+ All Categories
Home > Documents > From bacteria to human: A journey into the world of chitinases

From bacteria to human: A journey into the world of chitinases

Date post: 20-Dec-2016
Category:
Upload: mohammad-ali
View: 225 times
Download: 4 times
Share this document with a friend
10
UNCORRECTED PROOF 1 Research review paper Q2 From bacteria to human: A journey into the world of chitinases Sina Q1 Adrangi a , Mohammad Ali Faramarzi b, 4 a Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran 5 b Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 14176, Iran 6 7 abstract article info 8 Article history: 9 Received 7 August 2012 10 Received in revised form 26 September 2013 11 Accepted 28 September 2013 12 Available online xxxx 13 14 15 16 Keywords: 17 Chitinase 18 Chi-lectin 19 Gene evolution 20 Biocontrol 21 Biomarker 22 Chitinases, the enzymes responsible for the biological degradation of chitin, are found in a wide range of 23 organisms from bacteria to higher plants and animals. They participate in numerous physiological processes 24 such as nutrition, parasitism, morphogenesis and immunity. Many organisms, in addition to chitinases, produce 25 inactive chitinase-like lectins that despite lacking enzymatic activity are involved in several regulatory functions. 26 Most known chitinases belong to families 18 and 19 of glycosyl hydrolases, however a few chitinases that belong 27 to families 23 and 48 have also been identied in recent years. In this review, different aspects of chitinases and 28 chi-lectins from bacteria, fungi, insects, plants and mammals are discussed. 29 © 2013 Published by Elsevier Inc. 30 31 32 33 34 35 Contents 36 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 37 2. Classication, structure and catalytic mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 38 3. Bacterial chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 39 4. Fungal chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 40 5. Insect chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 41 6. Plant chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 42 7. Mammalian chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 43 8. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 44 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 45 46 1. Introduction 47 Chitin is the second most abundant natural carbohydrate polymer 48 after cellulose and consists of β-(1 4)-linked units of N- 49 acetylglucosamine (GlcNAc). It is a major component of fungal cell 50 walls and invertebrate exoskeletons. In nature, chitin occurs in two 51 different crystalline forms, α and β (Aam et al., 2010). α-Chitin is the 52 dominant form and it is composed of linear chains of GlcNAc arranged 53 in an antiparallel manner. On the other hand, β-chitin consists of parallel 54 chains. Chitinolytic enzymes are produced by a wide range of organisms 55 including bacteria, fungi, insects, plants and animals for different 56 purposes such as nutrition, morphogenesis, and defense against chitin- 57 containing pathogens (Adrangi et al., 2010). Many of theses organisms 58 possess several genes that encode chitinolytic enzymes. For example, 59 most lamentous fungi have 10 to 20 different chitinolytic genes, 60 while in mycoparasitic species the number of such genes may reach 61 30 or even higher (Hartl et al., 2012). These enzymes act in a synergetic 62 or successive manner to degrade chitin (Patil et al., 2000). Higher 63 organisms such as Arabidopsis have also been reported to contain a 64 large number of chitinolytic genes (Hossain et al., 2010). However, not all of these gene codes are Q3 for active enzymes. Many organisms 66 including plants, invertebrates and higher animals express genes 67 encoding so called chitinase-like lectins (chi-lectins) that are devoid of 68 chitinolytic activity due to substitutions in their key catalytic residues 69 (Arakane and Muthukrishnan, 2010; Hossain et al., 2010; Vega and Biotechnology Advances xxx (2013) xxxxxx Abbreviations: AMCase, acidic mammalian chitinase; CBM, carbohydrate-binding module; GH, glycosyl hydrolase; GlcNAc, N-acetylglucosamine; IAD, innovation, amplica- tion, and divergence; ISP, ice structuring protein; MDN, mutation during non-functionality; PR, pathogenesis-related. Corresponding author. Tel./fax: +98 21 66954712. E-mail address: [email protected] (M.A. Faramarzi). JBA-06743; No of Pages 10 0734-9750/$ see front matter © 2013 Published by Elsevier Inc. http://dx.doi.org/10.1016/j.biotechadv.2013.09.012 Contents lists available at ScienceDirect Biotechnology Advances journal homepage: www.elsevier.com/locate/biotechadv Please cite this article as: Adrangi S, Faramarzi MA, From bacteria to human: A journey into the world of chitinases, Biotechnol Adv (2013), http:// dx.doi.org/10.1016/j.biotechadv.2013.09.012
Transcript
Page 1: From bacteria to human: A journey into the world of chitinases

1

2Q2

3Q1

45

6

789101112131415161718192021

32

33

3435

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

Biotechnology Advances xxx (2013) xxx–xxx

JBA-06743; No of Pages 10

Contents lists available at ScienceDirect

Biotechnology Advances

j ourna l homepage: www.e lsev ie r .com/ locate /b iotechadv

Research review paper

From bacteria to human: A journey into the world of chitinases

F

Sina Adrangi a, Mohammad Ali Faramarzi b,⁎a Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iranb Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 14176, Iran

O

Abbreviations: AMCase, acidic mammalian chitinasmodule; GH, glycosyl hydrolase; GlcNAc,N-acetylglucosamtion, anddivergence; ISP, ice structuringprotein;MDN,muPR, pathogenesis-related.⁎ Corresponding author. Tel./fax: +98 21 66954712.

E-mail address: [email protected] (M.A. Faramarzi)

0734-9750/$ – see front matter © 2013 Published by Elsehttp://dx.doi.org/10.1016/j.biotechadv.2013.09.012

Please cite this article as: Adrangi S, Faramarzdx.doi.org/10.1016/j.biotechadv.2013.09.012

O

a b s t r a c t

a r t i c l e i n f o

D

22

23

24

25

26

27

28

29

Article history:Received 7 August 2012Received in revised form 26 September 2013Accepted 28 September 2013Available online xxxx

Keywords:ChitinaseChi-lectinGene evolutionBiocontrolBiomarker

PROChitinases, the enzymes responsible for the biological degradation of chitin, are found in a wide range of

organisms from bacteria to higher plants and animals. They participate in numerous physiological processessuch as nutrition, parasitism, morphogenesis and immunity. Many organisms, in addition to chitinases, produceinactive chitinase-like lectins that despite lacking enzymatic activity are involved in several regulatory functions.Most known chitinases belong to families 18 and 19 of glycosyl hydrolases, however a few chitinases that belongto families 23 and 48 have also been identified in recent years. In this review, different aspects of chitinases andchi-lectins from bacteria, fungi, insects, plants and mammals are discussed.

© 2013 Published by Elsevier Inc.

3031

E

Contents

RRECT

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 02. Classification, structure and catalytic mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03. Bacterial chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 04. Fungal chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 05. Insect chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 06. Plant chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 07. Mammalian chitinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 08. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

54

55

56

57

58

59

60

61

62

UNC1. Introduction

Chitin is the second most abundant natural carbohydrate polymerafter cellulose and consists of β-(1 → 4)-linked units of N-acetylglucosamine (GlcNAc). It is a major component of fungal cellwalls and invertebrate exoskeletons. In nature, chitin occurs in twodifferent crystalline forms, α and β (Aam et al., 2010). α-Chitin is thedominant form and it is composed of linear chains of GlcNAc arrangedin anantiparallelmanner. On theother hand,β-chitin consists of parallel

63

64

65Q3

66

67

68

69

e; CBM, carbohydrate-bindingine; IAD, innovation, amplifica-tationduringnon-functionality;

.

vier Inc.

iMA, From bacteria to human

chains. Chitinolytic enzymes are produced by awide range of organismsincluding bacteria, fungi, insects, plants and animals for differentpurposes such as nutrition, morphogenesis, and defense against chitin-containing pathogens (Adrangi et al., 2010). Many of theses organismspossess several genes that encode chitinolytic enzymes. For example,most filamentous fungi have 10 to 20 different chitinolytic genes,while in mycoparasitic species the number of such genes may reach30 or even higher (Hartl et al., 2012). These enzymes act in a synergeticor successive manner to degrade chitin (Patil et al., 2000). Higherorganisms such as Arabidopsis have also been reported to contain alarge number of chitinolytic genes (Hossain et al., 2010). However, notall of these gene codes are for active enzymes. Many organismsincluding plants, invertebrates and higher animals express genesencoding so called chitinase-like lectins (chi-lectins) that are devoid ofchitinolytic activity due to substitutions in their key catalytic residues(Arakane and Muthukrishnan, 2010; Hossain et al., 2010; Vega and

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 2: From bacteria to human: A journey into the world of chitinases

T

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

t1:1

t1:2

t1:3

t1:4

t1:5

t1:6

t1:7

t1:8

t1:9t1:10

t1:11

t1:12

t1:13

t1:14

2 S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

CO

RREC

Kalkum, 2012). Despite lacking catalytic activity, these proteins retainthe capacity to bind chitin. Owing to their widespread presence anddiverse biological functions, chitinolytic enzymes have found severalapplications. For example they can be used in the production ofsingle-cell proteins, isolation of fungal protoplasts, estimation offungal biomass, development of 3D cell culture scaffolds, biocontrolof plant-pathogenic fungi and insect vectors, and the production ofchitooligosaccharides, glucosamine, GlcNAc, neoglycoproteins and arti-ficial polysaccharides (Adrangi et al., 2010; Jamialahmadi et al., 2011; Liet al., 2008; Lu et al., 2012; Ortiz-Rodriguez et al., 2010; Tajdini et al.,2010; Zakariassen et al., 2011). Recent studies in the field of chitinolyticenzymes have demonstrated that both the diversity and the physiolog-ical roles of these enzymes are far beyond those previously recognized.

2. Classification, structure and catalytic mechanism

Based on theirmode of action, chitinolytic enzymes are classified intotwo categories: chitinases (EC 3.2.1.14) that cleave the chitin chain atinternal sites in a random manner, and β-N-acetylhexosaminidases(EC 3.2.1.52) that catalyze the successive removal of GlcNAc residuesfrom the non-reducing end of the chain (Adrangi et al., 2010). Chitinasesoccur in families 18, 19, 23, and 48 of glycosyl hydrolases (GH), whileβ-N-acetylhexosaminidases are included in GH3, GH18, GH20, andGH84 (Table 1). The classification of GH families is based on sequencehomology and a continuously updated list of these families is availablethrough the CAZy database (Cantarel et al., 2009). The catalytic domainsof members of each GH family fold into a common three-dimensionalstructure (Fig. 1). Families GH18, GH20, and GH84 all have similar(β/α)8 barrel domains (Sumida et al., 2011). On the other hand, GH19andGH23 enzymes adopt anα+β structure, while the catalytic domainof GH3 enzymes has a bipartite structure comprising a (β/α)8 barrelfollowed by an (α/β)6 sandwich (Wohlkonig et al., 2010; Yoshidaet al., 2010). Some GH3 enzymes, however, lack the (α/β)6 sandwich(Yoshida et al., 2010). Finally, GH48 enzymes have an (α/α)6 barrelstructure characterized by six centralα-helices surrounded by six exter-nalα-helices (Yennamalli et al., 2011). Most chitinases aremodular pro-teins that, in addition to a catalytic domain, contain auxiliary domainssuch as the carbohydrate-binding module (CBM) (Guillen et al., 2010).The CBM enhances the activity of the enzyme towards insoluble sub-strates by anchoring the enzyme to the substrate and disrupting thecrystalline structure of the substrate resulting in the formation of freechain ends (Guillen et al., 2010; Vaaje-Kolstad et al., 2005). Like catalyticmodules, CBMs are classified into families of homologous proteinswhichmay be accessed at the CAZy database. A second feature that helps toovercome the low accessibility of insoluble substrates is the presenceof a deep and narrow substrate-binding cleft lined with aromaticresidues in some chitinolytic enzymes (Fig. 2) (Horn et al., 2006a,b;Zakariassen et al., 2010). Such enzymes act in a processive manner,meaning once attached to a substrate chain, they thread the chainthrough their catalytic cleft performing several hydrolytic cuts insteadof releasing the substrate after each cleavage. The aromatic residues

UN

Table 1Characteristics of chitinolytic enzymes belonging to different GH families.

