Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

REVIEW

Glutaraldehyde: behavior in aqueous solution,


reaction with proteins, and application to
enzyme crosslinking
Isabelle Migneault, Catherine Dartiguenave, Michel J. Bertrand, and Karen C. Waldron

BioTechniques 37:790-802 (November 2004)

Glutaraldehyde possesses unique characteristics that render it one of the most effective protein crosslinking reagents. It can be pres-
ent in at least 13 different forms depending on solution conditions such as pH, concentration, temperature, etc. Substantial literature
is found concerning the use of glutaraldehyde for protein immobilization, yet there is no agreement about the main reactive species
that participates in the crosslinking process because monomeric and polymeric forms are in equilibrium. Glutaraldehyde may react
with proteins by several means such as aldol condensation or Michael-type addition, and we show here 8 different reactions for
various aqueous forms of this reagent. As a result of these discrepancies and the unique characteristics of each enzyme, crosslinking
procedures using glutaraldehyde are largely developed through empirical observation. The choice of the enzyme-glutaraldehyde
ratio, as well as their final concentration, is critical because insolubilization of the enzyme must result in minimal distortion of its
structure in order to retain catalytic activity. The purpose of this paper is to give an overview of glutaraldehyde as a crosslinking
reagent by describing its structure and chemical properties in aqueous solution in an attempt to explain its high reactivity toward
proteins, particularly as applied to the production of insoluble enzymes.

CHEMICAL BEHAVIOR OF
INTRODUCTION aldehydes in generating thermally and GLUTARALDEHYDE IN
chemically stable crosslinks (15). In AQUEOUS SOLUTION
Among the many available protein fact, studies of collagen crosslinking
crosslinking agents, glutaraldehyde reactions with monoaldehyde (formal- Knowledge of the structure and
has undoubtedly found the widest dehyde) and dialdehydes having chain mechanism of crosslinking reagents is
application in various fields such as lengths of two to six carbon atoms (gly- important for their use. However, the
histochemistry (1–3), microscopy oxal, malonaldehyde, succinaldehyde, structure of glutaraldehyde in aqueous
(1,4,5), cytochemistry (6), leather tan- glutaraldehyde, and adipaldehyde) solution has been the subject of more
ning industry (7,8), enzyme technology demonstrated that the reactivity in this debate than any of the other crosslink-
(9–13), chemical sterilization (14), and series is maximized at five carbons; ing reagents. In fact, glutaraldehyde
biomedical (15) and pharmaceutical thus glutaraldehyde is the most effec- structure in aqueous solution is not lim-
sciences (16). Glutaraldehyde, a linear, tive crosslinking agent (18). ited to the monomeric form (Figure 1,
5-carbon dialdehyde, is a clear, color- Glutaraldehyde has found wide- structure I). Figure 1 gives an overview
less to pale straw-colored, pungent oily spread use for enzyme immobilization. of the possible molecular forms of glu-
liquid that is soluble in all proportions in Despite the success of this reagent, its taraldehyde in aqueous solution based
water and alcohol, as well as in organic chemistry has been quite controversial on reports covering the past 40 years
solvents. It is mainly available as acidic (19). In fact, the simple structure of (10,20–29). The different glutaralde-
aqueous solutions (pH 3.0–4.0), rang- glutaraldehyde is not indicative of the hyde structures (Figure 1) have been
ing in concentration from less than 2% complexity of its behavior in aqueous numbered as found in the literature and
to 70% (w/v). Glutaraldehyde has had solution and its reactivity. Our purpose are discussed in chronological order.
great success because of its commercial here is to review the literature on glutar- In 1962, Aso and Aito (20,30) stud-
availability and low cost in addition aldehyde by first presenting its chemical ied the polymerization of glutaralde-
to its high reactivity. It reacts rapidly behavior in aqueous solution and then its hyde using cationic catalysts, and they
with amine groups at around neutral reactivity with proteins, focusing on its found that a similar polymerization
pH (17) and is more efficient than other application for enzyme immobilization. occurred spontaneously in aqueous

Université de Montréal, Montréal, Canada

790 BioTechniques Vol. 37, No. 5 (2004)


solutions, at room temperature in the amounts of α,β-unsaturated aldehydes
absence of a catalyst. The final product (structure VI) by UV-spectrophotomet-
(structure VIII) was proposed to be a ric analysis, which was in agreement
soluble tetramer or pentamer containing with the findings of Hardy et al. (21).
approximately one free aldehyde group They proposed, after H-NMR analysis,
per molecule formed through the intra- that aqueous solutions of glutaralde-
molecular-intermolecular propagation hyde consisted of free glutaraldehyde
polymerization with ring formation. (structure I), the cyclic hemiacetal of its
In 1968, Richards and Knowles (10) hydrate (structure IV), and oligomers of
studied glutaraldehyde solutions by this (structure V), all in equilibria and
proton (H) nuclear magnetic resonance in different proportions as a function of
(H-NMR). The NMR data (i.e., types of the temperature. In 1974, Whipple and
protons and peak integrations) did not Ruta (32) studied aqueous glutaralde-
agree with those expected from dimer, hyde using 13C-NMR and concluded
cyclic dimer, trimer, or bicyclic trimer that aqueous glutaraldehyde consisted
only, but rather was consistent with a primarily of the cyclic hemiacetal
mixture of polymeric forms of these (structure IV), but they added that this
oligomers as well as higher polymeric form was equally distributed between
species. The authors concluded that two geometrical isomeric forms (cis
commercial solutions were largely and trans).
polymeric and contained significant Monsan et al. (23), in 1975, con-
amounts of α,β-unsaturated aldehydes firmed, for acidic glutaraldehyde solu-
(structure VI) that were able to form tions, the results obtained by Hardy et
rings (structure VII) by loss of water al. (21), Korn et al. (22), and Whipple
molecules by aldol condensation. The and Ruta (32) using gel and thin-layer
structure VI represents the average chromatography, mass spectrometry
structure of the unsaturated polymer- (MS), H-NMR, and infrared spectros-
ized glutaraldehyde (α,β-unsaturated copy (IR). For glutaraldehyde solutions
compound), and Hooper (31) reported at neutral or slightly alkaline pH, they
that the pendent aldehyde groups of observed the production of molecular
structure VI would be scarcely hydrated forms, which could precipitate. They
because the carbonyl form is stabilized also noted the formation of an abundant
by conjugation. precipitate, named poly-glutaraldehyde,
Hardy et al. (21), in 1969, used when sodium hydroxide was added to
ultraviolet (UV) spectrophotom- 25% aqueous glutaraldehyde solution
etry in combination with H-NMR to to reach pH 11.0. MS and IR analysis
investigate glutaraldehyde solutions. allowed the identification of the solid as
They also found α,β-unsaturated being the result of aldolic condensation
aldehydes (structure VI), but only as a of glutaraldehyde molecules (structures
very minor component of the organic VI and/or VII).
content because of the relatively weak In 1980, Margel and Rembaum (25)
absorbance observed at 235 nm. Fur- investigated the aldol condensation of
thermore, after purification of glutaral- glutaraldehyde in the pH range of 7.0
dehyde by liquid extraction with ethyl to 13.5. They obtained poly-glutaralde-
ether, these authors obtained 50% pure hyde and proposed the structure IX after
glutaraldehyde (structure I) with the spectroscopic and electrochemical anal-
expected H-NMR spectrum. Addi- ysis. They also reported that poly-glu-
tional H-NMR investigations showed taraldehyde may be soluble or insoluble
that purified glutaraldehyde undergoes in water and may have different con-
very rapid hydration upon dissolution centrations of aldehyde, hydroxyl, and
in water, which agreed with the results carboxylic functional groups depending
of Aso and Aito (20,30). Thus, Hardy et on the polymerization conditions, such
al. (21) postulated that glutaraldehyde as pH and oxygen content, leading to
monomer (structure I) existed as a mix- the structures XII and XIII.
ture of hydrated forms (structures II, III, In 1991, Tashima et al. (27) obtained
and IV) in aqueous solution, all of them a new dimer when glutaraldehyde was
being in equilibria. treated by aqueous alkaline solution (pH
In their work a few years later, 8.5). Their analyses by UV and IR sug-
Korn et al. (22) did not find significant gested the existence of α,β-unsaturated
Vol. 37, No. 5 (2004) BioTechniques 791
REVIEW

