 90
Ovalbumin (1 mg/mL) 45,000 90
Bovine Serum Albumin (1 mg/mL) 67,000 90
Bovine IgG Fraction II (1 mg/mL) 156,000 90

Protocol
Ordering Information
Description Membrane NMWL Qty/Pk* Catalogue No.

Disassemble Centriprep 3,000 3,000 24 4302


Device and Centrifugal Filters 96 4303
Add Sample 10,000 10,000 24 4304
96 4305
Reassemble 30,000 30,000 24 4306
and Place in 96 4307
Centriprep
50,000 50,000 24 4310
96 4311
Centrifuge Until
Equilibrium is *Additional package sizes available. Contact Millipore.
Reached

Decant Filtrate
and Centrifuge
Again

Disassemble and
Recover Retentate

22
Centricon Plus-70 Centrifugal Filters

30 70 m L
The Centricon Plus-70 device can concentrate
most 70 mL solutions down to 350 L in less than 25
Convenient
minutes, making it a convenient alternative to stirred Alternative
cells. Typical sample recoveries are >90% with minimal to Stirred
sample loss due to non-specific binding. Cells

High concentration factors, with samples typically


concentrated in the 50X to 200X range
Low hold-up volume
Invert spin method of recovery
Low binding Ultracel regenerated cellulose mem-
brane
Low-binding polypropylene housing

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n C e n t rico n P lu s -70 C e n t rif u g a l F i l te r s


Typical Protein Recovery
Retentate Recovery (%)
Solute (Concentration) Nominal MW 3,000 10,000 30,000 50,000
Cytochrome c (0.25 mg/mL) 12,400 94
Bovine Serum Albumin (1 mg/mL) 67,000 85 95 96 84
IgG Fraction II (1 mg/mL) 156,000 94 91 91

Protocol Typical Spin Times


80
Filtrate Volume (mL)

60
Add Sample

40 PBS
0.25 mg/mL BSA
1.0 mg/mL BSA
20 5.0 mg/mL BSA

0
0 10 20 30 40
Time (min)
Invert into
Receiver 5K NMWL Biomax membrane spun at 3500 x g in a
swinging bucket rotor.

Ordering Information
Recover Purified
Sample Description Membrane NMWL Qty/Pk* Catalogue No.
Centricon Plus-70 Biomax 5,000 8 UFC7 005 08
Centrifugal Filters Ultracel 10,000 8 UFC7 010 08
30,000 8 UFC7 030 08
100,000 8 UFC7 100 08

*Additional package sizes available. Contact Millipore.

For additional information, visit www.millipore.com/centricon70

23
MultiScreen Filter Plate with
mL
< 0.5

Ultracel-10 Membrane
The first automation high throughput ultrafiltration
plate for protein purification. 10,000 NMWL
Ultracel regenerated cellulose membrane provides
low non-specific binding and high protein recovery.
Processes from 50 to 500 L
95% typical retention of Cytochrome c
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n M u l t i S cre e n F i l te r P l a te w i t h U l t r a ce l -10 M e m b ra n e

Compatible with standard microtiter receiver


plates (300 L, 700 L deep well, 150 L
conical bottom)
Fast, easy centrifugal protocol
Ideally suited for instrumentation and
liquid handling equipment
Uniform performance from well to well

Typical Spin Times


300
Typical Protein Recovery 250
Buffer
Cytochrome c (1 mg/mL
Filtrate Volume (L)

BSA (1 mg/mL)
Membrane Protein Typical Protein 200 Fetal Bovine Serum
NMWL Solute Retention (%) 150
10,000 Cytochrome c, 95
100
12,400 daltons
(1 mg/mL) 50

10,000 BSA, 99 0
0 10 20 30 40 50 60
67,000 daltons Spin Time (min)
(1 mg/mL)
300 L sample spun at 2000 x g in swinging
300 L sample spun at 2000 x g in swinging bucket rotor.
bucket rotor.

Ordering Information
Description Qty/Pk* Catalogue No.
MultiScreen Filter 10 MAUF 010 10
Plate with Ultracel-10
membrane

*Additional package sizes available. Contact Millipore.

For additional information, visit www.millipore.com/ultracel

24
Ultrafree Centrifugal Filters

0.12 m L
for Sample Clarification
Ultrafree-MC Centrifugal Filters
For sample clarification with low hold-up
Maximum starting volume: 500 L
Hold-up volume: <5 L

Ultrafree-CL Centrifugal Filters


For sample clarification with high recovery
Maximum starting volume: 2 mL
Hold-up volume: <10 L Ultrafree-MC

Ordering Information

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n Ul t r af re e C e n t rif u g a l F i l te r s


Description Membrane Pore Size (m) Sterility Qty/Pk Catalogue No.
Ultrafree-MC Durapore (PVDF) 0.1 Non-sterile 25 UFC3 0VV 25
Centrifugal 100 UFC3 0VV 00
Filters 0.22 Non-sterile 25 UFC3 0GV 25
100 UFC3 0GV 0S
Sterile 50 UFC3 0GV 05
0.45 Non-sterile 25 UFC3 0HV 25
Non-sterile 100 UFC3 0HV 00
Sterile 50 UFC3 0HV 0S
0.65 Non-sterile 25 UFC3 0DV 25
Ultrafree-CL
100 UFC3 0DV 00
Sterile 50 UFC3 0DV 0S
5.0 Non-sterile 100 UFC3 0SV 00
Protocol Hydrophilic PTFE 0.22 Non-sterile 25 UFC3 0LG 25
0.45 Non-sterile 25 UFC3 0LH 25
Ultrafree-CL Durapore (PVDF) 0.1 Non-sterile 25 UFC4 0VV 25
Add Centrifugal 100 UFC4 0VV 00
Sample Filters 0.22 Non-sterile 25 UFC4 0GV 25
100 UFC4 0GV 00
0.45 Non-sterile 25 UFC4 0HV 25
100 UFC4 0HV 00
Close 0.65 Non-sterile 25 UFC4 0DV 25
Lid
5.0 Non-sterile 25 UFC4 0SV 25
Hydrophilic PTFE 0.22 Non-sterile 25 UFC4 0LG 25
0.45 Non-sterile 25 UFC4 0LH 25

Additional package sizes available. Contact Millipore.

Collect
Particulates
Sample

Filtrate

25
Pellicon XL Cassettes and
12 L

Labscale TFF System


Pellicon XL cassettes allow you to easily concentrate
or diafilter samples using tangential flow filtration.
The cassettes are available with either microfiltration
or ultrafiltration membranes.
Use cassettes with Ultracel regenerated
cellulose or Biomax polyethersulfone
(ultrafiltration) membranes for
concentrating, desalting proteins,
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n Pe l l ico n X L C a s s e t te s a n d L a b s c a l e T F F Sy s te m

polysaccharides, lipid solutions,


cell suspensions, and mammalian cells.
Use cassettes with Durapore PVDF
(microporous) membrane for sample preparation, The Labscale TFF system is specifically designed
washing, or cell harvesting, or for clarifying cell to operate Pellicon XL modules. The system reservoir
cultures, lysates, or fermentation broths. accepts direct docking of the device, eliminating the
need for tubing connections.
Provides the lowest working volumes, with typical
sample concentrations of >25X
An optional 100 mL reservoir is available for
low volume applications

Typical Processing Times


Pellicon XL
150

125
Process Flux (Lmh)

100

75

50

25

0
0 1 10 100
Protein Concentration (g/L)

Pellicon XL cassettes provide rapid, efficient concen-


tration of protein-containing solutions. Data show
concentration of BSA with a 10K NMWL cassette
with Ultracel regenerated cellulose membrane on a
LabScale TFF system operating at 13 C with 40 psi
inlet pressure and 20 psi outlet pressure.

26
Ordering Information

12 L
Description Membrane NMWL Catalogue No.
Pellicon XL Ultracel Regenerated 5,000 PXC0 05C 50
Cassettes Cellulose 10,000 PXC0 10C 50
30,000 PXC0 30C 50
300,000 PXC3 00C 50
1,000,000 PXC0 1MC 50
Biomax 5,000 PXB0 05A 50
Polyethersulfone 8,000 PXB0 08A 50
10,000 PXB0 10A 50
30,000 PXB0 30A 50
50,000 PXB0 50A 50
100,000 PXB1 00C 50
300,000 PXB3 00C 50 Multi-Manifold accessory allows

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n Pe l l ico n X L C a s s e t te s a n d L a b s c a l e T F F Sy s te m


500,000 PXB5 00C 50 up to three cassettes to be mounted
on the Labscale system for processing
1,000,000 PXB0 1MC 50
multi-liter samples.

Description Membrane Pore Size (m) Catalogue No.


Pellicon XL Durapore PVDF 0.10 PXVV PPC 50
Cassettes 0.22 PXGV PPC 50
0.45 PXHV MPC 50
0.65 PXDV PPC 50

Description Pore Size (m) Catalogue No.


System kit includes pump module, stir base, 500 mL and
100 mL reservoirs, stand for 100 mL reservoir, and multi-manifold.
Labscale TFF System 115 V XX42 LSS 11
230 V XX42 LSS 12
GB-230 V XX42 LSS 13

27
Pellicon 2 Mini-Cassettes
L
10

Cassette-style Pellicon 2 modules are available


with Durapore PVDF (microporous) and Ultracel
regenerated cellulose and Biomax polyethersulfone
(ultrafiltration) membranes for high performance
in large volume concentration and diafiltration
applications, as well as scale-up applications.
Cassettes for low viscosity (A-screen), low to
medium viscosity (C-screen), and high viscosity/
high concentration (V-screen) solutions.
Larger Pellicon cassette systems are available
for processing 250 L or more. Contact Millipore
or visit our web site for more information.
Stainless steel Pellicon 2 Mini-Cassette Holder
can operate up to three mini-cassettes.

Ordering Information
M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n Pe l l ico n 2 M i n i - C a s s e t te s

Description Membrane NMWL A Screen C Screen V Screen


Pellicon 2 Biomax 5,000 P2B0 05A 01 P2B0 05V 01
Mini-Cassettes Polyethersulfone 8,000 P2B0 08A 01 P2B0 08V 01
10,000 P2B0 10A 01 P2B0 10V 01
30,000 P2B0 30A 01 P2B0 30V 01
50,000 P2B0 50A 01 P2B0 50C 01 P2B0 50V 01
100,000 P2B1 00A 01 P2B1 00C 01 P2B1 00V 01
300,000 P2B3 00C 01 P2B3 00V 01
500,000 P2B5 00C 01 P2B5 00V 01
1,000,000 P2B0 1MC 01 P2B0 1MV 01
Ultracel 5,000 P2C0 05C 01 P2C0 05V 01
Regenerated 10,000 P2C0 10C 01 P2C0 10V 01
Cellulose 30,000 P2C0 30C 01 P2C0 30V 01
100,000 P2C1 00C 01 P2C1 00V 01
300,000 P2C3 00C 01 P2C3 00V 01
1,000,000 P2C0 1MC 01 P2C0 1MV 01

Accessories
Description Catalogue No.
Pellicon 2 Mini Holder XX42 PMI NI
Pellicon 2 Mini Holder Fitting Kit XX42 PFK 01

28
Prep/Scale Spiral Wound
Filter Cartridges
Prep/Scale spiral wound filter cartridges are
available in three sizes for easy, reliable preparation
of samples ranging from 100 liters down to 100 mL.
Self-contained design ensures leak-free filtration
Hose barb connectors for easy setup
Can be operated with standard 1 to
16 L/minutes peristaltic pumps
Prep/Scale Holder includes fittings and basic

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n P re p / S c a l e S p i r a l Wo u n d F i l te r C a r t ri d g e s
instrumentation

Three Cartridge Sizes


Prep/Scale- Prep/Scale- Prep/Scale-
TFF-1 TFF-2 TFF-6
Filter Area (m2) 0.1 0.23 0.54
Minimum Working 100 150 250
Volumes (mL)*
*Including pump tubing

Ordering Information
Description Membrane NMWL Prep/Scale-TFF-1 Prep/Scale-TFF-2 Prep/Scale-TFF-6
Prep/Scale Spiral Regenerated 1,000 CDUF 001 LA CDUF 002 LA CDUF 006 LA
Wound Filer Cellulose 3,000 CDUF 001 LB CDUF 002 LB CDUF 006 LB
Cartridges 5,000 CDUF 001 LC CDUF 002 LC CDUF 006 LC
10,000 CDUF 001 LG CDUF 002 LG CDUF 006 LG
30,000 CDUF 001 LT CDUF 002 LT CDUF 006 LT
100,000 CDUF 001 LH CDUF 002 LH CDUF 006 LH
300,000 CDUF 001 LM CDUF 002 LM CDUF 006 LM
Polyethersulfore 10,000 CDUF 001 TG CDUF 002 TG CDUF 006 TG
30,000 CDUF 001 TT CDUF 002 TT CDUF 006 TT
50,000 CDUF 001 TQ CDUF 002 TQ CDUF 006 TQ
100,000 CDUF 001 TH CDUF 002 TH CDUF 006 TH
300,000 CDUF 001 TM CDUF 002 TM CDUF 006 TM

Accessories
Description Catalogue No.
Prep/Scale Module Holder XX42 PS0 01
Peristaltic Pumps for TFF-1 use XX8200
for TFF-2 and TFF-6 use XX80 EL

29
Stirred Cells
Concentrate, diafilter, and exchange buffers
for macromolecule solutions including proteins,
enzymes, antibodies and viruses.

Series 8000 Stirred Cells


Five different sizes handle volumes from
3 mL to 400 mL
High flow rates with solutions up to
10% macrosolute concentration

Solvent-Resistant Stirred Cells


Two sizes: 75 mL (47 mm disc) and
300 mL (76 mm disc)
Borosilicate glass cylinder and PTFE Series 8000 Stirred Cell

components for broad chemical compatibility

Series 8000 Models


l s ce s s e s

Model 8003 Model 8010 Model 8050 Model 8200 Model 8400
b r adn eC ePl ro

Maximum Process Volume (mL) 3 10 50 200 400


Minimum Process Volume (mL) 0.075 1.0 2.5 5.0 10.0
e mi rre

Membrane Diameter (mm) 25 25 44.5 63.5 76


ic ei os n M St

Effective Membrane Area (cm ) 2


0.9 4.1 13.4 28.7 41.8
Hold-up Volume (mL) 0.07 0.2 0.5 1.2 1.5
D e rv at
a f ilt

Ordering Information
s arnd

Description Catalogue No.


f oarneUlt

Series 8000 Stirred Cells Model 8003 5125


Model 8010 5121
ct inD ge vMiceems br

Model 8050 5122


Model 8200 5123
Model 8400 5124
Solvent-Resistant Stirred Cells For 47 mm membranes XFUF 047 01
e taoneSse laend

For 76 mm membranes XFUF 076 01

Solvent-Resistant
Stirred Cell
e m br
Guid
M

30
Ultrafiltration Discs
High Recovery Ultracel Ultrafiltration Membranes
Hydrophilic, tight microstructure for highest
possible retention with lowest possible protein
adsorption
For use when concentrating or desalting
extremely dilute solutions or whenever your
sample is hydrophobic

High Flow Biomax Ultrafiltration Membranes


Open microstructure speeds concentration
To concentrate and desalt higher volumes or
more concentrated samples

Ordering Information

M e m br a ne s a nd D e v ic e s f o r Ult r a f ilt r at i o n Ul t r af i l t r a t io n D i s c s
Ultracel Ultrafiltration Discs
Regenerated Cellulose
NMWL
Filter
Diameter (mm) Qty/Pk 1,000 3,000 5,000 10,000 30,000 100,000
25 10 PLAC 025 10 PLBC 025 10 PLCC 025 10 PLGC 025 10 PLTK 025 10 PLHK 025 10
44.5 10 PLAC 043 10 PLBC 043 10 PLCC 043 10 PLGC 043 10 PLTK 043 10 PLHK 043 10
47 10 PLAC 047 10 PLBC 047 10 PLCC 047 10 PLGC 047 10 PLTK 047 10 PLHK 047 10
63.5 10 PLAC 062 10 PLBC 062 10 PLCC 062 10 PLGC 062 10 PLTK 062 10 PLHK 062 10
76 10 PLAC 076 10 PLBC 076 10 PLCC 062 10 PLGC 076 10 PLTK 076 10 PLHK 076 10
90 5 PLAC 090 05 PLBC 090 05 PLCC 090 05 PLGC 090 05 PLHK 090 05 PLHK 090 05
150 5 PLAC 150 05 PLBC 150 05 PLCC 150 05 PLGC 150 05 PLTK 150 05 PLHK 150 05

Biomax Ultrafiltration Discs


Polyethersulfone
NMWL
Filter
Diameter (mm) Qty/Pk 5,000 10,000 30,000 50,000 100,000
25 10 PBCC 025 10 PBGC 025 10 PBTK 025 10 PBQK 025 10 PBHK 025 10
44.5 10 PBCC 043 10 PBGC 043 10 PBTK 043 10 PBQK 043 10 PBHK 043 10
47 10 PBCC 047 10 PBGC 047 10 PBTK 047 10 PBQK 047 10 PBHK 047 10
63.5 10 PBCC 062 10 PBGC 062 10 PBTK 062 10 PBQK 062 10 PBHK 062 10
76 10 PBCC 076 10 PBGC 076 10 PBTK 076 10 PBQK 076 10 PBHK 076 10
90 5 PBCC 090 05 PBGC 090 05 PBTK 090 05 PBQK 090 05 PBHK 090 05
150 5 PBCC 150 05 PBGC 150 05 PBTK 150 05 PBQK 150 05 PBHK 150 05

31
32
Guid e t o S e l e ct in g M e m br a ne s a nd D e v ic e s
Protocols
Concentration, Desalting and Buffer Exchange . 34

Detergent Removal . . . . . . . . . . . . . . . . . . . . 38

for Proteins Two-Dimensional Electrophoresis


Sample Preparation . . . . . . . . . . . . . . . . . . . . 40

Purification of Serum Peptides for Biomarker . . . 42

Rapid Antibody Concentration . . . . . . . . . . . . . 45

Affinity Purification . . . . . . . . . . . . . . . . . . . . . 46

Removal of Unincorporated Label


from Labeled Protein . . . . . . . . . . . . . . . . . . . . 50

Rapid Purification of Monoclonal Antibodies . . . 52

A Simple Strategy for Protein Fractionation . . . . 54

Urine Concentration . . . . . . . . . . . . . . . . . . . . 56
Use of Centrifugal Filter Devices as an
Alternative to Stirred Cells . . . . . . . . . . . . . . . . 58

P ro t o c o l s f o r P ro t e in s

33
Concentration, Desalting, and
Buffer Exchange with Amicon Ultra
or Microcon Centrifugal Filters
Introduction Method
Amicon centrifugal devices from Millipore are ideal 1. Select the device with the appropriate NMWL
for removal or exchange of salts, sugars, nucleo- and volume for the application.
tides, and non-aqueous solvents, as well as other 2. Add the sample to the reservoir of the
materials of low molecular weight. They also serve centrifugal device.
to separate free from bound species.
3. If the sample is smaller than the maximum
Millipore centrifugal concentrators provide fast,
volume, it can be diluted up to the maximum
convenient, high-recovery alternatives to dialysis
volume before the first centrifugation step.
and ethanol precipitation. Sample dilution, often
This will help increase the salt removal.
associated with spin columns, is not a problem.
4. Centrifuge at the specified g-force for the
Salt transfer across the membrane is efficient and
recommended amount of time.
independent of microsolute concentration or size.
5. Remove the initial filtrate from the filtrate tube
P ro t o c o l s f o r P ro t e in s C o n ce n t r a t io n, D e s a l t i n g a n d Bu f f e r E xch a n g e

Millipores Amicon Ultra-4 and -15 centrifugal


filters are designed for high speed with high and set aside.
recovery. The devices incorporate low-binding 6. Add enough buffer or water to the device to
Ultracel regenerated cellulose ultrafiltration mem- bring the sample volume up to 4 or 15 mL.
brane for sample concentration and purification of 7. Centrifuge again.
solutions containing dilute or purified protein solutes, 8. Set aside the filtrate.
antigens, antibodies, enzymes, or microorganisms. 9. Recover the concentrated, de-salted sample.
Their speed and excellent recovery make them ideal
for desalting and buffer exchange applications. NOTE: Both of the filtrates should be retained until
One of the most common applications for Amicon the concentrated sample has been analyzed.
Ultra devices is concentration and desalting of
column fractions during protein purification by Results
various chromatography methods. Two examples The transfer of salts across a membrane filter is
below demonstrate the use of Amicon Ultra devices independent of sample concentration or size. There
for high protein and enzymatic activity recovery. is no change in the composition of the buffer when
desalting using ultrafiltration. For example, a solution
containing 500 mM salt still contains that concen-
tration after the initial centrifugation. Adding another
volume of salt-free buffer or water to the retentate
and centrifuging again will reduce the salt concen-
tration. This process, known as diafiltration, can
be repeated to achieve maximum salt removal.
Diafiltration can also be used when it is desirable to
have the sample in a different buffer. The sample is
concentrated and then repeatedly diluted with the
desired buffer and concentrated again.

34
As the results show in Tables 1 and 2, the Concentration of Indoleamine 2,3-Dioxygenase
efficient design of the Millipore devices allowed
Courtesy of Eduardo Vottero,
>90% of the salt to be removed during the first
University of British Columbia
centrifugation step. Typically, only one subsequent
Indoleamine 2,3-dioxygenase (IDO; MW 48,000)
centrifugation step was needed to increase the
is a heme-containing enzyme that is the first and
typical salt removal to 99% with >90% recovery
rate-limiting enzyme in human tryptophan metabo-
of the sample.
lism. IDO processes 98% of the total tryptophan
Protein purification by chromatography usually
available in the human body and is critical in
involves the collection of multiple column fractions,
suppression of immunoresponse by blocking
with only some of those fractions containing the
T-lymphocyte proliferation locally [Swanson et al,
protein of interest. After the fractions are combined, a
Am J Respir Cell Mol Biol [manuscript in prepara-
protein concentration step is often required for protein
tion] (2003); Sarkhosh et al, J Cell Biochem 90,
storage, or concentration with buffer ex-change may
206 (2003); Mellor et al, J Immunol 171, (2003)].
be needed for downstream separations.

