ArticlePDF AvailableLiterature Review

Abstract and Figures

The process of protein crosslinking comprises the chemical, enzymatic, or chemoenzymatic formation of new covalent bonds between polypeptides. This allows (1) the site-directed coupling of proteins with distinct properties and (2) the de novo assembly of polymeric protein networks. Transferases, hydrolases, and oxidoreductases can be employed as catalysts for the synthesis of crosslinked proteins, thereby complementing chemical crosslinking strategies. Here, we review enzymatic approaches that are used for protein crosslinking at the industrial level or have shown promising potential in investigations on the lab-scale. We illustrate the underlying mechanisms of crosslink formation and point out the roles of the enzymes in their natural environments. Additionally, we discuss advantages and drawbacks of the enzyme-based crosslinking strategies and their potential for different applications.
Content may be subject to copyright.
MINI-REVIEW
Enzyme-catalyzed protein crosslinking
Tobias Heck &Greta Faccio &Michael Richter &
Linda Thöny-Meyer
Received: 25 September 2012 /Revised: 1 November 2012 / Accepted: 2 November 2012
#The Author(s) 2012. This article is published with open access at Springerlink.com
Abstract The process of protein crosslinking comprises the
chemical, enzymatic, or chemoenzymatic formation of new
covalent bonds between polypeptides. This allows (1) the
site-directed coupling of proteins with distinct properties
and (2) the de novo assembly of polymeric protein net-
works. Transferases, hydrolases, and oxidoreductases can
be employed as catalysts for the synthesis of crosslinked
proteins, thereby complementing chemical crosslinking
strategies. Here, we review enzymatic approaches that are
used for protein crosslinking at the industrial level or have
shown promising potential in investigations on the lab-scale.
We illustrate the underlying mechanisms of crosslink for-
mation and point out the roles of the enzymes in their natural
environments. Additionally, we discuss advantages and
drawbacks of the enzyme-based crosslinking strategies and
their potential for different applications.
Keywords Cross-linking .Conjugation .Ligation .Fusion
proteins .Transglutaminase .Sortase A
Introduction
The process of joining protein molecules through intermo-
lecular covalent bonds is commonly referred to as protein
crosslinking. It gives rise to the creation of new macromo-
lecular assemblies that frequently reveal physicochemical
properties and functionalities different from those of the
sole parent compounds. As illustrated by Wong and
Jameson (2012), the terms protein crosslinkingand
protein conjugationare sometimes used to discriminate
between the covalent linkage of proteins that have a natural
affinity in vivo and the covalent coupling of two unrelated
protein species. To avoid confusion in the terminology, we
shall use the term protein crosslinkingin a general sense
throughout this review article to specify the formation of
one or more covalent bonds between proteins.
Consequently, we will cover approaches that result either
in the targeted fusion of distinct protein molecules by a
single covalent bond or in the formation of protein networks
by multiple covalent bonding.
Modifying the properties of proteins by site-directed
fusion or network formation is of great significance for
applications in many fields such as food processing, leather
and textile fabrication, tissue engineering as well as biochem-
ical and biomedical research. The generation of covalent
bonds between proteins can be induced physicochemically
by application of heat, alkaline conditions, mechanical agita-
tion, or photooxidative treatment (Gerrard 2002;Singh1991)
by addition of chemical crosslinkers or by enzyme catalysis.
Chemical crosslinking methodologies take advantage of the
enormous diversity of available crosslinking reagents that
differ in chemical functionality, reactivity, and size (Wong
and Jameson 2012). Homobifunctional and heterobifunctional
chemical crosslinkers carry two reactive groups to target pro-
teins at the same or at different functional groups, respectively.
Among these, glutaraldehyde probably represents the most
commonly used crosslinking reagent (Migneault et al. 2004).
Additionally, there are a few examples of multifunctional
crosslinkers that can be used to target proteins at more than
two functional groups simultaneously. Protein crosslinking
with bi- and multifunctional crosslinkers leads to the incorpo-
ration of molecular spacer groups of defined length and com-
position between the reaction partners. By contrast,
monofunctional crosslinkers (e.g., formaldehyde) react such
that with the exception of the reactive moiety no additional
T. Heck :G. Faccio :M. Richter :L. Thöny-Meyer (*)
Empa, Swiss Federal Laboratories for Materials Science and
Technology, Laboratory for Biomaterials, Lerchenfeldstrasse 5,
CH-9014 St. Gallen, Switzerland
e-mail: linda.thoeny@empa.ch
Appl Microbiol Biotechnol
DOI 10.1007/s00253-012-4569-z
linker is introduced into the final product. Furthermore, acti-
vating agents such as carbodiimides are widely used to direct-
ly connect proteins without incorporating a spacer (Wong and
Jameson 2012).
In addition to the prevalent chemical crosslinking
reagents, enzymes are increasingly employed as catalysts
to promote the introduction of covalent bonds between
protein molecules. In this article, we review the diversity
of enzymatic approaches that can be used for protein cross-
linking in vitro. By addressing the physiological back-
grounds of the enzymes and their underlying coupling
mechanisms, we illustrate how these biomimetic approaches
have emerged to complement the toolbox of protein cross-
linking strategies. Eventually, we discuss advantages and
limitations of choosing and utilizing enzyme-based cross-
linking strategies for different practical scenarios.
Enzymatic protein crosslinking in vivo
The posttranslational covalent modification of proteins is
essential to pro- and eukaryotic cells in order to increase
the structural and functional diversity of the proteome
(Walsh 2006; Walsh et al. 2005). Most of these modifica-
tions are catalyzed by specific enzymes that have evolved
for their respective tasks. Enzymatically introduced modifi-
cations of proteins occur at the functional groups of amino
acid side chains, which are, in some cases, embedded in the
context of sequence-specific recognition motifs. They com-
prise (1) the addition of organic molecules including cofac-
tors (http://www.ebi.ac.uk/thornton-srv/databases/
CoFactor), oligosaccharides, nucleotides, lipids, and small
moieties such as methyl, acetyl, and phosphoryl groups; (2)
intramolecular transformations such as disulfide bond for-
mation and proteolytic processing; and (3) intermolecular
crosslinking by covalent bond formation between individual
protein molecules (Fig. 1).
One of the most extensively studied cellular protein
crosslinking events is the enzyme-catalyzed covalent tether-
ing of ubiquitin to target proteins. Ubiquitin represents a
small protein of 76 amino acid residues with a size of
~8 kDa. The process of ubiquitin attachment, designated
as ubiquitylationor ubiquitination,is traditionally asso-
ciated with protein trafficking to the eukaryotic 26S protea-
some for specific degradation (Hershko and Ciechanover
1998; Hershko et al. 1982). Proteins directed to the protea-
some carry a minimum of four covalently linked ubiquitin
molecules that are connected through highly proteolysis-
resistant isopeptide bonds between the ε-amine of Lys48
and the C-terminus of the successive subunit. The activation
and transfer of ubiquitin to target proteins is catalyzed by a
cascade of three enzymes (E1, E2, and E3) in an adenosine
triphosphate (ATP)-dependent manner (Fig. 2). Nowadays,
it is evident that modifications of proteins with ubiquitin and
other ubiquitinlike proteins influences a great variety of
cellular signaling and regulatory processes depending on
the type and length of the attached ubiquitin chain (mono-
or polyubiquitination) and on the lysine residues involved in
the connection between the individual ubiquitin subunits
(Hochstrasser 2009; Spasser and Brik 2012; Weissman et
al. 2011). Only recently, even a prokaryotic ubiquitin-like
protein (Pup) from Mycobacterium tuberculosis has been
described (Darwin 2009), which is coupled to target proteins
by the action of the ligase PafA (Guth et al. 2011).
Lipidation
N-Myristoylation
S-Palmitoylation
S-Isoprenylation
(farnesyl or geranylgeranyl)
Glycosyl phosphatidylinositol
(GPI) anchorin
g
Intermolecular protein crosslinking
Ubiquitination
Fibrin crosslinking
Collagen and elastin crosslinking
Protein attachment to bacterial surfaces
Bacterial pilus assembly
Intramolecular protein
modification
Disulfide-bond formation
Proteolytic processing
(e.g. signal-peptide removal,
pro-protein maturation)
Attachment of small
functional groups
N-, O-, S-andC-Methylation
N-Acetylation
Phosphorylation
Sulfurylation
Hydroxylation
Cofactor attachment
Biotinylation
Phosphopantetheinylation
Lipoic-acid attachment
Tet r a
py
rrole/heme li
g
ation
TARGET
PROTEIN
Glycosylation
O-, N-andC-Glycosylation
ADP-Ribosylation
Fig. 1 Schematic illustration of
enzyme-catalyzed covalent
modifications of proteins in
vivo; for a more comprehensive
overview we refer to Walsh
(2006) and Walsh et al. (2005).
The cartoon depicted in the
center was created with the
program PyMOL and shows the
structure of ubiquitin (PDB ID:
1ubq) (Schrodinger 2010)
Appl Microbiol Biotechnol
Many physiologically important protein crosslinking reac-
tions in higher eukaryotes are accomplished by transglutami-
nases. Eukaryotic transglutaminases are calcium-dependent
enzymes that catalyze the formation of protein networks by
introducing glutamyl-lysyl isopeptide bonds between target
proteins (Fig. 3). The most prominent member of the mamma-
lian tissue transglutaminases is the fibrin-stabilizing factor XIII,
which participates in blood coagulation by crosslinking anti-
parallel fibrin chains to mechanically stable clots. For addition-
al information on the functional diversity of eukaryotic
transglutaminases, we refer to a review article by Lorand and
Graham (2003). In the late 1980s, the first calcium-independent
transglutaminase from the microbial strain Streptomyces
mobaraensis (formerly classified as Streptoverticillium mobar-
aense) was described in the patent and peer-reviewed literature
(Ando et al. 1989; Motoki et al. 1993). Diverse physiological
functions including mycelium growth and differentiation in S.
mobaraensis (Pasternack et al. 1998) as well as spore coat
formation in Bacillus subtilis (Zilhão et al. 2005) are associated
with microbial transglutaminases.
Another naturally occurring crosslinking reaction between
proteins is catalyzed by sortases that constitute a group of
calcium-dependent enzymes embedded in the membrane of
Gram-positive bacteria (http://nihserver.mbi.ucla.edu/Sortase/).
Based on their primary amino acid sequence, sortases are
currently assigned to six different classes (AF) that exert
highly site-specific transpeptidation reactions (Fig. 3)atthe
bacterial cell surface (Spirig et al. 2011). These include the
anchoring of diverse functional proteins to the growing cell
wall by sortase A (Marraffini et al. 2006; Mazmanian et al.
1999) and the assembly of pili from individual pilin subunits by
sortase C (Hendrickx et al. 2011).
In contrast to the transamidation reactions catalyzed by
transglutaminases and sortases, lysyl oxidases induce the
oxidative crosslinking of collagen and elastin chains in the
extracellular matrix of eukaryotic cells (Lucero and Kagan
2006). The enzymatic step catalyzed by lysyl oxidases com-
prises the deamination of lysyl side chains in collagen and
elastin with the concomitant formation of hydrogen perox-
ide (Fig. 4). The resulting aldehydes can react spontaneous-
ly either with a second aldehyde to yield the corresponding
aldol condensation product or add to the primary amine of a
neighboring lysine residue to generate a Schiff base. The so-
formed crosslinked chains of collagen and elastin constitute
fibrous network structures that largely contribute to the
stabilization of eukaryotic tissues.
Enzymes used for protein crosslinking in vitro
The capability of enzymes to crosslink proteins in vivo
immediately suggests their use for diverse applications in
vitro. Of the aforementioned enzymes, mainly transglutami-
nases have been successfully applied for the introduction of
OH
O
+ ATP AMP + PPi
S
O
SH
NH
On
E 1
E 2
S
O
E 2
E 3
NH2
SH
E 1
Protein 1
Protein 1
Ubiquitin
Ubiquitin
Ubiquitin
Ubiquitin
Fig. 2 Simplified schematic
illustration of the enzymatic
cascade leading to
ubiquitination of target proteins
in eukaryotic cells (Spasser and
Brik 2012)
Fig. 3 Protein crosslinking through transamidation reactions catalyzed by transglutaminase and sortase A. The reactive thioester intermediates
generated at the active site of the enzymes are framed
Appl Microbiol Biotechnol
crosslinks into various protein matrices (Kuraishi et al.