formyl and hydroxyl groups in this mol- for gel chromatography, chloroform/ intensities is known to be not quanti-
ecule, and gas chromatography (GC) acetone for thin-layer chromatography, tative in 13C-NMR (34). Thus, these
MS analysis indicated the molecular and deuterated chloroform or carbon observations led Kawahara et al. (28)
formula C10H14O3 (molecular weight: tetrachloride for H-NMR). In fact, the to investigate the molecular structure of
182 g/mol). Moreover, after derivatiza- equilibrium between monomeric and glutaraldehyde in aqueous solution by
tion followed by two-dimensional NMR polymeric glutaraldehyde in anhydrous UV absorption and light scattering. The
analysis, they proposed the structure X, solvents could possibly shift to the 70% (w/v) glutaraldehyde solution used
in equilibrium with structure XI, for the latter to produce water. Other studies for their study was found to contain a
glutaraldehyde dimer. showed similar problems (10,21,32). large quantity of polymeric species with
In 1992, Kawahara et al. (28) reported For example, Richards and Knowles cyclic hemiacetal structure (V). Upon
that most of the studies summarized (10) and Hardy et al. (21) conducted dilution, the polymerized glutaralde-
above (10,21,23,32) neglected possible their experiments in deuterated water, hyde slowly converted to monomers,
solvent effects on the glutaraldehyde but the exchange of deuterium for thus inducing a great variation in the
structure. In fact, water is the medium hydrogen bound to the α-carbon could relative abundances of monomeric and
in which commercial glutaraldehyde is give erroneous results when comparing polymeric species, according to the glu-
supplied and in which the crosslinking the peak intensities by H-NMR (22). taraldehyde concentration. In fact, they
reaction with proteins is carried out, Moreover, the hydration equilibrium found that in dilute solution and in the
and glutaraldehyde was found to react constants for monoaldehyde (formal- pH range of 3.0 to 8.0, glutaraldehyde
with this solvent in various ways. Thus, dehyde) are reported to differ in water was almost monomeric, predominantly
according to Kawahara et al. (28), there and deuterated water (33), and this in cyclic hemiacetal form (structure
was a considerable problem in the stud- probably occurs with glutaraldehyde. IV). In 1997, the same authors (29)
ies carried out by Monsan et al. (23) Whipple and Ruta (32) used 13C-NMR found that glutaraldehyde structure was
because their analyses were conducted to analyze glutaraldehyde solutions, similar for aqueous solutions up to 10%
only in organic solvent (tetrahydrofuran but direct comparison of the peak (w/v) and concluded that the content of

Figure 1. Summary of the possible forms of glutaraldehyde in aqueous solution.

792 BioTechniques Vol. 37, No. 5 (2004)


α,β-unsaturated structures (structure time at the beginning of the 1960s for
VI) was negligible regardless of the the fixation of tissues (1), and since this
glutaraldehyde concentration. time many other applications have been
In summary, several studies (10,20– developed. The high reactivity of glu-
29) have shown that commercially taraldehyde toward proteins at around
available glutaraldehyde represents neutral pH is based on the presence of
multicomponent mixtures, but knowing several reactive residues in proteins and
which of these components is the most molecular forms of glutaraldehyde in
efficient for reactions with proteins is aqueous solution, leading to many dif-
debatable. In fact, in aqueous solution, ferent possible reaction mechanisms.
glutaraldehyde can exist in its simplest Enzyme immobilization represents a
form, a monomeric dialdehyde, but also good example to illustrate the use of
as a dimer, trimer, and polymer. There- glutaraldehyde as protein crosslinking
fore, the effectiveness of glutaraldehyde reagent.
immobilization and the controversies Glutaraldehyde can react with sev-
surrounding its chemical behavior could eral functional groups of proteins, such
be rationalized with the multiplicity of as amine, thiol, phenol, and imidazole
structures, which depends on the solu- (39) because the most reactive amino
tion conditions. acid side-chains are nucleophiles. Vari-
ous data on aldehyde reactivity (at pH
from 2.0 to 11.0) with the following
APPLICATION TO SUBSTANCES amino acids have been reported in the
OF BIOLOGICAL INTEREST literature: lysine (18); tyrosine, tryp-
tophan, and phenylalanine (40); histi-
Carbohydrates, Lipids, and Nucleic dine, cysteine, proline, serine, glycine,
Acids glycylglycine, and arginine (41). These
authors investigated the ability of dif-
There is little information on the use ferent aldehydes to react with amino
of aldehydes to fix carbohydrates (35) or acids, and they ranked the reactive moi-
lipids (6). Most lipids do not react well eties of the amino acids in decreasing
with glutaraldehyde, with the exception order of reactivity as follows: ε-amino,
of some phospholipids that contain pri- α-amino, guanidinyl, secondary amino,
mary amines (e.g., phosphatidylserine and hydroxyl groups. Avrameas and
and phosphatidylethanolamine; Refer- Ternynck (42) concluded that either
ence 36). In the case of nucleic acids, glutaraldehyde did not react with the
formaldehyde is by far the most effec- amine function of the guanidinyl group
tive agent for their fixation by reaction (arginine) or that in protein molecules
with amino groups of DNA nucleotides the more reactive groups prevented
(37), but little information is available the observation of arginine reactivity
on the interaction between glutaralde- with glutaraldehyde. Okuda et al. (17)
hyde and DNA (38). noted that glutaraldehyde reacted with
thiol groups only in the presence of a
Proteins: General Case primary amino group. Glutaraldehyde
reacts reversibly with amino groups
Glutaraldehyde was used for the first over a wide pH range (≥pH 3.0), except

Figure 2. Schiff base (1) and Michael-type (2) reactions of glutaraldehyde with proteins.

