You are on page 1of 10

Appl Microbiol Biotechnol (2006) 71: 773–782

DOI 10.1007/s00253-005-0183-7

MINI-REVIEW

Neetu Dahiya . Rupinder Tewari .


Gurinder Singh Hoondal

Biotechnological aspects of chitinolytic enzymes: a review

Received: 11 June 2005 / Revised: 4 September 2005 / Accepted: 7 September 2005 / Published online: 21 July 2006
# Springer-Verlag 2006

Abstract Chitin and chitinases (EC 3.2.1.14) have an the exoskeleton of arthropods, the outer shell of crusta-
immense potential. Chitinolytic enzymes have wide-rang- ceans, nematodes, etc. Approximately 75% of the total
ing applications such as preparation of pharmaceutically weight of shellfish, such as shrimp, crab, and krill, is con-
important chitooligosaccharides and N-acetyl D-glucos- sidered waste, and chitin comprises 20 to 58% of the dry
amine, preparation of single-cell protein, isolation of pro- weight of the said waste (Wang and Chang 1997). Chitin
toplasts from fungi and yeast, control of pathogenic fungi, has a broad range of applications in biochemical, food, and
treatment of chitinous waste, and control of malaria trans- various chemical industries. It has antimicrobial, anti-
mission. In this review, we discuss the occurrence and cholesterol and antitumor activities (Patil et al. 2000;
structure of chitin, the types and sources of chitinases, their Gooday 1999). Chitin and its related materials are also used
mode of action, chitinase production, as well as molecular in wastewater treatment (Flach et al. 1992), drug delivery
cloning and protein engineering of chitinases and their (Kadowaki et al. 1997), wound healing, and dietary fiber
biotechnological applications. (Dixon 1995; Muzzarelli 1977; Muzzarelli et al. 1999).
Chitinases, which hydrolyze chitin, occur in a wide range
of organisms including viruses, bacteria, fungi, insects,
Introduction higher plants, and animals (Park et al. 1997). The roles of
chitinases in these organisms are diverse. In vertebrates,
Enzyme technology is an interdisciplinary field, and chitinases are usually part of the digestive tract. In insects
enzymes are routinely used in many environmental-friendly and crustaceans, chitinases are associated with the need for
industrial sectors. With the advancement in biotechnology partial degradation of old cuticle. Chitinases have been
especially in the area of genetics, protein engineering, implicated in plant resistance against fungal pathogens
developments in bioinformatics, and the availability of because of their inducible nature and antifungal activities in
sequence data have opened a new era of enzyme appli- vitro (Taira et al. 2002). Chitinase in fungi is thought to have
cations in many industrial processes. autolytic, nutritional, and morphogenetic roles. In viruses,
Chitin, a β-(1,4)-linked polymer of N-acetyl D-glucos- chitinases are involved in pathogenesis (Patil et al. 2000).
amine (GlcNAc), is widely distributed in nature, particu- In bacteria, chitinases play a role in nutrition and
larly as a structural polysaccharide in fungal cell walls in parasitism. In addition to the above potential applications,
chitinases can be used for the production of chitooligosac-
N. Dahiya (*) charides, which have been found to function as antibacterial
Genes and Proteins Laboratory, agents, elicitors of lysozyme inducers, and immunoenhanc-
National Institute of Immunology, ers (Wen et al. 2002). Chitinases can also be used in
Aruna Ashaf Ali Marg, J.N.U. Campus,
New Delhi 110067, India agriculture to control plant pathogens (Dahiya et al. 2005a;
e-mail: ineetudahiya@yahoo.com Karasuda et al. 2003).
Fax: +91-11-6162125 The findings in the catalytic and substrate-binding
mechanisms of chitinolytic enzymes as well as their se-
R. Tewari
Department of Biotechnology, quence homology and applications to plant protection
Panjab University, against fungal pathogens and insect pests were reviewed in
Chandigarh, 160014, India detail by Fukamizo (2000). Various molecular and bio-
technological aspects such as regulation strategies, gene
G. S. Hoondal
Department of Microbiology,
cloning of chitinase from microorganism and plants, and
Panjab University, various industrial and agricultural applications of chitin-
Chandigarh, 160014, India ases have been described by Patil et al. (2000).
774