2001; Zhu and Tramper 2008), and also, the crosslinking
reactions catalyzed by sortase A and lysyl oxidase have
recently gained increasing attention. By contrast, the enzy-
matic cascade responsible for protein ubiquitination has not
been investigated for crosslinking applications in vitro, and
indeed, its practical usefulness seems very limited due to the
ATP dependence of the reaction and the complex interplay
of the enzymes E1, E2, and E3 (Fig. 2). Interestingly, further
oxidoreductases that catalyze reactions with nonproteino-
genic substrates in vivo have been investigated for the
synthesis of new covalent bonds between proteins in vitro.
Regarding the enzymatic mechanisms, two types of cross-
linking reactions can be distinguished, i.e., (1) direct cova-
lent bonding catalyzed by transferases (EC 2) and
hydrolases (EC 3) via proteinylenzymethioester inter-
mediates and (2) enzyme-mediated covalent bonding via
reactive species that are enzymatically generated by oxidor-
eductases (EC 1) and spontaneously react further with pro-
teins to form protein networks. In this section, we outline the
physiological functions and biochemical properties of
enzymes relevant to protein crosslinking applications in
vitro. For summaries of the enzymes discussed herein and
the proposed underlying crosslinking reactions, we refer to
Table 1and to Figs. 3and 4, respectively.
Transferases and hydrolases
The common crosslinking reaction catalyzed by transferases
and hydrolases entirely occurs at the active site of the
Fig. 4 Crosslinking of proteins mediated through oxidation by oxidor-
eductases. The reactive species generated by the enzymes are framed.
The illustration of the laccase- and peroxidase-catalyzed oxidation
reactions is simplified and does not show the stoichiometry of
substrates and products (laccase oxidizes four substrate molecules per
molecule of oxygen, whereas peroxidase oxidizes only two substrate
molecules per molecule of hydrogen peroxide)
Appl Microbiol Biotechnol
Table 1 Overview of enzymes used for protein crosslinking in vitro
Enzyme
(EC number)
Main
function
in vivo
Cofactor/metal
ion dependence
Main applications of enzymatic
crosslinking reaction
Target moiety
for crosslink
formation
Mechanism of
crosslink formation
Recent review articles
Transferases
Transglutaminase
(EC 2.3.2.13)
Crosslinking of
diverse proteins
Calcium (only in
eukaryotic trans-
glutaminases)
Network formation of various food
proteins; production of protein
hydrogels; modification of protein
fibers and leather; site-specific
fusion; and immobilization of
tagged target proteins
Glutamyl side chains
of proteins
Formation of glutamyl-
lysyl isopeptide bonds
Griffin et al. (2002),
Lorand and Graham
(2003), Serafini-Fracassini
and Del Duca (2008),
Yokoyama et al. (2004)
Hydrolases
Sortase A
(EC 3.4.22.70)
Attachment of cell-
surface proteins
Calcium Site-specific coupling and immobilization
of engineered target proteins
Threonineglycine
peptide bond of
LPXTG sorting motif
Formation of threonine
glycine peptide bonds
Popp and Ploegh (2011),
Spirig et al. (2011)
Subtilisin
(EC 3.4.21.62)
Extracellular proteolysis Peptide fragment ligation C-terminally esterified
amino acids of
polypeptides
Formation of peptide bonds Gupta et al. (2002)
Oxidoreductases
Tyrosinase
(EC 1.14.18.1)
Melanin formation Copper Network formation of food proteins
(e.g., in meat, fish, yoghurt, wheat-
based products, and edible protein
films); enzyme crosslinking
Tyrosyl side chains
of proteins; phenolic
compounds
Quinone formation;
subsequent spontaneous
1,4-additions
Claus and Decker (2006),
Faccio et al. (2012), Fairhead
and Thöny-Meyer (2012),
Halaouli et al. (2006)
Laccase
(EC 1.10.3.2)
Lignin degradation
and biosynthesis
Copper Network formation of food proteins
(e.g., in meat, fish, wheat-based
products, and edible protein films)
Tyrosyl side chains of
proteins; phenolic
compounds
Radical formation;
subsequent spontaneous
radical couplings
Mayer and Staples (2002),
Quintanar et al. (2007),
Santhanam et al. (2011),
Witayakran and Ragauskas
(2009)
Peroxidase
(EC 1.11.1.x)
Oxidation of diverse
compounds
Mostly heme Network formation of food proteins
(e.g., in wheat-based products);
production of protein hydrogels;
site-specific fusion of tagged target
proteins
Tyrosyl side chains
of proteins; phenolic
compounds
Radical formation;
subsequent spontaneous
radical couplings
Koua et al. (2009), Veitch
(2004)
Lysyl oxidase
(EC 1.4.3.13)/amine
oxidase (EC 1.4.3.6)
Crosslinking of
collagen and elastin
Lysine tyrosinyl
quinone (LTQ);
copper
Production of protein hydrogels Lysyl side chains
of proteins
Aldehyde formation;
subsequent spontaneous
formation of aldol
condensation and Schiff
base products
Lucero and Kagan (2006)
Appl Microbiol Biotechnol
enzymes and proceeds via formation of a covalent pro-
teinylenzymethioester intermediate. Nucleophilic attack
by an incoming amine nucleophile at the carbonyl group
of the thioester intermediate releases the bound protein
moiety from the enzyme, leading to formation of a new
peptide bond between the target molecules (Fig. 3).
Transglutaminases (EC 2.3.2.13)
As outlined previously, transglutaminases catalyze a trans-
amidation reaction between glutamyl and lysyl side chains
of target proteins. The catalytic reaction proceeds via gluta-
mine deamination and formation of a proteinglutamyl
thioester at the active site of the enzyme. Nucleophilic attack
by a lysyl ε-amino group of a second protein at the carbonyl
moiety of the thioester intermediate generates isopeptide-
crosslinked proteins that are largely resistant to proteolysis
by common peptidases (Mariniello et al. 2007). In contrast
to eukaryotic transglutaminases, transglutaminases from mi-
crobial origin are calcium-independent, which represents a
major advantage for their practical use (Griffin et al. 2002;
Yokoyama et al. 2004).
Peptidases (EC 3.4.x)
An exceptional enzyme, which crosslinks proteins in a highly
site-specific manner, is the sortase SrtA (EC 3.4.22.70)
from Staphylococcus aureus (Mazmanian et al. 1999).
Although SrtA is formally assigned to the class of pepti-
dases, the enzyme in vivo catalyzes a transpeptidation reac-
tion to tether surface proteins to the cell wall of Gram-
positive bacteria. SrtA recognizes target proteins containing
a conserved LPXTG amino acid motif, where X represents
any proteinogenic amino acid (Kruger et al. 2004). The
catalytic reaction proceeds via nucleophilic attack by the
enzyme's catalytic cysteinyl thiol group at the threonine
glycine amide bond within the sorting motif of the target
protein. Subsequently, a proteinylenzymethioester inter-
mediate is formed with concomitant release of the C-
terminal target protein moiety. An amine nucleophile, the
terminal amino group of an oligoglycine polypeptide in
vivo, eventually releases the covalently bound protein moi-
ety from the enzyme under formation of a peptide bond
crosslink.
Although common peptidases are associated with peptide
bond hydrolysis, examples of enzymes are described that
can, under certain conditions, catalyze peptide bond forma-
tion between polypeptide fragments (Bordusa 2002). In the
so-called kinetically controlled approach, a synthetic poly-
peptide ester serves as the substrate for enzymatic coupling
by peptidases. The crosslinking reaction is initiated by for-
mation of a covalent reaction intermediate between the
catalytic nucleophile of the peptidase and the carboxy
terminus of the substrate. Eventually, nucleophilic attack
by the terminal amino group of an acceptor polypeptide
leads to release of the bound acyl moiety from the enzyme
and crosslink formation between the polypeptide fragments
through a newly established peptide bond.
Oxidoreductases
With the exception of lysyl oxidase, most oxidoreductases
are not explicitly destined by nature to promote the forma-
tion of covalent bonds between proteins. Nevertheless,
members of the oxidoreductases have been widely investi-
gated for their potential to crosslink proteins in vitro because
they lack pronounced substrate specificity. Many oxidore-
ductases react with a broad range of small molecules such as
low molecular weight phenols and/or macromolecular sub-
strates including functional amino acid side chains of pro-
teins. Covalent bond formation between proteins initiated by
the action of oxidoreductases involves at least two sequen-
tial chemical steps (Fig. 4). Of these, only the initial redox
reaction with the primary substrate is directly catalyzed by
the enzyme. The resulting reactive species, i.e., quinones,
radicals, or aldehydes may subsequently undergo nonenzy-
matic conversions to form various types of covalent bonds.
In this article, we focus on oxidoreductases that have been
experimentally shown to catalyze redox reactions directly at
functional amino acid side chains of target proteins. Hence,
we do not include glucose oxidases (Wong et al. 2008) and
sulfhydryl oxidases (Faccio et al. 2011) that are proposed to
induce intermolecular crosslinking of food proteins mainly
by producing reactive hydrogen peroxide from small-
molecule sugars and thiols, respectively.
Tyrosinases (EC 1.14.18.1)
Tyrosinases are dicopper enzymes that are widespread in
pro- and eukaryotic organisms (Claus and Decker 2006;
Faccio et al. 2012; Halaouli et al. 2006). They initiate the
biosynthetic production of melanins by converting L-tyro-
sine to dopaquinone, which subsequently undergoes spon-
taneous reactions to yield melanin. Furthermore, they are
attributed to other distinct functions including phenol detox-
ification in bacteria and cuticle sclerotization in inverte-
brates. Activated tyrosinases have both monophenolase
and diphenolase activity as they are able to catalyze the
hydroxylation of monophenolic compounds to o-diphenols
as well as the subsequent oxidation of these o-diphenols to
the respective o-quinones with concomitant reduction of
molecular oxygen to water (Espín et al. 2000). In addition
to many low molecular weight mono- and diphenolic mol-
ecules, surface-exposed tyrosyl side chains of proteins may
also serve as substrates for tyrosinases that convert them to
the respective o-quinones (Ito et al. 1984;Matheisand
Appl Microbiol Biotechnol
Whitaker 1984a). These are proposed to react spontaneously
mainly via 1,4-additions with the side chains of lysine, tyro-
sine, histidine, and cysteine residues, depending on their
abundance and accessibility on the target protein, to form
covalent proteinprotein crosslinks (Bittner 2006). Proteins
with weakly defined three-dimensional structure and unfolded
proteins are the preferred targets for crosslinking by tyrosi-
nases, whereas globular proteins are poorly, if at all, converted
by the enzyme (Hellman et al. 2011;Selinheimoetal.2007b).
However, it has been shown that crosslink formation between
proteins that are not accessible to tyrosinases can be induced
by the addition of small-molecule phenolic compounds
(Fairhead and Thöny-Meyer 2010; Jus et al. 2012;
Thalmann and tzbeyer 2002). These molecules likely func-
tion as crosslinking mediators to overcome the absence of
surface-exposed tyrosine residues on the target proteins.
Laccases (EC 1.10.3.2)
Laccases are bluemulticopper oxidases that have been
discovered in fungi, plants, and bacteria (http://www.lcce
d.uni-stuttgart.de/). Most of the characterized laccases orig-
inating from fungi contribute to the degradation of lignins,
whereas laccases derived from plants are mainly related to
lignin and cell wall biosynthesis (Mayer and Staples 2002;
Singh Arora and Kumar Sharma 2010). Prokaryotic laccases
have been proposed to be involved in many intra- and
extracellular processes, but their exact physiological func-
tions remain mostly elusive (Santhanam et al. 2011). The
active sites of laccases harbor a total of four copper atoms
organized at three distinct sites (T1, T2, and T3) that can be
distinguished by their spectral properties (Quintanar et al.