Vol. 37, No. 5 (2004) BioTechniques 793


REVIEW

between pH 7.0 to 9.0 where only a taraldehyde (Figure 2). Richards and the facility of polymeric forms to revert
little reversibility is observed (17). Knowles (10) proposed that the reaction to the active monomer depended upon
The crosslinking of proteins, either involved the conjugate addition of pro- pH (i.e., the type of glutaraldehyde poly-
to a carrier (solid support) or between tein amino groups to ethylenic double mers in solution). He also considered
protein molecules (carrier-free), gener- bonds (Michael-type addition) of the that polymers existing in the alkaline
ally implies the ε-amino group of lysyl α,β-unsaturated oligomers found in the pH range cannot revert to the monomer
residues (43). The unprotonated amino commercial aqueous solutions of glutar- because time and temperature tend to
groups are very reactive as nucleophilic aldehyde that are usually used (Figure favor a more irreversible polymer, in
agents (44). It should be noted that lysyl 2, reaction 2). A few years later, Peters contrast to polymers that exist at acidic
ε-amino groups have pKa (acid dissoci- and Richards (47) showed work that and neutral pH. This was supported in
ation constant) > 9.5, but it is presumed supported this hypothesis because they 1990 when Ruijgrok et al. (50) showed
that the small percentage of amines found that, in the presence of an excess that glutaraldehyde polymers in the
present in their unprotonated form at of amino groups, nucleophilic addition neutral and acidic pH range could revert
lower pH is sufficient to react with glu- on the ethylenic double bond was pos- easily to the monomer under the influ-
taraldehyde, which then drives the acid- sible. Monsan et al. (23) proposed a ence of heating or ultrasonic radiation.
base equilibrium to deprotonation of slightly different mechanism in which As early as 1976, Hardy et al.
these groups for further reaction. Most an addition reaction occurred on the (51,52) and Lubig et al. (53) argued
proteins contain many lysine residues, aldehydic part of the α,β-unsaturated that the reaction of glutaraldehyde with
usually located on the protein surface polymers (and poly-glutaraldehyde) to proteins was not due to α,β-unsaturated
(i.e., exposed to the aqueous medium) give a Schiff base (imine) stabilized by aldehydes but may involve some dimer-
because of the polarity of the amine conjugation (Figure
group. Furthermore, lysine residues are 2, reaction 1).
generally not involved in the catalytic In the early
site, which allows moderate crosslink- 1970s, Boucher
ing to preserve protein conformation (48,49) proposed
(45) and thus biological activity (46). As that monomeric
stated previously, glutaraldehyde exists glutaraldehyde was
in multiple forms in aqueous solution, the active species
and all of these forms might be reactive involved in the
toward lysine residues (ε-amino group) crosslinking with Figure 3. Crosslinking of proteins with glutaraldehyde giving a quater-
of proteins. proteins and that nary pyridinium compound.
In spite of the substantial amount
of literature concerning the use of glu-
taraldehyde, there is still no agreement
about the main reactive species in glu-
taraldehyde solutions during the cross-
linking process. Aldehydes are expected
to form Schiff bases upon nucleophilic
attack by the ε-amino groups of lysine
residues in the protein (23). However,
Schiff bases are unstable under acidic
conditions and tend to break down to
regenerate the aldehyde and amine. In
contrast, the linkage formed by the reac-
tion of glutaraldehyde with an amino
group has shown exceptional stability
at extreme pHs and temperatures, thus
a simple Schiff base with both ends of
monomeric glutaraldehyde has been
ruled out as a mechanism for glutaral-
dehyde crosslinking with proteins. Sev-
eral alternative mechanisms have been
proposed.
Between 1968 and 1975, Richards
and Knowles (10) and Monsan et al.
(23) postulated pathways, both involv-
ing the reaction of the protein amino
group with α,β-unsaturated aldehydes
formed by aldol condensation of glu- Figure 4. Reaction of dimeric cyclic glutaraldehyde with proteins under basic conditions.

794 BioTechniques Vol. 37, No. 5 (2004)


ization in the presence of the amino more stable because of the conjugation
group, such as the formation of qua- of the internal aldehyde group with the
ternary pyridinium compounds (Figure C-C double bonds and a Michael addi-
3), rather than glutaraldehyde poly- tion product (VIb). In the presence of
mers reacting with amino groups. The excess amine, a mixed product (VIc) is
mechanism involved could result from seen, which is labile to acid hydrolysis
cyclization, dehydration, and internal because of the disruption of resonance
redox reactions of a Schiff base. Hardy stabilization. Because elimination of
et al. (52) reported the isolation of a water in the formation of Schiff bases is
pyridinium-type compound following reversible and because prolonged expo-
the reaction of glutaraldehyde with sure to buffer solutions, particularly at
amines and suggested this structure as a elevated pH, might impair binding and
stable crosslink. They showed that this lead to gradual release of the enzyme,
compound had an UV absorption maxi- reducing the double bonds of the Schiff
mum at 265 nm, which was consistent bases by application of suitable reduc-
with the original observation of Bowes ing agents (19) such as sodium borohy-
and Cater (18). dride (NaBH4) or sodium cyanoborohy-
In 1991, Tashima et al. (27) con- dride (NaCNBH3) has been proposed.
cluded that reaction of alkaline glutar- In both cases, the reduction of (VIa)
aldehyde solutions (mixture of dimers produces a secondary amine that is
X and XI) with proteins may involve a tolerant to variations in pH and is stable
Michael addition to the double bonds to even in acidic conditions. Sodium cya-
give (Xa) and (XIa), as shown in Figure noborohydride is preferred because it
4 (adapted from Reference 19). If these is a milder reagent (54). In fact, sodium
reactions occur, no reduction is neces- borohydride not only reduces Schiff
sary to stabilize the adducts. If an excess bases, but also aldehyde groups, leading
of amine is added, the compound (XIb) to a lower yield of conjugate formation
may be formed. (55). Even if the use of a reducing agent
In 1994, Walt and Agayn (19) pro- has been recommended, reduction is not
posed multiple reaction products for usually required (19).
the different glutaraldehyde structures In 1997, Kawahara et al. (29) specu-
in solution depending on the pH condi- lated on whether proteins could catalyze
tions because each form of glutaralde- the aldol condensation/polymerization
hyde might participate differently in of glutaraldehyde. They suggested that
crosslinking reactions with proteins. monomeric glutaraldehyde could be
Thus, under acidic or neutral conditions, converted to polymeric forms by the
glutaraldehyde exists as a mixture of action of amino groups and that this
monomers [i.e., free aldehyde form (I) product played a major role in the cross-
or cyclic hemiacetal (IV)] or as a poly- linking reaction of proteins. In fact, they
mer [i.e., cyclic hemiacetal oligomer proposed that the polymerization of
(V)]. Each of these structures would glutaraldehyde via aldol condensation
be expected to form Schiff bases upon proceeded in parallel with the cross-
nucleophilic attack by lysine residues in linking reaction and that the formation
a protein, as shown in Figure 5. How- of a Schiff base (imine) by one glu-
ever, as previously mentioned, Schiff taraldehyde molecule with one amino
bases are unstable under acidic condi- group enhances its aldol condensation
tions and thus Schiff base formation with other glutaraldehyde molecules.
(Figure 5, Equation 1) between a lysine The final crosslinked structure would
amino group and free aldehyde (struc- be a linear aldol-condensed oligomer
ture I) is not favored. It is more likely of glutaraldehyde, with several Schiff
that monomeric cyclic hemiacetal (IV) base linkages branching off. They also
and its multimeric form (V) react via observed that the dehydration step
reactions in Equations 2 and 3 of Figure following aldol condensation occurs
5, under acidic conditions. Under basic almost completely at the glutaraldehyde
conditions (Figure 6), the reaction of monomer units containing Schiff base
α,β-unsaturated oligomeric aldehydes imine, in contrast to the glutaraldehyde
(VI) with amine can give two products units containing no Schiff base, where
robust to acid hydrolysis: a Schiff base little dehydration occurs. Therefore,
(Figure 6, structure VIa), which was the formed Schiff base linkage eventu-
Vol. 37, No. 5 (2004) BioTechniques 795
REVIEW