The present review will focus on the microbial chitinases, Enzyme families
their characteristic molecular cloning and protein engineer-
ing of chitinases, and applications in environmental-friendly Based on amino acid sequence similarity, chitinolytic
industrial and agricultural sectors. enzymes are grouped into families 18, 19, and 20 of
glycosyl hydrolases (Henrissat and Bairoch 1993).
Family 18 is diverse in evolutionary terms and contains
Chitin occurrence and structure chitinases from bacteria, fungi, viruses, animals, and some
plant chitinases. Family 19 consists of plant chitinases
Chitin is widely distributed in nature, particularly as a (classes I, II, and IV) and some Streptomyces chitinases
structural polysaccharide in fungal cell walls, the exoskel- (Hart et al. 1995). The chitinases of the two families, that is,
eton of arthropods, the outer shell of crustaceans, and 18 and 19, do not share amino acid sequence similarity.
nematodes. It is one of the most abundant biopolymer They have completely different 3-D structures and molec-
found on earth, second to cellulose. Chitin is a β-(1,4)- ular mechanisms and are therefore likely to have evolved
linked GlcNAc polymer. Several reports are available from different ancestors (Suzuki et al. 1999). Family 20
regarding the presence of chitin in fungal cell walls. In includes the β-N-acetylhexosaminidases from bacteria,
filamentous fungi and basidiomycetes, it comprises 16% of Streptomycetes, and humans.
the dry weight of the organism. Cell walls of Mucoraceae Bacterial chitinases are clearly separated into three major
are known to have chitosan in addition to chitin. In yeast, subfamilies, A, B, and C, based on the amino acid sequence
the amount of chitin in the cell wall is much lower, but bud of individual catalytic domains (Watanabe et al. 1993).
scars have been shown to be largely composed of chitin Subfamily A chitinases have the presence of a third domain
(Kuranda and Robbins 1991). Approximately 75% of the corresponding to the insertion of an α+β fold region
total weight of shellfish, such as shrimp, crab, and krill, is between the seventh and eighth (α/β)8 barrel. On the other
considered waste, and chitin comprises 20 to 58% of the hand, none of the chitinases in subfamilies A and B have
dry weight of the said waste (Wang and Chang 1997). this insertion. Several chitinolytic bacteria that possess
Chitin is synthesized from the activated precursor uri- chitinases belonging to different subfamilies like Serratia
dine diphosphate N-acetyl-D-glucosamine by the enzyme marcescens (Suzuki et al. 1999), Bacillus circulans WL-12
chitin synthase. Since chitin is important in the growth of (Alam et al. 1995), and Streptomyces coelicolor A3(2)
fungi, it is most likely that there exists a system whereby it (Saito et al. 1999) are reported.
can be remodeled by a concerted breakdown and buildup to
allow a degree of plasticity in the wall, which would be
important in budding and in the elongation of hyphae. Proposed catalytic mechanism of chitinases
GlcNAc is a constituent part of an asparagine-linked oligo-
saccharide of glycoprotein (Shaikh and Desphande 1993). There are two general mechanistic pathways for acid-
The x-ray diffraction studies revealed that chitin occurs in catalyzed glycosyl hydrolysis that results in the following:
three polymorphic forms, that is, α-, β-, and γ-chitins,
1. Retention of the stereochemistry of the anomeric oxygen
which differ in the arrangement of molecular chains within
at C-1 relative to the initial configuration (scheme 1) or
the crystal cell. In α-chitin, chains are arranged in an
2. Inversion of the stereochemistry (scheme 2; Tews et al.
antiparallel fashion, whereas, in β-chitin, chains are parallel.
1997)
In γ-chitin, chains are in mixed form (Peberdy 1985).
Among these, α-chitin is the most abundant form of chitin. An example of retaining mechanism is hen egg white
lysozyme. The mechanism is believed to proceed as follows.
The β-(1,4) glycosidic oxygen is first protonated (leading to
Chitin-degrading enzymes an oxocarbenium ion intermediate), then stabilized by a
second carboxylate (either through covalent or electrostatic
Chitinases can be classified into two major categories. interactions). Nucleophilic attack by water yields the hydro-
Endochitinases (EC 3.2.1.14) cleave chitin randomly at lysis products, which necessarily retains the initial anomeric
internal sites, generating low molecular mass multimers of configuration. This is commonly referred to as the double-
GlcNAc, such as chitotetraose, chitotriose, and diacetylchi- displacement mechanism of hydrolysis (Fig. 1a). Although
tobiose. Exochitinases can be divided into two subcategories: the x-ray crystal structure of family 19 chitinase reveals a
chitobiosidases (EC 3.2.1.29), which catalyze the progres- lysozyme-like fold (suggesting a double-displacement
sive release of diacetylchitobiose starting at the nonreducing mechanism), the hydrolysis products for two family 19
end of chitin microfibril, and β-(1,4) N-acetyl glucosamini- chitinases show inversion of anomeric configuration. This
dases (EC 3.2.1.30), which cleave the oligomeric products of leads to the second commonly discussed hydrolysis mech-
endochitinases and chitobiosidases, generating monomers of anism, a concerted single-displacement reaction in which a
GlcNAc (Sahai and Manocha 1993). An alternative pathway bound water molecule acts as the nucleophile (Fig. 1b).
involves the deacetylation of chitin to chitosan, which is The crystal structure suggested that the second catalytic
finally converted to glucosamine residues by the action of carboxylate may be sufficiently close to allow coordination
chitosanase (EC 3.2.1.132). Characteristics of some chi- of a water molecule consistent with a single-displacement
tinases and their sources are summarized in Table 1. mechanism (Brameld and Goddard 1998a,b).
775
Table 1 Physiological properties of purified chitinase from microbial sources
Microorganism Optimum Optimum Application Inhibitors Reference
pH temperature (°C)

Enterobacter sp. 5.5 45 Release of fungal protoplasts, production of N-bromosuccinamide Dahiya et al.
NRG4 GlcNAc, antifungal potential 2005a,b,c
Enterobacter sp. 7.0 40 n.s. EDTA, PCMB Park et al. 1997
G-1
Enterobacter 6.0 55 n.s. Hg2+, Co2+, Mg2+ Tang et al. 2001
aerogenes
Alcaligenes 5.0 50 Antifungal potential Cu2+, Na+ Vaidya et al.
xylosoxydans 2003
Vibrio alginolyticus 4.0, 9.0 45 Preparation of chitopentoase and chitotriose n.s. Murao et al. 1992
TK-22
Bacillus sp. BG-11 7.5–9.0 45–55 Antifungal potential Allosamidin, EDTA, Bhushan and
iodoacetic acid Hoondal 1998
Bacillus sp.NCTU2 7.0 60 n.s. n.s. Wen et al. 2002
Bacillus sp. 13.26 7.0–8.0 60 n.s. Mn2+, Ca2+ Yuli et al. 2004
Serratia marces- 4.0–7.0 30 n.s. n.s. Roberts and
cens QMB1466 Cabib 1982
Serratia plymuthica 5.4 55 Antifungal potential Co2+, Cu2+ Frankowski et al.
HRO-C48 2001
n.s. Not specified

Family 18 chitinases are reported to yield hydrolysis ion intermediate formed through a chimeric assistance by the
products that retain the anomeric configuration at C-1. neighboring N-acetyl group (Tews et al. 1997).
However, the x-ray crystal structure of two family 18
chitinases reveals the absence of a second acidic residue in
the active site, which is capable of stabilizing the oxo- Synergistic action of multiple forms of chitinases
carbenium ion. Thus, neither the single- nor the double-
displacement mechanism is consistent with the observed Most of the chitinolytic organisms produce multiple
structure and hydrolysis products. An increasing body of isomeric forms of chitinases, which may result from post-
experimental and theoretical evidence points an oxazoline translational processing of a single-gene product or, more

Fig. 1 a The double-displacement hydrolysis mechanism proposed configuration. b The single-displacement hydrolysis mechanism
for family 18 chitinases. Protonation of a GlcNAc residue in a boat proposed for family 19 chitinases. Two acidic residues are required
conformation leads to an oxazoline intermediate, which may be in the active site, and the hydrolysis product shows inversion of the
hydrolyzed to form a product with retention of the anomeric anomeric configuration (Brameld and Goddard 1998b)
776

