Sei sulla pagina 1di 41

C H A P T E R

11
(Strept)avidin–Biotin Systems

One of the most popular methods of noncovalent site for biotin, or vitamin H, and one oligosaccharide
conjugation is to make use of the natural strong bind- modification (Asn-linked). The tetrameric protein is
ing of (strept)avidin for the small molecule biotin. The highly basic, having a pI of about 10. The biotin inter-
strength of the (strept)avidin–biotin interaction has action with avidin is among the strongest noncovalent
made it a useful tool in specific targeting applications affinities known, exhibiting a dissociation constant of
and assay design. Since each (strept)avidin molecule about 1.3 × 10−15 M. Tryptophan and lysine residues in
contains a maximum of four biotin-binding sites, the each subunit are known to be involved in forming the
interaction can be used to enhance the signal strength in binding pocket (Gitlin et al., 1987, 1988).
immunoassay systems. The tetrameric native structure of avidin is resistant
Modification reagents that can add a functional bio- to denaturation under extreme chaotropic conditions.
tin group to proteins, nucleic acids, and other molecules Even in 8-M urea or 3-M guanidine hydrochloride
now come in many shapes and reactivities (Section 6, the protein maintains structural integrity and activity
this chapter, and Chapter  18, Section 1.3). Depending (Green, 1963). When biotin is bound to avidin, the inter-
on the functionality present on the biotinylation com- action promotes even greater stability to the complex.
pound, specific reactive groups on antibodies or other An avidin–biotin complex (ABC) is resistant to break-
proteins may be modified to create a (strept)avidin down in the presence of up to 8-M guanidine at pH 5.2.
binding site. Amines, carboxylates, sulfhydryls, and A minimum of 6- to 8-M guanidine at pH 1.5 is required
carbohydrate groups can be specifically targeted for bio- for inducing complete dissociation of the avidin–biotin
tinylation through the appropriate choice of biotin deriv- interaction complex (Cuatrecasas and Wilchek, 1968;
ative. In addition, photoreactive biotinylation reagents Bodanszky and Bodanszky, 1970). Since the subunits
(Section 6.5, this chapter) are used to add nonselectively in avidin are not held together by disulfide bonds, con-
a biotin group to molecules containing no convenient ditions that cause denaturation also result in subunit
functional groups for modification. In this manner, oligo- disassociation.
nucleotide probes often are modified for detection with The strength of the noncovalent avidin–biotin inter-
(strept)avidin conjugates (Chapter 23, Section 2.3). action along with its resistance to breakdown makes
The following sections discuss the concept and use it extraordinarily useful in bioconjugate chemistry.
of the (strept)avidin–biotin interaction in bioconjugate Biotinylated molecules and avidin conjugates can “find”
techniques. Preparation of biotinylated molecules and each other under the most extreme conditions to bind
(strept)avidin conjugates also are reviewed with sug- and complex together. The biospecificity of the interac-
gested protocols. For a discussion of the major bio- tion is similar to antibody–antigen or receptor–ligand
tinylation reagents, see Section 6, this chapter, and recognition, but on a much higher level with respect to
Chapter 18, Section 1.3. affinity constants. Variations in buffer salt, pH, the pres-
ence of denaturants or detergents, and extremes of tem-
perature will not prevent the interaction from occurring
1.  THE (STREPT)AVIDIN–BIOTIN (Ross et al., 1986).
INTERACTION The only disadvantage to the use of avidin is its ten-
dency to bind nonspecifically with components other
Avidin is a glycoprotein found in egg-whites that than biotin due to its high pI and carbohydrate con-
contains four identical subunits of 16,400 Daltons each, tent. The strong positive charge on the protein causes
giving an intact molecular weight of approximately ionic interactions with more negatively charged mol-
66,000 (Green, 1975). Each subunit contains one binding ecules, especially cell surfaces. In addition, carbohydrate

Bioconjugate Techniques, Third Edition.


DOI: http://dx.doi.org/10.1016/B978-0-12-382239-0.00011-X 465 © 2013 Elsevier Inc. All rights reserved.
466 11.  (Strept)avidin–Biotin Systems

binding proteins on cells can interact with the polysac- the amount of nonspecific binding due to ionic interac-
charide portions on the avidin molecule to bind them in tion with other molecules. Of additional significance
regions devoid of targeted biotinylated molecules. These is the fact that streptavidin is not a glycoprotein, thus
nonspecific interactions can lead to elevated background there is no potential for binding to carbohydrate recep-
signals in some assays, preventing the full potential of tors. These factors lead to better signal-to-noise ratios in
the avidin–biotin amplification process to be realized. assays using streptavidin–biotin interactions than those
Streptavidin is a biotin-binding protein similar to employing avidin–biotin.
avidin, but it is of bacterial origin and originates from Both avidin and streptavidin can be conjugated
Streptomyces avidinii. Due to streptavidin’s structural to other proteins or labeled with various detection
differences, however, it can overcome some of the non- reagents without loss of biotin-binding activity. The
specific binding deficiencies of avidin (Chaiet and Wolf, biotin-binding proteins can also be immobilized onto
1964). Similar to avidin, streptavidin contains four sub- surfaces, chromatography supports, microparticles, and
units, each with a single biotin-binding site. After some nanoparticles for use in coupling biotinylated molecules
postsecretory modifications, the intact tetrameric protein (Xie et  al., 2011; Williams et  al., 2012; Yu et  al., 2012).
has a molecular mass of about 60,000 Daltons, slightly Streptavidin is slightly less soluble in water than avidin,
less than that of avidin (Bayer et al., 1986, 1989). but both are extremely robust proteins that can tolerate a
The primary structure of streptavidin is consider- wide range of buffer conditions, pH values, and chemi-
ably different than that of avidin, despite the fact that cal modification processes. Bioconjugate techniques can
they both bind biotin with similar avidity. The molecu- utilize the ε- or N-terminal amines on these proteins
lar model illustrates the four identical subunits of strep- for direct conjugation or employ modification reagents
tavidin as colored ribbons with four biotins shown as to transform their existing functional groups into other
space-filling molecules bound deep within the binding reactive groups (Chapter 2, Section 4).
pockets of each polypeptide chain (Le Trong et al., 2011) In this chapter and throughout this book, the use of
(PDB structure 3RY1). The variation in the amino acid the term “(strept)avidin” is meant to infer that either
sequence from avidin results in a much lower isoelectric avidin or streptavidin can be used in the associated
point for streptavidin (pI 5–6) compared to the highly protocols, conjugates, and applications. However, due
basic pI of 10 for the egg-white protein. Moderation in to its enhanced properties, streptavidin has effectively
the overall charge of streptavidin substantially reduces replaced native avidin in most capture or detection

BIOCONJUGATE TECHNIQUES
2.  Use of (Strept)Avidin–Biotin Interactions in Assay Systems 467
methods designed for use with biomolecules. There are Substrate
some chemically modified avidin preparations, such as
NeutrAvidin (Thermo Fisher), that have eliminated its Streptavidin–enzyme
conjugate E
negative properties through deglycosylation and reduc- Colored or
ing its pI through covalent modification of charged fluorescent product
residues. These modified avidin preparations perform
S
much better than native avidin and for certain applica-
tions even perform better than streptavidin. Biotinylated
Antibody binding antibody
to antigen

2.  USE OF (STREPT)AVIDIN–BIOTIN


INTERACTIONS IN ASSAY SYSTEMS
Microplate
well
The specificity of biotin binding to (strept)avidin pro-
vides the basis for developing assay systems to detect FIGURE 11.1  The basic design of the labeled avidin–biotin (LAB)
or quantify analytes. Biotinylated molecules can be assay system.
targeted in complex mixtures by using the appropriate
(strept)avidin conjugates. If the biotinylated component Substrate
has affinity for binding a particular antigen, then the
Biotinylated
antigen can be located through the use of a (strept)avi- enzyme
din conjugate containing a detectable molecule. A series Colored or
fluorescent product
of (strept)avidin–biotin interactions can be built upon E E
each other—utilizing the multivalent nature of each tet-
rameric (strept)avidin molecule—to further enhance the
detection capability for the target. Immunoassays built S

on the layering of (strept)avidin–biotin interactions Biotinylated


can result in amplified sensitivities and lower limits of Antibody binding
antibody
detection than assays using antibodies directly target- to antigen
ing an analyte (He et al., 2012).
A common application for (strept)avidin–biotin
Microplate
chemistry is in immunoassays. The specificity of anti-
well
body molecules provides the targeting capability to rec-
ognize and bind particular antigen molecules. If there
FIGURE 11.2  The basic design of the bridged avidin–biotin
are biotin labels on the antibody, it creates multiple sites (BRAB) assay system.
for the binding of (strept)avidin. If (strept)avidin is in
turn labeled with an enzyme, fluorophore, etc., then a
very sensitive antigen-detection system is created. The tests, substrate development then provides the chemi-
potential for more than one labeled (strept)avidin to cal detectability necessary to quantify the antigen
become attached to each antibody through its multiple (Guesdon et al., 1979).
biotinylation sites is the key to dramatic increases in In a slightly more complex design, the bridged avi-
assay sensitivity over that obtained through the use of din–biotin (BRAB) system uses (strept)avidin’s multiple
antibodies directly labeled with a detectable tag. biotin-binding sites to create an assay of potentially
There are several basic immunoassay designs that higher sensitivity than that of the LAB assay. Again
make use of the enhanced sensitivity afforded by the the biotinylated antibody is allowed to bind to its tar-
(strept)avidin–biotin interaction. Most of these assays get, but in the next step an unmodified (strept)avidin
use conjugates of (strept)avidin with enzymes, such is introduced to bind with the biotin binding sites on
as horseradish peroxidase (HRP) or alkaline phospha- the antibody. Finally, a biotinylated enzyme is added
tase, although other labels (such as fluorophores) can to provide a detection vehicle (Figure 11.2). Since
be used as well. In the simplest assay design, called the bound (strept)avidin still has additional biotin-
the labeled avidin–biotin (LAB) system (Figure 11.1), a binding sites available, the potential exists for more
biotinylated antibody is allowed to incubate and bind than one biotinylated enzyme to interact with each
with its target antigen. Next, a (strept)avidin–enzyme bound (strept)avidin. In some cases, sensitivity can
conjugate is introduced and allowed to interact with be increased over that of the LAB technique by using
the available biotin sites on the bound antibody. Just as this bridging ability of (strept)avidin (Seydack, 2008;
in other enzyme-linked immunosorbent assay (ELISA) Jedrychowski et al., 2009).

BIOCONJUGATE TECHNIQUES
468 11.  (Strept)avidin–Biotin Systems

Similar techniques can be used to devise (strept)


avidin–biotin assay systems for detection of nucleic
acid hybridization. DNA probes labeled with biotin
Colored or
fluorescent product
can be detected after they bind their complementary
DNA target through the use of (strept)avidin-labeled
Substrate
complexes (Bugawan et  al., 1990; Lloyd et  al., 1990).
E
Direct detection of hybridized probes can be accom-
plished, similar to the LAB method, by incubating with
S a (strept)avidin–enzyme conjugate followed by sub-
E strate development. BRAB-like and ABC-like assays can
E E also be utilized to further enhance a DNA probe signal
(Chapter 23, Section 2.3).
S Biotinylated Non-enzyme assay systems can be designed with the
S enzyme molecules
S (strept)avidin–biotin interaction, as well. Fluorescently
labeled (strept)avidin molecules can be used to detect
E Streptavidin a biotinylated molecule after it has bound its target. In
E
bridges
fact, a single preparation of a fluorescent (strept)avidin
derivative can be used as a universal detection reagent
S for any biotinylated targeting molecule. The main
Biotinylated application of this technique is in cytochemical staining
antibody wherein the fluorescence signal is used to localize anti-
Antibody
binding gen or receptor molecules in cells and tissue sections. In
to antigen addition, detection of analytes on arrays is commonly
carried out using fluorescently labeled (strept)avidin
Microplate
well
conjugates to bind to biotinylated primary antibodies
interacting with specific targets on the array surface.
FIGURE 11.3  The assay design of the avidin–biotin complex Other tags or probes can be coupled to (strept)avidin
(ABC) system. and used in a similar fashion. For instance, radiolabeled
(strept)avidin can be employed as a universal detec-
tion reagent in radioimmunoassay designs (Wojchowski
A modification on this theme can be used to produce and Sytkowski, 1986) (Chapter  12, Section 2). (Strept)
one of the most sensitive enzyme-linked assay systems avidin labeled with 125I can be used to localize bioti-
known. The ABC system (for avidin–biotin complex) nylated monoclonal antibodies directed against tumor
increases the detectability of antigen beyond that pos- cells in vivo for imaging purposes (Paganelli et al., 1988).
sible with either the LAB or BRAB designs by forming Chemical tags such as in hydrazide–(strept)avidin deriv-
a polymer of biotinylated enzyme and (strept)avidin atives can be made to site-direct (strept)avidin’s interac-
before its addition to an antigen-bound, biotinylated tion toward oxidized carbohydrate residues for specific
antibody (Bayer et  al., 1988). When (strept)avidin and detection of glycoconjugates (Section 5, this chapter).
a biotinylated enzyme are mixed together in solution Colloidal gold-labeled (strept)avidin can be used as
in the proper proportion, the multiple binding sites on highly sensitive detection reagents for microscopy tech-
(strept)avidin create a linking matrix to form a high- niques (Cubie and Norval, 1989; Krager et al., 2012)
molecular-weight complex. If the biotinylated enzyme (Chapter  14, Section 6.6). Finally, cytotoxic substances
is not in large enough excess to block all the binding coupled to (strept)avidin can be used to direct cell-kill-
sites on (strept)avidin, then additional sites will still be ing activity toward a tumor-cell-bound, biotinylated
available on this complex to bind a biotinylated anti- monoclonal antibody (or other targeting molecule) for
body which is bound to its complementary antigen. cancer therapy (Hashimoto et al., 1984; Haugland and
The large complex provides multiple enzyme mol- Bhalgat, 2008) (Chapter 20, Section 3).
ecules to enhance the sensitivity of detecting antigen Universal detection reagents can also be constructed
(Figure 11.3). Thus, the ABC procedure is currently through biotinylation techniques. Modification of
among the highest-sensitivity methods available for immunoglobulin-binding proteins with biotin tags, for
immunoassay work. It is a common assay technique instance, creates a reagent useful for the general assay
used for microplate-based ELISA assays and for immu- of antibody molecules. In this sense, biotinylated pro-
nohistochemistry (IHC) procedures (Hadisaputri et al., tein A or biotinylated protein G can be used to detect
2011; Inouye et al., 2012; Li et al., 2012). the binding of any primary IgG to its antigen target

BIOCONJUGATE TECHNIQUES
3.  Preparation of (Strept)Avidin Conjugates 469
(provided there is no other antibody molecule presence The following sections discuss three main methods
to cause nonspecific binding of the protein A compo- for preparing these types of (strept)avidin–protein
nent). Subsequent addition of a labeled (strept)avidin conjugates: (1) the use of an N-hydroxysuccinimide
molecule binds to the biotinylated protein A, complet- (NHS) ester–maleimide heterobifunctional crosslinker,
ing the formation of a detection complex (Jagannath (2) the use of the carbohydrate on glycoproteins for
and Sehgal, 1989). reductive amination coupling, and (3) employing the
To develop assay systems using the (strept)avidin– old technique of homobifunctional crosslinking with
biotin interaction, it is first necessary to produce the glutaraldehyde.
associated (strept)avidin conjugates and/or biotinylated
components. When the LAB technique is employed, 3.1.  NHS Ester–Maleimide-Mediated
the (strept)avidin conjugate is made using crosslink-
Conjugation Protocols
ing agents, not biotinylation reagents, in order to main-
tain the binding capacity of the (strept)avidin tetramer Heterobifunctional crosslinking agents can be used to
toward other biotinylated molecules. In the BRAB control the degree of protein conjugation, thus limiting
assay system, (strept)avidin is left unconjugated and polymerization and controlling the molar ratio of each
acts merely as the multivalent bridging molecule, while component in the final complex (Chapter 6). Particularly
both the targeting molecule and the detection molecule useful heterobifunctionals include the amine- and
are biotinylated. The components for the ABC assay are sulfhydryl-reactive NHS ester–maleimide crosslinkers
identical to the BRAB system. discussed in Chapter  6, Section 1. Chief among these
The following sections discuss the main techniques is succinimidyl-4-(N-maleimidomethyl)cyclohexane-
used to make (strept)avidin conjugates and various 1-carboxylate (SMCC) or sulfo-SMCC (Chapter  6,
biotinylated components. Section 6 of this chapter and Section 1.3), which contains a reasonably long spacer
Chapter 18, Section 1.3, should be consulted for a com- and a relatively stable maleimide group due to the adja-
plete overview of biotinylation reagents. cent cyclohexane ring in its cross-bridge.
Conjugations carried out with SMCC usually involve
up to three steps. In the first stage, one of the proteins is
3.  PREPARATION OF (STREPT)AVIDIN modified at its amine groups via the NHS ester end of the
CONJUGATES crosslinker to form amide linkages, which upon modifi-
cation then create derivatives that terminate in reactive
Conjugates of (strept)avidin with other protein mol- maleimide groups. If the other protein to be conjugated
ecules must be prepared to design systems using the LAB does not contain sulfhydryl residues necessary to react
assay technique. Suitable protein molecules attached to with the maleimide-activated protein, it must be modi-
(strept)avidin either possess indigenous detectability, fied to contain them (Chapter  2, Section 4.1). Finally,
such as in the case of ferritin or phycobiliproteins, or pos- the two reactive components are mixed together in the
sess catalytic activity (enzymatic) that can be utilized to proper ratio to effect the conjugation reaction (Shuvaev
produce a detectable substrate product. The majority of et al., 2011; Lillich et al., 2012).
conjugation procedures for making (strept)avidin–protein For the preparation of (strept)avidin–enzyme conju-
conjugates use the amines, sulfhydryls, or carbohydrates gates, either protein may first be modified with SMCC
on each protein as functional groups for crosslinking. and the other one modified to contain –SH groups. Since
Perhaps the most common conjugates of (strept) (strept)avidin does not possess any free sulfhydryls—
avidin involve attaching enzyme molecules for use and the disulfides present in (strept)avidin are inacces-
in ELISA systems. As in the case of antibody–enzyme sible to easy reduction—it must be modified with either
conjugation schemes (Chapter  20, Section 1), by far a crosslinker or with a thiolation agent before conjuga-
the most commonly used enzymes for this purpose tion. If the enzyme employed contains free sulfhydryls
are horseradish peroxidase and alkaline phosphatase. in its native state, such as β-galactosidase, then it is con-
Other enzymes such as β-galactosidase and glucose oxi- venient to activate (strept)avidin with SMCC and simply
dase are used less often, especially with regard to assay add the sulfhydryl-containing protein to it for conjuga-
tests for clinically important analytes (Chapter 22). tion. If the enzyme does not contain free sulfhydryls
Other proteins commonly crosslinked to (strept)avi- (as is the case with alkaline phosphatase or horseradish
din are chromogenic or fluorescent molecules, such as peroxidase), then the choice of which component gets
ferritin or phycobiliproteins (Chapter  10, Section 7). maleimide-activated and which gets thiolated is up to
These conjugates can be used in microscopy techniques the individual.
to stain and localize certain antigens or receptors in The following protocol describes the activation of
cells or tissue sections. (strept)avidin with sulfo-SMCC and its subsequent