attachment to water-insoluble carriers


via glutaraldehyde is one of the sim-
plest and most gentle coupling meth-
ods in enzyme technology (43). The
first reported application of the use of
a bifunctional reagent was by Zahn in
the 1950s (56), which was followed by
studies on the chemistry of crosslinking
with glutaraldehyde for the prepara-
tion of stable protein crystals for X-ray
diffraction studies (45) or for the fixa-
tion of tissue samples for microscopic
investigation (57). Later, glutaralde-
hyde was widely used as a mild cross-
linking agent for the immobilization of
enzymes because the reaction proceeds
in aqueous buffer solution under condi-
tions close to physiological pH, ionic
strength, and temperature. Essentially,
two methods have been used: (i) the for-
mation of a three-dimensional network
as a result of intermolecular crosslink-
ing and (ii) the binding to an insoluble
carrier (e.g., nylon, fused silica, con-
trolled pore glass, crosslinked proteins
such as gelatin and bovine serum albu-
Figure 5. Reactions of glutaraldehyde with proteins under acidic or neutral conditions.
min (BSA), and polymers with pendant
amino groups).
ally constitutes a conjugate system with glutaraldehyde is present in different Immobilization can be achieved for
the adjacent ethylenic double bond. forms even for specific and controlled many enzymes under a wide range of
Once such conjugation is formed, the reaction conditions, several of the pos- conditions, which should be chosen
resonance interaction is reported to sible reaction mechanisms presented according to the specific results
make Schiff base linkages stable to acid above could proceed simultaneously. required. These conditions have often
hydrolysis (23). been determined by trial and error
In conclusion, the chemical nature Enzymes because insolubilization is critically
of the reaction of glutaraldehyde with dependent on a delicate balance of fac-
proteins is not clearly understood, and Immobilized enzymes are currently tors such as the nature of the enzyme
the mechanisms of protein crosslinking the subject of considerable interest (42,58,59), the concentration of both
reactions remain open to speculation. because of their advantages over sol- enzyme (60) and reagent (58), the pH
However, it seems that no single mech- uble enzymes or alternative technolo- (61) and ionic strength (62) of the solu-
anism is responsible for glutaraldehyde gies, and their applications are steadily tion, the temperature (63), and the reac-
reaction with proteins. In fact, because increasing. Immobilization by covalent tion time (64).
The nature of the
enzyme, particularly its
lysine content, has an
effect on its insolubi-
lization by glutaralde-
hyde (42,58,59). More-
over, if only a small
amount of enzyme is
available or if extensive
modification must be
avoided, the addition
of inert, lysine-rich
protein (e.g., BSA)
has been suggested by
Broun (58).
As mentioned above,
Figure 6. Reaction of polymeric glutaraldehyde with proteins under basic conditions. the concentrations of
796 BioTechniques Vol. 37, No. 5 (2004)
REVIEW

enzyme and glutaraldehyde must be care- the formation of insoluble active chy-
fully considered to obtain water-insoluble motrypsin (EC 3.4.21.1) was most rapid
enzyme derivatives via crosslinking (60); at pH 6.2 (pI 8.6) and chymotrypsinogen
low concentrations of enzyme and glutar- A at pH 8.2 (pI 9.5). The existence of an
aldehyde tend to induce intramolecular optimum pH suggests the important role
crosslinking by enhancing the probability of the protein charge on the intermolec-
that glutaraldehyde functional groups ular crosslinking required for insolubi-
will react with the same enzyme mol- lization. The charge on the protein may
ecule (60). Thus, conditions should be regulate crosslinking, which was maxi-
chosen carefully to favor intermolecular mal when the repulsive charges were
crosslinking between enzyme molecules minimal. Furthermore, Tomimatsu et al.
instead of unwanted intramolecular links, (62) and Broun (58) concluded that the
which could also be formed (58,65,66). lower the ionic strength of the reaction
Broun (58) reported that the amount of medium, the more rapid the crosslink-
crosslinking agent used affects the degree ing of chymotrypsin. On the other hand,
or extent of crosslinking. He indicated the choice of pH should also be taken
that low concentrations of glutaralde- into account regarding the reactivity of
hyde were not able to form sufficient aqueous glutaraldehyde, most immobi-
crosslinkages to effect precipitation of lizations being conducted in the neutral
the enzyme. At higher concentrations, the or slightly alkaline pH range.
extent of crosslinking was high enough to The influence of temperature and
form a tight structure by excluding water reaction time on insolubilization of
molecules to insolubilize the enzyme enzymes has been reported by Broun
derivative. Chui and Wan (67) indicated (58). In early reports on enzyme immo-
that enzymatic activity was inversely bilization, the reactions were carried out
proportional to the concentration of at low temperature (4°C), which was
glutaraldehyde used because extensive preferred for labile molecules, but the
crosslinking may result in a distortion of immobilization process required long
the enzyme structure (i.e., the active site reaction times (6–18 h; Reference 59).
conformation). With this distortion, the Ottesen et al. (63) and Bullock (35) sug-
accessibility and accommodation of the gested that the reaction of glutaralde-
substrate may be reduced, thus affecting hyde with lysine residues was progres-
the retention of biological activity. Fur- sive with time, probably depending on
thermore, the relative concentration of the accessibility of the ε-amino groups.
enzyme to glutaraldehyde should also be Currently, ambient temperature is used
considered (17). We found that crosslink- for glutaraldehyde immobilization of
ing of the enzyme trypsin (EC 3.4.21.4) enzymes within 4 h or less (68,69).
with glutaraldehyde could be achieved The catalytic activity of water-
over a wide range of relative mole ratios insoluble enzyme derivatives prepared
in 50 mM sodium phosphate buffer at using multifunctional reagents such as
pH 6.8 but that the time required to com- glutaraldehyde can vary considerably
mence precipitation ranged from 0.5 (61,63,70) and has been shown to be
to 120 min for enzyme:glutaraldehyde dependent on the amount of crosslinking
ratios of 1:500 to 1:25, respectively (I. reagent used during insolubilization, as
Migneault, unpublished data). well as on other factors (71). Moreover,
The reaction of glutaraldehyde with the kinetic behavior of immobilized
enzymes to give soluble and insoluble enzymes is, in many respects, different
products has been extensively stud- from that of free enzyme in solution
ied, and the reaction was shown to be (72), these differences being related to
pH-dependent (39). Jansen et al. (61) the diverse microenvironments gener-
showed that the optimum pH for glutar- ated by the enzymatic hydrolysis of the
aldehyde insolubilization varied from substrate. Kinetic properties of soluble
protein to protein. In fact, they observed enzymes are expressed in terms of
that the pH values for the most rapid Michaelis-Menten parameters. In the
insolubilization of BSA, soybean tryp- case of immobilized enzymes, apparent
sin inhibitor, lysozyme (EC 3.2.1.17), kinetic properties are used because the
and papain (EC 3.4.22.2) were found overall kinetic behavior of the enzy-
to be nearly the same as the isoelectric matic preparation is the sum of isolated
points (pIs) of these proteins, whereas contributions of each individual enzyme
798 BioTechniques Vol. 37, No. 5 (2004)
molecule, which can be immobilized with glutaraldehyde lost its transami- fragile crystals become much more
via different amino groups, leading nase activity but was still able to form sturdy and robust, so that there is much
to different exposures of the catalytic complexes with its antibody (75). less chance of damage during handling,
centers (73). Our work (74) on trypsin Stabilities (thermal, chemical, and while remaining permeable to dissolved
immobilization with glutaraldehyde mechanical) of water-insoluble enzyme solutes. Among enzymes, proteases
either by covalent attachment to ami- derivatives have also been described such as trypsin are of great interest
nopropyl controlled pore glass (CPG) (46,76). Most notably, thermal stabil- because of their numerous applica-
particles or by crosslinking of trypsin ity of immobilized enzymes has been tions in many fields. However, most
in solution showed an increase in the shown to vary from greater down to of the commonly used proteases are
apparent Michaelis constant, KM,app a lesser extent relative to the native marginally stable in their soluble form,
(i.e., a decrease in enzyme-substrate enzyme. The stability of an enzyme the prominent cause of their irrevers-
affinity) relative to free trypsin, which (protein) can typically be increased by ible inactivation being autoproteolytic
was more pronounced for glutaralde- crosslinking because intra- and intermo- digestion. Therefore, stabilization by
hyde-crosslinked trypsin compared to lecular crosslinks lead to a more rigid immobilization has been the subject of
CPG-trypsin. Thus, according to our molecule that can resist conformational considerable research. For example, we
results, the crosslinking procedure led changes (77). In fact, the covalent bonds digested denaturated lysozyme using
to a more constrained enzyme. Fur- created during the crosslinking reaction two immobilized trypsin preparations
thermore, the shapes of the pH-activity are stable, even in the presence of sub- (enzyme either covalently attached to
curves depend on the nature of the prod- strate or high ionic strength solutions aminopropyl CPG particles or cross-
ucts liberated as well as on the kinetic (59). Moreover, pH and temperature linked with glutaraldehyde) and did not
parameters. Changes in the specificity can be varied over a wide range without observe autoproteolysis (I. Migneault,
of certain immobilized enzymes have dissolution or deterioration of the cross- C. Dartiguenave, J. Vinh, M.J. Ber-
been reported. For example, glutamic linked crystals (78). The crosslinking trand, and K.C. Waldron, submitted
transaminase (EC 2.6.1.1) crosslinked confers mechanical advantages because data). Moreover, these immobilized
REVIEW