often, the products of multiple genes. The heterogeneity of Chitinase production


chitinases was attributed to posttranlational modifications
such as differential glycosylation and/or proteolysis. Microbial chitinase has been produced by liquid batch
Multiple chitinolytic enzymes have been reported in fermentation, continuous fermentation, and fed-batch fer-
several microorganisms such as S. marcescens (Suzuki et mentation. In addition to these, solid-state fermentation and
al. 2002), Aeromonas sp. No. 10S-24 (Ueda et al. 1995), biphasic cell systems have also been used for the pro-
Pseudomonas aeruginosa K-187 (Wang and Chang 1997), duction of chitinase. Generally, chitinase produced from
B. circulans WL-12 (Mitsutomi et al. 1998), Bacillus microorganisms is inducible in nature.
licheniformis X-74 (Takayanagi et al. 1991), Streptomyces Extracellular chitinase production is reported to be
sp. J. 13-3 (Okazaki et al. 1995), and Streptomyces griseus influenced by media components such as carbon sources,
HUT 6037 (Itoh et al. 2002). nitrogen sources, and agricultural residues such as rice
Suzuki et al. (2002) reported the synergistic action of bran, wheat bran, etc. (Bhushan 1998; Dahiya et al. 2005b).
chitinases Chi A, Chi B, and Chi C1 of S. marcescens 2170 An enhancing effect of glucose on chitinase production
on chitin degradation. They proposed that despite having was reported by Bhushan (1998) when glucose was used
similar catalytic domains, Chi A and Chi B were con- with chitin in production medium. However, a suppressing
sidered to digest chitin chains in the opposite direction. Chi effect of glucose on chitinase production was reported by
A was proposed to degrade the chitin chain from the Miyashita et al. (1991).
reducing end, whereas Chi B, from the nonreducing end. Several other physical factors such as aeration, pH, and
Addition of Chi A after treatment of powdered chitin with incubation temperature also affect chitinase production.
Chi B and vice versa generally improved chitin degradation The addition of amino acids and their analogs such as
efficiency. tryptophan, tyrosine, glutamine, and arginine (0.1 mM) in
A thermophilic bacterium, B. licheniformis X-74, posses- the growth medium stimulated chitinase production from
ses four chitinases, I, II, III, and IV. Chitinases II, III, and IV Bacillus sp. BG-11 (Bhushan 1998).
produced (GlcNAc)2 and GlcNAc, whereas chitinase I Some other methods, such as cell immobilization
predominantly produced (GlcNAc)2. Chitinases II, III, and (Bhushan 1998), biphasic cell systems (Chen and Lee
IV also catalyzed a transglycosylation reaction that con- 1995), solid-state fermentations, etc., have been used for
verted (GlcNAc)4 into (GlcNAc)6 (Takayanagi et al. 1991). improving chitinase production from different microorgan-
isms (Bhushan 1998). In an immobilized system, whole
cell immobilization of an organism to a solid support such
Chitinase inhibitors as polyurethane foam was applied. Chitinase production
was enhanced up to 4.8-fold over a period of 72 h in
In addition to general enzyme inhibitors, such as organic submerged fermentation.
compounds and oxidizing/reducing agents, a number of Enhanced production of extracellular chitinase by S.
reports are available on the natural chitinase inhibitors. marcescens in an aqueous two-phase system (ATP) of PEG
Allosamidin, an antibiotic produced by Streptomyces and dextran was reported by Chen and Lee (1995). They
sp., is a known specific inhibitor of chitinases from insects, reported a maximum chitinase activity of 41.5 units in
yeast, fungi, and human serum. It is similar to GlcNAc but ATPs [2% (w/w) PEG 20,000 and 5.0% (w/w) dextran T
lacks a pyranose ring oxygen and contains an oxazoline 500] compared with 13.6 units in a polymer-free system.
ring in which the methyl group is substituted by dimethyl- In solid substrate fermentation using flake chitin as the
amine. Allosamidin is a competitive inhibitor. Allosamidin solid substrate, the maximum chitinase yield obtained from
exerts its inhibitory effect by acting as a nonhydrolyz- Enterobacter sp. NRG4 was 616 U/g solid substrate after
able analog of the oxazolinium ion intermediate (Tews et 168 h of growth at 30°C and 75% moisture level. When
al. 1997). It was reported that allosamidin inhibited the wheat bran was used in combination with chitinous sub-
Bacillus sp. BG-11 chitinase by 50 and 70% at a con- strates, the chitinase yield increased. Maximum enzyme
centration of 30 and 50 μg/ml of enzyme solution, re- yield was 1,475 U/g at wheat bran to flake chitin ratio, 1;
spectively, with an IC50 value of 40 μM (Bhushan 1998). moisture, 80%; and inoculum, 2.6 ml after 168 h (Dahiya
Psammaplin A, a brominated tryrosine-derived com- et al. 2005b).
pound, was found to be a noncompetitive inhibitor of
chitinase B from S. marcescens, a family 18 chitinase.
Crystallographic studies suggest that a disordered Psam- Molecular cloning and protein engineering
maplin A molecule is bound near the active site (Tabudravu
et al. 2002). Omura et al. (2000) isolated a compound A number of attempts have been made to clone and express
named argifin from Gliocladium sp. FTD 0668. The IC50 genes from several organisms such as B. circulans WL-12
value of argifin against Lucilia cuprina chitinase was 3.7 mM. (Mitsutomi et al. 1998), Enterobacter agglomerans
Another chitinase inhibitor, argadin, was isolated from (Chernin et al. 1997), and S. marcescens 2170 (Suzuki et
Clonostachys sp. FO-7314 by Arai et al. (2000). Its IC50 al. 1998) into E. coli.
values against L. cuprina chitinase were 34 nM at 20°C and These proteins were expressed in E. coli to study the
150 nM at 37°C. A new chitinase inhibitor, CI-4, was isolated degradation of chitin by chitinases Chi A, Chi B, and Chi C
from Pseudomonas sp. IZ08 by Izumida et al. (1996).
777