Family Catalytic mechanism Proton donor Base/nucleoph

ChitinasesGH18 Retaining Glu Substrate acetGH19 Inverting Glu Glu/aspGH23 Inverting Glu Not known

GH48 Inverting Glu Not known

β-N-acetylhexosaminidasesGH3 Retaining Glu AspGH18 Retaining Glu Substrate acetGH20 Retaining Glu Substrate acetGH84 Retaining Asp Substrate acet

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

provide the necessary environment for the flexible binding and move-ment of the substrate through the active site. Although processivitycomprises an efficient strategy for the hydrolysis of insoluble substrates,it is usually associated with reduced activity towards soluble or moreaccessible polymeric substrates (Aam et al., 2010).

Like other glycosyl hydrolases, chitinolytic enzymes generally cata-lyze the depolymerization of their substrate through one of the twopathways known as single- and double-displacement mechanisms(Aam et al., 2010; Andersen et al., 2005; Cantarel et al., 2009; Li andGreene, 2010; Slamova et al., 2010; Tang et al., 2004; Udaya Prakashet al., 2010). In both pathways, two distinct catalytic groups areinvolved. One of these is a carboxyl group that acts as a proton donorand is usually provided by a conserved glutamate residue at the activesite of the enzyme, although in some cases, for example in familyGH84, an aspartate residue may fulfill this role (Table 1). The secondcatalytic group may act either as a base (as in the single-displacementmechanism) or a nucleophile (as in the double-displacement mecha-nism). This group may be a carboxyl moiety provided by a conservedglutamate or aspartate residue, or it may be the N-acetyl group of thesugar positioned in the −1 subsite of the enzyme (Aam et al., 2010;Udaya Prakash et al., 2010). Subsites are numbered from –n to +n,where negative sign represents the non-reducing end of the chain withcleavage occurring between the −1 and +1 subsites (Davies et al.,1997). Since the single-displacementmechanism results in the inversionof the anomeric configuration of the hydrolyzedGlcNAc residue, it is alsoknown as the inverting mechanism. On the other hand, in the double-displacement mechanism, also referred to as the retaining mechanism,the anomeric configuration is retained.

3. Bacterial chitinases

Bacterial chitinases occur in families GH18, GH19, andGH23 (Dahiyaet al., 2006; Udaya Prakash et al., 2010; Ueda et al., 2009).Most bacterialchitinases belong to the GH18 family (Larsen et al., 2011) (Fig. 3). Basedon sequence homology, bacterial GH18 chitinases are classified intothree subfamilies A, B and C (Li and Greene, 2010). It should be notedthat the nomenclature of bacterial chitinases does not follow this classi-fication; for example, chitinase B from Serratia marcescens belongs tosubfamily Awhile Bacillus circulans chitinase D is classified in subfamilyB (Watanabe et al., 1999). Some bacterial GH18 chitinases besidescatalytic and CBM domains contain a fibronectin type III-like domainwhich plays a role in substrate binding (Horn et al., 2006a). On theother hand, the distribution of GH19 chitinases among bacteria appearsto be restricted to actinobacteria and purple bacteria (Udaya Prakashet al., 2010). It has been proposed that actinobacteria and purple bacte-ria may have acquired GH19 chitinase genes from plants and, in turn,transferred them to arthropods and nematodes (Udaya Prakash et al.,2010). To date, only one GH23 chitinase has been identified in bacteria(Ueda et al., 2009). This enzyme was isolated from Ralstonia sp.A-471 and comprises an N-terminal chitin-binding domain linked toa C-terminal catalytic domain that shows homology to goose-type

ile Structure Reference

amido group (β/α)8 CAZY; Sumida et al. (2011)α+ β CAZY; Wohlkonig et al. (2010)α+ β CAZY; Wohlkonig et al. (2010);

Arimori et al. (2013)(α/α)6 CAZY; Yennamalli et al. (2011)

(β/α)8+ (α/β)6 CAZY; Yoshida et al. (2010)amido group (β/α)8 CAZY; Sumida et al. (2011)amido group (β/α)8 CAZYamido group (β/α)8 CAZY

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 3: From bacteria to human: A journey into the world of chitinases

T

F

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225Q4

Fig. 1. Three-dimensional structure of representative GH enzymes. (A) A GH18 chitinase from Aspergillus fumigatus showing a (β/α)8 barrel (PDB ID: 2XVP). (B) A GH19 chitinase fromCarica papaya showing α+ β structure (PDB ID: 3CQL). In most cases, like this example, the β regions are actually composed of isolated bridges rather than extended strands (residuesshown in stick representation). (C) A GH3 glucohydrolase from Hordeum vulgare showing a bipartite domain consisting of a (β/α)8 barrel (lower right) and an (α/β)6 sandwich (upperleft) (PDB ID: 1IEQ). (D) AGH48 cellobiohydrolase from Clostridium thermocellum showing an (α/α)6 barrel (PDB ID: 1L1Y). All figureswere prepared using VMD (Humphrey et al., 1996).

3S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

RREC

lysozyme. The gene encoding this enzyme also appears to be acquiredvia horizontal gene transfer (Ueda et al., 2009).

Bacteria produce chitinases for different purposes such as nutritionand parasitism (Dahiya et al., 2006; Faramarzi et al., 2009). For somespecies, chitinases may play an essential role in providing the bacteriumwith required nutrients or precursors. The spirochete Borrelia burgdorferi,for example, is unable to produce GlcNAc required for cell wall synthesisand in an environment such as the tick midgut where free GlcNAc is notavailable, the ability to degrade the chitin-rich peritrophic membranewould allow the bacterium to obtain the necessary GlcNAc (Rhodeset al., 2010). It has also been shown that the chitinase system ofPseudoalteromonas is involved in nitrogen metabolism (Delpin andGoodman, 2009). However, for many other heterotrophic bacteriachitinases are probably not crucial in this respect since in most habitatsdifferent nutrient sources are available (Cottrell et al., 2000). On theother hand, autotrophic microorganisms such as the cyanobacteriumAnabaena probably produces chitinolytic enzymes as part of their allelo-pathic system to inhibit the growth of competitor organisms such asfungi (Prasanna et al., 2010). Chitinases are also involved in bacterialpathogenesis. In cases where the host is known to contain chitin, suchas in insects, the mechanism seems straightforward. For example, theentomopathogenic bacterium Yersinia entomophaga produces an ABC-type protein toxin complex that can kill the host within 72h of infection(Busby et al., 2012; Landsberg et al., 2011). It has been shown that thiscomplex includes two subunits with chitinase activity that anchor thecomplex to, and facilitate its penetration through the peritrophicmembrane (Busby et al., 2012). Vector-borne bacterial pathogens ofnon-chitinous organisms such as mammals and plants also producechitinases to colonize the insect vector by digesting the peritrophic

UNCO

Fig. 2. Substrate-binding cleft of processive and non-processive chitinases. (A) A processive chitbrasiliensis (PDB ID: 1KQY). Cocrystallized substrates are shown in stick representation.

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

Omembrane (Chandler et al., 2011; Killiny et al., 2010). However, evenin non-chitinous hosts, chitinases may directly contribute to bacterialvirulence, probably by binding to alternative substrates such as thecarbohydrate moieties of glycoproteins involved in the host's immuneresponse (Chaudhuri et al., 2010). In the human pathogen Listeriamonocytogenes, the virulence gene regulator PrfA induces, among othergenes, chitinase expression indicating that the biological functions ofthis enzyme are not limited to the external environment (Larsen et al.,2010). Chitinase production is also stimulated by the quorum-sensingLuxR–LuxI homologous system that is involved in virulence, biofilmformation and surface motility in some pathogens such as Pseudomonasaeruginosa and Chromobacterium violaceum (Alipour et al., 2010; Stauffand Bassler, 2011). Clostridium difficile produces a bifunctional proteinwith chitinase and peroxiredoxin activities that appears to be partlyresponsible for the symptoms of C. difficile infection (Permpoonpattanaet al., 2011).

Bacteria also produce chitin-binding proteins comprised exclusivelyof non-catalytic CBMs. For example, S. marcescens secretes a proteinnamed CBP21 that belongs to CBM family 33 and seems to be involvedin chitin digestion despite lacking hydrolytic activity (Vaaje-Kolstadet al., 2005). CBP21 probably promotes the degradation of insolublechitin via non-enzymatic disruption of the crystalline structure of thesubstrate. Nevertheless, recent studies suggest that CBP21may actuallypossess oxidoreductase activity (Tran et al., 2011; Vaaje-Kolstad et al.,2010). The majority of bacterial CBM proteins belong to the CBM33family while their eukaryotic counterparts mostly belong to CBM fami-lies 14 and 18 (Purushotham et al., 2012; Tran et al., 2011).

It has been shown that some bacteria such as Streptomyces andBacillus licheniformis produce different chitinase inhibitors; however

inase from Serratia marcescens (PDB ID: 1E6N). (B) A non-processive chitinase from Hevea

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 4: From bacteria to human: A journey into the world of chitinases

CTED P

RO

OF

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

Fig. 3.A cladogramshowing the relationship of selected chitinases fromall bacterial genera representedwith at least oneGH18 chitinase sequence in theNCBI RefSeq database. Chitinase Dfrom Bacillus circulans (Swiss-Prot Accession: P27050) was used as query for a BLAST search against the RefSeq database. A total number of 439 chitinase sequences from 68 bacterialgenera were obtained (E value b 0.01) and aligned using MEGA5 (Tamura et al., 2011). For each genus, the entry showing the highest degree of similarity to the query sequence wasselected and included in the cladogram. It should be noted that most of these genera are represented in RefSeq with several species and each species with more than one chitinase;as a result, depending on the selected sequences, the topology of the resulting tree may vary.

4 S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

UNCO

RREthe biological functions of these inhibitors are not completely clarified

(Kumar and Rao, 2010; Suzuki et al., 2008). Allosamidin is apseudotrisaccharide produced by some Streptomyces species thatmimics the structure of chitin and competitively inhibits GH18chitinases. Although allosamidin can suppress chitinase activity inallosamidin-nonproducer strains, it is not secreted to the medium bythe producer strain and rather tends to accumulate in the myceliawhere it probably acts as a signal molecule for chitinase production(Suzuki et al., 2008). The peptidic aspartic protease inhibitor ofB. licheniformis, on the other hand, is secreted to the medium andshows a non-competitive inhibition pattern (Kumar and Rao, 2010).