trypsin preparations showed excel- (84), phenalanyl-lysine coated polysty- CONCLUSIONS


lent digestion reproducibility based on rene beads (85), and polyethyleneimine
liquid chromatographic and capillary treated magnetite (86). Immobilized The success of glutaraldehyde as
electrophoretic peptide maps. Insoluble enzymes are also used in biosensors a crosslinking agent is a result of its
trypsin preparations were found to be (87), chromatographic packings and multicomponent nature, where several
considerably more stable than native detectors (88), online solid-phase reac- forms are present in equilibrium in the
trypsin in the alkaline pH range (79). tors (89), and in the field of medical reagent solution at a given pH. How-
Glassmeyer and Ogle (80) reported diagnostics and therapy (90). ever, the exact molecular composition
that an insoluble trypsin preparation There is an ongoing interest in car- of glutaraldehyde solutions, as well as
could be used repeatedly without loss of rier-free immobilized enzymes, such as which component is the most reactive,
activity and could be left standing in pH crosslinked enzyme crystals (CLECs; is debatable despite plenty of knowl-
8.0 buffer at room temperature for 40 h References 45 and 91), crosslinked edge. As a result, the reaction mecha-
with only a 9% loss of activity. How- dissolved enzymes (CLEs; References nism of glutaraldehyde with protein
ever, the activity of insoluble trypsin 64, 70, and 74) and crosslinked enzyme amino groups is not clearly understood,
preparations was completely destroyed aggregates (CLEAs; Reference 92). as illustrated by the large number of
upon incubation at 100°C in pH 8.0 CLECs results from the chemical sta- mechanisms reported in the literature
buffer for 15 min (80). Walsh et al. (81) bilization of the crystalline lattice of and summarized in this review. No
described the enhanced chemical stabil- enzyme molecules by glutaraldehyde, single mechanism seems to be respon-
ity of crosslinked, monolayered trypsin forming highly concentrated immo- sible for glutaraldehyde crosslinking
in the presence of urea, and Habeeb (70) bilized enzyme particles that can be with proteins. All reported forms of
reported a higher stability of trypsin lyophilized and stored for a long time glutaraldehyde exhibit the ability to
crosslinked with glutaraldehyde during at room temperature. CLEs are obtained react and crosslink proteins, leading
continuous use for casein digestions. by the crosslinking of dissolved to the formation of a broad range of
The storage stability of several enzymes, which leads to enzymes with conjugates. A fairly rigid control of
water-insoluble enzyme derivatives has enhanced thermostability. CLEAs were reaction conditions is needed to achieve
been examined because an insoluble synthesized in a simple and effective efficient insolubilization of each dif-
enzyme derivative should retain activity way by physical aggregation of the ferent enzyme due to their structural
for considerable periods of time to be enzyme penicillin G acylase (penicillin variability. Nonetheless, the resulting
useful. For example, Jansen and Olson amidohydrolase, E.C. 3.5.1.11) using enzyme derivatives generally show
(64) reported that papain (EC 3.4.22.2) a precipitant (e.g., tert-butyl alcohol), good stability and thus can be reused.
crosslinked with glutaraldehyde showed followed by chemical crosslinking with Although partial enzyme inactivation
no detectable decrease in esterase activ- glutaraldehyde. due to chemical modification is often
ity after 5 months at 4°C. Glassmeyer A different utilization of glutar- unavoidable, in most cases enough
and Ogle (80) stored insoluble prepa- aldehyde’s crosslinking ability, first catalytic activity is retained. More work
rations of trypsin in water at 4°C for reported in the late 1970s, is related to is needed to provide additional infor-
several months and, in most cases, at the preparation of microspheres for a mation on the exact structure of these
most a 15% drop in activity was found. variety of immunological and therapeu- crosslinked products.
Silman and Katchalski (79) did not note tic applications (24). Glutaraldehyde
a marked loss of activity after lyophili- was used for the preparation of micro-
zation and storage at room temperature. spheres from proteins (e.g., gelatin) or COMPETING INTERESTS
On the contrary, catalase (EC 1.11.1.6) alginate because of its excellent fixative STATEMENT
crosslinked with glutaraldehyde showed properties. Gelatin microspheres have
a decrease of about 20% in its initial been widely evaluated as drug carriers The authors declare no competing
activity after 2 weeks at 4°C in aqueous (93). Unfortunately, gelatin dissolves interests.
solution, after which no further decrease rather rapidly in aqueous environments,
in activity occurred after 5 months of making the use of this polymer difficult
storage (11). Thus, storage of immobi- for the production of controlled and/or REFERENCES
lized enzyme preparations depends on long-term delivery systems (94). Thus,
1.Sabatini, D.D., K. Bensch, and R.J. Bar-
the nature of the product and should be the use of a crosslinking procedure that rnett. 1963. Cytochemistry and electron mi-
tested for each case. leads to the formation of nonsoluble croscopy. The preservation of cellular ultra-
The application of chemical cross- networks within the microsphere wall structure and enzymatic activity by aldehyde
linking is multidisciplinary, ranging is required to reduce dissolution and fixation. J. Cell. Biol. 17:19-58.
2.Anderson, P.J. 1967. Purification and quan-
from basic protein biochemistry to premature drug release (95). Sodium titation of glutaraldehyde and its effect on
applied biotechnology, engineering, alginate microspheres have been several enzyme activities in skeletal muscle. J.
and medicine. Since the 1960s, glu- crosslinked with glutaraldehyde in the Histochem. Cytochem. 15:652-661.
taraldehyde has been used to couple presence of calcium chloride (67), and 3.Hopwood, D. 1967. The behavior of vari-
ous glutaraldehydes on Sephadex G-10 and
enzyme (and protein) to carriers such as applications of alginate microspheres some implications for fixation. Histochemie
cellulosic materials (82), aminoalkyl- have been reported in medicine (96) as 11:289-295.
silylated glass (83), polyacrylhydrazide well as in agriculture (97). 4.Hayat, M.A. 1981. Fixation for Electron Mi-