from S. marcescens (Suzuki et al. 2002). The chitinase provide precise information about their structures, cyto-
gene from S. marcescens was also expressed in E. coli and chemical localization studies can reveal the functional
Pseudomonasfluorescens 701E1, and it was reported that specialization of these polymers. Wheat germ agglutinin–
the gene was expressed more efficiently in E. coli, as gold complex and chitinase gold complex have been used
compared with Pseudomonas (Fuchs et al. 1986). The as probes for the detection of GlcNAc residues in the
chitinase gene from Streptomyces lividans was cloned in E. secondary cell walls of plants and in pathogenic fungi
coli to study induction pattern (Miyashita et al. 1991). They (Benhamou and Asselin 1989).
suggested the role of a 12-bp direct repeat in the induction Grenier et al. (1991) report the tagging of a barley
of chitinase by chitin and its repression by glucose. chitosanase with colloidal gold particles for the localization
Recently, chitinase A from Enterobacter sp. G-1 and of chitosan in spore and hyphal cell walls of fungi. This
chitosanase A from Matsuebacter chitosanotabidus were technique was used for the detection of chitosan in the cell
cloned in the yeast Schizosaccharomyces pombe to study walls of Ophiostoma ulmi and Aspergillus niger. Chitinase
the functional expression of these enzymes and their effect gold-labeled complexes have also been used for the
on morphogenesis in S. pombe. In this host, chitinase was immunocytochemical and cytochemical localization of
expressed inside the cells, whereas chitosanase was chitin and N-acetyl-D-glucosamine residues in a biotrophic
expressed as a secretion product (Shimono et al. 2002). mycoparasite, Piptocephalis virginiana (Manocha and
A number of reports are available on the cloning of Zhonghua 1997).
chitinases either to increase biocontrol efficiency of Bacillus
thuringiensis to prepare highly active chitinase preparation
or to produce transgenic plants for increased resistance Production of single-cell protein
against insects. Sampson and Gooday (1998) reported
chitinolytic activities of two strains of B. thuringiensis. The solid waste from shellfish processing is mainly
They reported enhanced virulence of B. thuringiensis with composed of chitin, CaCO3, and protein. Revah-Moiseev
increased chitinase production. Two chitinases, Chi 35 and and Carrod (1981) suggested the use of shellfish waste for
Chi 25, from Streptomyces thermophilus OPC 520 were the bioconversion of chitin to yeast single-cell protein
cloned in E. coli, and it was reported that the polysaccharide- (SCP) using chitinolytic enzymes. They used the S.
binding domain of Chi 35 is involved in the hydrolysis of marcescens chitinase system to hydrolyze the chitin and
insoluble chitin and antifungal activity (Tsujibo et al. 2001). Pichia kudriavazevii to yield SCP (with 45% protein and
A family 18 chitinase gene, chiA, from thermophile 8–11% nucleic acids). The commonly used fungi as the
Rhodothermus marinus was cloned and expressed in E. source of SCP are Hansenula polymorpha, Candida
coli. It was reported to be the most thermostable chitinase tropicalis, Saccharomyces cerevisiae, and Myrothecium
isolated from bacteria (Hobel et al. 2005). In another report, verrucaria. Vyas and Deshpande (1991) utilized the
two chitinase genes encoding ChiCH and ChiCW of Bacil- chitinolytic enzymes of M. verrucaria and S. cerevisiae
lus cereus 28-9 were cloned in pGEX-6P-1 and expressed in for the production of SCP from chitinous waste. The total
E. coli cells as soluble glutathione S-transferase–chitinase protein content was reported to be 61%, with very low
fusion proteins (Huang and Chen 2005). contents of nucleic acids (3.1%). Cody et al. (1990)
Such studies are essential for designing a more efficient suggested the enzymatic conversion of chitin to ethanol.
chitinase producer and production of transgenic plants that The criteria used to evaluate SCP production are growth
can be used for the control of fungal and insect pathogens. yield, total protein, and nucleic acid contents. The protein
Furthermore, biochemical and molecular studies could lead content in organisms used was between 39 and 73%,
to a better understanding of the chitinase secretory process whereas the nucleic acid contents were 1–11%. The best
and the development of cloning strategies suitable for reported was that of S. cerevisiae, which exhibited >60%
secretion of desired products. proteins and 1–3% nucleic acid contents.

Application of chitinases Isolation of protoplasts

Chitinases have shown an immense potential for increasing Fungal protoplasts have been used as an effective
the production of several useful products in the most experimental tool in studying cell wall synthesis, enzyme
economic way. The major applications of chitinases are synthesis, and secretion, as well as in strain improvement
discussed in the following subsections. for biotechnological applications. Since fungi have chitin
in their cell walls, the chitinolytic enzyme seems to be
essential along with other wall-degrading enzymes for
Cytochemical localization of chitin/chitosan using protoplast formation from fungi. Dahiya et al. (2005a)
chitinase chitosanase gold complexes reported the effectiveness of Enterobacter sp. NRG4
chitinase in the generation of protoplasts from Trichoderma
Chitin and chitosan are the most ubiquitous polymers of reesei, Pleurotus florida, Agaricus bisporus, and A. niger.
fungal cell walls. Although biochemical analysis can Mizuno et al. (1997) isolated protoplast from Schizophyl-
778

lum commune using the culture filtrate of B. circulans (2002) produced GlcNAc from α-chitin using crude
KA-304. An enzyme complex from B. circulans WL-12 chitinolytic enzymes from Aeromonas hydrophila H-2330.
with high chitinase activity was effective in generating
protoplasts from Phaffia rhodozyme (Johnson et al.
1979). Chitinase as a target for biopesticides

Chitin is present in the exoskeleton and gut lining of


Production of chitooligosaccharides, glucosamine, insects. The molting enzyme chitinase has been described
and GlcNAc from Bombyx mori (silkworm), Manduca sexta (tobacco
hawkmoth), and several other species. Similarly, chitinases
Chitooligosaccharides, glucosamines, and GlcNAc have an have been implicated in different morphological events in
immense pharmaceutical potential. Chitooligosaccharides fungi (Villagomez-Castro and Lopez-Romero 1996).
are potentially useful in human medicines. For example, Allosamidin, a potent inhibitor of chitinase, was found to
chitohexaose and chitoheptaose showed antitumor activity. be inhibitory to the growth of mite (Tetranychus urticae)
A chitinase from Vibrio alginolyticus was used to prepare and a housefly larva (Musca domestica) after ingestion
chitopentaose and chitotriose from colloidal chitin (Murao (Sakuda et al. 1987). Chitinase inhibitors can be explored
et al. 1992). A chitinase preparation from S. griseus was used as potential biopesticides.
for the enzymatic hydrolysis of colloidal chitin. The chito-
biose produced was subjected to chemical modifications to
give novel disaccharide derivatives of 2-acetamido 2-deoxy Estimation of fungal biomass
D-allopyranose moieties that are potential intermediates for
the synthesis of an enzyme inhibitor, that is, N,N′-diacetyl-β- A variety of methods have been described to quantify fungi
chitobiosyl allosamizoline (Terayama et al. 1993). in soil. The techniques include direct microscopic observa-
Specific combinations of chitinolytic enzymes would be tion and extraction of fungus-specific indicator molecules
necessary to obtain the desired chain length of the oligomer. such as glucosamine ergosterol. A strong correlation has
For example, the production of chitooligosaccharides requires been reported between chitinase activity and fungal popu-
high levels of endochitinase and low levels of N-acetyl- lation in soils. Such correlation was not found for bacteria
glucosaminidase and exochitinase, whereas the production and actinomycetes. Thus, chitinase activity appears to be a
of GlcNAc requires higher proportion of exochitinase and suitable indicator of actively growing fungi in soil. Miller et
N-acetylglucosaminidase (Aloise et al. 1996). Alterna- al. (1998) reported the correlation of chitinase activity with
tively, transglycosylation activity of a variety of endochi- the content of fungus-specific indicator molecules 18:2ωb
tinases and N-acetylglucosaminidases will also be useful to phospholipid fatty acid and ergosterol using specific
generate desired chitooligomers, oligomers with changed methylumbelliferyl substrates. Similarly, chitinase and chi-
glycosidic linkages and glycopeptides. tin-binding proteins can be used for the detection of fungal
Nanjo et al. (1989) observed the accumulation of infections in humans (Laine and Lo 1996).
hexamer when tetramer or pentamer was incubated with
Nocardia orientalis chitinase. A chitinase from T. reesei
also exhibited a similar type of efficient transglycosylation Mosquito control
reaction. They reported the accumulation of hexamer and
dimer as the major product when the enzyme was reacted The worldwide socioeconomic aspects of diseases spread
with tetramer (Usui et al. 1990). They also observed a by mosquitoes made them potential targets for various pest
chain elongation from dimer to hexamer and heptamer control agents. In case of mosquitoes, entomopathogenic
using lysozyme catalysis in the presence of 30% ammo- fungus such as Beauveria bassiana could not infect the
nium sulfate in a buffered medium. Chi-26 from Strep- eggs of Aedes aegypti, a vector of yellow fever and dengue,
tomyces kurssanovii showed the accumulation of hexamer and other related species due to the aquatic environment.
in the reaction mixture containing tetramer and pentamer The scarabaeid eggs laid in the soil were found to be
(Stoyachenko et al. 1994). susceptible to B. bassiana (Ferron 1985). M. verrucaria, a
The transglycosylation reaction of Mucor hiemalis saprophytic fungus, produces a total complex of an insect
endo-β-N-acetyl glucosaminidase was used for the prep- cuticle-degrading enzyme (Shaikh and Desphande 1993).
aration of sugar derivatives modified at C-1 or C-2 for the It has been seen that both first and fourth instar larvae of
synthesis of glycopeptides (Yamanoi et al. 2004). mosquito A. aegypti can be killed within 48 h with the help
A chitinolytic enzyme preparation from N. orientalis of the crude preparation from M. verrucaria (Mendonsa et
IFO12806 was used for the preparation of GlcNAc from al. 1996). Though 100% mortality was observed within
chitooligosaccharides (Sakai et al. 1991). Crude bacterial 48 h, purified endochitinase lethal times (LT50) were 48
chitinases from Burkholderia cepacia TU09 and B. and 120 h for first and fourth instar larvae, respectively.
licheniformis SK-1 were used for the hydrolysis of α- However, the time period was found to be decreased,
chitin (from crab shells) and β-chitin (from squid pens) to corresponding to 24 h and 48 h, when the purified chitinase
produce GlcNAc (Pichyangkura et al. 2002). Sashiwa et al. was supplemented with lipolytic activity.
779