BIOCONJUGATE TECHNIQUES
470 11.  (Strept)avidin–Biotin Systems

+ +
NH2 Cl NH2 Cl
S NH2 + S N
SH
S
H

Streptavidin molecule 2-Iminothiolane Modification producing


containing amine groups a terminal sulfhydryl group

O
H
N N
E
O
O

SMCC-activated enzyme
containing maleimide groups

+
NH2 Cl O
H
S N
S
N N
E
H
O
O

Streptavidin–enzyme conjugate
formation through thioether bond

FIGURE 11.4  Streptavidin can be modified with 2-iminothiolane (Traut’s reagent) to produce sulfhydryl groups. Subsequent reaction with a
maleimide-activated enzyme produces a thioether-linked conjugate.

conjugation with an enzyme modified to contain sulf- 2. Add 1.0 mg of sulfo-SMCC (Thermo Fisher) to each
hydryls using N-succinimidyl-S-acetylthioacetate ml of (strept)avidin solution. Mix to dissolve.
(SATA) (Chapter 2, Section 4.1). A method for the oppo- 3. React for 30 to 60 min at room temperature. Since
site approach, wherein the enzyme is activated with maleimide groups are labile in aqueous solution,
SMCC and the (strept)avidin component is thiolated, is extended reaction times should be avoided.
presented immediately after this protocol. This strategy 4. Immediately purify the maleimide-activated (strept)
may be the most common approach to forming these avidin away from excess crosslinker and reaction
conjugates (Figure 11.4). In addition, since there are byproducts by gel filtration on a desalting resin. A spin
enzymes commercially available that are activated with column will facilitate the most rapid purification. Use
SMCC (Thermo Fisher), their use may be the easiest 0.1-M sodium phosphate, 0.15-M NaCl, pH 7.2, as the
solution to forming conjugates. chromatography buffer. Pool the fractions containing
protein (the first peak eluting from the column). After
Protocol for the Conjugation of SMCC-Activated
elution, adjust the protein concentration to 10 mg/ml
(Strept)avidin with Thiolated Enzyme
for the conjugation reaction (centrifugal concentrators
ACTIVATION OF (STREPT)AVIDIN WITH SMCC work well for this step). At this point, the maleimide-
1. Dissolve (strept)avidin (Thermo Fisher) in 0.1-M activated (strept)avidin may be frozen and lyophilized
sodium phosphate, 0.15-M NaCl, pH 7.2, at a to preserve its maleimide activity. The modified
concentration of 10 mg/ml. protein is stable for at least 1 year in a

BIOCONJUGATE TECHNIQUES
3.  Preparation of (Strept)Avidin Conjugates 471
freeze-dried state. If kept in solution, the maleimide- separation between the thiolated protein and
activated (strept)avidin is labile and should be used excess hydroxylamine and reaction byproducts,
immediately to conjugate with a thiolated enzyme the sample size applied to the column should be at
following the procedure described below. a ratio of no more than 5% sample volume to the
total column volume. Collect 0.5-ml fractions. Pool
the fractions containing protein by measuring the
MODIFICATION OF ENZYME WITH SATA
absorbance of each fraction at 280 nm.
If β-galactosidase is used to conjugate with an
SMCC-activated (strept)avidin, then there is no need to PRODUCTION OF CONJUGATE
thiolate the enzyme, since it contains sulfhydryls in its 1. Immediately mix the thiolated enzyme with an
native state (Fujiwara, 1988; Sivakoff and Janes, 1988). amount of maleimide-activated (strept)avidin to
For conjugations using horseradish peroxidase, alkaline obtain the desired molar ratio of enzyme-to-(strept)
phosphatase, or glucose oxidase, however, thiolation is avidin in the conjugate. Use of a 4 : 1 (enzyme : avidin)
necessary to add the necessary sulfhydryls. molar ratio in the conjugation reaction usually results
in high-activity conjugates suitable for use in many
1. Dissolve the enzyme to be modified in 0.1-M sodium enzyme-linked immunoassay procedures employing
phosphate, 0.15-M NaCl, pH 7.2, at a concentration the LAB approach.
of 10 mg/ml. 2. React for 30 to 60 min at 37°C or 2 h at room
2. Prepare a stock solution of SATA (Thermo Fisher) by temperature. The conjugation reaction may also be
dissolving it in DMSO at a concentration of 13 mg/ml. carried out at 4°C overnight.
Use a fume hood to handle the organic solvent.
3. Add 25 μl of the SATA stock solution to each A variation of the above method can be used,
ml of 10 mg/ml enzyme solution. For different wherein the enzyme is first activated with SMCC and
concentrations of enzyme in the reaction medium, conjugated to a thiolated (strept)avidin molecule. This
proportionally adjust the amount of SATA addition; approach is probably the most common way of prepar-
however, do not exceed 10% DMSO in the aqueous ing (strept)avidin–enzyme conjugates, and since the
reaction medium. activated enzymes are readily available (Thermo Fisher)
4. React for 30 min at room temperature. it may also be the easiest.
5. To purify the SATA-modified enzyme perform a gel
filtration separation using a desalting resin or dialyze Protocol for the Conjugation of SMCC-Activated
against 0.1-M sodium phosphate, 0.15-M NaCl, pH Enzymes with Thiolated (Strept)avidin
7.2, containing 10-mM EDTA. Purification is not ACTIVATION OF ENZYME WITH SULFO-SMCC
absolutely required, since the following deprotection The following protocol describes the activation of
step is carried out using hydroxylamine at a horseradish peroxidase (HRP) with sulfo-SMCC. Other
significant molar excess over the initial amount of enzymes may be activated in a similar manner. The
SATA added. Whether a purification step is carried activated enzyme possesses maleimide groups that are
out or not, at this point, the derivative is stable and relatively unstable in aqueous solution. Therefore, the
may be stored under conditions which favor long- thiolation reaction should be coordinated with the acti-
term enzyme activity. vation process so that the final conjugation can be car-
6. Deprotect the acetylated sulfhydryl groups on the ried out immediately. Note: If activated enzymes are
SATA-modified enzyme according to the following obtained (Thermo Fisher), this step may be eliminated.
protocol:
1. Dissolve HRP in 0.1-M sodium phosphate, 0.15-M
a. Prepare a 0.5-M hydroxylamine solution in 0.1-M NaCl, pH 7.2, at a concentration of 10 mg/ml.
sodium phosphate, pH 7.2, containing 10-mM 2. Add 3.3 mg of sulfo-SMCC (Thermo Fisher) to each
EDTA. ml of the HRP solution. Mix to dissolve and react
b. Add 100 μl of the hydroxylamine stock solution for 30 min at room temperature. Alternatively, two
to each ml of the SATA-modified enzyme. Final equal additions of crosslinker may be carried out—
concentration of hydroxylamine in the enzyme the second one after 15 min of incubation—to obtain
solution is 50-mM. even more efficient modification.
c. React for 2 h at room temperature. 3. Immediately purify the maleimide-activated
d. Purify the thiolated enzyme by gel filtration on HRP away from excess crosslinker and reaction
a desalting resin using 0.1-M sodium phosphate, byproducts by gel filtration on a desalting column.
0.1-M NaCl, pH 7.2, containing 10-mM EDTA as Use 0.1-M sodium phosphate, 0.15-M NaCl, pH
the chromatography buffer. To obtain efficient 7.2, as the chromatography buffer. HRP can be

BIOCONJUGATE TECHNIQUES
472 11.  (Strept)avidin–Biotin Systems

observed visually as it flows through the column high-activity conjugates suitable for use in many
due to the color of its heme ring. Pool the fractions enzyme-linked immunoassay procedures employing
containing the HRP peak. After elution, adjust the the LAB approach.
HRP concentration to 10 mg/ml for the conjugation 8. React for 30 to 60 min at 37°C or 2 h at room
reaction. At this point, the maleimide-activated temperature. The conjugation reaction also may be
enzyme may be frozen and lyophilized to preserve done at 4°C overnight.
its maleimide activity. The modified enzyme is stable
for at least one year in a freeze-dried state. If kept 3.2.  Conjugation Using Periodate Oxidation/
in solution, the maleimide-activated HRP should be
Reductive Amination
used immediately to conjugate with thiolated (strept)
avidin following the protocols outlined below. Glycoproteins may be conjugated with another
amine-containing protein through the process of peri-
THIOLATION OF (STREPT)AVIDIN odate oxidation and reductive amination. Periodate
1. Dissolve (strept)avidin in 0.1-M sodium phosphate, oxidation of polysaccharide components on the glyco-
0.15-M NaCl, pH 7.2, at a concentration of 10 mg/ml. protein results in the formation of reactive aldehyde
2. Prepare a stock solution of SATA by dissolving it in residues by cleavage of carbon–carbon bonds and oxi-
DMSO at a concentration of 13 mg/ml. Use a fume dation of the associated adjacent hydroxyls (Chapter 2,
hood to handle the organic solvent. Section 4.4). Conjugation with another protein may be
3. Add 25 μl of the SATA stock solution to each ml achieved by reacting the aldehydes with amines to form
of 10 mg/ml (strept)avidin solution. For different intermediate Schiff bases with subsequent reduction
concentrations of protein in the reaction medium, using sodium cyanoborohydride to create stable sec-
proportionally adjust the amount of SATA addition; ondary amine bonds.
however, do not exceed 10% DMSO in the aqueous This method of conjugation is particularly well-
reaction medium. suited for coupling HRP or ferritin with (strept)avidin
4. React for 30 min at room temperature. (Dhar et  al., 2012). Both HRP and (strept)avidin are
5. To purify the SATA-modified (strept)avidin use gel glycoproteins that can be oxidized with sodium peri-
filtration on a desalting column or dialyze against odate to generate aldehydes. Thus, HRP–(strept)avidin
0.1-M sodium phosphate, 0.15-M NaCl, pH 7.2, and ferritin–(strept)avidin may be prepared by reduc-
containing 10-mM EDTA. At this point, the derivative tive amination. Ferritin is a large, complex protein of
is stable and may be stored under conditions which molecular weight 750,000. Its structure is comprised of
favor long-term (strept)avidin activity. a protein shell of diameter approximately 12 nm that
6. Deprotect the acetylated sulfhydryl groups on the surrounds a micelle core consisting of ferric hydroxide
SATA-modified protein according to the following of about 6-nm diameter. This core contains more than
protocol: 2000 iron atoms, making the protein extremely elec-
tron dense and thus perfect for electron microscopy
a. Prepare a 0.5-M hydroxylamine solution in 0.1-M applications. The properties of HRP are described in
sodium phosphate, pH 7.2, containing 10-mM Chapter 22, Section 1.
EDTA. The following protocol is adapted from Bayer et al.
b. Add 100 μl of the hydroxylamine stock solution to (1976).
each ml of the SATA-modified (strept)avidin. Final
concentration of hydroxylamine in the solution is Protocol for the Conjugation of (Strept)avidin with
50-mM. Ferritin Using Reductive Amination
c. React for 2 h at room temperature. 1. Dissolve (strept)avidin in 0.1-M sodium acetate,
d. Purify the thiolated protein by gel filtration on 0.15-M NaCl, pH 4.5, at a concentration of 3 mg/ml.
Sephadex G-25 using 0.1-M sodium phosphate, 2. Dissolve ferritin in 0.1-M sodium acetate, 0.15-M
0.1-M NaCl, pH 7.2, containing 10-mM EDTA as NaCl, pH 4.5, at a concentration of 100 mg/ml.
the chromatography buffer. 3. Add 1 ml of ferritin solution to every 5 ml of (strept)
avidin solution. Chill on ice.
CONJUGATION OF SMCC-ACTIVATED ENZYME WITH 4. Dissolve sodium periodate in water at a
THIOLATED (STREPT)AVIDIN concentration of 100-mM. Prepare fresh and protect
7. Immediately mix the SMCC-activated enzyme with from light.
an amount of thiolated (strept)avidin to obtain the 5. Add 110 μl of sodium periodate solution to each ml
desired molar ratio of enzyme-to-(strept)avidin in of (strept)avidin/ferritin solution.
the conjugate. Use of a 4 : 1 (enzyme:avidin) molar 6. React for 3 h on ice with periodic mixing. Protect
ratio in the conjugation reaction usually results in from light.

BIOCONJUGATE TECHNIQUES
3.  Preparation of (Strept)Avidin Conjugates 473
FIGURE 11.5  Oxidation of the polysac-
charide components of HRP produces reactive
E E
aldehyde groups. Conjugation to streptavidin
HO HO may then be achieved by reductive amination.
O O
NaIO4
O O O O

HO OH O O

HRP containing Oxidation producing


polysaccharide chains reactive aldehyde groups
NaCNBH3

S NH2
O

S NH E
O

Streptavidin molecule
containing amine groups
O

O
H

Reductive amination coupling


forming secondary amine linkage

7. Remove excess periodate by gel filtration on a 4. React in the dark for 15 min at room temperature.
column of Sephadex G-25 or by overnight dialysis A color change will be apparent as the reaction
against 50-mM sodium borate, 0.15-M NaCl, pH 8.5. proceeds—changing from the brownish/gold color
8. Dissolve 10 mg of sodium borohydride in 1 ml of of concentrated HRP to green. Longer reaction times
10-mM NaOH. Prepare fresh. Add 83 μl of this will result in a decrease in HRP enzymatic activity.
reducing solution to each ml of (strept)avidin/ 5. Immediately purify the oxidized enzyme by gel
ferritin solution. filtration using a column of Sephadex G-25. The
9. React for 1 h on ice. chromatography buffer is 0.01-M sodium phosphate,
10. Remove excess reductant by gel filtration using a 0.15-M NaCl, pH 7.2. Collect 0.5-ml fractions and
column of Sephadex G-25 or by extensive dialysis monitor for protein at 280 nm. HRP may also be
against 20-mM sodium phosphate, 0.15-M NaCl, detected by its absorbance at 403 nm. In oxidizing
pH 7.4. large quantities of HRP, the fraction collection
process may be done visually by pooling the
Conjugation of HRP by reductive amination can be
colored HRP peak as it comes off the column.
achieved by oxidizing the carbohydrate on the enzyme
6. Pool the fractions containing protein. Adjust
and subsequently coupling to the amines on (strept)avi-
the enzyme concentration to 10 mg/ml for the
din (Figure 11.5).
conjugation step. The periodate-activated HRP may
be stored frozen or freeze-dried for extended periods
Protocol for the Preparation of (Strept)avidin–HRP
without loss of activity. However, do not store the
by Reductive Amination
preparation in solution at room temperature or
OXIDATION OF HRP WITH SODIUM PERIODATE 4°C, since precipitation will occur over time due to
1. Dissolve HRP in water or 0.01-M sodium phosphate, self-polymerization.
0.15-M NaCl, pH 7.2, at a concentration of 10 to 20 mg/ml.
2. Dissolve sodium periodate in water at a CONJUGATION OF PERIODATE-OXIDIZED HRP WITH
concentration of 0.088-M. Protect from light. (STREPT)AVIDIN
3. Immediately add 100 μl of the sodium periodate 1. Dissolve (strept)avidin at a concentration of 10 mg/
solution to each ml of the HRP solution. This results ml in 0.2-M sodium bicarbonate, pH 9.6, at room
in an 8-mM periodate concentration in the reaction temperature. The high pH buffer will result in very
mixture. Mix to dissolve. Protect from light. efficient Schiff base formation and conjugation