800 BioTechniques Vol. 37, No. 5 (2004)


croscopy. Academic Press, New York. 24.Rembaum, A., S. Margel, and J. Levy. 1971. Reaction of dialdehyde with functional
5.Kiernan, J.A. 2000. Formaldehyde, formalin, 1978. Polyglutaraldehyde: a new reagent for groups in collagen. Rev. Tech. Ind. Cuir. 63:5-
paraformaldehyde and glutaraldehyde: what coupling proteins to microspheres and for 14.
they are and what they do. Microsc. Today labeling cell-surface receptors. J. Immunol. 42.Avrameas, S. and T. Ternynck. 1969. The
00-1:8-12. Methods 24:239-250. cross-linking of proteins with glutaraldehyde
6.Hopwood, D. 1972. Theoretical and practi- 25.Margel, S. and A. Rembaum. 1980. Synthesis and its use for the preparation of immunoad-
cal aspects of glutaraldehyde. Histochem. J. and characterization of poly(glutaraldehyde). sorbents. Immunochem. 6:53-66.
4:267-303. A potential reagent for protein immobilization 43.Weetall, H.H. 1974. Immobilized enzymes:
7.Fein, M.L., E.H. Harris Jr, J. Naghski, and and cell separation. Macromolecules 13:19- analytical applications. Anal. Chem. 46:602A-
E.M. Filachione. 1959. Tanning with glutar- 24. 604A.
aldehyde. I. Rate studies. J. Amer. Leather 26.Tashima, T., U. Kawakami, M. Harada, 44.Guisán, J.M. 1988. Aldehyde-agarose gels as
Chem. Ass. 54:488-502. T. Sakata, N. Satoh, T. Nakagawa, and H. activated supports for immobilization—stabi-
8.Filachione, E.M., M.L. Fein, and E.H. Har- Tanaka. 1987. Isolation and identification of lization of enzymes. Enzyme Microb. Tech-
ris Jr. 1964. Tanning with glutaraldehyde. J. new oligomers in aqueous solution of glutar- nol. 10:375-382.
Amer. Leather. Chem. Ass. 59:281-292. aldehyde. Chem. Pharm. Bull. 35:4169-4180. 45.Quiocho, F.A. and F.M. Richards. 1964. In-
9.Quiocho, F.A. and F.M. Richards. 1966. The 27.Tashima, T., I. Masahiro, T. Kuroda, S. termolecular cross-linking of a protein in the
enzymic behavior of carboxypeptidase-A in Yagi, and N. Terumichi. 1991. Structure of crystalline state: carboxypeptidase A. Proc.
the solid state. Biochemistry. 5:4062-4076. a new oligomer of glutaraldehyde produced Natl. Acad. Sci. USA 52:833-839.
10.Richards, F.M. and J.R. Knowles. 1968. by aldol condensation reaction. J. Org. Chem. 46.Avrameas, S. 1969. Coupling of enzymes to
Glutaraldehyde as a protein cross-linkage re- 56:694-697. proteins with glutaraldehyde. Use of the con-
agent. J. Mol. Biol. 37:231-233. 28.Kawahara, J., T. Ohmori, T. Ohkubo, S. jugates for the detection of antigens and anti-
11.Schejter, A. and A. Bar-Eli. 1970. Prepara- Hattori, and M. Kawamura. 1992. The bodies. Immunochemistry 6:43-52.
tion and properties of crosslinked water-in- structure of glutaraldehyde in aqueous solu- 47.Peters, K. and F.M. Richards. 1977. Chemi-
soluble catalase. Arch. Biochem. Biophys. tion determined by ultraviolet absorption and cal cross-linking: reagents and problems in
136:325-330. light scattering. Anal. Biochem. 201:94-98. studies of membrane structure. Annu. Rev.
12.Fishman, S.N., B.I. Chodorov, and M.V. 29.Kawahara, J., K. Ishikawa, T. Uchimaru, Biochem. 46:523-551.
Volkenstein. 1971. Molecular mechanisms of and H. Takaya. 1997. Chemical cross-link- 48.Boucher, R.M.G. 1972. Advances in ster-
membrane ionic permeability changes. Bio- ing by glutaraldehyde between amino groups: ilization techniques. State of the art and re-
chim. Biophys. Acta 225:1-10. its mechanism and effects, p. 119-131. In G. cent breakthroughs. Amer. J. Hosp. Pharm.
13.Monsan, P. 1978. Influence of the conditions Swift, C.E. Carraher Jr., and C.N. Bowman 29:661-672.
of trypsin immobilization onto Spherosil on (Eds.), Polymer Modification. Plenum Press, 49.Boucher, R.M.G. 1974. Potentiated acid 1,5
coupling efficiency. Eur. J. Appl. Microbiol. New York. pentanedial solution--a new chemical steril-
Biotechnol. 5:1-11. 30.Aso, C. and Y. Aito. 1962. Polymerization of izing and disinfecting agent. Amer. J. Hosp.
14.Gorman, S.P., E.M. Scott, and A.D. Russell. bifunctional monomers. II. Polymerization of Pharm. 31:546-557.
1980. Antimicrobial activity, uses and mecha- glutaraldehyde. Makromol. Chem. 58:195- 50.Ruijgrok, J.M., M.E. Boon, and J.R. De
nism of action of glutaraldehyde. J. Appl. 203. Wijn. 1990. The effect of heating by micro-
