Srivastava 2016

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 88

   

Production, properties, and applications of endo-β-mannanases

Praveen Kumar Srivastava, Mukesh Kapoor

PII: S0734-9750(16)30136-7
DOI: doi:10.1016/j.biotechadv.2016.11.001
Reference: JBA 7082

To appear in: Biotechnology Advances

Received date: 28 December 2015


Revised date: 12 October 2016
Accepted date: 7 November 2016

Please cite this article as: Srivastava Praveen Kumar, Kapoor Mukesh, Produc-
tion, properties, and applications of endo-β-mannanases, Biotechnology Advances (2016),
doi:10.1016/j.biotechadv.2016.11.001

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
ACCEPTED MANUSCRIPT

Title: Production, properties, and applications of endo-β-mannanases

Authors: Praveen Kumar Srivastava and Mukesh Kapoor§*

T
IP
Addresses: Department of Protein Chemistry and Technology, CSIR-Central Food

R
Technological Research Institute, Mysuru-570 020

SC
§
Academy of Scientific and Innovative Research (AcSIR), CSIR-CFTRI Campus,

Mysuru, India

NU
MA
*Corresponding Author: E-mail: mkapoor@cftri.res.in; Phone: +91-821-2515331
D
P TE
CE
AC

1
ACCEPTED MANUSCRIPT

Abstract

The present review provides up-to-date information on the occurrence and

methodologies used for producing and purifying endo-β-mannanases and a

T
comprehensive comparison of their biochemical properties. The amalgamation of

IP
biochemical, molecular and structural biology approaches which have been used for

R
understanding endo-β-mannanase families, catalytic mechanism, substrate binding,

SC
non-catalytic modules, trans-glycosylation, and multi-functional enzyme complexes

has been given critical attention. A separate section entailing the state-of-the-art about

NU
thermostable endo-β-mannanases, which has emerged as an exciting field of both
MA
basic and applied research, is also deliberated. The remarkable progress made by

endo-β-mannanases in various industrial sectors like food, feed, detergents, biofuel


D

and oil drilling is also emphasized.


TE

Key words: Endo-β-mannanase, Biochemical properties, Genetic engineering

Structural organization, Thermo-stability, Industrial application


P
CE
AC

2
ACCEPTED MANUSCRIPT

1. Introduction

Plant cell walls are macromolecular structures comprising of an intricate intermix of

cellulose and a matrix of polysaccharides, glycoproteins, proteoglycans, low-

T
molecular-weight compounds, and ions (Willats et al., 2001). They represent an

IP
abundant, inexpensive, uniquely sustainable, and renewable resource that offers

R
enormous potential in the production of numerous food and non-food consumer

SC
products such as prebiotics, fuels, organic chemicals, and polymeric materials (Broda,

1992).

NU
Cellulose, hemicellulose and lignin in an approximate ratio of 2:1:1 makes up
MA
to 90% of the plant cell wall (Van Zyl et al., 2010). Cellulose, the most abundant

biopolymer, comprises of a bundle of linear poly β-1,4-glucan chains organized by


D

intra-chain and inter-chain H-bonding networks (Kuhad et al., 1997). Hemicelluloses


TE

comprise 25-30% of total wood dry weight, and are composed of more than 20

different monosaccharides in diverse combinations (Filho, 1998; Pérez et al., 2002).


P

Xylans represent the pre-dominant hemicellulosic component in hardwood from


CE

angiosperms, while mannans represent the most abundant hemicelluloses in


AC

softwoods, and specialized structures like plant seeds (Beg et al., 2001; Pérez et al.,

2002).

The structural and heterogeneous nature of mannans requires close association

and synergy between a variety of main chain and side chain cleaving enzymes like

endo-β-mannanase (EC 3.2.1.78), exo-β-mannosidase (EC 3.2.1.25), β-glucosidase

(EC 3.2.1.21), acetyl mannan esterases (EC 3.1.1.6), and α-galactosidase (EC

3.2.1.22) for their biodegradation (Dhawan and Kaur, 2007; Malgas et al., 2015a,

2015b). Mannan-degrading enzyme systems can be found in several glycosyl

hydrolase families (like GH 1, GH 2, GH 3, GH 5, GH 26, GH 27, GH 113 etc.)

3
ACCEPTED MANUSCRIPT

(www.cazy.org). They contribute to 1) microbial metabolism by the generation of

energy equivalents (simpler monosaccharides/oligosaccharides) from substrate

hydrolysis, 2) in plant‘s growth, maturation, and ripening by the metabolism of cell

T
wall mannans (Moreira and Filho, 2008).

IP
Mannanases obtained from various GH families are quite different with

R
respect to their primary structure, but are similar in their spatial arrangement, (β/α)8-

SC
barrel protein fold and are grouped into GH-A clan (Hilge et al., 1998). They often

display modular architectures generally comprising of catalytic domain(s),

NU
carbohydrate binding module(s) (CBM), and additional functional domain(s) (Sunna,
MA
2010). X-ray crystallographic and site-directed mutagenesis studies from a wide range

of species elucidated that endo-β-mannanases 1) require at least five substrate binding


D

sub-sites to carry out efficient catalysis of oligo- or polysaccharide substrates and 2)


TE

have an open cleft shaped active site with strictly conserved acid/base catalyst

(Glutamate at the beta-strand 4 end) and nucleophile catalyst (Glutamate at the beta-
P

strand 7 end) which are 5.5 Ǻ apart (Hilge et al., 1998; Hogg et al., 2003; Yan et al.,
CE

2008; Tailford et al., 2009). Recently, these enzymes have gathered significant
AC

attention from both academia and industry due to their potential applications in

bioconversion of lignocellulosic residues, food, feed, detergents, oil drilling, and

pharmaceuticals (Chauhan et al., 2012; Malgas et al., 2015a).

The recent progress made in the occurrence, structure, properties and

industrial applications of β-mannan and endo-β-mannanases (GH 5, GH 26, and GH

113) are discussed in the present review. Structural determinants, families, reaction

mechanism of endo-β-mannanases and a holistic view of thermo-stable endo-β-

mannanases are also covered. However, the present review does not cover other

mannan degrading enzymes like exo-β-mannosidases, α-galactosidases and esterases

4
ACCEPTED MANUSCRIPT

etc. or plant polysaccharide-degrading enzymes and synergistic interactions thereof,

and thus, the reader is advised to refer to earlier reviews (Gilbert et al., 2008; Malgas

et al., 2015a; Chauhan and Gupta, 2016) for further details.

T
2. β-Mannan: An important hemicellulose

IP
Mannans show a great deal of structural diversity and are sub-divided into four types

R
viz. pure/linear mannan, glucomannan, galactomannan, and galactoglucomannan on

SC
the basis of their sugar backbone composition (Popa and Spiridon, 1998) (Fig. 1).

They perform a variety of functions in plants, like acting as storage polysaccharides

NU
especially in endosperm or vacuoles of different seeds and vegetative tissues (Meier
MA
and Reid, 1982), providing structural integrity to the cell wall by binding to cellulose

and in addition, they also function like signalling molecules in growth and
D

development (Liepman et al., 2007). Currently, there is a great deal of interest in


TE

mannan-based polysaccharides as they provide multi-faceted benefits like immuno-

pharmacological, therapeutic, bio-medical, and prebiotic properties which find usage


P

in food, feed, and pharmaceuticals sectors (Maier et al., 1993; Moreira and Filho,
CE

2008; Ferreira et al., 2012; Yamabhai et al., 2016).


AC

2.1. Structure, occurrence and properties of β-mannan

2.1.1. Linear mannan

Linear mannan consists of a linear chain of β-1,4 linked D-mannosyl residues. They

are the major structural components in softwoods, hardwoods and plant seeds

(Aspinall, 1959). A neutral and water-soluble polysaccharide isolated from the current

pseudobulbs of Onicidium was a linear β, 1-4 linked mannan (Wang et al., 2006). The

linear mannan in the seed endosperm of Phytelephas macrocarpa and seeds derived

from Umbelliferae species provided protection from mechanical damage (Reid and

Edward, 1995). In some algal species, pure mannans have been believed to replace

5
ACCEPTED MANUSCRIPT

cellulose as the main cell wall glycan and are called as α-cellulose (Fernández et al.,

2012). Sulphated mannans (carbohydrates: sulphate, 2.7:1) in genus Codium form an

amorphous central layer and act as an interphase region between the neutral and

T
acidic layers (Fernández et al., 2012). Linear mannan act as a reserve material in

IP
families‘ viz. Apiaceae, Rubiaceae, and Asteraceae, while in Schizolobium

R
parahybae, family Caesalpiniaceae it did not act as a reserve material (Petkowicz et

SC
al., 2007).

2.1.2. Galactomannan

NU
Galactomannans are composed of 1,4-linked β-D-mannopyranosyl residues
MA
substituted by single 1,6-linked α-D-galactopyranosyl groups at the C-6 position in

sugar residues along the polysaccharide chain (McCleary et al., 1985).


D

Galactomannans in general, form highly viscous and stable aqueous solutions which
TE

are used as stiffeners and stabilizers in various food products (Srivastava and Kapoor,

2005). In the pharmaceutical industry, galactomannans are used as controlled drug


P

release agents (Toti and Aminabhavi, 2004). The frequency of side group substitution
CE

varies in galactomannans. For example, galactomannans isolated from guar seeds


AC

(Cyamopsis tetragonolobus, average molecular weight 220 kDa), tara gum,

(Caesalpinea spinosa) and carob seeds (Ceratonia siliqua, average molecular weight

310 kDa) have mannose-to-galactose ratios of 2:1, 3:1, and 4:1, respectively. The

average degree of polymerization (DP) of guar gum, tara gum, and carob gum ranges

between 3000-5000 (McCleary et al., 1985; Mudgil et al 2012a, b; Sébastien et al.,

2014). Galactomannans represents the most abundant constituents of coffee beans.

The β-(1→4)-linked mannan chains in coffee are substituted at O-6 position with

single galactose residues approximately every 100 residues (Campos-Vega et al.,

2015). Simões et al. (2010) have shown that pattern of acetylation was quite different

6
ACCEPTED MANUSCRIPT

in galactomannans found in coffee residues and infusions. Recently, galactomannans

have been isolated and characterized from the seeds of Gleditsia triacanthos,

Caesalpiniapul cherrima and Adenanthera pavonina. Electron spray ionization-

T
tandem mass spectrometry (ESI-MS/MS) analysis of G. tricanthos galactomannan

IP
indicated the presence of acetyl and pentose groups in addition to galactose

R
substitution (Cerqueira et al., 2011). A galactomannan from the seeds of Cassia

SC
grandis had a constant mannose/galactose ratio of 2.44:1 (Albuquerque et al., 2014),

while galactomannan from the seeds of vinal (Prosopis ruscifolia) had

NU
mannose/galactose ratio of 1.6, with traces of arabinose and glucose residues (Busch
MA
et al., 2015). 3D-molecular modelling studies established that intra-molecular

interactions between hairy (substituted) and smooth (unsubstituted) regions in


D

galactomannan from Sophora alopecuroides L. seeds are forming intra-molecular


TE

junction zones via hydrogen bonding in galactomannans with low mannose/galactose

ratios, e.g., 1.48-1.84 (Guo et al., 2016) (Fig. 2).


P

2.1.3. Glucomannan
CE

Glucomannan (average DP >200) is characterized by the presence of β-1,4-linked D-


AC

mannosyl and D-glucosyl residues, generally in 3:1 ratio, and are frequently

acetylated at the C-2, C-3 or C-6 positions (Northcote, 1972). Acetylated

glucomannans constitute about 60-80% of Konjac tuber (Maeda et al., 1980), and are

also reported from Lupinus varius (Ishurd et al., 2006). Konjac glucomannan has been

well-exploited in food and pharmaceutical sectors for the management of weight and

prevention of chronic diseases (Ishurd et al., 2006; Liang et al., 2015; Shah et al.,

2015). All the major fractions of O-acetyl-glucomannan from Dendrobium officinale

shared same major structure (2, 3-O-acetyl-glucomannan) but had different

mannose/glucose ratio, molecular weight, and intrinsic viscosity (Xing et al., 2014).

7
ACCEPTED MANUSCRIPT

Amorphophallus oncophyllus glucomannan, when compared to commercial

glucomannan, showed higher solubility and degree of acetylation, but lower viscosity,

water holding capacity and DP (Harmayani et al., 2014). Glucomannan from Bletilla

T
striata showed relative mole ratio of 2.4:1 mannose to glucose and molecular size of

IP
135 kDa (Wang et al., 2015).

R
2.1.4. Galactoglucomannan

SC
Galactoglucomannans are characterized by the presence of β-1,4-linked D-mannosyl

and D-glucosyl residues substituted with α-1,6-linked galactosyl side group in 3:1:1

NU
molar ratio (Puls, 1993; Popa and Spiridon, 1998). Schröder et al. (2001) reported
MA
galactoglucomannan from ripe kiwifruit with 1:2:2 galactose–glucose–mannose ratio

and molecular weight in the range of 16–42 kDa. Water extracted fractions of
D

rapeseed straw were rich in galactoglucomannan (Svärd et al., 2015). Acetylated


TE

galactoglucomannans are classified into two groups viz. galactose rich and acetyl poor

and galactose poor and acetyl rich (Willför et al., 2008). They form the predominant
P

portions of hemicelluloses found in wood from gymnosperms like Norway spruce


CE

(Picea abies) (Ekholm et al., 2012), angiosperms like black berry (Rubus fruticosus)
AC

(Cartier et al., 1988), spruce (Wilför et al., 2008) and ferns (Bailey and Pain, 1971).

Galactoglucomannan oligosaccharides alleviated cadmium stress in Arabidopsis

thaliana (Kučerová et al., 2014), while galactoglucomannan from Dendrobium

huoshanense has been suggested as anti-fibrotic agent for the prevention of liver

injury and fibrosis (Pan et al., 2012). Galactoglucomannans are also used as raw

material for packaging films and as an oxygen barrier (Jansson et al., 2014; Kisonen

et al., 2015). Recently, spruce galactoglucomannan and its carboxymethyl derivatives

were found to inhibit lipid oxidation (Lehtonen et al., 2016). For more information

8
ACCEPTED MANUSCRIPT

about mannans, readers are advised to refer to earlier reviews (Dhawan and Kaur,

2007; Moreira and Filho, 2008).

3. Endo-β-mannanase production from microorganisms

T
Mannanases are ubiquitous in nature and have been isolated from bacteria, fungi,

IP
actinomycetes, plants, and animals (Table 1). Submerged fermentation (SmF) and

R
solid state fermentation (SSF) are used predominantly for producing microbial endo-

SC
β-mannanases (Dhawan and Kaur, 2007; Chauhan et al., 2012). Several research

groups have attempted to improve endo-β-mannanase production by process

NU
optimization (Chantorn et al., 2013; Vijayalaxmi et al., 2013; Yin et al., 2013;
MA
Chauhan et al., 2014a).

3.1. Submerged fermentation (SmF)


D

SmF is the method of choice for producing endo-β-mannanase from microorganisms


TE

(Chauhan et al., 2012, 2014a; Srivastava and Kapoor, 2014). Wild-type microbial

endo-β-mannanases are extra-cellular, and their production is influenced by physico-


P

chemical and nutritional parameters like incubation time, pH, temperature, N2 content,
CE

and C-source. Approaches like one-factor-at-a-time, statistical methods like Plackett


AC

burman, Box Behnken, and central composite design or their combinations have been

used to improve endo-β-mannanase production (Ozturk et al., 2010; Srivastava and

Kapoor, 2014) (Table 2).