Table 1. Removal of sodium chloride and recovery of protein with Amicon Ultra-15 and Ultra-4 devices
Cytochrome c Cytochrome c BSA BSA IgG
0.25 mg/mL 0.25 mg/mL 1 mg /mL 1 mg/mL 1 mg/mL
NMWL 5 kDa 10 kDa 30 kDa 50 kDa 100 kDa
% Protein % NaCl % Protein % NaCl % Protein % NaCl % Protein % NaCl % Protein % NaCl
Spin

P ro t o c o l s f o r P ro t e in s C o n ce n t r a t io n, D e s a l t i n g a n d Bu f f e r E xch a n g e
Recovery Removal Recovery Removal Recovery Removal Recovery Removal Recovery Removal
1 94.5 97.2 97.3 97.9 96.0 98.2 98.9 97.1 99.9 97.7
2 92.6 99.9 95.6 99.9 94.4 99.9 92.4 99.9 97.1 99.5
Three Amicon Ultra-15 devices of each cut-off were tested with 15 mL of solute. 500 mM NaCl was added to each solution. Each spin
was performed at 4000 x g for 30 minutes. After the first spin, the retentate was brought up to 15 mL with ultrapure water from a Milli-Q
(Millipore) system. OD readings were taken at 410 nm for Cytochrome c and 280 nm for BSA and IgG.

Table 2. Removal of riboflavin and


recovery of IgG with Microcon filter
device with Ultracel membrane
Spin % NaCI % IgG
Number Remaining Recovered
1 9 95
2 2 93
3 <1 93
4 <1 92
500 L of a 50:50 mixture of riboflavin and IgG were
spun in a Microcon 3K NMWL device at 12,000 x g
for 75 minutes at room temperature in 55 angle rotor.
After the initial spin, the retentate was twice diluted
with 500 L of PBS and spun again. After each spin,
concentration of riboflavin and IgG in the filtrate and
retentate were monitored.

35
Recombinant IDO was expressed in E. coli BL21 developed for PKR, and PKR has been purified using
(DE3) cells utilizing the pET 28a (+) vector system. three chromatography steps on Agarose-Heparin,
In this system, a hexahistidyl tag was fused to full- Agarose-Poly (I), Poly (C) and Sephacryl S-200 gel
size IDO at the N-terminus with a spacer sequence filtration columns. After the last step, PKR-containing
and a thrombin cleavage site. The protein was column fractions were pooled and concentrated
purified by conventional His-tag purification using Amicon Ultra-15 30K NMWL devices. The
methods and eluted with imidazole. The histidine concentration step was necessary for long-term
tag was removed by thrombin cleavage. Final protein storage. Table 3 shows the protein recovery
purification was done by gel filtration chromatogra- results obtained after four concentrations. Over 90%
phy G-75. Amicon Ultra-15 centrifugal devices recovery was obtained and no protein loss to the
were used to concentrate the IDO fractions from filtrate was observed.
an initial concentration of ~0.5 mg/mL to a Amicon Ultra-15 30K NMWL devices were also
final concentration of 10 mg/mL. Samples were used for exchanging buffer for PKR autophosphory-
analyzed by SDS-PAGE using a 12.5% polyacryl- lation (activation) assay. 200 L of 6.301 mg/mL
amide gel (Figure 1). In addition, it was shown that PKR in Protein Storage buffer (20 mM HEPES,
no IDO activity loss was observed after concentra- 1 M NaCl, 10 mM b-mercaptoethanol, 0.1 mM
tion using an Amicon Ultra device. EDTA, 10% glycerol, pH 7.5) was diluted to 15 mL
with Phosphorylation Buffer (20 mM HEPES,
Concentration of PKR and Buffer Exchange 50 mM KCl, 5 mM MgCl2, 0.1 mM EDTA, 1 mM
Courtesy of Peter A. Lemaire and DTT, pH 7.5) and re-concentrated three times using
Dr. James Cole, University of Connecticut
P ro t o c o l s f o r P ro t e in s C o n ce n t r a t io n, D e s a l t i n g a n d Bu f f e r E xch a n g e

Human protein kinase R (PKR) is one of the major


proteins induced by interferon as part of the host Figure 1.
defense against viral infection14. PKR is synthesized
in a latent form and is activated by autophosphory-
lation induced upon binding dsRNA. Once
phosphorylated, active PKR phosphorylates the
40 kDa
eukaryotic translation initiation factor elF2a leading
to a block in protein synthesis in virally infected cells.
35 kDa
PKR has been implicated as a participant in various
signal transduction pathways associated with cellular
processes including transcription79, differentiation10,
apoptosis11, splicing14 and transformation5,6.
However, difficulties in purifying PKR in large
SDS-PAGE of purified indoleamine 2,3-dioxygenase
amounts has limited rigorous biophysical character-
before and after concentration using Amicon Ultra-15
ization of the mechanisms of PKR activation. A high- centrifugal devices.
yield prokaryotic expression system has been

Table 3. PKR concentration results


Spin 1 Spin 2 Spin 3 Spin 4
Starting Volume (mL) 15 15 15 15
Starting Concentration (mg/mL) 0.229 0.229 0.169 0.169
Volume of Retentate (L) 590 490 203 225
Total Amount of PKR (mg)
Before UF 3.4350 3.4350 2.5320 2.5320
After UF 3.1005 3.1548 2.4081 2.3983
Filtrate 0.0000 0.0000 0.0000 0.0000
% Recovery 90.26 91.84 95.11 94.72

36
Amicon Ultra devices at 3000 x g for 20 minutes at
4 C. The filtrates from the three steps were pooled Figure 2.
and the total amount of protein in all samples was 100
determined by UV absorption A280.
80

Relative Activity of PKR


Protein activity was tested by autophosphoryla-
tion assay. The protein in the storage buffer was
60
supplemented with 5 mM MgCl2 and the samples
were allowed to undergo autophosphorylation at 40
30 C for 20 minutes in the presence of 3 mM ATP
20
and 3 Ci [g32P] ATP. The activity was determined
by autoradiography and quantified by liquid 0
scintillation counting. As shown in Figure 2, the No Buffer Buffer
Exchange Exchange
activity of PKR when no buffer exchange was only
Comparison of PKR autophosphorylation with buffer
6% of that when buffer exchange step was per-
exchange by ultrafiltration and without buffer
formed. Hence, the UF successfully exchanged the exchange.
buffer while maintaining the activity of the protein.

References
1. Samuel CE. Antiviral actions of interferon. 8. Cuddihy AR, et al. Double-stranded-RNA-
Interferon-regulated cellular proteins and activated protein kinase PKR enhances tran-
their surprisingly selective antiviral activities. scriptional activation by tumor suppressor p53.

P ro t o c o l s f o r P ro t e in s C o n ce n t r a t io n, D e s a l t i n g a n d Bu f f e r E xch a n g e
Virology 1991;183(1):111. Mol Cell Biol 1999;19(4):247584.
2. Hovanessian AG. The double stranded RNA- 9. Demarchi F, Gutierrez MI, Giacca M. Human
activated protein kinase induced by interferon: immunodeficiency virus type 1 Tat protein
dsRNA-PK. J Interferon Res 1989;9(6):641 7. activates transcription factor NF-kappaB
3. Lebleu B, et al. Interferon, double-stranded through the cellular interferon-inducible, double-
RNA, and protein phosphorylation. Proc Natl stranded RNA-dependent protein kinase, PKR.
Acad Sci USA 1976;73(9):310711. J Virol 1999; 73(8):70806.
4. Samuel CE. Mechanism of interferon action: 10. Petryshyn R, et al. Effect of interferon on protein
phosphorylation of protein synthesis initiation translation during growth stages of 3T3 cells.
factor eIF-2 in interferon-treated human cells by Arch Biochem Biophys 1996;326(2):2907.
a ribosome-associated kinase processing site 11. Barber GN. Host defense, viruses and apopto-
specificity similar to hemin-regulated rabbit sis. Cell Death Differ 2001;8(2):11326.
reticulocyte kinase. Proc Natl Acad Sci USA 12. Tan SL, Katze MG. The emerging role of the
1979;76(2):6004. interferon-induced PKR protein kinase as a
5. Koromilas AE, et al. Malignant transformation apoptotic effector: A new face of death?
by a mutant of the IFN-inducible dsRNA- J Interferon Cytokine Res 1999;19:54354.
dependent protein kinase. Science 1992; 13. Balachandran S, et al. Activation of the
257:16859. dsRNA-dependent protein kinase, PKR, induces
6. Meurs E, et al. Tumor supressor function of apoptosis through FADD-mediated death
interferon-induced double-stranded RNA signaling. EMBO J, 1998;17(23):6888902.
activated protein kinase. Proc Natl Acad 14. Osman F, et al. A cis-acting element in the 3-
Sci USA 1993;90:2326. untranslated region of human TNF-alpha mRNA
7. Wong AH, et al. Physical association between renders splicing dependent on the activation of
STAT1 and the interferon-inducible protein protein kinase PKR. Genes Dev 1999;13(24):
kinase PKR and implications for interferon and 328093.
double-stranded RNA signaling pathways.
EMBO J, 1997;16(6):1291304.

37
Detergent Removal with Microcon
and Amicon Ultra Centrifugal Filters
Introduction Method and Results
Microcon and Amicon Ultra centrifugal filters are In a series of studies, Millipore researchers used
efficient laboratory tools for removing small mol- Total Organic Carbon (TOC) analysis to measure
ecules from solutions of proteins or nucleic acids. detergent removal using either Microcon or Amicon
Often, the molecule to be removed is one of a Ultra concentrators after a single centrifugation spin
number of commonly used detergents or protein (note that complete detergent removal generally
solubilizing agents. The chemical nature of most requires 35 spins). As the results in the Tables 1
detergents allows for micelle formation above a and 2 indicate, detergent removal depends both
critical concentration limit (Critical Micelle on the original detergent concentration and
Concentration, CMC). Micelle formation results in the NMWL of the centrifugal units. All measure-
aggregation of the detergent and leads to gross ments shown were made using detergent/
changes in molecular structure. This affects the distilled water solutions.
amount of the detergent that can be removed from
NOTE: Temperature, the presence of salts in
a solution by centrifugal devices with specific
the solution, and/or macromolecule/detergent
nominal molecular weight limit (NMWL)
interactions may lower the CMC for a particular
membranes.
detergent. Use the tables only as general guidelines
For example, the monomer of Triton X-100
in assessing the efficiency of detergent removal with
has a molecular weight of 500650 daltons.
the Microcon and Amicon Ultra devices.
Triton X-100 should pass readily through the
10,000 NMWL membrane in an Amicon Ultra
device. However, at concentrations above 0.01%
(0.2 mM), Triton X-100 forms micelles composed
of approximately 140 monomeric units. During
P ro t o c o l s f o r P ro t e in s D ete r g e n t Re m ova l

ultrafiltration, the micelles behave like 70,000


90,000 dalton globular proteins. As a result,
more than 90% of Triton is retained by the ultra-
filtration membrane. Therefore, above the CMC
of Triton X-100, an Amicon Ultra-4 100K NMWL
concentrator would be required to remove the
detergent effectively.

38
Table 1. Percent detergent removal after one spin with Table 2. Percent detergent removal
Microcon centrifugal devices after one spin with Amicon Ultra-4
centrifugal devices
NMWL
Detergent (%) 3 kDa 10 kDa 30 kDa 50 kDa 100 kDa NMWL
SDS Detergent (%) 10 kDa 30 kDa
0.001 >90% >90% >90% >90% >90% SDS
0.1 >90% >90% >90% >90% >90% 0.1 74% 75%
1 4089% 4089% 4089% 4089% 4089% 1 13% 11%
5 <40% <40% <40% <40% <40% 5 0.5% 2.0%
NaDeoxycholate Tween-20
0.1 >90% >90% >90% >90% >90% 0.1 47% 69%
1 >90% >90% >90% >90% >90% 1 36% 42%
5 4089% 4089% 4089% >90% >90% 5 39% 47%
CAPS Triton X-100
5 >90% >90% >90% >90% >90% 0.10 40% 70%
CPCl 1 1 26% 37%
0.01 >90% >90% >90% >90% >90% 5 37% 39%
0.1 4089% 4089% 4089% 4089% 4089% CHAPS
1 <40% <40% <40% <40% <40% 0.1 96% 81%
5 <40% <40% <40% <40% <40% 1 62% 96%
TDMABr2 5 36% 80%
0.1 >90% >90% >90% >90% >90%
1 <40% <40% <40% 4089% <90%
5 <40% <40% <40% <40% <90%
Digitonin
0.01 >90% >90% >90% >90% >90%
0.1 4089% 4089% 4089% 4089% 4089%
1 <40% <40% <40% <40% <40%
Tween-20
0.01 <40% <40% 4089% 4089% >90%
0.1 <40% <40% <40% 4089% >90%

P ro t o c o l s f o r P ro t e in s D ete r g e n t Re m ova l
1 <40% <40% <40% <40% 4089%
5 <40% <40% <40% <40% <40%
Triton X-100
0.01 4089% 4089% 4089% 4089% >90%
0.1 <40% <40% <40% <40% >90%
1 <40% <40% <40% <40% 4089%
5 <40% <40% <40% <40% <40%
CHAPS
0.1 >90% >90% >90% >90% >90%
1 4089% 4089% >90% >90% >90%
5 <40% <40% 4089% >90% >90%
1
Cetylpyridinium chloride
2
Tetradecyltrimethylammonium bromide

39
Two-Dimensional Electrophoresis
Sample Preparation with
Amicon Ultra Centrifugal Filters
Two-dimensional electrophoresis (2DE) is one of the Usually protein concentration is achieved by
most commonly used methods in proteome analysis. protein precipitation with acetone or TCA. The
Briefly, proteins are separated by their isoelectric disadvantage of protein precipitation is that some
point (first dimension separation) followed by SDS- of the proteins become insoluble and can not be
PAGE separation by molecular weight (second resolubilized in IPG buffer. Another disadvantage is
dimension separation). Although it is a powerful that many salts become insoluble in acetone and
method for simultaneously displaying hundreds precipitate along with the proteins.
of proteins, 2DE presents a challenge for sample Ultrafiltration (UF) can achieve protein concentra-
preparation. Salts and ionic detergents are common tion and desalting in one step. Figures 1 and 2
chemicals contaminating biological samples that present two examples of protein preparation for
are not compatible with 2DE separation; however 2DE by acetone precipitation and ultrafiltration.
P ro t o c o l s f o r P ro t e in s Tw o - D i m e n s io n a l E l e c t ro p h o re s i s S a m p l e P re p a r a t io n

they are often required to solubilize proteins from Both examples demonstrate that UF provides more
cells and tissues. The high concentration of salts efficient salt removal and allows better separations
combined with the relatively low protein content and improved resolution of protein spots in
make samples completely unsuitable for isoelectric two-dimensional electrophoresis.
focusing.

Figure 1.

Two-dimensional electrophoresis of yeast cell lysate prepared by acetone precipitation (left) and ultrafiltration (right).
Sacharomyces cerevisiae strain s288c was grown to log phase. The cells were pelleted and resuspended in Cellular
and Organelle Membrane Solubilizing Reagent from the ProteoPrep kit (Sigma) and lysed by sonication. Cellular
debris was removed by centrifugation and the supernatants were reduced and alkylated with 5 mM tributylphosphine
and 10 mM acrylamide. Lysates were acetone-precipitated to remove residual Tris and alkylating reagent or were
filtered through Amicon Ultra-4 10K NMWL devices. Samples were redissolved in ProteomIQ Resuspension Reagent
(Proteome Systems), focused in broad range (pH 310) immobilized pH gradients (IPGs) on the IsoelectrIQ IEF
instrument. Second dimension gels were 615% polyacrylamide gradient GelChips (Proteome Systems) run on
an ElectrophoretIQ 2D instrument (Proteome Systems). Data courtesy of Dr. G. Smejkal, Proteome Systems, Inc.,
Woburn, MA, USA.

40
Figure 2.

P ro t o c o l s f o r P ro t e in s Tw o - D i m e n s io n a l E l e c t ro p h o re s i s S a m p l e P re p a r a t io n
Two-dimensional electrophoresis of endothelial cell lysates prepared by acetone precipitation (left) and ultrafiltration
with Amicon Ultra devices (right). Whole cell lysates of endothelial cells were prepared in 7 M urea, 2 M thiourea,
4% CHAPS, 10 mM DTT, 20 mM Tris buffer. The samples were very dilute and presumably contained DNA fragments,
salts and lipids. 300 mg of the total protein was either acetone-precipitated or desalted in Amicon Ultra devices.
After acetone precipitation with four volumes of cold acetone, samples were incubated overnight at 20 C,
precipitated by centrifugation, rinsed with cold acetone, and pelleted again. The resulting pellet was resuspended
in rehydration buffer (7 M urea, 2 M thiourea, 4% CHAPS, 10 mM DTT, 20 mM Tris). Alternatively, samples were
filtered through Amicon Ultra-4 10K NMWL devices after being mixed with nine volumes of 20 mM Tris HCl, pH 7.
The sample was concentrated to approximately 40 L and adjusted to 350 L with rehydration buffer. The proteins
were separated by IEF in 18 cm IPG Drystrips, pH 310 (GE Healthcare). A second dimension separation was
performed in 10% SDS-PAGE gel. The gels were silver stained and scanned. Data courtesy of Dr. Leonid Kryazhev,
Genome Quebec, Montreal, Canada.

41
Purification of Serum Peptides for
Biomarker Research with Amicon Ultra
Centrifugal Filters
Introduction Methods
A biomarker can be defined as a molecule that Preparation of Serum Peptides by UF and SPE
indicates an alteration in physiology. Biomarkers One milliliter of human serum, with or without
play an essential role in the drug discovery and acetonitrile, was filtered using Amicon Ultra-4
development process. They provide powerful clues 10K NMWL centrifugal devices. The devices were
to genetic susceptibility, disease progression, and centrifuged in a swinging bucket rotor for 15 to
predisposition, as well as drug response and the 30 minutes at 3000 x g. Ten microliters of the
physiological and metabolic profiles of diseases filtrate was acidified with 5 L of 1% TFA, concen-
and drug responses. Biomarkers can also provide trated with ZipTipC18 pipette tips. Co-elution was
valuable diagnostic and prognostic information that
P ro t o c o l s f o r P ro t e in s P u rif i c a t io n of S e ru m Pe p t i d e s f o r Bio m a rke r Re s e a rch

performed directly onto a MALDI target with 2 L of


can facilitate personalized medicine. Peptide and -cyano-4-hydroxycinnamic acid matrix (5 mg/mL
protein patterns have been linked to ovarian cancer, in 50% acetonitrile, 0.1% TFA). If acetonitrile was
breast cancer, prostate cancer and astrocytoma15. added to the serum prior to the filtration, the samples
Most diagnostic tests are based on blood or were briefly evaporated in a Speed Vac centrifuge
urine analysis5. Serum is a key source of putative to remove solvent before ZipTip purification.
protein biomarkers, and, by its nature, can elucidate
organ-confined events. Mass spectrometry, coupled Peptide Analysis by Mass Spectrometry
with bioinformatics, is capable of distinguishing Peptide-containing ultrafiltrates from cell lysates
between serum protein pattern signatures in late- or human serum were acidified with 1% TFA and
stage and early-stage ovarian cancer patients6. concentrated on ZipTipC18 or ZipTipSCX pipette tips
One of the major impediments to the discovery of following the procedure outlined in the user guide.
new biomarkers is the presence of salts, proteins, All samples were overlaid with 1 L of -cyano-
and lipids in plasma or serum that makes it difficult to 4-hydroxycinnamic acid matrix (5 mg/mL in
detect and analyze peptides by mass spectrometry. 50% acetonitrile, 0.1% TFA) and analyzed on
When untreated serum is spotted onto a MALDI-TOF Voyager-DE Workstation (Applied Biosystems)
plate, it does not produce any usable signal in mass in linear mode.
spectrometry. Multiple protocols have been devel-
Preparation of Serum Peptides by
oped to extract and enrich peptides from tissues and
Acetonitrile Precipitation
body fluids, such as batch reversed phase chroma-
tography over C18 resin and extraction with 0.1% Acetonitrile was added to human serum at a
trifluoroacetic acid (TFA) or 50% acetonitrile to 1:1 ratio (v:v), and samples were centrifuged to
selectively precipitate large proteins while enhancing precipitate larger proteins. The supernatant was
the solubility of smaller proteins and peptides. dried in a Speed-Vac centrifuge, resuspended in
Ultrafiltration has previously been reported as a 0.1% TFA, and then desalted and concentrated with
sample preparation tool to prepare low molecular ZipTipC18 pipette tips. Co-elution was performed
weight fractions for biomarker analysis710. In this directly onto a MALDI target with 2 L of -cyano-
study we show that ultrafiltration in combination with 4-hydroxycinnamic acid matrix (5 mg/mL in
solid phase extraction (SPE) on C18 resin can be a 50% acetonitrile, 0.1% TFA).
convenient and efficient method for serum peptide
purification. The approach provides more peptides
for mass spectrometry analysis than the acetonitrile
precipitation method.
42
Results One of the common methods for serum peptide
preparation for mass spectrometry is acetonitrile
While human serum contains numerous peptides
fractionation, where the addition of 5070%
and small proteins, they are not accessible by direct
acetonitrile precipitates larger proteins, while the
mass spectrometry analysis. Even after reverse
peptides stay soluble in the supernatant. Another
phase concentration and desalting, only a few
way to produce relatively protein-free filtrates is
peptides are detectable in the mass spectrum
ultrafiltration. Figure 2 presents the MALDI-TOF
(data not shown). This can be explained by the
spectra of (A) straight rat serum; (B) serum superna-
high concentration of proteins and lipids competing
tant after 50% acetonitrile precipitation; and (C)
with the peptides to bind to the resin (Figure 1A).

Figure 1.

A B C

P ro t o c o l s f o r P ro t e in s P u rif i c a t io n of S e ru m Pe p t i d e s f o r Bio m a rke r Re s e a rch


MALDI-TOF spectra of (A) unprocessed rat serum; (B) serum peptides in 50% acetonitrile supernatant; and (C) serum ultrafiltrate
processed with Amicon Ultra-4 30K NMWL centrifugal device.

Figure 2.

A B

C D

MALDI-TOF spectra of rat serum peptides after concentration and desalting by reversed
phase chromatography: (A) rat serum processed with ZipTipC18 pipette tip; (B) serum
supernatant after 50% acetonitrile precipitation processed with ZipTipC18 pipette tip;
(C) 30K serum ultrafiltrate processed with ZipTipC18 pipette tip; and (D) the same as (C)
but 20% acetonitrile was added to the serum prior to ultrafiltration.