4. Fungal chitinases

Fungal chitinases are members of the GH18 family (Hartl et al.,2012). They can be further divided into three subgroups, namely A, B,and C (not to be confused with bacterial GH18 chitinase subfamilies).This classification scheme primarily makes use of sequence similaritybut other distinctive features have also been described (Seidl, 2008).Subgroup A chitinases contain a single catalytic domain and no CBMs.These are processive enzymes with a deep substrate-binding cleft. Sub-group B chitinases are non-processive and in addition to their catalyticdomain comprise either a C-terminal CBM or an unstructured serine/threonine-rich domain. Subgroup C chitinases consist of an N-terminalCBM linked to a C-terminal catalytic domain. Like subgroup A, subgroupC chitinases have a deep narrow substrate-binding cleft that indicatesa processive nature. An interesting feature of subgroup C chitinases isthe presence of several LysM (lysin motif; also known as CBM50)domains (Hartl et al., 2012). The LysM domain was first identified in

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

bacteriophage and bacterial cell wall degrading enzymes as a substrate(peptidoglycan) binding domain (Bateman and Bycroft, 2000). It waslater shown to bind other glycans such as chitooligosaccharides aswell (Buist et al., 2008). In plants, specific LysM-containing cell surfacereceptors (known as LysM-RLK) are involved in the perception of rhizo-bial nodulation (Nod) factors (Radutoiu et al., 2003). Nod factors arelipochitooligosaccharide signaling molecules secreted by Rhizobiumbacteria that induce the formation of symbiotic root nodules in legumi-nous plants (Knogge and Scheel, 2006). Since these molecules aresubjected to species-specific substitutions, they are believed to play amajor role in the specific recognition of Rhizobium species by the hostplants (Gressent et al., 2002; Streng et al., 2011). In fact, it has beenshown that the heterologous expression of the enzymes involved inthe acylation of Nod factors may be used to extend the host range ofRhizobium bacteria (Pacios Bras et al., 2000). Plant LysM-RLKs alsoplay a role in defense against fungal pathogens (Brotman et al., 2012;Petutschnig et al., 2010). Based on thesefindings, aswell as the observa-tion that subgroup C chitinases are present at higher numbers inmycoparasitic fungi, it was hypothesized that it may be possible tocategorize fungal chitinases as self- and non-self degrading enzymes(Gruber and Seidl-Seiboth, 2012). However, detailed analysis of theexpression profile of subgroup C chitinases from several mycoparasiticTrichoderma species revealed that these enzymes are not expressed ina mycoparasitism-specific manner suggesting that they are involved inthe degradation of both self and non-self cell walls (Gruber and Seidl-Seiboth, 2012; Gruber et al., 2011a,b). The protection of the fungus'sown cell wall is probably achieved by limiting the accessibility ofchitin by cell wall proteins rather than the speciation of individualchitinases (Gruber and Seidl-Seiboth, 2012). In situations where self

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 5: From bacteria to human: A journey into the world of chitinases

T

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

5S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

EC

cell wall degradation is required, e.g. in hyphal branching, a localizedde-protection of chitin occurs. In addition to mycoparasitic fungi, non-mycoparasitic fungi have also been shown to secret proteins withchitinolytic activity in order to inhibit the growth of competitive fungi.For example, the α subunit of Kluyveromyces lactis killer toxin, whichis essential for the antifungal activity of the toxin, is actually achitinolytic enzyme (Colussi et al., 2005).

In nature, entomopathogenic fungi play an important role in control-ling insect pests, especially those that are resistant to viral and bacterialinfections (Fang et al., 2012; Schrank and Vainstein, 2010). Sincechitinases are an important part of the enzymatic arsenal of thesefungi, improved strains overexpressing native or engineered chitinaseshave been produced and demonstrated to exhibit increased virulenceagainst insects (Fan et al., 2007; Fang et al., 2005). Such improvedstrains can potentially be used in the biocontrol of pest and diseasevector insects (Fang et al., 2012; Jami Al Ahmadi et al., 2008;Tasharrofi et al., 2011). An alternative approach is to use fungal orbacterial chitinases directly as biopesticides (Binod et al., 2007; Kimand Je, 2010; Martinez et al., 2012). The insecticidal activity of suchpreparations may be improved by the addition of adjuvants such aspolyoxyethylene-(3)-isotridecyl ether that promote the penetration ofthe enzymes through epicuticle (Kim et al., 2010). In practice, however,the applicability of this method is limited by several factors includingthe rapid evaporation of the aqueous phase especially under dryingconditions. Nematode-pathogenic fungi such as Clonostachys roseathat produce several proteases and chitinases to target eggs and larvaehave also been exploited as biological control agents to suppressplant or even animal parasitic nematodes (Baloyi et al., 2012; Zouet al., 2010).

Invasive fungal infections are amajor cause of mortality in immuno-suppressed patients mostly due to the fact that current diagnosticmethods usually fail to detect fungal infections at early stages (Vegaand Kalkum, 2012). It has been proposed that radiolabeled chitinasesand chitin-binding proteins may be used for the early detectionof such infections (Lupetti et al., 2011). Other methods that detectthe host responses to fungal infections such as the use of immuno-fluorescence stains for chitinase have also been investigated (Guoet al., 2012). Nevertheless, further studies are needed before theseapproaches are ready for clinical use.

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

UNCO

RR

5. Insect chitinases

With the exception of a GH48 chitinase identified in the leaf beetleGastrophysa atrocyanea (Fujita et al., 2006), all known insect chitinasesbelong to the family GH18 (J. Zhang et al., 2011). Based on sequencehomology and domain architecture, GH18 insect chitinases have beenassigned into eight distinct groups denoted by Roman numerals I–VIII(Arakane and Muthukrishnan, 2010; H. Zhang et al., 2011). Group Iare characterized by the presence of an N-terminal catalytic domainjoined to a CBM14 domain via a serine/threonine-rich linker. The linkerregion serves as a site for glycosylation which increases the stability ofthe enzyme against proteases. Group II are much larger proteins with4–5 catalytic domains and 4–7 CBMs. Some catalytic domains of groupII chitinases seem to be enzymatically inactive as their catalytic asparticacid residue is replaced by other residues such as histidine or serine(Arakane and Muthukrishnan, 2010). Group III chitinases consist oftwo catalytic domains and a single CBM. Groups IV, V, VII and VIII havea single catalytic domain and usually no CBM, and are distinguishedfrom each other by sequence homology. Group V includes chitinase-like proteins such as the imaginal disk growth factors (IDGFs) that lackchitinase activity and are involved in growth regulation and immunity.Group VI chitinases have one catalytic domain, one CBM and a longC-terminal stretch with a high percentage of serine and threonineresidues. Other features such as transmembrane and signal regionsmay also be present in all groups.

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

Insect chitinases play an important role in chitin turnover especiallyduring molting. However, their expression should be tightly controlledsince premature exposure can lead to growth inhibition and mortality(H. Zhang et al., 2011). To achieve such a control, insects produce severalfunctionally specialized chitinases that are differentially expressedthrough the various stages of metamorphosis (Zhu et al., 2008). Atleast some of these enzymes are regulated by hormonal control(Merzendorfer and Zimoch, 2003; Royer et al., 2002; Takahashi et al.,2002; Zheng et al., 2003). For example, in Tribolium castaneum, TcCHT5is required for pupal–adult molting only while TcCHT10 is involved inlarval–larval, larval–pupal and pupal–adult molting (Zhu et al., 2008).Since during a normal molting cycle the synthesis of the new cuticleand the degradation of the old cuticle occur simultaneously, it is alsoimportant to protect the nascent cuticle from chitinases and othermolting enzymes (Chaudhari et al., 2011). This is mainly achievedthrough the action of specific proteins that colocalize and directly inter-act with newly synthesized chitin and organize it into laminae confer-ring resistance to chitinolytic enzymes (Chaudhari et al., 2011; Petkauet al., 2012). Knickkopf is one such protein that shows homology tothe CBM9 family (Chaudhari et al., 2011; Iyer et al., 2007). Thecolocalization of Knickkopf with chitin appears to be controlled by yetanother protein known as Obstructor-A which is encoded by a memberof the obstructor multigene family (Petkau et al., 2012). The obstructorfamily was first identified in Drosophila melanogaster and shown tocode for putative chitin-binding proteins (Behr and Hoch, 2005).Chitinases (and chi-lectins) are also produced by the venomand salivaryglands of some insect species (Arakane and Muthukrishnan, 2010).In endoparasitoid wasps such as Chelonus inanitus and Toxoneuronnigriceps the venom chitinases are probably involved in the degradationof the host cuticle facilitating the egression or ingression of parasitoidlarvae through such barriers (Consoli et al., 2005; Vincent et al., 2010).However, it has been shown that the venom of ectoparasitoid specieslike Nasonia vitripennis also contains chitinases (Danneels et al., 2010).Since in ectoparasitoids no larval egression or ingression occurs, otherfunctions should be ascribed to these enzymes (Danneels et al., 2010).The physiological role of chi-lectins found in the saliva of some insectssuch as the anopheline mosquito is not well understood either. Ithas been proposed that these chi-lectins may be involved in the self-defense mechanism of the anopheline mosquito against malariaparasites through a complex sequence of events (Owhashi et al.,2008). Plasmodium falciparum, the causative parasite of malaria, duringits sporogonic cycle in the midgut of the anopheline vector, produces achitinase that disrupts the peritrophic membrane and allows theparasite to migrate to the salivary glands of the vector (Giansanti et al.,2007). Inhibition of this enzyme results in complete disruption ofsporogonic development (Bhatnagar et al., 2003; Isaacs et al., 2012).Following a mosquito bite, the mosquito saliva proteins includingchi-lectins are introduced to the immune system of the host wherethey induce antibody production. Such anti-chitinase antibodies arefrequently encountered in the sera of residents of malaria-endemicareas (Owhashi et al., 2008). Ingestion of these antibodies from bloodmeals may contribute to the defense mechanism of mosquitoes byinhibiting the chitinolytic activity of malaria parasites (Owhashi et al.,2008). Insect chi-lectins have been shown to contribute to otherprocesses such as growth regulation and immunity as well (Aronsteinet al., 2010; Nakabachi et al., 2010).

6. Plant chitinases

Based on sequence similarity, plant chitinases are classified intoseven classes I–VII (Kasprzewska, 2003; Patil et al., 2009; Sarma et al.,2012). Class I, II, IV, VI, and VII chitinases belong to the GH19 familywhile class III and V chitinases are members of the GH18 family(Ohnuma et al., 2011, 2012). Chitinases comprise an important part ofpathogenesis-related (PR) proteins; a group of proteins includinghydrolytic enzymes, enzyme inhibitors, and membrane-permeabilizing

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 6: From bacteria to human: A journey into the world of chitinases

T

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

6 S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

UNCO

RREC

peptides that are produced by plants in response to invading pathogensand abiotic factors (Ebrahim et al., 2011; Edreva, 2005; Li and Yi, 2012;Sels et al., 2008). It should be noted that although this definition includesabiotic factors, induction by such factors alone is not a sufficient criterionfor categorizing a particular protein as a PR protein (Edreva, 2005). PRproteins show antifungal, antibacterial, insecticidal, nematicidal, andantiviral effects and participate in the systemic acquired resistance(SAR) of plants against a broad range of pathogens. PR proteins arecurrently classified into 17 families with chitinases occurring in familiesPR-3, PR-4, PR-8, and PR-11 (Sels et al., 2008). Considering the impor-tant role of chitinases in plant defense against chitin-containing patho-gens, efforts have been made to produce transgenic plants expressingseveral chitinases or a combination of chitinases and other PR proteinsto increase their resistance against such pathogens and promisingresults have been achieved (Al Ahmadi et al., 2008; Amian et al., 2011;Ramana Rao et al., 2011). An alternative approach is to clone chitinasegenes in symbiotic rhizobacteria such as Burkholderia vietnamiensis(Zhang et al., 2012). There are legitimate concerns about the environ-mental consequences of using such procedures; nevertheless field stud-ies have demonstrated that these methods do not negatively affectthe decomposition dynamics of the plants or the soil fungal biomass(Duc et al., 2011; Stefani et al., 2010).