3.2. Solid state fermentation (SSF)

SSF offers colossal benefits for microbial endo-β-mannanase bioprocesses which

include, increased process productivity, less waste-water production, eco-friendly

nature, a higher concentration of enzyme, simple equipments, and low levels of

catabolic repression (Beg et al., 2000; Srivastava and Kapoor, 2014; Kuhad and

Kapoor, 2015). However, problems associated with scale up, downstream processing,

9
ACCEPTED MANUSCRIPT

biomass estimation, and control over process parameters has negatively affected its

adoption at industrial scale. Only a few microorganisms, mostly fungi, can produce

endo-β-mannanase under SSF conditions (Barrios-González, 2012; Thomas et al.,

T
2013) (Table 2).

IP
4. Heterologous cloning and expression of endo-β-mannanase gene

R
A large number of endo-β-mannanases have been cloned and expressed in

SC
heterologous hosts (Table 3) in order to a) make them suitable for industrial

applications by increasing their thermal stability, protease resistance, and pH stability

NU
(Sunna, 2010; Li et al., 2013; Xu et al., 2013), b) understand structure-function
MA
relationships, c) delineate the role of particular amino acid residues (Yan et al., 2008),

d) increase production (Songsiriritthigul et al., 2010; Kaira et al., 2016; Srivastava et


D

al., 2016), and e) purify enzyme in single step with high yield and specific activity
TE

(Huang et al., 2014).

Endo-β-mannanse gene from bacterial sources are usually overexpressed in


P

Escherichia coli (Srivastava et al., 2016) but there are also reports of other
CE

heterologous hosts like Bacillus megaterium (Summpunn et al., 2011), Brevibacillus


AC

brevis (Zhou et al., 2013), Pichia pastoris (Qiao et al., 2010), and Kluyveromyces

cicerisporous (Pan et al., 2011). Fungal endo-β-mannanase genes are heterologously

expressed most commonly in P. pastoris but Aspergillus sp. (Van Zyl et al., 2010) has

also been reported to express endo-β-mannanase. The β-mannanase encoding man5A

gene from acidophilic Bispora sp. MEY-1 was heterologously overexpressed in seeds

of maize plant (Xu et al., 2013). The size of endo-β-mannanase gene reported from

prokaryotic and eukaryotic sources ranges between 978-2010 kb. The fully mature

recombinant endo-β-mannanase proteins contain anywhere between 326-669 amino

acid residues with molecular weight ranging from 31-65 kDa (Table 3).

10
ACCEPTED MANUSCRIPT

Highest enzyme titres obtained after cloning and expression of endo-β-

mannanase gene are reported from Aspergillus aculeatus (expressed in Yarrowia

lipolytica) 1575 IU/ml (Yang et al., 2009) and Bacillus sp. CFR1601 [expressed in

T
Hi-Control E.coli BL21 (DE3)] 8,406 U/ml (Kaira et al., 2016). Recombinant endo-β-

IP
mannanase from Aspergillus usamii constituted more than 75% of the total protein

R
secreted into the culture supernatant (Li et al., 2014). A β-mannanase gene from

SC
Aspergillus sulphureus was codon optimized and expressed in P. pastoris with a yield

of 3 mg/ml and 35% increase in enzyme activity (Chen et al., 2009). Similarly, β-

NU
mannanase gene from B. subtilis MA139 was codon optimized and resulting
MA
recombinant enzyme had higher enzyme activity (Qiao et al., 2010).

5. Purification of endo-β-mannanases
D

Many protocols involving clarification, concentration/precipitation of enzyme


TE

followed by 2-6 chromatographic steps depending on the properties of endo-β-

mannanase(s) have been reported in the literature for the purification of endo-β-
P

mannanases (Table 4).


CE

6. Structural determinants, families, and reaction mechanism of endo-β-


AC

mannanases

6.1. Endo-β-mannanase families

Endo-β-mannanases from various organisms have been classified in glycoside

hydrolase (GH) families 5, 26, and 113 on the basis of amino acid sequence similarity

(www.cazy.org). A recent study (Shimizu et al., 2015) identified a novel mannanase

from Aspergillus nidulans which had no amino acid sequence similarity with other 1,4

endo-β-mannanases or to any protein with a known function and had been placed in a

new glycoside hydrolase family 134. Endo-β-mannanases obtained from various GH

families are quite different with respect to their primary structure, however

11
ACCEPTED MANUSCRIPT

interestingly they are very similar in their spatial arrangement, (β/α)8-barrel protein

fold and are grouped into GH-A clan (Fig. 3). A) GH 5 endo-β-mannanases: They are

primarily produced by eukaryotic organisms like fungi, plants, and animals (Larsson

T
et al., 2006; Van Zyl et al., 2010). However, few bacteria like Cellulosimicrobium sp.

IP
HY-13 (Kim et al., 2011a, 2011b), Vibrio sp. strain MA-138 (Tanaka et al., 2009),

R
Thermotoga petrophila (Dos Santos et al., 2012), and Bacillus licheniformis (Ethier et

SC
al., 1998) have also been reported to produce GH5 endo-β-mannanase. B) GH 26

endo-β-mannanases: They are found predominantly in bacteria like Bacillus sp.

NU
CFR1601 (Srivastava et al., 2014, 2016), Bacillus subtilis WL-3 (Yoon et al., 2008),
MA
B. subtilis B 23 (Zhou et al., 2013), Clostridium cellulovorans (Jeon et al., 2011),

Paenibacillus polymyxa (Cho et al., 2006), Pantoea agglomerans (Wang et al., 2010),
D

Pseudomonas fluorescens (Bolam et al., 1996), and Cellulomonas fimi (Hekmat et al.,
TE

2010). Presence of GH 5 (Ethier et al., 1998) and GH 26 (Songsiriritthigul et al.,

2010) mannanases have been reported from two different strains of B. licheniformis,
P

whereas coprophilic ascomycete Podospora anserina has been reported to produce


CE

both GH 5 and GH 26 endo-β-mannanase (Couturier et al., 2013a, 2013b).


AC

6.2. Substrate binding sub-sites

Endo-β-mannanase requires at least five substrate binding sub-sites to carry out

efficient catalysis of oligo- or polysaccharide substrates. According to Davies et al.

(1997), these sub-sites are numbered as +4, +3, +2, +1, -1, -2, -3 etc. starting from the

non-reducing end-to-the-reducing end, with hydrolysis of the O-glycosidic bond

placed between -1 and +1 sub-sites. Though the site of cleavage is same (-1 and +1

sub-sites) but the sub-sites used for interaction with the glycoside substrate may

differ. For instance, for interacting with mannotriose, a GH 5 endo-β-mannanase from

Thermobifida fusca (Hilge et al., 1998) (Fig. 4) employs -2, -3, and -4 enzyme sub-

12
ACCEPTED MANUSCRIPT

sites, while a GH26 endo-β-mannanase from Cellvibrio japonicas (Hogg et al., 2003)

makes use of -1, -2, and -3 sub-sites. GH5 enzyme, BaMan5A, derived from Bacillus

agaradhaerens can accommodate glucose or mannose at both -2 and +1 subsites,

T
while GH26 B. subtilis mannanase, BsMan26A, displays strong specificity only for

IP
mannose residues at its negative binding sites (Tailford et al., 2009). Therefore,

R
BaMan5A was able to hydrolyze glucomannan because the polar residue at the -2

SC
sub-site can make productive contact with the substrate 2-OH group in either its axial

(as in mannose) or its equatorial (as in glucose) configuration. However, other distal

NU
sub-sites do not exploit the 2-OH group as a specificity determinant. The tighter
MA
mannose recognition at the -2 sub-site in BsMan26A is mediated by interactions of

polar residues with the axial 2-OH group of a 4C1 ground state mannoside and is well
D

supported by previous studies on the C. japonicus GH26 mannanases CjMan26A


TE

(Hogg et al., 2003) and CjMan26C (Cartmell et al., 2008). The mutagenic variants of

CjMan26A, in which polar residues have been removed, do not distinguish between
P

mannose and glucose at the -2 subsite. Arg-361, His-377, and Glu-121 made pivotal
CE

productive polar interactions with mannose at the -2 subsite, but arginine, through its
AC

multiple interactions with O2 and O3, made the most significant contribution towards

substrate binding (Tailford et al., 2009). GH26 endo-β-mannanases select mannose at

both the -1 and -2 sub-sites and preferentially attack homo-polymers of mannose like

mannooligosaccharides or soluble mannans such as galactomannans. However, GH5

mannanases appear to display relaxed specificity for glucose or mannose at the

majority of the sub-sites and thus are optimized for glucomannan attack which is

found in the cell walls of angiosperms (Tailford et al., 2009). Kumagai et al. (2015)

highlighted the captivating differences in the properties of GH5 mannanases from

Streptomyces thermolilacinus (StMan) and T. fusca (TfMan) with respect to substrate

13
ACCEPTED MANUSCRIPT

recognition. StMan and TfMan hydrolysed galactosylmannooligosaccharide (GGM5;

6III,6IV-α-D-galactosyl mannopentaose) to GGM3 and M2, and GGM4 and M1,

respectively (Fig. 5). Both loop 7 and 8 were found as key regions for determining

T
substrate specificity.

IP
The presence of cellulose-specific non-catalytic carbohydrate binding modules

R
in many of GH5 enzymes functionally corroborates plant cell wall as the prime target

SC
of GH 5 enzymes (Hogg et al., 2003; Tailford et al., 2009). Couturier et al. (2013b)

have recently carried out structural and biochemical analyses of glycoside hydrolase

NU
families 5 (PaMan5A) and 26 (PaMan26A) β-(1,4)-mannanases from coprophilic
MA
fungus P. anserina. They have reported that the P. anserina mannanase (PaMan26A)

does not seem to agree with the previous models suggested for GH26 endo-β-
D

mannanases from B. subtilis and C. japonicus as it had atypical hydrolysis pattern


TE

with the release of mannotetrose and mannose from mannopentose resulting from a

predominant binding mode involving the -4 sub-site.


P

It could be concluded that many characteristics of endo-β-mannanases with


CE

respect to substrate specificity are conserved, but there are certain subtle differences
AC

in the key regions which interact with the substrate and leads to heterogeneity in the

sub-sites used for interaction with the sugar moieties.

6.3. Reaction mechanism

Endo-β-mannanases carry out catalysis by the double displacement mechanism with

retention of configuration (Couturier et al., 2013a, 2013b). In the first stage of

reaction, a glycosyl-enzyme intermediate is formed by a combination of nucleophilic

attack of the anomeric carbon and protonation of the glycosidic oxygen. In the later

stage of the reaction, the enzyme-substrate complex is cleaved by water attacking the

anomeric carbon of glycosyl group and releasing the enzyme in its original state

14
ACCEPTED MANUSCRIPT

(Bolam et al., 1996). GH 26 family mannanase (Pseudomonas cellulosa Man26A)


1
follows S5B2,5oS2 pyranoside ring inter-conversions by adopting B2,5

conformation in the transition state. The key specificity determinant in these

T
pseudoaxial ring inter-conversions for the hydrolysis of β-mannosides is the O3 atom.

IP
The 1S5 conformation of mannose orients the susceptible glycosidic linkage into an

R
axial position which simultaneously permits unhindered attack by nucleophilic

SC
glutamate at the opposite face of anomeric carbon (Ducros et al., 2002). Analysis of

computed Free Energy Surface of retaining β-D-mannosidases further corroborated

NU
this catalytic itinerary (Ardèvol et al., 2010). Similarly, other reports also support the
MA
fact that β-D-mannosidases catalyse the hydrolysis of sugar backbone via a boat like

conformation (Vincent et al., 2004; Tailford et al., 2008). However, other

glycosidases like cellulases or xylanases display 1S34H34C1 pseudo-rotational


D
TE

itinerary (Ducros et al., 2002; Vincent et al., 2004) (Fig. 6).

X-ray crystallographic and site directed mutagenesis studies of GH 26 endo-β-


P

mannanase from B. subtilis z-2 (Yan et al., 2008), P. fluorescens ssp. (Bolam et al.,
CE

1996), P. cellulosa (Hogg et al., 2001), C. fimi (Hekmat et al., 2010),and C. japonicus
AC

(Le Nours et al., 2005; Tailford et al., 2009) elucidated an open cleft shaped active

site with strictly conserved acid/base catalyst (Glutamate at the beta-strand 4 end) and

nucleophile catalyst (Glutamate at the beta-strand 7 end) which are 5.5 Ǻ apart (Hogg

et al., 2001). The catalytic acid-base and nucleophile residues in endo-β-mannanases

from different organisms are summarized in Table 5.

6.4. Non-catalytic carbohydrate binding modules (CBMs): Implications on substrate

hydrolysis

Endo-β-mannanases like other glycosyl hydrolases often display modular

architectures generally comprising of catalytic domain(s), CBM, and additional

15
ACCEPTED MANUSCRIPT

functional domain(s). A CBM is defined as a contiguous amino acid sequence within

a carbohydrate-active enzyme with independent fold and function and have

carbohydrate-binding activity. Their major function is to bind the enzyme to the

T
substrate and thereby enhance its catalytic activity, especially towards insoluble

IP
substrates. At present, 71,527 CBMs have been classified on the basis of amino acid

R
sequence, binding specificity and structure into 74 families (www.cazy.org). On the

SC
basis of three dimensional structure, CBMs have been divided into seven different

‗fold families‘; with most CBMs belonging to the β-sandwich fold family (Hägglund

NU
et al., 2003; Hervé et al., 2010; Zhang et al., 2014).
MA
Mannanases from Trichoderma reesei and Agaricus bisporus have been

shown to comprise of catalytic domain (CD) 5 at the N-terminal and CBM of family 1
D

at the C-terminal (Hägglund et al., 2003; Tang et al., 2001). Among bacteria,
TE

mannanases from T. petrophila (Da Silva et al., 2014), Caldocellulosiruptor

saccharolyticus (Morris et al., 1995), and Clostridium thermocellum ATCC 27405


P

(Ghosh et al., 2013) have been shown to contain CBM of families 27 at the C-
CE

terminal, 27 at the central position, and 35 appended at the N-terminal, respectively.


AC

The full length (CtManf) endo-β-mannanase from C. thermocellum ATCC 27405

exhibited higher activity and hydrolyzing capacity at higher temperatures than its

truncated derivative (CtManT) containing only catalytic domain. These observations

were attributed to the appended N-terminal CtCBM35 domain 1) facilitating increased

catalysis by concentrating the catalytic module in the vicinity of the substrate and 2)

prolonging the binding of the substrate (Ghosh et al., 2014a).

The properties of the CBM encoded by T. reesei β-mannanase Man5A was

studied by constructing a mutant lacking the part encoding the CBM (Man5A∆CBM).

Both Man5A∆CBM and wild-type enzyme were able to hydrolyse mannopentaose,

16
ACCEPTED MANUSCRIPT

soluble locust bean gum galactomannan and insoluble ivory nut mannan in similar

fashion. However, Man5A∆CBM showed a significant decrease in hydrolysis of

mannan/cellulose complex when compared with wild-type protein (Hägglund et al.,

T
2003).