BIOCONJUGATE TECHNIQUES
474 11.  (Strept)avidin–Biotin Systems

with the highest possible incorporation of enzyme with amine groups to create crosslinks by one of sev-
molecules per (strept)avidin molecule. To produce eral routes (Chapter  5, Section 6.2, and Chapter  15,
lower molecular weight conjugates (using less Section 2.1). In aqueous solution, the molecule under-
efficient Schiff base formation conditions), dissolve goes a number of transformations through hemiacetal
the proteins at a concentration of 10 mg/ml in 0.1-M and aldol products to create a variety of reactive inter-
sodium phosphate, 0.15-M NaCl, pH 7.2. mediates. Although glutaraldehyde is a bifunctional
2. The periodate-oxidized HRP (prepared above) is aldehyde molecule, it doesn’t react at both ends as one
finally purified using 0.01-M sodium phosphate, might expect through aldehyde–amine Schiff base for-
0.15-M NaCl, pH 7.2. For conjugation using mation and reductive amination. The complex activa-
the lower-pH-buffered environment, this HRP tion and conjugation reactions described in Chapter 15
preparation can be used directly at 10 mg/ml for the use of glutaraldehyde to immobilize affinity
concentration. For conjugation using the higher pH ligands onto chromatography supports can be refer-
carbonate buffer, dialyze the HRP solution against enced as a basis for the conjugation reactions that can
0.2-M sodium carbonate, pH 9.6 for 2 h at room occur for bioconjugation in solution. The reagent is
temperature prior to use. highly efficient at protein conjugation, but it also has a
3. Mix the (strept)avidin solution with the enzyme tendency to form high-molecular-weight polymers due
solution at a ratio of 1 : 6.6 (v/v). Since (strept)avidin to its bifunctional or multifunctional nature. Single-step
has a molecular weight of about 66,000 and HRP’s protocols using glutaraldehyde are particularly noto-
molecular weight is 40,000, this ratio of volumes will rious at resulting in some degree of insoluble protein
result in a molar ratio of HRP:(strept)avidin equal oligomers (Porstmann et  al., 1985). Two-step methods
to 4 : 1. For conjugates consisting of greater enzyme- somewhat alleviate this problem, but the potential for
to-(strept)avidin ratios, proportionally increase the conjugate precipitation is still present.
amount of enzyme solution as required. Typically, Preparation of (strept)avidin conjugates with other
molar ratios of 2 : 1 to 10 : 1 (enzyme:avidin) give proteins can be accomplished using either a one- or
acceptable conjugates useful in a variety of ELISA two-step glutaraldehyde procedure. Both methods may
techniques. result in some degree of oligomer formation; however,
4. React for 2 h at room temperature to form the initial the two-step protocol may keep insoluble material to
Schiff base interactions. a minimum. Although the following procedures are
5. In a fume hood, add 10 μl of 5-M sodium described using particular proteins, they may be used
cyanoborohydride (Sigma) per ml of reaction as a general guide for coupling enzymes, ferritin, phy-
solution. Caution: Cyanoborohydride is extremely cobiliproteins, or other detectable proteins to (strept)
toxic. All operations should be carried out with care avidin. Some optimization may be necessary to obtain
in a fume hood. Also, avoid any contact with the the best yield of active conjugate.
reagent, as the 5-M solution is prepared in 1-N NaOH.
6. React for 30 min at room temperature (in a fume
hood). Protocol for the One-Step Glutaraldehyde
7. Block unreacted aldehyde sites by addition of 50 μl Conjugation of Ferritin to (Strept)avidin
of 1-M ethanolamine, pH 9.6, per ml of conjugation This protocol is adapted from Bayer and Wilchek
solution. Approximately 1-M ethanolamine solution (1980).
may be prepared by addition of 300 μl ethanolamine
1. Prepare a solution containing 5 mg/ml (strept)avidin
to 5 ml of deionized water. Adjust the pH of the
and 25 mg/ml ferritin in 0.02-M sodium phosphate,
ethanolamine solution by addition of concentrated
0.15-M NaCl, pH 7.4, at room temperature. Note: For
HCl, keeping the solution cool on ice.
the coupling of other proteins to (strept)avidin, their
8. React for 30 min at room temperature.
concentration may be reduced from the 25 mg/ml
9. Purify the conjugate from excess reactants by dialysis
stated for ferritin.
or gel filtration using Sephadex G-25. Use 0.01-M
2. In a fume hood, add 10 μl of 25% glutaraldehyde per
sodium phosphate, 0.15-M NaCl, pH 7.0, as the
ml of (strept)avidin/ferritin solution. Mix well.
buffer for either operation. Use a fume hood, since
3. React for 1 h at room temperature.
cyanoborohydride will be present in some of the
4. To reduce the resultant Schiff bases and any excess
fractions.
aldehydes, add sodium borohydride to a final
concentration of 10 mg/ml.
3.3.  Glutaraldehyde Conjugation Protocol Note: Some protocols do not call for a reduction
Glutaraldehyde is one of the oldest homobifunc- step. The addition of borohydride at this level may
tional reagents used for protein conjugation. It reacts result in disulfide bond cleavage and loss of protein

BIOCONJUGATE TECHNIQUES
4.  Preparation of Fluorescently Labeled (Strept)Avidin 475
activity in some cases. As an alternative to reduction, 4.  PREPARATION OF FLUORESCENTLY
add 50 μl of 0.2-M lysine in 0.5-M sodium carbonate, LABELED (STREPT)AVIDIN
pH 9.5, to each ml of the conjugation reaction to block
excess reactive sites. Block for 2 h at room tempera- Fluorophore modification of (strept)avidin creates
ture. Other amine-containing small molecules may be a reagent system that can be used to detect and local-
substituted for lysine—such as glycine, Tris buffer, or ize biotinylated targeting molecules. The application
ethanolamine. of such reagents in immunohistochemical staining
5. Reduce for 1 h at 4°C. techniques is significant (Bonnard et  al., 1984). A bio-
6. To remove any insoluble polymers that may have tinylated antibody directed against a particular tis-
formed, centrifuge the conjugate or filter it through a sue antigen can be allowed to bind its target in situ,
0.45-μm filter. Purify the conjugate by gel filtration or and then a fluorescently tagged (strept)avidin may
dialysis using PBS, pH 7.4. be added to bind and visualize the antibody-bound
antigenic sites by luminescence. Individual cellular
A two-step glutaraldehyde protocol may result in
structures can be labeled in similar assay strategies
lower molecular weight conjugates, thus limiting the
and detected by fluorescent microscopy or cell sort-
degree of insoluble material formed during the cross-
ing techniques (Sternberger, 1986; Abou-Samra et  al.,
linking process. The following protocol is adapted from
1990). Biotinylated targeting molecules like antibod-
Avrameas (1969).
ies can have some nonspecific binding potential due
Protocol for the Two-Step Glutaraldehyde to the presence of a hydrophobic biotin tag. However,
Conjugation of Enzymes to (Strept)avidin if hydrophilic biotinylation compounds are used in the
1. Dissolve the enzyme at a concentration of 10 mg/ml modification reaction (such as those containing a PEG
in 0.1-M sodium phosphate, 0.15-M NaCl, pH 6.8. spacer; see Chapter  18, Section 1.3), then the degree
2. Add glutaraldehyde to a final concentration of of nonspecific binding in the conjugate can be kept to
1.25%. a minimum. The multivalent nature of (strept)avidin’s
3. React overnight at room temperature. biotin-binding sites combined with the potential of
4. Purify the activated enzyme from excess more than one biotin tag per antibody creates a system
glutaraldehyde by gel filtration (using Sephadex of much greater potential sensitivity than when using
G-25) or by dialysis against PBS, pH 6.8. fluorescently modified antibodies alone. The com-
5. Dissolve (strept)avidin at a concentration of plex formed from the (strept)avidin–biotin interaction
10 mg/ml in 0.5-M sodium carbonate, pH 9.5. Mix the amplifies the fluorescent signal beyond that capable in
activated enzyme with the (strept)avidin solution directly labeled antibody techniques.
at the desired molar ratio to effect the conjugation. Double labeling systems can also be developed
Mixing the equivalent of 1 to 2 moles of enzyme per using the (strept)avidin–biotin interaction. If two pri-
mole of (strept)avidin usually results in acceptable mary antibodies directed against separate antigenic
conjugates. determinants are labeled, one with biotin and the
6. React overnight at 4°C. other with another detection component (such as a
7. To reduce the resultant Schiff bases and any excess fluorophore, enzyme, gold particles, etc.), then both
aldehydes, add sodium borohydride to a final may be used to simultaneously localize different anti-
concentration of 10 mg/ml. gens in tissue sections. The biotinylated antibody may
subsequently be detected by the addition of a fluores-
Note: Some protocols avoid a reduction step, as it cently labeled (strept)avidin reagent. An example of
can lead to disulfide bond cleavage and detrimental a double label (strept)avidin–biotin detection system
effects on protein activity. As an alternative to reduc- is that of Feller et  al. (1983). A pair of tonsil antigens
tion, add 50 μl of 0.2-M lysine in 0.5-M sodium carbon- was visualized using two monoclonal antibodies, one
ate, pH 9.5, to each ml of the conjugation reaction to fluorescein-labeled, and the other biotinylated. The
block excess reactive sites. Block for 2 h at room tem- biotinylated antibody was detected by using a phycoer-
perature. Other amine-containing small molecules may ythrin-labeled (strept)avidin conjugate (Section 4.4, this
be substituted for lysine—such as glycine, Tris buffer, or chapter, and Chapter 10, Section 7). Even triple-labeling
ethanolamine. systems may be developed using this strategy (van
8. Reduce for 1 h at 4°C. Dongen et al., 1985).
9. To remove any insoluble polymers that may have The following sections present suggested protocols
formed, centrifuge the conjugate or filter it through a for labeling (strept)avidin with selected fluorophores.
0.45-μm filter. Purify the conjugate by gel filtration or Other fluorescent probes may be constructed using the
dialysis using PBS, pH 7.4. reagents and methods discussed in Chapter 10.

BIOCONJUGATE TECHNIQUES
476 11.  (Strept)avidin–Biotin Systems

FIGURE 11.6  The reaction of FITC


with streptavidin produces a fluorescent
probe via isothiourea bonds.
S NH2 O O O

O
Streptavidin containing
amine groups
O
+
O O O NH
S NH C

O S

O
Thiourea bond formation

N
C
S
FITC

4.1.  Modification with FITC Protocol


Fluorescein isothiocyanate (FITC) has been one 1. Dissolve (strept)avidin in 0.1-M sodium carbonate,
of the most common fluorescent labels used to mod- pH 9.5, at a concentration of 2 to 4 mg/ml.
ify proteins and other biomolecules (Chapter  10, 2. Dissolve FITC in DMF at a concentration of 2 mg/ml.
Section 1). The isothiocyanate group reacts with Protect from light.
amines in protein molecules to form a stable thiourea 3. Add 50 to 100 µl of the FITC solution to each ml of
linkage (Figure 11.6). (Strept)avidin may be tagged the (strept)avidin solution.
with this reagent to yield highly fluorescent deriva- Note: The optimal level of fluorescein modifica-
tives useful both in single-staining and double-stain- tion should be determined experimentally to obtain a
ing techniques (Bayer and Wilchek, 1980; Bakkus bright conjugate without the tendency to precipitate or
et al., 1989; Szabo et al., 1989). Optimal modification fluorescently quench due to over-labeling. For relatively
levels for fluorescein are in the range of 3 to 8 fluoro- hydrophobic dyes such as FITC, this may mean react-
phores per (strept)avidin molecule. Lower incorpora- ing the (strept)avidin with no more than a 5-fold mole
tion levels will result in low luminescence and poor excess of dye to obtain about two to three fluorescent
sensitivity. Higher levels may cause fluorescein–fluo- labels per molecule. Excessive labeling also causes non-
rescein quenching effects, resulting in decreased fluo- specific binding of dye-labeled conjugates in assays and
rescence. Too high a modification level may also result detection applications.
in nonspecific binding of the derivatized proteins to
non-targeted components in assay systems or even 4. React overnight at 4°C in the dark.
result in precipitation of the modified protein due to 5. Remove excess fluorescein by gel filtration using a
hydrophobic aggregation of the dyes. column of Sephadex G-25.
Although FITC and other reactive fluorescein deriva-
tives are still widely used to label (strept)avidin and
4.2.  Modification with Lissamine Rhodamine B
other proteins, better fluorescence yield and stability
will be obtained if one of the newer hydrophilic fluores-
Sulfonyl Chloride
cein dyes is used, such as DyLight 488 (Thermo Fisher). Rhodamine derivatives are popular probes to use in
See Chapter  10, Section 1, for additional details on tandem with fluorescein labels. The Lissamine deriva-
labeling proteins with fluorescein. tives of rhodamine (Chapter  10, Section 2) are intensely

BIOCONJUGATE TECHNIQUES
4.  Preparation of Fluorescently Labeled (Strept)Avidin 477

S NH2
N O N
+

Streptavidin containing
amine groups
SO3

+
+ O
N O N S
S N
H O

SO3 Sulfonamide bond formation


(red fluorescent probe)

SO2Cl

Lissamine rhodamine
sulfonyl chloride

FIGURE 11.7  Streptavidin can be labeled with Lissamine rhodamine sulfonyl chloride to form a fluorescent probe.

mole excess of dye over the amount of (strept)avidin in


fluorescent, strongly emitting in the red region of the
solution may result in the best performing conjugate.
spectrum. The red luminescence of Lissamine rhodamine
contrasts sharply with the green emission of fluorescein 4. React for 1 h at room temperature in the dark.
derivatives. Lissamine rhodamine B sulfonyl chloride 5. Remove excess fluorophore by gel filtration using a
can be used to modify proteins at their ε- and N-terminal column of Sephadex G-25 or by dialysis.
amine functional groups. The resultant derivatives are
Modification of (strept)avidin with Texas Red sulfo-
linked through stable sulfonamide bonds, resulting in
nyl chloride may be done similarly, except the fluoro-
rhodamine’s fluorescent character being incorporated
phore is first dissolved in acetonitrile prior to addition
into the modified molecules. (Strept)avidin derivatives of
to the aqueous reaction mixture.
this fluorophore are particularly popular for use in fluo-
rescent assay systems (Figure 11.7).
4.3.  Modification with AMCA–NHS
Protocol
AMCA derivatives possess fluorescent properties
1. Dissolve (strept)avidin in 0.1-M sodium carbonate/
within the blue region of the visible spectrum (Chapter 10,
bicarbonate buffer, pH 9.0, at a concentration of
Section 3). Their emission range is well removed from
1 to 5 mg/ml.
other common fluorophores, making them excellent
2. Dissolve Lissamine rhodamine B sulfonyl chloride in
choices for use in double-labeling techniques, for exam-
DMF at a concentration of 1 to 2 mg/ml. Protect from
ple with fluorescein-labeled molecules. Coumarin-based
light and use immediately. Do not use DMSO as the
fluorescent probes are very good donors for excited-state
solvent, as sulfonyl chlorides react with it.
energy transfer to fluorescein dye derivatives. AMCA–
3. In a darkened lab and with gentle mixing, slowly
NHS reacts with amine-containing molecules to result in
add 50 to 100 µl of the fluorophore solution to each
stable amide-bond derivatives (Figure 11.8). (Strept)avi-
ml of the (strept)avidin solution.
din may be labeled with this reagent to give probes useful
Note: Determine the optimal level of dye labeling for immunohistochemical staining of biotinylated target-
experimentally to avoid over-labeling, which can cause ing molecules. AMCA-labeled proteins are fairly stable to
fluorescent quenching, nonspecific binding in assays, photoquenching and exhibit a large Stokes shift, allowing
or even precipitation of the conjugate. For hydrophobic sensitive measurements to be made without interference
dyes, a reaction including no more than about a 5-fold from scattered excitation light.

BIOCONJUGATE TECHNIQUES
478 11.  (Strept)avidin–Biotin Systems

S NH2

Streptavidin containing
amine groups CH3
H
S N
+ O
O O NH2
O CH3 O
O HO Amide bond formation
N N (blue fluroescent probe)
O
O O O NH2 O
NHS
AMCA–NHS

FIGURE 11.8  AMCA–NHS reacts with the amine groups of streptavidin to produce amide bonds.

Protocol 30-fold higher than labeling with small, synthetic fluo-


1. Dissolve (strept)avidin) in 50-mM sodium borate, pH rophores. Their ability to be monitored by fluorescing
8.5, at a concentration of 10 mg/ml. Other buffers may in the red region of the spectrum decreases potential
be used for an NHS ester reaction, including 0.1-M interferences from indigenous biological fluorescence.
sodium phosphate, pH 7.5 (Chapter 3, Section 1.4). Phycoerythrin-labeled (strept)avidin probes can be used
2. Dissolve AMCA–NHS (Thermo Fisher) in DMSO at a in double-staining procedures with a fluorescein-labeled
concentration of 2.6 mg/ml. Protect from light. antibody, detecting two antigens in the same tissue sec-
3. In subdued lighting conditions, slowly add 50–100 μl tion simultaneously by excitation at 488 nm (Feller et al.,
of the AMCA–NHS stock solution to each ml of the 1983; Agata et al., 2012).
(strept)avidin solution, with gentle mixing. The bilin content of these fluorescent proteins ranges
from a low of 4 prosthetic groups in C-phycocyanin to
Note: The optimal level of fluorescent labeling the 34 groups of B- and R-phycoerythrin. Phycoerythrin
should be determined experimentally to obtain the best derivatives, therefore, can be used to create the most
conjugate in the intended application. Avoid over-label- intensely fluorescent probes possible using these pro-
ing as this may cause conjugate precipitation or non- teins. (Strept)avidin–phycoerythrin conjugates, for
specific binding in assays. Relatively hydrophobic dyes example, have been used to detect as few as 100 bioti-
may perform best if the substitution level of the con- nylated antibodies bound to receptor proteins per cell
jugate is not greater than about three to five dyes per (Zola et al., 1990).
(strept)avidin molecule. Conjugates of (strept)avidin with these fluorescent
4. React for 1 h at room temperature in the dark. probes may be prepared by activation of the phyco-
5. Remove excess reagent and reaction byproducts by biliprotein with SPDP to create a sulfhydryl-reactive
gel filtration using a column of Sephadex G-25 or by derivative, followed by modification of (strept)avidin
dialysis. with 2-iminothiolane or SATA (Chapter  2, Section 4.1)
to create the free sulfhydryl groups necessary for con-
jugation. The protocol for SATA modification of (strept)
4.4.  Conjugation with Phycobiliproteins avidin can be found in Section 3.1, this chapter. The
Phycobiliproteins are incredibly fluorescent due to procedure for SPDP activation of phycobiliproteins can
their multiple chromophoric bilin prosthetic groups, con- be found in Chapter  10, Section 7. Reacting the SPDP-
ferring extremely high absorbance coefficients to each activated phycobiliprotein with thiol-labeled (strept)
protein molecule (Chapter  10, Section 7). Conjugates of avidin at a molar ratio of 2 : 1 will result in highly fluo-
these biliproteins with targeting molecules form extraor- rescent biotin-binding probes.
dinarily luminescent probes. Labeling with phycobili- Other fluorescent probes may also be used to label
protein derivatives can provide absorption coefficients (strept)avidin molecules for detection of biotinylated

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 479
precise localization of glycoconjugates. Detection in a
O
H single step using this strategy is possible using preformed
S COOH + H2N
N
N
NH2 complexes of hydrazide-activated (strept)avidin and a
H
O
biotinylated enzyme (Figure 11.10).
The activation of (strept)avidin with adipic dihy-
Streptavidin containing
Adipic acid dihydrazide drazide may be done using the method of Bayer et  al.
carboxylate groups (1987). A summary of this protocol is given below.