2007). They catalyze single-electron abstractions from a
wide range of phenols and anilines among other substrates
(Reiss et al. 2011; Witayakran and Ragauskas 2009). The
oxidation of four substrate molecules by laccases goes along
with the concomitant reduction of one equivalent of molec-
ular oxygen to water that is the sole by-product of the
reaction. The formed radicals may undergo subsequent cou-
pling reactions of various types leading to the formation of
different covalently linked products. In proteins, mainly
exposed tyrosyl side chains serve as substrates for oxidation
by laccases, and the resulting phenoxy radicals may sponta-
neously initiate subsequent protein crosslinking reactions
(Mattinen et al. 2006). As for tyrosinases, the addition of
phenolic compounds as crosslinking mediators can be neces-
sary to promote protein crosslinking by laccases (Steffensen et
al. 2008).
Peroxidases (EC 1.11.1.x)
The group of peroxidases encompasses miscellaneous
enzymes that universally occur in microorganisms, plants,
and animals. Presently, members of the peroxidases are
assigned to 20 classes with different EC numbers. Due to
the great complexity and diversity of peroxidases, the data-
base PeroxiBase has been created to summarize information
on properties, functions, and regulation of these enzymes
(https://peroxibase.toulouse.inra.fr/) (Koua et al. 2009).
Most peroxidases contain a prosthetic heme group that is
required for the electron transfer reaction, and also, perox-
idases with other cofactors including flavin adenine dinu-
cleotide (FAD) are described. Peroxidases abstract single
electrons from a broad range of substrates of mostly aro-
matic nature (Gumiero et al. 2010), including surface-
accessible tyrosine residues of proteins (Matheis and
Whitaker 1984b). The oxidation of two substrate molecules
is accompanied by the concomitant reduction of hydrogen
peroxide to water. The phenoxy radicals resulting from
tyrosine oxidation by peroxidases may undergo various
spontaneous reactions including radical couplings, which
mainly leads to the formation of different dityrosyl-type
crosslinks between proteins (Heijnis et al. 2011; Matheis
and Whitaker 1984a).
Lysyl oxidases (EC 1.4.3.13)
As described above, lysyl oxidases initiate the crosslinking
of collagen and elastin chains in the extracellular matrix of
higher eukaryotes (Lucero and Kagan 2006). Hence, they
are among the few enzymes that promote the formation of
protein crosslinks in their physiological environments and
likewise have been used for practical crosslinking applica-
tions in vitro (Bakota et al. 2011). Lysyl oxidases require
two cofactors for catalysis, namely, a covalently bound lysine
tyrosinyl quinone (LTQ) cofactor and a copper ion. They
oxidize the primary amine groups of accessible lysyl side
chains on target proteins to the corresponding aldehydes,
which may undergo subsequent reactions to form the cova-
lently crosslinked aldol condensation or Schiff base product.
Formation of protein networks
The covalent assembly of proteins into macromolecular net-
works by enzyme catalysis has been extensively investigat-
ed in various areas of applications where biological protein
matrices with material-like structural and mechanical prop-
erties are required. The ability of enzymes to form cross-
linked protein networks has, for instance, been exploited to
modify the texture and appearance of food products, to
develop new biomimetic tissue scaffolds, or to strengthen
protein-based fibers for textile fabrication. In this section,
we give an overview of enzymatic approaches that are used
in different fields to create products composed of covalent
protein networks.
Appl Microbiol Biotechnol
Food processing
The modification of food proteins by enzymatic crosslink-
ing can affect the long-term stability required for storage or
the organoleptic properties such as structure, texture, ap-
pearance, and flavor of food products. There is extensive
literature available on the enzymatic crosslinking of proteins
in foods, and many aspects of this field have been covered in
a comprehensive book and review articles (Whitehurst and
van Oort 2010; Buchert et al. 2010; Gerrard 2002; Singh
1991). For this reason, we restrict our discussion to the gen-
eral aspects of enzyme-catalyzed crosslinking approaches that
are used to modify the properties of proteins in food products,
and we explicitly highlight selected studies from this rapidly
progressing field of research.
Enzyme preparations suitable for applications in the food
industry must be tested for toxicity and immunogenicity and
certified with the status Generally Recognized As Safe
(GRAS) defined by the US Food and Drug Association
(FDA). For instance, a GRAS status has been assigned to
transglutaminase preparations from S. mobaraensis for pro-
tein crosslinking in seafood, meat, dairy, and cereal products
(FDA/CFSAN agency response letters: GRAS notice numb-
ers 000004 (1998), 000029 (1999), 000055 (2001), and
000095 (2002)). Nowadays, microbial transglutaminase is
produced on large scale and distributed under the trade name
ACTIV by Ajinomoto US, Inc. (http://www.transgluta
minase.com). Transglutaminase is commonly referred to as
meat gluedue to its ability to assemble pieces of meat by
crosslink formation, thereby restoring the structure of meat
from low-quality fresh meat cuts (Kuraishi et al. 1997)or
improving the textural properties of processed meat gels
(Sun and Arntfield 2011). The transglutaminase-catalyzed
crosslinking reaction in meat and fish products is predomi-
nantly directed towards the myofibrillar protein myosin
(Chanarat et al. 2012; Huang et al. 1992; Lantto et al.
2005), which represents the primary constituent of thick
muscle filaments. In addition to muscle proteins, microbial
transglutaminase shows high crosslinking activity with
caseins from milk (Sharma et al. 2001) and glutenins from
wheat grains (Autio et al. 2005; Basman et al. 2002). This
makes the use of the enzyme attractive for applications in
other areas of the food industry such as the dairy and the
baking sectors (Jaros et al. 2006;Joyeetal.2009). For
example, transglutaminase has been used to induce protein
crosslinking during yoghurt making with the aim of super-
seding the addition of dry matter stabilizers that are com-
monly added to enhance the strength of acidified caseinate
gels (Bönisch et al. 2007; Jaros et al. 2006). In noodle and
bread making, transglutaminase-catalyzed crosslinking has
been shown to change the rheological properties and the
microstructure of dough preparations as compared to un-
treated doughs (Kim et al. 2008; Steffolani et al. 2010;Wu
and Corke 2005). For more detailed summaries on the
applications of transglutaminase in the food sector, we refer
to review articles by Motoki and Kumazawa (2000)and
Kuraishi et al. (2001).
The generation of protein crosslinks by oxidative
enzymes including tyrosinases, laccases, and peroxidases
hasalsobeeninvestigatedinseveralareasofthefood
industry (reviewed in Buchert et al. (2010); Whitehurst
and van Oort (2010)). However, in comparison to trans-
glutaminase, relatively little is known on how exactly these
enzymes affect the chemical compositions of protein matri-
ces in foods. It should be noted here that the reaction of
quinones generated by tyrosinases with free amino acids
may influence the color and aroma of the food product,
which may limit the use of tyrosinases in certain applica-
tions of the food industry (Bittner 2006). Nevertheless,
tyrosinase from the filamentous fungus Trichoderma reesei
has been shown to induce the gelation of acidified milk gels.
In contrast to transglutaminase, the studied tyrosinase did
not require preheating of the milk to allow the enzymatic
crosslinking reaction, which could be advantageous for the
production of certain milk products (Ercili Cura et al. 2010).
It has been proposed that the allergenic properties of the
milk protein casein might be mitigated by enzymatic cross-
linking (Stanic et al. 2010). Selinheimo et al. (2007a) eval-
uated the application of tyrosinase from T. reesei and laccase
from Trametes hirsuta on dough preparations from wheat
flour for bread making. Although protein crosslinking of
gliadins and glutenins induced by these enzymes made the
doughs harder and less extensible, the resulting breads were
more voluminous and had softer crumbs than ordinary
breads.
Food packaging
The development of polysaccharide- and protein-based
polymeric films has recently received attention due to the
need for biocompatible and biodegradable materials that
possess mechanical, swelling, and barrier properties suitable
for food packaging. Edible thin films from various different
proteins have been prepared by enzyme-catalyzed crosslink-
ing with microbial transglutaminase (Di Pierro et al. 2006;
Mariniello et al. 2010), and recently, also with tyrosinase
and laccase (Juvonen et al. 2011). For example, films con-
taining transglutaminase-crosslinked whey proteins embed-
ded in a chitosan matrix showed (1) low solubility in water
(2) reduced permeability for oxygen, carbon dioxide, and
water vapor, and (3) enhanced elongation to break but lower
deformability than noncrosslinked films (Di Pierro et al.
2006). Similar changes regarding the barrier properties were
observed in thin film preparations of grapefruit albedo
homogenates combined with the bean protein phaseolin
after transglutaminase-catalyzed crosslinking. However, in
Appl Microbiol Biotechnol
contrast to the whey protein films studied by Di Pierro et al.
(2006), the transglutaminase-treated albedo homogenate
phaseolin films not only revealed higher tensile strength
but also an approximately twofold increase in their elasticity
(Mariniello et al. 2010).
Biomimetic materials
Protein- and polysaccharide-based hydrogels (i.e., highly
water-absorbent polymeric networks) have emerged as bio-
logical scaffolds to support three-dimensional tissue formation
because their mesh structures represent good mimics of the
natural extracellular matrix and offer the possibility to entrap
bioactive molecules such as growth factors. Extensive re-
search has been directed towards the development of enzy-
matic approaches for hydrogel network formation (Moreira
Teixeira et al. 2012). The use of enzymes has gained increas-
ing attention particularly for the synthesis of injectable in situ-
forming hydrogels because enzymes usually react under phys-
iological conditions and, in contrast to many chemicals, are
mostly regarded as biocompatible. Protein hydrogels have
been created by enzyme-catalyzed crosslinking from various
protein sources, mostly from collagen or its denatured form
gelatin. For example, Yung et al. (2007) used microbial trans-
glutaminase to generate crosslinked gelatin hydrogels that are
thermally stable in saline solution at body temperature, allow
proliferation of encapsulated HEK293 cells, and facilitate the
transport of secreted therapeutic proteins such as interleukin-2
(Yung et al. 2010). Moreover, synthetic glutamine- and lysine-
rich oligopeptides have been crosslinked with two eukaryotic
transglutaminases to yield modular cytocompatible protein
hydrogels (Davis et al. 2010). Besides hydrogels, a microfluidic
device composed of a transglutaminase-crosslinked gelatin
mold has been designed to analyze attached cell cultures in an
in vivo-like environment mimicking the extracellular matrix
(Paguirigan and Beebe 2006).
In addition to transglutaminases, horseradish peroxidase
has been exploited to induce the gelation of mainly
polysaccharide-based building blocks including chitosan or
dextran derivatives for applications in tissue engineering
(Jin et al. 2009; Jin et al. 2010). Furthermore, Sofia et al.
(2002) demonstrated the peroxidase-catalyzed crosslinking
of protein hydrogels from differently substituted polyas-
partic acid derivatives. Fetal bovine serum, which contains
lysyl oxidase as a natural enzymatic component, and com-
mercially available plasma amine oxidase have been
employed as catalysts for the production of hydrogels by
oxidative crosslinking of lysine-rich synthetic peptide nano-
fibers (Bakota et al. 2011). Gradually advancing gelation
with time was observed, going along with a steady increase
of robustness of the formed peptide hydrogels as determined
by rheology. Lau et al. (2006) reported the development of a
gene delivery system that allowed gene uptake and local
expression of lysyl oxidase by fibroblasts embedded in a
collagen layer mimicking the extracellular matrix. After
3 weeks of culturing, collagen gels with lysyl oxidase-
transfected cells showed threefold increased mechanical
strength over gels harboring control transgenic cells. This
effect could be attributed to additional crosslinking of the
collagen fibers induced by expression of lysyl oxidase in the
matrix-embedded cells.
Textile and leather manufacturing
The treatment of wool proteins, i.e., mainly keratins, with
proteolytic enzymes is a field of active research in textile-
processing applications. Limited enzymatic hydrolysis of
keratin fibers has been shown to improve the shrink resis-
tance and antifelting behavior of woolen fabrics (Wang et al.
2011). However, the breakdown of keratin has to be tightly
controlled because excessive proteolysis causes drastic fiber
damage and reduces tensile strength. The introduction of
additional crosslinks into the keratin network by the use of
enzymes represents an approach to compensate for the loss
of tensile strength caused by peptidase-catalyzed hydrolysis.
Several studies indicated that keratin fibers crosslinked by
microbial transglutaminase maintain higher fabric strength
after (1) proteinase treatment (Cortez et al. 2004; Du et al.