Morphogenesis and health. Bhushan and Hoondal (1998) studied the


compatibility of a thermostable chitinase from Bacillus
Chitinases play an important role in yeast and insect sp. BG-11 with the commonly used fungicides and
morphogenesis. Kuranda and Robbins (1991) reported the insecticides.
role of chitinases in cell separation during growth in S. A Fusarium chlamydosporum strain, a mycoparasite of
cerevisiae, and Shimono et al. (2002) studied the functional groundnut rust (Puccinia arachidis), produces endochitin-
expression of chitinase and chitosanase and their effects on ase that inhibits germination of uredospores of rust fungus
morphogenesis in the yeast S. pombe. When the chiA gene (Mathivanan et al. 1998). Chitinolytic enzymes of T.
was expressed in S. pombe, yeast cells grow slowly and harzianum were found to be inhibitory to a wide range
cells become elongated, but when the choA gene was of fungi than similar enzymes from other sources (Lorito
expressed, cells become swollen. Expression of both chiA et al. 1993). Govindsamy et al. (1998) reported the use
and choA genes resulted in elongated and fat cells. of purified preparation of M. verrucaria chitinase to con-
trol a groundnut rust, P. arachidis. Penicillium janthi-
nellum P9 caused mycelial damage in Mucor plumbus
Medical application and Cladosporium cladosporiodes (Giambattista et al.
2001).
Chitinases can be employed in human health care, such as Partially purified chitinase from T. harzianum destroys
making ophthalmic preparations with chitinases and micro- the cell wall of Crinipellis perniciosa, the casual agent of
biocides. A direct medical use has been suggested for witches’ broom disease of cocoa (DeMarco et al. 2000).
chitinases in the therapy for fungal diseases in potentiating Chitinase from B. cereus YQ 308 inhibited the growth of
the activity of antifungal drugs (Pope and Davis 1979; plant pathogenic fungi such as Fusarium oxysporum,
Orunsi and Trinci 1985). They can also be used as potential Fusarium solani, and P. ultimum (Chang et al. 2003).
additives in antifungal creams and lotions due to their topical
applications.
Conclusion and future prospects