Bacteriol. 48:161-190. 31.Hooper, D.L. 1967. Nuclear magnetic reso- wave irradiation and by conventional heating
15.Nimni, M.E., D. Cheung, B. Strates, M. nance measurements of equilibria involving on the aldehyde concentration in aqueous glu-
Kodama, and K. Sheikh. 1987. Chemically hydration and hemiacetal formation from taraldehyde solutions. Histochem. J. 22:389-
modified collagen: a natural biomaterial for some carbonyl compounds. J. Chem. Soc. 393.
tissue replacement. J. Biomed. Mater. Res. B:169-170. 51.Hardy, P.M., A.C. Nicholls, and H.N. Ry-
21:741-771. 32.Whipple, E.B. and M. Ruta. 1974. Structure don. 1976. The nature of the cross-linking of
16.Gosselin, R.E., H.C. Hodge, R.P. Smith, and of aqueous glutaraldehyde. J. Org. Chem. proteins by glutaraldehyde. Part I. Interaction
M.N. Gleason. 1976. Clinical Toxicology of 39:1666-1668. of glutaraldehyde with the amino-groups of 6-
Commercial Products. Acute Poisoning, 4th 33.Gruen, L.C. and P.T. McTigue. 1963. Hydra- aminohexanoic acid and of alpha-N-acetyl-ly-
ed. The Williams & Wilkins Co., Baltimore, tion equilibria of aliphatic aldehydes in H2O sine. J. Chem. Soc. Perkin Trans. 1:958-962.
MD. and D2O. J. Chem. Soc. (Nov):5217-5223. 52.Hardy, P.M., G.J. Hughes, and H.N. Rydon.
17.Okuda, K., I. Urabe, Y. Yamada, and H. 34.Silverstein, R.M., G.C. Bassler, and T.C. 1979. The nature of the cross-linking of pro-
Okada. 1991. Reaction of glutaraldehyde Morrill. 1981. Spectrometric Identification teins by glutaraldehyde. Part 2. The formation
with amino and thiol compounds. J. Ferment. of Organic Compounds, 4th ed. John Wiley & of quaternary pyridinium compounds by the
Bioeng. 71:100-105. Sons, New York. action of glutaraldehyde on proteins and the
18.Bowes, J.H. and C.W. Cater. 1968. The 35.Bullock, G.R. 1984. The current status of identification of a 3-(2-piperidyl)pyridinium
interaction of aldehydes with collagen. Bio- fixation for electron microscopy: a review. J. derivative, anabilysine, as a cross-linking en-
chim. Biophys. Acta 168:341-352. Microsc. 133:1-15. tity. J. Chem. Soc. Perkin Trans. I:2282-2288.
19.Walt, D.R. and V. Agayn. 1994. The chem- 36.Russell, A.D. and D. Hopwood. 1976. The 53.Lubig, R., P. Kusch, K. Roper, and H.
istry of enzyme and protein immobilization biological uses and importance of glutaralde- Zahn. 1981. Zum reaktionsmechanismus von
with glutaraldehyde. Trends Anal. Chem. hyde. Prog. Med. Chem. 13:271-301. glutaraldehyd mit proteinen. Monatsh. Chem.
13:425-430. 37.Hopwood, D. 1975. The reactions of glutar- 112:1313-1323.
20.Aso, C. and Y. Aito. 1962. Intramolecular- aldehyde with nucleic acids. Histochem. J. 54.Jentoft, N. and D.G. Dearborn. 1979. Label-
intermolecular polymerization of glutaralde- 7:267-276. ling of proteins by reductive methylation us-
hyde. Bull. Chem. Soc. Japan 35:1426. 38.Adami, R.C. and K.G. Rice. 1999. Meta- ing sodium cyanoborhydride. J. Biol. Chem.
21.Hardy, P.M., A.C. Nicholls, and H.N. Ry- bolic stability of glutaraldehyde cross-linked 254:4359-4365.
don. 1969. The nature of glutaraldehyde in peptide DNA condensates. J. Pharm. Sci. 55.Hermanson, G.T. 1996. Bioconjugate Tech-
aqueous solution. J. Chem. Soc. Chem. Com- 88:739-746. niques. Academic Press, San Diego, CA.
mun. 10:565-566. 39.Habeeb, A.F.S.A. and R. Hiramoto. 1968. 56.Zahn, H. 1955. Bridge reactions in amino
22.Korn, A.H., S.H. Feairheller, and E.M. Fi- Reaction of proteins with glutaraldehyde. acids and fibrous proteins. Angew. Chem.
lachione. 1972. Glutaraldehyde: nature of the Arch. Biochem. Biophys. 126:16-26. 67:561-572.
reagent. J. Mol. Biol. 65:525-529. 40.Hopwood, D., C.R. Callen, and M. McCabe. 57.Robertson, E.A. and R.L. Schultz. 1970.
23.Monsan, P., G. Puzo, and H. Mazarguil. 1970. The reactions between glutaraldehyde The impurities in commercial glutaraldehyde
1975. Étude du mécanisme d’établissement and various proteins. An investigation of their and their effect on the fixation of brain. J. Ul-
des liaisons glutaraldehyde protéines. Biochi- kinetics. Histochem. J. 2:137-150. trastruct. Res. 30:275-287.
mie 57:1281-1292. 41.Alexa, G., D. Chisalita, and G. Chirita. 58.Broun, G.B. 1976. Chemically aggregated