2007), (2) repeated washing cycles with proteinase-
containing detergents (Cortez et al. 2005), or (3) bleaching
with hydrogen peroxide (Montazer et al. 2011) as compared
to the corresponding fibers without enzyme treatment.
Furthermore, transglutaminase has been employed to graft
casein (Cui et al. 2011), gelatin (Cui et al. 2009), and silk
proteins (Cortez et al. 2007) onto wool yarns, generating
products with improved physicomechanical properties such as
increased tensile strength and higher smoothness. In a similar
approach, wool fibers have been functionalized with ε-linked
polylysine, an FDA-approved biomolecule with antibacterial
properties (Wang et al. 2010). Recent studies demonstrated
that tyrosinase can be employed to oxidize tyrosyl side chains
of keratin, allowing crosslinking of proteins such as collagen
andelastintowoolfibers(Jusetal.2009;Lanttoetal.2012).
In the leather industry, transglutaminase-crosslinked prepara-
tions of gelatin and casein have been examined as low-cost
filling materials to stuff voids in animal hides. The crosslinked
fillers were shown to be homogeneously distributed in the
hides, to resist the washing steps during leather processing and
to improve properties of the leather such as grain smoothness
and fullness (Liu et al. 2011; Taylor et al. 2009; Taylor et al.
2007).
Stabilization of technical enzymes
Approaches using chemical crosslinking reagents such as
glutaraldehyde are extensively used for the preparation of
Appl Microbiol Biotechnol
crosslinked enzyme aggregates (CLEAs) from synthetically
relevant enzymes. CLEAs generally exhibit properties desir-
able for process development such as good operational stabil-
ity, high productivity, and recyclability (Sheldon 2011). To the
best of our knowledge, only one enzyme-based approach to
yield CLEAs has been reported in the scientific literature.
Using the tyrosinase from Verrucomicrobium spinosum in
the presence of phenol, Fairhead and Thöny-Meyer (2010)
reported the formation of catalytically active CLEAs from
Candida antarctica lipase B (CALB). A further evaluation
of enzymatically formed CLEAs is needed in order to assess
their usefulness for biocatalytic transformations as compared
to CLEAs produced by traditional chemical crosslinking strat-
egies. Especially factors including catalyst activity and stabil-
ity, production costs, sustainability, and possibility of
enzymatically introduced cross-contaminations should be
addressed.
Site-directed protein fusion
In accordance with the definition given in the Introduction,
protein crosslinking not only covers the formation of cova-
lently linked protein networks with material-like properties
but also includes the specific linkage of two proteins on the
molecular level. Traditionally, this is achieved by genetic
assembly of the respective encoding DNA regions and subse-
quent heterologous expression of the fusion protein. This
allows combination of the properties of distinct protein spe-
cies, for example, the binding specificity of an antibody with
the immunoregulatory function of a cytokine to create anti-
bodycytokine fusion proteins for anticancer therapy
(Kontermann 2012). However, if expression or correct folding
of the fusion protein fails, alternative methods are required to
link two separately expressed proteins in a site-specific man-
ner. Native chemical ligation (NCL) represents the most ro-
bust and widely applied strategy for the chemoselective
ligation of two synthetically produced, unprotected polypep-
tide chains (Dawson and Kent2000). In NCL, a native peptide
bond is formed between one polypeptide bearing a chemically
introduced C-terminal thioester and a second polypeptide
containing an N-terminal cysteine residue. NCL has been
complemented by intein-based methods, namely, expressed
protein ligation (EPL) and protein trans-splicing (PTS); for a
review on EPL and PTS, we refer to Muralidharan and Muir
(2006). Besides these synthetic and semisynthetic techniques,
hydrolytic enzymes have been used for the formation of native
peptide bonds between polypeptide fragments. The best
known example is the blockwise assembly of fully active
ribonuclease A from six esterified peptide segments in aque-
ous solution using a genetically optimized double mutant of
subtilisin BPN(named subtiligase)fromBacillus amyloli-
quefaciens (Jackson et al. 1994).
Due to its site specificity for the LPXTG sorting motif,
the sortase SrtA from S. aureus has recently emerged as an
enzymatic tool for the site-directed assembly of proteins by
transpeptidation (Tsukiji and Nagamune 2009). Examples
for the SrtA-catalyzed fusion of proteins include the site-
specific coupling of various globular proteins to an IgG
antibody (Levary et al. 2011) and to a single-chain antibody
(Ta et al. 2011). Under optimized reaction conditions, prod-
uct yields of 4085 % were reached and it was shown that
the antibody specificity was retained in the fusion proteins.
It is worth mentioning that SrtA can also be employed for
the oriented immobilization of proteins on glycine-modified
solid supports (Parthasarathy et al. 2007), for the site-
specific introduction of various molecular probes (Popp et
al. 2009), and for protein circularization (Antos et al. 2009).
In addition to SrtA, microbial transglutaminase (Takazawa
et al. 2004; Tanaka et al. 2004)aswellashorseradish
peroxidase (Minamihata et al. 2011) have been investigated
as catalysts to promote the site-specific coupling of proteins
carrying engineered peptide tags.
Advantages, challenges, and limitations of enzymatic
crosslinking strategies
The ability of enzymes to promote the formation of covalent
crosslinks between amino acid residues of proteins offers
versatile possibilities for protein modifications, thereby
complementing the toolbox of chemical crosslinking meth-
odologies. As shown in the previous sections, only a very
limited number of enzymes are frequently used for protein
crosslinking purposes. Because these enzymes differ funda-
mentally with regard to their biochemical properties, reac-
tion mechanisms, and substrate specificities (Table 1and
Figs. 3and 4), they can be exploited to access miscellaneous
crosslinked protein products ranging from fusion proteins
connected through a single covalent bond to protein net-
works linked through a multitude of intermolecular covalent
bonds.
Polymeric networks composed of proteins feature
material-like structural and mechanical properties that are
desirable in many fields of applications, as outlined above.
Unlike many chemicals, enzymes are most active under
mild aqueous reaction conditions and their crosslinking
reactions can often be controlled by modifying temperature,
pH, or ionic strength (as shown, for example, in Heijnis et
al. 2010). For the purpose of generating or extending cova-
lent protein networks, enzymes with a low degree of spec-
ificity towards the amino acid sequence of their target
proteins are preferred. In this respect, microbial transgluta-
minase from S. mobaraensis is by far the most extensively
studied and applied enzyme because (1) it is commercially
available in large quantities, (2) it is rated as nontoxic and
Appl Microbiol Biotechnol
nonimmunogenic by the FDA, (3) it does not require cofac-
tors, and (4) it is active over a wide pH range and resists
temperatures up to 50 °C (Motoki and Kumazawa 2000).
Transglutaminase shows residue, but not sequence specific-
ity, and forms defined isopeptide crosslinks between gluta-
mine and lysine residues of proteins. Similarly, phenol
oxidation by tyrosinases, laccases, and peroxidases can be
regarded as a residue-specific reaction in the context of the
22 proteinogenic amino acids because tyrosine represents
the only proteinogenic amino acid that contains a phenolic
moiety. Nevertheless, crosslink formation mediated by oxi-
doreductases is difficult to control because only the initial
activation of the tyrosyl side chains is enzyme-catalyzed,
and the resulting reactive species may undergo spontaneous
follow-up reactions to form a wide array of diverse covalent
crosslinks involving CC, CO, CN, and CS bonds
(Fig. 4). Transglutaminases as well as oxidoreductases ac-
cept diverse proteins as substrates for crosslinking, provided
they carry the respective amino acid residue required for the
enzymatic reaction at a surface-exposed position. On the
one hand, this implies that any protein lacking accessible
target amino acids will not be enzymatically crosslinked. On
the other hand, the enzyme itself may potentially serve as
substrate for the crosslinking reaction, which could conse-
quently cause its incorporation into the crosslinked protein
network. It should be noted here that oxidoreductases may
also abstract electrons from a wide range of phenols of low
molecular weight such as ferulic or caffeic acid. These
molecules, once activated to the corresponding quinone or
radical species, may act as mediators to crosslink target
proteins that are not directly converted by the enzyme due
to a lack of enzyme-accessible tyrosyl side chains.
While the generation of protein-based polymer networks
relies on the formation of multiple covalent bonds among
individual polypeptide chains, the oriented intermolecular
fusion of two distinct proteins requires the precise formation
of a single covalent bond at a defined position between the
reaction partners. Therefore, the coupling process has to be
tightly controlled and demands a reaction that ensures site
rather than residue specificity. Unlike densely crosslinked
protein matrices, which are commonly intended for indus-
trial bulk applications, site specifically linked fusion pro-
teins are usually produced on a comparably small scale.
Their applications are highly specialized and mostly restrict-
ed to the areas of biochemical and biomedical research. In
addition to the prevalent synthetic and semisynthetic liga-
tion strategies (NCL, EPL, and PTS) (Dawson and Kent
2000; Muralidharan and Muir 2006), proteolytic enzymes
may be employed to catalyze the site-specific formation of
peptide bonds. Although, in most cases, protease-catalyzed
approaches have been exploited to produce peptides of only
short-chain length (Bordusa 2002), the potential of commer-
cially available proteases to assemble intact proteins from
synthetic polypeptide esters has also been demonstrated
(Jackson et al. 1994; Machova et al. 2003). The sortase
SrtA from S. aureus catalyzes transpeptidation reactions of
proteins in vivo and probably represents the most straight-
forward enzymatic approach in vitro to generate site specif-
ically linked fusion proteins. While the aforementioned
chemical- and protease-based approaches to fusion protein
synthesis ensure site specificity by the presence of synthet-
ically introduced C-terminal ester or thioester groups, site
specificity of the SrtA-catalyzed coupling reaction is pro-
vided by the presence of a short LPXTG amino acid motif,
which can be introduced genetically near the C-terminus of
the recombinant target polypeptide (Fig. 3). Advantages
associated with the use of SrtA for the generation of fusion
proteins include (1) facile and efficient production of the
enzyme in E. coli, (2) minimal requirements for target
protein modification due to small recognition motifs, (3)
mild aqueous reaction conditions that sustain the native
conformation of most target proteins, and (4) high yields
of transpeptidation products. However, it is important to
mention that despite the site specificity of SrtA for the
LPXTG-sorting motif, unproductive side reactions may oc-
cur due to the presence of different nucleophiles competing
for the release of the enzyme-bound protein substrate. While
nucleophilic attack by the N-terminal oligoglycine amino
moiety of the fusion partner leads to formation of the direc-
tionally linked protein product (Fig. 3), hydrolysis of the
recognition motif and formation of isopeptidyl-crosslinked
fusion products may also occur as side reactions (Möhlmann
et al. 2011). Thus, thorough reaction optimization and a final
purification step may be necessary for obtaining the desired
fusion protein in a sufficiently pure form. Furthermore, the
need for engineered protein substrates may limit the applica-
tion of the SrtA-catalyzed reaction to the production of high-
value protein products that are difficult to access by chemical
fusion strategies.
Besides the enzymes described in this review, chemical
crosslinking reagents with a broad variety of chemical fea-
tures are available to target the functional groups present in
the amino acid side chains of proteins (Wong and Jameson
2012). While some chemical crosslinkers possess residue
specificity in the context of the naturally occurring amino
acids, others may selectively target different functional
groups in amino acids showing similar chemical reactivity.
Crosslinking strategies based on chemical crosslinkers are
widely used for diverse applications, ranging from funda-
mental studies of proteinprotein interactions at the molec-
ular level to the large-scale production of crosslinked
aggregates of technical enzymes. Furthermore, crosslinking
reagents may be employed to provide target proteins with
new noncanonical functionalities for subsequent chemical
modifications, to attach molecular probes such as fluoro-
phores, or to covalently immobilize proteins on solid
Appl Microbiol Biotechnol
carriers. In contrast to enzymes that serve a catalytic func-
tion and thus are restored after each cycle of crosslink
formation, chemical reagents are consumed during the reac-
tion and hence are usually supplied in at least stoichiometric
amounts in order to allow the crosslinking reaction to pro-
ceed to completion.