Control of plant pathogenic fungi Several aspects of chitinases have stimulated research in
the study of biochemical, regulatory, and molecular aspects
Biological control or the use of microorganisms or their of chitinolytic enzyme systems. Researchers are now
secretions to prevent plant pathogens and insect pests offers directing their studies toward the discovery and engineer-
an attractive alternative or supplement for the control of ing of novel enzymes that are more robust with respect to
plant diseases. Therefore, biological control tactics have their kinetics and the identification of active site residues.
become an important approach to facilitate sustainable In the future, protein engineering will offer the possibility
agriculture (Wang et al. 2002). of generating chitinases with entirely new functions.
Chitin application increased the population of chitino-
lytic actinomycetes, fungi, and bacteria. The increase is
Acknowledgements The authors thank the Council of Scientific
shown to be correlated with the reduction in pathogenic and Industrial Research (CSIR), Government of India for financial
fungi and nematodes and, more importantly, with the support in carrying out these investigations.
reduction of infectivity and, hence, crop damage (Wang et
al. 2002). A biological control agent of fungal root
pathogen should exert a sufficient amount of antagonistic References
activity.
The chitinase produced by Enterobacter sp. NRG4 was Alam MM, Nikaidou N, Tanaka H, Watanabe T (1995) Cloning and
highly active toward Fusarium moniliforme, A. niger, sequencing of chic gene of Bacillus circulans WL-12 and
Mucor rouxi, and Rhizopus nigricans (Dahiya et al. 2005a). relationship of its product to some other chitinases and chitinase
like proteins. J Ferment Bioeng 80:454–461
The chitinase from Alcaligenes xylosoxydans inhibited the Aloise PA, Lumme M, Haynes CA (1996) N-acetyl D-glucosamine
growth of Fusarium udum and Rhizoctonia bataticola production from chitin waste using chitinase from Serratia
(Vaidya et al. 2001). marcescens. In: Muzzarelli RAA (ed) Chitin enzymology, vol
Mahadevan and Crawford (1997) reported the antag- 2. Eur Chitin Soc, Grottammare, pp 581–594
Arai N, Shiomi K, Yamaguchi Y, Masuma R, Iwai Y, Turberg A,
onistic action of Streptomyces lydicus WXEC108 against Kolbl H, Omura S (2000) Argadin, a new chitinase inhibitor,
Pythium ultimum and Rhizoctonia solani, which cause produced by Clonostachy sp. FO. 7314. Chem Pharm Bull
disease in cotton and pea. Horsch et al. (1997) suggested (Tokyo) 48:1442–1446
the use of N-acetylhexosaminidase as a target for the Benhamou N, Asselin A (1989) Attempted localization of a
substrate for chitinase in plant cells reveals abundant N-acetyl
design of low molecular weight antifungals. Chitinases D-glucosamine residues in secondary walls. Biol Cell 67:341–
can be added as a supplement to the commonly used 350
fungicides and insecticides not only to make them more Bhushan B (1998) Isolation, purification, characterization and scale-
potent but also to minimize the concentration of chemi- up production of a thermostable chitinase from an alkalophilic
cally synthesized active ingredients of the fungicides and microorganism. Ph.D. thesis, Department of Microbiology,
Panjab University, Chandigarh
insecticides that are otherwise harmful to the environment
780
Bhushan B, Hoondal GS (1998) Isolation, purification and proper- Henrissat B, Bairoch A (1993) New families in the classification of
ties of a thermostable chitinase from an alkalophilic Bacillus sp. glycosylhydrolases based on amino acid sequence similarities.
BG-11. Biotechnol Lett 20:157–159 Biochem J 293:781–788
Brameld KA, Goddard III WA (1998a) Substrate distrortion to a Hobel CF, Hreggvidsson GO, Marteinsson VT, Bahrani-Mougeot F,
boat conformation at subsite-1 is critical in the mechanism of Einarsson JM, Kristjansson JK (2005) Cloning, expression and
family 18 chitinases. J Am Chem Soc 120:3571–3580 characterization of a highly thermostable family 18 chitinase
Brameld KA, Goddard III WA (1998b) The role of enzyme distortion from Rhodothermus marinus. Extremophiles 9:53–64
in the single displacement mechanism of family 19 chitinases. Horsch M, Mayer C, Sennhauser U, Rast DM (1997) β-N-
Proc Natl Acad Sci U S A 120:4276–4281 acetylhexosaminidase: a target for the design of antifungal
Chang WT, Chen CS, Wang SL (2003) An antifungal chitinase agents. Pharma 76:187–218
produced by Bacillus cereus with shrimp and crab shell powder Huang CJ, Chen CY (2005) High level expression and character-
as carbon source. Curr Microbiol 47:102–108 ization of two chitinases ChiCH and ChiCW of Bacillus cereus
Chen JP, Lee MS (1995) Enhanced production of Serratia 28-9 in Escherichia coli. Biochem Biophys Res Commun
marcescens chitinase in PEG/dextran aqueous two-phase 372:8–17
system. Enzyme Microb Technol 17:1021–1027 Itoh Y, Kawase T, Nikajdou N, Fukada H, Mitsutomi M, Watanabe
Chernin LS, De la Fuente L, Sobolev V (1997) Molecular cloning T, Itoh Y (2002) Functional analysis of the chitin binding
structural analysis and expression in Escherichia coli of a domain of a family 19 chitinase from Streptomyces griseus
chitinase gene from Enterobacter agglomerans. Appl Environ HUT6037: substrate-binding affinity and cis-dominant increase
Microbiol 63:834–839 of antifungal function. Biosci Biotechnol Biochem 66:
Cody RM, Davis ND, Lin J, Shaw D (1990) Screening micro- 1084–1092
organisms for chitin hydrolysis and production of ethanol from Izumida H, Imamura N, Sano H (1996) A novel chitinase inhibitor
aminosugars. Biomass 21:285–295 from a marine bacterium, Pseudomonas sp. J Antibiot (Tokyo)
Dahiya N, Tewari R, Tiwari RP, Hoondal GS (2005a) Production of 49:76–80
an antifungal chitinase from Enterobacter sp. NRG4 and its Johnson EA, Villa TG, Lewis MJ, Phaff HJ (1979) Lysis of the cell
application in protoplast production. World J Microbiol wall of yeast Phaffia rhodozyme by lytic complex from Bacillus
Biotechnol (in press) circulans WL-12. J Appl Biochem 1:273–282
Dahiya N, Tewari R, Tiwari RP, Hoondal GS (2005b) Chitinase Kadowaki S, Saskiawan I, Watanabe J, Yamamoto K, Bunno M,
production in solid state fermentation by Enterobacter sp. Ichihara Y, Kumagai H (1997) Transglycosylation activity of β-
NRG4 using statistical experimental design. Curr Microbiol N-acetylhexosaminidase from Penicillium oxalicum and its