In addition to their role in defense against pathogens, plantchitinases also serve several other physiological functions that maynot be directly related to their hydrolytic activity. For example, someplant chitinases show ice structuring activity (Goni et al., 2010; Yaishet al., 2006). Ice structuring proteins (ISPs) are a group of proteinsproduced by different organisms including plants that bind ice crystalsand affect their growth and morphology providing cold or freezingtolerance for the organism (Hassas-Roudsari and Goff, 2012). ISPsprobably interactwith crystals through an icebinding surface composedof charge-conserved amino acids located apart from the enzyme'sactive site (Yaish et al., 2006). Plant chitinases are also up-regulatedby certain heavy metals such as lead and cadmium (Cai et al., 2011;Walliwalagedara et al., 2010). This may simply represent a commonstress response, although there is some evidence that chitinases maycounteract oxidative stress (Walliwalagedara et al., 2010). It has alsobeen shown that chitinases can act as calcium storage proteins. Yanget al. (2011) have isolated a class III chitinase from Punica granatumseeds that binds calcium ionswith high capacity. Although the presenceof calcium improves the stability of P. granatum seed chitinase, it hasnegligible effect on its activity. The subcellular localization pattern ofthis enzyme is also in agreement with a role in calcium storage.

Chi-lectins are produced by a broad range of plants and, like activechitinases, appear to be involved in different physiological processes(Van Damme et al., 2007). In Arabidopsis thaliana and Oryza sativachi-lectins play an essential role in cellulose biosynthesis (Sanchez-Rodriguez et al., 2012; Wu et al., 2012). The underlying mechanism ofthis observation is not completely understood but evidence suggeststhat chi-lectins bind to emerging cellulose microfibrils and affect theircrystallinity or the association of crystalline and amorphous regions(Sanchez-Rodriguez et al., 2012). In another study it was demonstratedthat chi-lectins affect root structure architecture in A. thaliana throughsimilar mechanisms (Hermans et al., 2010). On the other hand, studieson fruit ripening in banana have shown that some chi-lectins mayserve as storage proteins to support the synthesis of ripening-associated proteins by providing the required amino acids (Peumanset al., 2002). Plant chi-lectins also showantifungal and insecticidal activ-ity (Vasconcelos et al., 2011;Wasano et al., 2009). It has been proposedthat the antifungal activity of chi-lectins results from their ability toinhibit fungal glycosidases such as xylanases, however other mecha-nisms may also be involved (Vasconcelos et al., 2011). With regard totheir insecticidal activity, chi-lectins probably exert their toxic effect bydisrupting the formation of the peritrophic membrane (Wasano et al.,2009). Plant chi-lectins are up-regulates in response to abiotic stressessuch as drought, heat, and high salinity as well (Kwon et al., 2007).

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

7. Mammalian chitinases

All known mammalian chitinases belong to the GH18 family(Shuhui et al., 2009). The first human chitinase to be identified was achitotriosidase produced by macrophages of Gaucher patients (Bootet al., 1995). Since this enzyme showed antifungal properties, it wasproposed that it may be involved in defense against chitin-containingpathogens. This observation was supported by other studies (Bussinket al., 2006). In contrast to these findings, later studies revealed thatabout 5% of Caucasian populations are completely deficient in activechitotriosidase suggesting that this enzyme may no longer be a directeffector of the innate immune response (Boot et al., 2005). This deficiencyresults from a 24-bp duplication in the CHIT1 gene (the gene encodinghuman chitotriosidase) that leads to aberrant splicing and, consequently,the production of an enzymatically inactive protein. Despite the factthat the relatively high incidence of this polymorphism precludes adirect defensive function for chitotriosidase, this enzyme still appearsto affect the host's defense system through other mechanisms. In arecent study, it was shown that the aforementioned duplication eventis strongly associatedwith an increased rate of decline in the lung func-tion of smokers with chronic obstructive pulmonary disease (COPD)(Aminuddin et al., 2011). This suggests a protective role against rapiddisease progression for human chitotriosidase although the underlyingmechanism still remains to be elucidated. Chitotriosidase may alsoexert some protective effects against atherosclerosis (Kitamoto et al.,2013). A few years after the discovery of chitotriosidase, a secondchitinase named acidic mammalian chitinase (AMCase) was isolatedand shown to be expressed primarily in the gastrointestinal tract andlung of both mouse and human (Boot et al., 2001). Based on its expres-sion profile, a dual function in innate immunity and chitin digestionwasproposed for AMCase (Bussink et al., 2007). Recent studies performedon several bat species suggest that AMCase may actually be involvedin chitin digestion in insectivorous mammals (Strobel et al., 2013).This is probably not the case for humans, however, as AMCase expres-sion level in the human gastrointestinal tract is significantly lowerthan that observed in mouse (Ohno et al., 2013). In the respiratorytract, AMCase also appears to be involved in Th2-mediated inflamma-tion (Kawada et al., 2007). Originally, it was hypothesized that inresponse to Th2 cytokines such as IL-13, airway epithelial cells andmacrophages produce AMCase that in turn induces the production ofseveral chemokines resulting in the recruitment of T cells, eosinophilsand macrophages (Lee, 2009; Shuhui et al., 2009; Zhu et al., 2004).This assumption was based on the observation that neutralizingAMCase with anti-AMCase serum or chitinase inhibitors reducedinflammatory infiltration in mouse asthma models (Shuhui et al.,2009). However, a recent study has shown that AMCase may actthrough altering the Th1/Th2 balance in lungs (Fitz et al., 2012). Oneshould note that some of the physiological functions of AMCase areindependent from its enzymatic activity (Hartl et al., 2009), hence,considering the different nature of the neutralizing agents used inthese studies, it can be assumed that these two proposed mechanismsmay represent two different pathways through which AMCase exertsits effects. AMCase and/or chitotriosidase may also be implicated inconjunctivitis, nasal polyp pathogenesis, adenoid hypertrophy, neuro-myelitis, and gastritis (Bucolo et al., 2011; Correale and Fiol, 2011;Cozzarini et al., 2009; Heo et al., 2011; Park et al., 2011). Both AMCaseand chitotriosidase, in addition to an N-terminal catalytic domain, con-tain a C-terminal CBM that belongs to the CBM14 family (Funkhouserand Aronson, 2007), and the genes coding for both enzymes havebeen identified in all mammals for which complete genome data areavailable (Bussink et al., 2007).

In addition to active chitinolytic enzymes, mammalian genomesalso code for enzymatically inactive chi-lectins (Shuhui et al., 2009). Inhumans three chi-lectins have been identified: human chitinase 3-likeprotein 1 (CHI3L1; also known as YKL-40), human chitinase 3-like pro-tein 2 (CHI3L2; also known asYKL-39), and oviductin (OVGP1) (Bussink

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 7: From bacteria to human: A journey into the world of chitinases

T

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

7S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

UNCO

RREC

et al., 2007; Shuhui et al., 2009). The mouse genome contains at leastseven chi-lectin genes. The physiological function of chi-lectins is notcompletely understood but they appear to be involved in different pro-cesses such as tissue remodeling, fertilization, and innate immunity(Bussink et al., 2006; Vega and Kalkum, 2012). For example, it hasbeen shown that CHI3L1 mediates mammary tissue remodeling duringinvolution possibly by inhibiting epithelial cell differentiation andpolarization and inducing cell motility (Scully et al., 2011). However,elevated levels of CHI3L1 are also associated with pathologic conditionssuch as breast malignancies which are characterized by poor differenti-ation (Scully et al., 2011). In fact, the murine homologue of CHI3L1 wasfirst discovered in breast cancer cells and named breast regressionprotein 39 (BRP-39) (Lee et al., 2009). In Th2-dependent immuneresponses, on the other hand, CHI3L1 appears to be involved in macro-phage and dendritic cell activation and apoptosis prevention (Lee et al.,2009), and thus it is not surprising that increased expression ofchi-lectins has been reported in several inflammatory diseases such asneuromyelitis (Correale and Fiol, 2011), colitis (Aomatsu et al., 2011),COPD (Sakazaki et al., 2011), and hepatitis (Lebensztejn et al., 2007).CHI3L1 appears to exert its effect by binding to interleukin-13 receptorα2 (He et al., 2013). It has been proposed that chi-lectins may be usedas biomarkers to evaluate the stage, prognosis and therapeutic responseof different diseases (Agapov et al., 2009; Roslind and Johansen, 2009).For example, CHI3L1 plasma level may be used to estimate the severityof preeclampsia (Seol et al., 2009), to evaluate the prognosis of chronicheart failure (Bilim et al., 2010), or to monitor the therapeutic responseof psoriasis (Imai et al., 2011). A limitation to this strategy is that inmany cases the plasma concentration of the chi-lectin does not directlycorrelate with the clinical parameters of the disease under investigation(Lebensztejn et al., 2007;Mathiasen et al., 2011). Itmay also be possibleto use chi-lectins as diagnostic biomarkers. However, since chi-lectinsare overexpressed in a wide range of inflammatory and oncogenicpathologies, they are of little diagnostic value when used alone. Thesensitivity and selectivity of diagnosis can be increased by measuringa panel of biomarkers at the same time (Chang et al., 2009). Changet al. (2009) have developed a multiplex proximity ligation assayusing three biomarkers (CHI3L1, osteopontin, and carbohydrate antigen19-9) for the diagnosis of pancreatic cancer. In another study, a similarapproach was used to improve the accuracy of ovarian cancer diagnosis(Fredriksson et al., 2008). Further studies are required before thesemethods can be exploited in clinic.

8. Concluding remarks

Chitinases and/or chi-lectins are ubiquitous proteins that are widelydistributed among all kingdoms of life. These proteins take part in awide range of physiological processes including nutrition, morphogen-esis, pathogenesis, parasitism, growth regulation, and immunity. Inmicroorganisms, they are primarily involved in the metabolism ofchitin, both endogenous (e.g. in fungi) and exogenous (e.g. in bacteria).However, in higher organisms, their regulatory function seems to bedominant. This is especially true for mammals since, as stated inSection 7, preliminary evidence suggests that mammalian chitinasesdo not directly participate in defense against chitinous pathogens. Asimilar argument can be made for plants. Although plant chitinasesare classified as PR proteins, it is not clear whether their increasedexpression in response to infections is a pathogen-specific process orit simply reflects a general stress-induced response in which chitinasesact as signaling intermediates (refer to Section 6). In this context, it istempting to look at chitinases as “living” examples of gene evolution.This idea becomes particularly attractive when we consider thatthe acquisition of (new) regulatory functions by chitinases can beexplained by both the mutation during non-functionality (MDN) andinnovation, amplification, and divergence (IAD) models of gene evolu-tion (Bergthorsson et al., 2007). Both models are based on the assump-tion that new genes always emerge from duplicated genes. The MDN

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

model indicates that gene duplication precedes the acquisition of newfunctions and, in order to prevent the loss of the newly duplicatedgene due to drift or other processes, some selective pressure must bepresent. Severalmechanisms such as the protective effect of redundancy,stabilization by subfunctionalization, and the effect of increased genedosage participate in the maintenance of the duplicated gene, amongwhich the latter may have played an important role in the evolution ofchitinases. As described in Section 1,many organisms such as fungi con-tain several chitinase genes thatmight have provided them competitiveadvantage. Once a duplicated gene has been stabilized, it may, overtime, acquire the rare mutations that provide a new function. On theother hand, according to the IAD model, innovation (acquisition ofminor side activities) occurs before gene duplication. These activitiesare neither beneficial nor detrimental per se, but they may becomevaluable following a change in the surrounding environment that favorsone of these newly acquired functions. In this instance, the newfunctionmay act as a selectable trait. Such non-enzymatic side activitieshave been documented for mammalian and plant chitinases, asdiscussed in Sections 6 and 7. The IAD model can also explain theemergence of chi-lectins. It can be hypothesized that with the elimina-tion of the need for active chitinases, the selective pressure preventing“forbidden mutations” in critical regions such as the catalytic activesite of the enzyme was removed. However, these genes were stillselectively maintained for their (previously) side activities, giving riseto new non-enzymatic proteins with regulatory functions. Other exam-ples of such “dead enzymes” with regulatory functions have also beenidentified (Adrain and Freeman, 2012). Since young proteins evolvemore quickly than old proteins (Vishnoi et al., 2010), it is not surprisingthat the number and diversity of chi-lectin genes in mammaliangenomes appear to be higher than their active counterparts (refer toSection 7).