Despite their broad commercial availability, reaction ef-
ficiency, and versatility, the use of chemical crosslinking
reagents can be unfavorable particularly when applied in
the areas of food processing and tissue engineering, because
many of these compounds are rated as toxic or may form
harmful by-products leaching from the crosslinked protein
matrix. To overcome these limitations, the study of enzymes
that catalyze the formation of protein crosslinks in vitro has
gained broad attention. However, except the established
approaches based on microbial transglutaminase, the field
of enzymatic protein crosslinking is still in its infancy. There
is a need not only to explore and further improve the protein
crosslinking technology using the currently known enzymes
but also to direct future investigations towards the discovery
of enzymes with new crosslinking activities in order to
broaden the range of target amino acids and protein sub-
strates accessible to enzymatic crosslinking.
Open Access This article is distributed under the terms of the Creative
Commons Attribution License which permits any use, distribution, and
reproduction in any medium, provided the original author(s) and the
source are credited.
References
Ando H, Adachi M, Umeda K, Matsuura A, Nonaka M, Uchio R,
Tanaka H, Motoki M (1989) Purification and characteristics of a
novel transglutaminase derived from microorganisms. Agr Biol
Chem Tokyo 53:26132617
Antos JM, Popp MWL, Ernst R, Chew GL, Spooner E, Ploegh HL
(2009) A straight path to circular proteins. J Biol Chem
284:1602816036. doi:10.1074/jbc.M901752200
Autio K, Kruus K, Knaapila A, Gerber N, Flander L, Buchert J (2005)
Kinetics of transglutaminase-induced cross-linking of wheat pro-
teins in dough. J Agric Food Chem 53:10391045. doi:10.1021/
Jf0485032
Bakota EL, Aulisa L, Galler KM, Hartgerink JD (2011) Enzymatic
cross-linking of a nanofibrous peptide hydrogel. Biomacromole-
cules 12:8287. doi:10.1021/Bm1010195
Basman A, Köksel H, Ng PKW (2002) Effects of transglutaminase on
SDS-PAGE patterns of wheat, soy, and barley proteins and their
blends. J Food Sci 67:26542658. doi:10.1111/j.1365-
2621.2002.tb08794.x
Bittner S (2006) When quinones meet amino acids: chemical, physical
and biological consequences. Amino Acids 30:205224.
doi:10.1007/s00726-005-0298-2
Bönisch MP, Huss M, Weitl K, Kulozik U (2007) Transglutaminase
cross-linking of milk proteins and impact on yoghurt gel proper-
ties. Int Dairy J 17:13601371. doi:10.1016/j.idairyj.2007.01.019
Bordusa F (2002) Proteases in organic synthesis. Chem Rev 102:4817
4867. doi:10.1021/Cr010164d
Buchert J, Ercili Cura D, Ma H, Gasparetti C, Monogioudi E, Faccio
G, Mattinen M, Boer H, Partanen R, Selinheimo E, Lantto R,
Kruus K (2010) Crosslinking food proteins for improved func-
tionality. Annu Rev Food Sci Technol 1:113138. doi:10.1146/
annurev.food.080708.100841
Chanarat S, Benjakul S, H-Kittikun A (2012) Comparative study on
protein cross-linking and gel enhancing effect of microbial trans-
glutaminase on surimi from different fish. J Sci Food Agric
92:844852. doi:10.1002/Jsfa.4656
Claus H, Decker H (2006) Bacterial tyrosinases. Syst Appl Microbiol
29:314. doi:10.1016/j.syapm.2005.07.012
Cortez J, Bonner PLR, Griffin M (2004) Application of transglutami-
nases in the modification of wool textiles. Enzyme Microb Tech-
nol 34:6472. doi:10.1016/j.enzmictec.2003.08.004
Cortez J, Bonner PLR, Griffin M (2005) Transglutaminase treatment of
wool fabrics leads to resistance to detergent damage. J Biotechnol
116:379386. doi:10.1016/j.jbiotec.2004.12.007
Cortez J, Anghieri A, Bonner PLR, Griffin M, Freddi G (2007) Trans-
glutaminase mediated grafting of silk proteins onto wool
fabrics leading to improved physical and mechanical proper-
ties. Enzyme Microb Technol 40:16981704. doi:10.1016/
j.enzmictec.2006.10.013
Cui L, Wang Q, Wang P, Huan Q, Fan X (2009) Transglutaminase-
mediated crosslinking of gelatin onto wool surfaces to improve the
fabric properties. J Appl Polym Sci 113:25982604. doi:10.1002/
App.30300
Cui L, Fan X, Wang P, Wang Q, Fu G (2011) Casein and
transglutaminase-mediated modification of wool surface. Eng
Life Sci 11:201206. doi:10.1002/elsc.201000110
Darwin KH (2009) Prokaryotic ubiquitin-like protein (Pup), pro-
teasomes and pathogenesis. Nat Rev Microbiol 7:485491.
doi:10.1038/Nrmicro2148
Davis NE, Ding S, Forster RE, Pinkas DM, Barron AE (2010)
Modular enzymatically crosslinked protein polymer hydrogels
for in situ gelation. Biomaterials 31:72887297. doi:10.1016/
j.biomaterials.2010.06.003
Dawson PE, Kent SBH (2000) Synthesis of native proteins by chem-
ical ligation. Annual Rev Biochem 69:923960. doi:10.1146/
annurev.biochem.69.1.923
Di Pierro P, Chico B, Villalonga R, Mariniello L, Damiao AE, Masi P,
Porta R (2006) Chitosanwhey protein edible films produced in
the absence or presence of transglutaminase: analysis of their
mechanical and barrier properties. Biomacromolecules 7:744
749. doi:10.1021/Bm050661u
Du G, Cui L, Zhu Y, Chen J (2007) Improvement of shrink-resistance and
tensile strength of wool fabric treated with a novel microbial trans-
glutaminase from Streptomyces hygroscopicus. Enzyme Microb
Technol 40:17531757. doi:10.1016/j.enzmictec.2006.12.001
Ercili Cura D, Lille M, Partanen R, Kruus K, Buchert J, Lantto R
(2010) Effect of Trichoderma reesei tyrosinase on rheology and
microstructure of acidified milk gels. Int Dairy J 20:830837.
doi:10.1016/j.idairyj.2010.06.008
Espín JC, Varón R, Fenoll LG, Gilabert MA, García-Ruíz PA, Tudela J,
García-Cánovas F (2000) Kinetic characterization of the substrate
specificity and mechanism of mushroom tyrosinase. Eur J Bio-
chem 267:12701279. doi:10.1046/j.1432-1327.2000.01013.x
Faccio G, Nivala O, Kruus K, Buchert J, Saloheimo M (2011) Sulfhydryl
oxidases: sources, properties, production and applications. Appl
Microbiol Biotechnol 91:957966. doi:10.1007/s00253-011-3440-y
Faccio G, Kruus K, Saloheimo M, Thöny-Meyer L (2012) Bacterial
tyrosinases and their applications. Process Biochem. doi:10.1016/
j.procbio.2012.08.018
Fairhead M, Thöny-Meyer L (2010) Cross-linking and immobilisation
of different proteins with recombinant Verrucomicrobium spino-
sum tyrosinase. J Biotechnol 150:546551. doi:10.1016/
j.jbiotec.2010.10.068
Appl Microbiol Biotechnol
Fairhead M, Thöny-Meyer L (2012) Bacterial tyrosinases: old enzymes
with new relevance to biotechnology. New Biotechnol 29:183
191. doi:10.1016/j.nbt.2011.05.007
Gerrard JA (2002) Proteinprotein crosslinking in food: methods,
consequences, applications. Trends Food Sci Technol 13:391
399. doi:10.1016/S0924-2244(02)00257-1
Griffin M, Casadio R, Bergamini CM (2002) Transglutaminases:
nature's biological glues. Biochem J 368:377396. doi:10.1042/
BJ20021234
Gumiero A, Murphy EJ, Metcalfe CL, Moody PCE, Raven EL (2010)
An analysis of substrate binding interactions in the heme perox-
idase enzymes: a structural perspective. Arch Biochem Biophys
500:1320. doi:10.1016/j.abb.2010.02.015
Gupta R, Beg QK, Lorenz P (2002) Bacterial alkaline proteases:
molecular approaches and industrial applications. Appl Microbiol
Biotechnol 59:1532. doi:10.1007/s00253-002-0975-y
Guth E, Thommen M, Weber-Ban E (2011) Mycobacterial ubiquitin-
like protein ligase PafA follows a two-step reaction pathway with
a phosphorylated Pup intermediate. J Biol Chem 286:44124419.
doi:10.1074/jbc.M110.189282
Halaouli S, Asther M, Sigoillot JC, Hamdi M, Lomascolo A (2006)
Fungal tyrosinases: new prospects in molecular characteristics,
bioengineering and biotechnological applications. J Appl Micro-
biol 100:219232. doi:10.1111/j.1365-2672.2006.02866.x
Heijnis WH, Wierenga PA, Berkel WJH, Gruppen H (2010) Directing
the oligomer size distribution of peroxidase-mediated cross-linked
bovine α-lactalbumin. J Agr Food Chem 58:56925697.
doi:10.1021/Jf100168x
Heijnis WH, Dekker HL, de Koning LJ, Wierenga PA, Westphal AH,
de Koster CG, Gruppen H, van Berkel WJH (2011) Identification
of the peroxidase-generated intermolecular dityrosine cross-link
in bovine α-lactalbumin. J Agr Food Chem 59:444449.
doi:10.1021/Jf104298y
Hellman M, Mattinen ML, Fu B, Buchert J, Permi P (2011) Effect of
protein structural integrity on cross-linking by tyrosinase evidenced
by multidimensional heteronuclear magnetic resonance spectrosco-
py. J Biotechnol 151:143150. doi:10.1016/j.jbiotec.2010.11.006
Hendrickx APA, Budzik JM, Oh SY, Schneewind O (2011) Architects at
the bacterial surfacesortases and the assembly of pili with isopep-
tide bonds. Nat Rev Microbiol 9:166176. doi:10.1038/Nrmicro2520
Hershko A, Ciechanover A (1998) The ubiquitin system. Annu Rev
Biochem 67:425479. doi:10.1146/annurev.biochem.67.1.425
Hershko A, Eytan E, Ciechanover A, Haas AL (1982) Immunochem-
ical analysis of the turnover of ubiquitin-protein conjugates in
intact cellsrelationship to the breakdown of abnormal proteins.
J Biol Chem 257:1396413970
Hochstrasser M (2009) Origin and function of ubiquitin-like proteins.
Nature 458:422429. doi:10.1038/nature07958
Huang YP, Seguro K, Motoki M, Tawada K (1992) Cross-linking of
contractile proteins from skeletal muscle by treatment with mi-
crobial transglutaminase. J Biochem Tokyo 112:229234
Ito S, Kato T, Shinpo K, Fujita K (1984) Oxidation of tyrosine residues
in proteins by tyrosinaseformation of protein-bonded 3,4-dihy-
droxyphenylalanine and 5-S-cysteinyl-3,4-dihydroxyphenylala-
nine. Biochem J 222:407411
Jackson DY, Burnier J, Quan C, Stanley M, Tom J, Wells JA (1994) A
designed peptide ligase for total synthesis of ribonuclease A with
unnatural catalytic residues. Science 266:243247. doi:10.1126/
science.7939659
Jaros D, Partschefeld C, Henle T, Rohm H (2006) Transglutaminase in
dairy products: chemistry, physics, applications. J Texture Stud
37:113155. doi:10.1111/j.1745-4603.2006.00042.x
Jin R, Moreira Teixeira LS, Dijkstra PJ, Karperien M, van Blitterswijk
CA, Zhong ZY, Feijen J (2009) Injectable chitosan-based hydro-
gels for cartilage tissue engineering. Biomaterials 30:25442551.
doi:10.1016/j.biomaterials.2009.01.020
Jin R, Moreira Teixeira LS, Dijkstra PJ, van Blitterswijk CA, Karperien
M, Feijen J (2010) Enzymatically-crosslinked injectable hydrogels
based on biomimetic dextran-hyaluronic acid conjugates for carti-
lage tissue engineering. Biomaterials 31:31033113. doi:10.1016/
j.biomaterials.2010.01.013
Joye IJ, Lagrain B, Delcour JA (2009) Use of chemical redox agents
and exogenous enzymes to modify the protein network during
breadmakinga review. J Cereal Sci 50:1121. doi:10.1016/
j.jcs.2009.04.001
Jus S, Kokol V, Guebitz GM (2009) Tyrosinase-catalysed coating of
wool fibres with different protein-based biomaterials. J Biomater
Sci 20:253269. doi:10.1163/156856209X404523
Jus S, Stachel I, Fairhead M, Meyer M, Thöny-Meyer L, Guebitz GM
(2012) Enzymatic cross-linking of gelatine with laccase and ty-
rosinase. Biocatal Biotransfor 30:8695. doi:10.3109/
10242422.2012.646036
Juvonen H, Smolander M, Boer H, Pere J, Buchert J, Peltonen J (2011)
Film formation and surface properties of enzymatically cross-
linked casein films. J Appl Polym Sci 119:22052213.