IP
In Caldibacillus cellulovorans, putative ManA protein was encoded by

R
complete manA ORF2 and had multi-domain structure comprising of four domains: a

SC
putative N-terminal domain (D1) of unknown function, an internal carbohydrate

binding domain (CBD) (D2), a β-mannanase catalytic domain (D3), and a C-terminal

NU
CBD (D4). These domains were linked to each other via proline-threonine-rich
MA
peptides (Sunna et al., 2000). Modular β-mannanase (Man5C) from Vibrio sp. strain

MA-138 consisted of a catalytic module connected through a linker region to a C-


D

terminal CBM27. The presence of C-terminal CBM27 contributed to the binding of


TE

substrate to the modular enzyme and thereby increased the rate of depolymerisation of

soluble mannan. The truncated derivative without CBM27 exhibited 1.5–3.5-fold


P

lower catalytic efficiency (kcat/Km) towards soluble mannans as compared to the full-
CE

length Man5C (Tanaka et al., 2009). The modular organization of PaMan26A shows
AC

a family 35CBM at its N-terminal end, a short linker (12 residues, R133 to N141)

region rich in proline residues and no glycosylation sites. Moreover, the linker was

tightly bound to the CD and made sure that the CBM and CD are in close association

with each other (Couturier et al., 2013a, 2013b).

Despite the ability of CBMs to increase the affinity of the modular enzyme

towards its substrate, few studies have shown a decrease in turnover number (kcat) and

the kinetic efficiency (kcat/Km) of truncated derivatives upon removal of CBMs

(Sunna, 2010; Wang et al., 2014) indicating that the catalytic process was less

efficient in their presence. Sterical hindrances in substrate catalysis imposed by CBMs

17
ACCEPTED MANUSCRIPT

and unproductive binding due to competition in between CBM and catalytic module

to soluble substrate have been proposed as the plausible reasons (Sunna, 2010).

Truncation of CBM (ThMan5A△CBM) from mannanase from Trichoderma

T
harzianum improved its expression efficiency and properties of encoded enzyme. The

IP
recombinant enzyme (ThMan5A△CBM) produced 2,460 ± 45.1 U/ml of mannanase

R
activity in the culture supernatant which was 2.3-fold higher than the full-length

SC
ThMan5A gene (Wang et al., 2014). As most of the CBM functional studies have

been carried out with purified substrates, the likelihood of them mimicking the

NU
molecular complexity of the natural hemicellulose present in plant cell walls are
MA
abysmally low.

7. Biochemical properties of endo-β-mannanases


D

7.1. pH
TE

Fungal sources: Acidic pH optima has been reported for GH 5 endo-β-mannanases

from fungal sources like A. sulphureus (pH 2.4) (Chen et al., 2007), A. aculeatus (pH
P

2.5) (Pham et al., 2010), Penicillium oxalicum GZ2 (Liao et al., 2014), and
CE

Trichoderma reesi (pH 3.5) (Eneyskaya et al., 2009). However, a novel endo-β-
AC

mannanase belonging to a new GH family 134 from A. nidulans was found to be

optimally active at near neutral pH (Shimizu et al., 2015). With respect to pH

stability, endo-β-mannanases obtained from many fungal sources retained appreciable

(80%) activity in acidic to neutral pH range (4-7). Bacterial sources: They are

reported to be maximally active at neutral to alkaline pH. For example, endo-β-

mannanase (GH 5) from Bacillus nealsonii PN11 (Chauhan et al., 2014b) and Bacillus

N16-5 (GH 26) (Lin et al., 2007) were optimally active at pH 8 and 9.6, respectively.

An endo-β-mannanase (GH 26) from Bacillus sp. CFR1601 was optimally active at

pH 7 (Srivastava and Kapoor, 2015). Bacterial mannanases showed significant

18
ACCEPTED MANUSCRIPT

stability in the pH range 6-9 (Table 6). Interestingly, endo-β-mannanase from

Acinetobacter sp. ST 1-1 retained more than 80% activity in a broad pH range 3-10

(Titapoka et al., 2008).

T
7.2. Temperature

IP
Most endo-β-mannanases reported till date showed maximal activity in the

R
temperature range of 40-650C (Table 6). However, GH 5 endo-β-mannanase from

SC
Cryptopygus antarcticus displayed maximum activity at 300C (Song et al., 2008),

while Thermotoga neapolitana 5068 and P. oxalicum GZ2 GH 5 endo-β-mannanases

NU
exhibited maximum activity at 920C (Duffaud et al., 1997) and 800C (Liao et al.,
MA
2014), respectively. Endo-β-mannanases from Bacillus N16-5 (GH 26) (Lin et al.,

2007), A. aculeatus (GH 5) (Pham et al., 2010), and Caldicellulosiruptor Rt 8B.4 (GH

26) (Sunna, 2010) displayed maximum activity at temperatures more than 700C.
D
TE

7.3. Molecular weight and isoelectric point

The molecular weight of endo-β-mannanases reported from various sources is within


P

30-80 kDa range (Table 6). However, endo-β-mannanases belonging to GH 26 family


CE

from C. thermocellum (Ghosh et al., 2014a, 2014b) and unclassified family from
AC

Penicillium occitanis (Blibech et al., 2010) showed low molecular weight of 15kDa

and 18 kDa, respectively. Thermoanaerobacterium polysaccharolyticum produced a

high molecular weight (120 kDa) endo-β-mannanase (Cann et al., 1999), while C. fimi

produced three endo-β-mannanase having molecular weights of 30 kDa, 75 kDa and

100 kDa) (Stoll et al., 1999). The isoelectric point (pI) of most endo-β-mannanases is

reported in the range 4-8 (Table 6). In some cases, multiple forms of enzymes are

isoforms, produced from the same gene but differ due to subtle post-translational

modifications (Puchart et al., 2004) whereas in other organisms these enzymes are

encoded by different genes (Millward-Sadler et al., 1996).

19
ACCEPTED MANUSCRIPT

7.4. Kinetic parameters

The Km and Vmax of endo-β-mannanases are reported to be in the range of 0.0095-27.4

mg/ml and 29-16666.7 µmol/ml or mg/min, respectively. Locust bean gum and

T
Konjac mannan are the most commonly used substrates for the estimation of kinetic

IP
parameters (Table 6).

R
7.5. Effect of metal ions, surfactants, and chemical reagents

SC
Among additives, the effect of metal ions on endo-β-mannanases activity is quite

intriguing. For example, Fe3+ and Fe2+ inhibited activity of endo-β-mannanase from

NU
Paenibacillus cookie (Yin et al., 2012), whereas Fe3+ activated and Fe2+ had no effect
MA
on endo-β-mannanase from B. subtilis WY34 (Jiang et al., 2006). Similarly, Ag2+ also

showed differential effects wherein some cases it acted as activator while in other

cases as an inhibitor (Jiang et al., 2006; Luo et al., 2009; Yin et al., 2012). Co2+ and
D
TE

Hg2+ activated and inhibited many endo-β-mannanases, respectively (Jiang et al.,

2006; Luo et al., 2009; Kim et al., 2011b; Katrolia et al., 2012; Yin et al., 2012).
P

Metal ion chelators (EDTA/EGTA/citrate) inhibited endo-β-mannanases from


CE

Cellulosimicrobium sp. strain HY-13 (Kim et al., 2011a), C. thermocellum (Ghosh et


AC

al., 2013), and P. cookie (Yin et al., 2012) suggesting the importance of metal ions

during catalytic reaction. However, endo-β-mannanase from Sphingomonas sp. JB13

(Zhou et al., 2012) was stimulated by EDTA.

Disulphide reducing agents (iodoacetamide/β-mercaptoethanol/dithiothrietol)

had no major effect on endo-β-mannanase activity from P. cookie (Yin et al., 2012)

and B. subtilis WY34 (Jiang et al., 2006), but inhibited endo-β-mannanase from B.

nealsonii PN11 (Chauhan et al., 2014b). However, in other cases they activated endo-

β-mannanases from Cellulosimicrobium sp. strain HY-13 (Kim et al., 2011a), Bispora

antennata CBS 126.38 (Liu et al., 2012), and Rhizomucor miehei (Katrolia et al.,

20
ACCEPTED MANUSCRIPT

2013). N-Bromosuccinimide, a tryptophan modifying agent, inhibited endo-β-

mannanases from Cellulosimicrobium sp. strain HY-13 (Kim et al., 2011a, 2011b)

and B. subtilis WY34 (Jiang et al., 2006). Alkylating agent N-ethylemaleimide

T
slightly inhibited endo-β-mannanases from P. cookie (Yin et al., 2012).

IP
Non-ionic surfactants have mostly stimulated or had an insignificant effect on

R
the activity of endo-β-mannanases (Kim et al., 2011a, 2011b; Chauhan et al., 2014b).

SC
However, ionic surfactants like SDS and CTAB led to a drastic reduction in endo-β-

mannanase activity (Jiang et al., 2006; Zhou et al., 2012; Chauhan et al., 2014b;

NU
Srivastava and Kapoor, 2015). In few studies, organic solvents have been shown to
MA
stimulate (Chauhan et al., 2014b) or inhibit ( Zhou et al., 2012; Chauhan et al., 2014b)

the activity of endo-β-mannanases.


D

8. Transglycosylation activity
TE

The transglycosylation activity leads to the synthesis of new glycosides or

oligosaccharides which are longer than the original substrate. The glycone substrate,
P

forming the glycosyl enzyme intermediate, serve as a donor and the incoming
CE

carbohydrate hydroxyl group act as an acceptor molecule rather than water. Synthesis
AC

of mannose-containing glycol-conjugates using traditional organic chemistry-based

approaches is laborious and time-consuming. Endo-β-mannanases because of their

retaining double displacement mechanism are able to perform the transglycosylation

reactions and thus offers an easy alternative. GH 5 or 113 family endo-β-mannanase

from P. anserina (GH 5) (Couturier et al., 2013b), A. nidulans FGSC A4 (GH 5)

(Dilokpimol et al., 2011), Mytilus edulis (GH 5) (Larsson et al., 2006), and

Alicyclobacillus acidocaldarius Tc-12-31(GH 113) (Zhang et al., 2008) were able to

catalyze transglycosylation reactions by producing longer manno-oligosaccharides of

DP 4–6 from donor manno-oligosaccharides. Rosengren et al. (2014) recently showed

21
ACCEPTED MANUSCRIPT

very high transglycosylation activity in A. nidulans GH 5 endo-β-mannanase.

Acidophilic β-mannanase from Gloeophyllum trabeum CBS900.73 was able to

produce mannooligosaccharides with higher degree of polymerization from

T
mannotetrose by transglycosylation (Wang et al., 2016). There is no evidence at

IP
present for transglycosylation activity in GH 26 endo-β-mannanases.

R
9. Mannanase as a part of multi-functional enzyme complex

SC
Many enzymes are secreted individually by microorganisms in extracellular medium

but a few forms a multi-enzyme complex (MEC). MEC gives a competitive edge to

NU
microorganisms in ecological terms by improving the substrate hydrolysis and energy
MA
generation. Cellulosome which coordinate the deconstruction of cellulose and

hemicellulose and play a major role in carbon turnover represents one of the finest
D

examples of cell bound multienzyme complex (Fontes and Gilbert, 2010). The
TE

cellulosome of C. thermocellum, an anaerobic bacterium, showcases co-operative

interactions within an MEC for sustaining cell growth and survival. C. thermocellum
P

does not utilize mannose and xylose but still produces hemicellulose degrading
CE

enzymes like xylanase and mannanases as part of cellulosome which is dominated by


AC

enzymes (endoglucanases, exoglucanases and β-glucosidases) active in the

degradation of cellulose. This paradox is explained by the pivotal role of xylanase and

mannanases in cleavage of mannan/xylan matrix surrounding cellulose thereby

facilitating the access to cellulose by cellulases leading to production of sugars which

can be assimilated by the microorganism (Halstead et al., 1999). A large gene cluster

for cellulosome from Clostridium cellulovorans comprises of various genes (cbpA-

exgS-engH-engK-hbpA-engL-manA-engM-engN; 22 kb) which include the scaffolding

protein cellulose binding protein A (cbpA), exoglucanase (exgS), several

endoglucanases (engH, engK, engL, engM, and engN) of family 9, mannanase

22
ACCEPTED MANUSCRIPT

(manA), hydrophobic protein (hbpA) containing a surface layer homology domain,

and a hydrophobic (or cohesin) domain (Tamaru et al., 2000). The β-mannanase

reported from anaerobic extreme thermophile Caldocellum saccharolyticum is also a

T
part of a multi-domain enzyme. The amino-terminal catalytic domain has β-

IP
mannanase activity, while the carboxy-terminal domain acts as an endoglucanase

R
(Gibbs et al., 1992). A multi-enzyme complex was reported from B. licheniformis

SC
SVD1 contained two endoglucanases, seven xylanases, two mannanases, and one

pectinase (Van Dyk et al., 2010). Cellulosomal complex of Clostridium cellulovorans

NU
also displayed endo-β-mannanase activity (Jeon et al., 2011). A fusion gene was
MA
isolated from a microbial metagenome which encodes an N-terminal endo-1,4-β-

mannanase and C-terminal 1,3-1,4-β-glucanase (Wong et al., 2013).


D

10. Thermal stability of endo-β-mannanases: delineating the contributions of


TE

structural elements

A substantive amount of work has been carried out in the past several years to shed
P

light on the molecular basis of endo-β-mannanase thermal stability by understanding


CE

its biochemical and structural properties especially from microbial sources like A.
AC

usamii (Tang et al., 2013) and Streptomyces (Kumagai et al., 2012, 2013a, 2013b). It

has been shown by several studies on full length endo-β-mannanase, its truncated

derivatives or mutants that several molecular interactions like disulphide bridges, H-

bonding, and structural features like metal binding site with affinity for bivalent metal

ions like Ca2+, Mg2+, Mn2+, Ni2+ and Zn2+, linker sequences rich in Thr/Ser, and

internal repeats play a significant role in endo-β-mannanase thermal stability (Yan et

al., 2008; Kumagai et al., 2012, 2013a, 2013b; Lu et al., 2014; Srivastava et al., 2016).

Recent observations have also suggested that CBMs also function in an

23
ACCEPTED MANUSCRIPT

unconventional manner towards endo-β-mannanase thermal stability (Sunna, 2010;

Dos Santos et al., 2012).

10.1. The role of metal binding site

T
The thermal stability of wild-type and mutant endo-β-mannanases from Streptomyces

IP
thermoluteus (StManII) and Streptomyces lividans (SlMan) was enhanced (up to 170C

R
of Tm value) by addition of Ca2+ and Mn2+. In the catalytic domain of these endo-β-

SC
mannanases, a bivalent ion-binding site which binds one ion per molecule of enzyme

was responsible for thermal stability. The bivalent ions (Mg2+, Ca2+, and Mn2+) shared

NU
the same single binding site. Moreover, presence of Ca2+ and Mn2+ also led to changes
MA
in the secondary structures of these endo-β-mannanases. Primary structure alignment

of many GH5 bacterial mannanases has revealed high conservation of residues at the
D

calcium-binding site (Kumagai et al., 2012, 2013a, 2013b).


TE

In endo-β-mannanases like BCman from B. subtilis Z-2 (Yan et al., 2008) and

ManB-1601 from Bacillus sp. CFR1601 (Srivastava et al., 2014, 2016), presence of a
P

metal-binding motif comprising of six-coordinate Zn2+ with the His(1)-Glu(336)-


CE

His(23) has been shown (Fig. 7). The metal-binding motif linked the flexible loops at
AC

the N-terminus and C-terminus and increased the rigidity of the protein eventually

leading to high thermal stability. Studies on ManB-1601 thermal stability elaborated

the quintessential function of metal ions as EDTA treatment of holoenzyme

promulgated the population of conformational state that unfolds at lower temperature

(Srivastava et al., 2014, 2016).