43
serum ultrafiltrate processed with an Amicon Ultra-4 References
30K NMWL device. We observed higher quality
1. Ardekani AM, Liotta LA, Petricoin III. Expert Rev
spectra and an increased number of MALDI-TOF
Mol Diagn 2002;2:12.
detected peptides if the serum was ultrafiltered.
Further improvement of spectra can be achieved 2. Carter D, Douglass JF, Cornellison CD, Retter
by reverse phase chromatography, which concen- MW, Johnson JC, Bennington AA, Fleming TP,
trates and desalts the peptides. The ZipTipC18 Reed SG, Houghton RL, Diamond TS, Vedvick
pipette tip is a convenient and efficient tool for TS. Biochemistry 2002;41:6714.
micro-scale sample preparation prior to mass 3. Wellmann A, Wollscheid V, Lu H, Ma ZL,
spectrometry. Figure 2 shows the MALDI-TOF Albers P, Schutze K, Rohde V, Behrens P,
spectra of rat serum peptides prepared with Dreschers S, Ko Y, Wernert N. Int J Mol Med
ZipTipC18 pipette tip out of (A) straight serum, 2002;9:341.
(B) 50% acetonitrile supernatant and (C) serum 4. Petricoin EF, Ardekani AM, Hitt BA, Levine PJ,
processed by ultrafiltration. The last method Fusaro VA, Steiberg SM, Mills GB, Simone C,
provided stronger MALDI-TOF signal, higher Fishman DA, Kohn EC, Liotta LA. Lancet 2002;
signal-to-noise ratio and double the number of 359:572.
detected peptides. 5. Bischoff R, Luider TM. J Chrom B 2004;803:
The addition of 5 to 10 % acetonitrile to serum 2740.
P ro t o c o l s f o r P ro t e in s P u rif i c a t io n of S e ru m Pe p t i d e s f o r Bio m a rke r Re s e a rch

and plasma samples (data not shown) prior to 6. Stevens EV, Liotta LA, Kohn EC. J Gynecol
ultrafiltration was shown to improve the detection Cancer 2003;13:1339.
of serum peptides, the MALDI spectrum and the
7. Schulz-Knappe P, Schrader M, Standker L,
overall signal intensity.
Richter R, Hess R, Jurgens M, Forssmann W-G.
J Chromatogr A 1997;776:125132.
Conclusion
8. Basso D, Valerio A, Seraglia R, Mazzza S,
For the analysis of serum peptides, complexity
Piva MG, Greco E, Fogar P, Gall N,
reduction by eliminating higher molecular weight
Pedrazzoli S, Tiengo A, and Plebani M.
proteins is critical for high resolution mass spec-
Pancreas 2002;24:814.
trometry. Efficient separation of peptides from the
9. Prazeres S, Santos MA, Ferreira HG, Sobrinho
majority of proteins and salts can be achieved by
LG. Clin Endocrinol (Oxf) 2003;58:68690.
sample ultrafiltration. We have shown the effective
use of Amicon Ultra-4 30K NMWL centrifugal 10. Tirumalai RS, Chan KC, Prieto DA, Issaq HJ,
devices for the preparation of peptides. Other Conrads TP, Veenstra TD, Mol Cell Proteomics
molecular weight cut-offs can be utilized depending 2003;10:1096103.
on the desired range of peptides. The method can 11. Chernokalskaya E, Gutierrez S, Pitt AM,
be used directly in combination with ZipTipC18 Leonard J. Electrophoresis 2004;25:
pipette tips for peptide identification by MS/MS 24612468.
or as a first step prior to further surface-mediated 12. Millipore Case Study PR0001EN00.
enrichment using SELDI-TOF methods. These sample Automating Multiplexed Cytokine Assays
preparation protocols may also be applicable to manuscript in preparation.
other low molecular weight markers such as drugs
and metabolites. Peptide purification using ultrafiltra-
tion to de-proteinize followed by SPE to de-salt
serum and plasma samples (data not shown) was
easily adapted to the 96-well format allowing large
numbers of samples to be simultaneously prepared.

44
Rapid Antibody Concentration
with Amicon Ultra Centrifugal Filters
Ultrafiltration offers a fast and convenient way to
concentrate antibodies purified from serum, ascites Figure 1.
fluid, or hybridoma supernatants. The traditional 40 12
purification protocol for immunoglobulins includes

IgG Concentration (mg/mL)


PROSEP-A
10
affinity binding to Protein A or G chromatography 30 PROSEP-G

IgG Volume (mL)


media, washing unbound proteins, and eluting with 8

a low pH buffer. Subsequently, purified antibodies 20 6


are often too dilute for their intended purpose or for
4
long-term storage. In addition, harsh elution 10
conditions sometimes require buffer exchange to 2
preserve protein activity. 0 0
Dialysis is often used for antibody concentration 0 3 10 15 20

and buffer exchange. However, ultrafiltration Centrifugation Time (min)


provides a quick, alternative method to concentrate Concentration of rabbit IgG with Amicon Ultra-15 devices. The IgGs were
and diafilter immunoglobulins with up to 99% purified using Montage PROSEP-A or PROSEP-G Antibody Purification Kits.
The lines show IgG volume reduction, while the bars show a proportional
recovery and one-step salt removal (see
increase in IgG concentration after 20 minutes of centrifugation time.
Concentration, Desalting, and Buffer Exchange with
Amicon Ultra or Microcon Centrifugal Filters, page
34).
To demonstrate the suitability of Amicon Ultra Figure 2.

P ro t o c o l s f o r P ro t e in s Ra p i d A n t i b o d y C o n ce n t r a t io n
devices for concentrating purified IgG, rabbit serum 1 2 3 4 5
SDS-PAGE gel of purified kDa
was processed with Montage Purification Kits with
rabbit IgG before and 116
PROSEP-A and PROSEP-G Antibody (Millipore) after concentration with
and then concentrated with Amicon Ultra-15 30K Amicon Ultra-15 devices.
66
NMWL devices. Figure 1 shows a decrease in the Lane 1: MW standards 55
Heavy
retentate volume proportional to the increase in Lanes 2, 3: PROSEP-A-purified IgG Chain

antibody concentration. A twenty-minute centrifuga- before concentration 36


(lane 2) and after
tion resulted in >95% recovery of immunoglobulins. concentration (lane 3) Light
21 Chain
Figure 2 shows an SDS-PAGE gel of purified rabbit Lanes 4, 5: PROSEP-G-purified IgG
before concentration 14
IgGs before and after ultrafiltration.
(lane 4) and after
concentration (lane 5)
Protein Load: 5 L in lanes 2 and 4;
1 L in lanes 3 and 5

45
Affinity Purification with
Ultrafree-MC Centrifugal Filters
Introduction
Affinity interaction chromatography is often the
single most effective step in any protein purification
procedure. Up to 95% purity can be achieved in
one step, depending on the nature of the interaction
and the starting composition of the protein solution.
Well known examples of highly specific affinity
interactions include antibodies and protein A/G;
multiple histidine tags and nickel; streptavidin and
biotin; antibodies and antigens; and many others.
Ultrafree-MC centrifugal filter unit
Less specific interactions are also used for enrich-
ment or depletion protocols, including albumin
depletion on cibacron blue resin, glycoprotein Another alternative for small-scale purification
enrichment on concavalin A resin, and capture are centrifugal devices with microporous membrane,
of nucleic acid-binding proteins on heparin resin. such as Ultrafree-MC centrifugal devices. The
Affinity chromatography is often employed in sample can be added to the filter basket and mixed
the small-scale batch mode as a quick method for for the needed residence time and then centrifuged.
microgram-scale protein purification. The typical The process removes the interstitial liquid but does
protocol involves: not dehydrate the beads. Washing and elution
1. Pipetting a small volume of affinity resin into can also be performed in a similar manner and
a microfuge tube that contains the sample are more effective due to the efficient removal of
2. Vortexing the tube for a few minutes buffer and/or eluant.
Ultrafree-MC centrifugal filter units with micropo-
P ro t o c o l s f o r P ro t e in s A f f i n i t y P u rif ic a t io n

3. Centrifuging the resin to the bottom


rous membrane come with low protein-binding
4. Pipetting off the supernatant
Durapore PVDF membrane in five different pore
5. Washing a few times (using steps 2 and 3) sizes from 0.1 to 5.0 m. Affinity resin can be
6. Eluting with a small amount of eluant loaded into the filter basket and the device used
Although the method is relatively simple, care as a home-made mini-spin column.
must be taken not to remove the chromatography We show the applicability of the device for
resin when pipetting off the supernatant. purification of rabbit IgG on PROSEP-A resin and
Pre-packed mini-spin columns are a convenient His-tagged C-RP protein on three different commer-
tool for small-scale protein purification. Operated cial metal-chelate resins.
by centrifugation, they usually complete the whole
procedure in less than an hour.

46
Materials Method for IgG Purification
Ultrafree-MC 0.45 m centrifugal devices Solutions
(Millipore cat. no. UFC3 0HV 00) PROSEP-A binding buffer A: 1.5 M Glycine/
PROSEP-A high capacity resin NaOH, 3 M NaCl, pH 9.0
(Millipore cat. no. 1131 118 26) PROSEP-A elution buffer B2: 0.2 M Glycine/
Rabbit serum Gibco (Invitrogen lot no. HCl, pH 2.5
1132782) PROSEP-A neutralization buffer: 1 M Tris/HCl,
Micro-centrifuge Biofuge Pico pH 9.0
(Heraeus instruments)
Procedure
Jouan CR1822 fixed angle rotor centrifuge
1. 200 mg of PROSEP-A media were placed in
Xcell SureLock Mini-cell vertical electrophoresis Ultrafree-MC 0.45 m filter basket.
system (Invitrogen cat. no. EI0001)
2. The columns were equilibrated with 400 L of
NuPage NOVEX Bis-Tris 4 12%, 1 mm thick, binding buffer A and centrifuged for 1 minute
15 well SDS gels, (Invitrogen cat. no. NP0323) at 100 x g.
NuPage Sample Reducing agent (10X) 3. 200 L of rabbit serum were diluted 1:1 with
(Invitrogen cat. no. NP009) binding buffer and the entire volume was loaded
NuPage SDS Sample Buffer (4X) into the spin column containing PROSEP-A resin.
(Invitrogen cat. no. NP007) 4. Devices were placed on a shaker for 15 minutes
at room temperature and centrifuged at 100 x g
SimplyBlue SafeStain Coomassie G-250 stain
for 5 minutes. Flow-through was collected for
(Invitrogen cat. no. LC6060)
future analysis.
5. Three consecutive washes of 400 L each were
performed by adding 400 L of binding buffer A
and centrifuging at 2,000 x g for 2 minutes each.
6. 200 L of elution buffer B2 were added and
centrifuged for 2 minutes at 2,000 x g.

P ro t o c o l s f o r P ro t e in s A f f i n i t y P u rif ic a t io n
7. 26 L of neutralization buffer were added to
each collection tube. A second elution was
collected after repeating the same process one
more time.

47
Method for His-tagged C-RP Purification 5. 500 L of the clarified lysate were added to
the resin.
Solutions
6. The His-tagged proteins were bound for
Lysis buffer: 50 mM sodium phosphate, 300 mM 30 minutes with light agitation.
sodium chloride, 10 mM immidazole, pH 7 7. The lysate was removed by centrifugation at
Binding buffer: 50 mM sodium phosphate, 500 x g for 10 minutes.
300 mM sodium chloride, 10 mM immidazole, 8. The resin was washed with 500 L of wash
pH 7 buffer for 5 minutes with agitation. The wash
Wash buffer: 50 mM sodium phosphate, 300 mM solution was removed by centrifugation for
sodium chloride, 20 mM immidazole, pH 7 5 minutes at 500 x g. This step was repeated
two more times.
Elution buffer: 50 mM sodium phosphate,
300 mM sodium chloride, 250 mM immidazole, 9. 250 L of elution buffer were added to the
pH 7 Ultrafree-MC device and mixed for 5 minutes.
Purified protein was recovered by centrifugation
1 mg/mL Lysozyme stock
at 500 x g for 1 minute.
Benzonase
Results
Procedure
The results of purifying rabbit IgG using Ultrafree-
1. Recombinant proteins were expressed in
MC centrifugal devices are shown in Figure 1.
Escherichia coli.
The device was challenged with approximately
2. Cells were prepared at a 10X concentration 14 mg of total protein, with an estimated IgG
using lysis buffer. Lysozyme was added to a content of 1.52 mg. The original serum, flow-
concentration of 0.1 mg/mL. To reduce the through, three washes and two eluted fractions
viscosity, benzonase was added to the lysate. were analyzed by SDS-PAGE. The total amount of
The lysates were clarified by centrifugation. purified IgG, as estimated by OD280, was 1.2 mg
3. 200 L of the 50% resin slurry were added to and 1.1 mg on two devices processed in parallel.
the Ultrafree-MC device and the residual fluid The whole procedure was completed in less than
was removed by centrifugation for 1 minute at one hour. This method can be useful for monitoring
P ro t o c o l s f o r P ro t e in s A f f i n i t y P u rif ic a t io n

500 x g. the titer of antigen-specific antibodies after immune


4. The resin was equilibrated with 500 L of activation, or whenever small amounts of IgG need
binding buffer and centrifuged for 2 minutes to be purified.
at 500 x g.

48
Figure 2 shows the results of His-tagged protein
purification in Ultrafree-MC devices using Ni-NTA Figure 1.
1 2 3 4 5 6 7 8
agarose and BD Talon resin. Two recombinant Rabbit IgG purification kDa
200
proteins were purified: C-RP, high-expressing protein on PROSEP-A resin in
of 26 kDa; and RT66, low-expressing protein of Ultrafree-MC devices. 116.3
97.4
66 kDa. Both purifications resulted in high purity Lane 1: Molecular 66.3
weight 55.4
proteins. The amount of proteins purified out of standards
500 L of lysate was 3235 g for C-RP protein Lane 2: Rabbit serum 36.5
and 8 g for RT66. Lane 3: Flow through 31.0

The data show that affinity batch purification Lanes 46: Three consec- 21.5
can be effectively performed on a small scale using utive washes
14.4
Lanes 7, 8: Eluted IgG
Ultrafree-MC devices loaded with resin. The
from two
6.0
process combines the high efficiency of batch devices
binding and washing with the handling conve-
nience of a mini-spin column. With minimal hands-
on time, the method provides flexibility of resin to
lysate ratio and binding conditions, independent of Figure 2.
centrifugation speed and rotor angle. This method 1 2 3 4 5 6 7
His-tagged protein kDa
200
is applicable to recombinant protein purification, purification using
antibody purification and immunoprecipitation. Ultrafree-MC devices. 116.3
97.4
Lane 1: Molecular 66.3 RT66
weight 55.4
standards
Lane 2: E. coli lysate 36.5
expressing 31.0
C-RP
C-RP protein
21.5
Lane 3: E. coli lysate
expressing 14.4
RT66 protein
Lanes 4, 5: Proteins 6.0
purified on

P ro t o c o l s f o r P ro t e in s A f f i n i t y P u rif ic a t io n
Ni-NTA resin
Lanes 6, 7: Proteins purified
on BD Talon resin

49
Removal of Unincorporated Label
from Labeled Protein with Amicon Ultra
Centrifugal Filters
Introduction Results
Centrifugal devices containing ultrafiltration mem- Figure 1 shows the SDS-PAGE gel of FITC-labeled
branes are ideal for the removal or exchange of BSA before and after each of four diafiltration cycles.
salts, sugars, nucleotides, non-aqueous solvents, Unincorporated FITC is clearly visible in the starting
and other materials of low molecular weight. They material and in the first filtrate. After only one cycle
also serve to separate free from bound species. of ultrafiltration, the majority of the free label is
For the removal of unincorporated label in protein removed. Subsequent cycles of filtration result in
labeling applications, Millipore centrifugal devices BSA that is virtually free of unincorporated FITC.
provide fast, convenient, high-recovery alternatives Fluorescence measurement offers a more sensitive
to gel filtration. Sample dilution, often associated method to monitor the ultrafiltration process. Figure
P ro t o c o l s f o r P ro t e in s Re m ova l of Un i n co rp o r a te d L a b e l f ro m L a b e l e d P ro te i n

with gel filtration, is not a problem. A few rounds of 2 shows the change in fluorescence of filtrate and
diafiltration can efficiently remove unincorporated retentate during four cycles of FITC-BSA diafiltration.
label. Two examples below demonstrate the use The results indicate that almost 80% of free FITC can
of Millipore centrifugal ultrafiltration devices for be removed after the first ultrafiltration and three
removal of unreacted fluorescent and isotope labels. rounds result in 98% removal. This demonstrates
the viability of ultrafiltration as an alternative to gel
Method for Removal of FITC from FITC-BSA filtration for cleaning up protein labeling reactions.
1. Bovine serum albumin was labeled with Using an Amicon Ultra-4 device, each ultrafiltration
fluorescein-isothiocyanate (FITC); the free is accomplished in 1015 minutes and allows high
unincorporated label was not removed. recovery of target protein. While gel filtration results
in diluted protein fractions, ultrafiltration offers the
2. Two mL of FITC-labeled BSA solution at
additional advantage of concentrating protein while
0.5 mg/mL were loaded into two Amicon
removing unincorporated label.
Ultra-4 10K NMWL devices and centrifuged
at 3,000 x g for 10 minutes.
3. Retentates (about 50 L each) were re-diluted
to 2 mL with water and centrifuged again.
This step was repeated twice.
4. After each ultrafiltration, the retentate and
filtrate were analyzed by SDS-PAGE and their
fluorescence measured on a SpectraFLUOR
plate reader (Tecan) at excitation 485 nm and
emission 530 nm.
5. The SDS PAGE gel was scanned on a Storm
(GE Healthcare) fluorescence scanner.

50
Figure 1. Figure 2.
SDS-PAGE of FITC-labeled BSA 60,000

Lane 1: Starting material 50,000 Retentate


Lane 2: Retentate after first ultrafiltration Filtrate

(Aribitrary Units)
40,000

Fluorescence
Lane 3: Retentate after 2 rounds of ultrafiltration
Lane 4: Retentate after 3 rounds of ultrafiltration 30,000
Lane 5: Retentate after 4 rounds of ultrafiltration
20,000
Lane 6: Filtrate after first ultrafiltration
Lane 7: Filtrate after 2 rounds of ultrafiltration 10,000
Lane 8: Filtrate after 3 rounds of ultrafiltration
0
Starting Spin 1 Spin 2 Spin 3 Spin 4
material
Centrifugation Time (min)
FITC-BSA
Fluorescence measurements of the filtrates and retentates after each of four
cycles of FITC-labeled BSA ultrafiltration. Free label was transferred through
the 10,000 NMWL membrane while labeled BSA was retained and
concentrated. All retentates and filtrates were volume adjusted to 2 mL
prior to the measurement.

P ro t o c o l s f o r P ro t e in s Re m ova l of Un i n co rp o r a te d L a b e l f ro m L a b e l e d P ro te i n
FITC

1 2 3 4 5 6 7 8

51
Rapid, Ultrafiltration-based Method for
Purification of Monoclonal Antibodies
Introduction desalted and exchanged into a biological buffer
using dialysis. The entire process typically requires
Monoclonal antibodies (MAb) continue to gain
several days to complete and can be particularly
importance as therapeutic and diagnostic agents
onerous if multiple MAbs are to be evaluated in
for many types of cancer. The process of screening
parallel. We describe here a new and simplified
hybridoma libraries for candidate MAbs is both time
method that minimizes the processing time to less
consuming and labor intensive. Once a hybridoma
than a day to obtain pure MAb.
cell line expressing a suitable MAb is established,
a bench-scale purification methodology (e.g., 50
Method
to 500 mL) must be developed to produce sufficient
The method involves clarification of the hybridoma
MAb for further characterization.
supernatant by microfiltration using a Stericup
A traditional method for purifying MAbs involves
vacuum filter cup, followed by concentration using
clarification of the hybridoma supernatant by
ultrafiltration1 with a Centricon Plus-70 device
centrifugation, followed by an ammonium sulfate
(see Table 1 and Figure 1 for method details).
precipitation to concentrate the MAbs. The precipi-
The MAb is further purified on protein A/G beads.
tate is then recovered by centrifugation, resolubi-
The purified MAb is desalted and buffer-exchanged
P ro t o c o l s f o r P ro t e in s Ra p i d P u rif ic a t io n of M o n o cl o n a l A n t i b o d ie s

lized and desalted using dialysis. After these steps,


using ultrafiltration.
the MAb is further purified using Protein A/G
affinity chromatography. The purified antibody is

Table 1. Comparison of traditional and ultrafiltration workflows

Workflow for Obtaining Concentrated Supernatant


Traditional Ultrafiltration
Ammonium Sulfate Precipitation Centrifugal Tangential Flow Filtration (TFF)
1. Start with 200 mL 1. Start with 200 mL 1. Start with 1000 mL
2. Weigh out ammonium sulfate 2. Add supernatant to 2. Add supernatant to Labscale
3. Add ammonium sulfate to Centricon Plus-70 Device TFF unit (30K MWCO)
clarified supernatant with (100K MWCO) 3. Apply pressure (40 psi) with
constant stirring (20-30 min). 3. Centrifuge (2030 min) constant stirring (~2 hrs)
Store at 4 C. 4. Invert spin to collect sample 4. Collect sample (5 min)
4. Centrifuge to recover pellet, (2 min)
resuspend pellet 5. Load on protein-G column
5. Prepare dialysis membrane
and check for integrity
6. Collect resuspended
precipitate in dialysis
tubing/device
7. Dialyze with three changes
of PBS (18-24 hrs)
8. Collect dialysate in
centrifuge tube(s) and
centrifuge (~30 min)
9. Load on protein-G column
Total time required ~24 hrs Total time required ~40 min Total time required ~2 hrs

52
Results
Figure 2.
This approach was successfully used to purify an
2 Abalone Commercial
anti c-myc antibody secreted by the hybridoma
clone 9E102 (Figure 1). The MAb purified by this
new method performed comparably to the commer-
cially purified MAb in downstream applications
such as western blotting and ELISA (Figure 2). The
data demonstrate that the new protocol is robust
and delivers MAb of a high purity and yield as
compared to the traditionally purified MAb. The Traditional UF-based

application of ultrafiltration to MAb purification


will be of considerable value to any researcher
interested in screening hybridoma libraries and
accelerating the purification of MAbs.