doi:10.1002/App.32943
Kim YS, Huang W, Du G, Pan Z, Chung O (2008) Effects of trehalose,
transglutaminase, and gum on rheological, fermentation, and bak-
ing properties of frozen dough. Food Res Int 41:903908.
doi:10.1016/j.foodres.2008.07.013
Kontermann RE (2012) Antibodycytokine fusion proteins. Arch Bio-
chem Biophys 526:194205. doi:10.1016/j.abb.2012.03.001
Koua D, Cerutti L, Falquet L, Sigrist CJA, Theiler G, Hulo N, Dunand
C (2009) PeroxiBase: a database with new tools for peroxidase
family classification. Nucleic Acids Res 37:D261D266.
doi:10.1093/Nar/Gkn680
Kruger RG, Otvos B, Frankel BA, Bentley M, Dostal P, McCafferty
DG (2004) Analysis of the substrate specificity of the Staphylo-
coccus aureus sortase transpeptidase SrtA. Biochemistry
43:15411551. doi:10.1021/bi035920j
Kuraishi C, Sakamoto J, Yamazaki K, Susa Y, Kuhara C, Soeda T
(1997) Production of restructured meat using microbial transglu-
taminase without salt or cooking. J Food Sci 62:488490.
doi:10.1111/j.1365-2621.1997.tb04412.x
Kuraishi C, Yamazaki K, Susa Y (2001) Transglutaminase: its utiliza-
tion in the food industry. Food Rev Int 17:221246. doi:10.1081/
FRI-100001258
Lantto R, Puolanne E, Kalkkinen N, Buchert J, Autio K (2005) Enzyme-
aided modification of chicken-breast myofibril proteins: effect of
laccase and transglutaminase on gelation and thermal stability. J Agr
Food Chem 53:92319237. doi:10.1021/Jf051602a
Lantto R, Ellis J, Fatarella E, Cortez J (2012) Influence of different
pretreatments on the accessibility of transglutaminase and tyros-
inase to wool fibre proteins. J Text Inst 103:5563. doi:10.1080/
00405000.2010.544103
Lau YKI, Gobin AM, West JL (2006) Overexpression of lysyl oxidase
to increase matrix crosslinking and improve tissue strength in
dermal wound healing. Ann Biomed Eng 34:12391246.
doi:10.1007/s10439-006-9130-8
Levary DA, Parthasarathy R, Boder ET, Ackerman ME (2011) Pro-
teinprotein fusion catalyzed by sortase A. PLoS One 6:e18342.
doi:10.1371/journal.pone.0018342.g002
Liu Q, Liu L, Li J, Zhang D, Sun J, Du G, Chen J (2011) Influence of
microbial transglutaminase modified gelatin-sodium caseinate, as
a filler, on the subjective mechanical and structural properties of
leather. J Am Leather Chem As 106:200207
Lorand L, Graham RM (2003) Transglutaminases: crosslinking
enzymes with pleiotropic functions. Nat Rev Mol Cell Bio
4:140156. doi:10.1038/Nrm1014
Lucero HA, Kagan HM (2006) Lysyl oxidase: an oxidative enzyme
and effector of cell function. Cell Mol Life Sci 63:23042316.
doi:10.1007/s00018-006-6149-9
Appl Microbiol Biotechnol
Machova Z, von Eggelkraut-Gottanka R, Wehofsky N, Bordusa F,
Beck-Sickinger AG (2003) Expressed enzymatic ligation for the
semisynthesis of chemically modified proteins. Angew Chem Int
Ed 42:49164918. doi:10.1002/anie.200351774
Mariniello L, Giosafatto CVL, Di Pierro P, Sorrentino A, Porta R
(2007) Synthesis and resistance to in vitro proteolysis of trans-
glutaminase cross-linked phaseolin, the major storage protein
from Phaseolus vulgaris. J Agr Food Chem 55:47174721.
doi:10.1021/Jf0637269
Mariniello L, Giosafatto CVL, Di Pierro P, Sorrentino A, Porta R
(2010) Swelling, mechanical, and barrier properties of albedo-
based films prepared in the presence of phaseolin cross-linked
or not by transglutaminase. Biomacromolecules 11:23942398.
doi:10.1021/Bm100566j
Marraffini LA, DeDent AC, Schneewind O (2006) Sortases and the art
of anchoring proteins to the envelopes of gram-positive bacteria.
Microbiol Mol Biol Rev 70:192221. doi:10.1128/
Mmbr.70.1.192-221.2006
Matheis G, Whitaker JR (1984a) Modification of proteins by polyphe-
nol oxidase and peroxidase and their products. J Food Biochem
8:137162. doi:10.1111/j.1745-4514.1984.tb00322.x
Matheis G, Whitaker JR (1984b) Peroxidase-catalyzed cross linking of
proteins. J Protein Chem 3:3548. doi:10.1007/BF01024835
Mattinen ML, Hellman M, Permi P, Autio K, Kalkkinen N, Buchert J
(2006) Effect of protein structure on laccase-catalyzed protein
oligomerization. J Agr Food Chem 54:88838890. doi:10.1021/
Jf062397h
Mayer AM, Staples RC (2002) Laccase: new functions for an old enzyme.
Phytochemistry 60:551565. doi:10.1016/S0031-9422(02)00171-1
Mazmanian SK, Liu G, Ton-That H, Schneewind O (1999) Staphylococ-
cus aureus sortase, an enzyme that anchors surface proteins to the
cell wall. Science 285:760763. doi:10.1126/science.285.5428.760
Migneault I, Dartiguenave C, Bertrand MJ, Waldron KC (2004) Glutar-
aldehyde: behavior in aqueous solution, reaction with proteins, and
application to enzyme crosslinking. Biotechniques 37:790802
Minamihata K, Goto M, Kamiya N (2011) Protein heteroconjugation
by the peroxidase-catalyzed tyrosine coupling reaction. Bioconju-
gate Chem 22:23322338. doi:10.1021/bc200420v
Möhlmann S, Mahlert C, Greven S, Scholz P, Harrenga A (2011) In
vitro sortagging of an antibody Fab fragment: overcoming unpro-
ductive reactions of sortase with water and lysine side chains.
Chembiochem 12:17741780. doi:10.1002/cbic.201100002
Montazer M, Lessan F, Pajootan E, Dadashian F (2011) Treatment of
bleached wool with trans-glutaminases to enhance tensile
strength, whiteness, and alkali resistance. Appl Biochem Biotech
165:748759. doi:10.1007/s12010-011-9293-0
Moreira Teixeira LS, Feijen J, van Blitterswijk CA, Dijkstra PJ, Kar-
perien M (2012) Enzyme-catalyzed crosslinkable hydrogels:
emerging strategies for tissue engineering. Biomaterials
33:12811290. doi:10.1016/j.biomaterials.2011.10.067
Motoki M, Kumazawa Y (2000) Recent trends in transglutaminase
technology for food processing. Food Sci Technol Res 6:151
160. doi:10.3136/fstr.6.151
Motoki M, Okiyama A, Nonaka M, Tanaka H, Uchio R, Matsuura A,
Ando H, Umeda K (1993) Production of novel transglutaminase
derived from Streptoverticillium. Japan Patent JP5023744
Muralidharan V, Muir TW (2006) Protein ligation: an enabling tech-
nology for the biophysical analysis of proteins. Nat Methods
3:429438. doi:10.1038/Nmeth886
Paguirigan A, Beebe DJ (2006) Gelatin based microfluidic devices for
cell culture. Lab Chip 6:407413. doi:10.1039/b517524k
Parthasarathy R, Subramanian S, Boder ET (2007) Sortase A as a
novel molecular staplerfor sequence-specific protein conjuga-
tion. Bioconjugate Chem 18:469476. doi:10.1021/Bc060339w
Pasternack R, Dorsch S, Otterbach JT, Robenek IR, Wolf S, Fuchsbauer
HL (1998) Bacterial pro-transglutaminase from Streptoverticillium
mobaraensepurification, characterisation and sequence of the
zymogen. Eur J Biochem 257:570576. doi:10.1046/j.1432-
1327.1998.2570570.x
Popp MWL, Ploegh HL (2011) Making and breaking peptide bonds:
protein engineering using sortase. Angew Chem Int Ed 50:5024
5032. doi:10.1002/anie.201008267
Popp MWL, Antos JM, Ploegh HL (2009) Site-specific protein label-
ing via sortase-mediated transpeptidation. Curr Protoc Protein Sci
15:3.115.3.9. doi:10.1002/0471140864.ps1503s56
Quintanar L, Stoj C, Taylor AB, Hart PJ, Kosman DJ, Solomon EI
(2007) Shall we dance? How a multicopper oxidase chooses its
electron transfer partner. Acc Chem Res 40:445452.
doi:10.1021/Ar600051a
Reiss R, Ihssen J, Thöny-Meyer L (2011) Bacillus pumilus laccase: a
heat stable enzyme with a wide substrate spectrum. BMC Bio-
technol 11:9. doi:10.1186/1472-6750-11-9
Santhanam N, Vivanco JM, Decker SR, Reardon KF (2011) Expres-
sion of industrially relevant laccases: prokaryotic style. Trends
Biotechnol 29:480489. doi:10.1016/j.tibtech.2011.04.005
Schrodinger, LLC (2010) The PyMOL molecular graphics system,
version 1.4.1.
Selinheimo E, Autio K, Krijus K, Buchert J (2007a) Elucidating the
mechanism of laccase and tyrosinase in wheat bread making. J
Agr Food Chem 55:63576365. doi:10.1021/Jf0703349
Selinheimo E, NiEidhin D, Steffensen C, Nielsen J, Lomascolo A,
Halaouli S, Record E, O'Beirne D, Buchert J, Kruus K (2007b)
Comparison of the characteristics of fungal and plant tyrosinases.
J Biotechnol 130:471480. doi:10.1016/j.jbiotech.2007.05.018
Serafini-Fracassini D, Del Duca S (2008) Transglutaminases: wide-
spread cross-linking enzymes in plants. Ann Bot London
102:145152. doi:10.1093/Aob/Mcn075
Sharma R, Lorenzen PC, Qvist KB (2001) Influence of transglutaminase
treatment of skim milk on the formation of ε-(γ-glutamyl)lysine and
the susceptibility of individual proteins towards crosslinking. Int
Dairy J 11:785793. doi:10.1016/S0958-6946(01)00096-6
Sheldon RA (2011) Characteristic features and biotechnological appli-
cations of cross-linked enzyme aggregates (CLEAs). Appl Micro-
biol Biotechnol 92:467477. doi:10.1007/s00253-011-3554-2
Singh H (1991) Modification of food proteins by covalent crosslinking.
Trends Food Sci Technol 2:196200. doi:10.1016/0924-2244
(91)90683-A
Singh Arora D, Kumar Sharma R (2010) Ligninolytic fungal laccases
and their biotechnological applications. Appl Biochem Biotech-
nol 160:17601788. doi:10.1007/s12010-009-8676-y
Sofia SJ, Singh A, Kaplan DL (2002) Peroxidase-catalyzed cross-
linking of functionalized polyaspartic acid polymers. J Mac-
romol Sci Part A: Pure Appl Chem 39:11511181. doi:10.1081/Ma-
120014843
Spasser L, Brik A (2012) Chemistry and biology of the ubiquitin signal.