10.2. The role of non-catalytic carbohydrate binding module (CBMs)

CBMs facilitate not only non-catalytic disruption of polysaccharide but also provide

molecular flexibility between the structural domains and optimize the geometry

between domains with respect to high temperatures (Blake et al., 2006; Hervé et al.,

24
ACCEPTED MANUSCRIPT

2010). Deletion of CBM 27 domain from GH5 β-mannanase from T. petrophila led

to reduction in midpoint of thermal denaturation from 1000C to 880C but had no effect

on catalytic efficiency or substrate specificity (Dos Santos et al., 2012). Thermo-

T
stability of GH5 β-mannanase (AuMan5A) derived from A. usamii YL-01-78 having

IP
only catalytic domain, has been improved by fusion with family 1 CBM of the T.

R
reesei cellobiohydrolase I (TrCBH I). However, the fusion protein showed increased

SC
Km and slightly altered Vmax values (Tang et al., 2013). Lu et al. (2014) and Wang et

al. (2014) provided striking evidence on improvement of thermal stability of GH5 β-

NU
mannanase (Man5XZ3) from A. nidulans XZ3 and ThMan5A from T. harzianum
MA
MGQ2 by removal of CBM, respectively.

The positioning of CBM domain with respect to catalytic domain affects

thermal sensitivity. It was shown in CsMan26, a modular β-mannanase from


D
TE

Caldicellulosiruptor strain Rt8.B4, consisting of two N-terminal family-27 CBMs

followed by a family 35 CBM and a family 26 glycoside hydrolase catalytic module


P

(Sunna, 2010).
CE

Linker sequences between catalytic domain and CBM also contribute towards
AC

thermo-stability of endo-β-mannanases. Removal of CBM1 (Man5ΔCBM) led to

improvement in thermo-stability of Man5XZ3, a multi-modular β-mannanase encoded

by A. nidulans XZ3, but removal of the linker region along with CBM1 (Man5ΔCL)

resulted in poor thermo-stability (Lu et al., 2014). These findings taken together with

the role of CBM‘s in substrate anchoring suggest that a) Presence of CBMs leads to

high efficiency of catalytic module towards the complex natural substrate like plant

cell wall by promoting molecular anchoring leading to close association and longer

contact with the substrate and b) CBMs work as thermo-stabilizing domains as

evident in many endo-β-mannanases but thermo-stabilising functions of CBM‘s still

25
ACCEPTED MANUSCRIPT

remains highly intriguing as there are reports of endo-β-mannanases wherein they do

not contribute towards thermo-stability. The latter observation also finds support from

CBM modules present in enzymes from mesophilic counterparts (Shoseyov et al.,

T
2006).

IP
10.3. Role of disulphide bridge, salt bridge, and H-bond

R
B. subtilis z-2 mannanase, BCman, showed a marked reduction in its thermal stability

SC
and loss of activity at 800C when the disulphide linkages were reduced with β-

mercaptoethanol. It has been postulated that conformational environment and solvent

NU
accessibility of disulphide bridges are the key requirements for the latter to act as
MA
stabilization factor in improving proteins thermo-stability (Yan et al., 2008).

However, no such role of disulphide bridge was evident in the thermal stability of
D

endo-β-mannanase from Bacillus sp. CFR1601 (Srivastava et al., 2014).


TE

The average ion-pair interaction per residue and utilization of arginine in such

ion-pairs (forming multiple hydrogen bonds with acidic partner) along with a
P

propensity to form salt bridges also helps in contributing towards thermo-stability of


CE

proteins. This has been shown for thermo-stable endo-β-mannanase (Q1.1) from
AC

Thermomonospora fusca which had 0.07 average ion pair interaction per residue.

Q1.1 and S. lividans endo-β-mannanases share high sequence similarity (54.0%).

However, Q1.1 with 10 additional salt bridges and four ion triplets (two of them

include an arginine forming hydrogen bond) has 270C higher temperature optima

(850C) than S. lividans endo-β-mannanases (580C) (Hilge et al., 1998).

11. Structure-based rational design and molecular evolution: Advent of endo-β-

mannanase with improved activity and stability

Endo-β-mannanase from P. agglomerans has been engineered using modified DNA

shuffling and site-directed mutagenesis. From a library of 19,700 clones, two mutant

26
ACCEPTED MANUSCRIPT

enzymes Gly267Ser and His134Arg/Phe141Leu displayed 1.14 and 3.3-fold increase

in catalytic efficiency (kcat/Km) towards locust bean gum, respectively. Enhancement

of polar contacts or positive charges by replacement of His 134 with either Arg or Lys

T
by site-directed mutagenesis resulted in 2.81- and 2.75-fold increase, respectively in

IP
the catalytic efficiency of the enzyme and decrease in Km (Man26P > His134Lys

R
>His134Arg) when compared with the native enzyme. The presence of weakly

SC
nucleophilic Ser (Gly267Ser) at 267 in place of Gly helped by forming new hydrogen

bonds which can interact with hydroxyl groups of the substrate and thereby facilitated

NU
the interaction of the enzyme with the substrate (Wang et al., 2013).
MA
In another study, site-directed mutagenesis has been employed to improve the

acid-stability of endo-β-mannanase from B. subtilis B10-02 by changing several


D

surface-exposed amino acid residues to acidic or neutral ones. Among the mutations,
TE

replacement of His 54 residue with Asp shifted the stability and optimal activity from

pH 6.5 to 5.5 (Xu et al., 2013).


P

With the rationale of improving hemicellulose hydrolysis, Guo et al. (2013)


CE

have constructed chimeras of xylanase and mannanase. The properties of xylanase


AC

and mannanase in chimera and their synergistic abilities in efficiently hydrolyzing

Luffa cylindrica fiber was shown to be influenced by the type of linker, the length of

linker, and by their order of integration. A structure-based rational design was also

employed to engineer GH-5 mannanase from Aspergillus niger BK01. Among the 23

mutants created, Tyr216Trp mutant showed an 18±2.7% improvement in its specific

activity as compared to the wild-type enzyme. The higher catalytic activity of this

mutant was attributed to 1) the extended aromatic ring of Trp leading to lower

substrate affinity by altering the interaction of polysaccharides, and 2) prevention of

the enzyme from retaining the substrate at the binding site (Huang et al., 2014). The

27
ACCEPTED MANUSCRIPT

Tm value and half-life of a mesophilic GH 5 β-mannanase from A. usamii was

improved by 12.10C and 48-fold, respectively by substitution of loop-structure using

rational design followed by megaprimer PCR (Dong et al., 2016).

T
Couturier et al. (2013a) have employed a random mutagenesis strategy using

IP
error-prone PCR and two steps of high-throughput enzymatic screening to generate

R
variants (10,800 mutants) of GH5 (PaMan5A) and GH26 (PaMan26A) endo-β-

SC
mannanases from ascomyceteous fungus P. anserina which displayed improved

activity (lowered Km and increased kcat/Km) towards galactomannan. One single

(PaMan5A-G311S), two double


NU
(PaMan5A-K139R/ Y223H, PaMan26A-
MA
P140L/D416G) and two triple mutants (PaMan5A-V256L/G276V/Q316H, PaMan5A-

W36R/I195T/V256A) were used for further characterization. Circular dichroism, pH


D

and temperature profiles of these mutants were similar to those of respective parental
TE

enzymes. However, all the selected variants displayed significant improvement in

activity as compared to wild-type enzymes. PaMan26A-P140L/D416G double mutant


P

having mutations in the linker region (P140L) and at the entrance of the active site
CE

(D416G) showed a 30% increase in kcat/Km compared to the parental enzyme. The
AC

reduced linker rigidity by virtue of first mutation (P140L) was given as the reason to

partially explain the increase in kcat/Km, while the lack of carboxylic side chain due to

second mutation (D416G) facilitated entry of substrate to the active site. Triple

mutant PaMan5A-W36R/I195T/V256A showed improved turnover towards

galactomannan and 1.8-fold higher catalytic efficiency. Single mutant PaMan5A-

G311S with almost unmodified structure except slight movement of the beta-strand 8

showed 8.2-fold increase in kcat/Km due to reduced Km possibly by the positioning of

W315 residue at the surface of enzyme by the interaction of hydroxyl group of serine

with nearby residues.

28
ACCEPTED MANUSCRIPT

12. Industrial applications of endo-β-mannanases

12.1. Functional food

Prebiotics and dietary fibres comprises of non-digestible oligosaccharides (NDO‘s)

T
and polysaccharides whose chemical structure prevents them from being digested in

IP
mammalian small intestine due to the absence of required digestive enzymes.

R
However, they are readily assimilated and metabolized by colonic probiotic

SC
microorganisms like Lactobacillus and Bifidobacteria. Fermentation of these

carbohydrates by probiotic strains is mediated through a pathway by glycolytic

NU
enzymes leading to the production of short chain fatty acids (SCFA) like acetic acid,
MA
propionic acid, and butyric acid. SCFA in turn impart various functional roles in the

body such as establishment of probiotic microbial community, growth inhibition of


D

pathogenic bacteria, as an energy source (mainly by acetate) in human and animal


TE

muscle, kidney, heart, and brain, hypo-cholesterolemic (mainly by propionate) effects,

protection against inflammatory bowel disease, ulcerative colitis, and Crohn‘s disease
P

(mainly by butyrate) (Li and Nie, 2015).


CE

Manno-oligosaccharides (MOS) represent an excellent prebiotic food


AC

ingredient. They are derived from the hydrolysis of mannan from guar gum, locust

bean gum or konjac gum. Several researchers have studied the potential of

mannanases from microorganisms like P. occitanis (locust bean gum) (Blibech et al.,

2011), B. subtilis CAe24 (copra meal) (Rungrassamee et al., 2014), Bacillus sp.

CFR1601 (guar gum, locust bean gum) (Srivastava and Kapoor, 2014; Srivastava et

al., 2015b) Bifidobacterium adolescentis (ivory nut mannan) (Kulcinskaja et al.,

2013), and Bifidobacterium animalis subsp. lactis Bl-04 (ivory nut mannan and locust

bean gum) (Morrill et al., 2015) in the production of MOS (Table 7).

29
ACCEPTED MANUSCRIPT

Guar gum (GG) is utilized in food products like dairy, soups, sauces, and

bakery as a source of dietary fibre and for hydration, thickening, and stabilizing

properties (Yoon et al., 2008). It also helps in reducing serum cholesterol and

T
alleviating symptoms of irritable bowel syndrome. However, the high viscosity of GG

IP
hampers its incorporation in food products besides affecting protein efficacy and

R
nutrient digestion. Partially hydrolysed guar gum (PHGG), which offers health

SC
benefits and stabilizing properties similar to GG, can be easily incorporated in food

products due to its high solubility and less viscosity and therefore, represents a

NU
feasible alternative to GG for the food industry. PHGG is also effective in treating
MA
irritable bowel syndrome (Yoon et al., 2008), and it has been shown recently that

PHGG could also serve as an ideal natural soluble fibre for delivering acute and long
D

term satiety effects and can reduce energy intake from whole day snacking (Rao et al.,
TE

2015). The partially hydrolysed galactomannan from Caesalpinia pulcherrima

presented suitable properties to be added as an alternative dietary fibre source in a


P

wide range of food products, particularly beverages (Buriti et al., 2014). PHGG is
CE

produced conventionally using physical (hydrothermal, ultra-sonication, and


AC

irradiation) and chemical (acid) methods which require high energy inputs and are

environment un-friendly (Gupta et al., 2015). Instead, low-cost and mild enzymatic

depolymerization of GG using endo-β-mannanases has been reported as a plausible

alternative (Srivastava and Kapoor, 2015).

12.2. Coffee and fruit juice clarification

Presence of mannans in coffee and fruit juice extracts leads to high viscosity, which

hampers their industrial processing and thus acts as a bottleneck in their effective

commercialization. Endo-β-mannanases from B. nealsonii PN11 (Chauhan et al.,

2014b), and Sclerotium rolfsii (Sachslehner et al., 2000) were successfully employed

30
ACCEPTED MANUSCRIPT

for reducing the dynamic viscosity of coffee extract. An immobilized mannanase was

shown to hydrolyse coffee galactomannan (Nicolas et al., 1998). Recently, application

of endo-β-mannanase from Lactobacillus plantarum M24 (Nadaroglu et al., 2015)

T
and Bacillus pumilus (M27) (Adiguzel et al., 2014) was reported in clarification of

IP
peach and orange, apricot, grape, and apple juices, respectively.

R
12.3. Feed sector

SC
Improved nutrient utilization, feed conversion efficacy, higher egg production,

improved growth performance and immunity, increased ileum protein digestibility,

NU
and reduced uric acid excretion were reported after usage of mannanases in poultry
MA
feed (Lee et al., 2003; Wu et al., 2005; Zou et al., 2006). Supplementation of multi-

enzyme complex (Natuzyme® and Hemicell®) which included endo-β-mannanase, to


D

fish feed significantly improved growth performance and feed utilization in Caspian
TE

salmon (Salmotrutta caspius) (Ali Zamini et al., 2014). Supplementation of β-

mannanase (480U/kg dry matter) to feed rich in mannan (palm kernel meal, copra
P

meal, soybean hull, etc.) stimulated growth, nitrogen utilization, and feed efficiency in
CE

growing Korean native goats (Lee et al., 2014).


AC

12.4. Detergent sector

Mannanases have also been used in detergent formulations for the removal of stains

containing mannan gums which otherwise are difficult to clean due to their ability to

get adsorbed onto cellulose fibres (Srivastava and Kapoor, 2014).

12.5. Saccharification and bioethanol

Mannan degrading enzymes can be employed synergistically for converting

lignocellulosic polysaccharides into fermentable sugars which can be subsequently

used for ethanolic fermentation (Galbe and Zacchi, 2002; Chandel et al., 2015).

Jørgensen et al. (2010) have shown the utilization of palm kernel cake (PKC) for the

31
ACCEPTED MANUSCRIPT

production of fermentable sugars by using a cocktail of xylanase and mannanase. A

crude solution of endo-β-mannanase and xylanase was shown to produce xylose and

reducing sugar from aqueous ammonia pre-treated corn cob powder (Zhang and Sang,

T
2015). Supplementation of fungal β-mannanase and β-mannosidase to the

IP
Talaromyces cellulolyticus cellulase system ameliorated the hydrolysis of softwood

R
(ball-milling-treated Douglas fir) (Inoue et al., 2015). P. anserina mannanases,

SC
PaMan5A (GH 5) and PaMan26A (GH 26), were cloned and expressed in Pichia

pastoris for their utilization in the enzymatic hydrolysis (saccharification) of

lignocellulosic biomass (spruce)


NU
by T. reesei. Results obtained showed
MA
supplementation of T. reesei industrial xylanase-rich enzyme cocktail (E508) with a

combination of PaMan5A and PaMan26A was better in releasing sugars (34%) as


D

compared to PaMan5A (28%) or PaMan26A (17%) alone (Couturier et al., 2011).


TE

12.6. Oil drilling

In oil drilling operations, now-a-days, guar gum is being used to flood the well along
P

with sand particles followed by pressurizing the bedrock until it fractures. To ease
CE

product flow, thinning of the polymer solution is carried out, and mannanases which
AC

can catalyse the hydrolysis of guar gum at elevated temperatures (>80°C) are proving

useful for this purpose. A β-mannanase from Enterobacter sp. strain N18 has been

used as a gel-breaker in place of chemical gel breakers for low-temperature oil wells

and other industrial fields (You et al., 2016).