Figure 1. 0.7
C Tr UF
C: Clarified supernatant UF-based method
0.6
Tr: Supernatant precipitated Traditional method
OD @ 450 mm

0.5
with ammonium sulfate and Commercially available
dialyzed using 10K MWCO 0.4

P ro t o c o l s f o r P ro t e in s Ra p i d P u rif ic a t io n of M o n o cl o n a l A n t i b o d ie s
membrane (Spectrapor,
Rancho Dominguez, CA) 0.3
UF: Supernatant concentrated 0.2
by ultrafiltration on
Centricon Plus 70 device 0.1

0.0
0 10 100

Concentration of mAB (g/mL)

Upper panel shows activity comparison measured in a western blot


between MAb (derived from HEK 293) purified using ultrafiltration method
versus traditional method. Three-fold serial dilutions of HEK 293 cell
nuclear extracts were run on 4 to 12% NuPAGE gels and transferred to
Immobilon-P membrane (Millipore). Blots were probed with the indicated
Conclusion primary antibodies and secondary anti-mouse alkaline phosphatase
Our data show that the combination of microfiltra- conjugate (Sigma, St. Louis, MO). The antibodies were detected with
Immobilon Western AP substrate (Millipore) and imaged on a Kodak
tion and ultrafiltration is a rapid method for MAb
scanner.
purification and that MAb purified on Montage Bottom panel shows activity comparison measured in an ELISA
centrifugal columns with Protein G media is compa- assay. HEK 293 cells were grown on 96-well plates. After fixation and
rable to the commercially available MAb in purity, epitope-retrieval by heating for 10 minutes in a microwave, cells were
permeabilized with 1% saponin (Sigma) in PBS + 2% normal donkey serum
activity and cost. The Ultrafiltration-based MAb
(Jackson Immunoresearch, West Grove, PA) and treated with serial dilutions
purification method, compared to the traditional of the indicated purified MAbs. The cells were then washed and treated
method, is faster (23 hours versus 2 days), easier with goat anti-mouse HRP-conjugate antibody (Sigma). The reactions were
to use, and yields higher recoveries. developed using a SureBlue TMB HRP substrate (KPL, Gaithersburg, MD).
The readings were measured on a SpectraMax plate reader (Molecular
Devices, Sunnyvale, CA).
References
1. Saha K, Case R, Wong PK. J Immunol Methods
1992;151(1-2):307-308.
2. Evan GI, Lewis GK, Ramsay G, Bishop JM. Mol
Cell Biol 1985;5(12):3610-3616.

53
A Simple Strategy for Protein
Enrichment Using Ultrafiltration
Introduction original concentration. These limitations can be
overcome by using centrifugal ultrafiltration devices
The human plasma proteome is a diverse universe
for enrichment.
of information and it will continue to be a vast
source of biomarker discovery and investigation.
Method
Ultrafiltration (UF) has been reported13 as a sample
This experiment demonstrates the use of ultrafiltration
preparation tool to prepare low molecular weight
devices to enrich both LMW and high molecular
(LMW, <10 kDa) fractions by reducing the complex-
weight (HMW) fractions from serum. Protein
ity of serum proteins prior to mass spectrometry for
separation was accomplished by serial filtration
biomarker analysis. Traditionally, enrichment of
through decreasing molecular weight cutoff
protein solutions on the basis of size has been
(MWCO) devices from 100K to 10K MWCO.
accomplished using gel filtration chromatography4,5.
This serial enrichment strategy enabled the com
However, gel filtration is laborious, limited by
partmentalization of proteins by size as well as an
sample size and time-consuming. Another conse-
increase in throughput as compared to directly
quence of gel filtration chromatography is the
enriching samples using a lower MWCO device.
significant dilution of the protein relative to the
P ro t o c o l s f o r P ro t e in s P ro te i n E n rich m e n t U s i n g U l t r af i l t r a t io n

Figure 1. Serial enrichment strategy Figure 2. Purification of Cytochrome c from a mixture


containing 10% fetal bovine serum (FBS) and the
Step 1: 100 kDa MWCO compartmentalization of protein fractions using
40 min at 1500 x g
Amicon Ultra devices

100 kDa Retentate 100 kDa Permeate M S R100 P100 R50 P50 R30 P30
(<100 kDa)

100 kDa
Step 2: 50 kDa MWCO
30 min at 1500 x g
50 kDa

50 kDa Retentate 30 kDa


50 kDa Permeate
(<50 kDa)
Cytochrome c Purified
(12.5 kDa) Cytochrome c

Step 3: 30 kDa MWCO


M: Marker R50: 50 kDa Rententate
30 min at 1500 x g
S: 10% FBS + 1 mg/mL Cytochrome c P50: 50 kDa Permeate
R100: 100 kDa Retentate R30: 30 kDa Retentate
30 kDa Retentate P100: 100 kDa Permeate P30: 30 kDa Permeate
30 kDa Permeate
(<30 kDa)
A serum-protein mixture spiked with purified Cytochrome c (12.5 kDa)
A complex mixture is centrifuged in a 100K device. was centrifuged in an Amicon Ultra 4 mL device using serially decreasing
The retentate is removed and saved for later analysis. MWCOs as described in Figure 1. The red, blue and green lines indicate
The permeate fraction is then centrifuged in a 50K approximate molecular weight ranges and represent the theoretical MWCOs.
device. This process is repeated for the 30K device. Colored dashed boxes represent size specific protein compartments.

54
The traditional methodology employing use of a References
single MWCO to remove HMW and enrich for
1. Forssmann WG, et al. J Chromatogr A
the LMW resulted in impure fractions compared
1997;776:125-132.
to those from serial enrichment.
2. Plebani M, et al. Pancreas 2002;24:8-14.
Conclusions 3. Sobrinho LG, et al. Clin Endocrinol (Oxf)
Protein fractionation using centrifugal ultrafiltration 2003;58:686-690.
devices is a simple and rapid means of reducing 4. Werner MJ. Chromatogr 1966;25(1):63-70.
sample complexity and compartmentalize proteins 5. Kent UM. Methods Mol Bio 1999;
based on molecular weight. Serial enrichment is 115:11-18.
faster than a direct single device enrichment and
protein fractions resulting from serial enrichment
have greater purity than those from a single device
method. Regenerated cellulose membranes out-
perform PES membranes in an enrichment paradigm.
Ultrafiltration is a potential alternative to gel filtration
chromatography for size based protein enrichment.

Figure 3. Serial enrichment is more efficient than Figure 4. Regenerated cellulose


direct enrichment in terms of purity and time membranes offer optimal separation
compared to PES
Time Required:

P ro t o c o l s f o r P ro t e in s P ro te i n E n rich m e n t U s i n g U l t r af i l t r a t io n
Serial Direct 100
100 min 130 min IgG (0.1 mg/mL)
Recovery of Cytochrome C (%)

90
(150300 L) (500600 L)
80 IgG (1.0 mg/mL)
M S R100 R50 R30 R10 M S R30 R10 70
60
50
100 kDa 40
30
20
10
0
Ultracel PES Device A PES Device B
50 kDa
30 kDa 30 kDa Regenerated cellulose membranes (Ultracel mem-
Light brane) outperform polyethersulfone (PES) membranes
Chains
used for enrichment. A binary mixture containing
Cytochrome c (1 mg/mL) and 155 kDa IgG (0.1 or
10 kDa 10 kDa
1.0 mg/mL) was centrifuged using 100K MWCO
devices. Cytochrome c recovery was measured in
M: Marker R50: 50 kDa Rententate
the permeates. Significantly greater Cytochrome c
S: 50% Adult Serum R30: 30 kDa Retentate
recovery was observed using Ultracel membrane
R100: 100 kDa Retentate R10: 10 kDa Permeate when the system was challenged with high levels
of IgG (1.0 mg/mL).
Direct enrichment is not recommended for unknown or highly concentrated
samples. The figure shows 50% adult bovine serum enriched directly
(right panel) using an Amicon Ultra-4 30K MWCO device. The device was
centrifuged for more than 2 hours at 1500 x g to achieve a final volume of
~500 L, an 8X concentration. Presence of BSA and other higher molecular
weight contaminating proteins clearly show the benefits of using a serial
enrichment strategy (left panel) as described in Figure 1. Permeates collected
from the 30K (R10) devices were concentrated with 10K devices prior to
protein visualization. An unknown protein ~12 kDa was purified using a
serial method, but not direct enrichment.

55
Urine Concentration with
Amicon Ultra Centrifugal Filters
Introduction Materials
The measurement of specific proteins in urine is Amicon Ultra-4 device, 4 mL, 10K NMWL
important for the diagnosis and management of
Centrifuge with fixed-angle or swinging bucket
disease states. In most cases, the content of these
rotor capable of 3400 x g
proteins in urine is too low to be detected and
needs to be concentrated. Amicon Ultra-4 devices Kit for microprotein determination
can be used to concentrate urine samples prior to (i.e. Sigma cat. no. 610-A/Brilliant blue G/
clinical laboratory analyses. For example, patients Coomassie blue)
with multiple myeloma exhibit a proliferation of one Pipetter with 200 L tip
antibody-producing plasma cell, which leads to
Electrophoresis (agarose gel) and immunofixation
excess production of free immunoglobulin light
equipment with apparatus and reagents
chains known as Bence-Jones proteins. After sample
enrichment in the Amicon Ultra-4 10K NMWL Method
device, immunofixation electrophoresis can be used
1. Determine the total protein in a 24-hour urine
to identify free light chains (Bence-Jones proteins)
specimen.
in urine by forming a light chain-antibody complex.
Also, agarose electrophoresis can be used to 2. Fill Amicon Ultra-4 device with 4 mL of urine.
quantitate light chains and identify additional low 3. Centrifuge at 3400 x g for 3045 minutes
molecular weight proteins such as albumin, a-1 (approximately 2550 L concentrate volume).
globulins, transferrin and IgG that can be present This produces up to a 160-fold increase in
in renal tubular disorders. Ultrafiltration of urine concentration.
samples in Amicon Ultra-4 devices provides 4. Insert a pipetter into the bottom of the filter unit
reproducible, high sample recovery for electropho- and withdraw the concentrated sample.
P ro t o c o l s f o r P ro t e in s Uri n e C o n ce n t r a t io n

retic analyses, usually in 45 minutes or less. 5. Perform agarose electrophoresis on the concen-
trate to quantitate light chains and identify other
proteins. Determine the percentage of light
chains with respect to the total number of
components in the urine. Then multiply the
percentage of light chains by the total 24-hour
protein concentration (grams per 24-hour volume).
6. Perform immunofixation electrophoresis on the
concentrate to identify light chains.

56
Acknowledgements Additional Notes
Research using Amicon Ultra devices for urine 1. Amicon Ultra devices can also be used to
concentration in this protocol was conducted by concentrate serum, plasma and cerebrospinal
Mark Merchant, Ph.D. at Helena Laboratories, fluid for similar analyses. A concentration of
Beaumont, TX. approximately 20 mg/mL is required in order to
detect free light chains from diseased patients by
References agarose electrophoresis. Detection by immuno-
1. Tietz, N. Clinical Guide to Laboratory Tests, fixation electrophoresis is 10 times more sensitive
2nd ed. Philadelphia: Saunders, WB; than by agarose electrophoresis.
1990;362363. 2. Normal heterogeneous immunoglobulins may
2. Kahns L. Clinical Chemistry 1991;37: also be seen in urine concentrate with immuno-
15571558. fixation electrophoresis. This ladder effect is
3. Cleveland Clinic homepage. Accessed July comprised of microheterogenous light chains.
2002. www.clevelandclinic.org/myeloma/ Bence-Jones proteins may be within this ladder.
DiagnosisAndTreatmentOf MultipleMyeloma.html To verify the presence of Bence-Jones proteins
requires additional analysis by two-dimensional
4. Christenson RH, et al. Clinical Chemistry
electrophoresis.
1983;29(6):10281030.
3. If there is excess antigen, dilution of the concen-
5. Christenson RH, and Russell ME. Clinical
trate will be required until equilibrium is achieved
Chemistry 1985;31(6):973.
between the antigen (Bence-Jones protein) and
the antibody.
4. Millipore also offers static concentrators
(Minicon devices) for concentration of
Bence-Jones protein in urine.

P ro t o c o l s f o r P ro t e in s Uri n e C o n ce n t r a t io n

57
Use of Centrifugal Filter Devices as
an Alternative to Stirred Cells
Introduction Abstract
Stirred cell devices have been successfully used to This study aims to achieve purification and concen-
purify proteins from large volumes of solution for tration of a fusion protein composed of the alpha
many years. However, the need for assembly and and gamma subunits of the human high affinity
cleaning between uses, and the lengthy separation IgE receptor. The alpha-gamma fusion protein is
times, can present difficulties for active, or under- purified and then coupled to Sepharose (GE)
staffed, laboratories. beads to produce an affinity column for isolating
Millipores large volume centrifugal filter devices human IgE antibodies.
P ro t o c o l s f o r P ro t e in s U s e of C e n t rif u g a l F i l te r D ev ice s a s a n A l te rn a t i ve to St i rre d C e l l s

come preassembled and ready to use. Individual


devices are available for processing 20 or 70 mL Method
volumes. For larger volumes, multiple devices can The alpha and gamma subunit components of the
be spun simultaneously. Spin times are typically high affinity IgE receptor were transfected into
measured in minutes. And unlike stirred cells, mouse myeloma cell line NS0 and the secreted
centrifugal devices run unattended. There is no fusion protein was purified on a Protein G column.
need to refill a reservoir and, therefore, less risk The eluted proteins were concentrated from
of adventitious contamination. volumes of 5001000 mL to 510 mL using a
The following protocol demonstrates the use Centricon Plus-70 centrifugal filter device in a
of Centricon Plus-70 centrifugal filter devices to swinging bucket rotor at 3000 x g for 10 minutes
purify and concentrate proteins from large volumes (60 mL per filter unit). Buffer exchange was also
of solution. performed subsequently using the same units in
tissue culture grade sodium azide-free PBS. The
concentrated samples were then filter-sterilized
Figure 1. under sterile conditions using a 0.2 m pore filter.

140
Results
120
Spectrophotometric evaluation of alpha-gamma
Absorbance Units

100 fusion protein recovery using Centricon Plus-70


80 devices showed a result of 18.8 mg/L of superna-
60 tant. HPLC trace analysis revealed the chromato-
gram shown in Figure 1.
40

20
Acknowledgement
0
0 5 10 15 20 25 30
Protocol courtesy of GKT School of Biomedical
Time (min) Sciences, London.

HPLC profile of alpha-gamma fusion component of the human FcRI

58
Protocols for
Purification of DNA Sequencing Reactions . . . . 60

Concentrating and Desalting DNA or RNA . . . . 63

Nucleic Acids Preparing Samples for Forensics ID Analysis . . . 66

Concentration of Genomic DNA for


Forensic Analysis . . . . . . . . . . . . . . . . . . . . . . 67

Purification of PCR Products . . . . . . . . . . . . . . . 68

Quantitative Recoveries of Nanogram


Amounts of Nucleic Acids . . . . . . . . . . . . . . . . 70

RNA Purification and Preparation of


Fluorescent cDNA Probe from Human mRNA . . 72

Purification of In Vitro Synthesized mRNA . . . . . 76

Effect of Centrifugal Ultrafiltration on


Large Fragment DNA Integrity . . . . . . . . . . . . . 78
DNA Extraction from Agarose Gels . . . . . . . . . 80

PCR Purification . . . . . . . . . . . . . . . . . . . . . . . 82

Enzyme Removal . . . . . . . . . . . . . . . . . . . . . . 84

P ro t o c o l s f o r Nuc l e ic Ac id s

59
Use of Microcon 100 Centrifugal Filter
Devices to Purify DNA Sequencing
Reactions
Introduction Method
DNA sequencing reactions performed using ABIs DNA was isolated according to standard protocols
BigDye terminator chemistries require the removal and amplified for sequencing analysis on an ABI
of unincorporated dye-terminators and excess salt model 3700 DNA sequencer using polymerase
prior to injection into a sequencer. The traditional reagents and ABIs BigDye Terminator v. 3.1.
method for purifying DNA sequencing reactions is Sample was purified using two experimental
ethanol precipitationa labor intensive method protocols. Results were assessed using electrophero-
requiring multiple spins, incubation, and drying that grams, Phred 20 read lengths, and estimation of
can take up to an hour. Care must also be taken to dye blobs.
retain the resulting pellet.
Microcon 100 protocol
Microcon 100 devices can remove contaminat-
ing salts and unincorporated dye terminators from 1. Sequence Product (10 L) + 100 L of Montage
P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of D N A S e q u e n ci n g Re a c t io n s

DNA sequencing reactions and deliver SEQ quality injection solution in a Microcon 100 device
and read lengths equal to or better than ethanol 2. Spin for 2.5 minutes at 12,000 x g
precipitation in 10 minutes. As capillary-based 3. Add 100 L of injection solution to device
DNA sequencers become more prevalent, the need 4. Spin for 2.5 minutes at 12,000 x g
for single sample purification of SEQ reactions also
5. Resuspend in 20 L of injection solution then
increases.
incubate for 2 minutes
The first protocol achieved a time savings of
6. Invert spin for 2 minutes at 1,000 x g
50 minutes over the traditional ethanol precipitation
method while a second protocol, which relies on 7. Inject into sequence analyzer
less vigorous centrifugation, took approximately The above steps were completed in 10 minutes
as long as ethanol precipitation but excelled in at least one third the time required for ethanol
experiments in which it was desirable to read precipitation.
close to the primer.
Results
Equipment Key parameters for DNA sequence reactions
Instrument: ABI sequencer cat. no.3700 include quality, read length, sequencing accuracy,
ability to read close to the primer, and the presence
Reagent: ABI BigDye Terminator v3.1
of dye blobs. The first three factors may be assessed
Injection parameters: existing parameters using Phred 20 scoring, which is performed auto-
developed for Montage Injection solution. matically via software. Phred 20 corresponds
Data output: Graphs and electropherograms roughly to a 99% probability that a base call is
that show A) removal of dye-terminators (blobs); accurate. Ability to read close to the primer is
B) Read length (Phred 20 scores); C) Primer measured by the Primer + value, which relies on
both software and visual verification. Dye blobs
are artifacts of unknown origin, occurring during
sequencing or cleanup, that may overshadow
peaks. Dye blob assessment is typically performed
by visually examining the electropherogram.

60
Table 1. Results from a sequence purification
using the Microcon 100 device
Dye Terminator Phred 20 Dye Blobs Primer +
1/2X 646 59 0.4 11 10
1/8X 656 42 None observed 87
n=48

Figure 1. Electropherogram of a 1/2X SEQ reaction, purified using the Microcon 100 protocol

P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of D N A S e q u e n ci n g Re a c t io n s
Microcon 100 at 12,000 x g Phred 20684 Primer + = 8 Dye blobs = 0

61
Conclusion Table 2. Results from a typical sequence purification
Microcon 100 ultrafiltration devices represent a using an alternate protocol
rapid and efficient alternative to ethanol precipita- Dye Terminator Phred 20 Dye Blobs Primer +
tion when preparing DNA for sequencing analysis. 1/2X 627 83 0.6 10
The high-spin protocol outlined above affords a 1/8X 653 73 None observed 1.7 3
significant reduction (10 minutes vs. 60 minutes) in Ethanol 605 88 0.5 2.6 4
sample preparation time. For experiments where low
Prime + is required, researchers can employ a low-
speed centrifugation protocol that provides read
lengths equal to or better than ethanol precipitation,
with far less opportunity for user error.

Figure 2. Electropherogram of a 1/8X SEQ reaction, purified using an alternate protocol


P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of D N A S e q u e n ci n g Re a c t io n s

Microcon 100 at 500 x g Phred 20 = 622 Primer + = 1 Dye blobs = 0

62
Concentrating and Desalting
DNA or RNA with Microcon and
Amicon Ultra Centrifugal Filters
Introduction Millipore ultrafiltration membranes are character-
ized with well-defined, globular solutes (proteins).
Centrifugal filter devices serve as powerful tools
Typically, the nominal molecular weight limit
in molecular biology applications, such as DNA
(NMWL) for a membrane is the point at which over
or RNA concentration and desalting procedures.
90% of a solute with that molecular weight will be
Ultrafiltration (UF) is a pressure-driven, convective
retained. To process DNA or RNA, the membrane
process that uses semipermeable membranes to
needs to be characterized according to the number
separate species by molecular size and shape.
of nucleotides in the fragment. Polynucleotides
UF is highly efficient, allowing for concentration
(DNA and RNA) have tertiary structures that are
and desalting at the same time. Unlike the use of
ordinarily more extended than those of typical
chemical precipitation methodologies (i.e., ethanol,
globular proteins of similar size. Millipore has
phenol/chloroform), there is no phase change,
determined nucleotide cut-offs (based on the
which often denatures labile species. DNA and
number of bases or base pairs in a fragment of

P ro t o c o l s f o r Nuc l e ic Ac id s C o n ce n t r a t i n g a n d D e s a l t i n g D N A o r R N A
RNA samples with starting concentrations as low as
DNA or RNA) that correspond to the NMWL of
5 ng/mL can be routinely concentrated in minutes
each of their low binding membranes. The nucleo-
with 99% recovery of starting material, and without
tide cut-off (NCO) indicates the fragment length of
the use of co-precipitants. Centrifugal concentrator
single- or double-stranded DNA or RNA that one
devices are ideal for separating high and low-
would expect to recover at 90% efficiency with a
molecular weight species. Ultrafiltration can also
unit of the named NCO. It is best to choose the
be used to change solvents by diafiltration. In this
NCO with about half the length of the fragment
process, the sample is concentrated, then diluted
of interest. For example, selecting a 30K NMWL
to the original volume with the desired buffer and
membrane for a 50 base pair fragment generally
concentrated again, thus washing out the
results in 90% product recovery. A 10K NMWL
original solvent.
membrane would provide for closer to 100%
recoveries but would take much longer to process
the sample. For nucleic acid samples >500 base
Table 1. Nucleotide cut-off guidelines for pairs, a 100K NMWL membrane is appropriate.
Microcon centrifugal devices (based on Table 1 offers guidelines for DNA/RNA retention
>90% recovery of nucleic acids) based on the nucleotide content of single- and
Single-Stranded* Double-Stranded double-stranded pieces. For example, more than
Nucleotide Nucleotide 90% of a single-stranded 30-mer will typically be
NMWL Cut-off (bp) Cut-off (bp)
retained by a Microcon 10K NMWL.
3K 10 10 Concentrating dilute DNA solutions is a key step
10K 30 20 for many subsequent preparative and analytical
30K 60 50 procedures. For example, standard plasmid
50K 125 100 preparations involving cesium chloride, equilibrium
100K 300 125 centrifugation, and gel filtration yield DNA in large
*Single-stranded nucleic acids with extensive secondary volumes that require concentrating prior to precipita-
structure will be better retained than those without. tion. DNA concentration is also necessary in
purifying restriction fragments from gels.