Vol. 37, No. 5 (2004) BioTechniques 801


REVIEW

enzymes, p. 263-280. In K. Mosbach (Ed.), capillary zone electrophoresis and mass spec- choline determination in brain tissue homog-
Methods in Enzymology, Academic Press, trometry. Electrophoresis 25:1367-1378. enates. Anal. Chem. 73:2875-2882.
New York. 75.Patramani, I., K. Katsiri, E. Pistevou, T. 89.Bartolini, M., V. Andrisano, and I.W. Wain-
59.Kennedy, J.F. and J.M.S. Cabral. 1983. Kalogeratos, M. Pavlatos, and A.E. Evan- er. 2003. Development and characterization
Immobilized enzymes, p. 253-391. In W.H. gelopoulos. 1969. Glutamic-aspartic transam- of an immobilized enzyme reactor based on
Scouten (Ed.), Solid Phase Biochemistry: inase-antitransaminase interaction: a method glyceraldehyde-3-phosphate dehydrogenase
Analytical and Synthetic Aspects. John Wiley for antienzyme purification. Eur. J. Biochem. for on-line enzymatic studies. J. Chromatogr.
& Sons, New York. 11:28-36. A 987:331-340.
60.Zaborsky, O.R. 1973. Immobilized Enzymes, 76.Broun, G., E. Selegny, S. Avrameas, and D. 90.Chandran, P., M. Thakur, and C.S. Pundir.
CRC Press, Cleveland, OH. Thomas. 1969. Enzymatically active mem- 2001. Improved determination of urinary oxa-
61.Jansen, E.F., Y. Tomimatsu, and A.C. Ol- branes: some properties of cellophane mem- late with alkylamine glass bound barley oxa-
son. 1971. Cross-linking of α-chymotrypsin branes supporting cross-linked enzymes. Bio- late oxidase. J. Biotechnol. 85:1-5.
and other proteins by reaction with glutaralde- chim. Biophys. Acta 185:260-262. 91.St. Clair, N.L. and M.A. Navia. 1992. Cross-
hyde. Arch. Biochem. Biophys. 144:394-400. 77.Torchilin, V.P., A.V. Maksimenko, V.N. linked enzyme crystals as robust biocatalysts.
62.Tomimatsu, Y., E.F. Jansen, W. Gaffield, Smirnov, I.V. Berezin, A.M. Klibanov, and J. Amer. Chem. Soc. 114:7314-7316.
and A.C. Olson. 1971. Physical chemical ob- K. Martinek. 1978. The principles of enzyme 92.Cao, L., F. van Rantwijk, and R.A. Shel-
servations on the α-chymotrypsin glutaralde- stabilization. III. The effect of the length of don. 2000. Cross-linked enzyme aggregates:
hyde system during formation of an insoluble intra-molecular cross-linkages on thermosta- a simple and effective method for the im-
derivative. J. Colloid Interface Sci. 36:51-64. bility of enzymes. Biochim. Biophys. Acta mobilization of penicillin acylase. Org. Lett.
63.Ottesen, M. and B. Svensson. 1971. Modifi- 522:277-283. 2:1361-1364.
cation of papain by treatment with glutaralde- 78.Quiocho, F.A. 1976. Immobilized proteins 93.Tabata, Y., S. Hijikata, M. Muniruzzaman,
hyde under reducing and non-reducing condi- in single crystals, p. 546-558. In K. Mosbach and Y. Ikada. 1999. Neovascularization ef-
tions. C .R. Trav. Lab. Carlsberg 38:171-185. (Ed.), Methods in Enzymology. Academic fect of biodegradable gelation microspheres
64.Jansen, E.F. and A.C. Olson. 1969. Proper- Press, New York. incorporating basic fibroblast growth factor. J.
ties and enzymatic activities of papain insolu- 79.Silman, I. and E. Katchalski. 1966. Water- Biomater. Sci. Polym. Ed. 10:79-94.
bilized with glutaraldehyde. Arch. Biochem. insoluble derivatives of enzymes, antigens, 94.Esposito, E., R. Cortesi, and C. Nastruzzi.
Biophys. 129:221-227. and antibodies. Annu. Rev. Biochem. 35:873- 1996. Gelatin microspheres: influence of
65.Bano, B. and M. Saleemuddin. 1980. Stud- 908. preparation parameters and thermal treatment
ies on chemically aggregated trypsin using 80.Glassmeyer, C.K. and J.D. Ogle. 1971. on chemico-physical and biopharmaceutical
glutaraldehyde. Ind. J. Biochem. Biophys. Properties of an insoluble form of trypsin. properties. Biomater. 17:2009-2020.
17:12-17. Biochem. 10:786-792. 95.Leo, E., M.A. Vandelli, R. Cameroni, and
66.Gupta, M.N. 1993. Cross-linking techniques: 81.Walsh, K.A., L.L. Houston, and R.A. Ken- F. Forni. 1997. Doxorubicin-loaded gelatin
application to enzyme and protein stabiliza- ner. 1970. Chemical modification of bovine nanoparticles stabilized by glutaraldehyde :
tion and bioconjugate preparation, p. 307- trypsinogen and trypsin, p. 56. In P. Desnu- involvement of the drug in the cross-linking
324. In M.E. Himmel and G. Georgiou (Eds.), elle, H. Neurath and M. Ottesen (Eds.), Struc- process. Int. J. Pharm. 155:75-82.
Biocatalyst Design for Stability and Specific- ture—Function Relationships of Proteolytic 96.Wan, L.S.C., P.W.S. Heng, and L.W. Chan.
ity. American Chemical Society, Washington, Enzymes. Academic Press, New York. 1992. Drug encapsulation in alginate micro-
DC. 82.Lilly, M.D., W.E. Hornby, and E.M. Crook. spheres by emulsification. J. Microencapsul.
67.Chui, W.K. and L.S. Wan. 1997. Prolonged 1966. The kinetics of carboxymethylcellulose- 9:309-316.
retention of cross-linked trypsin in calcium ficin in packed beds. Biochem. J. 100:718- 97.Kulkarni, A.R., K.S. Soppimath, T.M. Am-
alginate microspheres. J. Microencapsulation 723. inabhavi, A.M. Dave, and M.H. Mehta.
14:51-61. 83.Robinson, P.J., P. Dunnill, and M.D. Lilly. 2000. Glutaraldehyde crosslinked sodium
68.Krogh, T.N., T. Berg, and P. Hojrup. 1999. 1971. Porous glass as a solid support for im- alginate beads containing liquid pesticide for
Protein analysis using enzymes immobi- mobilization or affinity chromatography of soil application. J. Control. Release 63:97-
lized to paramagnetic beads. Anal. Biochem. enzymes. Biochim. Biophys. Acta 242:659- 105.
274:153-162. 661.
69.Tomer, S., J.G. Dorsey, and A. Berthod. 84.Rapatz, E., A. Travnicek, G. Fellhofer, and Address correspondence to:
2001. Nonionic micellar liquid chromatogra- F. Pittner. 1988. Studies on the immobiliza-
phy coupled to immobilized enzyme reactors. tion of glucuronidase (Part 1). Covalent im- Karen C. Waldron
J. Chromatogr. A 923:7-16. mobilization on various carriers. Appl. Bio- Department of Chemistry
70.Habeeb, A.F. 1967. Preparation of enzymi- chem. Biotechnol. 19:223-234. Université de Montréal,
cally active, water-insoluble derivatives of 85.Wood, W.G. and A. Gadow. 1983. Immobi- C.P. 6128, Succ. Centre-ville
trypsin. Arch. Biochem. Biophys. 119:264- lization of antibodies and antigens on macro Montréal, Canada, H3C 3J7
268. solid phases. A comparison between adsorp- e-mail: karen.waldron@umontreal.ca
71.Herzog, V. and H.D. Fahimi. 1974. Effect tive and covalent binding. A critical study of
of glutaraldehyde on catalase. Biochemical macro solid phases for use in immunoassay
and cytochemical studies with beef liver cata- systems, Part 1. J. Clin. Chem. Clin. Biochem.
lase and rat liver peroxisomes. J. Cell. Biol. 21:789-797.
60:303-311. 86.Dekker, R.F. 1989. Immobilization of a lac-
72.Mattiasson, B. and K. Mosbach. 1976. As- tase onto a magnetic support by covalent at-
say Procedures for Immobilized Enzymes, p. tachment to polyethyleneimine-glutaralde-
335-353. In K. Mosbach (Ed.), Methods in hyde-activated magnetite. Appl. Biochem.
Enzymology. Academic Press, New York. Biotechnol. 22:289-310.
73.Regan, D.L., P. Dunnill, and M.D. Lilly. 87.Badea, M., A. Curulli, and G. Palles-
1974. Immobilized enzyme reaction stability: chi. 2003. Oxidase enzyme immobilisation
attrition of the support material. Biotechnol. through electropolymerised films to assemble
Bioeng. XVI:333-343. biosensors for batch and flow injection analy-
74.Migneault, I., C. Dartiguenave, J. Vinh, sis. Biosens. Bioelectron 18:689-698.
M.J. Bertrand, and K.C. Waldron. 2004. 88.Guerrieri, A. and F. Palmisano. 2001. An
Comparison of two glutaraldehyde immo- acetylcholinesterase/choline oxidase-based
bilization techniques for solid-phase tryptic amperometric biosensors as a liquid chro-
peptide mapping of human hemoglobin by matography detector for acetylcholine and

802 BioTechniques Vol. 37, No. 5 (2004)

You might also like