13. Concluding remarks and future perspectives

Combinatorial and coherent methodologies like structure-based rational design,

directed evolution and fundamental biochemistry have resulted in the emergence of

endo-β-mannanases to suit the need of both academia and industry. Improvement in

scientific understanding by cutting-edge experimentation in the structure-activity

32
ACCEPTED MANUSCRIPT

relationship, catalytic mechanism, and factors involved in conferring stability to

enzymes would help to create more robust and active endo-β-mannanases so as to aid

hemicellulose hydrolysis under industrial conditions. Mining novel genes from

T
mother-nature using metagenomic libraries and next generation sequencing platforms

IP
can also help to unravel powerful endo-β-mannanases with hitherto unknown

R
properties.

SC
Acknowledgements

Authors thank Prof. Ram Rajashekharan, Director, CSIR-CFTRI, for constant

NU
encouragement. The support obtained under project MLP0116 is gratefully
MA
acknowledged. P.K.S thanks, University Grants Commission (UGC), New Delhi,

India, for Junior and Senior Research Fellowship.


D

References
TE

Adiguzel, A., Nadaroglu, H., Adiguzel, G., 2014. Purification and characterization of

β-mannanase from Bacillus pumilus (M27) and its applications in some fruit juices. J.
P

Food Sci. Technol. 52(8), 5292-5298.


CE

Albuquerque, P.B., Barros, W., Santos, G.R., Correia, M.T., Mourão, P.A., Teixeira,
AC

J.A., Carneiro-da-Cunha, M.G., 2014. Characterization and rheological study of the

galactomannan extracted from seeds of Cassia grandis. Carbohydr. Pol. 104, 127-

134.

Ali Zamini, A., Kanani, H.G., Azam Esmaeili, A., Ramezani, S., Zoriezahra, S.J.,

2014. Effects of two dietary exogenous multi-enzyme supplementation, Natuzyme®

and beta-mannanase (Hemicell®), on growth and blood parameters of Caspian salmon

(Salmo trutta caspius). Comp. Clin. Path. 23, 187-192.

33
ACCEPTED MANUSCRIPT

Ardèvol, A., Biarnés, X., Planas, A., Rovira, C., 2010. The conformational free-

energy landscape of β-D-mannopyranose: evidence for a 1S5→ B2,5→ OS2 catalytic

itinerary in β-mannosidases. J. Am. Chem. Soc. 132(45), 16058-16065.

T
Aspinall, G.O., 1959. Structural chemistry of the hemicelluloses. Adv. Carbohydr.

IP
Chem. 14, 429-468.

R
Bailey, R.W., Pain, V., 1971. Polysaccharide mannose in New Zealand

SC
ferns. Phytochemistry 10(5), 1065-1073.

Barrios-González, J., 2012. Solid-state fermentation: Physiology of solid medium, its

NU
molecular basis and applications. Process Biochem. 47, 175-185.
MA
Beg, Q., Kapoor, M., Mahajan, L., Hoondal, G.S., 2001. Microbial xylanases and

their industrial applications: a review. Appl. Microbiol. Biotechnol. 56, 326-338.


D

Beg, Q.K., Bhushan, B., Kapoor, M., Hoondal, G.S., 2000. Effect of amino acids on
TE

production of xylanase and pectinase from Streptomyces sp. QG-11-3. World J.

Microbiol. Biotechnol. 16, 211-213.


P

Blake, A.W., McCartney, L., Flint, J.E., Bolam, D.N., Boraston, A.B., Gilbert, H.J.,
CE

Knox, J.P., 2006. Understanding the biological rationale for the diversity of cellulose-
AC

directed carbohydrate-binding modules in prokaryotic enzymes. J. Biol. Chem. 281,

29321-29329.

Blibech, M., Chaari, F., Bhiri, F., Dammak, I., Ghorbel, R.E., Chaabouni, S.E., 2011.

Production of manno-oligosaccharides from locust bean gum using immobilized

Penicillium occitanis mannanase. J. Mol. Catal. B Enzym. 73, 111-115.

Blibech, M., Ghorbel, R.E., Fakhfakh, I., Ntarima, P., Piens, K., Bacha, A.B.,

Chaabouni, S.E., 2010. Purification and characterization of a low molecular weight

of β-mannanase from Penicillium occitanis Pol6. Appl. Biochem. Biotechnol. 160,

1227-1240.

34
ACCEPTED MANUSCRIPT

Bolam, D.N., Hughes, N., Virden, R., Lakey, J.H., Hazlewood, G.P., Henrissat, B.,

Braithwaite, K.L., Gilbert, H.J., 1996. Mannanase A from Pseudomonas fluorescens

ssp. cellulosa is a retaining glycosyl hydrolase in which E212 and E320 are the

T
putative catalytic residues. Biochemistry 35, 16195-16204.

IP
Bourgault, R., Oakley, A.J., Bewley, J.D., Wilce, M.C.J., 2005. Three-dimensional

R
structure of (1,4)-β-D-mannan mannanohydrolase from tomato fruit. Protein Sci. 14,

SC
1233-1241.

Broda, P., 1992. Biotechnology in the degradation and utilization of lignocellulose.

Biodegradation 3, 219-238.
NU
MA
Buriti, F.C.A., Dos Santos, K.M.O., Sombra, V.G., Maciel, J.S., Teixeira Sá, D.M.A,

Salles, H.O., Oliveira, G., de Paula, R.C.M., Feitosa, J.P.A, Monteiro Moreira,
D

A.C.O., Moreira, R.A., Egito, A.S., 2014. Characterisation of partially hydrolysed


TE

galactomannan from Caesalpinia pulcherrima seeds as a potential dietary fibre. Food

Hydrocoll. 35, 512-521.


P

Busch, V.M., Kolender, A.A., Santagapita, P.R., Buera, M.P., 2015. Vinal gum, a
CE

galactomannan from Prosopis ruscifolia seeds: Physicochemical characterization.


AC

Food Hydrocoll. 51, 495-502.

Cai, H., Shi, P., Huang, H., Luo, H., Bai, Y., Yang, P., Meng, K., Yao, B., 2011. An

acidic β-mannanase from Penicillium sp. C6: Gene cloning and over-expression in

Pichia pastoris. World J. Microbiol. Biotechnol. 27, 2813-2819.

Campos-Vega, R., Loarca-Piña, G., Vergara-Castañeda, H., Oomah, B.D., 2015.

Spent coffee grounds: A review on current research and future prospects. Trends Food

Sci. Technol. 45(1), 24-36.

Cann, I.K.O., Kocherginskaya, S., Michael, R., White, B.A., Mackie, R.I., 1999.

Molecular cloning, sequencing, and expression of a novel multidomain mannanase

35
ACCEPTED MANUSCRIPT

gene from Thermoanaerobacterium polysaccharolyticum. J. Bacteriol. 181, 1643-

1651.

Cartier, N., Chambat, G., Joseleau, J.P., 1988. Cell wall and extracellular

T
galactoglucomannans from suspension-cultured Rubus fruticosus cells.

IP
Phytochemistry 27(5), 1361-1364.

R
Cartmell, A., Topakas, E., Ducros, V.M., Suits, M.D., Davies, G.J., Gilbert, H.J.,

SC
2008. The Cellvibrio japonicus mannanase CjMan26C displays a unique exo-mode of

action that is conferred by subtle changes to the distal region of the active site. J. Biol.

Chem. 283, 34403-34413.


NU
MA
Cerqueira, M.A., Souza, B.W.S., Simões, J., Teixeira, J.A., Domingues, M.R.M.,

Coimbra, M.A., Vicente, A.A., 2011. Structural and thermal characterization of


D

galactomannans from non-conventional sources. Carbohydr. Pol. 83(1), 179-185.


TE

Chandel, A.K., Gonçalves, B.C., Strap, J.L., da Silva, S.S., 2015. Biodelignification

of lignocellulose substrates: An intrinsic and sustainable pretreatment strategy for


P

clean energy production. Crit. Rev. Biotechnol. 35(3), 281-293.


CE

Chantorn, S.T., Buengsrisawat, K., Pokaseam, A., Sombat, T., 2013. Optimization of
AC

extracellular mannanase production from Penicillium oxalicum KUB-SN2-1 and

application for hydrolysis property. Songklanakarin J. Sci. Technol. 35, 17-22.

Chauhan, P.S., Gupta, N., 2016. Insight into microbial mannosidases: a review. Crit.

Rev. Biotechnol. 1-12.

Chauhan, P.S., Bharadwaj, A., Puri, N., Gupta, N., 2014a. Optimization of medium

composition for alkali-thermostable mannanase production by Bacillus nealsonii PN-

11 in submerged fermentation. Int. J. Curr. Microbiol. Appl. Sci. 3, 1033-1045.

36
ACCEPTED MANUSCRIPT

Chauhan, P.S., Puri, N., Sharma, P., Gupta, N., 2012. Mannanases: Microbial sources,

production, properties and potential biotechnological applications. Appl. Microbiol.

Biotechnol. 93, 1817-1830.

T
Chauhan, P.S., Sharma, P., Puri, N., Gupta, N., 2014b. Purification and

IP
characterization of an alkali-thermostable β-mannanase from Bacillus nealsonii PN-

R
11 and its application in mannooligosaccharides preparation having prebiotic

SC
potential. Eur. Food Res. Technol. 238, 927-936.

Chauhan, P.S., Tripathi, S.P., Sangamwar, A.T., Puri, N., Sharma, P., Gupta, N.,

NU
2015. Cloning, molecular modeling, and docking analysis of alkali-thermostable β-
MA
mannanase from Bacillus nealsonii PN-11. Appl. Microbiol. Biotechnol. 99, 8917-

8925.
D

Chen, X., Cao, Y., Ding, Y., Lu, W., Li, D., 2007. Cloning, functional expression and
TE

characterization of Aspergillus sulphureus β-mannanase in Pichia pastoris. J.

Biotechnol. 128, 452-461.


P

Chen, X., Qiao, J., Yu, H., Cao, Y., 2009. Overexpression of an optimized Aspergillus
CE

sulphureus β-mannanase gene in Pichia pastoris. Biologia 64, 235-238.


AC

Cho, K.M., Hong, S.Y., Lee, S.M., Kim, Y.H., Kahng, G.G., Kim, H., Yun, H.D.,

2006. A cel44C-man26A gene of endophytic Paenibacillus polymyxa GS01 has

multi-glycosyl hydrolases in two catalytic domains. Appl. Microbiol. Biotechnol. 73,

618-630.

Couturier, M., Feliu, J., Bozonnet, S., Roussel, A., Berrin, J.G., 2013a. Molecular

engineering of fungal GH5 and GH26 β-(1, 4)-mannanases toward improvement of

enzyme activity. PloS one 8(11), e79800.

Couturier, M., Haon, M., Coutinho, P.M., Henrissat, B., Lesage-Meessen, L., Berrin,

J.G., 2011. Podospora anserina hemicellulases potentiate the Trichoderma reesei

37
ACCEPTED MANUSCRIPT

secretome for saccharification of lignocellulosic biomass. Appl. Environ.

Microbiol. 77(1), 237-246.

Couturier, M., Roussel, A., Rosengren, A., Leone, P., Stålbrand, H., Berrin, J.G.,

T
2013b. Structural and biochemical analyses of glycoside hydrolase families 5 and 26

IP
β-(1,4)-mannanases from Podospora anserina reveal differences upon manno-

R
oligosaccharide catalysis. J. Biol. Chem. 288, 14624-14635.

SC
Da Silva, V.M., Colussi, F., De Oliveira Neto, M., Braz, A.S.K., Squina, F.M.,

Oliveira, C.L.P., Garcia, W., 2014. Modular hyperthermostable bacterial endo-β-1,4-

NU
mannanase: Molecular shape, flexibility and temperature-dependent conformational
MA
changes. PLoS One 9 (3), e92996.

Davies, G.J., Wilson, K.S., Henrissat, B., 1997. Nomenclature for sugar-binding
D

subsites in glycosyl hydrolases. Biochem. J. 321, 557-559.


TE

Dhawan, S., Kaur, J., 2007. Microbial mannanases: an overview of production and

applications. Crit. Rev. Biotechnol. 27, 197-216.


P

Dias, F.M.V., Vincent, F., Pell, G., Prates, J.A.M., Centeno, M.S.J., Tailford, L.E.,
CE

Ferreira, L.M.A., Fontes, C.M.G.A., Davies, G.J., Gilbert, H.J., 2004. Insights into the
AC

molecular determinants of substrate specificity in glycoside hydrolase family 5

revealed by the crystal structure and kinetics of Cellvibrio mixtus mannosidase 5A. J.

Biol. Chem. 279, 25517-25526.

Dilokpimol, A., Nakai, H., Gotfredsen, C.H., Baumann, M.J., Nakai, N., Abou

Hachem, M., Svensson, B., 2011. Recombinant production and characterisation of

two related GH5 endo-β-1,4-mannanases from Aspergillus nidulans FGSC A4

showing distinctly different transglycosylation capacity. Biochim. Biophys. Acta -

Proteins Proteomics 1814, 1720-1729.

38
ACCEPTED MANUSCRIPT

Dirk, L.M.A., Van der Krol, A.R., Vreugdenhil, D., Hilhors, H.W.M., Bewley, J.D.,

1999. Galactomannan, soluble sugar and starch mobilization following germination of

Trigonella foenum-graecum seeds. Plant Physiol. Biochem. 37, 41-50.

T
Dong, Y.H., Li, J.F., Hu, D., Yin, X., Wang, C.J., Tang, S.H., Wu, M.C., 2016.

IP
Replacing a piece of loop-structure in the substrate-binding groove of Aspergillus

R
usamii β-mannanase, AuMan5A, to improve its enzymatic properties by rational

SC
design. Appl. Microbiol. Biotechnol. 100, 3989-3998.

Dos Santos, C.R., Paiva, J.H., Meza, A.N., Cota, J., Alvarez, T.M., Ruller, R., Prade,

NU
R.A., Squina, F.M., Murakami, M.T., 2012. Molecular insights into substrate
MA
specificity and thermal stability of a bacterial GH5-CBM27 endo-1,4-β-D-mannanase.

J. Struct. Biol. 177, 469-476.


D

Ducros, V.M.A., Zechel, D.L., Murshudov, G.N., Gilbert, H.J., Szabó, L., Stoll, D.,
TE

Withers, S.J., Davies, G.J., 2002. Substrate distortion by a β-mannanase: Snapshots of

the Michaelis and covalent intermediate complexes suggest a B2,5 conformation for
P

the transition-state. Angew. Chemie - Int. Ed. 114, 2948-2951.


CE

Duffaud, G.D., McCutchen, C.M., Leduc, P., Parker, K.N., Kelly, R.M., 1997.
AC

Purification and characterization of extremely thermostable β-mannanase, β-

mannosidase, and β-galactosidase from the hyperthermophilic eubacterium

Thermotoga neapolitana 5068. Appl. Environ. Microbiol. 63, 169-177.

Dulson, J., Bewley, J.D., 1989. Mannanase from Lactuca sativa: Metabolic

requirements for production and partial purification. Phytochemistry 28, 363-369.

Ekholm, F.S., Ardá, A., Eklund, P., André, S., Gabius, H.J., Jiménez‐Barbero, J.,

Leino, R., 2012. Studies related to Norway spruce galactoglucomannans: chemical

synthesis, conformation analysis, NMR spectroscopic characterization, and molecular

recognition of model compounds. Chem. Eur. J. 18(45), 14392-14405.

39
ACCEPTED MANUSCRIPT

Eneyskaya, E.V., Sundqvist, G., Golubev, A.M., Ibatullin, F.M., Ivanen, D.R.,

Shabalin, K.A., Brumer, H., Kulminskaya, A.A., 2009. Transglycosylating and

hydrolytic activities of the β-mannosidase from Trichoderma reesei. Biochimie 91,

T
632-638.