EDC Protocol
1. Dissolve 160 mg of adipic acid dihydrazide (Aldrich)
in 5 ml of 0.1-M sodium phosphate, pH 6.0. Some
heating of the tube under a hot-water tap may be
required to help solubilize the compound. Cool to
O
room temperature.
H H 2. Dissolve 50 mg of (strept)avidin in the adipic acid
N N
S C N
H
NH2 dihydrazide solution.
O O 3. Add 160 mg of the water soluble carbodiimide
EDC (Thermo Fisher) (Chapter 4, Section 1.1) to the
Hydrazide-activated streptavidin solution, and mix to dissolve.
4. React for 4 h at room temperature.
FIGURE 11.9  Reaction of adipic acid dihydrazide with strepta- 5. Dialyze against PBS, pH 7.2, to remove excess
vidin produces a hydrazide derivative that is highly reactive toward reagent and reaction byproducts.
periodate-oxidized polysaccharides.
Hydrazide-activated (strept)avidin may be stored as
a freeze-dried preparation without loss of activity.
targeting molecules. Chapter  10 reviews many addi-
tional fluorescent labels, such as quantum dots, lantha-
nide chelates, and cyanine dye derivatives, all of which
6.  BIOTINYLATION TECHNIQUES
may be used in similar protocols to create detection
conjugates for (strept)avidin–biotin-based assays.
The highly specific interaction of (strept)avidin with
the small vitamin biotin can be a useful tool in design-
ing assay, detection, and targeting systems for biologi-
5.  PREPARATION OF HYDRAZIDE cal analytes (see Sections 1 and 2). The extraordinary
ACTIVATED (STREPT)AVIDIN affinity of (strept)avidin’s interaction with biotin allows
biotin-containing molecules in complex mixtures to be
Hydrazide groups can react with aldehydes or ketones discretely bound with (strept)avidin conjugates. If the
to form hydrazone linkages (Chapter  3, Section 5.1). (strept)avidin–biotin complex contains detection com-
Proteins may be labeled with hydrazide residues by ponents, then the targeted analytes can be located or
reaction of their indigenous carboxylate groups with bis- quantified. This assay concept is made possible through
hydrazine compounds such as adipic acid dihydrazide or the ability of biotin to be covalently attached to other
carbohydrazide (Chapter  5, Section 8). A carbodiimide- targeting molecules, such as antibodies. In this sense,
mediated reaction between the protein and the bis-hydra- biotin derivatives may be prepared which contain
zine reagent forms diimide-bond derivatives terminating reactive portions able to couple with particular func-
in hydrazide groups (Figure 11.9). (Strept)avidin labeled tional groups in proteins and other molecules. Biotin
with adipic acid dihydrazide can form the basis of a car- modification of secondary molecules, called biotinyl-
bohydrate detection system using the (strept)avidin–bio- ation, results in covalent derivatives containing one or
tin interaction (Bayer et al., 1987, 1990; Bayer and Wilchek, more bicyclic biotin rings extending from the parent
1990). Glycoconjugates in tissue sections, cells, or blots structure. These biotinylation sites are still capable of
may be treated with sodium periodate or galactose oxi- binding avidin or streptavidin with the specificity and
dase to create aldehyde groups on the associated sugar nearly the same avidity of free biotin in solution. Since
components. Introduction of hydrazide-activated (strept) the biotin components are relatively small, macromol-
avidin causes hydrazone bonds to form between the ecules can be modified with these reagents without
hydrazides and aldehydes, thus specifically targeting gly- significantly affecting their physical or chemical proper-
coproteins and other carbohydrate-containing molecules. ties (Della-Penna et  al., 1986). Proteins, carbohydrates,
Subsequent detection with a biotinylated enzyme allows lipid molecules, and nucleic acids can be modified to

BIOCONJUGATE TECHNIQUES
480 11.  (Strept)avidin–Biotin Systems

Horseradish
peroxidase
(HRP)

HO Sodium HO
O periodate O
O O O O

HO OH O O

Glycoprotein containing Polysaccharide groups


carbohydrate residues containing reactive aldehydes

Hydrazide-
activated
streptavidin

HO
O
O O
O
H H
N N
O
S C N
H
N
H
O O

Specific labeling at glycoprotein sites

FIGURE 11.10  Glycoproteins can be oxidized with sodium periodate to generate aldehyde residues. These may be specifically labeled using
a hydrazide–streptavidin derivative through hydrazone bond formation. Subsequent detection may be done using biotinylated enzymes.

contain one or more biotins able to strongly interact Bicyclic


with (strept)avidin. The technique of biotinylation is Spacer Arm Ring System
made easier through the commercial availability of a
range of different biotin derivatives having a number of
important reactivity and property characteristics useful
in (strept)avidin–biotin chemistry.
The basic design of a biotin labeling compound is Reactive Group Valeric Acid
illustrated in Figure 11.11. Common to all such modifi- Side Chain
cation reagents is the presence of the bicyclic biotin ring
FIGURE 11.11  The basic design of a biotinylation reagent
at one end of the structure and a reactive group at the includes the bicyclic rings and valeric acid side chain of d-biotin at
other end that can be used to couple with other mole- one end and a reactive group to couple with target groups at the other
cules. Biotinylation reagents also possess various cross- end. Spacer groups may be included in the design to extend the biotin
bridges or spacer groups built off the valeric acid side group away from modified molecules, thus ensuring better interac-
tion capability with avidin or streptavidin probes.
chain of the molecule. Since the binding sites for biotin
on avidin and streptavidin are pockets buried about 9 Å
beneath the surface of the proteins, spacers can affect of an avidin or streptavidin probe to a biotinylated mol-
the accessibility of biotinylated compounds for effi- ecule is also affected by the length of spacer in the bio-
ciently binding avidin or streptavidin conjugates (Green tinylation reagent. When longer spacers are utilized to
et  al., 1971). In some applications, the use of a long make biotinylated macromolecules, it can potentially
spacer arm in the biotinylation reagent will result in the result in a 5-fold greater rate of streptavidin interaction
greatest potential assay sensitivity. The rate of binding (Bonnard et al., 1984).

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 481
Biotinylation reagents containing spacers can be 6.1.  Determination of the Level of Biotinylation
subdivided into several categories: (1) aliphatic spac-
ers, (2) PEG-based spacers, and (3) cleavable or non- It is often important to determine the extent of biotin
cleavable. The aliphatic spacer arms typically consist modification after a biotinylation reaction is complete.
of hydrocarbon chains, which may be substituted Measuring biotin incorporation into macromolecules
with amide bonds or disulfide linkages. These spacers can aid in optimizing a particular (strept)avidin–­biotin
increase the length of the biotin group from a modified assay system. It can also be used to ensure reproduc-
protein, but they may also confer considerable hydro- ibility in the biotinylation process. The most com-
phobicity to the biotinylation agent and molecules mon method of measuring the degree of biotinylation
labeled with them. In fact, many biotinylated proteins makes use of the HABA dye assay (Green, 1965).
may have a tendency to aggregate in solution or even HABA is 4′-hydroxyazobenzene-2-carboxylic acid. In
precipitate due to the overall increased hydrophobic- the absence of biotin, the dye is capable of specifically
ity of the biotin modifications. By contrast, biotinyl- forming noncovalent complexes with (strept)avidin
ation compounds built using hydrophilic PEG-based at its biotin-binding sites. Upon binding to (strept)avi-
spacers have excellent water-solubility properties and din in aqueous solution, HABA exhibits a character-
have little or no tendency to aggregate or precipitate istic absorption band at 500 nm (ε =  35,500 M−1 cm−1,
in solution. When using biotinylation agents having expressed as per mole of HABA bound). The addition
aliphatic spacers, such as the popular NHS–LC-biotin, of biotin to this complex results in displacement of
it is therefore advantageous to keep the level of bio- HABA from the binding site, since the affinity constant
tin modification at a minimum to avoid nonspecific of the (strept)avidin–biotin interaction (1.3 × 1015 M−1)
binding, aggregation, or precipitation caused by the is much greater than that for (strept)avidin–HABA
reagent. Antibodies biotinylated using such biotin (6 × 106 M−1). As HABA is displaced, the absorbance of
compounds often do well if there are only one to three the complex decreases proportionally. Thus, the amount
biotin groups per antibody molecule; however, higher of biotin present in the solution can be determined by
modification levels can quickly cause stability prob- plotting the (strept)avidin–HABA absorbance at 500 nm
lems as well as nonspecific binding in assays. This versus the absorbance modulation with increasing con-
problem is circumvented by use of the hydrophilic centrations of added biotin. Comparing an unknown
biotin compounds containing a PEG spacer arm. Even biotin-containing sample to this standard response
over-biotinylation of an antibody with a PEG-based curve, can result in the determination of the biotin con-
reagent usually won’t result in precipitation, espe- centration in the sample.
cially if the length of the PEG spacer is at least PEG4 Since a biotinylated molecule potentially is able to
or greater. These hydrophilic biotins are discussed in interact with (strept)avidin at its biotin-binding sites
detail in Chapter 18, Section 1.3. just as strongly as biotin in solution, the degree of bio-
Another variable to consider in choosing bioti- tinylation may be determined using the HABA method
nylation reagents is the use of a biotin analog such as as well. Comparison of the response of a biotinylated
iminobiotin that has a moderated affinity constant in protein, for example, with a standard curve of various
its binding of avidin or streptavidin (Section 6.2, this biotin concentrations allows calculation of the molar
chapter). Analogs may be useful if release of the (strept) ratio of biotin incorporation.
avidin–biotin bond is important for isolating a targeted Two variations of the HABA–dye assay for biotinyl-
analyte. Using native biotin, the interaction with avidin ated proteins are possible. In one approach, the bioti-
is so strong that up to 6- to 8-M guanidine at pH 1.5 is nylated protein is digested using the enzyme pronase
required to break the bond, possibly causing extensive prior to doing the assay. The digestion process breaks
denaturation of any other complexed molecules. By the protein into small fragments, some of which pos-
contrast, iminobiotinylated molecules can be released sess biotin modifications. The digestion is done to elimi-
simply by adjusting the pH down to 4. nate any sterically hindered biotinylation sites from not
The following sections discuss some of the more being able to interact with (strept)avidin. The second
common biotinylation reagents available for modifi- approach merely uses the intact biotinylated protein in
cation of proteins and other biomolecules. Each biotin the assay, assuming that the HABA assay results will
derivative contains a reactive portion (or can be made then provide a truer picture of the level of accessible bio-
to contain a reactive group) that is specific for coupling tin sites on the molecule. Pronase addition obviously
to a particular functional group on another molecule. is not necessary for assessing biotinylated molecules
Careful choice of the correct biotinylation reagent can which are not proteins.
result in directed modification away from active centers The following protocol describes both of these
or binding sites, and thus preserve the activity of the HABA-based tests for determining the level of
modified molecule. biotinylation.

BIOCONJUGATE TECHNIQUES
482 11.  (Strept)avidin–Biotin Systems

Protocol protein sample by using the standard curve. The


1. Dissolve (strept)avidin in 0.05-M sodium phosphate, number of moles of biotin divided by the moles
0.15-M NaCl, pH 6.0, at a concentration of of protein present gives the number of biotin
0.5 mg/ml. A total of 3 ml of the (strept)avidin modifications on each protein molecule.
solution is required to create a standard curve using
known concentrations of biotin and an additional
6.2.  Amine-Reactive Biotinylation Agents
3 ml is needed for each sample determination.
2. Dissolve the HABA dye (Sigma) in 10-mM NaOH Amine-reactive biotinylation reagents contain reac-
at a concentration of 2.42 mg/ml (10-mM). Prepare tive groups off biotin’s valeric acid side chain that are
about 100 μl of the HABA solution for each 3-ml able to form covalent bonds with primary amines in
portion of (strept)avidin solution required. proteins and other molecules. Two basic types are com-
3. Dissolve the biotinylated protein to be measured monly available: NHS esters and carboxylates. NHS
in 0.05-M sodium phosphate, 0.15-M NaCl, pH 6.0, esters spontaneously react with amines to form amide
at a concentration of 10 to 20 mg/ml. The amount linkages (Chapter  3, Section 1.4), whereas carboxylate-
required is about 100 μl of sample per determination. containing biotin compounds can be coupled to amines
4. Dissolve d-biotin in 0.05-M sodium phosphate, via a carbodiimide-mediated reaction using EDC
0.15-M NaCl, pH 6.0, at a concentration of 0.5-mM. (Chapter 4, Section 1.1).
5. For the proteolytic digestion procedure, dissolve
pronase in water at a concentration of 1% (w/v). d-Biotin and Biocytin
6. If pronase digestion of the biotinylated protein is d-Biotin (hexahydro-2-oxo-1 H-thieno[3,4-d]imidazole-
to be carried out, heat 100 μl of the sample at 56°C 4-pentanoic acid) is a naturally occurring growth fac-
for 10 min, then add 10 μl of the pronase solution. tor present in small amounts within every cell. It is a key
Allow the sample to digest enzymatically at room component in numerous processes involving carboxyl-
temperature overnight. If no pronase digestion ation reactions, wherein it functions as a cofactor and
is desired, simply use the biotinylated protein transporter of CO2 (coenzyme R). Biotin is mainly found
solution prepared in step three without further covalently attached to lysine ε-amine groups of proteins
treatment. via its valeric acid side chain. The compound was origi-
7. To construct a standard curve of various biotin nally discovered through symptoms of deficiency caused
concentrations, first zero a spectrophotometer by eating too many raw egg-whites. Biotin (or vitamin H)
at an absorbance setting of 500 nm with sample was found to be complexed and inactivated by the egg-
and reference cuvettes filled with 0.05-M sodium white protein avidin (Boas, 1927; du Vigneaud, 1940).
phosphate, 0.15-M NaCl, pH 6.0. Remove the buffer Treatment with additional vitamin H alleviated the
solution from the sample cuvette and add 3 ml of the symptoms.
(strept)avidin solution plus 75 μl of the HABA dye
solution. Mix well and measure the absorbance of
the solution at 500 nm. Next add 2-μl aliquots of the
biotin solution to this (strept)avidin–HABA solution,
mix well after each addition, and measure and record
the resultant absorbance change at 500 nm. With
each addition of biotin, the absorbance of the (strept)
avidin–HABA complex at 500 nm decreases. The
absorbance readings are plotted against the amount
of biotin added to construct the standard curve.
8. To measure the response of the biotinylated protein
sample, add 3 ml of the (strept)avidin solution plus
75 μl of the HABA dye to a cuvette. Mix well and
measure the absorbance of the solution at 500 nm.
Next, add a small amount of sample to this solution
and mix. Record the absorbance at 500 nm. If the
change in absorbance due to sample addition was
not sufficient to obtain a significant difference
from the initial (strept)avidin–HABA solution,
add another portion of sample and measure again.
Determine the amount of biotin present in the 

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 483
The interaction of biotin with the proteins avidin and The only potential deficiency in using d-biotin to
streptavidin is among the strongest noncovalent affini- directly modify a protein is the relatively short spacer
ties known (Ka = 1015 M−1). The binding occurs between arm afforded by the indigenous valeric acid group. Some
the bicyclic ring of biotin and a pocket within each of applications may require longer spacers to maintain
the four subunits of the proteins. The valeric acid por- good binding potential toward avidin or streptavidin.
tion is not directly involved with the interaction with Biocytin is ε-N-biotinyl-l-lysine, a derivative of
avidin (Green, 1975; Wilchek and Bayer, 1988), but d-biotin containing a lysine group coupled at its
elimination of its carboxylate group or changing the ε-amino side chain to the valeric acid carboxylate. It is
amide linkage in biotinylation compounds can affect its a naturally occurring complex of biotin that is typically
affinity for (strept)avidin. Amide derivatives of biotin’s found in serum and urine, and probably represents
valeric acid group, however, can be made without inter- breakdown products of recycling biotinylated proteins.
fering with its high-affinity interactions. This character- The enzyme biotinidase specifically cleaves the lysine
istic allows modification of the valeric acid side chain residue and releases the biotin component from biocy-
without affecting the binding potential toward avidin tin (Ebrahim and Dakshinamurti, 1986, 1987).
or streptavidin. Biocytin has been used extensively as a labeling
d-Biotin is thus the basic building block for con- reagent for intracellular components within neurons
structing biotinylation reagents. The molecule may (Horikawa and Armstrong, 1988; King et al., 1989; Izzo,
be attached directly to a protein via its valeric acid 1991; Granata and Kitai, 1992). It is particularly good
side chain or derivatized at this carboxylate with for anterograde tracing studies in the central nervous
other organic components to create spacer arms and system, since it can be easily injected into neurons
various reactive groups. Reaction of biotin with pri- using micropipettes. Subsequent visualization of bio-
mary amine groups on proteins can be done using cytin locations may be achieved using a (strept)avidin–
the water soluble carbodiimide EDC (Chapter  4, enzyme conjugate (Section 3, this chapter).
Section 1.1). EDC activates the carboxylate to create Biocytin should not be used in a carbodiimide reac-
a highly reactive, intermediate ester. This ester can tion to modify proteins or other molecules, since it
then couple to amines to form stable amide bond contains both a carboxylate and an amine group. A car-
derivatives (Figure 11.12). Biotinylated molecules bodiimide-mediated reaction, as suggested for d-biotin
thus formed retain the ability to bind avidin or strep- previously, would cause self-conjugation and polymer-
tavidin with high affinity. ization of this reagent.

FIGURE 11.12  D-Biotin can be directly coupled to amine-containing molecules using the water soluble carbodiimide EDC to form an amide
bond linkage.

BIOCONJUGATE TECHNIQUES
484 11.  (Strept)avidin–Biotin Systems

FIGURE 11.13  The active ester group of


NHS–biotin reacts with amine-containing com-
pounds to form amide bond linkages.