In recent years, chitinase research has made fast progress especiallyat molecular levels and chitinolytic activity, once considered to belimited to GH18 and GH19 families, and has been identified in otherGH families suggesting that the diversity of chitinases may be greaterthan previously anticipated. In a foreseeable future, with the elucidationof the different mechanisms through which they exert their physiolog-ical effects, chitinases and chi-lectins may found several medical, phar-maceutical, and agricultural applications. As discussed in Section 7,chi-lectins can potentially be used as biomarkers for the diagnosis ofdifferent types of cancers or for the evaluation of the therapeuticresponse and prognosis of several diseases in humans. Chitinase-producing microorganisms, on the other hand, have successfully beenexploited in the treatment of veterinary parasites (Section 4). Thepossibility of the direct application of parenteral chitinase preparationsin the treatment of systemic fungal infections has also been investigatedand promising results have been achieved (van Eijk et al., 2005). Alter-natively, it may be possible to control fungal infections using chitinaseinhibitors such as methylxanthines (Tsirilakis et al., 2012). However itshould be noted that these compounds may not be effective against allknown fungal pathogens. Chitinases can also be used in the biocontrolof plant pathogens and insect vectors of human diseases as describedin Sections 6 and 4, respectively. Finally, chitinases have been utilizedin the production of neoglycoproteins (Li et al., 2008) and syntheticpolysaccharides (Faijes and Planas, 2007). Neoglycoproteins are valu-able tools for structure-function studies (Kent, 2004) while syntheticpolysaccharides offer promising opportunities for the development ofprophylactic and therapeutic agents (Boltje et al., 2009). From an indus-trial viewpoint, screening alternative sources such as extreme habitatsfor chitinolytic enzymes may be advantageous as related glycosylhydrolases obtained from extremophiles often demonstrate superiorcharacteristics when catalytic reactions are performed under non-physiologic conditions such as high salinity and low water activity(Karan et al., 2012; Moshfegh et al., 2013; Niknejad et al., 2013). Thismay also be the case for chitinases. For example, such conditions maybe encountered in cases where chitinases are used as biocontrol agents

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 8: From bacteria to human: A journey into the world of chitinases

670

671

672

673

674675676677678679680681682683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739740741742743744745746747748749750

751752753754

8 S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

(Barranco-Florido et al., 2002). Similarly, thermostable chitinases mayoffer considerable advantages for the production of oligosaccharides athigh temperature and low pH (Hobel et al., 2005).

T

755756757758759760761762763764765766767768769770771772773774775776777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806807808809810811812813814815816817818819820821822823824825826827828829830831832833834835836

UNCO

RREC

References

Aam BB, Heggset EB, Norberg AL, Sorlie M, Varum KM, Eijsink VG. Production ofchitooligosaccharides and their potential applications in medicine. Mar Drugs2010;8:1482–517.

Adrain C, Freeman M. New lives for old: evolution of pseudoenzyme function illustratedby iRhoms. Nat Rev Mol Cell Biol 2012;13:489–98.

Adrangi S, Faramarzi MA, Shahverdi AR, Sepehrizadeh Z. Purification and characterizationof two extracellular endochitinases from Massilia timonae. Carbohydr Res 2010;345:402–7.

Agapov E, Battaile JT, Tidwell R, Hachem R, Patterson GA, Pierce RA, et al. Macrophagechitinase 1 stratifies chronic obstructive lung disease. Am J Respir Cell Mol Biol2009;41:379–84.

Al Ahmadi KJ, Yazdi MT, Najafi MF, Shahverdi AR, Faramarzi MA, Zarrini G, et al. Optimi-zation of medium and cultivation conditions for chitinase production by the newlyisolated: Aeromonas sp. Biotechnology 2008;7:266–72.

Jami Al Ahmadi K, Tabatabaei Yazdi M, Fathi Najafi M, Shahverdi AR, Faramarzi MA, et al.Isolation and characterization of a chitionolytic enzyme producing microorganism,Paenibacillus chitinolyticus JK2 from Iran. Res J Microbiol 2008;3:395–404.

Alipour M, Suntres ZE, Lafrenie RM, Omri A. Attenuation of Pseudomonas aeruginosavirulence factors and biofilms by co-encapsulation of bismuth-ethanedithiol withtobramycin in liposomes. J Antimicrob Chemother 2010;65:684–93.

Amian AA, Papenbrock J, Jacobsen HJ, Hassan F. Enhancing transgenic pea (Pisum sativumL.) resistance against fungal diseases through stacking of two antifungal genes(chitinase and glucanase). GM Crops 2011;2:104–9.

Aminuddin F, Akhabir L, Stefanowicz D, Pare PD, Connett JE, Anthonisen NR, et al. Geneticassociation between human chitinases and lung function in COPD. Hum Genet2011;131:1105–14.

Andersen OA, DixonMJ, Eggleston IM, van Aalten DM. Natural product family 18 chitinaseinhibitors. Nat Prod Rep 2005;22:563–79.

Aomatsu T, Imaeda H, Matsumoto K, Kimura E, Yoden A, Tamai H, et al. Faecal chitinase3-like-1: a novel biomarker of disease activity in paediatric inflammatory boweldisease. Aliment Pharmacol Ther 2011;34:941–8.

Arakane Y, Muthukrishnan S. Insect chitinase and chitinase-like proteins. Cell Mol Life Sci2010;67:201–16.

Arimori T, Kawamoto N, Shinya S, Okazaki N, Nakazawa M, Miyatake K, et al. Crystalstructures of the catalytic domain of a novel glycohydrolase family 23 chitinasefrom Ralstonia sp. A-471 reveals a unique arrangement of the catalytic residues forinverting chitin hydrolysis. J Biol Chem 2013;288:18696–706.

Aronstein KA, Murray KD, Saldivar E. Transcriptional responses in honey bee larvaeinfected with chalkbrood fungus. BMC Genomics 2010;11:391.

Baloyi MA, LaingMD, Yobo KS. Use of mixed cultures of biocontrol agents to control sheepnematodes. Vet Parasitol 2012;184:367–70.

Barranco-Florido JE, Alatorre-Rosas R, Gutiérrez-Rojas M, Viniegra-González G, Saucedo-Castañeda G. Criteria for the selection of strains of entomopathogenic fungiVerticillium lecanii for solid state cultivation. Enzyme Microb Technol 2002;30:910–5.

Bateman A, Bycroft M. The structure of a LysM domain from E. colimembrane-bound lyticmurein transglycosylase D (MltD). J Mol Biol 2000;299:1113–9.

Behr M, Hoch M. Identification of the novel evolutionary conserved obstructor multigenefamily in invertebrates. FEBS Lett 2005;579:6827–33.

Bergthorsson U, Andersson DI, Roth JR. Ohno's dilemma: evolution of new genes undercontinuous selection. Proc Natl Acad Sci U S A 2007;104:17004–9.

Bhatnagar RK, Arora N, Sachidanand S, Shahabuddin M, Keister D, Chauhan VS. Syntheticpropeptide inhibits mosquito midgut chitinase and blocks sporogonic developmentof malaria parasite. Biochem Biophys Res Commun 2003;304:783–7.

Bilim O, Takeishi Y, Kitahara T, IshinoM, Sasaki T, Suzuki S, et al. Serum YKL-40 predicts ad-verse clinical outcomes in patientswith chronicheart failure. J Card Fail 2010;16:873–9.

Binod P, Sukumaran RK, Shirke SV, Rajput JC, Pandey A. Evaluation of fungal culturefiltrate containing chitinase as a biocontrol agent against Helicoverpa armigera.J Appl Microbiol 2007;103:1845–52.

Boltje TJ, Buskas T, Boons GJ. Opportunities and challenges in synthetic oligosaccharideand glycoconjugate research. Nat Chem 2009;1:611–22.

Boot RG, Renkema GH, Strijland A, van Zonneveld AJ, Aerts JM. Cloning of a cDNAencoding chitotriosidase, a human chitinase produced by macrophages. J Biol Chem1995;270:26252–6.

Boot RG, Blommaart EF, Swart E, Ghauharali-van der Vlugt K, Bijl N, Moe C, et al. Identi-fication of a novel acidic mammalian chitinase distinct from chitotriosidase. J BiolChem 2001;276:6770–8.

Boot RG, Bussink AP, Verhoek M, de Boer PA, Moorman AF, Aerts JM. Marked differencesin tissue-specific expression of chitinases in mouse and man. J Histochem Cytochem2005;53:1283–92.

Brotman Y, Landau U, Pnini S, Lisec J, Balazadeh S, Mueller-Roeber B, et al. The LysMreceptor-like kinase LysM RLK1 is required to activate defense and abiotic-stressresponses induced by overexpression of fungal chitinases in Arabidopsis plants. MolPlant 2012;5:1113–24.

Bucolo C, Musumeci M, Musumeci S, Drago F. Acidic mammalian chitinase and the eye:implications for ocular inflammatory diseases. Front Pharmacol 2011;2:43.

Buist G, Steen A, Kok J, Kuipers OP. LysM, a widely distributed protein motif for binding to(peptido)glycans. Mol Microbiol 2008;68:838–47.

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

Busby JN, Landsberg MJ, Simpson RM, Jones SA, Hankamer B, Hurst MR, et al. Structuralanalysis of Chi1 chitinase from Yen-Tc: the multisubunit insecticidal ABC toxincomplex of Yersinia entomophaga. J Mol Biol 2012;415:359–71.

Bussink AP, van Eijk M, Renkema GH, Aerts JM, Boot RG. The biology of the Gaucher cell:the cradle of human chitinases. Int Rev Cytol 2006;252:71–128.

Bussink AP, Speijer D, Aerts JM, Boot RG. Evolution of mammalian chitinase(-like)members of family 18 glycosyl hydrolases. Genetics 2007;177:959–70.

Cai Y, Cao F, Wei K, Zhang G, Wu F. Genotypic dependent effect of exogenous glutathioneon Cd-induced changes in proteins, ultrastructure and antioxidant defense enzymesin rice seedlings. J Hazard Mater 2011;192:1056–66.

Cantarel BL, Coutinho PM, Rancurel C, Bernard T, LombardV,Henrissat B. TheCarbohydrate-Active EnZymes database (CAZy): an expert resource for Glycogenomics. Nucleic AcidsRes 2009;37:D233–8.

Chandler JC, Molins CR, Petersen JM, Belisle JT. Differential chitinase activity and produc-tion within Francisella species, subspecies, and subpopulations. J Bacteriol 2011;193:3265–75.