IP
Ethier, N., Talbot, G., Sygusch, J., 1998. Gene cloning, DNA sequencing, and

R
expression of thermostable β-mannanase from Bacillus stearothermophilus. Appl.

SC
Environ. Microbiol. 64, 4428-4432.

Fernández, P.V., Estevez, J.M., Cerezo, A.S., Ciancia, M., 2012. Sulfated β-D-

NU
mannan from green seaweed Codium vermilara. Carbohydr. Pol. 87(1), 916-919.
MA
Ferreira, S.A., Oslakovic, C., Cukalevski, R., Frohm, B., Dahlbäck, B., Linse, S.,

Gama, F.M., Cedervall, T., 2012. Biocompatibility of mannan nanogel—safe


D

interaction with plasma proteins. Biochim. Biophys. Acta - Gen. Subj. 1820, 1043-
TE

1051.

Filho, E.X.F., 1998. Hemicellulases and biotechnology, in: Pandalai, S.G., (ed)
P

Recent research developments in microbiology. Research Signpost, Trivandrum, pp.


CE

165-176.
AC

Flari, V., Matoub, M., Rouland, C., 1995. Purification and characterization of a β-

mannanase from the digestive tract of the edible snail Helix lucorum L. Carbohydr.

Res. 275, 207-213.

Fontes, C.M., Gilbert, H.J., 2010. Cellulosomes: highly efficient nanomachines

designed to deconstruct plant cell wall complex carbohydrates. Annu. Rev.

Biochem. 79, 655-681.

Freiesleben, V.P., Spodsberg, N., Blicher, T.H., Anderson, L., Jørgensen, H.,

Stålbrand, H., Meyer, A.S., Krogh, K.B., 2016. An Aspergillus nidulans GH26 endo-

40
ACCEPTED MANUSCRIPT

β-mannanase with a novel degradation pattern on highly substituted

galactomannans. Enzyme Microb. Technol. 83, 68-77.

Galbe, M., Zacchi, G., 2002. A review of the production of ethanol from

T
softwood. Appl. Microbiol. Biotechnol. 59(6), 618-628.

IP
Ghosh, A., Luís, A.S., Brás, J.L.A., Fontes, C.M.G.A., Goyal, A., 2013. Thermostable

R
recombinant β-(1→4)-mannanase from C. thermocellum: biochemical

SC
characterization and manno-oligosaccharides production. J. Agric. Food Chem. 61,

NU
12333-12344.

Ghosh, A., Verma, A.K., Gautam, S., Gupta, M.N., Goyal, A., 2014a. Structure and
MA
functional investigation of ligand binding by a family 35 carbohydrate binding

module (CTCBM35) of β-mannanase of family 26 glycoside hydrolase from


D

Clostridium thermocellum. Biochemistry (Moscow) 79, 672-686.


TE

Ghosh, A., Verma, A.K., Tingirikari, J.R., Shukla, R., Goyal, A., 2014b. Recovery

and purification of oligosaccharides from copra meal by recombinant endo-β-


P
CE

mannanase and deciphering molecular mechanism involved and its role as potent

therapeutic agent. Mol. Biotechnol. 57, 111-127.


AC

Gibbs, M.D., Reeves, R.A., Sunna, A., Bergquist, P.L., 1999. Sequencing and

expression of a β-mannanase gene from the extreme thermophile Dictyoglomus

thermophilum Rt46B.1, and characteristics of the recombinant enzyme. Curr.

Microbiol. 39, 351-357.

Gibbs, M.D., Saul, D.J., Luthi, E., Bergquist, P.L., 1992. The β-mannanase from

―Caldocellum saccharolyticum‖ is part of a multidomain enzyme. Appl. Environ.

Microbiol. 58, 3864-3867.

41
ACCEPTED MANUSCRIPT

Gilbert, H.J., Stålbrand, H., Brumer, H., 2008. How the walls come crumbling down:

recent structural biochemistry of plant polysaccharide degradation. Curr. Opin. Plant

Biol. 11(3), 338-348.

T
Gonçalves, A.M.D., Silva, C.S., Madeira, T.I., Coelho, R., De Sanctis, D., San

IP
Romão, M.V., Bento, I., 2012. Endo-β-D-1,4-mannanase from Chrysonilia sitophila

R
displays a novel loop arrangement for substrate selectivity. Acta Crystallogr. Sect. D

SC
Biol. Crystallogr. 68, 1468-1478.

Guo, N., Zheng, J., Wu, L.S., Tian, J., Zhou, H.B., 2013. Engineered bifunctional

NU
enzymes of endo-1,4-β-xylanase/endo-1,4- β-mannanase were constructed for
MA
synergistically hydrolyzing hemicellulose. J. Mol. Catal. B Enzym. 97, 311-318.

Guo, R., Ai, L., Cao, N., Ma, J., Wu, Y., Wu, J., Sun, X., 2016. Physicochemical
D

properties and structural characterization of a galactomannan from Sophora


TE

alopecuroides L. seeds. Carbohydr. Pol. 140, 451-460.

Gupta, S., Saurabh, C.K., Variyar, P.S., Sharma, A., 2015. Comparative analysis of
P

dietary fiber activities of enzymatic and gamma depolymerized guar gum. Food
CE

Hydrocoll. 48, 149-154.


AC

Hägglund, P., Eriksson, T., Collén, A., Nerinckx, W., Claeyssens, M., Stålbrand, H.,

2003. A cellulose-binding module of the Trichoderma reesei β-mannanase Man5A

increases the mannan-hydrolysis of complex substrates. J. Biotechnol. 101(1), 37-48.

Halmer, P., 1989. De novo synthesis of mannanase by the endosperm of Lactuca

sativa. Phytochemistry 28, 371-378.

Halstead, J.R., Vercoe, P.E., Gilbert, H.J., Davidson, K., Hazlewood, G.P., 1999. A

family 26 mannanase produced by Clostridium thermocellum as a component of the

cellulosome contains a domain which is conserved in mannanases from anaerobic

fungi. Microbiology 145(11), 3101-3108.

42
ACCEPTED MANUSCRIPT

Harmayani, E., Aprilia, V., Marsono, Y., 2014. Characterization of glucomannan

from Amorphophallus oncophyllus and its prebiotic activity in vivo. Carbohydr. Pol.

112, 475-479.

T
Hatada, Y., Takeda, N., Hirasawa, K., Ohta, Y., Usami, R., Yoshida, Y., Grant, W.D.,

IP
Ito, S., Horikoshi, K., 2005. Sequence of the gene for a high-alkaline mannanase from

R
an alkaliphilic Bacillus sp. strain JAMB-750, its expression in Bacillus subtilis and

SC
characterization of the recombinant enzyme. Extremophiles 9, 497-500.

Hekmat, O., Lo Leggio, L., Rosengren, A., Kamarauskaite, J., Kolenova, K.,

NU
Stålbrand, H., 2010. Rational engineering of mannosyl binding in the distal glycone
MA
subsites of Cellulomonas fimi endo-β-1,4-mannanase: Mannosyl binding promoted at

subsite -2 and demoted at subsite -3. Biochemistry 49, 4884-4896.


D

Hervé, C., Rogowski, A., Blake, A.W., Marcus, S.E., Gilbert, H.J., Knox, J.P., 2010.
TE

Carbohydrate-binding modules promote the enzymatic deconstruction of intact plant

cell walls by targeting and proximity effects. Proc. Natl. Acad. Sci. U.S.A. 107(34),
P

15293-15298.
CE

Hilge, M., Gloor, S.M., Rypniewski, W., Sauer, O., Heightman, T.D., Zimmermann,
AC

W., Winterhalter, K., Piontek, K., 1998. High-resolution native and complex

structures of thermostable β-mannanase from Thermomonospora fusca - substrate

specificity in glycosyl hydrolase family 5. Structure 6, 1433-1444.

Hogg, D., Pell, G., Dupree, P., Goubet, F., Martín-Orúe, S.M., Armand, S., Gilbert,

H.J., 2003. The modular architecture of Cellvibrio japonicus mannanases in glycoside

hydrolase families 5 and 26 points to differences in their role in mannan degradation.

Biochem. J. 371, 1027-1043.

43
ACCEPTED MANUSCRIPT

Hogg, D., Woo, E.J., Bolam, D.N., McKie, V.A., Gilbert, H.J., Pickersgill, R.W.,

2001. Crystal structure of mannanase 26A from Pseudomonas cellulosa and analysis

of residues involved in substrate binding. J. Biol. Chem. 276, 31186-31192.

T
Huang, J.W., Chen, C.C., Huang, C.H., Huang, T.Y., Wu, T.H., Cheng, Y.S., Ko,

IP
T.P., Lin, C.Y., Liu, J.R., Guo, R.T., 2014. Improving the specific activity of β-

R
mannanase from Aspergillus niger BK01 by structure-based rational design. Biochim.

SC
Biophys. Acta - Proteins Proteomics 1844, 663-669.

Inoue, H., Yano, S., Sawayama, S., 2015. Effect of β-mannanase and β-mannosidase

NU
supplementation on the total hydrolysis of softwood polysaccharides by the
MA
Talaromyces cellulolyticus cellulase system. Appl. Biochem. Biotechnol. 176, 1673-

1686.
D

Ishurd, O., Kermagi, A., Elghazoun, M., Kennedy, J.F., 2006. Structural of a
TE

glucomannan from Lupinus varius seed. Carbohydr. Pol. 65(4), 410-413.

Jansson, M., Danielsson, S., Saadatmand, S., Edlund, U., Albertsson, A.-C., 2014.
P

Upgrading of wood pre-hydrolysis liquor for renewable barrier design: a techno-


CE

economic consideration. Cellulose 21, 2045-2062.


AC

Jeon, S.D., Yu, K.O., Kim, S.W., Han, S.O., 2011. A celluloytic complex from

Clostridium cellulovorans consisting of mannanase B and endoglucanase E has

synergistic effects on galactomannan degradation. Appl. Microbiol. Biotechnol. 90,

565-572.

Jiang, Z., Wei, Y., Li, D., Li, L., Chai, P., Kusakabe, I., 2006. High-level production,

purification and characterization of a thermostable β-mannanase from the newly

isolated Bacillus subtilis WY34. Carbohydr. Pol. 66, 88-96.

44
ACCEPTED MANUSCRIPT

Jørgensen, H., Sanadi, A.R., Felby, C., Lange, N.E.K., Fischer, M., Ernst, S., 2010.

Production of ethanol and feed by high dry matter hydrolysis and fermentation of

palm kernel press cake. Appl. Biochem. Biotechnol. 161, 318-332.

T
Kaira, G.S., Panwar, D., Kapoor, M., 2016. Recombinant endo-mannanase (ManB-

IP
1601) production using agro-industrial residues: Development of economical medium

R
and application in oil extraction from copra. Bioresour. Technol. 209, 220-227.

SC
Katrolia, P., Zhou, P., Zhang, P., Yan, Q., Li, Y., Jiang, Z., Xu, Z., 2012. High level

NU
expression of a novel β-mannanase from Chaetomium sp. exhibiting efficient mannan

hydrolysis. Carbohydr. Pol. 87, 480-490.


MA
Katrolia, P., Yan, Q., Zhang, P., Zhou, P., Yang, S., Jiang, Z., 2013. Gene cloning and

enzymatic characterization of an alkali-tolerant endo-1,4-β-mannanase from


D

Rhizomucor miehei. J. Agric. Food Chem. 61, 394-401.


TE

Kim, D.Y., Ham, S.J., Lee, H.J., Cho, H.Y., Kim, J.H., Kim, Y.J., Shin, D.H., Rhee,

Y.H., Son, K.H., Park, H.Y., 2011a. Cloning and characterization of a modular
P
CE

GH5 β-1,4-mannanase with high specific activity from the fibrolytic bacterium

Cellulosimicrobium sp. strain HY-13. Bioresour. Technol. 102, 9185-9192.


AC

Kim, D.Y., Ham, S.J., Lee, H.J., Kim, Y.J., Shin, D.H., Rhee, Y.H., Son, K.H., Park,

H.Y., 2011b. A highly active endo-β-1,4-mannanase produced by Cellulosimicrobium

sp. strain HY-13, a hemicellulolytic bacterium in the gut of Eisenia fetida. Enzyme

Microb. Technol. 48, 365-370.

Kim, M.K., An, Y.J., Jeong, C.S., Song, J.M., Kang, M.H., Lee, Y.H., Cha, S.S. 2013.

Expression at 279 K, purification, crystallization and preliminary X-ray

crystallographic analysis of a novel cold-active β-1, 4-D-mannanase from the

Antarctic springtail Cryptopygus antarcticus. Acta Crystallogr. Sect. F Struct. Biol.

Cryst. Commun. 69(9), 1007-1010.

45
ACCEPTED MANUSCRIPT

Kisonen, V., Prakobna, K., Xu, C., Salminen, A., Mikkonen, K.S., Valtakari, D.,

Eklund, P., Seppälä, J., Tenkanen, M., Willför, S., 2015. Composite films of

nanofibrillated cellulose and O-acetyl galactoglucomannan (GGM) coated with

T
succinic esters of GGM showing potential as barrier material in food packaging. J.

IP
Mater. Sci. 50(8), 3189-3199.

R
Kučerová, D., Kollárová, K., Zelko, I., Vatehová, Z., Lišková, D., 2014.

SC
Galactoglucomannan oligosaccharides alleviate cadmium stress in Arabidopsis. J.

Plant Physiol. 171(7), 518-524.

NU
Kuhad, R.C., Kapoor, M., 2015. Method of producing alkalothermostable xylanase
MA
from Bacillus pumilus MK001 by solid state fermentation. Patent, India, IN 984 DEL

2008.
D

Kuhad, R.C., Singh, A., Eriksson, K.E.L., 1997. Microorganisms and enzymes
TE

involved in the degradation of plant fiber cell walls, in: Eriksson, K.-E.L.,

(Ed.), Biotechnology in the pulp and paper industry. Springer, Berlin Heidelberg, pp.
P

45-125.
CE

Kulcinskaja, E., Rosengren, A., Ibrahim, R., Kolenová, K., Stålbrand, H., 2013.
AC

Expression and characterization of a Bifidobacterium adolescentis β-mannanase

carrying mannan-binding and cell association motifs. Appl. Environ.

Microbiol. 79(1), 133-140.

Kumagai, Y., Kawakami, K., Mukaihara, T., Kimura, M., Hatanaka, T., 2012. The

structural analysis and the role of calcium binding site for thermal stability in

mannanase. Biochimie 94, 2783-2790.

Kumagai, Y., Kawakami, K., Uraji, M., Hatanaka, T., 2013a. Effect of the binding of

bivalent ion to the calcium-binding site responsible for the thermal stability of

46
ACCEPTED MANUSCRIPT

actinomycete mannanase: Potential use in production of functional

mannooligosaccharides. J. Mol. Catal. B Enzym. 94, 63-68.

Kumagai, Y., Kawakami, K., Uraji, M., Hatanaka, T., 2013b. Binding of bivalent ions

T
to actinomycete mannanase is accompanied by conformational change and is a key

IP
factor in its thermal stability. Biochim. Biophys. Acta - Proteins Proteomics 1834,

R
301-307.

SC
Kumagai, Y., Yamashita, K., Tagami, T., Uraji, M., Wan, K., Okuyama, M., Yao, M.,

Kimura, A., Hatanaka, T., 2015. The loop structure of Actinomycete glycoside

NU
hydrolase family 5 mannanases governs substrate recognition. FEBS J. 282, 4001-
MA
4014.