63
There are three major techniques currently sition. The salt concentration in a sample concen-
available for concentrating nucleic acids: trated by Microcon centrifugal filter devices will be
repeated extractions with n-butanol the same as in the original sample. For desalting,
the concentrated sample is diluted with water or
adsorption to ion exchange resin,
buffer to its original volume and spun again in a
followed by high salt elution
process called diafiltration. This removes the salt
lyophilization by the concentration factor of the ultrafiltration.
The first method has the disadvantage that For example, if a 500 L sample containing
n-butanol concentrates all solutes, including salt, 100 mM salt is concentrated to 25 L (20X
which tends to co-precipitate with DNA upon concentration factor), 95% of the total salt in the
addition of ethanol. The second method, ion sample will be removed. The salt concentration in
exchange, aside from requiring buffers of various the sample will remain at 100 mM. Rediluting the
ionic strengths, yields DNA in a high salt solution. sample to 500 L will bring the salt concentration
The third method, lyophilization, increases the to 5 mM. Concentrating to 25 L once more will
concentration of buffer components, which can remove 99% of the original total salt. The concen-
result in degradation of nucleic acids. trated sample will now be in 5 mM salt. For more
Ultrafiltration membranes retain DNA or RNA but complete salt removal, an additional redilution and
are permeable by smaller ionic buffer components. spinning cycle will remove 99.9% of the initial salt
Ultrafiltration alone does not change buffer compo- content (see Table 2).

Methods
P ro t o c o l s f o r Nuc l e ic Ac id s C o n ce n t r a t i n g a n d D e s a l t i n g D N A o r R N A

Table 2. Desalting and recovery with Microcon Microcon Device


30K NMWL devices
1. Select a Microcon unit with nucleotide cut-off
Spin % DNA % CsCl equal to or smaller than the molecular size of the
Number Recovered Removed
nucleic acid you want to retain (refer to Table 1).
1 88 99.9
2. Insert Microcon sample reservoir into one
2 89 100
of the two vials provided for each unit.
3 88 100
3. To concentrate (without affecting salt concentra-
Using 1 mL of 25 g/mL E. coli DNA in 6 M CsCl, sample was repeatedly con- tion): Pipette up to 500 L of DNA or RNA
centrated to 0.1 mL. Sample spun at 2000 x g in a 45 degree fixed angle rotor.
The concentrated sample was reconstituted to original 1 mL volume by adding
sample into the reservoir. Spin for recommended
50 mm Tris. After three spins, CsCl concentration was reduced by four orders of time, not exceeding recommended g-force
magnitude. guidelines shown in Table 3.
4. To exchange salt: Add the proper amount of
appropriate the diluent to bring the concentrated
Table 3. Recommended g-force and spin time sample to 500 L. Spin for the recommended
for Microcon devices time, not exceeding g-force shown in Table 3.
To achieve lower salt concentration, repeat the
Maximum Spin Time (min) Spin Time (min)
NMWL G-Force Rating at 4 C at 25 C entire step as necessary. NOTE: Do not let filtrate
3K 14,000 185 95 vial overfill.

10K 14,000 50 35 5. Remove reservoir from vial and invert into a


30K 14,000 15 8 new vial (save filtrate until sample has been
analyzed).
50K 14,000 10 6
100K 500 25 15 6. Spin for 2 minutes at 5001000 x g to recover
nucleic acid in the vial.
7. Remove reservoir. Cap vial to store.

64
Amicon Ultra Device Table 4. Nucleotide cut-off guidelines for Amicon Ultra-4
1. Select an Amicon Ultra unit with a nucleotide A. Nucleotide cut-off guidelines for single stranded DNA
cut-off equal to or smaller than the molecular size
Single-Stranded NMWL
of the nucleic acid you want to retain (refer to
Nucleotide Cut-off (bp) 3K (40 min) 10K (20 min)
Table 4).
10 98 97
2. To concentrate (without affecting salt concentra-
15 96 95
tion): Pipette up to 4 mL of DNA or RNA sample
20 94 94
into the reservoir. Spin for the recommended time,
25 96 96
not exceeding g-force shown in Table 5.
3. To exchange salt: Add the proper amount of the B. Nucleotide cut-off guidelines for double stranded DNA
appropriate diluent to bring the concentrated NMWL
Double-Stranded
sample to 4 mL. Spin for the recommended time, Nucleotide Cut-off (bp) 30K 50K 100K
not exceeding g-force shown in the Table 5. 137 95 95
To achieve lower salt concentration, repeat the
301 94
entire step as necessary. NOTE: Do not let filtrate
657 99
vial overfill.
1159 100 97
Based on >90% retention. Spinning 50 and 100K devices at higher than
recommended g-force may result in lower recovery of DNA.

P ro t o c o l s f o r Nuc l e ic Ac id s C o n ce n t r a t i n g a n d D e s a l t i n g D N A o r R N A
Table 5. Recommended g-force and spin time
for Amicon Ultra devices
Maximum Spin Time (min)
NMWL G-Force Rating at 25 C
3K 7,500 40
10K 7,500 20
30K 5,000 10
50K 2,000 20
100K 2,000 10

65
Preparing Samples for Forensics
Identification Analysis Using
Microcon Centrifugal Devices
Centrifugal filter devices with ultrafiltration (UF) Several suppliers offer STR assay kits. Each
membranes are used by forensics laboratories to supplier offers several versions of their kits with
isolate genomic DNA for human identification. The different protocols, modes of operation, and
source materials are typically blood stains and/or detection systems to suit the users needs. Millipores
other bodily fluids obtained from crime scenes, UF-based centrifugal devices are specified
criminal suspects, or human remains. in several of these protocols (AmpFLSTR Profiler,
The isolated genomic DNA is used to identify Applied Biosystems; GenePrint STR and GenePrint
individual persons based on their patterns of Short Fluorescent STR Systems, Promega).
P ro t o c o l s f o r Nuc l e ic Ac id s P re p a ri n g S a m p l e s f o r F o re n s ic s I d e n t if ic a t io n A n a l y s i s

Tandem Repeats. STRs are polymorphic DNA loci


that contain repeated DNA sequences. Because the Use of UF-based Centrifugal Devices in
repeats can vary from two to seven bases in length, New York City
many different alleles are possible for each locus. Depending on the type of evidence under investiga-
The parallel analysis of 9 to 13 polymorphic tion, the Department of Forensic Biology in New
STR loci can unequivocally identify an individual. York City had been preparing genomic DNA by
either (1) organic extraction (phenol/chloroform/
isoamylalcohol) followed by alcohol precipitation
Figure 1. or (2) direct lysis in the presence of Chelex beads
Genotype analysis of a (Bio-Rad) with no further purification.
semen sample before The Department of Forensic Biologys laboratory
A
and after concentration has replaced the ethanol precipitation step with
by a Microcon centrifu-
gal filter. Electrophero-
Microcon centrifugal filters with a 100K NMWL
gram A shows a partial ultrafiltration membrane. The centrifugal devices
profile with a high provide a faster way to concentrate the purified
molecular weight loci
DNA without the use of toxic chemicals.
in each color missing.
Electropherogram B The laboratory also uses the Microcon 100K
shows a full profile and NMWL device after Chelex extraction to remove
higher peak heights. inhibitors that can adversely affect PCR amplifica-
Amounts amplified were
tion. In addition, Chelex extraction of small blood-
0.62 ng for electrophe-
rogram A and 1 ng for stain samples often results in DNA concentrations
electropherogram B. below detection limits. Concentrating extracted
B The sample was extract- samples with Microcon 100K NMWL devices
ed with Chelex beads.
The peaks are labeled
improves the success rate of STR amplification and
with allele name, size yields a full STR allele profile even with minute
in base pairs, and blood or semen stains. According to the New York
fluorescent peak height.
City Department of Forensic Biology, several of the
The improvement in
results is due not only peaks shown in Figure 1 would have been below
to the increased DNA the detection threshold and identification would
input but also to the have been negative or inconclusive without sample
concentration step
cleanup using Microcon devices.
following extraction.

66
Concentration of Genomic DNA
for Forensic Analysis Using
Amicon Ultra-4 50K Centrifugal Devices
Introduction Protocol Recommendations for
Millipores Amicon Ultra-4 centrifugal filter devices Genomic DNA Isolation
provide fast ultrafiltration, with the capability for Dilute sample to 2.0 mL with TE Buffer
high concentration factors. The device design (10 mM Tris, pH 7.6, 1 mM EDTA)
incorporates the Millipore Ultracel low binding
Centrifuge at 2,000 x g for 20 minutes
regenerated cellulose membrane, providing
at room temperature
excellent sample recoveries from dilute and complex
sample matrices. The Amicon Ultra device is Repeat for the number of washes desired
configured in a vertical design, which minimizes Note: The centrifugation time is based on samples

P ro t o c o l s f o r Nuc l e ic Ac id s C o n ce n t r a t io n of G e n o m ic D N A f o r F o re n s ic A n a l y s i s
solute polarization and subsequent fouling of the containing up to 2 g of genomic DNA. For forensics
membrane. The vertical design and available samples, users will likely need to determine the
surface area provide for fast sample processing, appropriate centrifugation time as many factors
high retentate recovery (typically greater than (e.g., DNA concentration and temperature) will
90% of dilute protein or DNA concentrate), and affect centrifugation time.
the capability for very high concentration factors
(>80-fold). The Amicon Ultra-4 devices are suitable
for use with dilute nucleic acid samples which can
be quickly ultrafiltered, allowing for fast separation
Amicon Ultra has enabled our testing lab
of genomic DNA from low molecular weight (MW)
to process samples quicker and with a
compounds.
higher convenience and confidence as the
The Amicon Ultra device design is compatible
decreased number of handling steps allows
with the multiple spin recovery assays used for the
us to be free of cross-contamination.
purification of genomic DNA for forensic analysis.
The concentrate is collected from the filter unit Professor Miguel Paredes,
sample reservoir using a pipetter, while the ultrafil- Barcelona Laboratory of Instituto Nacional
trate can be collected in the provided centrifuge de Toxicologia y Ciencias Forenses
tube if desired. The filter device can be spun in a
swinging bucket or a fixed angle rotor. The Amicon
Ultra devices come fully assembled in a centrifuge
tube, ready for sample use.

67
Purification of PCR Products
with an Amicon Ultra Device
Introduction
Figure 2. Effect of MWCO and g-force
To enable the use of DNA fragments generated by
on the recovery of a 137 bp PCR product
the Polymerase Chain Reaction (PCR) in downstream
using Amicon Ultra-4 100K device
applications, unincorporated primers, nucleotides
100
and contaminating salts must to be quantitatively
removed. The application of ultrafiltration to PCR

% PCR Recovery
80

clean-up is well established and represents a 60


convenient and efficient approach to generate
40
concentrated, purified PCR products suitable for such
20
downstream applications such as cloning or DNA
sequencing. Millipore offers a number of products 0
2,000 x g 5,000 x g 7,500 x g
designed for the purification of PCR products in
96-well and 384-well plate and 1-up formats.
The protocol and data shown here demonstrate the achieved via dilution of the starting material
utility of Amicon Ultra-4 devices for the purification and running at a relatively low g-force. For
of PCR products. By extension, these data can be PCR fragments, an Amicon Ultra-4 device with
used to select an Amicon Ultra with the appropriate a 50K MWCO membrane generally strikes the
molecular weight cut-off (MWCO) for use in the best balance between speed and recovery. If the
purification of DNA used in other applications.
P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of P C R P ro d u c t s

DNA sample is in a buffer containing high salt and


cannot be diluted, or the device is run at a higher
Considerations than recommended g-force, a significantly reduced
For applications involving nucleic acids, strand DNA recovery will likely be observed. Under these
length is the most useful parameter for selecting the conditions, higher DNA or RNA recovery can
Amicon Ultra device appropriate for a particular be achieved by using the Amicon Ultra with the
application. However other parameters including, next tighter membrane, albeit with a much longer
DNA concentration, the magnitude of the driving processing time. Note that, for maximal removal of
force (g-force) and the salt concentration all act in unincorporated single-stranded primers from double-
concert to affect DNA recovery. When purifying stranded DNA fragments, the molecular weights of
PCR reactions for example, optimal yield can be the primer and DNA fragment should differ by at
least an order of magnitude.

Figure 1. PCR recovery with Method


Amicon Ultra-4 50K device DNA fragments (137, 301, 657 and 1159 bp)
100 were generated by PCR using plasmid DNA as
Recovery (% of Challenge)

the template. After pooling to minimize device-


80
to-device variability, 100 L aliquots of the PCR
60
reactions were diluted with 2.0 mL of TE Buffer
40 (10 mM Tris-HCL, pH 8.0, 1 mM EDTA) and
20
added to an Amicon Ultra-4 device with a 50K
MWCO membrane. The samples were centrifuged
0
(n=8) 137 301 657 1159 at 2,000 x g for 20 minutes, an additional 2.0 mL
PCR Fragmentation Size (bp) of TE buffer was added to the sampled and the PCR
products were concentrated by a second 20-minute

68
spin at 2,000 x g. The purified PCR products were Ultrafiltration-based purification relies on size
collected by pipette and the recovered DNA exclusion to affect the separation of single stranded
quantified by a fluorescent SYBR Green I assay primers from the double stranded PCR fragments.
(Molecular Probes). The final sample volume was As discussed above, the major challenge to
87 15 L. The recoveries shown in Figure 1 optimizing ultrafiltration for PCR purification is
demonstrate the high recovery that can be obtained with small, <1000 bp PCR products. Similarly, the
from PCR reactions using this approach. efficiency of primer removal becomes more
The recovery of PCR fragments purified by challenging as the size of the single stranded primer
ultrafiltration can be challenging when the resulting increases. As shown in Figure 4, the ability of an
DNA fragments are <1000 bp in length. The Amicon Ultra device with 30K MWCO membrane
amount of DNA recovered after purification can to remove primers is significantly diminished as the
be maximized by using an Amicon Ultra device primer length approaches 50 bases. In contrast,
with a lower MWCO membrane (e.g., 50K versus when a 50K MWCO membrane is used, primer
100K MWCO) and by centrifuging the devices at removal remains high and is consistent across the
2,000 x g as shown in Figure 2 for a 137 bp PCR range of PCR primers evaluated. Whereas the use
fragment. Although increasing the g-force will speed of a 100K would be expected to quantitatively
processing time, it will do so at the expense of DNA remove all but the largest of primers, purification
recovery. For larger DNAs, the effect is less of small (e.g., <300 bp) PCR products is suboptimal
dramatic (data not shown). when using a 100k MWCO membrane. Therefore,
The quantitative removal of single-stranded DNA an Amicon Ultra-4 device with a 50K MWCO
primers or oligonucleotides from PCR reactions is membrane provides the best balance of primer
critical for downstream applications. In order to removal and recovery when used for PCR
demonstrate the ability of Amicon Ultra devices to purification.
efficiently remove primers, unpurified PCR products
were spiked with 20 pmoles of a DNA reporter Conclusions

P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of P C R P ro d u c t s
primer that had been labeled with fluorescein on its The data presented herein demonstrate the utility of
5 end. This primer (TCAG)5 was designed such that Amicon Ultra-4 devices for the purification of PCR
it had minimal secondary structure. After purification products. By using devices with a 50K MWCO,
of the PCR products with an Amicon Ultra-4 device, users can expect to routinely obtain both high
the amount of primer remaining in the sample was recoveries and efficient primer removal that will
determined by measuring fluorescence against a facilitate downstream applications. Although the
standard curve. The average primer removal in this analysis performed for this study focused on PCR
experiment was 97.7 1.4%. This level of primer purification, the general principles are applicable
removal has been shown to be sufficient for DNA to purification of other nucleic acid preparations.
sequencing and other important PCR-based
applications.
Figure 4. Effect of MWCO on primer
removal in Amicon Ultra devices
Figure 3. Amicon Ultra device efficiently 100
Removal (% of Challenge)

removes unincorporated PCR primers


80
100
Removal (% of Challenge)

60
80
40
60
20
40
0
20 20 25 48
Primer Length (Bases)
0
137 301 657 1159 Amicon Ultra, 50k Amicon Ultra, 30k
PCR Fragmentation Size (bp)

69
Quantitative Recoveries of Nanogram
Amounts of Nucleic Acids with
Microcon Centrifugal Filters
Introduction were then centrifuged at 12,000 x g in a fixed-
angle microcentrifuge at 4 C for 15 minutes. The
Molecular cloning experiments often require
supernatants were carefully removed and pellets
concentration of nanogram quantities of DNA.
resuspended in 50 L of TE buffer. The radioactivity
When dealing with such small amounts of nucleic
in the precipitates and supernatants was then
acids, the use of coprecipitants (tRNA or purified
determined by counting Cherenkov radiation. The
glycogen) is essential for effective DNA recovery1.
P ro t o c o l s f o r Nuc l e ic Ac id s Q u a n t i t a t i ve Re cove rie s of N a n o g r a m A m o u n t s of Nu cl e i c A ci d s

percentage of recovered DNA was calculated from


However, when carrier tRNA cannot be used (for
precipitates and the initial counts. Each data point
example, when DNA is to be labeled with 32P dATP
in the tables represents the average of at least
and polynucleotide kinase), the only method of
three samples.
recovering DNA as a precipitate in ethanol has
To precipitate the oligomer, 60 L of TE buffer
been the ultracentrifugation method developed by
containing a given amount of 25-mer and 5 ng of
Shapiro2. Ultrafiltration, which has been traditionally
radiolabeled tracer was supplemented with 240 L
used to concentrate and desalt protein samples
of 5 M ammonium acetate and 750 L of EtOH.
simultaneously, can be an efficient alternative to
Subsequent steps were identical to those described
ethanol precipitation.
for DNA.
In this experiment, three popular ethanol
precipitation protocols for DNA and oligonucle- Ultrafiltration
otides are compared with ultrafiltration in Microcon A solution of DNA (500 L) was placed into a
centrifugal filter devices. For a detailed discussion Microcon 30K NMWL unit and spun at 12,000 x g
on the effects of incubation time, temperature and for 10 minutes. A 500 L solution of oligonucle-
centrifugation parameters on ethanol precipitation, otides in TE buffer was placed into a Microcon 3K
see Reference 3. NMWL unit and spun for 45 minutes at 12,000 x g.
The retentates were recovered by inverting the units
Methods and centrifuging at 5001000 x g for 2 minutes.
Plasmid pBR322 was digested with EcoR I and the While concentrating the oligomer samples, it is
3 recessed termini were filled in with alpha 32P important to avoid high salt concentrations which
dATP, using Klenow fragment of DNA polymerase I. promote binding of single-stranded nucleic acids
A 25-nucleotide mixed base oligomer was radio to the cellulose-based ultrafiltration membrane.
labeled by phosphorylation with bacteriophage T4 The radioactivity in the retentates and filtrates
polynucleotide kinase. was determined by counting Cherenkov radiation.
Data represent averages of six samples each.
Ethanol Precipitation
All DNA precipitations were performed in 250 L
Results
volume. Each tube contained a given amount of
Standard methods for ethanol precipitation of
DNA, supplemented with 10 ng of radioactively
nucleic acids and ultrafiltration are compared in
labeled pBR322 in 0.3 M sodium acetate buffer.
Tables 1 and 2. Recovery of DNA was assessed at
To precipitate the DNA, 2.5 volumes of 95% EtOH
close to 100%, indicating that no significant amount
were added to each tube. The content was well
of nucleic acid was lost to the membrane or device
mixed and incubated for the specified period of
due to adsorption.
time at 20 C or 70 C (dry ice). The solutions

70
In contrast, DNA recovery using EtOH precipita- References
tion varied from as little as 14% as in the case of
1. Wallace DM. Precipitation of Nucleic Acids.
DNA (10 ng/mL) precipitated for 15 minutes at
Methods in Enzymology 1987;152:418.
70 C to a maximum of 76% after overnight
incubation at 20 C. 2. Shapiro DJ. Quantitative Ethanol Precipitation of
Nanogram Quantities of DNA and RNA. Anal
Discussion Biochem 1981;110:22931.
Poor recovery of nucleic acids at very low concen- 3. Crouse J, Amorese D. Ethanol Precipitation:
tration with ethanol precipitation may be partially Ammonium Acetate as an Alternative to Sodium
due to the fact that small amounts of DNA do not Acetate. Focus 1987;9(2):35.
adhere well to the tube walls following sedimenta-
tion unless high g forces (ultracentrifugation) are
employed. The yield of DNA incubated at 70 C
is slightly reduced, in agreement with previous

P ro t o c o l s f o r Nuc l e ic Ac id s Q u a n t i t a t i ve Re cove rie s of N a n o g r a m A m o u n t s of Nu cl e i c A ci d s


studies. Also an overnight precipitation at 20 C
can significantly improve recovery. The ultrafiltration
devices were found to concentrate and desalt
nucleic acids effectively in one step resulting in
high recoveries and providing a quick, alternative
to ethanol precipitation.