Chang ST, Zahn JM, Horecka J, Kunz PL, Ford JM, Fisher GA, et al. Identification of abiomarker panel using a multiplex proximity ligation assay improves accuracy ofpancreatic cancer diagnosis. J Transl Med 2009;7:105.

Chaudhari SS, Arakane Y, Specht CA, Moussian B, Boyle DL, Park Y, et al. Knickkopf proteinprotects and organizes chitin in the newly synthesized insect exoskeleton. Proc NatlAcad Sci U S A 2011;108:17028–33.

Chaudhuri S, Bruno JC, Alonzo III F, Xayarath B, Cianciotto NP, Freitag NE. Contribution ofchitinases to Listeria monocytogenes pathogenesis. Appl Environ Microbiol 2010;76:7302–5.

Colussi PA, Specht CA, Taron CH. Characterization of a nucleus-encoded chitinase from theyeast Kluyveromyces lactis. Appl Environ Microbiol 2005;71:2862–9.

Consoli FL, Brandt SL, Coudron TA, Vinson SB. Host regulation and release of parasitism-specific proteins in the system Toxoneuron nigriceps–Heliothis virescens. CompBiochem Physiol B Biochem Mol Biol 2005;142:181–91.

Correale J, Fiol M. Chitinase effects on immune cell response in neuromyelitis optica andmultiple sclerosis. Mult Scler 2011;17:521–31.

Cottrell MT, Wood DN, Yu L, Kirchman DL. Selected chitinase genes in cultured andunculturedmarine bacteria in the alpha- and gamma-subclasses of the proteobacteria.Appl Environ Microbiol 2000;66:1195–201.

Cozzarini E, Bellin M, Norberto L, Polese L, Musumeci S, Lanfranchi G, et al. CHIT1 andAMCase expression in human gastric mucosa: correlation with inflammation andHelicobacter pylori infection. Eur J Gastroenterol Hepatol 2009;21:1119–26.

Dahiya N, Tewari R, Hoondal GS. Biotechnological aspects of chitinolytic enzymes: areview. Appl Microbiol Biotechnol 2006;71:773–82.

Danneels EL, Rivers DB, de Graaf DC. Venom proteins of the parasitoid waspNasonia vitripennis: recent discovery of an untapped pharmacopee. Toxins 2010;2:494–516.

Davies GJ, Wilson KS, Henrissat B. Nomenclature for sugar-binding subsites in glycosylhydrolases. Biochem J 1997;321(Pt. 2):557–9.

Delpin MW, Goodman AE. Nitrogen regulates chitinase gene expression in a marinebacterium. ISME J 2009;3:1064–9.

Duc C, Nentwig W, Lindfeld A. No adverse effect of genetically modified antifungal wheaton decomposition dynamics and the soil fauna community—a field study. PLoS One2011;6:e25014.

Ebrahim S, Usha K, Singh B. Pathogenesis-related (PR)-proteins: chitinase and β-1,3-glucanase in defense mechanism against malformation in mango (Mangifera indicaL.). Sci Hortic 2011;130:847–52.

Edreva A. Pathogenesis-related proteins: research progress in the last 15 years. Gen ApplPlant Physio 2005;31:105–24.

Faijes M, Planas A. In vitro synthesis of artificial polysaccharides by glycosidases andglycosynthases. Carbohydr Res 2007;342:1581–94.

Fan Y, Fang W, Guo S, Pei X, Zhang Y, Xiao Y, et al. Increased insect virulence in Beauveriabassiana strains overexpressing an engineered chitinase. Appl Environ Microbiol2007;73:295–302.

Fang W, Leng B, Xiao Y, Jin K, Ma J, Fan Y, et al. Cloning of Beauveria bassiana chitinasegene Bbchit1 and its application to improve fungal strain virulence. Appl EnvironMicrobiol 2005;71:363–70.

Fang W, Azimzadeh P, St Leger RJ. Strain improvement of fungal insecticides for control-ling insect pests and vector-borne diseases. Curr Opin Microbiol 2012;15:232–8.

Faramarzi MA, Fazeli M, Yazdi MT, Adrangi S, Al Ahmadi KJ, Tasharrofi N, et al. Optimiza-tion of cultural conditions for production of chitinase by a soil isolate of Massiliatimonae. Biotechnology 2009;8:93–9.

Fitz LJ, DeClercq C, Brooks J, Kuang W, Bates B, Demers D, et al. Acidic mammalianchitinase is not a critical target for allergic airway disease. Am J Respir Cell Mol Biol2012;46:71–9.

Fredriksson S, Horecka J, Brustugun OT, Schlingemann J, Koong AC, Tibshirani R, et al.Multiplexed proximity ligation assays to profile putative plasma biomarkers relevantto pancreatic and ovarian cancer. Clin Chem 2008;54:582–9.

Fujita K, Shimomura K, Yamamoto K, Yamashita T, Suzuki K. A chitinase structurallyrelated to the glycoside hydrolase family 48 is indispensable for the hormonallyinduced diapause termination in a beetle. Biochem Biophys Res Commun 2006;345:502–7.

Funkhouser JD, Aronson Jr NN. Chitinase family GH18: evolutionary insights from thegenomic history of a diverse protein family. BMC Evol Biol 2007;7:96.

Giansanti A, Bocchieri M, Rosato V, Musumeci S. A fine functional homology betweenchitinases from host and parasite is relevant for malaria transmissibility. ParasitolRes 2007;101:639–45.

Goni O, Sanchez-Ballesta MT, Merodio C, Escribano MI. Potent cryoprotective activity ofcold and CO2-regulated cherimoya (Annona cherimola) endochitinase. J Plant Physiol2010;167:1119–29.

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 9: From bacteria to human: A journey into the world of chitinases

T

837838839840841842843844845846847848849850851852853854855856857858859860861862863864865866867868869870871872873874875876877878879880881882883884885886887888889890891892893894895896897898899900901902903904905906907908909910911912913914915916917918919920921922

923924925926927928929930931932933934935936937938939940941942943944945946947948949950951952953954955956957958959960961962963964965966967968969970971972973974975976977978979980981982983984985986987988989990991992993994995996997998999100010011002100310041005100610071008

9S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

UNCO

RREC

Gressent F, Mantegazza N, Cullimore JV, Driguez H, Ranjeva R, Bono JJ. High-affinity nodfactor binding site from Phaseolus vulgaris cell suspension cultures. Mol Plant MicrobeInteract 2002;15:834–9.

Gruber S, Seidl-SeibothV. Self versus non-self: fungal cellwall degradation in Trichoderma.Microbiology 2012;158:26–34.

Gruber S, Kubicek CP, Seidl-Seiboth V. Differential regulation of orthologous chitinasegenes in mycoparasitic Trichoderma species. Appl Environ Microbiol 2011a;77:7217–26.

Gruber S, Vaaje-Kolstad G, Matarese F, Lopez-Mondejar R, Kubicek CP, Seidl-Seiboth V.Analysis of subgroup C of fungal chitinases containing chitin-binding and LysMmodules in the mycoparasite Trichoderma atroviride. Glycobiology 2011b;21:122–33.

Guillen D, Sanchez S, Rodriguez-Sanoja R. Carbohydrate-binding domains: multiplicity ofbiological roles. Appl Microbiol Biotechnol 2010;85:1241–9.

Guo C, Ghadersohi S, Kephart GM, Laine RA, Sherris DA, Kita H, et al. Improving the detec-tion of fungi in eosinophilic mucin: seeing what we could not see before. OtolaryngolHead Neck Surg 2012;147:943–9.

Hartl D, He CH, Koller B, Da Silva CA, Kobayashi Y, Lee CG, et al. Acidic mammalianchitinase regulates epithelial cell apoptosis via a chitinolytic-independent mecha-nism. J Immunol 2009;182:5098–106.

Hartl L, Zach S, Seidl-Seiboth V. Fungal chitinases: diversity, mechanistic properties andbiotechnological potential. Appl Microbiol Biotechnol 2012;93:533–43.

Hassas-Roudsari M, Goff HD. Ice structuring proteins from plants: mechanism of actionand food application. Food Res Int 2012;46:425–36.

He CH, Lee CG, Dela Cruz CS, Lee CM, Zhou Y, Ahangari F, et al. Chitinase 3-like 1 regulatescellular and tissue responses via IL-13 receptor alpha2. Cell Rep 2013;4:830–41.

Heo KW, Hur DY, Park SK, Yang YI, Kwak HH, Kim TY. Expression of chitinases inhypertrophied adenoids of children. Otolaryngol Head Neck Surg 2011;145:660–5.

Hermans C, Porco S, Verbruggen N, Bush DR. Chitinase-like protein CTL1 plays a role inaltering root system architecture in response to multiple environmental conditions.Plant Physiol 2010;152:904–17.

Hobel CF, HreggvidssonGO,MarteinssonVT, Bahrani-Mougeot F, Einarsson JM, KristjanssonJK. Cloning, expression, and characterization of a highly thermostable family 18chitinase from Rhodothermus marinus. Extremophiles 2005;9:53–64.

Horn SJ, Sikorski P, Cederkvist JB, Vaaje-Kolstad G, Sorlie M, Synstad B, et al. Costs andbenefits of processivity in enzymatic degradation of recalcitrant polysaccharides.Proc Natl Acad Sci U S A 2006a;103:18089–94.

Horn SJ, Sorbotten A, Synstad B, Sikorski P, Sorlie M, Varum KM, et al. Endo/exo mecha-nism and processivity of family 18 chitinases produced by Serratia marcescens. FEBSJ 2006b;273:491–503.

Hossain MA, Noh HN, Kim KI, Koh EJ, Wi SG, Bae HJ, et al. Mutation of the chitinase-likeprotein-encoding AtCTL2 gene enhances lignin accumulation in dark-grownArabidopsisseedlings. J Plant Physiol 2010;167:650–8.

Humphrey W, Dalke A, Schulten K. VMD: visual molecular dynamics. J Mol Graph1996;14:33–8.

Imai Y, Tsuda T, Aochi S, Futatsugi-Yumikura S, Sakaguchi Y, Nakagawa N, et al. YKL-40(chitinase 3-like-1) as a biomarker for psoriasis vulgaris and pustular psoriasis.J Dermatol Sci 2011;64:75–7.

Isaacs AT, Jasinskiene N, Tretiakov M, Thiery I, Zettor A, Bourgouin C, et al. TransgenicAnopheles stephensi coexpressing single-chain antibodies resist Plasmodiumfalciparum development. Proc Natl Acad Sci U S A 2012;109:E1922–30.

Iyer LM, Anantharaman V, Aravind L. The DOMON domains are involved in heme andsugar recognition. Bioinformatics 2007;23:2660–4.

Jamialahmadi K, Behravan J, NajafiMF, Yazdi MT, Shahverdi AR, Faramarzi MA. Enzymaticproduction of N-acetyl-D-glucosamine from chitin using crude enzyme preparation ofAeromonas sp. PTCC1691. Biotechnology 2011;10:292–7.

Karan R, Capes MD, Dassarma S. Function and biotechnology of extremophilic enzymes inlow water activity. Aquat Biosyst 2012;8:4.

Kasprzewska A. Plant chitinases—regulation and function. CellMol Biol Lett 2003;8:809–24.KawadaM, Hachiya Y, Arihiro A,Mizoguchi E. Role of mammalian chitinases in inflamma-

tory conditions. Keio J Med 2007;56:21–7.Kent S. Novel forms of chemical protein diversity — in nature and in the laboratory. Curr

Opin Biotechnol 2004;15:607–14.Killiny N, Prado SS, Almeida RP. Chitin utilization by the insect-transmitted bacterium

Xylella fastidiosa. Appl Environ Microbiol 2010;76:6134–40.Kim JS, Je YH. A novel biopesticide production: attagel-mediated precipitation of chitinase

from Beauveria bassiana SFB-205 supernatant for thermotolerance. Appl MicrobiolBiotechnol 2010;87:1639–48.