51:1–9 application to synthesis of a drug carrier. J Ferment Bioeng
Dahiya N, Tewari R Tiwari RP, Hoondal GS (2005c) Chitinase from 83:341–345
Enterobacter sp. NRG4: its purification, characterization and Karasuda S, Tanaka S, Kajihara H, Yamamoto Y, Koga D (2003)
reaction pattern. Electronic J Biotechnol (in press) Plant chitinase as a possible biocontrol agent for use instead
DeMarco JL, Lima CLH, Desousa MV, Felix CR (2000) A of chemical fungicides. Biosci Biotechnol Biochem 67:
Trichoderma harzianum chitinase destroys the cell walls of 221–224
the phytopathogen Crinipellis pernicosa, the casual agent of the Kuranda MJ, Robbins PW (1991) Chitinase is required for cell
witches broom disease of cocoa. World J Microbiol Biotechnol separation during growth of Saccharomyces cerevisiae. J Biol
16:383–386 Chem 266:19707–19758
Dixon B (1995) Using fungal dressings to heal wounds. Biotechnol Laine LA, Lo CJ (1996) Diagnosis of fungal infections with a
13:120–121 chitinase. PCT Int Appl Wo 9802742 A122 (CA 128, 86184)
Ferron P (1985) Fungal control. In: Kerkut GA, Gilbert LI (eds) Lorito M, Harman GE, Hayes CK, Broadway RM, Tronsmo A, Woo
Comprehensive insect biochemistry and pharmacology, vol 12. SL, DiPietro A (1993) Chitinolytic enzymes produced by
Pergamon, Oxford, pp 313–346 Trichoderma harzianum: antifungal activity of purified en-
Flach J, Pilet PE, Jolles P (1992) What’s new in chitinase research. dochitinase and chitobiosidase. Phytopathology 83:302–307
Experimentia 48:701–706 Mahadevan B, Crawford DL (1997) Properties of the antifungal
Frankowski J, Lorito M, Scala F, Schmid R, Berg G, Bahl H (2001) biocontrol agent Streptomyces lydicus WYEC 108. Enzyme
Purification and properties of two chitinolytic enzymes of Microb Technol 20:489–493
Serratia plymuthica HRO-C48. Arch Microbiol 176:421–426 Manocha MS, Zhonghua Z (1997) Immunochemical and cytochem-
Fuchs RL, Mcpherson SA, Drahos DJ (1986) Cloning of Serratia ical localization of chitinase and chitin in infected hosts of a
marcescens gene encoding chitinase. Appl Environ Microbiol biotrophic mycoparasite, Piptocephalis virginiana. Mycologia
51:504–509 89:185–194
Fukamizo T (2000) Chitinolytic enzymes: catalysis, substrate Mathivanan N, Kabilan V, Murugesan K (1998) Purification,
binding and their application. Curr Protein Pept Sci 1:105–124 characterization and anti-fungal activity from Fusarium
Giambattista RD, Federici F, Petruccioli M, Fence M (2001) The chlamydosporum, a mycoparasite to groundnut rust, Puccinia
chitinolytic activity of Penicillium janthinellum P9: purifica- arachidis. Can J Microbiol 44:646–651
tion, partial characterization and potential application. J Appl Mendonsa ES, Vartak PH, Rao JU, Deshpande MV (1996) An
Microbiol 91:498–505 enzyme from Myrothecium verrucaria that degrades insect
Gooday GW (1999) Aggressive and defensive roles of chitinases. cuticle for biocontrol of Aedes aegypti mosquito. Biotechnol
EXS 87:157–169 Lett 18:373–376
Govindsamy V, Gunaratna KR, Balasubramanian R (1998) Proper- Miller M, Palofarvi A, Rangger A, Reeslev M, Kjoller A ( 1998)
ties of extracellular chitinase from Myrothecium verrucaria, an The use of fluorogenic substrates to measure fungal presence
antagonist to the groundnut rust Puccinia arachidis. Can J Plant and activity in soil. Appl Environ Microbiol 64:613–617
Pathol 20:62–68 Mitsutomi O, Isono M, Uchiyama A, Nikaidov N, Ikegami T,
Grenier J, Benhamou N, Asselin A (1991) Colloidal gold- Watanabe T (1998) Chitosanase activity of the enzyme
complexed chitosanase: a new probe for ultrastructural local- previously reported as β-1,3-1,4-glucanase from Bacillus
ization of chitosan in fungi. J Gen Microbiol 137:2007–2015 circulans WL 12. Biosci Biotechnol Biochem 62:2107–2114
Hart PJ, Pfluger HD, Monzingo AF, Hoihi T, Robertus JD (1995) Miyashita K, Fujii T, Sawada Y (1991) Molecular cloning and
The refined crystal structure of an endochitinase from characterization of chitinase gene from Streptomyces lividans.
Hordeum vulgare L. seeds at 1.8Å resolution. J Mol Biol J Microbiol 137:2065–2072
248:402–413
781
Mizuno K, Kimura O, Tachiki T (1997) Protoplast formation from Shaikh SA, Desphande MV (1993) Chitinolytic enzymes their
Schizophyllum commune by a culture filtrate of Bacillus contribution to basic and applied research. World J Microbiol
circulans KA-304 grown on a cell-wall preparation of S. Biotechnol 9:468–475
commune as a carbon source. Biosci Biotechnol Biochem Shimono K, Matsuda H, Kawamukai M (2002) Functional expres-
61:852–857 sion of chitinase and chitosanase, and their effects on
Murao S, Kuwada T, Itoh H, Oyama H, Shin T (1992) Purification morphologies in the yeast Schizosaccharomyces pombe. Biosci
and characterization of a novel type of chitinase from Vibrio Biotechnol Biochem 66:1143–1147
alginolyticus TK-22. Biosci Biotechnol Biochem 56:368–369 Stoyachenko IA, Varlamov VP, Davankov VA (1994) Chitinases of
Muzzarelli RAA (1977) Chitin. Pergamon, Oxford Streptomyces kurssanovii: purification and some properties.
Muzzarelli RA, Mattioli-Balmonte M, Pugnaloni A, Biagini G Carboydr Polym 24:47–54
(1999) Biochemistry, histology and clinical uses of chitins and Suzuki K, Sugawara N, Suzuki M, Uchiyama T, Katouno F,
chitosans in wound healing. EXS 87:251–264 Nikaidou N, Watanabe T (2002) Chitinases, A, B and C1 of
Nanjo F, Sakai K, Ishikawa M, Isobe K, Usui T (1989) Properties Serratia marcescens 2170 produced by recombinant E. coli:
and transglycosylation reaction of a chitinase from Nocardia enzymatic properties and synergism on chitin degradation.
orientalis. Agric Biol Chem 53:2189–2195 Biosci Biotechnol Biochem 66:1075–1083
Okazaki K, Kato F, Watanabe N, Yasuda S, Masui Y, Hayakawa S Suzuki K, Suzuki M, Taiyoji M, Nikaidou N, Watanabe T (1998)
(1995) Purification and properties of two chitinases from Chitin binding protein (CBP21) in the culture supernatant of
Streptomyces sp. J-13-3. Biosci Biotechnol Biochem 59: Serratia marcescens 2170. Biosci Biotechnol Biochem 62:
1586–1587 128–135
Omura S, Arai N, Yamaguchi Y, Masuma R, Iwai Y, Namikashi M, Suzuki K, Taiyoji M, Sugawara N, Nikaidou N, Henrissat B,
Turberg A, Kolbl H, Shiomi K (2000) Argifin, a new chitinase Watanabe T (1999) The third chitinase gene (chiC) of Serratia
inhibitor, produced by Gliocladium sp. FDD-0668, I. Taxon- marcescens 2170 and the relationship of its product to other