Angew Chem Int Ed 51:225. doi:10.1002/anie.201200020
Spirig T, Weiner EM, Clubb RT (2011) Sortase enzymes in Gram-
positive bacteria. Mol Microbiol 82:10441059. doi:10.1111/
j.1365-2958.2011.07887.x
Stanic D, Monogioudi E, Dilek E, Radosavljevic J, Atanaskovic-
Markovic M, Vuckovic O, Raija L, Mattinen M, Buchert J,
Cirkovic Velickovic T (2010) Digestibility and allergenicity as-
sessment of enzymatically crosslinked β-casein. Mol Nutr Food
Res 54:12731284. doi:10.1002/mnfr.200900184
Steffensen CL, Andersen ML, Degn PE, Nielsen JH (2008) Cross-linking
proteins by laccase-catalyzed oxidation: importance relative to other
modifications. J Agric Food Chem 56:1200212010. doi:10.1021/
jf801234v
Steffolani ME, Ribotta PD, Pérez GT, León AE (2010) Effect of
glucose oxidase, transglutaminase, and pentosanase on wheat
proteins: relationship with dough properties and bread-making
quality. J Cereal Sci 51:366373. doi:10.1016/j.jcs.2010.01.010
Appl Microbiol Biotechnol
Sun XD, Arntfield SD (2011) Gelation properties of chicken myofi-
brillar protein induced by transglutaminase crosslinking. J Food
Eng 107:226233. doi:10.1016/j.jfoodeng.2011.06.019
Ta HT, Prabhu S, Leitner E, Jia F, von Elverfeldt D, Jackson KE, Heidt
T, Nair AKN, Pearce H, von zur Muhlen C, Wang X, Peter K,
Hagemeyer CE (2011) Enzymatic single-chain antibody tagging
a universal approach to targeted molecular imaging and cell
homing in cardiovascular disease. Circ Res 109:365373.
doi:10.1161/Circresaha.111.249375
Takazawa T, Kamiya N, Ueda H, Nagamune T (2004) Enzymatic
labeling of a single chain variable fragment of an antibody with
alkaline phosphatase by mircobial transglutaminase. Biotechnol
Bioeng 86:399404. doi:10.1002/Bit.20019
Tanaka T, Kamiya N, Nagamune T (2004) Peptidyl linkers for protein
heterodimerization catalyzed by microbial transglutaminase. Bio-
conjugate Chem 15:491497. doi:10.1021/Bc034209o
Taylor MM, Marmer WN, Brown EM (2007) Evaluation of polymers
prepared from gelatin and casein or whey as potential fillers. J Am
Leather Chem As 102:111120
Taylor MM, Lee J, Bumanlag LP, Cooke PH, Brown EM, Hernandez
Balada E (2009) Treatment of low-quality hides with fillers pro-
duced from sustainable resources: effect on properties of leather. J
Am Leather Chem As 104:324334
Thalmann CR, Lötzbeyer T (2002) Enzymatic cross-linking of proteins
with tyrosinase. Eur Food Res Technol 214:276281.
doi:10.1007/s00217-001-0455-0
Tsukiji S, Nagamune T (2009) Sortase-mediated ligation: a gift from
Gram-positive bacteria to protein engineering. Chembiochem
10:787798. doi:10.1002/cbic.200800724
Veitch NC (2004) Horseradish peroxidase: a modern view of a
classic enzyme. Phytochemistry 65:249259. doi:10.1016/
j.phytochem.2003.10.022
Walsh CT (2006) Posttranslational modification of proteins: expanding
nature's inventory. Roberts, Greenwood Village
Walsh CT, Garneau-Tsodikova S, Gatto GJ (2005) Protein posttransla-
tional modifications: the chemistry of proteome diversifications.
Angew Chem Int Ed 44:73427372. doi:10.1002/anie.200501023
Wang Q, Jin G, Fan X, Zhao X, Cui L, Wang P (2010) Antibacterial
functionalization of wool via mTGase-catalyzed grafting of ε-
poly-L-lysine. Appl Biochem Biotechnol 160:24862497.
doi:10.1007/s12010-009-8708-7
Wang P, Wang Q, Cui L, Gao M, Fan X (2011) The combined use of
cutinase, keratinase and protease treatments for wool bio-antifelting.
Fiber Polym 12:760764. doi:10.1007/s12221-011-0760-6
Weissman AM, Shabek N, Ciechanover A (2011) The predator
becomes the prey: regulating the ubiquitin system by ubiquityla-
tion and degradation. Nat Rev Mol Cell Bio 12:605620.
doi:10.1038/Nrm3191
Whitehurst RJ, van Oort M (2010) Enzymes in food technology.
Wiley-Blackwell, Chichester
Witayakran S, Ragauskas AJ (2009) Synthetic applications of
laccase in green chemistry. Adv Synth Catal 351:11871209.
doi:10.1002/adsc.200800775
Wong SS, Jameson DM (2012) Chemistry of protein and nucleic acid
cross-linking and conjugation, 2nd edn. Taylor & Francis, Boca Raton
Wong CM, Wong KH, Chen XD (2008) Glucose oxidase: natural occur-
rence, function, properties and industrial applications. Appl Micro-
biol Biotechnol 78:927938. doi:10.1007/s00253-008-1407-4
Wu J, Corke H (2005) Quality of dried white salted noodles affected by
microbial transglutaminase. J Sci Food Agric 85:25872594.
doi:10.1002/Jsfa.2311
Yokoyama K, Nio N, Kikuchi Y (2004) Properties and applications of
microbial transglutaminase. Appl Microbiol Biotechnol 64:447
454. doi:10.1007/s00253-003-1539-5
Yung CW, Wu LQ, Tullman JA, Payne GF, Bentley WE, Barbari TA
(2007) Transglutaminase crosslinked gelatin as a tissue engi-
neering scaffold. J Biomed Mater Res A 83A:10391046.
doi:10.1002/Jbm.A.31431
Yung CW, Bentley WE, Barbari TA (2010) Diffusion of interleukin-2 from
cells overlaid with cytocompatible enzyme-crosslinked gelatin hydro-
gels. J Biomed Mater Res A 95A:2532. doi:10.1002/Jbm.A.32740
Zhu Y, Tramper J (2008) Novel applications for microbial transgluta-
minase beyond food processing. Trends Biotechnol 26:559565.
doi:10.1016/j.tibtech.2008.06.006
Zilhão R, Isticato R, Martins LO, Steil L, Völker U, Ricca E, Moran
CP, Henriques AO (2005) Assembly and function of a spore coat-
associated transglutaminase of Bacillus subtilis. J Bacteriol
187:77537764. doi:10.1128/Jb.187.22.7753-7764.2005
Appl Microbiol Biotechnol
... Collagen fibrils can be altered significantly by age-related degradative processes driven by matrix metalloproteinases (e.g. collagenases) as well as post-translational enzymatic and non-enzymatic collagen modifications [3,5]. These mechanisms are vital for preserving tissue function [3,[5][6][7][8]. ...
... collagenases) as well as post-translational enzymatic and non-enzymatic collagen modifications [3,5]. These mechanisms are vital for preserving tissue function [3,[5][6][7][8]. While enzymatic collagen cross-linking positively impacts collagen structure, nonenzymatic cross-linking involves processes like glycation or oxidation of proteins, leading to the formation of advanced glycation end-products (AGEs) [3,7]. ...
... These findings highlight the accuracy of echogenicity changes in the LD layer, as indicative of collagen damage due to intrinsic factors values in the revision group 128. 5 ...
Article
Full-text available
Purpose The literature is scarce in exploring the role of imaging parameters like ultrasound (US) as a biomarker for surgical outcomes. The purpose of this study is to investigate the associations between skin US parameters and revision surgery following spine lumbar fusion. Methods Posterior lumbar fusion patients with 2-years follow-up were assessed. Previous fusion or revision not due to adjacent segment disease (ASD) were excluded. Revisions were classified as cases and non-revision were classified as controls. US measurements conducted at two standardized locations on the lumbar back. Skin echogenicity of the average dermal (AD), upper 1/3 of the dermal (UD), lower 1/3 of the dermal (LD), and subcutaneous layer were measured. Echogenicity was calculated with the embedded echogenicity function of our institution’s imaging platform (PACS). Statistical significance was set at p < 0.05. Results A total of 128 patients (51% female, age 62 [54–72] years) were included in the final analysis. 17 patients required revision surgery. AD, UD, and LD echogenicity showed significantly higher results among revision cases 124.5 [IQR = 115.75,131.63], 128.5 [IQR = 125,131.63] and 125.5 [IQR = 107.91,136.50] compared to the control group 114.3 [IQR = 98.83,124.8], 118.5 [IQR = 109.28,127.50], 114 [IQR = 94.20,126.75] respectively. Conclusion The findings of this study demonstrate a significant association between higher echogenicity values in different layers of the dermis and requiring revision surgery. The results provide insights into the potential use of skin US parameters as predictors for revision surgery. These findings may reflect underlying alterations in collagen. Further research is warranted to elucidate the mechanisms driving these associations.
... PNVCL, a non-crystalline polymer, exhibits a T g of 147 • C [27,32,33]. However, it is important to note that the T g of PNVCL can be influenced by several factors, including molecular weight, purity, moisture content, and compositional variations [34][35][36]. ...
... Conversely, chemically crosslinked samples undergo a bonding process whereby covalent bonds form between the polymer chains. The formation of covalent bonds leads to the samples' mechanical strength and stability [35]. Materials that undergo chemical crosslinking frequently demonstrate improved thermal and chemical stability as a result of the robust covalent bonds formed between the polymer chains [36]. ...
Article
Full-text available
Citation: Halligan, E.; Tie, B.S.H.; Colbert, D.M.; Alsaadi, M.; Zhuo, S.; Keane, G.; Geever, L.M. Synthesis and Characterisation of 4D-Printed NVCL-co-DEGDA Resin Using Stereolithography 3D Printing. Macromol 2024, 4, 150-164. https:// Abstract: The design and manufacturing of objects in various industries have been fundamentally altered by the introduction of D-dimensional (3D) and four-dimensional (4D) printing technologies. Four-dimensional printing, a relatively new technique, has emerged as a result of the ongoing development and advancements in 3D printing. In this study, a stimulus-responsive material, N-Vinylcaprolactam-co-DEGDA (NVCL-co-DEGDA) resin, was synthesised by Stereolithography (SLA) 3D printing technique. The N-Vinylcaprolactam-co-DEGDA resins were initiated by the Diphenyl (2,4,6-trimethylbenzoyl) phosphine oxide (TPO) photoinitiator. A range of Di(ethylene glycol) di-acrylate (DEGDA) concentrations in the NVCL-co-DEGDA resin was explored, ranging from 5 wt% to 40 wt%. The structural properties of the 3D printed objects were investigated by conducting Attenuated Total Reflectance-Fourier Transform Infrared Spectroscopy (ATR-FTIR). Additionally, the 3D printed samples underwent further characterisation through differential scanning calorimetry (DSC) and swelling analysis. The results revealed an inverse relationship between DEGDA concentration and T g values, indicating that higher concentrations of DEGDA resulted in lower T g values. Additionally, the pulsatile swelling studies demonstrated that increasing DEGDA concentration prolonged the time required to reach the maximum swelling ratio. These findings highlight the influence of DEGDA concentration on both the thermal properties and swelling behaviour of 3D printed samples.
... Enzymatic cross-linking where enzymes are utilized to create covalent bonds between protein-based materials has recently become prominent owing to its simplicity and nontoxic properties, unlike chemical crosslinkers. 118,127 Fibrinogen is cleaved by thrombin, resulting in the formation of fibrin which is a cross-linked type of hydrogel. 19,128 Therefore, fibrinogen and thrombin were used altogether in the studies that created 3D bioprinted GBM and NB models. ...
Article
Full-text available
Primary brain tumor is one of the most fatal diseases. The most malignant type among them, glioblastoma (GBM), has low survival rates. Standard treatments reduce the life quality of patients due to serious side effects. Tumor aggressiveness and the unique structure of the brain render the removal of tumors and the development of new therapies challenging. To elucidate the characteristics of brain tumors and examine their response to drugs, realistic systems that mimic the tumor environment and cellular crosstalk are desperately needed. In the past decade, 3D GBM models have been presented as excellent platforms as they allowed the investigation of the phenotypes of GBM and testing innovative therapeutic strategies. In that scope, 3D bioprinting technology offers utilities such as fabricating realistic 3D bioprinted structures in a layer-by-layer manner and precisely controlled deposition of materials and cells, and they can be integrated with other technologies like the microfluidics approach. This Review covers studies that investigated 3D bioprinted brain tumor models, especially GBM using 3D bioprinting techniques and essential parameters that affect the result and quality of the study like frequently used cells, the type and physical characteristics of hydrogel, bioprinting conditions, cross-linking methods, and characterization techniques.