Biocytin, however, can form the basis for construct- groups in proteins and other molecules. NHS esters
ing trifunctional crosslinking reagents (Chapter  7). The react by nucleophilic attack of an amine on the carbonyl
lysine component of the molecule contains a free car- group, releasing the NHS group and forming a stable
boxylate and an α-amine group that can be used to build amide linkage (Chapter  3, Section 1.4) (Figure 11.13).
spacers and reactive groups for crosslinking purposes. NHS–biotin is the simplest biotinylation reagent avail-
The biotin component is the third arm of the trifunctional able. Modification reactions are carried out under mildly
system, retaining its ability to bind (strept)avidin probes alkaline conditions, and they usually result in a high
after conjugation has occurred at its other two ends. Such efficiency of biotin incorporation.
a trifunctional derivative has been used to study the
hormone binding site of the insulin receptor (Wedekind
et  al., 1989). This compound, 4-azido-2-nitrophenyl-
biocytin-4-nitrophenyl ester, contains an amine-reactive
group and a photoreactive phenyl azide functionality
(Chapter 7, Section 1). The nitrophenyl ester reacts with
amines on proteins and other molecules to from stable
amide linkages. Once a molecule is modified in this man-
ner, it contains both a photosensitive group and a bio-
tin handle for conjugation and detection, respectively.
Interaction of the modified protein with another protein
followed by subsequent photolysis with UV light will
result in covalent crosslinking. Localization and detec-
tion of the crosslinked molecules can then be done using
an avidin or streptavidin conjugate. Another trifunc-
tional compound, sulfo-SBED (or sulfosuccinimidyl-
2-[6-(biotinamido)-2-(pazidobenzamido) hexanoamido]
ethyl-1,39-dithiopropionate), is also based on a biocytin
core (Chapter  7, Section 2). Additional information on
the properties and use of these trifunctional biotin com-
pounds based upon biocytin is described in Chapter  7 
and in Chapter 24, Section 3, related to the study of pro- NHS–biotin is insoluble in aqueous environments.
tein interactions. It must be dissolved first in organic solvent as a con-
centrated stock solution and an aliquot added to an
NHS–Biotin and Sulfo-NHS–Biotin aqueous reaction medium to facilitate dissolution.
The valeric acid carboxylate of d-biotin may be acti- Organic solvents such as dimethylformamide (DMF)
vated to an NHS ester for direct modification of amine or dimethyl sulfoxide (DMSO) are suitable for this

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 485
purpose. Addition of an NHS–biotin solution to a reac- 2. Immediately before use, dissolve sulfo-NHS–biotin
tion should not exceed a level of about 10% organic (Thermo Fisher) in water at a concentration of
solvent in the buffer to avoid protein precipitation 20 mg/ml. Alternatively, the compound may be
problems or precipitation of the buffer salt itself. Once dissolved in organic solvent to prevent hydrolysis
added to the reaction medium, NHS–biotin may appear prior to a reaction (i.e., dry DMF or DMSO). Adjust
as a cloudy or hazy suspension, indicating incom- the concentration and quantity of this stock solution
plete solubility. However, such micro-dispersions are to be prepared according to the amount of reagent
still effective at modification, often driving the bulk needed to biotinylate the desired amount of
of the reagent into solution as the NHS ester reacts. protein. If prepared in water, the sulfo-NHS–biotin
Biotinylation of peptides or other molecules that are stock solution must be used immediately, since
water insoluble may be carried out completely in the NHS ester is subject to hydrolysis in aqueous
organic solvent. For example, insulin can be biotinyl- environments.
ated with NHS–biotin in an organic medium (Hofmann 3. With mixing, add a quantity of the sulfo-NHS–biotin
et al., 1977). solution to the protein solution to obtain a 5- to
A water soluble analog of NHS–biotin containing 10-fold molar excess of biotinylation reagent over
a negatively charged sulfonate group on its NHS ring the quantity of protein present. For instance, for an
structure is also available. Sulfo-NHS–biotin may be immunoglobulin (MW 150,000) at a concentration
added directly to aqueous reactions without the need of 10 mg/ml, 10 μl of a sulfo-NHS–biotin solution
for organic solvent dissolution. A concentrated stock (containing 8 × 10−4 mmoles) should be added per ml
solution may be prepared in water to facilitate the addi- of antibody solution to obtain a 6-fold molar excess.
tion of a small quantity to a reaction, but hydrolysis of For more dilute protein solutions (e.g., 1–2 mg/ml),
the NHS ester will occur at a rapid rate, so the solution an increased amount of biotinylation reagent may
must be used immediately. This reagent is widely used be required (e.g., >10-fold molar excess) to obtain
to biotinylate antibodies and proteins for subsequent similar incorporation yields as when using more
detection using streptavidin-based conjugates or for concentrated protein solutions.
capture on immobilized streptavidin resins, especially
Note: To obtain biotinylation densities in the range
in pull-down assays (Lei et  al., 2012; Wu et  al., 2012)
of one to five biotins per protein, use no more than a
(Chapter 15).
5-fold mole excess of biotinylation reagent over the
The only disadvantage to the use of NHS–biotin or
concentration of protein present in the reaction. Some
sulfo-NHS–biotin is the lack of a long spacer group off
optimization may have to be done to obtain the best
the valeric acid side chain. Since the binding site for
biotinylation level for use in the anticipated application.
biotin on avidin and streptavidin is somewhat below
the surface of the proteins, some biotinylated molecules 4. React for 30 to 60 min at room temperature.
may not interact as efficiently with (strept)avidin as 5. Purify the biotinylated protein from excess reagent
when longer cross-bridges are used (Green et  al., 1971; and reaction byproducts by gel filtration using a
Bonnard et al., 1984). desalting resin or by dialysis against PBS.
NHS esters of d-biotin have been used in many
Determination of the degree of biotinylation can be
applications, including the biotinylation of rat IgE
­
done using the HABA assay (Section 6.1, this chapter).
to study receptors on murine lymphocytes (Lee and
Conrad, 1984), in the development of an immunochemi-
cal assay for a post-synaptic protein and its receptor NHS–LC-Biotin and Sulfo-NHS–LC-Biotin
(LaRochelle and Froehner, 1986), in the study of plasma NHS–LC-biotin is a derivative of d-biotin contain-
membrane domains by biotinylation of cell surface pro- ing a spacer arm off the valeric acid side chain, termi-
teins in Dictyostelium disoideum amoebas (Ingalls et  al., nating in an NHS ester. The compound is also known
1986), and for the detection of blotted proteins on nitro- as succinimidyl-6-(biotinamido)hexanoate or NHS-X-
cellulose membranes after transfer from polyacrylamide biotin. The 6-aminocaproic acid spacer provides greater
electrophoresis gels (LaRochelle and Froehner, 1986b). length between a covalently modified molecule and the
The following protocol is a generalized method for bicyclic biotin rings. The total distance from an attached
the biotinylation of a protein using sulfo-NHS–biotin. molecule to the biotin component is about 22.4 Å, sig-
nificantly greater than the 13.5-Å length of NHS–biotin
Protocol without a spacer arm. This increased distance can result
1. Dissolve the protein to be biotinylated in 0.1-M in better binding potential for avidin or streptavidin
sodium phosphate, 0.15-M NaCl, pH 7.2 to 7.5, at a probes, because the binding sites on these proteins are
concentration of 1 to 10 mg/ml. buried relatively deep inside the surface plane.

BIOCONJUGATE TECHNIQUES
486 11.  (Strept)avidin–Biotin Systems

In a study comparing NHS–LC-biotin with two other


derivatives of biotin, NHS–SS-biotin (Section 6.2, this
chapter) and biotin hydrazide (Section 6.4, this chap-
ter), it was found that modification through amines on
monoclonal antibodies resulted in 2.5 times more activ-
ity in binding a streptavidin–agarose affinity column
than when modification of carbohydrate residues using
hydrazide conjugation chemistry was done (Gretch
et al., 1987). This was probably due to the greater abun-
dance of amino groups over polysaccharide residues on
these antibodies or the limited accessibility of the gly-
can between the heavy chains in the Fc region.
NHS–LC-biotin can be used to add a biotin tag to
monoclonal antibodies directed at certain tumor anti-
gens. The biotinylated monoclonals are allowed to
bind to the tumor cell surfaces in vivo, and subsequent
 administration of an avidin or streptavidin conjugate
The NHS ester end of NHS–LC-biotin reacts with can form the basis for inducing cytotoxic effects or cre-
amine groups in proteins and other molecules to form ating traceable complexes for use in imaging techniques
stable amide-bond derivatives (Figure 11.14). Optimal (Hnatowich et al., 1987).
reaction conditions are at a pH of 7 to 9, but the higher The reagent has also been used in a unique tRNA-
the pH the greater will be the hydrolysis rate of the mediated method of labeling proteins with biotin for
ester. Avoid amine-containing buffers that will compete nonradioactive detection of cell-free translation prod-
in the acylation reaction. NHS–LC-biotin is insoluble in ucts (Kurzchalia et  al., 1988), in creating one- and two-
aqueous reaction conditions and must be solubilized in step noncompetitive avidin–biotin immunoassays
organic solvent prior to the addition of a small quan- (Vilja, 1991), for immobilizing streptavidin onto solid
tity to a buffered reaction. Preparation of concentrated surfaces using biotinylated carriers with subsequent
stock solutions may be carried out in DMF or DMSO. use in a protein avidin–biotin capture system (Suter and
Nonaqueous reactions may also be carried out with this Butler, 1986), and for the detection of DNA on nitrocel-
reagent for the modification of molecules insoluble in lulose blots (Leary et al., 1983).
water. The molar ratio of NHS–LC-biotin to a protein Sulfo-NHS–LC-biotin, a water soluble analog of
in a reaction can be from about 2 : 1 to about 50 : 1, with NHS–LC-biotin, is also available (Thermo Fisher),
higher levels resulting in better incorporation yields which contains a negatively charged sulfonate group
(Gretch et al., 1987). on its NHS ring structure. The presence of the negative

FIGURE 11.14  NHS–LC-biotin provides an


extended spacer arm to allow greater distance
between the biotin rings and a modified molecule.
Reaction with amines forms amide linkages.

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 487
charge creates enough polarity within the molecule to Note: To obtain a lower level of biotinylation (better
allow direct solubility in aqueous reaction mediums. All to maintain solubility and avoid aggregation of bioti-
other properties of the sulfonated version of the reagent nylated proteins), react the biotin compound at a level
are the same as those of NHS–LC-biotin. Sulfo-NHS– of no more than a 5-fold mole excess over the amount
LC-biotin has been used to develop a dual-labeling of protein present in the reaction medium.
method for virus imaging (Liu et al., 2012), as a biotinyl-
4. React for a total of 30 to 60 min at room temperature
ation agent having reduced membrane permeability for
or several hours at 4°C.
cell labeling (Strassberger et  al., 2011), and to develop
5. Remove unreacted biotinylation reagent and reaction
a pull-down assay to investigate pore opening in ion
byproducts by gel filtration using a desalting resin or
channels (Tolino et al., 2011).
dialysis against PBS.
Although NHS–LC-biotin and sulfo-NHS–LC-biotin
6. Assay for the level of biotin incorporation using the
are very popular reagents for biotinylation, they both
HABA dye procedure (Section 6.1, this chapter).
result in hydrophobic aliphatic biotin modifications on
proteins and antibodies. Unfortunately, these groups
have a tendency to aggregate in aqueous solution and NHS–Iminobiotin
may cause protein precipitation or loss of activity over NHS–iminobiotin is N-hydroxysuccinimido-2-
time, even when using low amounts of biotin substi- iminobiotin, the guanidino analog of NHS–biotin that
tution. Many commercial applications of biotinylated has a lower affinity constant for binding avidin or strep-
antibodies, for instance, use a low substitution level of tavidin. Iminobiotin replaces the 2-oxo-imidazole upper
biotin to avoid the aggregation issue as much as possi- ring structure of d-biotin with a 2-imino-imidazole
ble. As an alternative, the use of more hydrophilic PEG- structure, causing moderated interaction with the avi-
based biotin compounds of approximately the same din or streptavidin binding sites. This biotin analog can
spacer length are a better choice for maintaining water be used in situations requiring mild dissociation of the
solubility of modified proteins (Chapter 18, Section 1.3). (strept)avidin-biotin complex. Normally, breaking the
The following protocol is a suggested method for the (strept)avidin–biotin interaction requires 6- to 8-M gua-
biotinylation of proteins with either NHS–LC-biotin or nidine hydrochloride at a pH of 1.5, an environment
sulfo-NHS–LC-biotin. too severe for most proteins to maintain native struc-
ture or recover activity. Iminobiotin, by contrast, can
Protocol be bound to avidin or streptavidin at a pH wherein the
1. Dissolve the protein to be biotinylated in 0.1-M guanidino group is unprotonated and thus uncharged.
sodium phosphate, 0.15-M NaCl, pH 7.2 to 7.5, at a Binding occurs at pH values above 9.5 (typically done
concentration of 10 mg/ml. with good affinity at pH 11), and elution can be accom-
2. Dissolve NHS–LC-biotin (Thermo Fisher) in dry plished simply by changing the pH to 4.0—an environ-
DMF at a concentration of 40 mg/ml. This stock ment that protonates the 2-imino group and creates a
solution is stable for reasonable periods, although positive charge, thus effectively dissociating the interac-
long-term storage is not recommended. For use tion (Figure 11.15).
of the water soluble sulfo-NHS–LC-biotin, a stock
solution may be prepared in either organic solvent or
water. If a solution in water is made to facilitate the
addition of a small quantity of reagent to a reaction,
then the solution should be prepared quickly and
used immediately to prevent hydrolysis of the NHS
ester. Sulfo-NHS–LC-biotin may be dissolved in
water at a concentration of 20 mg/ml.
3. Add 50 μl of the NHS–LC-biotin solution in DMF
to each ml of the protein solution in two aliquots
apportioned 10 min apart. Alternatively, add a
quantity of the sulfo-NHS–biotin solution prepared
in water to the protein solution to obtain a 5- to
10-fold molar excess of biotinylation reagent over 
the quantity of protein present. For instance, for an NHS–iminobiotin can be used to label amine-­
immunoglobulin (MW 150,000) at a concentration containing molecules with an iminobiotin tag, pro-
of 10 mg/ml, 10 μl of the sulfo-NHS–biotin solution viding reversible binding potential with avidin or
(8 × 10−4 mmoles) should be added per ml of streptavidin. The NHS ester reacts with proteins and
antibody solution to obtain a 6-fold molar excess. other amine-containing molecules to create stable

BIOCONJUGATE TECHNIQUES
488 11.  (Strept)avidin–Biotin Systems

precipitation problems. Optimal conditions for protein


derivatization include non-amine-containing buffers
at a pH of 7 to 9. The following protocol is a suggested
method for labeling antibodies with NHS–iminobiotin.
Some optimization may have to be carried out for par-
ticular derivatization needs.

Protocol
FIGURE 11.15  At pH 4, the protonated form of iminobiotin 1. Dissolve the antibody to be modified in 50-mM
does not interact with the binding sites on avidin or streptavidin. At sodium borate, pH 8.0, at a concentration of
pH 11, the imino group is unprotonated and regains binding capabil- 5 mg/ml.
ity toward these proteins.
2. Dissolve NHS–iminobiotin in DMF at a concentration
of 1 mg/ml. Prepare fresh.
3. Add 100 μl of the NHS–iminobiotin solution to each
ml of the antibody solution. Mix well to dissolve.
Note: In the beginning, some turbidity may be pres-
ent in the reaction due to incomplete dissolution of
the NHS–iminobiotin. The solution may look cloudy
or have a micro-particulate suspension present. This
is normal for many water-insoluble reagents when
added to an aqueous solution in an organic solvent. As
the reaction takes place, the NHS–iminobiotin will be
driven into solution, both by coupling to the protein
and by hydrolysis of the NHS ester.
Note: The level of biotinylation of the protein to be
modified should be controlled to prevent over-labeling,
which can result in precipitation of the conjugate and
nonspecific binding in the intended application. Reacting
at a mole excess of no more than about 5-fold over the
FIGURE 11.16  NHS–iminobiotin can be used to label amine- amount of protein present may result in the best per-
containing molecules, creating amide linkages.
forming biotinylated proteins, especially when using
relatively hydrophobic biotin compounds. Some experi-
amide bond derivatives (Figure 11.16). An iminobioti- mentation may be necessary to determine the optimal
nylated molecule can then be used to target and purify conjugate for a particular application.
other components in biological samples. For instance, 4. React for 30 to 60 min at room temperature or for 3 h
a targeting molecule, such as an antibody, can be imi- at 4°C.
nobiotinylated and allowed to bind its target in com- 5. Remove unreacted NHS-iminobiotin and reaction
plex mixtures (such as tissue sections, cell extracts, or byproducts by dialysis or gel filtration using a
homogenates). The antibody–antigen complex sub- desalting resin.
sequently can be purified using an affinity column of
immobilized avidin with binding at pH 10 to 11 and
simple elution at pH 4 (Orr, 1981; Zeheb et al., 1983). The Sulfo-NHS–SS-Biotin
relatively mild elution condition allows recovery of the Sulfo-NHS–SS-biotin (also known as NHS–SS-
bound antigen without exposure to severely denaturing biotin) is sulfosuccinimidyl-2-(biotinamido)ethyl-
conditions. 1,3-dithiopropionate, a long-chain cleavable bio-
The iminobiotin–avidin interaction can also be uti- tinylation reagent that can be used to modify
lized in the opposite approach. Immobilized imino- amine-containing proteins and other molecules
biotin affinity columns can be used to purify avidin- or (Thermo Fisher). The cross-bridge of the compound
streptavidin-containing complexes under mild elution provides a 24.3-Å spacer arm that creates plenty of
conditions (Hofmann et al., 1980). distance between the modified molecule and the bio-
NHS–iminobiotin is insoluble in aqueous solution. It tin end. Using a long-chain biotinylation reagent can
can be dissolved in organic solvent (DMF) prior to addi- increase the efficiency of biotinylated molecules to bind
tion of a small aliquot to a buffered reaction medium. avidin or streptavidin conjugates, thus enhancing the
Do not exceed 10% DMF in the reaction to avoid protein potential sensitivity of assay systems.

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 489


After molecules modified with sulfo-NHS–SS-biotin
are allowed to interact with avidin or streptavidin
probes, the complexes can be cleaved at the disul-
fide bridge by treatment with 50-mM DTT. Reduction
releases the biotinylated molecule from the avidin
or streptavidin capture reagent without breaking the
(strept)avidin interaction. The use of disulfide biotinyl-
ation reagents thus provides much gentler conditions
to break the complex than would be required if the avi-
din–biotin interaction itself were disrupted (which dis- FIGURE 11.17  Sulfo-NHS–SS-biotin reacts with amine groups to
form amide bonds. The biotin group can be later cleaved off the modi-
sociates only at 6- to 8-M guanidine, pH 1.5). fied molecule by reduction of its internal disulfide linkage.
The use of a cleavable biotinylation reagent also pro-
vides a means to purify targeted molecules using affin-
ity chromatography on a column of immobilized avidin
pH of 7 to 9, avoidance of any amine-containing buffers
or streptavidin. For instance, an antibody modified with
or other components that may compete in the reaction
sulfo-NHS–SS-biotin can be allowed to bind its target in
(including imidazole buffers which catalyze hydrolysis
complex mixtures (such as tissue sections, cell extracts,
of these esters), and avoidance of reducing agents that
or homogenates). The antibody–antigen complex sub-
could cleave the disulfide bridge.
sequently can be isolated using an affinity column of
The following protocol is a suggested method for
immobilized avidin or streptavidin in a “pull-down
biotinylating antibody molecules with sulfo-NHS–
assay” (Knezevic et  al., 2011; Hayashi, 2012). Elution
SS-biotin. Some optimization may have to be done with
from the column with DTT breaks the disulfide bonds,
each application to ensure good biotin incorporation
releasing the antibody and its bound antigen. The iso-
with retention of antigen binding activity. Other pro-
lation of herpes virus proteins (Gretch et  al., 1987) and
teins and amine-containing molecules may be biotinyl-
the recovery of DNA binding proteins (Shimkus et  al.,
ated using similar conditions.
1985) were both carried out using this approach. Other
methods of immunoprecipitation using non-cleavable
biotinylation agents result in the inability to recover Protocol
the captured proteins except under severely denaturing 1. Dissolve the antibody to be biotinylated in 50-mM
conditions. sodium bicarbonate, pH 8.5, at a concentration
Due to the presence of the negatively charged sul- of 10 mg/ml. Other buffers and pH conditions
fonate group, sulfo-NHS–SS-biotin is a water soluble between pH 7 and 9 can be used as long as no
biotinylation reagent that may be added directly to amine-containing buffers like Tris are present. Avoid
aqueous reactions without prior dissolution in organic also the presence of disulfide reducing agents that
solvent. For the addition of small quantities of reagent, can cleave the disulfide group of the biotinylation
the compound may be dissolved in water and an ali- reagent.
quot transferred to the reaction medium. If an aque- 2. Add 0.3 mg of sulfo-NHS–SS-biotin (Thermo Fisher)
ous stock solution of sulfo-NHS–SS-biotin is prepared, to each ml of the antibody solution. To measure
it must be dissolved rapidly and used immediately to out small amounts of the biotinylation reagent,
prevent hydrolysis of the active ester. The NHS ester it may first be dissolved in water (or DMF) at a
reaction forms stable amide linkages with amine-con- concentration of at least 1 mg/ml. Immediately
taining proteins and other molecules (Figure 11.17). transfer the appropriate amount to the antibody
Optimal conditions for the NHS ester reaction include a solution.