omy fermentation and biological activities. J Antibiot (Tokyo) bacterial chitinases. Biochem J 343:587–596
53:603–608 Tabudravu JN, Eijsink VA, Gooday GW, Jaspar M, Komander D,
Orunsi NA, Trinci APJ (1985) Growth of bacteria on chitin, fungal Legg M, Synstad B, Van-Aalten DM (2002) Psammaplin A, a
cell walls and fungal biomass, and the effect of extracellular chitinase inhibitor isolated from the Fijian marine sponge
enzymes produced by these cultures on the antifungal activity Aplysinella rhax. Bioorg Med Chem 10:1123–1128
of amphotericin B. Microbiol 43:17–30 Taira T, Ohnuma T, Yamagami T, ASO Y, Ishiguro M, Ishihara M
Park JK, Morita K, Fukumoto I, Yamasaki Y, Nakagawa T, (2002) Antifungal activity of rye (Secale cereale) seed
Kawamukai M, Matsuda H (1997) Purification and character- chitinases: the different binding manner of class I and class II
ization of the Chitinase (ChiA) from Enterobacter sp. G-1. chitinases to the fungal cell wall. Biosci Biotechnol Biochem
Biosci Biotechnol Biochem 61:684–689 66:970–977
Patil SR, Ghormade V, Deshpande MV (2000) Chitinolytic Takayanagi T, Ajisaka K, Takiguchi Y, Shimahara K (1991) Isolation
enzymes: an exploration. Enzyme Microb Technol 26:473–483 and characterization of thermostable chitinases from Bacillus
Peberdy JF (1985) Mycolytic enzymes. In: Peberdy JF, Ferenczy L licheniformis X-74. Biochim Biophys Acta 1078:404–410
(eds) Fungal protoplasts—applications in biochemistry and Tang Y, Zhao J, Ding S, Liu S, Yang Z (2001) Purification and
genetics. Marcel Dekker, New York, pp 31–44 properties of chitinase from Enterobacter aerogenes. Wei
Pichyangkura R, Kudan S, Kultiyawong K, Sukwattanasinitt M, Sheng Wu Xue Bao 41:82–86
Aiba SI (2002) Quantitative production of 2-acetamido-2- Terayama H, Takahashi S, Kuzuhara H (1993) Large scale
deoxy-D-glucose from crystalline chitin by bacterial chitinase. preparation of N,N′-diacetylchitobiose by enzymatic degrada-
Carbohydr Res 337:557–559 tion of chitin and its chemical modification. J Carbohydr Chem
Pope AMS, Davis DAL (1979) The influence of carbohydrates on 12:81–93
the growth of fungal pathogens in vitro and in vivo. Postgrad Tews I, Scheltinga AC, Terwisscha V, Perrakis A, Wilson KS,
Med J 55:674–676 Dijkstra BW (1997) Substrate assisted catalysis unifies two
Revah-Moiseev S, Carrod PA (1981) Conversion of the enzymatic families of chitinolytic enzymes. J Am Chem Soc 119:
hydrolysate of shellfish waste chitin to single cell protein. 7954–7959
Biotechnol Bioeng 23:1067–1078 Tsujibo H, Okamoto T, Hatano N, Miyamoto K, Watanabe T,
Roberts RL, Cabib E (1982) Serratia marcescens chitinase: one step Mitsutomi M, Inamori Y (2001) Family 19 chitinases from
purification and use for the determination of chitin. Anal Streptomyces thermoviolaceus OPC-520: molecular cloning
Biochem 127:402–412 and characterization. Biosci Biotechnol Biochem 64:
Sahai AS, Manocha MS (1993) Chitinases of fungi and plants: their 2445–2453
involvement in morphogenesis and host parasite interaction. Ueda M, Fujiwara A, Kawaguchi T, Arai M (1995) Purification and
FEMS Microbiol Rev 11:317–338 some properties of six chitinases from Aeromonas sp. No. 10S-
Saito A, Fujii T, Yoneyama T, Redenbach M, Ohno T, Watanabe T, 24. Biosci Biotechnol Biochem 59:2162–2164
Miyashita K (1999) High multiplicity of chitinase genes in Usui T, Matsui H, Isobe K (1990) Enzymic synthesis of chito-
Streptomyces coelicolor A3(2). Biosci Biotechnol Biochem oligosaccharides utilizing transglycosylation by chitinolytic
63:710–718 enzymes in a buffer containing ammonium sulfate. Carbohydr
Sakai K, Uchiyama T, Matahira Y, Nanjo F (1991) Immobilization Res 203:65–77
of chitinolytic enzymes and continuous production of N-acetyl Vaidya RJ, Shah IM, Vyas PR, Chhatpar HS (2001) Production of
D-glucosamine with the immobilized enzymes. J Ferment chitinase and its optimization from a novel isolate Alcaligenes
Bioeng 72:168–172 xylosoxydans: potential antifungal biocontrol. World J Microbiol
Sakuda S, Isogai A, Matsumoto S, Suzuki A (1987) Search for Biotechnol 1:62–69
microbial insect growth regulators. II. Allosamidin, a novel Vaidya R, Roy S, Macmil S, Gandhi S, Vyas P, Chhatpar HS (2003)
insect chitinase inhibitor. J Antibiot 40:296–300 Purification and characterization of chitinase from Alcaligenes
Sampson MN, Gooday GW (1998) Involvement of chitinases of xylosoxydans. Biotechnol Lett 25:715–717
Bacillus thuringiensis during pathogenesis in insects. Microbiol Villagomez-Castro JC, Lopez-Romero E (1996) Identification and
144:2189–2194 partial characterization of three chitinases from Entamoeba
Sashiwa H, Fujishima S, Yamano N, Kawasaki N, Nakayama A, invadans with emphasis on their inhibition by allosamidin.
Muraki E, Hiraga K, Oda K, Aiba S (2002) Purification of N- Antonie Van Leeuwenhoek 70:41–48
acetyl D-glucosamine from alpha-chitin by crude enzymes Vyas PR, Deshpande MV (1991) Enzymatic hydrolysis of chitin by
from Aeromonas hydrophila H-2330. Carbohydr Res 337: Myrothecium verrucaria chitinase complex and its utilization to
761–763 produce SCP. J Gen Appl Microbiol 37:267–275
782
Wang SL, Chang WT (1997) Purification and characterization of Wen CM, Tseng CS, Cheng CY, Li YK (2002) Purification,
two bifunctional chitinases/lysozymes extracellularly produced characterization and cloning of a chitinase from Bacillus sp.
by Pseudomonas aeruginosa K-187 in shrimp and crab shell NCTU2. Biotechnol Appl Biochem 35:213–219
powder medium. Appl Environ Microbiol 63:380–386 Yamanoi T, Tsutsumida M, Oda Y, Akaike E, Osumi K,
Wang SL, Yen YH, Tsiao WJ, Chang WT, Wang CL (2002) Yamamamoto K, Fujita K (2004) Transglycosylation reaction
Production of antimicrobial compounds by Monascus purpur- of Mucor hiemalis endo-β-N-acetylglucosaminidase using
eus CCRC31499 using shrimp and crab shell powder as a sugar derivatives modified at C-1 or C-2 as oligosaccharide
carbon source. Enzyme Microb Technol 31:337–344 acceptors. Carbohydr Res 339:1403–1406
Watanabe T, Kobori K, Miyashita K, Fujii T, Sakai H, Uschida M, Yuli PE, Suhartono MT, Rukayadi Y, Hwang JK, Pyun YR (2004)
Tanaka H (1993) Identification of glutamic acid 204 and Characteristics of thermostable chitinase enzymes from the
aspartic acid 200 in chitinase A1 of Bacillus circulans WL-12 Indonesian Bacillus sp. 13.26. Enzyme Microb Technol
as essential residues for chitinase activity. J Biol Chem 35:147–153
268:18567–18572

You might also like