Kim JS, Je YH, Woo EO. Roles of adjuvants in aphicidal activity of enzymes from Beauveriabassiana (Ascomycota: Hypocreales) SFB-205 supernatant. J Asia Pac Entomol2010;13:345–50.

Kitamoto S, Egashira K, Ichiki T, Han X, McCurdy S, Sakuda S, et al. Chitinase inhibitionpromotes atherosclerosis in hyperlipidemic mice. Am J Pathol 2013;183:313–25.

Knogge W, Scheel D. LysM receptors recognize friend and foe. Proc Natl Acad Sci U S A2006;103:10829–30.

Kumar A, Rao M. A novel bifunctional peptidic aspartic protease inhibitor inhibitschitinase A from Serratia marcescens: kinetic analysis of inhibition and binding affin-ity. Biochim Biophys Acta 2010;1800:526–36.

Kwon Y, Kim SH, Jung MS, Kim MS, Oh JE, Ju HW, et al. Arabidopsis hot2 encodes anendochitinase-like protein that is essential for tolerance to heat, salt and droughtstresses. Plant J 2007;49:184–93.

Landsberg MJ, Jones SA, Rothnagel R, Busby JN, Marshall SD, Simpson RM, et al. 3D struc-ture of the Yersinia entomophaga toxin complex and implications for insecticidalactivity. Proc Natl Acad Sci U S A 2011;108:20544–9.

Larsen MH, Leisner JJ, Ingmer H. The chitinolytic activity of Listeria monocytogenes EGD isregulated by carbohydrates but also by the virulence regulator PrfA. Appl EnvironMicrobiol 2010;76:6470–6.

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

Larsen T, Petersen BO, Storgaard BG, Duus JO, Palcic MM, Leisner JJ. Characterization of anovel Salmonella Typhimurium chitinasewhichhydrolyzes chitin, chitooligosaccharidesand an N-acetyllactosamine conjugate. Glycobiology 2011;21:426–36.

Lebensztejn DM, Skiba E, Werpachowska I, Sobaniec-Lotowska ME, Kaczmarski M. Serumlevel of YKL-40 does not predict advanced liver fibrosis in children with chronichepatitis B. Adv Med Sci 2007;52:120–4.

Lee CG. Chitin, chitinases and chitinase-like proteins in allergic inflammation and tissueremodeling. Yonsei Med J 2009;50:22–30.

Lee CG, Hartl D, Lee GR, Koller B, Matsuura H, Da Silva CA, et al. Role of breast regressionprotein 39 (BRP-39)/chitinase 3-like-1 in Th2 and IL-13-induced tissue responses andapoptosis. J Exp Med 2009;206:1149–66.

Li H, Greene LH. Sequence and structural analysis of the chitinase insertion domainreveals two conserved motifs involved in chitin-binding. PLoS One 2010;5:e8654.

Li L, Yi H. Differential expression of Arabidopsis defense-related genes in response to sulfurdioxide. Chemosphere 2012;87:718–24.

Li C, Huang W, Wang LX. Chemoenzymatic synthesis of N-linked neoglycoproteinsthrough a chitinase-catalyzed transglycosylation. BioorgMed Chem2008;16:8366–72.

Lu HF, Narayanan K, Lim SX, Gao S, Leong MF, Wan AC. A 3D microfibrous scaffold forlong-term human pluripotent stem cell self-renewal under chemically defined condi-tions. Biomaterials 2012;33:2419–30.

Lupetti A, de Boer MG, Erba P, Campa M, Nibbering PH. Radiotracers for fungal infectionimaging. Med Mycol 2011;49:S62–9.

Martinez CP, Echeverri C, Florez JC, Gaitan AL, Gongora CE. In vitro production of twochitinolytic proteins with an inhibiting effect on the insect coffee berry borer,Hypothenemus hampei (Ferrari) (Coleoptera: Curculionidae) and the fungus Hemileiavastatrix the most limiting pests of coffee crops. AMB Express 2012;2:22.

Mathiasen AB, Harutyunyan MJ, Jorgensen E, Helqvist S, Ripa R, Gotze JP, et al. PlasmaYKL-40 in relation to the degree of coronary artery disease in patients with stableischemic heart disease. Scand J Clin Lab Invest 2011;71:439–47.

Merzendorfer H, Zimoch L. Chitin metabolism in insects: structure, function and regula-tion of chitin synthases and chitinases. J Exp Biol 2003;206:4393–412.

Moshfegh M, Shahverdi AR, Zarrini G, Faramarzi MA. Biochemical characterization ofan extracellular polyextremophilic alpha-amylase from the halophilic archaeonHalorubrum xinjiangense. Extremophiles 2013;17:677–87.

Nakabachi A, Shigenobu S, Miyagishima S. Chitinase-like proteins encoded in the genomeof the pea aphid, Acyrthosiphon pisum. Insect Mol Biol 2010;19(Suppl. 2):175–85.

Niknejad F, Moshfegh M, Najafzadeh MJ, Houbraken J, Rezaei S, Zarrini G, et al.Halotolerant ability and α-amylase activity of some saltwater fungal isolates. IranJ Pharm Res 2013;12:113–9.

Ohno M, Togashi Y, Tsuda K, Okawa K, Kamaya M, Sakaguchi M, et al. Quantification ofchitinase mRNA levels in human and mouse tissues by real-time PCR: species-specific expression of acidic mammalian chitinase in stomach tissues. PLoS One2013;8:e67399.

Ohnuma T, Numata T, Osawa T, Mizuhara M, Lampela O, Juffer AH, et al. A class Vchitinase from Arabidopsis thaliana: gene responses, enzymatic properties, andcrystallographic analysis. Planta 2011;234:123–37.

Ohnuma T, Numata T, Osawa T, Inanaga H, Okazaki Y, Shinya S, et al. Crystal structure andchitin oligosaccharide-binding mode of a ‘loopful’ family GH19 chitinase from rye,Secale cereale, seeds. FEBS J 2012;279:3639–51.

Ortiz-Rodriguez T, de la Fuente-Salcido N, Bideshi DK, Salcedo-Hernandez R, Barboza-Corona JE. Generation of chitin-derived oligosaccharides toxic to pathogenic bacteriausing ChiA74, an endochitinase native to Bacillus thuringiensis. Lett Appl Microbiol2010;51:184–90.

OwhashiM, HaradaM, Suguri S, OmaeH, Ishii A. Identification of aneosinophil chemotacticfactor from anopheline mosquitoes as a chitinase family protein. Parasitol Res2008;102:357–63.

Pacios Bras C, Jorda MA, Wijfjes AH, Harteveld M, Stuurman N, Thomas-Oates JE, et al.A Lotus japonicus nodulation system based on heterologous expression of the fucosyltransferase NodZ and the acetyl transferase NoIL in Rhizobium leguminosarum. MolPlant Microbe Interact 2000;13:475–9.

Park SK, Heo KW,Hur DY, Yang YI. Chitinolytic activity in nasal polyps. Am J Rhinol Allergy2011;25:12–4.

Patil RS, Ghormade VV, Deshpande MV. Chitinolytic enzymes: an exploration. EnzymeMicrob Technol 2000;26:473–83.

Patil DN, Datta M, Chaudhary A, Tomar S, Sharma AK, Kumar P. Isolation, purification,crystallization and preliminary crystallographic studies of chitinase from tamarind(Tamarindus indica) seeds. Acta Crystallogr Sect F Struct Biol Cryst Commun2009;65:343–5.

Permpoonpattana P, Tolls EH, Nadem R, Tan S, Brisson A, Cutting SM. Surface layers ofClostridium difficile endospores. J Bacteriol 2011;193:6461–70.

Petkau G, Wingen C, Jussen LC, Radtke T, Behr M. Obstructor-A is required for epithelialextracellular matrix dynamics, exoskeleton function and tubulogenesis. J Biol Chem2012;287:21396–405.

Petutschnig EK, Jones AM, Serazetdinova L, Lipka U, Lipka V. The lysin motif receptor-likekinase (LysM-RLK) CERK1 is a major chitin-binding protein in Arabidopsis thalianaand subject to chitin-induced phosphorylation. J Biol Chem 2010;285:28902–11.

Peumans WJ, Proost P, Swennen RL, Van Damme EJ. The abundant class III chitinasehomolog in young developing banana fruits behaves as a transient vegetative storageprotein and most probably serves as an important supply of amino acids for thesynthesis of ripening-associated proteins. Plant Physiol 2002;130:1063–72.

Prasanna R, Gupta V, Natarajan C, Chaudhary V. Bioprospecting for genes involved in theproduction of chitosanases and microcystin-like compounds in the production ofchitosanase and microcystin-like compounds in Anabaena strains. World J MicrobiolBiotechnol 2010;26:717–24.

Purushotham P, Arun PV, Prakash JS, Podile AR. Chitin binding proteins act synergisticallywith chitinases in Serratia proteamaculans 568. PLoS One 2012;7:e36714.

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://

Page 10: From bacteria to human: A journey into the world of chitinases

T

10091010101110121013101410151016101710181019102010211022102310241025102610271028102910301031103210331034103510361037103810391040104110421043104410451046104710481049105010511052105310541055105610571058105910601061106210631064106510661067106810691070107110721073107410751076107710781079108010811082108310841085

10861087108810891090109110921093109410951096109710981099110011011102110311041105110611071108110911101111111211131114111511161117111811191120112111221123112411251126112711281129113011311132113311341135113611371138113911401141114211431144114511461147114811491150115111521153115411551156115711581159116011611162

1164

10 S. Adrangi, M.A. Faramarzi / Biotechnology Advances xxx (2013) xxx–xxx

UNCO

RREC

Radutoiu S, Madsen LH, Madsen EB, Felle HH, Umehara Y, Gronlund M, et al. Plantrecognition of symbiotic bacteria requires two LysM receptor-like kinases. Nature2003;425:585–92.

Ramana Rao MV, Parameswari C, Sripriya R, Veluthambi K. Transgene stacking andmarker elimination in transgenic rice by sequential Agrobacterium-mediatedco-transformation with the same selectable marker gene. Plant Cell Rep2011;30:1241–52.

Rhodes RG, Atoyan JA, Nelson DR. The chitobiose transporter, chbC, is required for chitinutilization in Borrelia burgdorferi. BMC Microbiol 2010;10:21.

Roslind A, Johansen JS. YKL-40: a novel marker shared by chronic inflammation andoncogenic transformation. Methods Mol Biol 2009;511:159–84.

Royer V, Fraichard S, Bouhin H. A novel putative insect chitinase with multiple catalyticdomains: hormonal regulation during metamorphosis. Biochem J 2002;366:921–8.

Sakazaki Y, Hoshino T, Takei S, Sawada M, Oda H, Takenaka S, et al. Overexpressionof chitinase 3-like 1/YKL-40 in lung-specific IL-18-transgenic mice, smokers andCOPD. PLoS One 2011;6:e24177.

Sanchez-Rodriguez C, Bauer S, Hematy K, Saxe F, Ibanez AB, Vodermaier V, et al.Chitinase-like1/pom-pom1 and its homolog CTL2 are glucan-interacting proteinsimportant for cellulose biosynthesis in Arabidopsis. Plant Cell 2012;24:589–607.