BIOCONJUGATE TECHNIQUES
490 11.  (Strept)avidin–Biotin Systems

Note: This level of sulfo-NHS–SS-biotin addition rep- biotinylated cell surface proteins analyzed or isolated
resents about an 8-fold molar excess over the amount of using (strept)avidin reagents.
antibody present. This should result in a molar incor-
poration of approximately two to four biotins per 6.3.  Sulfhydryl-Reactive Biotinylation Agents
immunoglobulin molecule. Some optimization of the bio-
Sulfhydryl-reactive biotinylation reagents allow modi-
tinylation level may have to be carried out to obtain the
fication at cysteine –SH groups or at sites of specific thio-
best performing conjugate for a particular application.
lation within proteins and other molecules. Targeting
3. React for 30 to 60 min at room temperature or for sulfhydryls for modification, as opposed to amines, usu-
2 to 4 h at 4°C. ally results in more limited derivatization, often away from
4. Remove unreacted biotinylation reagent and reaction active centers or binding sites. Directed coupling of biotin
byproducts by dialysis or gel filtration using a in this manner can aid in preserving activity. For instance,
desalting resin. antibodies may be treated by reduction of their disulfide
groups (mainly in the hinge region), which forms free sulf-
Sulfo-NHS–SS-biotin can also be used to label cell sur-
hydryls that are removed from the antigen-binding sites
face proteins for subsequent detection or isolation using
(Chapter  20, Section 1.1). Biotinylation at these sites pro-
(strept)avidin reagents. The negative charge character
duces a derivative that can bind efficiently to both antigen
of the compound prior to its reaction with an amine on
and (strept)avidin probes without steric hindrance.
a protein prevents it from penetrating the cell membrane
Sulfhydryl groups also can be added to 5’-phos-
bilayer. Thus, proteins on the outer surface of the cell can
phate end of DNA probes (Chapter  23, Section 2.2).
be specifically tagged with a biotin group. The disulfide
Biotinylation at these sites avoids disruption of base
cross-bridge of sulfo-NHS–SS-biotin allows recovery of
pairing with complementary DNA targets, since the
labeled proteins after capture on an immobilized (strept)
point of modification is restricted to a single end posi-
avidin support. Reduction of the disulfide using DTT or
tion on the oligonucleotide.
TCEP releases the proteins without the severe denatur-
The following sections discuss three sulfhydryl-
ing conditions usually required to break the (strept)avi-
reactive biotinylation reagents that utilize maleimide-,
din–biotin interaction. This allows isolation of cell surface
pyridyl disulfide-, and iodoacetyl-reactive groups,
proteins under non-denaturing conditions for subsequent
respectively. The maleimide and iodoacetyl options
analysis (Schuberth et al., 1996; DeBlaquiere and Burgess,
produce nonreversible, covalent thioether linkages
1999; Ellerbroek et al., 2001; Jang and Hanash, 2003).
with target –SH groups. The pyridyl disulfide chemis-
The following protocol is based on the method of
try results in disulfide bonds that are reversible through
Thermo Fisher, as found in the instructions for the cell
cleavage with a reducing agent.
surface biotinylation kit.
Biotin–BMCC
Protocol
Biotin–BMCC is 1-biotinamido-4-[4′-(maleimidomethyl)
1. Grow cells in four T75-cm2 flasks until they are
cyclohexane-carboxamido]butane, a biotinylation
90 to 95% confluent.
reagent containing a maleimide group at the end of an
2. Remove the media and wash the cells twice with 8 ml
extended spacer arm (Thermo Fisher). The maleimide
of cold 0.1-M sodium phosphate, 0.15-M NaCl, pH
end reacts with sulfhydryl groups in proteins and other
7.2 (PBS). Note: This buffer contains a high buffer salt
molecules to form stable thioether linkages (Figure
content to stabilize the pH during the biotinylation
11.18). The reaction is highly specific for –SH groups in
reaction. Do not allow the cells to remain in contact
the range of pH 6.5 to 7.5. The long spacer arm (32.6 Å)
with it for more than 5 s to prevent detachment from
provides more than enough distance between modified
the flask surface.
molecules and the bicyclic biotin end to allow efficient
3. Dissolve 12 mg of sulfo-NHS–SS-biotin in 48 ml of
binding of avidin or streptavidin probes.
cold PBS and immediately add 10 ml of the solution
to each flask containing the washed cells.
4. React with gentle rocking for 30 min at 4°C.
5. Quench the reaction by the addition of 1 ml of 1-M
Tris, pH 7.2.
6. Scrape the cells from each flask and transfer them
into a 50-ml tube. Wash each flask using a single
10-ml portion of 0.025-M Tris, 0.15-M NaCl, pH 7.2,
and add the solution to the scraped cells.
7. The isolated cells may be lysed using standard
mechanical or detergent methods and the 

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 491
response in yeast (Wang et  al., 2012), and to study
S-nitrosylation of proteins (Cheng et al., 2012).
The following protocol is a suggested method for
modifying sulfhydryl-containing proteins with biotin–
BMCC. Some optimization of biotinylation levels may
have to be carried out for particular applications.

Protocol
1. Dissolve the protein to be biotinylated (containing
one or more free sulfhydryls) in 0.1-M sodium
phosphate, 0.15-M NaCl, 10-mM EDTA, pH 6.5 to
7.5, at a concentration of 2.5 mg/ml.
2. Dissolve biotin–BMCC (Thermo Fisher) in DMSO at
a concentration of 5 mg/ml.
3. Add 100 μl of the biotin–BMCC solution to each ml of
the protein solution. Mix well.
FIGURE 11.18 Biotin–BMCC provides sulfhydryl reactivity 4. React for at least 2 h at room temperature.
through its terminal maleimide group. The reaction creates a stable 5. Remove excess biotinylation reagent and reaction
thioether linkage. byproducts by dialysis or gel filtration using a
desalting resin.

The reagent is similar to another maleimide-contain- Biotin–HPDP


ing biotinylation reagent, 3-(N-maleimidopropionyl) bio- Biotin–HPDP is N-[6-(biotinamido)hexyl]-3′-(2′-
cytin, a compound used to detect sulfhydryl-containing pyridyldithio)propionamide (Thermo Fisher). The
molecules on nitrocellulose blots after SDS–electropho- reagent contains a 1,6-diaminohexane spacer group
resis separation (Bayer et al., 1987). Biotin–BMCC should which is attached to biotin’s valeric acid side chain. The
be useful in similar detection procedures. terminal amino group of the spacer is further modified
Biotin–BMCC is insoluble in water and must be dis- via an amide linkage with the acid precursor of SPDP
solved in an organic solvent prior to addition to an (Chapter 6, Section 1.1) to create a terminal, sulfhydryl-
aqueous reaction mixture. Preparing a concentrated reactive group. The pyridyl disulfide end of biotin–
stock solution in DMF or DMSO allows transfer of a HPDP can react with free thiol groups in proteins and
small aliquot to a buffer reaction. The upper limit of other molecules to form a disulfide bond with loss of
biotin–BMCC solubility in DMSO is approximately pyridine-2-thione (Figure 11.19). This leaving group
33-mM or 17 mg/ml. In DMF, it is only soluble to a level may be monitored by its characteristic absorbance at
of about 7-mM (4 mg/ml). Upon addition of an organic 343 nm to assess the level of biotinylation. However,
solution of the reagent to an aqueous environment (do since its extinction coefficient is rather low (about
not exceed 10% organic solvent in the aqueous medium 8 ×  103 M−1 cm−1), small-scale biotinylation reactions
to prevent protein precipitation), biotin–BMCC may may not be quantifiable using this technique.
form a micro-emulsion. This is normal, and during the
course of the reaction the remainder of the compound
will be driven into solution as it couples or hydrolyzes.
The required sulfhydryl groups for biotin–BMCC
modification may be indigenous in molecules, formed
through reduction of disulfides or created by the use of
thiolation reagents (Chapter  2, Section 4.1). At physi-
ological pH, the rate of the maleimide reaction toward
sulfhydryls is almost 1000-fold faster than its reac-
tion toward amines. However, at higher pH values
the maleimide will couple to amines quite readily (Wu 
et al., 1976; Ishi and Lehrer, 1986). Maleimides can also Modifications carried out with biotin–HPDP produce
undergo a ring-opening hydrolysis reaction which biotinylated compounds with long spacer arms (29.2 Å),
increases in rate with pH, effectively inactivating the which typically provide good binding efficiency with
reactive group for thiol coupling. avidin or streptavidin probes. After coupling to sulf-
Biotin–BMCC has been used to investigate palmi- hydryl-containing molecules, the biotin–HPDP com-
toylation (Fairbank, 2012), to study the heat-shock ponent can be cleaved back off by treatment with

BIOCONJUGATE TECHNIQUES
492 11.  (Strept)avidin–Biotin Systems

reaction include a pH range of 6 to 9 in buffer systems


that do not contain any extraneous sulfhydryl com-
pounds or reducing agents such as DTT, 2-mercapto-
ethanol, or TCEP. If reducing agents are used to create
sulfhydryls in the protein to be biotinylated, these must
be completely removed by dialysis or gel filtration
before reacting with biotin-HPDP.
A suggested protocol for the use of biotin–HPDP in
the modification of sulfhydryl-containing proteins fol-
lows. Similar procedures may be used when biotinylat-
ing other molecules containing thiols.

Protocol
1. Dissolve the sulfhydryl-containing protein to be
biotinylated in 0.1-M sodium phosphate, 0.15-M
NaCl, 10-mM EDTA, pH 7.2, at a concentration of at
least 2 mg/ml.
FIGURE 11.19  Biotin–HPDP reacts with sulfhydryl-containing 2. Dissolve biotin–HPDP (Thermo Fisher) in DMSO at a
molecules through its pyridyl disulfide group, forming reversible concentration of 4-mM (2.1 mg/ml).
disulfide bonds. The biotin group may be released from modified 3. Add 100 μl of the biotin–HPDP stock solution to each
molecules by reduction with DTT.
ml of the protein solution. Mix well.
4. React for 90 min at room temperature.
disulfide reducing agents, such as DTT. Breaking this 5. Purify the biotinylated protein by gel filtration using
bond releases the biotin modifications and regenerates a desalting resin or by dialysis. The PBS/EDTA
the original sulfhydryl-containing molecule. This cleav- buffer described in step 1 is suitable for either
ability also provides a means of recovering target com- operation.
plexes after purification of the biotinylated molecules
by affinity chromatography on immobilized avidin or Iodoacetyl–LC-Biotin
streptavidin. Thus, biotin–HPDP-modified antibod- Iodoacetyl–LC-biotin is N-iodoacetyl-N-biotinyl­
ies directed against some specific cellular antigen can hexylenediamine, a sulfhydryl-reactive biotinylation
be used to aid in the isolation of targeted components agent (Thermo Fisher). The reagent contains a 1,6-diami-
using affinity chromatography (immunoprecipitation) nohexane spacer group that is attached to biotin’s valeric
followed by elution with a disulfide reductant. acid side chain. The terminal amino group of the spacer
Using a similar approach, C1q has been modified is further modified via an amide linkage with an iodoace-
with biotin–HPDP and allowed to interact with its spe- tyl group to provide the sulfhydryl reactivity. Coupling to
cific receptor. Subsequent purification of the C1q recep- sulfhydryl-containing proteins or other molecules creates
tor was accomplished through binding to immobilized nonreversible thioether bonds (Figure 11.20). Modifications
streptavidin (Chapter  15) and subsequent cleavage of carried out with iodoacetyl–LC-biotin produce bioti-
the disulfide bridge of the biotinylation reagent using nylated compounds with sufficiently long spacer arms
DTT (Ghebrehiwet et al., 1988). Similar pull-down assays (27.1 Å), which help to ensure excellent binding potential
continue to be done with biotin–HPDP to study protein with avidin or streptavidin probes.
interactions in cells (Seth and Stamler, 2011) or to ana-
lyze post-translational modifications (Lee et al., 2011).
Biotin–HPDP is water insoluble and therefore must
be dissolved in an organic solvent prior to addition to
an aqueous reaction medium. Suitable solvents include
DMSO and DMF. Concentrated stock solutions may be
prepared in DMSO and a small aliquot transferred to a
buffered reaction solution. Do not add more than 10%
organic solvent to the aqueous reaction to prevent pre-
cipitation or denaturation of biological molecules. After
addition, a micro-emulsion may result. This is normal
for many water-insoluble reagents. The solution usu- 
ally will become clearer during the course of the reac- Iodoacetyl–LC-biotin is water insoluble and there-
tion. Optimal conditions for the disulfide interchange fore must be dissolved in an organic solvent prior to

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 493
Protocol
1. Dissolve the sulfhydryl-containing protein to be
biotinylated in 50-mM Tris, 0.15-M NaCl, 10-mM
EDTA, pH 8.3, at a concentration of 4 mg/ml.
2. Dissolve iodoacetyl–LC-biotin (Thermo Fisher) in
DMF at a concentration of 4-mM (2 mg/ml). Protect
from light.
3. Add 50 μl of the iodoacetyl–LC-biotin solution to
each ml of the protein solution. Mix well. This level
of addition represents a 3.28-fold molar excess of
biotinylation reagent over the quantity of protein
present if the protein has a molecular weight of
67,000 and possesses one sulfhydryl. Adjustments
to the amount of reagent addition may have to be
made to be appropriate for other proteins of different
molecular weight. Consideration of the number of
sulfhydryls present per protein molecule should
also be carried out. React the biotinylation reagent
at no more than a 3- to 5-fold molar excess over the
FIGURE 11.20  This biotinylation reagent reacts with sulfhydryl
groups through its iodoacetamide end to form thioether bonds.
amount of sulfhydryls present to ensure specificity
of the iodoacetyl group for only –SH groups. Higher
ratios of reagent-to-protein may cause reaction with
addition to an aqueous reaction medium. Suitable sol- amine groups present on the protein.
vents include DMSO and DMF. Concentrated stock 4. React for 90 min in the dark at room temperature.
solutions may be prepared in DMSO and a small ali- 5. Remove excess reactants and reaction byproducts by
quot transferred to a buffered reaction solution. Do dialysis or gel filtration using a desalting resin.
not add more than 10% organic solvent to the aqueous
reaction to prevent precipitation or denaturation of
biological molecules. After addition, a micro-emulsion 6.4.  Carbonyl- or Carboxyl-Reactive
may result. This is normal for many water-insoluble Biotinylation Agents
reagents. The solution usually will become clear during
the course of the reaction. Optimal conditions for cou- Hydrazide- or amine-containing biotinylation com-
pling using iodoacetyl-containing reagents include a pounds can be used to modify carbonyl or carboxyl
pH range of 7.5 to 8.5 in buffer systems that do not con- groups on other molecules. Hydrazides spontane-
tain any extraneous sulfhydryl compounds. In addition, ously react with aldehydes or ketones to give hydra-
protect all solutions containing iodoacetyl–LC-biotin zone linkages. The reaction may be further accelerated
from light, since photolysis may cause liberation of by the addition of an aniline catalyst (see Chapter  15,
iodine, degrading the activity of the compound and Section 2.4, for additional information on this reaction
possibly causing modification of tyrosine or histidine mechanism). The resulting hydrazone bonds may be
residues by iodination. further stabilized by reduction with sodium cyanobo-
Iodoacetyl–LC-biotin has been used to localize the rohydride. The amine-containing biotinylation reagents
SH1 thiol of myosin by use of an avidin–biotin complex (or the hydrazide ones) may be coupled to carboxyl-
visualized by electron microscopy (Sutoh et al., 1984), to ate groups using a carbodiimide reaction (Chapter  4,
determine the spatial relationship between SH1 and the Section 1.1). In addition, amine- or hydrazide-contain-
actin binding site on the myosin subfragment-1 surface ing biotinylation reagents may be coupled to cytosine
(Yamamoto et  al., 1984), to study markers for pancre- residues in DNA or RNA by transamination catalyzed
atic cancer (Zhu et al., 2012), and to assay protein–DNA by bisulfite (Chapter 23, Section 2.3).
interactions (Ritzefeld and Sewald, 2012).
The following protocol is a suggested method for Biotin–Hydrazide and Biotin–LC-Hydrazide
biotinylating sulfhydryl-containing proteins using iodo- Biotin–hydrazide is cis-tetrahydro-2-oxothieno[3,4-d]­
acetyl–LC-biotin. The required sulfhydryl groups may -imidazoline-4-valeric acid hydrazide, the hydrazine
be provided through reductive cleavage of disulfide derivative of d-biotin off its valeric acid carboxylate
bonds or by the use of thiolation reagents (Chapter  2, (Thermo Fisher). The hydrazide functionality reacts
Section 4.1). Other molecules may be modified with with aldehyde and ketone groups to give hydrazone
iodoacetyl–LC-biotin using similar techniques. linkages. Although formyl groups are not common in

BIOCONJUGATE TECHNIQUES
494 11.  (Strept)avidin–Biotin Systems

biological molecules, they may be created by oxidation


of diols with sodium periodate (Chapter  2, Section 4.4).
Thus, glycoconjugates may be targeted specifically at
their sugar residues. Biotinylation of these oxidized car-
bohydrates with biotin–hydrazide produces modifica-
tions that may be away from active centers or binding
sites on proteins (Figure 11.21). Particularly, immuno-
globulins may be biotinylated with this reagent at their
polysaccharide groups which typically are present in the
Fc region of the IgG molecule. Directed modification in
this manner avoids the antigen-binding sites at the ends
of the heavy and light chains, thus preserving antibody
activity and allowing avidin or streptavidin probes
to dock without blocking or interfering with antigen
binding (although care should be taken in this respect,
as some antibodies contain carbohydrate near their
­antigen-binding sites).

FIGURE 11.21  Biotin–hydrazide can be used to label aldehyde-


containing molecules, creating hydrazone bonds.