Table 1. Efficiency of different methods for concentration of DNA


DNA Concentration
10 ng/mL 25 ng/mL 50 ng/mL 250 ng/mL 1000 ng/mL
Method % Recovery % Recovery % Recovery % Recovery % Recovery
EtOH, 70 C, 15 min 14 15 23 52 55
EtOH, 20 C, 30 min 13 20 25 60 72
EtOH, 20 C, 18 hrs 31 45 45 76 67
Microcon 30K NMWL Device 95 95 98 98 99

Table 2. Efficiency of different methods for concentration of oligonucleotides


Oligomer Concentration
10 ng/mL 25 ng/mL 50 ng/mL 250 ng/mL 1000 ng/mL
Method % Recovery % Recovery % Recovery % Recovery % Recovery
EtOH, 70 C, 15 min 4 4 5 6 33
EtOH, 20 C, 30 min 6 5 6 4 33
EtOH, 20 C, 18 hrs 17 20 15 30 58
Microcon 30K NMWL Device 93 94 93 95 95

71
RNA Purification and Preparation of
Fluorescent cDNA Probe from Human
mRNA with Microcon Centrifugal Filters
Introduction tion of high quality fluorescently labeled probes
P ro t o c o l s f o r Nuc l e ic Ac id s R N A P u rif ic a t io n a n d P re p a r a t io n of F lu o re s ce n t cD N A P ro b e f ro m Hu m a n m R N A

suitable for use with microarrays. The protocol


When working with RNA, introduction of RNAse
below describes a method for the generation and
contamination during sample preparation is of major
purification of fluorescently labeled probes using a
concern. Sample yield and integrity can impact the
Microcon 30K NMWL centrifugal filter device.
efficiency of subsequent translations, hybridizations,
protein binding, antisense, or ribozyme studies. A RNA Sample Integrity
series of experiments was performed to determine The primary objective of the study was to determine
the effectiveness of using Amicon centrifugal the effect of centrifugation on RNA integrity. The
ultrafiltration devices from Millipore to concentrate second objective was to determine any gross
and diafilter RNA samples. The results indicate changes in the RNA that could be a result of
remarkably high RNA recoveries and low adsorption contact of the sample with the ultrafiltration devices
losses, especially with prior membrane passivation. themselves. An RNA transcript was made, using
Microarrays are efficient tools that enable the MEGAscript T7 RNA polymerase (Ambion) and
high throughput identification of genes that are DNA template Riboprobe Gemini (Promega)
differentially regulated in response to disease, drugs according to manufacturers protocols. This transcript
or other stimuli. With the completion of several key served as the starting RNA sample for the experi-
genome sequencing projects, scientists now have ments discussed below. Figure 1 is an autoradio-
the ability to custom design DNA microarrays gram of labeled RNA. It compares an untreated
specific to their research interests. The recent sample to samples that were concentrated in
advances in robotics, bioinformatics and detection Microcon 100K NMWL and 30K NMWL devices,
technologies have greatly simplified the manufacture used directly as supplied. The Microcon 100K
and analysis of microarrays. NMWL unit was spun at a force of 3,000 x g and
However, the successful application of microarray the Microcon 30K NMWL unit at 12,000 x g. As
technology requires highly purified, fluorescently is evident from the autoradiogram, both unfiltered
labeled cDNA probes. The application of ultrafiltra- samples are virtually indistinguishable from the
tion technology to this challenge has resulted in starting material.
a robust, efficient and rapid method for the genera-

Figure 1.
Concentration of RNA transcript. RNA is intact
1 2 3
and recovered with high efficiency.

Lane 1: Starting material 1.4 kb


Lane 2: RNA concentrated in a Microcon
100K NMWL device
Lane 3: RNA concentrated in a
Microcon 30K NMWL device 0.68 kb

72
Method 7. Neutralize by addition of 15 L of 0.1 N HCl.
8. Add 380 L of TE (10 mM Tris, 1 mM EDTA) to
Generation and Purification of Fluorescently a Microcon 30K NMWL device column. Next
Labeled Probes add the 60 L of Cy5 probe and the 60 L of
1. To anneal primer, mix 2 g of mRNA or Cy3 probe to the same Microcon device.
50100 g total RNA with 4 g of a regular NOTE: If re-purification of Cy dye flow-through is
or anchored oligo-dT primer in a total volume desired, do not combine probes until Wash 2.
of 15.4 L as shown in Table 1.

P ro t o c o l s f o r Nuc l e ic Ac id s R N A P u rif ic a t io n a n d P re p a r a t io n of F lu o re s ce n t cD N A P ro b e f ro m Hu m a n m R N A
9. Wash 1: Spin column for 7 to 8 minutes at
2. Heat to 65 C for 10 minutes and cool on ice.
14,000 x g.
3. Add 14.6 L of reaction mixture each to Cy3
10. Wash 2: Remove flow-through and add
and Cy5 reactions as shown in Table 2.
450 L TE and spin for 78 minutes at
4. Incubate at 42 C for 1 hour.
14,000 x g. It is a good idea to save the flow-
5. Add 1 L SSII (RT booster) to each sample. through for each set of reactions in a separate
Incubate for an additional 0.5 1 hours. microcentrifuge tube.
6. Degrade RNA and stop reaction by addition
15 L of 0.1 N NaOH, 2 mM EDTA and
incubate at 6570 C for 10 minutes. If
starting with total RNA, degrade for 30 minutes
instead of 10 minutes.

Table 1. Preparation of primer


Primer Cy3 (L) Cy5 (L) Notes
mRNA (1 g/L) x y 2 g of each if mRNA; 50100 g if total RNA
Oligo-dT (4 g/L) 1 1 Anchored: 5-T T T T T T T T T T T T T T T T T T T T V N-3
Purified H2O (DEPC) to 15.4 to 15.4
Total volume 15.4 15.4

Table 2. Preparation of probe


Reaction Mixture Volume (L) Unlabeled dNTPs Volume (L) Final Concentration (mM)
5X first-strand buffer* 6.0 dATP (100 mM) 25 25
0.1 M DTT 3.0 dCTP (100 mM) 25 25
Unlabeled dNTPs 0.6 dGTP (100 mM) 25 25
Cy3 or Cy5
3.0 dTTP (100 mM) 10 10
(1 mM, GE Healthcare)
Superscript II 2.0 Purified H2O 15
(200 U/L, Gibco-BRL)
Total volume 14.6 Total volume 100
*5X first-strand buffer: 250 mM Tris-HCl (pH 8.3), 375 mM KCl, 15 mM MgCl2

73
11. Wash 3: Remove flow-through and add Results
450 L 1X TE, 20 g of Cot1 human DNA
RNA recovery is a function of the initial RNA
(20 g/L, Gibco-BRL), 20 g polyA RNA
concentration and the buffer salt concentration.
(10 g/L, Sigma cat. no. P9403) and
Figure 2 shows RNA transcript recovery in
20 g tRNA (10 g/L, Gibco-BRL cat.
Microcon 100K.
no. 15401-011). Spin 710 minutes at
NMWL unit as a function of RNA concentration.
14,000 x g. Look for concentration of the probe
For all concentrations evaluated, retentate recovery
in the Microcon device. The probe usually has
is above 85%, even at an initial concentration as
P ro t o c o l s f o r Nuc l e ic Ac id s R N A P u rif ic a t io n a n d P re p a r a t io n of F lu o re s ce n t cD N A P ro b e f ro m Hu m a n m R N A

a purple color at this point. Concentrate to a


low as 25 ng/mL.
volume of less than or equal to the volume
Figure 2 also displays the material monitored in
listed in the Probe and TE column in Table 3.
the filtrate and on the membranes of the Microcon
These low volumes are attained after the center
units. (Membranes were removed and counted
of the membrane is dry and the probe forms
without further treatment.) The amount of RNA in the
a ring of liquid at the edges of the membrane.
filtrate does not vary significantly as a function of
Do not dry the membrane completely.
concentration. The RNA recovered on the mem-
12. Invert the Microcon device into a clean tube branes varies from <1% at high initial concentrations
and spin briefly at 14,000 RPM to recover to 4% at the lowest concentration tested. Millipores
the probe. Amicon centrifugal ultrafiltration devices contain
13. Select the appropriate row from Table 3. Adjust low-binding cellulosic Ultracel-YM membranes.
the probe volume to the value indicated in the Nitrocellulose is commonly used to immobilize RNA,
Probe and TE column. generally in high salt concentrations.
14. For final probe preparation add 4.25 L 20X Figure 3 shows RNA recovery as a function of
SSC and 0.75 L 10% SDS. When adding buffer salt concentration. The data show that both
the SDS, be sure to wipe the pipette tip with retentate and total recoveries fall as salt concentra-
clean, gloved fingers to remove excess SDS. tion increases. An increase in the amount of material
Avoid introducing bubbles and never vortex on the membrane is also seen (from 2% to 6%) as
after adding SDS. The probe is ready for the salt concentration increases from 10 to 500 mM.
hybridization. Similar trends are seen in the use of Microcon

Table 3. Final probe preparation


Cover Slip Size Total Hybrid Volume (L) Probe and TE (L) 20X SSC* (L) 10% SDS (L)
22 x 22 15 12 2.55 0.45
22 x 40 25 20 4.25 0.75
22 x 60 35 28 5.95 1.05
*20X SSC: 3.0 M NaCl, 300 mM NaCitrate (pH 7.0)

74
100K NMWL devices, except that retentate
recoveries fall to 60% at 500 mM salt. It was Figure 2.
possible to count the devices themselves in addition 100%

to the filtrate and membrane. The results show a


80%
slight increase in counts on the membrane with

RNA Recovered
increasing salt concentration. Significantly more
60%
(from 9% to 17%) material remained on the device
as salt concentration increased. Passivation in- 40%

P ro t o c o l s f o r Nuc l e ic Ac id s R N A P u rif ic a t io n a n d P re p a r a t io n of F lu o re s ce n t cD N A P ro b e f ro m Hu m a n m R N A
creased RNA recovery to 80%, regardless of initial
20%
salt concentration. This is attributed to a decrease
in the amount of RNA adhering to the device. 0%
When the devices were passivated, RNA loss 0.025 0.25 0.5 1.0 5.0 10.0
RNA Concentration (g/mL)
due to adsorption was reduced to 2%, regardless
of salt concentration. Microcon Microcon Microcon
Filtrate Membrane Retentate

Conclusion RNA recovery in Microcon concentrators. RNA was concentrated as


The results shown here demonstrate that RNA described in text. Cherenkov counts of Microcon retentate, filtrate and
samples can be concentrated or diafiltered with membrane (n=3 units) were compared to total starting counts.

Microcon concentrators without loss of RNA


integrity. RNA recovery in the retentate is typically
>85%. As the salt concentration of the buffer system
Figure 3.
increases, RNA retentate recovery decreases,
principally due to RNA adhering to the device 100%

rather than to the membrane. RNA recovery from


80%
solutions with high initial salt concentrations can be
RNA Recovered

significantly improved by pre-treating the devices 60%


with a 5% SDS solution.
40%

Acknowledgements
20%
Fluorescent probe preparation protocol courtesy
of Patrick Brown, Max Diehn, and Ash Alizadeh, 0%
10 100 200 500
Stanford University School of Medicine. NaCl Concentration (mM)

Filtrate Membrane Device Retentate

Improvement in RNA recoveries using Microcon devices (n=6) which were


pre-treated with a 5% SDS solution.

75
Purification of In Vitro Synthesized
mRNA with Microcon Centrifugal Filters
Introduction Methods
In vitro transcription reactions employing T3, T7 or RNA Transcription
SP6 phage-encoded RNA polymerases are widely For our studies we chose plasmid pGEM-luc
used to synthesize RNA from recombinant vectors containing the luciferase gene (luc) in the center of
containing appropriate promoters. Production of a multiple cloning cassette of the pGEM-11Zf (-)
large amounts of specific RNA is valuable in the plasmid (Promega). DNA template was linearized
preparation of hybridization probes and in vitro with XhoI, followed by enzyme and salt removal by
translation studies; in the synthesis of ribozymes, diafiltration in Microcon 100K NMWL devices.
rRNA, SRP, antisense RNA and substrates for RNA Linearized template was transcribed, using
splicing; and in RNA-protein interaction studies. MEGAscript kit (Ambion) according to the recom-
Microcon centrifugal filters are well suited for mended protocol. After the reaction was completed
the purification of radiolabeled RNA transcripts1. (3 to 4 hours), template DNA was degraded with
Ultrafiltration can simultaneously and efficiently DNase I and the reaction mix added to a Microcon
remove unincorporated ribonucleotides and salts 30K NMWL device filled with 450 L of water. The
P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of I n Vi t ro Sy n t h e s i ze d mR N A

from the transcripts and concentrate the RNA. RNA device was spun for 20 minutes at 12,000 x g in a
molecules retain their integrity and are recovered temperature-controlled centrifuge at 4 C. Purified,
with high yields. concentrated RNA was recovered by inverted spin.
Purity of a transcript is especially important when For the preparation of capped transcript, cap
it is used in in vitro translation systems. Trace analog m7G (5) ppp (5) G (New England
amounts of ethanol, phenol, salts or excess cap Biolabs, Inc.) was included in the transcription
analog used during the synthesis of capped mRNA reaction and the level of GTP reduced (4:1 ratio
can cause a dramatic decrease in translation of cap analog to GTP). To purify the transcript by
efficiency. phenol/chloroform extraction, the reaction mix
After the transcription reaction is complete, was diluted with water and a one-tenth volume of
template DNA is usually degraded by the addition ammonium acetate stop solution was added. The
of DNase I. The RNA is purified by two phenol/ mixture was extracted once with phenol/chloro-
chloroform extractions followed by ethanol form, followed by chloroform extraction. RNA was
precipitation. Other, less popular methods are gel precipitated with isopropanol and the pellet
purification (used predominantly when separation of resuspended in distilled water. Alternatively, LiCl
full-length transcript from shorter RNAs is important, precipitation solution (one-half volume) was added
e.g., ribonuclease protection assays) or LiCl to the reaction mix, followed by incubation at minus
precipitation. 20 C for 1 hour. RNA was pelleted by centrifuga-
A series of experiments was performed in our tion and dissolved in water. Size and integrity of
laboratory to determine the effectiveness of using the in vitro transcription products were assessed by
Microcon devices to purify in vitro synthesized running an aliquot of the purified RNA transcript on
mRNA and in vitro translation studies. Results a formaldehyde/formamide agarose gel. Ethidium
indicate that ultrafiltration can efficiently remove bromide was added to the RNA before lading on
inhibitory contaminants from mRNA preparations, the gel to stain the RNA sample and keep back-
leading to increased translational efficiencies. ground fluorescence low2.

76
Translation In Vitro References
In vitro translations were performed in the Flexi
1. Krowczynska AM. BioSolutions 1993;2(1):12.
Rabbit Reticulate Lysate System (Promega) according 2. Ogretmen B, Ratajczak H, Kats A, Stark BC.
to standard luciferase RNA translation conditions Biotechniques 1993;14(6):932-5.
with minor modifications (Rnasin Ribonuclease
3. Polayes D. Focus 1991;13(4):1302.
inhibitor was omitted and 35S-methionine added).
4. Dasso MC, Jackson RJ. Nucl Acid Res 1989;
Results of translation were analyzed by determina-
17:3129.
tion of percent incorporation of 35S-methionine and
fold stimulation, compared to controls without RNA.
Minimum acceptable stimulation was 8-fold.
Figure 1.
1 2 3 4
Results Comparison of pGem-luc transcript purification
methods. Transcripts were synthesized in 20 L
Aliquots of RNA transcript purified by different
reactions. After DNase I treatment, RNA was
methods (ultrafiltration, phenol extraction and LiCl purified from the reaction mix. 500 ng of
precipitation) were run on a denaturing agarose/ purified RNA were run on 1% agarose/
formaldehyde gel. Results are shown in Figure 1. formaldehyde gel.

The banding pattern of the 1.7 kb RNA transcripts Lane 1: 0.161.77 kb RNA ladder (Gibco BRL)
is identical regardless of purification method. Similar Lane 2: RNA purified in Microcon 30K NMWL
device
results were obtained in the case of capped
Lane 3: RNA purified by phenol extraction
transcript (results not shown).

P ro t o c o l s f o r Nuc l e ic Ac id s P u rif ic a t io n of I n Vi t ro Sy n t h e s i ze d mR N A
Lane 4: RNA purified by LiCl precipitation
The effect of increasing the mRNA concentration
on the translational efficiencies was examined.
At low mRNA levels, the capped luc mRNA was
translated three times more efficiently than the Figure 2.
uncapped mRNA (Figure 2). At higher mRNA levels,
140
the translation of both transcripts was comparable.
120
Similar behavior was observed with CAT mRNA3.
Even relatively high levels of mRNA did not cause 100
cpm x 104

the decrease in translational efficiencies noted by 80


other groups4. This result could be attributed in part 60
Capped pGem-luc RNA
to the lack of inhibitory contaminants in the mRNA pGem-luc RNA
40
preparation.
20
We also checked the effect of the RNA clean-up
0
method on in vitro translational efficiency. For details
0 0.5 1.0 1.5 2.0
of various procedures, see the Methods section. g RNA/50 L Reaction
RNA purified by each of the methods (1 g) was
Effect of pGem-luc RNA concentration on in vitro translation. Increasing
translated and analyzed. While there were no
amounts of uncapped Luc RNA transcripts and capped Luc RNA were used
observable differences between these RNAs by gel in translation reactions. Incorporation of 35S-methionine was determined
electrophoresis analysis (Figure 1), RNA purified by by TCA precipitation. Both RNA transcripts were purified with Microcon
ultrafiltration gave twice the translation efficiencies 30K NMWL devices.

of phenol-extracted RNA.

77
Effect of Centrifugal Ultrafiltration on
Large Fragment DNA Integrity
Introduction again with TE buffer (to 500 L or 2 mL, depending
on the device used) and spun once more, as
Centrifugal ultrafiltration provides a fast and easy
described above. The dilution step was repeated
method for the concentration and desalting of
a third time. After the third concentration spin, the
biological molecules. Previous reports document
retentate was collected by placing the sample
the use of ultrafiltration for desalting samples of
reservoirs upside down in new vials and spinning
nucleic acids1, removing excess primers from PCR2
d sid s E f f e c t of C e n t rif u g a l U l t r af i l t r a t io n o n L a r g e F r a g m e n t D N A In te g ri t y

the units for 1 minute at 1,000 x g.


reactions3, or concentrating DNA/RNA samples
The concentrated DNA was run on a 0.9%
without the need to use ethanol precipitation4.
SeaKem GTG agarose gel (FMC) and stained
Microcon filters offer a convenient, reproducible
with ethidium bromide to monitor sample integrity
means for centrifugal ultrafiltration of samples from
(Figure 1). DNA bands in lane 1 (starting material)
50 L to 2 mL. They insure high sample recovery
are indistinguishable from the bands on lanes 2 and
with their patented inverted recovery spin.
3, which were spun repeatedly in the ultrafiltration
The analysis of complex genomes depends on
device. There is no evidence that any supercoiled
the ability of the researcher to prepare pure, high
DNA was converted to the relaxed or linear forms
molecular weight DNA. When preparing high
during the concentration procedure, which would
molecular weight DNA samples for cosmid cloning
be the case had single-strand nicks been introduced
or other applications, it is important to treat the
during centrifugation.
DNA gently to avoid shearing or other damage to
the sample. As virtually all protocols for the prepa- Discrete Size Plasmids
ration of high molecular weight DNA require, at The next set of experiments monitored the effect
some point, buffer exchange and/or concentration, of centrifugal ultrafiltration on single population,
centrifugal ultrafiltration can provide an efficient discrete size plasmids. The plasmids used were
alternative to standard procedures. pBR322 (4,361 bp; New England Biolabs, Inc.),
To be useful, centrifugation must not cause pSPT18 (3,104 bp; Boehringer Mannheim) and
breakage of the DNA during the ultrafiltration pXTl (10,400 bp; Stratagene). Samples (1 g) of
procedure. This article summarizes the results of a each plasmid were spun in Microcon 30K NMWL
series of experiments designed to evaluate the effect units, as described previously. The starting material
of g forces on various samples of high molecular and retentates were run on a 1% agarose gel and
weight DNA during centrifugal ultrafiltration in stained with ethidium bromide.
Microcon units.
cl eli ce icA ciAc

Methods
Figure 1.
Nuc

1 2 3
DNA Ladder
P ro t o c o l s fo rNu

Supercoiled DNA Ladder.


As a model system to check for the introduction of
Lane 1: Starting material
single-strand nicks during the ultrafiltration spin,
Lanes 2, 3: Retentate from
supercoiled Ladder DNA (Gibco-BRL, Gaithersburg, DNA ladder spun
MD) which ranges in size from 2 to 8 kb was 3 times at 12,000 x g
in Microcon 30K
used. DNA diluted with TE buffer was loaded into NMWL device
Microcon 30K NMWL devices. The Microcon
units were spun for 7 minutes at 12,000 x g. The
concentrated DNA sample (retentate) was diluted

78
The results are shown in Figure 2. As in the
case of the DNA Ladder, the concentrated samples Figure 2.
1 2 3 4 5 6
(lanes 2, 4 and 6) appear to be identical with their Specific plasmid DNA
corresponding starting material. Again, there is no Lane 1: pBR322 starting material
evidence of conversion of the supercoiled form to Lane 2: pBR322 spun in Microcon
the relaxed form after exposure to g forces of 30K NMWL device
5,000 x g for 90 minutes and 12,000 x g for Lane 3: pSPT18 starting material

21 minutes during the concentration spins. Lane 4: pSPT18 spun in Microcon


30K NMWL device
Genomic Size DNA Lane 5: pXT1 starting material
Lane 6: pXT1 spun in Microcon
To monitor the integrity of molecules in the size
30K NMWL device
range of genomic DNA after centrifugation, lambda

P ro t o c o l s f o r Nuc l e ic Ac id s E f f e c t of C e n t rif u g a l U l t r af i l t r a t io n o n L a r g e F r a g m e n t D N A In te g ri t y
DNA (49 kb; Boehringer Mannheim, Indianapolis,
IN) and Bsu36 l digested BacPAK6 DNA (125 kb)
was used. The samples were diluted and centri- Figure 3.
fuged as described above. The retentates and 1 2 3 4
Large fragment DNA
starting material were run on a 1% agarose gel in
Lane 1: Lambda DNA starting material
a CHEF-DR II pulsed field electrophoresis system
Lane 2: Lambda DNA spun in
Bio-Rad, Richmond, CA). Electrophoresis was Microcon 30K NMWL device
performed at 200 V at 14 C in 0.5X TBE with Lane 3: BacPAK DNA starting material
ramped pulse from 1 to 6 seconds over 14 hours. Lane 4: BacPAK DNA spun in Microcon
The results with the lambda DNA mimic those 30K NMWL device
of the other samples run in these sets of experiments.
No adverse effects are noted after spinning the
DNA at g forces up to 12,000 x g in the ultrafiltra-
tion units. However, the larger BacPAK6 DNA
does show some degradation after ultrafiltration at
both 5,000 and 12,000 x g (Figure 3, smearing
in lane 4). Although a large percentage of the
sample appears to be intact, there was loss of
integrity of the BacPAK6 DNA sample after the
concentration procedure.