... It has already been described that the waterproofing and hardening of the eggshells of insects, including R. prolixus, are related to the crosslinking of proteins catalyzed by peroxidasemediated oxidation at the expense of hydrogen peroxide. This crosslinking occurs in tyrosine residues, forming dityrosine, which emits intrinsic fluorescence under ultraviolet excitation (Margaritis, 1985;Dias et al., 2013;Heck et al., 2013). We found that the silencing of Sec16 resulted in chorions with a 30% reduction Figures 6C,D). ...
Article
Full-text available
In nearly every species of insect, embryonic development takes place outside of the mother’s body and is entirely dependent on the elements that the mother had previously stored within the eggs. It is well known that the follicle cells (FCs) synthesize the eggshell (chorion) components during the process of choriogenesis, the final step of oogenesis before fertilization. These cells have developed a specialization in the massive production of chorion proteins, which are essential for the protection and survival of the embryo. Here, we investigate the function of Sec16, a protein crucial for the endoplasmic reticulum (ER) to Golgi traffic, in the oocyte development in the insect Rhodnius prolixus. We discovered that Sec16 is strongly expressed in vitellogenic females’ ovaries, particularly in the choriogenic oocyte and it is mainly associated with the FCs. Silencing of Sec16 by RNAi caused a sharp decline in oviposition rates, F1 viability, and longevity in adult females. In the FCs, genes involved in the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), and autophagy were massively upregulated, whereas the mRNAs of Rp30 and Rp45—which code for the two major chorion proteins - were downregulated as a result of Sec16 silencing, indicating general proteostasis disturbance. As a result, the outer surface ultrastructure of Sec16-silenced chorions was altered, with decreased thickness, dityrosine crosslinking, sulfur signals, and lower amounts of the chorion protein Rp30. These findings collectively demonstrate the critical role Sec16 plays in the proper functioning of the FCs, which impacts the synthesis and deposition of particular components of the chorion as well as the overall reproduction of this vector.
... Transglutaminase (TG; EC 2.3.2.13) catalyzes the formation of an isopeptide bond between the γ-carboxamide groups of glutamine residue and the ε-amino groups of lysine residues (Ando et al., 1989;Heck et al., 2013). In three previous studies, TG enhanced the overall texture and juiciness of the PBMA patties in a presence-or concentrationdependent manner (Chen et al. 2023;Kim et al. 2022;Sakai et al., 2021). ...
Article
An unresolved issue with plant-based meat analog (PBMA) products is their lower texture and juiciness than animal-derived meat products. Almost all commercial PBMA patties are constructed using methylcellulose (MC). Thus, this study aimed to develop better formulations of binding systems for PBMA patties that simulate the physical properties of beef patties than MC binder. We investigated the formulations of binding systems using crosslinking-catalyzed enzymes (transglutaminase [TG] and laccase [LC]) and 2–4 % carbohydrate binders (MC and sugar beet pectin [SBP]). Texture profile analysis using a rheometer and sensory scores by nine panelists revealed that TG-catalyzed PBMA patties with 3 % MC and LC-catalyzed PBMA patties with 4 % SBP were similar to beef patties than PBMA patties constructed with any MC concentration. These findings suggest that binding systems using crosslinking-catalyzed enzymes and carbohydrate binders could bridge the gap in physical properties between the current MC-based PBMA and beef patties.
Article
Full-text available
Enzymes play an important role in numerous natural processes and are increasingly being utilized as environmentally friendly substitutes and alternatives to many common catalysts. Their essential advantages are high catalytic efficiency, substrate specificity, minimal formation of byproducts, and low energy demand. All of these benefits make enzymes highly desirable targets of academic research and industrial development. This review has the modest aim of briefly overviewing the classification, mechanism of action, basic kinetics and reaction condition effects that are common across all six enzyme classes. Special attention is devoted to immobilization strategies as the main tools to improve the resistance to environmental stress factors (temperature, pH and solvents) and prolong the catalytic lifecycle of these biocatalysts. The advantages and drawbacks of methods such as macromolecular crosslinking, solid scaffold carriers, entrapment, and surface modification (covalent and physical) are discussed and illustrated using numerous examples. Among the hundreds and possibly thousands of known and recently discovered enzymes, hydrolases and oxidoreductases are distinguished by their relative availability, stability, and wide use in synthetic applications, which include pharmaceutics, food and beverage treatments, environmental clean-up, and polymerizations. Two representatives of those groups—laccase (an oxidoreductase) and lipase (a hydrolase)—are discussed at length, including their structure, catalytic mechanism, and diverse usage. Objective representation of the current status and emerging trends are provided in the main conclusions.
Article
Full-text available
Flexible wearable and implantable devices are contributing to the healthcare field by enabling intuitive interfaces with the tissues of human body through their thin form factors, which enable more accurate monitoring of signals and effective delivery of therapy. The development of such devices is accompanied with an increasing interest in strategies and technologies for conformally fixing and adhering flexible biomedical devices in place to acquire high‐quality biosignals over a long period, even with the subtle movements of the target. Owing to the various mechanical properties and wet or dynamic environments of human tissues, it is necessary to use different adhesion strategies that consider the biocompatibility and cohesive properties for each case. This paper provides an in‐depth analysis of recent bio‐adhesives technologies and their practical applications in the healthcare field by classifying them into: 1) Conventional Fixation, 2) Mechanical Fixation, and 3) Chemical Adhesion, based on the mechanism, and 4) Functional Adhesion and 5) Biomimetic Adhesion, based on the unique properties. Furthermore, the principles and detailed mechanisms of each adhesion strategy based on the design and characteristics of the bioadhesives are thoroughly reviewed to provide valuable insights and an overall summary of the prospects and challenges of future bioadhesives technology.
Article
Full-text available
Glutaraldehyde possesses unique characteristics that render it one of the most effective protein crosslinking reagents. It can be present 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.
Article
Full-text available
We recently demonstrated that fillers could be formed inside leather when gelatins alone or mixed proteins, such as gelatin and casein or gelatin and whey, were added to wet blue that had been pretreated with microbial transglutaminase. To monitor these reactions, we had added fluorescently labeled proteins to stock solutions and examined the resultant filled leather using a microscope equipped with an epi-fluorescent attachment. In this present study, based on our understanding of the effect that enzyme modification will have on physical properties of crosslinked proteins, we polymerized potential filler products prior to their addition to the wet blue. We characterized the products with respect to their physical properties and molecular weight distribution (degree of polymerization). These products were applied to wet blue and evaluated, again using fluorescent labels to monitor their filling capability. It was shown that the proteins were evenly distributed throughout the hide and, more importantly, were not removed during the washing steps. Micrographs showing the location of fillers using fluorescent labels are presented.
Book
Since the publication of the first edition of Chemistry of Protein Conjugation and Cross-Linking in 1991, new cross-linking reagents, notably multifunctional cross-linkers, have been developed and synthesized. The completion of the human genome project has opened a new area for studying nucleic acid and protein interactions using nucleic acid cross-linking reagents, and advances have also been made in the area of biosensors and microarray biochips for the detection and analysis of genes, proteins, and carbohydrates. In addition, developments in physical techniques with unprecedented sensitivity and resolution have facilitated the analysis of cross-linked products. Updated to reflect the advances of the 21st century, this book offers: An overview of the chemical principles underlying the processes of cross-linking and conjugation A thorough list of cross-linking reagents published in the literature since the first edition, covering monofunctional, homobifunctional, heterobifunctional, multifunctional, and zero-length cross-linkers Reviews of the use of these reagents in studying protein tertiary structures, geometric arrangements of subunits within complex proteins and nucleic acids, near-neighbor analysis, protein-to-protein or ligand-receptor interactions, and conformational changes of biomolecules Discusses the application of immunoconjugation for immunoassays, immunotoxins for targeted therapy, microarray technology for analysis of various biomolecules, and solid state chemistry for immobilizations.
Book
The second edition of this successful book highlights the widespread use of enzymes in food processing improvement and innovation, explaining how they bring advantages. The properties of different enzymes are linked to the physical and biochemical events that they influence in food materials and products, while these in turn are related to the key organoleptic, sensory and shelf life qualities of foods. Fully updated to reflect advances made in the field over recent years, new chapters in the second edition look at the use of enzymes in the reduction of acrylamide, in fish processing and in non-bread cereal applications such as flour confectionery. Genetic modification of source organisms (GMO) has been used to improve yields of purer enzymes for some time now but the newer technology of protein engineering (PE) of enzymes has the potential to produce purer, more targeted products without unwanted side activities, and a chapter is also included on this important new topic. Authors have been selected not only for their practical working knowledge of enzymes but also for their infectious enthusiasm for the subject. The book is aimed at food scientists and technologists, ingredients suppliers, geneticists, analytical chemists and quality assurance personnel.
Article
This work investigated the influence of filling process, using gelatin-sodium caseinate modified by microbial transglutaminase (MTG) on the subjective, mechanical, and structural properties of leather. The results indicate that in comparison with the control (without filling process), the MTG modified gelatin-sodium caseinate as the fillers improved the subjective properties such as grain tightness, grain smoothness, and fullness, but did not significantly affected the mechanical properties such as tensile strength, elongation at break, and tongue tear. The scanning electron microscopic analysis indicates that the microstructure of the leather treated with the fillers became more regular and tighter compared with the control. The results obtained here may be useful for the application of MTG in the leather processing in improving certain subjective and structural properties of leather.
Article
A microorganism producing transglutaminase was screened as an indication of hydroxamateform ing activity. The microbial transglutaminase was purified from the culture filtrate of the strain, S-8112, which was supposed to belong to the genus Streptoverticillium. The molecular weight of the purified enzyme was found to be about 40, 000 on SDS-polyacrylamide gel electrophoresis, the isoelectric point 8.9 and the optimal pH of the reaction 6-7. The present enzyme requires no calcium ions for its activity. Thus, it clearly differs from known transglutaminases derived from mammalian organs, which have been defined as calcium-dependent enzymes.
Article
Cloverphenes are graphene-type polyarenes, the structures of which resemble the threefold symmetry of clover leafs. In their Communication (10.1002/anie.201104935), D. Peña and co-workers present the synthesis and properties of a nanosized [16]cloverphene derivative, which consists of 16 fused benzene rings (22 benzene rings in total) and 102 sp(2) -hybridized atoms. The synthesis involves sequential [4+2] and [2+2+2] aryne cycloadditions.
Article
Conventional cross-linking of proteins involves the use of toxic chemicals. Here, cross-linking of gelatine and gelatine hydrolysates with tyrosinases from Botryosphaeria obtusa (BoT1 and BoT2), Agaricus bisporus (AbT) and from Verrucomicrobium spinosum (VsT) and with laccases from Trametes hirsuta (ThL) and T. versicolor (TvL) was demonstrated. Enzymatic oxidation of tyrosine residues was indicated by UV/VIS and fluorescence spectroscopy and further confirmed by oxygen consumption measurements. Using a model substrate (Tyr-Ala) dimerization was demonstrated by using RP-HPLC and LC-MS. Enzymatic cross-linking significantly increased the molecular weight of the soluble material up to the point of precipitation as demonstrated by both SDS-PAGE and size exclusion chromatography. The effect of cross-linking was further enhanced in the presence of phenolic molecules such as catechin.
Article
magnified image Laccases (benzenediol:oxygen oxidoreductase, EC 1.10.3.2), multi‐copper‐containing oxidoreductase enzymes, are able to catalyze the oxidation of various low‐molecular weight compounds, specifically, phenols and anilines, while concomitantly reducing molecular oxygen to water. Because of their high stability, selectivity for phenolic substructures, and mild reaction conditions, laccases are attractive for fine chemical synthesis. This review provides a discussion of the recent applications of this interesting enzyme in synthetic chemistry, including laccase and laccase‐mediator catalyzed reactions. In addition, the review also includes a brief discussion of the distribution of laccase in nature, enzyme structure, and the catalytic mechanism which are of relevance to their applications as biocatalysts.