6-aminocaproic acid extension off its valeric acid group


(Thermo Fisher). The increased length of this spacer
(24.7  Å) provides more efficient interaction potential
with avidin or streptavidin probes, possibly increasing
the sensitivity of assay systems. The reactions of biotin–
LC-hydrazide are identical to those of biotin–hydrazide.
Both biotin–hydrazide and biotin–LC-hydrazide can
be used to identify sites of carbonylation in biomol-
ecules, such as those caused by oxidative stress. These
sites typically involve some formation of aldehyde and
ketone groups, which can be specifically tagged using
 hydrazide-containing biotinylation agents. The hydra-
Biotin–hydrazide may also be used to couple with zide group forms a hydrazone bond at the sites of car-
carboxylate-containing molecules. Hydrazides can be bonylation, thus allowing detection using streptavidin
coupled with carboxylic acid groups by using the carbodi- conjugates or isolation by using immobilized strepta-
imide reaction (Chapter 4, Section 1.1). The carbodiimide vidin in pull-down assays (Frohnert et al., 2011; Curtis
activates a carboxylate to an o-acylisourea intermedi- et al., 2012).
ate. Biotin–hydrazide can react with this intermediate via The following protocol describes the use of biotin–
nucleophilic addition to form a stable covalent bond. hydrazide to label glycosylated proteins at their car-
Biotin–hydrazide has been used to biotinylate bohydrate residues. Control of the periodate oxidation
antibodies at their oxidized carbohydrate residues level can result in specific labeling of sialic acid groups
(O’Shanessy et  al., 1984, 1985, 1987; Hoffman and or general sugar residues (Chapter 2, Section 4.4).
O’Shannessy, 1988), to modify the low-density lipopro-
tein (LDL) receptor (Wade et  al., 1985), to biotinylate Protocol
nerve growth factor (NGF) (Rosenberg et  al., 1986), and 1. Dissolve a periodate-oxidized glycoprotein (i.e.,
to modify cytosine groups in oligonucleotides to produce antibodies—see Chapter 20, Section 1.3) in 0.1-M
probes suitable for hybridization assays (Reisfeld et  al., sodium phosphate, 0.15-M NaCl, pH 7.4, at a
1987) (Chapter 23, Section 2.3). concentration of 2 mg/ml. Note: The buffer, 0.1-M
An analog of this biotinylation reagent with a longer sodium acetate, pH 5.5, is typical of literature
spacer arm also exists. Biotin–LC-hydrazide contains a references for reaction of a hydrazide compound

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 495
with an aldehyde-containing molecule to form a
hydrazone linkage. Alternative buffer conditions
using higher pH values also work well. Physiological
pH conditions with the use of a reducing agent such
as sodium cyanoborohydride (step four) produce the
most efficient labeling yields when using hydrazide-
containing reagents.
2. Add biotin–hydrazide or biotin–LC-hydrazide to a
final concentration of 5-mM.
3. React for 2 h at room temperature.
4. To reduce the hydrazone bonds to more stable
linkages, cool the solution to 4°C and add an equal
volume of 30-mM sodium cyanoborohydride in
PBS. Incubate for 40 min. Note: If the presence of
a reducing agent is detrimental to protein activity,
eliminate this step. In most cases, the hydrazone
linkage is stable enough for avidin–biotin detection
experiments.
5. Remove excess reactants by dialysis or gel filtration
using a desalting column. FIGURE 11.22  Biocytin hydrazide reacts with aldehyde-contain-
ing molecules to form hydrazone bonds.
Biocytin Hydrazide
Another biotinylation reagent that can spontane- treatment with neuraminidase (Chapter  2, Section 4.4).
ously couple with aldehyde- or ketone-containing The use of this approach for labeling glycoproteins
molecules is biocytin hydrazide (Thermo Fisher). in situ was found to be optimal, due to the other poten-
Produced by forming the hydrazine derivative of bio- tial side-reactions that may occur when using sodium
cytin—a lysine–biotin complex often found naturally periodate.
in serum (Section 6.2, this chapter)—the compound The reactivity and use of biocytin hydrazide is simi-
has better solubility in aqueous solutions than either lar to that described for biotin–hydrazide in Section 6.4,
biotin–hydrazide or biotin–LC-hydrazide discussed this chapter. The following protocol for labeling glyco-
previously. The solubility enhancement of biocytin proteins at oxidized carbohydrate (galactose) sites is
hydrazide is due to the presence of lysine’s α-amino from Bayer and Wilcheck (1992).
group, which is protonated and positively charged at
physiological pH. The reagent can be used to label car- Protocol
bohydrate-containing molecules, such as glycoproteins, 1. Dissolve the glycoprotein to be labeled in 0.1-M
after they have been oxidized to contain reactive alde- sodium phosphate, 0.15-M NaCl, pH 7.4, containing
hydes (Chapter  2, Section 4.4). The hydrazide group 1-mM CaCl2 and 1-mM MgCl2 (labeling buffer), at a
forms a hydrazone linkage with the aldehydes, thus concentration of 1 mg/ml.
directing the biotinylation reaction toward the polysac- 2. Dissolve biocytin hydrazide (Thermo Fisher) in
charide regions of glycoconjugates (Figure 11.22). 0.1–M sodium phosphate, 0.15-M NaCl, pH 7.4
(PBS), at a concentration of 20 mg/ml.
3. To each ml of glycoprotein solution, add 30 μl
of neuraminidase (1 unit/ml as supplied by
Behringwerke AF), then 30 μl of galactose oxidase
(previously dissolved at 100 units/ml in the labeling
buffer of step one), and finally 100 μl of the biocytin
hydrazide solution.
4. React for 2 h at 37°C.
5. Remove unreacted reagents by dialysis or gel
filtration.

Biocytin hydrazide was used to label specifically 5-(Biotinamido)pentylamine
sialic acid residues, galactose residues, and for general The d-biotin derivative, 5-(biotinamido)pentyl-
sugar modification (Bayer et  al., 1988). The galactose amine,  contains a 5-carbon cadaverine spacer group
residues were oxidized using galactose oxidase after attached to the valeric acid side chain (Thermo Fisher).

BIOCONJUGATE TECHNIQUES
496 11.  (Strept)avidin–Biotin Systems

The compound can be used in a carbodiimide reac- post-translational protein modification as well as vari-
tion process to label carboxylate groups in proteins ous disease states.
and other molecules, forming amide bond linkages 5-(Biotinamido)pentylamine is able to participate
(Chapter  4, Section 1). However, the main use of this in the acyltransferase reaction, becoming covalently
biotinylation reagent is in the determination of factor attached to protein substrates at their glutamine resi-
XIIIa or transglutaminase enzymes in plasma, cell, or dues (Figure 11.23). Lee et  al. (1988) used this biotinyl-
tissue extracts. ation reagent to quantify factor XIII in plasma (D’Eletto
et al., 2012; Kuper et al., 2012). Transglutaminase activity
resulted in the modification of an N,N’-dimethylcasein
substrate which was subsequently detected by an avi-
din–biotin assay procedure. The assay may be carried
out in microplates using wells coated with the substrate
protein and quantifying the enzyme activity with strep-
tavidin–alkaline phosphatase (Slaughter et  al., 1992).
Jeon et  al. (1989) subsequently applied the assay to the
measurement of transglutaminase activity in cells.
Components biotinylated in cellular systems also can be
 isolated by use of affinity chromatography on immobi-
Factor XIII, also known as plasma transglutamin- lized avidin (Lee et al., 1992).
ase, is an enzyme of the blood coagulation cascade. It
is activated by thrombin and calcium to factor XIIIa, at 6.5.  Photoreactive Biotinylation Agents
which point it catalyzes covalent crosslinks between the
ε-amine group of lysine side chains and the γ-glutamyl Biotin derivatives containing a photoreactive group
side chain of glutamine residues. Abnormal levels of provide nonselective biotinylation potential at certain
factor XIII in plasma are clinically important, being reactive hydrogen sites or nucleophilic groups. They
associated with cancer, liver or renal dysfunction, or can be used to incorporate an avidin-binding bio-
various bleeding disorders. The assay of transgluta- tin group into molecules that do not possess amines,
minase activity therefore is important for investigating sulfhydryls, or other easily modifiable functional
the activity and function of this enzyme as it relates to groups. Many of these photoreactive derivatives

FIGURE 11.23  5-(Biotinamido)pentylamine can


be used to label glutamine residues in proteins by
enzymatic action of transglutaminase.

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 497
utilize the phenyl azide-type of photosensitive group, used to detect flavivirum RNA in infected cells (Khan
which can be activated by exposure to UV light to an and Wright, 1987), to detect single-copy genes and low-
intermediate nitrene or the nucleophile-reactive dehy- abundance mRNA (McInnes et  al., 1987), for the diag-
droazepine (Chapter  3, Section 7.1, and Chapter  6, nosis of barley yellow dwarf virus (Habili et  al., 1987),
Section 3). However, additional photoreactive groups to assay luteinizing hormone β mRNA in individual
that are also useful include a psoralen ring system gonadotropes (Childs et  al., 1987), to perform DNA
and a benzophenone group. A psoralen-based bio- mapping using a cross-hybridization technique (Chetrit
tinylation agent is presented in this section, while a et al., 1989), to create patterns of fluorescent streptavidin
benzophenone-containing biotin compound with a molecules on dextran-coated surfaces (Ahn et al., 2011),
water soluble PEG spacer is discussed in Chapter  18, and to form multilayer films on titanium dioxide sub-
Section 1.3. strates (Weng et al., 2011).
Photobiotin can be dissolved in water or buffer at
Photobiotin a concentration of 1 mg/ml and stored in the dark at
Perhaps the most common photoreactive biotin −20°C until needed. As long as no exposure to light is
derivative is N-(4-azido-2-nitrophenyl)-aminopropyl- permitted, the compound is stable for at least 1 year
N’-(N-d-biotinyl-3-aminopropyl)-N’-methyl-1,3- under these conditions.
propanediamine, simply called photoactivatable biotin The protocol for modifying DNA probes with pho-
or photobiotin (Forster et al., 1985) (Thermo Fisher). The tobiotin can be found in Chapter  23, Section 2.3. It is
compound contains a 9-atom diamine spacer group on based on the method of Forster et al. (1985). The follow-
the biotin valeric acid side chain at one end, while the ing method is a suggested protocol for the modifica-
other end of the spacer terminates in an aryl azide reac- tion of proteins using a photoreactive biotin derivative.
tive group. The presence of a nitro group on the phe- Some optimization may be necessary to obtain the best
nyl azide ring allows for photoactivation at higher UV incorporation levels.
wavelengths approaching the visible region of the spec-
trum, thus avoiding potential breakdown of biological
molecules through UV exposure. Photolyzing with light
at a wavelength of 350 nm causes rapid activation with
nitrene formation. The nitrene can couple to replace-
able hydrogen sites in target molecules, add to double
bonds within van der Waals distance, or undergo ring
expansion to the dehydroazepine. If ring expansion
occurs, the principal target group for coupling is a
nucleophile, such as a primary amine (Figure 11.24).


Photobiotin has been used to biotinylate numer-
ous macromolecules, including proteins and nucleic
acids. The biotinylation of alkaline phosphatase was
carried out with complete retention of activity (Forster
et  al., 1985). Tubulin was labeled with photobiotin and
detected on dot blots down to a level of 10 pg of sam-
ple using an avidin–enzyme conjugate (Lacey and
FIGURE 11.24  Photobiotin can be made to couple spontaneously
Grant, 1987). DNA and RNA were labeled for use in with nucleophiles by exposure to UV light. The phenyl azide ring
hybridization assays (Forster et  al., 1985; Keller, 1989). undergoes ring expansion to a highly reactive dehydroazepine inter-
For instance, photobiotin-modified probes have been mediate, which can react with amines.

BIOCONJUGATE TECHNIQUES
498 11.  (Strept)avidin–Biotin Systems

Protocol The psoralen photoreactive group provides better inser-


1. Dissolve the protein to be biotinylated at a tion yields than typical phenyl azide-based systems,
concentration of at least 1 mg/ml in water or dilute such as the standard photobiotin probe discussed previ-
buffer at neutral pH. ously in this section.
2. In subdued light, dissolve photobiotin (Thermo
Protocol
Fisher) in water at a concentration of 1 mg/ml.
3. Add a quantity of photobiotin solution to the protein 1. Dissolve the DNA sample to be modified at a
solution to give at least a 5-fold molar excess of concentration of 20 to 100 μg/ml in 10-mM Tris, 1-mM
biotinylation reagent. EDTA, pH 7.4. Note: The sample may be heated to
4. Place in an ice bath and irradiate from above (about denature and solubilize genomic DNA and then
10 cm away) for 15 min using a sunlamp (such cooled to form dsDNA for modification.
as Philips Ultrapnil MLU 300 W, General Electric 2. Dissolve the psoralen–PEG3–biotin reagent in DMF
sunlamp RSM 275 W, or National Self-Ballasted at a concentration of 20-mM (use a fume hood).
BHRF 240–250 V 250 W W-P lamp). Protect from light.
5. Remove excess photobiotin by dialysis or gel 3. Add a quantity of the psoralen–PEG3–biotin solution
filtration using a desalting column. to the DNA solution to result in a final concentration
of 200 μM. Mix well.
Psoralen–PEG3–Biotin 4. Expose the solution to long-wavelength UV light at
Psoralen–PEG3–biotin is a photoreactive biotinyl- about 365 nm (Philips TL 20 W/09 UV light works
ation reagent containing a psoralen group at one end well) for 10 to 30 min. The solution may be cooled
and a triethylene glycol (PEG-based) spacer in the mid- on ice to prevent heating during the irradiation
dle (Thermo Fisher). This compound is water soluble process.
due to the presence of the hydrophilic PEG arm. It is 5. Precipitate the sample to remove unreacted
able to photo-insert into double-stranded DNA and to a biotinylation reagent by adding 0.1-M potassium
lesser extent into double-stranded regions of RNA. The acetate and ethanol (1 : 2 ratio). Centrifuge and wash
reaction occurs upon exposure to UV light in the range the biotinylated DNA pellet with ethanol, then
of 320 to 400 nm, which forms an excited triplet state dry it under nitrogen. The purified sample may be
intermediate that can insert in certain double-bond dissolved in water or buffer.
structures, especially at the 5,6-double bond of thymine
bases. 6.6.  Active Hydrogen-Reactive
The psoralen ring system can intercalate within dou- p-Aminobenzoyl Biocytin, Diazotized
ble-stranded DNA or RNA and induce the formation of
adducts with adjacent thymine bases (Stutz et al., 2011) p-Aminobenzoyl biocytin contains a 4-aminoben-
(Figure 11.25). The furan side and pyrone side of the zoic acid amide derivative off the α-amino group
tricyclic rings in psoralen can both form cycloaddition of biocytin (Section 6.2, this chapter) lysine residue
products with the 5,6-double bond of thymine residues, (Thermo Fisher). The aromatic amine can be treated
which results in crosslinks between the DNA strands with sodium nitrite in dilute HCl to form a highly reac-
with a PEG–biotin label sticking out. tive diazonium group (Figure 11.26), which is able to
Psoralen–PEG3–biotin has been used to label dou- couple with active hydrogen-containing compounds.
ble-stranded DNA for detection using (strept)avidin A diazonium reacts rapidly with histidine or tyro-
reagents (Henriksen et al., 1991; Wygrecka et al., 2007). sine residues within proteins, forming covalent diazo

36.86 Å

O O
HN
O O O
NH
O O O
N O N
H H S
9.24 Å O
Psoralen–PEG3–Biotin
MW: 688.79

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 499
O O
HN
O O O
NH
O O O
N O N
H H S
O
Psoralen–PEG3–Biotin

Double-stranded
DNA containing thymidine nucleotides

UV Light

DNA
O O
O N HN
O O O
NH
HN O O O
N O N
CH3 H H S
O
O
H3C
Intercalation and cycloaddition
O to thymine bases
N
DNA
HN
O

FIGURE 11.25  The photoreactive compound psoralen–PEO3–biotin can intercalate into double-stranded DNA or RNA segments and cova-
lently link to thymine bases via a photoreaction process.

bonds (Wilchek et  al., 1986) (Figure 11.27). It can also


react with guanidine residues within DNA at posi-
tion eight of the base (Rothenberg and Wilchek, 1988)
(Figure 11.28). Biotinylation via diazo linkages is
reversible by treatment with a 10-fold molar excess
of Na2S2O4 (sodium dithionite) in 50-mM Tris, pH
8.5 (Gorecki et  al., 1971) (Chapter  3, Section 6.1, and
Chapter 5, Section 9).

FIGURE 11.26  The aminophenyl group of this biotin derivative


can be transformed into a diazonium reactive group by treatment
with sodium nitrite in dilute HCl. 

BIOCONJUGATE TECHNIQUES
500 11.  (Strept)avidin–Biotin Systems

FIGURE 11.27  The diazonium group of p-diazobenzoylbiocytin can react with tyrosine or histidine residues in proteins to form diazo bonds.

The following procedure describes the process for cre- Protocol for Biotinylation of Proteins on
ating a diazonium derivative of p-aminobenzoyl biocytin Blots Using the Diazonium Derivative of
along with the subsequent coupling of the activated spe- p-Aminobenzoyl Biocytin
cies to a protein or a nucleic acid probe. 1. Dilute the diazonium derivative of p-aminobenzoyl
biocytin with 0.2-M sodium borate, pH 8.4, to a
Protocol for Formation of the Diazonium concentration of 10 μg/ml.
Derivative 2. Transfer proteins onto a nitrocellulose membrane
1. Dissolve 2 mg of p-aminobenzoyl biocytin (Thermo using any appropriate procedure, including dot
Fisher) in 40 μl of 1-N HCl (concentration of blotting the protein solution onto the surface.
50 mg/ml). Cool the solution on ice. 3. Incubate the membrane with the biotin derivative at
2. Dissolve 7.7 mg of sodium nitrite in 1 ml of ice-cold a ratio of 1 ml/cc3 of membrane.
water. Prepare fresh. 4. React for 1 h at room temperature.
3. Mix 40 μl of the p-aminobenzoyl biocytin solution 5. Wash the membrane thoroughly with 0.1-M Tris,
with 40 μl of the sodium nitrite solution. 0.15-M NaCl, pH 7.5.
4. React for 5 min on ice to create the diazonium 6. Block nonspecific sites on the membrane with an
derivative. appropriate blocking component (such as BSA) and
5. Stop the reaction by the addition of 35 μl of 1-N detect the biotinylated proteins using an avidin or
NaOH. Use immediately for biotinylation. streptavidin conjugate.