Conclusions References
DNA samples of up to 49 kb were concentrated 1. Takagi S, Kimura M, Katsuki M. BioTechniques
repeatedly without any loss of sample integrity. 1993;14(2):21821.
Some loss of integrity was observed with a 125 kb 2. PCR is covered by U.S. patents issued to
sample, although it was not complete and repre- Hoffmann-LaRoche, Inc.
sents a small percentage of the total DNA in the
3. Sheng N, Zhang J, Whitton JL, McKee T.
sample. For large fragments of DNA, centrifugal
BioTechniques 1993;14(5):7814.
ultrafiltration provides a fast and efficient method to
4. Ruano G, Pagliaro EM, Schwartz TR, Lamy K,
concentrate or desalt the sample. It results in high
Messina D, Gaensslen RE, Lee HC.
recovery of intact product.
BioTechniques 1992;13(2):26674.

79
DNA Extraction from Agarose Gels
with Montage Gel Extraction Kit or
Ultrafree-DA Centrifugal Filters
Introduction DNA prepared with the Ultrafree-DA centrifugal
filter requires no further purification for most applica-
The Ultrafree-DA device is designed to recover
tions, including cloning and radioisotopic or
100 to 10,000 bp DNA from agarose gel slices in
fluorescent DNA sequencing. Since agarose gel
one 10-minute spin. It consists of a pre-assembled
electrophoresis has high resolving power, the small
sample filter cup with an agarose gel nebulizer and
and large non-specific amplification products that
a microcentrifuge vial. The device uses gel com-
frequently interfere with cloning and sequencing
pression to extract DNA from the agarose.
after PCR (polymerase chain reaction) are com-
Centrifugal force collapses the gel structure, drives
pletely removed from the product.
the agarose through a small orifice in the gel
nebulizer and captures the resultant gel slurry in the
Materials
sample filter cup. As the agarose is compressed at
5,000 x g, DNA is extruded from the gels pores. Microcentrifuge
The gel matrix is retained by the microporous Pre-assembled Ultrafree-DA centrifugal filter
membrane, and the DNA passes freely through device or Montage Gel Extraction Kit
the membrane. DNA can then be recovered in
P ro t o c o l s f o r Nuc l e ic Ac id s D N A E x t r a c t io n f ro m A g a ro s e G e l s

Modified TAE* electrophoresis buffer (40 mM


the filtrate vial.
Tris-acetate, pH 8.0, 0.1 mM Na2EDTA)
The Montage Gel Extraction Kit consists of
50 Ultrafree-DA centrifugal filters as well as a SeaKem agarose (FMC BioProducts) or equivalent
modified TAE buffer that allows the casting and Long-wavelength UV lamp
running of the gel from which the DNA fragment
Scalpel or razor blade
is to be extracted.

*Modified TAE is recommended rather than TBE for the following reasons: (1) TBE buffer strongly inhibits DNA sequencing reac-
tions while modified TAE buffer does not. (2) Modified TAE has 0.1 mM Na2EDTA while regular TAE has 1.0 mM Na2EDTA. The
EDTA level at 0.1 mM Na2EDTA will not interfere with the magnesium concentration in sequencing reactions and other down-
stream enzymatic treatments, many of which are dependent on magnesium.

80
Procedure 4. Spin at 5,000 x g for 10 minutes. Centrifugation
forces the agarose through the gel nebulizer,
1. Electrophorese 30 L of PCR product or other
converting it to a fine slurry that is captured by
DNA through a <1.25% ordinary agarose gel,
the sample filter cup. Extruded DNA in electro-
prepared in modified TAE buffer with ethidium
phoresis buffer passes through the microporous
bromide (0.5 g/mL).
membrane in the sample filter cup and collects in
2. Locate the band of interest with a long wave- the filtrate vial.
length UV lamp or transilluminator. With a razor
5. DNA in the filtrate is now ready for sequencing
blade or scalpel, cut out the slice of agarose
or cloning without further purification. Discard the
(<100 L or 100 mg) containing the band
gel nebulizer and sample filter cup and store the
of interest. Trim any away excess agarose
DNA in the capped filtrate vial.
from band.
3. Place the gel slice into the gel nebulizer/sample Results
filter cup/filtrate vial assembly and seal the
Gel compression is a quick and easy technique for
device with the cap attached to vial.
recovering DNA from an agarose gel slice.

Table 1. Effect of gel disruption on typical DNA recoveries


from agarose gels
%DNA Recovered from
% DNA Recovered Gel Disrupted by Gel
DNA Size (bp) from Intact Gel Nebulizer
100 74 78

P ro t o c o l s f o r Nuc l e ic Ac id s D N A E x t r a c t io n f ro m A g a ro s e G e l s
400 39 ND
700 43 71
1000 55 77
2027 * 47
4361 14 35
9416 * 32
23130 * 29
* = Not detectable

81
PCR Purification with
Montage PCR Filter Units
Introduction
Figure 1.
After polymerase chain reaction (PCR)1, amplified
DNA must be separated from excess reaction Primer DNA
Salt dNTP
components that can interfere with subsequent
manipulations such as cloning or sequencing.
The Montage PCR filter unit is a single-use device
that simplifies the purification of PCR products.
It concentrates amplified DNA and removes primers
and unincorporated dNTPs, while providing
excellent capacity, high recovery, and high purity.
The method is quick and highly reproducible, which
makes it ideal for processing one-up or multiple Step 1 Step 2 Step 3
samples in middle-throughput operations.

Materials
Results
Variable speed microcentrifuge
Montage PCR filter units were used to purify 100 L
Montage PCR filter unit PCR reactions according to the specified protocol.
Purified water (such as Milli-Q water) or TE buffer After recovering the DNA fragments (n = 10) using
a reverse spin, the samples were separated by
PCR reaction (aqueous phase)
agarose gel electrophoresis. Recoveries of the
Method various PCR products were determined by densitom-
etry (Figure 1).
P ro t o c o l s f o r Nuc l e ic Ac id s P C R P u rif ic a t io n

1. Insert the Montage PCR sample reservoir into


The Montage PCR filter unit is a convenient
one of the two vials provided.
method for single-sample PCR purification. The
2. Fill the reservoir with 300 L purified water or high-performance device purifies PCR products
TE buffer. Add 100 L PCR reaction to the in a single centrifugation step. Purified samples
reservoir (step 1). Smaller volumes of PCR are ready for downstream applications with no
product may be used, but the volume should additional purification steps.
be adjusted to a final volume of 400 L.
3. Spin the Montage PCR device at 1000 x g References
for 15 minutes (step 2). NOTE: For optimal 1. PCR is covered by U.S. patents issued to
recovery, do not centrifuge longer than the Hoffmann-LaRoche, Inc.
specified 15 minutes or greater than 1000 x g.
4. To recover DNA, remove the sample reservoir
from filtrate collection vial and place in a
clean vial.
5. Add 20 L purified water or TE buffer to sample
reservoir.
6. Invert the reservoir and spin at 1000 x g for
2 minutes to retrieve the purified PCR (step 3).

82
Figure 2.
100%

80%
Percent Recovery

60%

40%

20%

0%
137 bp 301 bp 500 bp 657 bp 1159 bp

Purification of PCR Products Using Montage PCR Filters

Figure 3.

P ro t o c o l s f o r Nuc l e ic Ac id s P C R P u rif ic a t io n

Typical electropherogram shows an 1159 bp PCR product purified with a Montage PCR filter unit. Note the uniform signal intensity and
long read lengths. Purified samples are ready for cloning or sequencing with no additional purification steps.

83
Enzyme Removal with Micropure-EZ
Centrifugal Filters
Introduction (Figure 1) before centrifuging again. The purified
DNA is suitable for cloning or for other enzymatic
The Micropure-EZ device is a convenient device
manipulation.
for removing enzymes from double-stranded (ds)
While most enzymes were removed by the
DNA (20 bp to >50,000 bp) solutions in a single
Micropure-EZ device (Table 1, on page 86),
centrifugation. It can be used to remove enzymes
Millipore felt it was equally important to notify
whenever heat inactivation or phenol/chloroform
customers of several enzymes that were not
extraction is impossible or impractical. After an
removed (Table 2 , on page 86). Millipore scientists
enzyme reaction, the modified DNA solution
were very careful to rule out enzyme inhibition as a
containing dilute protein is simply pipetted into a
mechanism. The sensitivity of each assay to detect
Micropure-EZ device that has been inserted into a
trace amounts of restriction enzymes in Micropure-
vial (which is supplied) and centrifuged for 1 minute
EZ filtrates was determined by carrying out a
at 12,00014,000 x g. The DNA passes freely
dilution series (this is the method used by restriction
into the vial (typically with 8590% DNA recovery)
enzyme manufacturers to estimate total activity).
while the enzyme remains bound to the proprietary
membrane in the Micropure-EZ device. Removal is
Method
defined as the complete absence of detectable
In order to ensure that each enzyme assay was
enzyme activity or, as in the isolated case of T4
detecting trace amounts of restriction enzymes in
polynucleotide kinase, inconsequential residual
Micropure-EZ filtrates, serial dilutions of Bgl I
activity (<0.08%). For simultaneous concentration
(representing 2 units, 0.4 units, 0.08 units, and
of the enzyme-free DNA, place the Micropure-EZ
0.016 units of total activity in 10 L) were carried
device into a Microcon microconcentrator
out in a reaction mix* prepared with either sterile
DI water (sdH2O) or with sdH2O that had been
P ro t o c o l s f o r Nuc l e ic Ac id s E n z y m e Re m ova l

filtered through Micropure-EZ devices and then


Figure 1. incubated. Comparison of the resultant restriction
Remove Enzyme
Restriction and digests after agarose gel electrophoreses verified
other enzymes
DNA Enzyme Salt
Micropure-EZ are absorbed by that aqueous extractables from Micropure-EZ devices
Micropure-EZ.
dsDNA passes did not inhibit the enzyme. Also, these standard
freely. curves determined the sensitivity of the enzyme
Remove Enzyme
Restriction and assay (i.e., the theoretical amount of residual
other enzymes Concentrate DNA
DNA is retained by enzyme activity that could be detected, if present).
Micropure-EZ are absorbed by Microcon
Micropure-EZ. Assembly ultrafilter in Microcon. To stress test Micropure-EZ devices sufficiently
dsDNA passes Salts pass freely.
freely. with enzymes and DNA, several extreme operating
Vial conditions were used consistently. Micropure-EZ
Recover devices were challenged with 50 units of Bgl I in
85% DNA
the presence of decoy DNA (1 g of pBR322) and
5 g of bovine serum albumin (BSA). Devices were
spun at 14,000 x g for 30 seconds. The filtrates
30 seconds 3 minutes were then assayed for residual enzyme activity by
Operation of Micropure-EZ device adding 1 L of pUC19 DNA and 0.5 L of 100X
BSA to the filtrate, mixing thoroughly and incubating

*15.8 L of sdH2O, 2 L of 10X NEBuffer 2, 0.2 L of 100X BSA, and 2 L of Bgl I (10 units/L)

84
at 37 C for 1 hour. After a brief centrifugation to which was added directly to the filtrate and
collect the condensate at the bottom of the micro- incubated, appeared completely intact (lanes
centrifuge tubes, 1 L of 0.5 M disodium EDTA 1114). Since the standard curve indicated that
and 10 L of 5X loading buffer were added to 0.08 units would be detected if it were present,
stop the reaction. we calculated that at least 99.8% of Bgl I was
The negative control was a portion of DNA removed and/or inactivated. Removal, rather than
master mix. Control digests of pBR322 DNA and inactivation, is the probable mechanism by which
pUC19 DNA were carried out separately. Micropure-EZ devices operate, since in separate
experiments we were unable to detect any enzyme
Results and Discussion (irrespective of activity) in Micropure-EZ filtrates
The Bgl I standard curve made with the Micropure- using very sensitive HPLC methods (data not shown).
EZ-filtered sdH2O was indistinguishable from
the standard curve made with untreated sdH2O. Summary
The plasmid DNA (1 g of pUC19 and 1 g of Enzyme removal and excellent DNA recovery are
pBR322) was cut to completion by 2 units of Bgl I accomplished with Micropure-EZ devices in a single
after one hour at 37 C (Figure 2, lanes 1 and 6). 60-second spin without any pre-wetting, binding,
None of the enzymes shown in Tables 1 or 2 was washing or elution step. Cumbersome phenol/
measurably inhibited by Micropure-EZ-filtered chloroform extraction and the associated hazardous
sdH2O. The standard curves indicated that as little waste accumulation are avoided. An additional line
as 0.08 units of Bgl I could be detected by this of evidence for the suitability of Micropure-EZ
assay (lanes 3 and 8). devices in molecular biology applications is that
The devices removed the 50 units of Bgl I as restriction digest purified with Micropure-EZ devices
evidenced by an absence of detectable activity in alone are readily ligated and cloned into plasmid
the filtrates. In the lanes corresponding to the Bgl I DNA (M.H.A.L., unpublished observations).
challenged Micropure-EZ inserts, some activity was Micropure-EZ devices used alone or with Microcon
observed against the decoy pBR322 DNA, as devices permit rapid and efficient sequential
expected during its brief exposure to Bgl I (particu- enzymatic DNA manipulations.
larly evident in lane 14). However, the pUC19,

P ro t o c o l s f o r Nuc l e ic Ac id s E n z y m e Re m ova l
Figure 2.
Analytical agarose gel, demonstrating the detection limits for Bgl I and Bgl I removal by Micropure-EZ devices.

Lanes 14: Serial dilutions of BgI I representing 2 units, 0.4 units, 0.08 units, and 0.016 units of activity (respectively)
against the DNA master mix prepared in non-filtered sdH2O
Lane 5: Undigested DNA master mix used as substrate for the standard curve containing pUC19 and pBR322 DNA
Lanes 69: Inhibition control: serial dilutions of BgI I representing 2 units, 0.,4 units, 0.08 units, and 0.016 units of activity (respectively)
against the DNA master mix prepared in Micropure-EZ-filtered sdH2O
Lane 10: Molecular weight standard: 1 kb DNA ladder (Gibco-BRL, Gaithersburg, MD)
Lanes 1114: Filtrates incubated with pUC19 DNA after challenging Micropure-EZ devices with 50 units of BgI I, BSA and
decoy pBR322 DNA
Lane 15: Uncut pBR322
Lane 16: BgI Icut pBR322
Lane 17: Uncut pUC19
Lane 18: BgI Icut pUC19

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18

85
Table 1. Enzymes removed by Micropure-EZ devices

Enzyme Challenge Enzyme Challenge Enzyme Challenge


AMV reverse transcriptase 50 U Bgl I 50 U Nru I 50 U
Calf intestinal alkaline 10 U BsiW I 70 U Pst I 100 U
phosphatase BssH II 20 U Sac I 100 U
DNase I (bovine pancreas) 10 U BstN I 50 U Sac II 100 U
Exonuclease III (E. coli) 100 U Dpn I 100 U Sal I 100 U
MMLV reverse 600 U EcoR I 100 U Sca I 50 U
transcriptase
Hae III 100 U Sph I 25 U
Mung bean nuclease 50 U
Hinc II 50 U Sst I 50 U
Proteinase K (Amresco) 5 g
Hind III 100 U Xho I 100 U
T4 DNA ligase 2,000 U*
Hpa I 25 U In tests, all enzymes were removed
T4 DNA polymerase 15 U
Kpn I 50 U from solutions containing DNA or
T4 polynucleotide kinase** 50 U RNA (in the case of RNase A). Five
Mbo I 25 U
Taq DNA polymerase 5U g of bovine serum albumin were
Mlu I 50 U also present during removal of restric-
Terminal deoxynucleotidyl 45 U
Nco I 50 U tion enzymes. Removal was indicated
transferase by undetectable or inconsequential
Nde I 100 U
Acc I 50 U enzyme activity in filtrate (<0.08%
NgoM I 50 U residual in the case of T4 polynucleo-
Apa I 100 U
Nhe I 25 U tide kinase).
BamH I 100 U
Not I 50 U
Bcl I 50 U

*New England Biolabs unit definition


**T4 polynucelotide kinase is not recommended with oligonucleotides (dsDNA only). The results suggest that
this kinase mediates the binding of oligos to the membrane in Micropure-EZ device, causing oligo loss.

Table 2. Enzymes not removed by Micropure-EZ devices


d sid s E n z y m e Re m ova l

Enzyme Challenge Enzyme Challenge


Bacterial alkaline 0.6 U Eae I 15 U
phosphatase EcoR V 50 U
DNA polymerase I 20 U Hinf I 50 U
(Klenow)
Msp I 100 U
Exonuclease I 50 U
Pvu I 25 U
Pfu DNA polymerase 2.5 U
Pvu II 50 U
Vent DNA polymerase 4U
Sau3A I 20 U
cl eli ce icA ciAc

Shrimp alkaline 1U
Sfi I 50 U
phosphatase
Sma I 25 U
Nuc

RNase A (bovine 1 g
P ro t o c o l s fo rNu

pancreas) Xba I 50 U
ApaL I 10 U In tests, Micropure-EZ devices did
Bgl II 50 U not remove the indicated number of
units of the listed enzymes. It may be
BsoB I 50 U effective in removing a lower number
Cla I 25 U of units.

*T4 RNA ligase is not recommended. Results suggest this ligase mediates binding of
nucleic acids to the membrane in Micropure-EZ devices, causing sample loss.

86
Protocols for
Concentration of Bacteriophage
Using Ultrafiltration . . . . . . . . . . . . . . . . . . . . . 88

Virus Concentration
Concentration of Animal Viruses
Using Ultrafiltration . . . . . . . . . . . . . . . . . . . . . 91

P ro t o c o l s f o r V iru s C ONC EN T RAT ION

87
Concentration of Bacteriophage
Using Ultrafiltration
Introduction Ultrafiltration is an alternative method to achieve
high bacteriophage titer. When complete purifica-
Bacteriophages were first discovered nearly a
tion from cell and media proteins is not required,
century ago1. Since then, phage biology has
phage can be quickly concentrated using centrifu-
drawn researchers attention because of their
gal ultrafiltration9. Ultrafiltration can also be used to
potent bactericidal capacity that can be used to
concentrate phage from environmental sources10.
treat human infections2. Bacteriophage and animal
For larger volume lysates, recirculating tangential
viruses have been successfully utilized as DNA
flow filtration (TFF) is a better choice11,12. Correct
carriers, or cloning vectors. The advantage to
membrane material, pore size, centrifugation speed
P ro t o c o l s f o r V iru s C ONC EN T RAT ION C o n ce n t r a t io n of B a c te rio p h a g e U s i n g U l t r af i l t ra t i o n

lambda-based vectors is that they can carry


and final retentate volume are critical to obtaining
fragments of DNA up to 25,000 bp. Another
high phage recovery and maintaining its infectivity.
application, phage display, is a novel genetic
This protocol describes the use of Millipore
selection system for cloning proteins from cDNA
ultrafiltration devices to concentrate Pseudomonas
libraries. In addition, there are many similarities
pseudoalcaligenes bacteriophage phi-6, an
between bacteriophages and animal cell viruses.
enveloped dsRNA virus. The virions are spherical
Thus, bacteriophages can be viewed as model
and 86 nm in diameter.
systems for animal cell viruses.
The standard bacteriophage propagation
Method
protocol normally involves infection of the host
bacterial strain, 1216 hours of bacterial growth, Phi-6 production
followed by the phage recovery from the superna- Note: All manipulations should be performed using
tant. Purification of phage usually includes a aseptic technique in a laminar flow hood.
density gradient centrifugation, followed by 1. Grow fresh culture of Pseudomonas pseudoalca-
extensive dialysis3,4. Phage titers are then deter- ligenes until it reaches the desired optical density
mined by plaque assays. of 0.1 absorbance units at 600 nm.
For some experiments, high titer bacteriophage
2. Inoculate the culture with enough phi-6 to
stock is desired. Traditionally, phage particles were
achieve a multiplicity of infection (MOI) of 0.5.
concentrated by polyeteylene glycol (PEG) and
3. Return the phi-6 containing Pseudomonas
dextran sulfate precipitation, precipitation by acid,
pseudoalcaligenes culture to the platform
or differential or equilibrium centrifugation5. Recently,
shaker and incubate at 26 C, 125 RPM,
different modifications of the Sambrook method are
for 1824 hours.
being used6. Phage is concentrated by precipitation
with sodium chloride and PEG and resuspended 4. After the overnight incubation, aliquot the culture
to the desired titer. The process requires 612 into four 250 mL centrifuge bottles.
hours of incubation with PEG and a lengthy pellet 5. Centrifuge the bottles for 30 minutes at
resuspension process to achieve high particle 16,000 x g.
recovery 7,8. 6. Remove the supernatant and transfer to a
sterile container.

88
Concentration using Amicon Ultra-4 or Ultra-15 6. Set retentate pressure by slowly adjusting
devices with 50 kDa NMWL membrane retentate valve clockwise until retentate
1. Clarify phi-6 supernatant using a Stericup pressure is 10 psi.
filter unit (Millipore cat. no. SCGP U05 RE), 7. Adjust pump speed and retentate valve
a Steriflip filter unit (Millipore cat. no. restriction to achieve desired feed pressure
SCGP 005 25), or a Millex-GP syringe filter of 30 psi and retentate pressure of 10 psi.
(Millipore cat. no. SLGP 033 RB) with low- Note: Do not exceed feed pressure of 60 psi.
binding 0.22 m PES membrane.
8. Filter the solution until the desired concentration
2. Load appropriate volume of clarified phi-6
or volume is obtained.
supernatant (4 or 15 mL) into an Amicon Ultra-4
9. Stop pump.
device (Millipore cat. no. UFC8 050 24) or
an Amicon Ultra-15 device (Millipore cat. no. 10. Disconnect pump outlet tubing from pump
UFC9 050 24) containing 50,000 NMWL outlet port and place in product recovery
regenerated cellulose membrane. collection vessel.