Kurakake, M., Sumida, T., Masuda, D., Oonishi, S., Komaki, T., 2006. Production of

galacto-manno-oligosaccharides from guar gum by β-mannanase from Penicillium


D
TE

oxalicum SO. J. Agric. Food Chem. 54, 7885-7889.

Larsson, A.M., Anderson, L., Xu, B., Muñoz, I.G., Usón, I., Janson, J.C., Stålbrand,
P

H., Ståhlberg, J., 2006. Three-dimensional crystal structure and enzymic


CE

characterization of β-mannanase Man5A from blue mussel Mytilus edulis. J. Mol.


AC

Biol. 357, 1500-1510.

Le Nours, J., Anderson, L., Stoll, D., Stålbrand, H., Lo Leggio, L., 2005. The

structure and characterization of a modular endo-β-1,4-mannanase from Cellulomonas

fimi. Biochemistry 44, 12700-12708.

Lee, J.-J., Seo, J., Jung, J.K., Lee, J., Lee, J.-H., Seo, S., 2014. Effects of β-mannanase

supplementation on growth performance, nutrient digestibility, and nitrogen

utilization of Korean native goat (Capra hircus coreanae). Livest. Sci. 169, 83-87.

47
ACCEPTED MANUSCRIPT

Lee, J.T., Bailey, C.A., Cartwright, A.L., 2003. β-Mannanase ameliorates viscosity-

associated depression of growth in broiler chickens fed guar germ and hull fractions.

Poult. Sci. 82, 1925-1931.

T
Lehtonen, M., Teräslahti, S., Xu, C., Yadav, M.P., Lampi, A.M., Mikkonen, K.S.,

IP
2016. Spruce galactoglucomannans inhibit lipid oxidation in rapeseed oil-in-water

R
emulsions. Food Hydrocoll. 58, 255-266.

SC
Li, H., Liu, Z., Wang, C., Huang, S., Zhao, M., 2015. Secretory expression and

characterization of a novel thermo-stable, salt-tolerant endo-1,4-β-mannanase of

NU
Bacillus subtilis WD23 by Pichia pastoris. Eur. Food Res. Technol. 240, 671-677.
MA
Li, J., Wei, X., Tang, C., Wang, J., Zhao, M., Pang, Q., Wu, M., 2014. Directed

modification of the Aspergillus usamii β-mannanase to improve its substrate affinity


D

by in silico design and site-directed mutagenesis. J. Ind. Microbiol. Biotechnol. 41,


TE

693-700.

Li, J.F., Zhao, S.G., Tang, C.D., Wang, J.Q., Wu, M.C., 2012. Cloning and functional
P

expression of an acidophilic β-mannanase gene (Anman5A) from Aspergillus niger


CE

LW-1 in Pichia pastoris. J. Agric. Food Chem. 60, 765-773.


AC

Li, J.-M., Nie, S.-P., 2015. The functional and nutritional aspects of hydrocolloids in

foods. Food Hydrocoll. 53, 46-61.

Li, Y., Hu, F., Wang, X., Cao, H., Liu, D., Yao, D., 2013. A rational design for

trypsin-resistant improvement of Armillariella tabescens β-mannanase MAN47 based

on molecular structure evaluation. J. Biotechnol. 163(4), 401-407.

Liang, H., Ye, T., Zhou, B., Li, J., He, L., Li, Y., Liu, S., Chen, Y., Li, B., 2015.

Fabrication of gastric floating controlled release tablet based on konjac glucomannan.

Food Res. Int. 72, 47-53.

48
ACCEPTED MANUSCRIPT

Liao, H., Li, S., Zheng, H., Wei, Z., Liu, D., Raza, W., Shen, Q., Xu, Y., 2014. A new

acidophilic thermostable endo-1,4-β-mannanase from Penicillium oxalicum GZ-2:

cloning, characterization and functional expression in Pichia pastoris. BMC

T
Biotechnol. 14, 90.

IP
Liepman, A.H., Nairn, C.J., Willats, W.G., Sørensen, I., Roberts, A.W., Keegstra, K.,

R
2007. Functional genomic analysis supports conservation of function among cellulose

SC
synthase-like a gene family members and suggests diverse roles of mannans in

plants. Plant Physiol. 143(4), 1881-1893.

NU
Lin, S.S., Dou, W.F., Xu, H.Y., Li, H.Z., Xu, Z.H., Ma, Y.H., 2007. Optimization of
MA
medium composition for the production of alkaline β-mannanase by alkaliphilic

Bacillus sp. N16-5 using response surface methodology. Appl. Microbiol. Biotechnol.
D

75, 1015-1022.
TE

Liu, H.-X., Gong, J.-S., Li, H., Lu, Z.-M., Li, H., Qian, J.-Y., Xu, Z.-H., Shi, J.-S.,

2015. Biochemical characterization and cloning of an endo-1,4-β-mannanase from


P

Bacillus subtilis YH12 with unusually broad substrate profile. Process Biochem. 50,
CE

712-721.
AC

Liu, Q., Yang, P., Luo, H., Shi, P., Huang, H., Meng, K., Yao, B., 2012. A novel

endo-1,4-β-mannanase from Bispora antennata with good adaptation and stability

over a broad pH range. Appl. Biochem. Biotechnol. 166, 1442-1453.

Lu, H., Luo, H., Shi, P., Huang, H., Meng, K., Yang, P., Yao, B., 2014. A novel

thermophilic endo-β-1,4-mannanase from Aspergillus nidulans XZ3: Functional roles

of carbohydrate-binding module and Thr/Ser-rich linker region. Appl. Microbiol.

Biotechnol. 98, 2155-2163.

Luo, H., Wang, Y., Wang, H., Yang, J., Yang, Y., Huang, H., Yang, P., Bai, Y., Shi,

P., Fan, Y., Yao, B., 2009. A novel highly acidic β-mannanase from the acidophilic

49
ACCEPTED MANUSCRIPT

fungus Bispora sp. MEY-1: Gene cloning and overexpression in Pichia pastoris.

Appl. Microbiol. Biotechnol. 82, 453-461.

Maeda, M., Shimahara, H., Sugiyama, N., 1980. Detailed examination of the

T
branched structure of konjac glucomannan. Agric. Biol. Chem. 44(2), 245-252.

IP
Maier, H., Anderson, M., Karl, C., Magnuson, K., Whistler, R.L., 1993. Guar, locust

R
bean, tara, and fenugreek gums, in: BeMiller, J., Whistler, R., (Eds.), Industrial gums:

SC
Polysaccharides and their derivatives. Academic press, New York, pp. 215-218.

Malgas, S., Van Dyk, J.S., Pletschke, B.I., 2015a. A review of the enzymatic

NU
hydrolysis of mannans and synergistic interactions between β-mannanase, β-
MA
mannosidase and α-galactosidase. World J. Microbiol. Biotechnol. 31, 1167-1175.

Malgas, S., Van Dyk, S.J., Pletschke, B.I., 2015b. β-Mannanase (Man26A) and α-

galactosidase (Aga27A) synergism – A key factor for the hydrolysis of


D
TE

galactomannan substrates. Enzyme Microb. Technol. 70, 1-8.

Marraccini, P., Rogers, J.W., Allard, C., André, M.L., Caillet, V., Lacoste, N.,
P

Lausanne, F., Michaux, S., 2001. Molecular and biochemical characterization of


CE

endo-β-mannanases from germinating coffee (Coffea arabica) grains. Planta 213,


AC

296-308.

McCleary, B.V., Clark, A.H., Dea, I.C., Rees, D.A., 1985. The fine structures of carob

and guar galactomannans. Carbohydr. Res. 139, 237-260.

Meier, H., Reid, J.S.G., 1982. Reserve polysaccharides other than starch in higher

plants, in: Loewus, F.A., Tanner, W., (eds), Encyclopedia of Plant. vol. 13A.

Springer, Berlin, pp. 418-471.

Mikkelson, A., Maaheimo, H., Hakala, T.K., 2013. Hydrolysis of konjac

glucomannan by Trichoderma reesei mannanase and endoglucanases Cel7B and

50
ACCEPTED MANUSCRIPT

Cel5A for the production of glucomannooligosaccharides. Carbohydr. Res. 372, 60-

68.

Millward-Sadler, S.J., Hall, J., Black, G.W., Hazlewood, G.P., Gilbert, H.J., 1996.

T
Evidence that the Piromyces gene family encoding endo-1,4-β mannanases arose

IP
through gene duplication. FEMS Microbiol. Lett. 141, 183-188.

R
Mizutani, K., Tsuchiya, S., Toyoda, M., Nanbu, Y., Tominaga, K., Yuasa, K.,

SC
Takahashi, N., Tsuji, A., Mikami, B., 2012. Structure of β-1,4-mannanase from the

common sea hare Aplysia kurodai at 1.05 Å resolution. Acta Crystallogr. Sect. F

NU
Struct. Biol. Cryst. Commun. 68, 1164-1168.
MA
Moreira, L.R.S., Filho, E.X.F., 2008. An overview of mannan structure and mannan-

degrading enzyme systems. Appl. Microbiol. Biotechnol. 79, 165-178.


D

Morrill, J., Kulcinskaja, E., Sulewska, A.M., Lahtinen, S., Stålbrand, H., Svensson,
TE

B., Hachem, M.A., 2015. The GH5 1,4-β-mannanase from Bifidobacterium animalis

subsp. lactis Bl-04 possesses a low-affinity mannan-binding module and highlights


P

the diversity of mannanolytic enzymes. BMC Biochem. 16(1), 26.


CE

Morris, D.D., Reeves, R.A, Gibbs, M.D., Saul, D.J., Bergquist, P., 1995. Correction
AC

of the β-mannanase domain of the celC pseudogene from Caldicellulosiruptor

saccharolyticus and activity of the gene product on kraft pulp. Appl. Environ.

Microbiol. 61, 2262-2269.

Mudgil, D., Barak, S., Khatkar, B.S., 2012a. Effect of enzymatic depolymerization on

physicochemical and rheological properties of guar gum. Carbohydr. Pol. 90, 224-

228.

Mudgil, D., Barak, S., Khatkar, B.S., 2012b. X-ray diffraction, IR spectroscopy and

thermal characterization of partially hydrolyzed guar gum. Int. J. Biol

Macromol. 50(4), 1035-1039.

51
ACCEPTED MANUSCRIPT

Nadaroglu, H., Adiguzel, A., Adiguzel, G., 2015. Purification and characterisation of

β-mannanase from Lactobacillus plantarum (M24) and its applications in some fruit

juices. Int. J. Food Sci. Technol. 50(5), 1158-1165.

T
Nicolas, P., Raetz, E., Reymond, S., Sauvegeat, J.L., 1998. Hydrolysis of the

IP
galactomannans of coffee extract with immobilized β-mannanase. Patent, US

R
5714183.

SC
Northcote, D.H., 1972. Chemistry of the plant cell wall. Annu. Rev. Plant. Physiol.

23, 113-132.

NU
Ootsuka, S., Saga, N., Suzuki, K.I., Inoue, A., Ojima, T., 2006. Isolation and cloning
MA
of an endo-β-1,4-mannanase from Pacific abalone Haliotis discus hannai. J.

Biotechnol. 125, 269-280.

Ozturk, B., Cekmecelioglu, D., Ogel, Z.B., 2010. Optimal conditions for enhanced β-
D
TE

mannanase production by recombinant Aspergillus sojae. J. Mol. Catal. B Enzym. 64,

135-139.
P

Pan, L.H., Lu, J., Luo, J.P., Zha, X.Q., Wang, J.H., 2012. Preventive effect of a
CE

galactoglucomannan (GGM) from Dendrobium huoshanense on selenium-induced


AC

liver injury and fibrosis in rats. Exp. Toxicol. Pathol. 64(7), 899-904.

Pan, X., Zhou, J., Tian, A., Le, K., Yuan, H., Xue, Y., Ma, Y., Lu, H., 2011. High

level expression of a truncated β-mannanase from alkaliphilic Bacillus sp. N16-5 in

Kluyveromyces cicerisporus. Biotechnol. Lett. 33, 565-70.

Pérez, J., Muñoz-Dourado, J., de la Rubia, T., Martinez, J., 2002. Biodegradation and

biological treatments of cellulose, hemicellulose and lignin: an overview. Int.

Microbiol. 5, 53-63.

52
ACCEPTED MANUSCRIPT

Petkowicz, C.L.O., Schaefer, S., Reicher, F., 2007. The mannan from Schizolobium

parahybae endosperm is not a reserve polysaccharide. Carbohydr. Pol. 69(4), 659-

664.

T
Pham, T.A., Berrin, J.G., Record, E., To, K.A., Sigoillot, J.C., 2010. Hydrolysis of

IP
softwood by Aspergillus mannanase: Role of a carbohydrate-binding module. J.

R
Biotechnol. 148, 163-170.

SC
Popa, V.I., Spiridon, J., 1998. Hemicelluloses: structure and properties, in: Dumitriu,

S., (Ed.), Polysaccharides: structural diversity and functional versatility. Marcel

Dekker, New York, pp. 297-311.


NU
MA
Pradeep, G.C., Cho, S.S., Choi, Y.H., Choi, Y.S., Jee, J.P., Seong, C.N., Yoo, J.C.,

2016. An extremely alkaline mannanase from Streptomyces sp. CS428 hydrolyzes


D

galactomannan producing series of mannooligosaccharides. World J. Microbiol.


TE

Biotechnol. 32(5), 1-9.

Puchart, V., Vršanská, M., Svoboda, P., Pohl, J., Ögel, Z.B., Biely, P., 2004.
P

Purification and characterization of two forms of endo-β-1,4-mannanase from a


CE

thermotolerant fungus, Aspergillus fumigatus IMI 385708 (formerly Thermomyces


AC

lanuginosus IMI 158749). Biochim. Biophys. Acta - Gen. Subj. 1674, 239-250.

Puls, J., 1993. Chemistry of hemicelluloses: relationship between hemicellulose

structure and enzymes required for hydrolysis, in: Coughlan, M.P., Hazlewood, G.P.,

(Eds.), Hemicellulose and Hemicellulases. Portland, London, pp. 1-27.

Qiao, J., Rao, Z., Dong, B., Cao, Y., 2010. Expression of Bacillus subtilis MA139 β-

mannanase in Pichia pastoris and the enzyme characterization. Appl. Biochem.

Biotechnol. 160, 1362-1370.

Rao, T.P., Hayakawa, M., Minami, T., Ishihara, N., Kapoor, M.P., Ohkubo, T.,

Juneja, L.R., Wakabayashi, K., 2015. Post-meal perceivable satiety and subsequent

53
ACCEPTED MANUSCRIPT

energy intake with intake of partially hydrolysed guar gum. Br. J. Nutr. 113(09),

1489-1498.

Reid, J.S.G., Edwards, M.E., 1995. Galactomannans and other cell wall storage

T
polysaccharides in seeds, in: Food Science and Technology, Marcel Dekker, New

IP
York, pp. 155-155.

R
Ren, Y.F., He, J.Y., Wang, X.F., 2007. Changes in activities of three enzymes

SC
degrading galactomannan during and following rice seed germination. Rice Sci. 14,

295-301.

NU
Rosengren, A., Reddy, S.K., Sjöberg, J.S., Aurelius, O., Logan, D.T., Kolenová, K.,
MA
Stålbrand, H., 2014. An Aspergillus nidulans β-mannanase with high

transglycosylation capacity revealed through comparative studies within glycosidase


D

family 5. Appl. Microbiol. Biotechnol. 98, 10091-10104.


TE

Rungrassamee, W., Kingcha, Y., Srimarut, Y., Maibunkaew, S., 2014.