Sarma K, Dehury B, Sahu J, Sarmah R, Sahoo S, Sahu M, et al. A comparative proteomicapproach to analyse structure, function and evolution of rice chitinases: a steptowards increasing plant fungal resistance. J Mol Model 2012;18:4761–80.

Schrank A, Vainstein MH. Metarhizium anisopliae enzymes and toxins. Toxicon 2010;56:1267–74.

Scully S, Yan W, Bentley B, Cao QJ, Shao R. Inhibitory activity of YKL-40 in mammaryepithelial cell differentiation and polarization induced by lactogenic hormones:a role in mammary tissue involution. PLoS One 2011;6:e25819.

Seidl V. Chitinases of filamentous fungi: a large group of diverse proteins with multiplephysiological functions. Fungal Biol Rev 2008;22:36–42.

Sels J, Mathys J, De Coninck BM, Cammue BP, De Bolle MF. Plant pathogenesis-related(PR) proteins: a focus on PR peptides. Plant Physiol Biochem 2008;46:941–50.

Seol HJ, Lee ES, Jung SE, Jeong NH, Lim JE, Park SH, et al. Serum levels of YKL-40 andinterleukin-18 and their relationship to disease severity in patients with preeclamp-sia. J Reprod Immunol 2009;79:183–7.

Shuhui L, Mok YK, Wong WS. Role of mammalian chitinases in asthma. Int Arch AllergyImmunol 2009;149:369–77.

Slamova K, Bojarova P, Petraskova L, Kren V. β-N-acetylhexosaminidase: what's in aname…? Biotechnol Adv 2010;28:682–93.

Stauff DL, Bassler BL. Quorum sensing in Chromobacterium violaceum: DNA recognitionand gene regulation by the CviR receptor. J Bacteriol 2011;193:3871–8.

Stefani FO, Tanguay P, Pelletier G, Piche Y, Hamelin RC. Impact of endochitinase-transformed white spruce on soil fungal biomass and ectendomycorrhizal symbiosis.Appl Environ Microbiol 2010;76:2607–14.

Streng A, op den Camp R, Bisseling T, Geurts R. Evolutionary origin of rhizobium Nodfactor signaling. Plant Signal Behav 2011;6:1510–4.

Strobel S, Roswag A, Becker NI, Trenczek TE, Encarnacao JA. Insectivorous bats digestchitin in the stomach using acidic mammalian chitinase. PLoS One 2013;8:e72770.

Sumida T, Fujimoto K, Ito M. Molecular cloning and catalytic mechanism of a novelglycosphingolipid-degrading beta-N-acetylgalactosaminidase from Paenibacillus sp.TS12. J Biol Chem 2011;286:14065–72.

Suzuki S, Nakanishi E, Furihata K, Miyamoto K, Tsujibo H, Watanabe T, et al. Chitinaseinhibitor allosamidin promotes chitinase production of Streptomyces generally. IntJ Biol Macromol 2008;43:13–9.

Tajdini F, Amini MA, Nafissi-Varcheh N, Faramarzi MA. Production, physiochemical andantimicrobial properties of fungal chitosan from Rhizomucor miehei and Mucorracemosus. Int J Biol Macromol 2010;47:180–3.

Takahashi M, Kiuchi M, Kamimura M. A new chitinase-related gene, BmChiR1, is inducedin the Bombyx mori anterior silk gland at molt and metamorphosis by ecdysteroid.Insect Biochem Mol Biol 2002;32:147–51.

Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S. MEGA5: molecular evolu-tionary genetics analysis using maximum likelihood, evolutionary distance, andmaximum parsimony methods. Mol Biol Evol 2011;28:2731–9.

Tang CM, ChyeML, Ramalingam S, Ouyang SW, Zhao KJ, UbhayasekeraW, et al. Functionalanalyses of the chitin-binding domains and the catalytic domain of Brassica junceachitinase BjCHI1. Plant Mol Biol 2004;56:285–98.

Tasharrofi N, Adrangi S, Fazeli M, Rastegar H, Khoshayand MR, Faramarzi MA. Optimiza-tion of chitinase production by Bacillus pumilus using Plackett–Burman design andresponse surface methodology. Iran J Pharm Res 2011;10:759–68.

Tran HT, Barnich N, Mizoguchi E. Potential role of chitinases and chitin-binding proteinsin host-microbial interactions during the development of intestinal inflammation.Histol Histopathol 2011;26:1453–64.

Tsirilakis K, Kim C, Vicencio AG, Andrade C, Casadevall A, Goldman DL. Methylxanthineinhibit fungal chitinases and exhibit antifungal activity. Mycopathologia 2012;173:83–91.

Udaya Prakash NA, Jayanthi M, Sabarinathan R, Kangueane P, Mathew L, Sekar K. Evolu-tion, homology conservation, and identification of unique sequence signatures inGH19 family chitinases. J Mol Evol 2010;70:466–78.

1163

Please cite this article as: Adrangi S, FaramarziMA, From bacteria to humandx.doi.org/10.1016/j.biotechadv.2013.09.012

ED P

RO

OF

Ueda M, Ohata K, Konishi T, Sutrisno A, Okada H, Nakazawa M, et al. A novel goose-typelysozyme genewith chitinolytic activity from themoderately thermophilic bacteriumRalstonia sp. A-471: cloning, sequencing, and expression. Appl Microbiol Biotechnol2009;81:1077–85.

Vaaje-Kolstad G, Horn SJ, van Aalten DM, Synstad B, Eijsink VG. The non-catalytic chitin-binding protein CBP21 from Serratia marcescens is essential for chitin degradation.J Biol Chem 2005;280:28492–7.

Vaaje-Kolstad G, Westereng B, Horn SJ, Liu Z, Zhai H, Sorlie M, et al. An oxidative enzymeboosting the enzymatic conversion of recalcitrant polysaccharides. Science 2010;330:219–22.

Van Damme EJ, Culerrier R, Barre A, Alvarez R, Rouge P, Peumans WJ. A novel family oflectins evolutionarily related to class V chitinases: an example of neofunctionalizationin legumes. Plant Physiol 2007;144:662–72.

van Eijk M, van Roomen CP, Renkema GH, Bussink AP, Andrews L, Blommaart EF, et al.Characterization of human phagocyte-derived chitotriosidase, a component of innateimmunity. Int Immunol 2005;17:1505–12.

Vasconcelos EA, Santana CG, Godoy CV, Seixas CD, Silva MS, Moreira LR, et al. A newchitinase-like xylanase inhibitor protein (XIP) from coffee (Coffea arabica) affectsSoybean Asian rust (Phakopsora pachyrhizi) spore germination. BMC Biotechnol2011;11:14–22.

Vega K, Kalkum M. Chitin, chitinase responses, and invasive fungal infections. IntJ Microbiol 2012;2012:920459.

Vincent B, Kaeslin M, Roth T, Heller M, Poulain J, Cousserans F, et al. The venom compo-sition of the parasitic wasp Chelonus inanitus resolved by combined expressedsequence tags analysis and proteomic approach. BMC Genomics 2010;11:693.

Vishnoi A, Kryazhimskiy S, BazykinGA,Hannenhalli S, Plotkin JB. Youngproteins experiencemore variable selection pressures than old proteins. Genome Res 2010;20:1574–81.

Walliwalagedara C, Atkinson I, van Keulen H, Cutright T, Wei R. Differential expression ofproteins induced by lead in the dwarf sunflower Helianthus annuus. Phytochemistry2010;71:1460–5.

Wasano N, Konno K, Nakamura M, Hirayama C, Hattori M, Tateishi K. A unique latex pro-tein, MLX56, defends mulberry trees from insects. Phytochemistry 2009;70:880–8.

Watanabe T, Kanai R, Kawase T, Tanabe T, Mitsutomi M, Sakuda S, et al. Family 19chitinases of Streptomyces species: characterization and distribution. Microbiology1999;145(Pt. 12):3353–63.

Wohlkonig A, Huet J, Looze Y, Wintjens R. Structural relationships in the lysozyme super-family: significant evidence for glycoside hydrolase signature motifs. PLoS One2010;5:e15388.

Wu B, Zhang B, Dai Y, Zhang L, Shang-Guan K, Peng Y, et al. Brittle Culm15 encodes amembrane-associated chitinase-like protein required for cellulose biosynthesis inrice. Plant Physiol 2012;159:1440–52.

Yaish MW, Doxey AC, McConkey BJ, Moffatt BA, Griffith M. Cold-active winter ryeglucanases with ice-binding capacity. Plant Physiol 2006;141:1459–72.

Yang H, Zhang T, Masuda T, Lv C, Sun L, Qu G, et al. Chitinase III in pomegranate seeds(Punica granatum Linn.): a high-capacity calcium-binding protein in amyloplasts.Plant J 2011;68:765–76.

Yennamalli RM, Rader AJ, Wolt JD, Sen TZ. Thermostability in endoglucanases isfold-specific. BMC Struct Biol 2011;11:10.

Yoshida E, Hidaka M, Fushinobu S, Koyanagi T, Minami H, Tamaki H, et al. Role of a PA14domain in determining substrate specificity of a glycoside hydrolase family 3beta-glucosidase from Kluyveromyces marxianus. Biochem J 2010;431:39–49.

Zakariassen H, Klemetsen L, Sakuda S, Vaaje-Kolstad G, Vårum KM, SørlieM, et al. Effect ofenzyme processivity on the efficacy of a competitive chitinase inhibitor. CarbohydrPolym 2010;82:779–85.

Zakariassen H, Hansen MC, Joranli M, Eijsink VG, Sørlie M. Mutational effectson transglycosylating activity of family 18 chitinases and construction of ahypertransglycosylating mutant. Biochemistry 2011;50:5693–703.

Zhang H, Liu M, Tian Y, Hu X. Comparative characterization of chitinases from silkworm(Bombyx mori) and bollworm (Helicoverpa armigera). Cell Biochem Biophys2011a;61:267–75.

Zhang J, Zhang X, Arakane Y, Muthukrishnan S, Kramer KJ, Ma E, et al. Comparative geno-mic analysis of chitinase and chitinase-like genes in the African malaria mosquito(Anopheles gambiae). PLoS One 2011b;6:e19899.

Zhang X, Huang Y, Harvey PR, Ren Y, Zhang G, Zhou H, et al. Enhancing plant disease sup-pression by Burkholderia vietnamiensis through chromosomal integration of Bacillussubtilis chitinase gene chi113. Biotechnol Lett 2012;34:287–93.

Zheng YP, Retnakaran A, Krell PJ, Arif BM, Primavera M, Feng QL. Temporal, spatial andinduced expression of chitinase in the spruce budworm, Choristoneura fumiferana.J Insect Physiol 2003;49:241–7.

Zhu Z, Zheng T, Homer RJ, Kim YK, Chen NY, Cohn L, et al. Acidic mammalian chitinase inasthmatic Th2 inflammation and IL-13 pathway activation. Science2004;304:1678–82.

Zhu Q, Arakane Y, Beeman RW, Kramer KJ, Muthukrishnan S. Functional specializationamong insect chitinase family genes revealed by RNA interference. Proc Natl AcadSci U S A 2008;105:6650–5.

Zou CG, Tao N, Liu WJ, Yang JK, Huang XW, Liu XY, et al. Regulation of subtilisin-like pro-tease prC expression by nematode cuticle in the nematophagous fungus Clonostachysrosea. Environ Microbiol 2010;12:3243–52.

: A journey into theworld of chitinases, Biotechnol Adv (2013), http://


Recommended