P ro t o c o l s f o r V iru s C ONC EN T RAT ION C o n ce n t r a t io n of B a c te rio p h a g e U s i n g U l t r af i l t ra t i o n


3. Centrifuge the device (4 or 15 mL) for 15 11. Disconnect retentate tubing from retentate
minutes at 1500 g. in port. Fluid should now drain by gravity.
If additional drainage is required, a syringe
4. The recoverable volume should be 80100 L
can be placed on the end of the retentate
for Amicon Ultra-4 devices and 350500 L
tube and fluid can be blown down.
for Amicon Ultra-15 devices.
12. Replace retentate tubing in retentate port.
Note: Avoid concentrating samples below Reconnect pump outlet tubing.
recommended volumes as this may lead to
13. Disconnect FEED IN tubing and place in
decreased phage recovery.
collection vessel. Open tank outlet valve,
Concentration using Pellicon XL50 device turn pump speed up to drain reservoir.
with PLCTK membrane and Millipores 14. Reconnect the pump outlet tubing to the
Labscale TFF system FEED IN port.
Note: The Labscale TFF system (Millipore cat. no.
Results
XX42 LSS 13) is not required to use Pellicon XL50.
A peristaltic pump may alternatively be used Millipore ultrafiltration devices with Ultracel regener-
(see Millipore user guide P60085). ated cellulose membrane offer low non-specific
binding and high retentate recovery. The vertical
1. Perform flush, integrity, and permeability tests
membrane reduces concentration polarization for
on Pellicon XL50 device with PLCTK membrane
ultra-fast concentration times and is especially
(Millipore cat. no. PXCO 30C 50) as indicated
beneficial for turbid and viscous solutions.
in the Pellicon XL50 User Guide.
As shown in the protocol above, phage titer
2. Remove reservoir cover and load up to 500 mL can be increased up to 30-fold in 15 minutes by
phi-6 supernatant into reservoir of Labscale centrifugal ultrafiltration for under 15 mL volumes.
TFF system. Amicon Ultra-4 and Ultra-15 devices with 50 kDa
3. Ensure vent port is open by removing plug from NMWL membranes can be used successfully to
VENT port and leaving open or installing a concentrate the phage phi-6 out of bacterial culture
Millex filter. supernatant. The same devices with 100 kDa
4. Open tank outlet valve. NMWL membranes are not recommended because
5. Turn on pump and increase speed until feed they lead to a low virus recovery and allow virus to
pressure reaches 20 psi. break through into the filtrate.

Note: Check system connections for leaks.


Tighten connections as required.

89
For large volume concentration, tangential flow 5. Bachrach U, Friedmann A. Appl Microbiol
filtration (TFF) should be used. Table 1 shows phage 1971;22(4): 706-715.
concentration results using a Pellicon XL50 cassette 6. Sambrook J, Fritsch EF, Maniatis T. Molecular
with the Labscale TFF system. The Pellicon XL50 cloning: a laboratory manual, 2nd ed. Cold
cassette couples Millipores ultrafiltration membranes Spring Harbor Laboratory Press, Cold Spring
with the linearly scalable TFF cassette for processing Harbor, N.Y. 1989
501000 mL volumes. The Ultracel membrane with 7. Bahns JT, Liu CM, Chen L. Protein Science
30 kDa NMWL is recommended. Twenty-fold 2004;13:2578-2587.
concentration of phage solution is usually achieved
8. Gill JJ, et al. Chemother 2006;50(9):
in 3050 minutes.
2912-2918.

References 9. Meile L, Abendschein P, Leisinger T.


J Bacteriol 1990;172(6):3507-3508.
1. Pennazio S. Riv Biol 2006;99(1):103-29.
10. Middelboe M, et al. Aquat Microb Ecol
2. Fischetti VA, Nelson D, Schuch R. Nature
2003;33:1-10.
P ro t o c o l s f o r V iru s C ONC EN T RAT ION C o n ce n t r a t io n of B a c te rio p h a g e U s i n g U l t r af i l t ra t i o n

Biotechnology 2006;24:1508-1511.
11. Rembhotkar GW, Khatri GS. Anal Biochem
3. Hansen MR, Mueller L, Pardi A. Nature
1989;176(2):373-4.
Structural Biology 1998;5:1065-1074.
12. Alonso MC, Rodriguez J, Borrego J. J Internatl
4. Zhilenkov EL, et al. Virology Journal
Microbiol 1999;2:227-232.
2006;3:50-55.

Table 1. Bacteriophage phi-6 concentration using Millipore ultrafiltration devices


Amicon Ultra-4, Amicon Ultra-4, Amicon Ultra-15, Pellicon XL50 TFF,
100 kDa 50 kDa 50 kDa 30 kDa
Initial volume (mL) 4 4 15 500
Initial viral titer, pFU/mL 2.3 x 10 10
2 x 10 10
2.02 x 10 10
1.9 x 107
Final volume (mL) 0.1 0.5 0.5 25
Final viral titer, pFU/mL 2.06 x 10 11
3.7 x 10 11
7.8 x 10 11
5.6 x 108
Concentration factor 40 8 30 20
Phage recovery (%)* 22 >100 >100 >100
Phage in the filtrate, pFU/mL 2.3 x 105 <100 <100 7 x 103
*We consistently observe higher than 100% recovery after phage concentration. This can be explained by increased infectivity of phage particles at
higher concentrations and agrees with other published data12.

90
Concentration of Animal Viruses
Using Ultrafiltration
Introduction and adeno-associated virus (AAV)10. Challenges
associated with the use of viral vectors in clinical
Despite the rapid progress in genomics, the biologi-
trials include production of sufficient quantities of
cal function of the majority of the human genes is
clinical grade material and maintaining biosafety of
still unknown. The identification of gene function is
the viral vector. Production and purification methods
an increasingly relevant issue, especially in the
for viral vectors vary both in terms of methodology
search for new targets for improved human disease
and virus yield and quality.
therapy. Functional genomics approaches, which
Generation and purification of viral vectors can
aim at the identification of genes via phenotypes
be labor intensive, costly and may require special

P ro t o c o l s f o r V iru s C ONC EN T RAT ION C o n ce n t r a t io n of A n i m a l V i ru s e s U s i n g U l t r af i l t ra t i o n


induced in biological systems, require measurement
equipment (such as ultracentrifuges). The standard
of gene function on the genomic scale in cell-based
method for purification of adenovirus, lentivirus
assays. cDNA expression libraries representing the
and AAV uses density gradient ultracentrifugation
population of expressed genes in a given cell/tissue
followed by extensive dialysis. More recently,
type have classically been cloned into plasmids,
column or membrane chromatographies have
which can be introduced into cells by transient
become the methods of choice for adenovirus
transfection. However, transfection efficiency varies
purification11,12. The yield of virus normally is
widely between cell types. Therefore, a variety of
determined by plaque assay, TCID50 assay and
virus-derived vector systems have been developed
more recently by ELISA assay. For most applications,
for improved cDNA transduction, expression, and
recombinant virus must be purified to a high titer.
screening in mammalian cells. These systems include
In order to achieve high titer stock, it is usually
simian virus 40 replicons in COS cells1,2 and
necessary to concentrate purified virus particles.
cDNA cloning and expression vectors derived
This Protocol describes the use of Millipore centrifu-
from retrovirus3,4 baculovirus5, alphavirus6, human
gal ultrafiltration devices for concentration of three
immunodeficiency virus7, vaccinia virus8, adenovirus9
most common virus types: lentivirus, adenovirus and
adeno-associated virus (AAV).
Figure 1. Primary culture of rat cortical
neurons infected with lentivirus Method
1. Packaging cells: Host cells are plated to achieve
8085% confluence
2. Transfection: Cells are either transfected with
recombinant plasmid(s), or infected with viable
virus particles.
3. Harvesting virus: Depending on the virus, either
cell supernatant or pelleted cells, or both, are
used to purify the recombinant virions. Three to
For virus propagation, HEK-293T cells were trans- four rounds of freeze/thaw cycles are usually
fected with helper plasmids and lentiviral plasmid
applied to release virus from cells. Cell debris is
expressing copGFP (System Bioscience). 48 hours
after transfection viral supernatant was collected then separated from cell lysate by centrifugation.
and 10 mL was concentrated using Amicon Ultra-15 4. Virus purification: Virus is purified from cell
100 kDa MWCO membrane to 200 L. 50 L of
lysate by density gradient ultracentrifugation or
concentrated viral stock was used for infection of
10E6 cells. column/membrane chromatography. In some
Data courtesy of Drs. Leila Aminova and Ambreena Siddiq, cases, affinity chromatography with heparin
Neuroprotection Laboratory of Beth Israel Medical Center. or mucin sepharose13,14,15 can be used.

91
Purified virus solutions obtained via these methods Table 1 shows typical results of concentration of
are then buffer exchanged using either passive adenovirus, lentivirus and AAV from both crude cell
dialysis or diafiltration into an appropriate storage lysate and virus purified by chromatography.
solution. It is important to remember that most of the viruses
cannot be efficiently concentrated and stored in
Virus Concentration
low salt buffers. Centrifugal concentration of the
In order to achieve a high titer of viral stock, the virus in <200 mM salt solutions will lead to virus
purified virus can be concentrated using centrifugal aggregation and decreased infectivity. An example
ultrafiltration. The correct choice of device, mem- of a recommended storage solution is 20 mM Tris,
brane material, MWCO, centrifugation speed, pH 8.0, 250 mM NaCl, 10 mM MgCl2,
centrifugation time and buffer composition are 5% sorbitol and 0.001% PF68 (pluronic acid)16.
critical for high recovery of infective viral particles. Concentrated viral stock can also be filter
Amicon Ultra centrifugal ultrafiltration devices with sterilized through 0.22 m filters. For example,
a 50 KDa NMWL Ultracel regenerated cellulose Steriflip-GP filter units (cat. no. SCGP 005 25)
membrane have been demonstrated to successfully
P ro t o c o l s f o r V iru s C ONC EN T RAT ION C o n ce n t r a t io n of A n i m a l V i ru s e s U s i n g U l t r af i l t ra t i o n

can be used for volumes of 50 mL or less or


concentrate adenovirus and AAV solutions. For Ultrafree-MC GV (cat. no. UFC3 0GV 0S)
lentivirus concentration, devices with both 50 and can be used for volumes of <0.5 mL.
100 kDa membranes can be used.

Table 1. Concentration of virus using Millipore ultrafiltration devices


Starting Starting Final Spin Time at Recovery Concentration
Starting solution Device Virus Titer Volume (mL) volume (L) 1500 x g (min) (%)* Factor
Adenovirus in crude Amicon Ultra-4, 1 x 108 4 150 20 >100 27X
cell lysate 50 kDa MW
Adenovirus purified, Amicon Ultra-4, 1 x 108 4 80 10 85 45X
in 1M salt buffer 50 kDa MW
AAV in crude Amicon Ultra-4, 2 x 107 4 200 35 50 20X
cell lysate 50 kDa MW
AAV in crude Amicon Ultra-15, 2 x 107 10 400 45 76 25X
cell lysate, diluted 50 kDa MW
in 100 mM Tris,
200 mM NaCl buffer
AAV in crude Amicon Ultra-4, 0.5 x 107 2 200 20 100 20X
cell lysate, diluted 50 kDa MW
in 100 mM Tris,
200 mM NaCl buffer
Lentivirus in cell Amicon Ultra-4, 9 x 104 4 40 30 >100 100X
culture supernatant 100 kDa MW
Lentivirus in cell Amicon Ultra-4, 9 x 104 4 40 30 93 100X
culture supernatant 50 kDa MW
Lentivirus in cell Centricon 70 Plus, 8 x 103 65 800 30 81 80X
culture supernatant 100 KDa MW
*We do not recommend concentrating the virus to higher than 1013 particles/mL due to potential virus aggregation17.

92
References
1. Gearing DP, et al. EMBO J 1989;8:3667.
2. Yokota T, et al. Proc Natl Acad Sci 1984;81:
1070.
3. Kitamura T, et al. Proc Natl Acad Sci 1995;
92:9146.
4. Shaughnessy L, et al. J Mol Neurosci 2004;
24:23.
5. Granziero L, et al. J Immunol Methods 1997;
203:131.
6. Koller D, et al. Nat Biotechnol 2001;19:851.
7. Van Maanen M, et al. Mol Ther 2003;8:167.
8. Smith ES, et al. Methods Mol Biol 2004;

P ro t o c o l s f o r V iru s C ONC EN T RAT ION C o n ce n t r a t io n of A n i m a l V i ru s e s U s i n g U l t r af i l t ra t i o n


269:65.
9. Elahi SM, et al. Gene Ther 2002;9:1238.
10. Choi VW, et al. Curr Gene Ther 2005;
5:299.
11. Blanche F, et al. Gene Therapy 2000;7:
1055-1062.
12. Shabram PW, et al. Hum Gene Therapy 1997;
9:453-465.
13. Clark KR, et al. Hum Gene Therapy 1999;
10:1031-1039.
14. Zolotukhin S, et al. Gene Therapy 1999;6:
973-985.
15. Auricchio A, et al. Hum Gene Therapy 2001;
12:71-76.
16. Alejandra E, Arbetman, AE, et al. Journal of
Virology 2005;79(24):15238-15245.
17. Galdiero F. Arch Virol. 1979;59(1-2):99-105.

93
High Throughput Applications
The MultiScreen filter plate with Ultracel-10 Purification of Serum Peptides for
membrane provides a new method for high Biomarkers Research
throughput sample prep. The ultrafiltration-based AN2010EN00
filter plate is designed for use with centrifugation This study is a high throughput version of the
and is compatible with standard microtiter plates, application note on page 42 of the Protocols
instrumentation, and liquid handling equipment. section of this handbook.
All of the publications summarized below
can be supplied by your local Millipore office or Enzymatic Activity Recovery
downloaded from www.millipore.com/ultracel. AN2011EN00
This study demonstrates the use of the MultiScreen
Nucleic Acid Purification and Concentration plate with Ultracel-10 membrane for concentrating
AN1040EN00 alkaline phosphatase without loss of biological
The MultiScreen Filter Plate with Ultracel-10 activity and with high reproducibility across
membrane can be used to purify and concentrate the plate.
single-stranded oligonucleotides and double-
stranded DNA fragments, as well as plasmid
and genomic DNA.

Concentration of Proteins in Cell Lysate


AN1424EN00
The MultiScreen filter plate with Ultracel-10
membrane can be used to concentrate whole
cell lysates without loss of protein and with high
reproducibility across the plate. Applications include
parallel protein purification, protein concentration
and buffer exchange in cell lysates for subsequent
separation or assaying.
App e nd i x H i g h T h ro u g h p u t A p p l i c a t io n s

MultiScreen Filter Plate with Ultracel-10 ultrafiltration


membrane

94
Glossary
Asymmetric Membrane1 Fouling Reverse Osmosis1
Membrane constituted of two or more Irreversible decline in membrane flux due to Liquid-phase pressure-driven separation
structural planes of non-identical deposition and accumulation of submicron process in which applied transmembrane
morphologies. particles and solutes on the membrane pressure causes selective movement of
surface. Also, crystallization and precipita- solvent against its osmotic pressure
Batch Process
tion of small solutes on the surface and in difference.
A fixed volume of solution contained in a
the pores of the membrane. Not to be
tank to which the concentrate is returned Tangential Flow Filtration (TFF)
confused with concentration polarization.
during the process. Flow through a membrane device in which
Membrane1 the fluid on the upstream side moves
Composite Membrane1
Structure having lateral dimensions much parallel to the membrane surface.
Membrane having chemically or structurally
greater than its thickness, through which
distinct layers. Transmembrane Pressure (TMP)
mass transfer may occur under a variety
The driving force in ultrafiltration. In a stirred
Concentration Polarization of driving forces.
cell, equivalent to gas pressure. In centrifu-
Accumulation of rejected solute on the
Molecular Weight Cut-off (MWCO) gal devices, it is related to g-force. In a
membrane surface. Depends on interactions
See Nominal Molecular Weight Limit. flowing system, TMP decreases as the
of pressure, viscosity, crossflow (tangential)
stream moves from inlet to outlet. Average
velocity, fluid flow conditions, flow channel Nanofiltration1
TMP = [(Pin + Pout)/2] Ppermeate
conditions and temperatures. Pressure-driven membrane-based separation
process in which particles and dissolved Ultrafiltration1
Crossflow (Tangential Flow)
molecules smaller than about 2 nm are Pressure-driven membrane-based separation
Solution flows across (tangential to) a
rejected. process in which particles and dissolved
membrane surface. Facilitates back diffusion
molecules smaller than 0.1 m and larger
of solute from that surface into the bulk Nucleotide Cut-off (NCO)
than about 2 nm are rejected.
solution, counteracting concentration The number of nucleotides in a DNA
polarization. fragment (single- or double-stranded) at Yield
which 90% of the fragment is retained by Amount of species recovered at the end of
Diafiltration
the membrane. the process as a percentage of the amount
Removal of small components from the
present in the feed solution.
retained species during ultrafiltration by Nominal Molecular Weight Limit (NMWL)
adding water or buffer solution to the The molecular weight at which at least 90%
retentate. See page 7 for further discussion. of a globular solute of that MW is retained References
by the membrane. 1. Terminology for membranes and
Dialysis
Diffusive transport of ions or other small Permeate (Filtrate, Ultrafiltrate) membrane processes (IUPAC
molecules through a membrane barrier The solution passing through the membrane, Recommendations 1996). IUPAC,
that contacts solvent on both sides. containing solvent and solutes not retained Journal
by the membrane. of Membrane Science.1996;120(1):
Downstream1 14959.
Side of a membrane from which the Plugging
permeate emerges. Accumulation of debris on the fluid flow
path, restricting or blocking flow.
Feed (Sample)
The starting solution (sometimes the solution Rejection App e nd i x G l o s s a r y
remaining upstream of the membrane). The fraction of solute held back by the
membrane. Can be measured at any point
Fluid Velocity
in the process or averaged over the run.
The flow rate of solution across the
membrane surface in cross (tangential) flow. Retentate (Reject Stream, Concentrate)
Related to hydraulic pressure drop. The solution containing the retained
(rejected) species.
Flux
The filtration rate through the membrane Retention Factor1 (rF)
per unit area. Parameter defined as one minus the ratio
of permeate concentration to the retentate
concentration of a component. Note:
rF = 1 [p]/[r] where [p] = concentration
of solute in permeate, and [r] = concentra-
tion of solute in retentate.

95
Acknowledgements
Millipore also wishes to acknowledge the following
individuals for their contributions:

Eduardo Vottero
University of British Columbia

Peter A. Lemaire and Dr. James Cole


University of Connecticut

Gary Smejkal
Proteome Systems
Woburn, MA

Leonid Kryazhev
Genome Quebec
Montreal, Canada

Mathew L. Thakur
Thomas Jefferson University Hospital
Philadelphia, PA

Mark Merchant
Helena Laboratories
Beaumont, TX

Department of Forensic Biology


App e nd i x

New York City

Patrick O. Brown, Max Diehn, Ash Alizadeh


Stanford University School of Medicine

Dr. Sophia N. Karagiannis


GKT School of Biomedical Sciences
London

96
Patent
PCR is covered by US patents issued to Hoffmann-LaRoche, Inc.

Trademarks
Millipore, Amicon, Biomax, Centricon, Centriprep, Durapore, Immobilon, Labscale, Microcon,
Micropure, Milli-Q, Minicon, Montage, MultiScreen, Pellicon, Prep/Scale, PROSEP, Steriflip,
Ultracel, Ultrafree, Zip, and ZipTip are trademarks of Millipore Corporation.
AmpFLSTR, BigDye and Voyager-DE are trademarks of Applera Corporation or a subsidiary.
BacPAK and Talon are trademarks of Becton, Dickinson and Company.
Biofuge is a trademark of Kendro Laboratory Products, Gmbh.
CHEF-DR and Chelex are trademarks of Bio-Rad Laboratories.
DryStrips, Sephacryl, Sepharose, and Storm are trademarks of GE Healthcare.
GenePrint, Flexi and Riboprobe are trademarks of Promega Corporation.
Kodak is a trademark of Eastman Kodak Company.
MEGAscript is a trademark of Ambion Inc.
NuPage, SimplyBlue, Superscript and Xcell Surelock are trademarks of Invitrogen Corporation.
ProteoPrep is a trademark of Sigma-Aldrich Biotechnology LP.
ProteomIQ, ElectrophoretIQ, IsoelectrIQ, and GelChips are trademarks of Proteome Systems.
SeaKem is a trademark of FMC.
SpectraFLUOR is a trademark of Tecan Group AG.
SpectraMax is a trademark of Molecular Devices Corporation.
Speed Vac is a trademark of Thermo Savant.
SureBlue is a trademark of Kirkegaard & Perry Laboratories, Inc.
SYBR is a trademark of Molecular Probes, Inc.
Triton is a trademark of Union Carbide Chemicals & Plastics Technology Corporation.
Tween is a trademark of Atlas Powder Company.
Vent is a trademark of New England Biolabs, Inc.
Additional Resources for Life Scientists

Ultrafiltration Application and Product Guide


Cell Biology at Millipore. Filter with Millipore for
Visit Millipores new Fast, High-Quality Results.
home for Cell Biology Millipore offers
research. You can hundreds of mem
easily access educa brane-based devices
tional content, labora for sterile filtration, antibodies forensics
tory protocols, literature chromatography,
and troubleshooting tips sample preparation,
along with many other and almost any other
scientific resources. Our application in the life proteins
intuitive cell biology site
lets you navigate, review
sciences laboratory.
To request a copy of the
serum
and purchase directly online thousands of Millipore Analytical Sample Preparation
products across many cell biology workflows. and General Filtration catalogue, visit
Visit www.millipore.com/cellbiology. www.millipore.com/source4filters.
enzymes viruses
Discover Our New Range of Laboratory Water Systems
Immunodetection Tools. Millipore provides total solutions from bench-top
Easily access
educational content,
systems to custom-engineered purification chains
for laboratory buildings. Youll find Millipore water
nucleic acids
laboratory protocols, systems installed in over 70,000 laboratories world-
literature and trouble wide supplying pure water for electrophoresis, PCR,
shooting tips along chromatography and other life science applications.
with many other To learn more, visit www.millipore.com
scientific resources.
You can also browse
and purchase online
Technical Support
thousands of immuno
Millipore Technical Service Specialists support
detection products, including more than
ultrafiltration and many other life science
10,000 antibodies formerly sold by Chemicon

Ultrafiltration
applications, including blotting, DNA
and Upstate, which are now part of Millipore.
sequencing sample preparation, sterile
Visit www.millipore.com/immunodetection.
filtration, and MS sample prep. To contact a
Specialist, call your local office or submit a
question at www.millipore.com/techservice.
To access our library of frequently asked
questions, go to www.millipore.com/faqs.
Application and Product Guide

Lit. No. TP0040EN00 Rev. C 05/08 Printed in U.S.A. BS GEN-08-00329


www.millipore.com
2008 Millipore Corporation, Billerica, MA 01821 U.S.A. All rights reserved.

Potrebbero piacerti anche