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 501
O
H 2N N NH 2 HN
O
+ NH
HO
S
2,6-Diaminopyridine D-Biotin
(in excess)

EDC, NHS

H2N O
HN
N O
NH
N
H S
BAP
Biotinylated diaminopyridine
Mol. Wt.: 335.43

FIGURE 11.29  The synthesis of BAP can be done by the reaction


of an excess of diaminopyridine with biotin in the presence of EDC
and NHS.

then be reacted with amine- or hydrazide-containing bio-


tinylation compounds to couple with the open aldehyde
group at the reducing end, thus forming a hydrazone
FIGURE 11.28  The diazonium group of p-diazobenzoylbiocytin linkage. The hydrazone bond may be reduced to stabi-
can couple to the C-8 position of guanidine bases in nucleic acids,
lize the bond (recommended when reacting with amine-
forming diazo bonds.
containing biotin compounds) or left as the unreduced
hydrazone, which typically is carried out when coupling
with hydrazide-biotin compounds. Alternatively, the
6.7.  Glycan Biotinylation Reagents reducing end of a carbohydrate can be reacted with an
Biotinylated oligosaccharides are convenient probes amine to form a glycosylamine derivative without open-
of carbohydrate interactions, because the biotin label ing the hemiacetal ring, thus better preserving the native
can be captured or detected using an avidin or strep- structure of a glycan, which is important in some studies
tavidin derivative. For instance, immobilized strepta- involving protein interactions.
vidin can be used to purify glycoconjugates that have A recent addition to the methods of glycan biotinyl-
been labeled with a biotin group, potentially isolating ation makes use of the Staudinger ligation reaction with
glycoproteins or carbohydrate binding proteins (see a phosphine–biotin derivative (see also Chapter  17,
Chapter  15). Enzyme-labeled or fluorescently labeled Section 6). Carbohydrates containing azide derivatives
avidin or streptavidin can be used to probe for biotin- have been modified with this biotin compound to probe
labeled carbohydrates in cells or tissue samples. In for glycoconjugates in vivo. This reaction is particularly
addition, a biotinylated glycan can be displayed on avi- useful for doing cell-based assays, because the ligation
din or streptavidin to make an immunogen for develop- reaction is completely orthogonal to any biological reac-
ing specific antibodies to the carbohydrate. tions or interactions.
Complex glycans on glycoproteins or other carbohy- The following sections describe fluorescent biotinyl-
drate-bearing molecules can be modified with a bioti- ation reagents that can be used to study carbohydrate
nylation reagent using a number of reaction strategies. function and interactions.
Oxidation with sodium meta periodate can be used to
create aldehyde residues from diols on sugars, and this Biotinylated Aminopyridine
technique has been used to specifically modify sialic acids BAP (biotinylated aminopyridine or 2-amino-(6-ami-
on glycans by reductive amination with a biocytin-hydra- dobiotinyl)pyridine) is a derivative of d-biotin made by
zide compound (see Section 6.4, this chapter) (Bayer et al., reacting the NHS ester of this vitamin with 2,6-diami-
1988). Other procedures make use of released glycans nopyridine (DAP) in large molar excess, typically car-
from glycoproteins or other glycoconjugates, which con- ried out using a carbodiimide EDC/NHS reaction
tain reducing ends upon cleavage. The reducing ends can (Figure 11.29). The resultant compound has fluorescent

BIOCONJUGATE TECHNIQUES
502 11.  (Strept)avidin–Biotin Systems

OH containing biotin. BAP–carbohydrate adducts have


H2 N O
R O been shown to be high affinity binders of both strep-
O OH HN
HO + N O
NH tavidin and avidin, despite the relatively short spacer
NH
O N afforded by the diaminopyridine–biotin linker (Toomre
H
CH 3
S and Varki, 1994).
N-Acetyl glucosamine
BAP The biotin group of BAP makes the compound some-
Biotinylated diaminopyridine
residue at glycan what hydrophobic, but attached to glycans the conju-
reducing end
gates should display good water solubility due to the
Sodium cyanoborohydride
abundance of hydroxyl groups in addition to potentially
or borane dimethylamine having other charged groups on the sugars. BAP solubil-
O ity in water was reported to be about 1 mg/ml, but with
the addition of less than 1% DMSO, this solubility can be
HN NH increased more than 10-fold (Toomre and Varki, 1994).
OH
Fluorescence of the diaminopyridine group allows
R OH H
O N O S detection of conjugates down to the picomole range,
HO
NH N with excitation and emission maxima at 345 nm and
O NH 400 nm, respectively. For detection of BAP and its conju-
CH 3 gates, the optimal buffer environment is less than pH 5,
Fluorescent glycan–BAP Conjugate because its fluorescent properties are pH dependent. A
preferred buffer is sodium acetate at pH 4.
FIGURE 11.30  BAP can be used to label the reducing end of After BAP conjugation to saccharides or glycans, sep-
released glycans by reductive amination in the presence of a reducing
aration of the conjugates and unreacted BAP can be fluo-
agent.
rescently followed using size exclusion chromatography
(SEC) on a TSK-G3000PW column, which successfully
properties due to the present of the aminopyridine ring, resolves most of the lower molecular weight conjugate
and its remaining free amine group may be used to species, including single-sugar adducts through three-
modify reducing saccharides and glycans by reductive sugar carbohydrates. BAP–glycan conjugates containing
amination (Figure 11.30). BAP can be used to label oli- more than three sugars elute early in the separation and
gosaccharides under mild conditions and without the do not resolve into discrete peaks, as smaller adducts
carbohydrate structural degradation that results using do. Unreacted BAP elutes last in the SEC separation.
periodate oxidation of carbohydrates. After modifica- Alternatively, separations can be carried out by anion-
tion, the glycans or carbohydrates can be analyzed by exchange chromatography using a column packed with
chromatography, electrophoresis, or mass spectrometry the HPLC support TSK-DEAE-2SW, which effectively
(Harvey, 2011; Nakano et al., 2011). resolves negatively charged carbohydrates, such as those
Rothenberg et  al. (1993) demonstrated the utility of containing sialic acid residues. BAP–glycan conjugates
BAP for highly sensitive fluorescence detection and sepa- will elute according to their degree of negative charge
ration of oligosaccharides by reverse-phase HPLC, with character. Sulfate-containing sugars in general will inter-
limits of detection down to about the 50-femtomole level act more strongly with the matrix and have longer reten-
(low picomole levels if using a cuvette reader with a 1-cm tion times than those containing only carboxylates.
path length). Toomre and Varki (1994) subsequently pub- BAP may be prepared by the reaction of NHS–biotin
lished an improvement on the synthesis and use of the with DAP. The biotinylation compound is commercially
BAP reagent. In addition, the biotin group of BAP-labeled available (Thermo Fisher) or it may be formed in situ by
glycans can be used to create neoglycoproteins by interac- reaction of d-biotin with EDC and NHS. An optimized
tion with tetrameric avidin or streptavidin molecules. The protocol for preparation of the reactive intermediate
resultant glyco-complexes have been shown to be potent ester and the final BAP compound can be determined
immunogens for evoking an IgG immune response in from Rothenberg et  al. (1993) with modifications by
mice toward the glycan components (Srikrishna, 2001). Toomre and Varki (1994). Basically, a solution of 0.3-M
BAP-modified glycans also can be used to probe DAP is prepared in 40 ml of 50-mM MES, pH 6.5, and
for receptors or binding proteins, which can then be 10 ml of 0.1-M d-biotin in DMSO is added. The reac-
detected by use of streptavidin conjugates or isolated tion is initiated by the addition of EDC and NHS to a
by affinity chromatography on immobilized strep- final concentration of 150-mM and 50-mM, respectively.
tavidin or immobilized monomeric avidin (Thermo The reaction is allowed to continue overnight at room
Fisher). The use of immobilized monomeric avidin is temperature with mixing before purification of BAP
convenient, because the biotinylated glycans can be on a C18 sample prep cartridge. The reaction mixture is
released by elution with acid pH or by using a solution applied to the cartridge and reaction byproducts and

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 503
DAP removed by washing with water and 10% aceto-
nitrile. BAP is finally eluted in high purity by washing
with 50% acetonitrile. O
The conjugation of BAP to oligosaccharides can O
HN
be achieved by the following protocol based on the H NH
N
method of Toomre and Varki (1994). H2N N
H S
O
Protocol
BNAH
1. Dissolve an oligosaccharide or glycan having a Biotinyl-L-3-(2-naphthyl)-alanine hydrazide
reducing end to be modified in 2 : 1 pyridine/glacial Mol. Wt.: 455.57
acetic acid (vol/vol) with a total reaction volume of 
10 to 100 μl. If the carbohydrate initially is insoluble In some cases, the ability to modify glycans at the
in the reaction solution, a prior dissolution in a reducing end without reduction preserves the carbo-
minimal amount of DMSO or water can be carried hydrate’s native structure sufficiently to allow interac-
out and then an aliquot transferred to the reaction tions with proteins that would otherwise not interact
medium. if the bond were reduced. Therefore, depending on the
2. Add to the solution a 50-fold molar excess of BAP ultimate use of the biotinylated carbohydrate, using
over the estimated amount of carbohydrate present a hydrazide-mediated conjugation process can have
in the reaction mixture. advantages over the use of amine–biotin compounds.
3. Heat at 80°C in a sealed Reactivial (Thermo Fisher) In the case of BNAH, however, it was determined that
for 1 h. the resultant linkage with the reducing end of an oligosac-
4. Add to the reaction mixture an equal volume of charide or glycan was not a hydrazone bond, but a gly-
the reducing agent borane-dimethylamine (BDA) cosylhydrazide derivative, which preserves the pyranose
complex, which was previously prepared in the ring structure of the sugar (Figure 11.31). This finding is
reaction buffer at a concentration of 125 mg/ml. the main reason a BNAH-modified glycan effectively dis-
5. React for another hour at 80°C. plays a near-native conformation at the reducing end. In
6. Purify the BAP–glycan conjugate from unreacted addition, the biotin label in this configuration is reversible
glycans by use of a C18 sample prep cartridge, as and can be released by incubation under acidic condi-
described above for the synthesis of BAP. Separation tions, thus allowing recovery of the carbohydrate.
of excess BAP from the conjugates may be carried Another advantage of the BNAH derivative is that the
out by SEC or anion-exchange chromatography, conjugation reaction with reducing sugars can be carried
depending on the size of carbohydrates being out in aqueous conditions and in an environment that
modified and their intrinsic charge. Follow the permits carbohydrate and biotinylation reagent solubility.
separations by visualization of BAP fluorescence
with a hand-held UV lamp or through the use of a
OH
fluorescence detector.
R O O
O OH
Biotinyl-l-3-(2-Naphthyl)-Alanine Hydrazide HO + HN
NH O
(BNAH) O
H
N
NH
H2N N
BNAH is a biotin–hydrazide derivative containing a CH3 H S
O
UV-absorbing and fluorescent naphthalene group (bio- N-Acetyl glucosamine
residue at glycan BNAH
tinyl-l-3-(2-naphthyl)-alanine hydrazide) (Leteux et  al., reducing end
1998). Unlike BAP described previously, BNAH has a
hydrazide group for coupling to the reducing end of
carbohydrates, instead of an amine. While both groups
can be successfully conjugated to an aldehyde of a O
released glycan, the biotin–amine compounds require OH
HN
O
a reducing agent to stabilize the resultant hydrazone H NH
R O N
bond. BNAH may be coupled to reducing sugars with- O HN N
H
HO S
out reduction, since the linkage formed between a NH O

hydrazide and an aldehyde is much more stable than O


Fluorescent Glycan–BNAH Conjugate
CH 3
that with an amine. The modified glycans or carbohy-
drates can be detected using the fluorescent properties FIGURE 11.31  BNAH contains a hydrazide group that can be
of the naphthalene group or captured by immobilized used to label the reducing end of released glycans through the forma-
streptavidin for further analysis (Harvey, 2011). tion of a hydrazone bond.

BIOCONJUGATE TECHNIQUES
504 11.  (Strept)avidin–Biotin Systems

Modified carbohydrates may be stored for at least one Apply the sample to a 5-μm C18-silica HPLC column
year at −20°C in a solution of water/methanol (9 : 1, v/v) (250 × 4.6 mm, Nucleosil). Elute with a gradient of
without degradation. water to acetonitrile at a flow rate of 1 ml/min over
The following protocol is based on the method of a time course of 30 min. Free BNAH and BNAH–
Leteux et al. (1998). glycan derivatives can be monitored by absorbance
at 275 nm. The conjugate peak will also be positive
Protocol for carbohydrate by reaction with orcinol, which can
1. Dissolve 100 nmol of the carbohydrate to be modified be detected by spray after spotting a small eluted
and 500 nmol of BNAH in 25 μl of methanol (20-mM sample on a TLC plate.
BNAH solution).
2. Evaporate the solution to dryness and re-dissolve in Biotin–PEG3–Phosphine
25 μl an acidic solution of methanol/water/acetic acid Saxon and Bertozzi (2000) reported on the synthesis and
(74 : 8 : 8, v/v) or in a neutral solution consisting of use of a novel biotinylation compound containing a phos-
methanol/water (9 : 1, v/v), depending on the relative phine group for coupling to azide containing molecules.
solubility of the carbohydrate. The reagent has a biotin handle at one end, a triethylene-
3. React for at least 5 h at 60°C if using the acidic glycol (PEG) diamine spacer imparting increased water
reaction solution or for a total of 16 h at 60°C if using solubility in the middle, and a 3-(diphenylphosphino)-
the neutral solution. 4-(methoxycarbonyl)benzamide group on the other end.
4. Purification of the conjugates may be achieved by Biotin–PEG3–phosphine reacts with azide derivatives of
reverse-phase HPLC separation. Dry the reaction amino acids, sugars, and cross-linkers to form an interme-
solution under a nitrogen stream and reconstitute in diate aza-ylide, which spontaneously rearranges in aque-
a minimum volume of acetonitrile/water (1 : 1, v/v). ous solution to create a stable amide bond (Figure 11.32).

OH

OH O
HO
OH
–N H O
N+ N
N Glycan Protein
HO
O
Azido–sialic acid–glycan

O
+ P
HN O
NH
CH3
O
H H
N O N
S O O
O O
Biotin–PEG3–Phosphine

OH

OH O
O HO
P OH
HN O O
NH H O
N
N Glycan Protein
H H H HO
N O N O
S O O
O O
Biotinylated glycan through
amide bond formation

FIGURE 11.32  Azido–sialic acid-containing glycans can be labeled in vivo with biotin–PEG–phosphine using the Staudinger ligation
reaction, which forms an amide bond.

BIOCONJUGATE TECHNIQUES
6.  Biotinylation Techniques 505
This reaction is a modified Staudinger ligation that can be biotinylated azido–glycoconjugates after cell lysis and
used to target azide-containing glycans or proteins in vivo. provide a method for studying glycoprotein function
and interactions.
The following protocol is based on the methods of
O Saxon and Bertozzi (2000) and Prescher et al. (2004).
P
HN O
NH
O
CH3 Protocol
H H
S
N
O
O
O
N 1. Treat and grow cells in the presence of 20-μM
O O
Ac4ManNAz for at least 3 days. The azide–mannose
derivative may be solubilized in 70% DMSO as
Biotin–PEG3–Phosphine
Mol. Wt.: 764.87 a more concentrated stock solution and then an
aliquot added to the media containing the cells.

Alternatively, mice may be treated with the azide–
Cells grown in the presence of azide analogs of
sugar derivative in aqueous DMSO at a level of
certain amino acids or sugars will incorporate these
100 to 300 mg/kg, using an injection of 200 μl
derivatives into proteins or carbohydrates through
administered intraperitoneally daily for 7 days.
enzymatic synthesis using the native cell machinery.
2. When working with cells, first wash them several
Azides thus displayed on biomolecules are unreac-
times with PBS, pH 7.4, to remove any remaining
tive with other substances typically found within the
Ac4ManNAz from the media. Alternatively, if
cell, but the azide derivatives may be targeted for con-
working with animals, isolate the tissue type desired
jugation using the modified Staudinger reaction (see
and prepare the cells to be labeled in PBS, pH 7.4.
Chapter 17, Section 6, for additional information on this
3. To label cell surface azide–glycans, the cells are
reaction and its use in biological labeling).
reacted for 1 h using a final concentration of 1-mM
Biotin–PEG3–phosphine can be used to label glycans
biotin–PEG3–phosphine reagent dissolved in PBS,
or proteins that have been modified to contain azide
pH 7.4.
groups. The beautiful specificity of this reaction and its
4. Wash the cells several times with PBS, pH 7.4, to
lack of toxic side reactions or additives make it suitable
remove excess biotinylation compound.
for use in living organisms, including cells and animals.
5. The labeled glycans may be analyzed by cell
The reaction is completely orthogonal to functional
sorting after staining with fluorescently modified
groups and reactions found in living systems, so the
streptavidin. Alternatively, the cells may be lysed
biotinylation process proceeds with no cross-reactions
and the labeled glycans isolated using immobilized
with other biomolecules. In addition, no cell toxicity
streptavidin.
has been observed due to the phosphine or the phos-
phine oxide byproduct of the coupling reaction. The Additional biotinylation reagents are discussed in
phosphine also does not appear to be capable of reduc- Chapter  18, Section 1.3, which describes newer hydro-
ing disulfides within proteins. philic biotin compounds containing PEG spacers. These
Prescher et al. (2004) have shown that mice fed with reagents offer significant advantages over the more tra-
the peracetylated azido–mannose sugar derivative ditional aliphatic compounds, because the pronounced
Ac4ManNAz efficiently convert it through deacety- water solubility of the PEG cross-bridge can prevent
lation by cytosolic esterases into an azido–sialic acid aggregation of biotinylated molecules. With some lon-
(SiaNAz), which then gets incorporated enzymati- ger chain biotin compounds that contain hydrophobic
cally into cell surface glycans. Biotinylation of these hydrocarbon spacers, proteins can precipitate or lose
aberrant carbohydrates provides a method of detect- activity over time due to the insolubility of the biotin
ing or isolating glycoproteins or other glycoconjugates modifications. Biotinylated antibodies are particularly
(Vainauskas et  al., 2012). For instance, fluorescently susceptible to aggregation and loss of antigen binding
labeled streptavidin can be used to image the cell sur- ability if they are modified using hydrophobic biotin
face structures containing the biotinylated azido–sugar compounds. The PEG-based biotin reagents show better
derivatives. Alternatively, immobilized streptavidin or solubility and longer stability than their corresponding
immobilized monomeric avidin can be used to purify aliphatic biotinylation compounds of equivalent size.

BIOCONJUGATE TECHNIQUES

Potrebbero piacerti anche