Mannooligosaccharides from copra meal improves survival of the Pacific white


P

shrimp ( Litopenaeus vannamei ) after exposure to Vibrio harveyi. Aquaculture 434,


CE

403-410.
AC

Sabini, E., Schubert, H., Murshudov, G., Wilson, K.S., Siika-Aho, M., Penttilä, M.,

2000. The three-dimensional structure of a Trichoderma reesei β-mannanase from

glycoside hydrolase family 5. Acta Crystallogr. D Biol. Crystallogr. 56(1), 3-13.

Sachslehner, A., Foidl, G., Foidl, N., Gübitz, G., Haltrich, D., 2000. Hydrolysis of

isolated coffee mannan and coffee extract by mannanases of Sclerotium rolfsii. J.

Biotechnol. 80, 127-134.

Schröder, R., Nicolas, P., Vincent, S.J.F., Fischer, M., Reymond, S., Redgwell, R.J.,

2001. Purification and characterisation of a galactoglucomannan from kiwifruit

(Actinidia deliciosa). Carbohydr. Res. 331(3), 291-306.

54
ACCEPTED MANUSCRIPT

Sébastien, G., Christophe, B., Mario, A., Pascal, L., Michel, P., Aurore, R., 2014.

Impact of purification and fractionation process on the chemical structure and

physical properties of locust bean gum. Carbohydr. Pol. 108, 159-168.

T
Shah, B.R., Li, B., Wang, L., Liu, S., Li, Y., Wei, X., 2015. Health benefits of konjac

IP
glucomannan with special focus on diabetes. Bioact. Carbohydr. Diet. Fibre 5(2), 179-

R
187.

SC
Shi, P., Yuan, T., Zhao, J., Huang, H., Luo, H., Meng, K., Wang, Y., Yao, B., 2011.

Genetic and biochemical characterization of a protease-resistant mesophilic β-

NU
mannanase from Streptomyces sp. S27. J. Ind. Microbiol. Biotechnol. 38, 451-458.
MA
Shimizu, M., Kaneko, Y., Ishihara, S., Mochizuki, M., Sakai, K., Yamada, M.,

Murata, S., Itoh, E., Yamamoto, T., Sugimura, Y., Hirano, T., Takaya, N., Kobayashi,

T., Kato, M., 2015. Novel β-1,4-mannanase belonging to a new glycoside hydrolase
D
TE

family in Aspergillus nidulans. J. Biol. Chem. 290, 27914-27927.

Shoseyov, O., Shani, Z., Levy, I., 2006. Carbohydrate binding modules: biochemical
P

properties and novel applications. Microbiol. Mol. Biol. Rev. 70(2), 283-295.
CE

Simões, J., Nunes, F.M., Domingues, M.D.R.M., Coimbra, M.A., 2010. Structural
AC

features of partially acetylated coffee galactomannans presenting immunostimulatory

activity. Carbohydr. Pol. 79(2), 397-402.

Song, J.M., Nam, K.W., Kang, S.G., Kim, C.G., Kwon, S.T., Lee, Y.H., 2008.

Molecular cloning and characterization of a novel cold-active β-1,4-D-mannanase

from the Antarctic springtail, Cryptopygus antarcticus. Comp. Biochem. Physiol. - B

Biochem. Mol. Biol. 151, 32-40.

Songsiriritthigul, C., Buranabanyat, B., Haltrich, D., Yamabhai, M., 2010. Efficient

recombinant expression and secretion of a thermostable GH26 mannan endo-1,4-β-

mannosidase from Bacillus licheniformis in Escherichia coli. Microb. Cell Fact. 9, 20.

55
ACCEPTED MANUSCRIPT

Sornlake, W., Matetaviparee, P., Rattanaphan, N., Tanapongpipat, S., Eurwilaichitr,

L., 2013. β-Mannanase production by Aspergillus niger BCC4525 and its efficacy on

broiler performance. J. Sci. Food Agric. 93, 3345-3351.

T
Sozzi, G., Cascone, O., Fraschina, A.A., 1996. Effect of a high-temperature stress on

IP
endo-mannanase and α- and β-galactosidase activities during tomato fruit ripening.

R
Postharvest Biol. Technol. 9, 49-63.

SC
Srivastava, M., Kapoor, V.P. 2005. Seed galactomannans: An overview. Chem.

Biodivers. 2(3), 295-317.

NU
Srivastava, P., Appu Rao A.G., Kapoor M., 2014. Structural insights into the thermal
MA
stability of endo-mannanase belonging to family 26 from Bacillus sp. CFR1601.

FASEB J. 580.2 (1 supplement).


D

Srivastava, P.K., Appu Rao, A.G., Kapoor, M., 2016. Metal-dependent thermal
TE

stability of recombinant endo-mannanase (ManB-1601) belonging to family GH 26

from Bacillus sp. CFR1601. Enzyme Microb. Technol. 84, 41-49.


P

Srivastava, P.K., Kapoor, M., 2013. Extracellular endo-mannanase from Bacillus sp.
CE

CFR1601: Economical production using response surface methodology and


AC

downstream processing using aqueous two phase system. Food Bioprod. Process. 91,

672-681.

Srivastava, P.K., Kapoor, M., 2014. Cost-effective endo-mannanase from Bacillus sp.

CFR1601 and its application in generation of oligosaccharides from guar gum and as

detergent additive. Prep. Biochem. Biotechnol. 44, 392-417.

Srivastava, P.K., Kapoor, M., 2015. Recombinant GH-26 endo-mannanase from

Bacillus sp. CFR1601: Biochemical characterization and application in preparation of

partially hydrolysed guar gum. Lebensm. Wiss. Technol. 64, 809-816.

56
ACCEPTED MANUSCRIPT

Srivastava, P.K., Panwar, D., Harish Prashanth, K.V., Kapoor, M., 2015b. Hydrolysis

of locust bean gum by GH-26 endo-mannanase: Generation of un-substituted and

terminally-substituted mannooligosaccharides and their In-vitro fermentability (Under

T
communication).

IP
Stoll, D., Stålbrand, H., Warren, R.A.J., 1999. Mannan-degrading enzymes from

R
Cellulomonas fimi. Appl. Environ. Microbiol. 65, 2598-2605.

SC
Summpunn, P., Chaijan, S., Isarangkul, D., Wiyakrutta, S., Meevootisom, V., 2011.

Characterization, gene cloning, and heterologous expression of β-mannanase from a

NU
thermophilic Bacillus subtilis. J. Microbiol. 49, 86-93.
MA
Sunna, A., 2010. Modular organisation and functional analysis of dissected modular

β-mannanase CsMan26 from Caldicellulosiruptor Rt8B.4. Appl. Microbiol.


D

Biotechnol. 86, 189-200.


TE

Sunna, A., Gibbs, M.D., Chin, C.W.J., Nelson, P.J., Bergquist, P.L., 2000. A gene

encoding a novel multidomain β-1,4-mannanase from Caldibacillus cellulovorans and


P

action of the recombinant enzyme on kraft pulp. Appl. Environ. Microbiol. 66, 664-
CE

670.
AC

Svärd, A., Brännvall, E., Edlund, U., 2015. Rapeseed straw as a renewable source of

hemicelluloses: Extraction, characterization and film formation. Carbohydr. Pol. 133,

179-186.

Tailford, L.E., Ducros, V.M.A, Flint, J.E., Roberts, S.M., Morland, C., Zechel, D.L.,

Smith, N., Bjørnvad, M.E., Borchert, T.V., Wilson, K.S., Davies, G.J., Gilbert, H.J.,

2009. Understanding how diverse β-mannanases recognize heterogeneous substrates.

Biochemistry 48, 7009-7018.

Tailford, L.E., Offen, W.A., Smith, N.L., Dumon, C., Morland, C., Gratien, J., Heck,

M.P., Stick, R.V., Blériot, Y., Vasella, A., Gilbert, H.J., 2008. Structural and

57
ACCEPTED MANUSCRIPT

biochemical evidence for a boat-like transition state in β-mannosidases. Nat. Chem.

Biol. 4(5), 306-312.

Tamaru, Y., Karita, S., Ibrahim, A., Chan, H., Doi, R.H., 2000. A large gene cluster

T
for the Clostridium cellulovorans cellulosome. J. Bacteriol. 182, 5906-5910.

IP
Tanaka, M., Umemoto, Y., Okamura, H., Nakano, D., Tamaru, Y., Araki, T., 2009.

R
Cloning and characterization of a β-1,4-mannanase 5C possessing a family 27

SC
carbohydrate-binding module from a marine bacterium, Vibrio sp. strain MA-138.

Biosci. Biotechnol. Biochem. 73, 109-116.

NU
Tang, C.D., Li, J.F., Wei, X.H., Min, R., Gao, S.J., Wang, J.Q., Yin, X., Wu, M.C.,
MA
2013. Fusing a carbohydrate-binding module into the Aspergillus usamii β-mannanase

to improve its thermostability and cellulose-binding capacity by in silico design. PLoS


D

One 8, e64766.
TE

Tang, C.M., Waterman, L.D., Smith, M.H., Thurston, C.F., 2001. The cel4 gene of

Agaricus bisporus encodes a β-Mannanase. Appl. Environ. Microbiol. 67, 2298-2303.


P

Thomas, L., Larroche, C., Pandey, A., 2013. Current developments in solid-state
CE

fermentation. Biochem. Eng. J. 81, 146-161.


AC

Titapoka, S., Keawsompong, S., Haltrich, D., Nitisinprasert, S., 2008. Selection and

characterization of mannanase-producing bacteria useful for the formation of

prebiotic manno-oligosaccharides from copra meal. World J. Microbiol. Biotechnol.

24, 1425-1433.

Toti, U.S., Aminabhavi, T.M., 2004. Modified guar gum matrix tablet for controlled

release of diltiazem hydrochloride. J. Controlled Release, 95(3), 567-577.

Tsukagoshi, H., Nakamura, A., Ishida, T., Otagiri, M., Moriya, S., Samejima, M.,

Igarashib, K., Kitamotoa, K., Arioka, M., 2014a. The GH26 β-mannanase RsMan26H

from a symbiotic protist of the termite Reticulitermes speratus is an endo-processive

58
ACCEPTED MANUSCRIPT

mannobiohydrolase: Heterologous expression and characterization. Biochem. Biophy.

Res. Comm. 452(3), 520-525.

Tsukagoshi, H., Nakamura, A., Ishida, T., Touhara, K.K., Otagiri, M., Moriya, S.,

T
Samejima, M., Igarashi, K., Fushinobu, S., Kitamoto, K., Arioka, M., 2014. Structural

IP
and biochemical analyses of glycoside hydrolase family 26 β-mannanase from a

R
symbiotic protist of the termite Reticulitermes speratus. J. Biol. Chem. 289, 10843-

SC
10852.

NU
Van Dyk, J.S., Sakka, M., Sakka, K., Pletschke, B.I., 2010. Identification of

endoglucanases, xylanases, pectinases and mannanases in the multi-enzyme complex


MA
of Bacillus licheniformis SVD1. Enzyme Microb. Technol. 47, 112-118.

Van Zyl, W.H., Rose, S.H., Trollope, K., Görgens, J.F., 2010. Fungal β-mannanases:
D

Mannan hydrolysis, heterologous production and biotechnological applications.


TE

Process Biochem. 45, 1203-1213.

Vijayalaxmi, S., Prakash, P., Jayalakshmi, S.K., Mulimani, V.H., Sreeramulu, K.,
P
CE

2013. Production of extremely alkaliphilic, halotolerent, detergent, and thermostable

mannanase by the free and immobilized cells of Bacillus halodurans PPKS-2.


AC

Purification and characterization. Appl. Biochem. Biotechnol. 171, 382-95.

Vincent, F., Gloster, T.M., Macdonald, J., Morland, C., Stick, R.V., Dias, F., Prates,

J.A., Fontes, C.M., Gilbert, H.J., Davies, G.J., 2004. Common inhibition of both

β‐glucosidases and β‐mannosidases by isofagomine lactam reflects different

conformational itineraries for pyranoside hydrolysis. Chembiochem 5(11), 1596-

1599.

Wang, A., Li, J., Zhang, B., Xu, X., Bewley, J.D., 2009. Expression and location of

endo-β-mannanase during the ripening of tomato fruit, and the relationship between

its activity and softening. J. Plant Physiol. 166, 1672-1684.

59
ACCEPTED MANUSCRIPT

Wang, C., Luo, H., Niu, C., Shi, P., Huang, H., Meng, K., Bai, Y., Wang, K., Hua, H.,

Yao, B., 2015. Biochemical characterization of a thermophilic β-mannanase from

Talaromyces leycettanus JCM12802 with high specific activity. Appl. Microbiol.

T
Biotechnol. 99, 1217-1228.

IP
Wang, C., Zhang, J., Wang, Y., Niu, C., Ma, R., Wang, Y., Bai, Y., Luo, H., Yao, B.,

R
2016. Biochemical characterization of an acidophilic β-mannanase from

SC
Gloeophyllum trabeum CBS900.73 with significant transglycosylation activity and

feed digesting ability. Food Chem. 197, 474-481.

NU
Wang, H.L., Yeh, K.W., Chen, P.R., Chang, C.H., Chen, J.M., Khoo, K.H., 2006.
MA
Isolation and characterization of a pure mannan from Oncidium (cv. Gower Ramsey)

current pseudobulb during initial inflorescence development. Biosci. Biotechnol.


D

Biochem. 70(2), 551-553.


TE

Wang, J., Shao, Z., Hong, Y., Li, C., Fu, X., Liu, Z., 2010. A novel β-mannanase from

Pantoea agglomerans A021: Gene cloning, expression, purification and


P

characterization. World J. Microbiol. Biotechnol. 26, 1777-1784.


CE

Wang, J., Zeng, D., Liu, G., Wang, S., Yu, S., 2014. Truncation of a mannanase from
AC

Trichoderma harzianum improves its enzymatic properties and expression efficiency

in Trichoderma reesei. J. Ind. Microbiol. Biotechnol. 41, 125-133.

Wang, J., Zhang, Q., Huang, Z., Liu, Z., 2013. Directed evolution of a family 26

glycoside hydrolase: Endo-β-1, 4-mannanase from Pantoea agglomerans A021. J.

Biotechnol. 167, 350-356.

Wang, Y., Azhar, S., Gandini, R., Divne, C., Ezcurra, I., Aspeborg, H., 2015.

Biochemical characterization of the novel endo-β-mannanase AtMan5-2 from

Arabidopsis thaliana. Plant Sci. 241, 151-163.

60
ACCEPTED MANUSCRIPT

Wang, Y., Liu, J., Li, Q., Wang, Y., Wang, C., 2015. Two natural glucomannan

polymers, from Konjac and Bletilla, as bioactive materials for pharmaceutical

applications. Biotechnol. Lett. 37(1), 1-8.

T
Willats, W.G.T., McCartney, L., Mackie, W., Knox., J.P., 2001. Pectin: cell biology

IP
and prospects for functional analysis. Plant Mol. Biol. 47, 9-27.

R
Willför, S., Sundberg, K., Tenkanen, M., Holmbom, B., 2008. Spruce-derived

SC
mannans–A potential raw material for hydrocolloids and novel advanced natural

materials. Carbohydr. Pol. 72(2), 197-210.

NU
Wong, W.S.D., Chan, J.V., McCormack, A.A., 2013. Comparative characterization of
MA
a bifunctional endo-1,4-β-Mannanase/1,3-1,4-β-glucanase and its individual

domains. Protein Pept. Lett. 20(5), 517-23.


D

Wozniewski, T., Blaschek, W., Franz, G., 1992. Isolation and characterization of an
TE

endo-β-mannanase of Lilium testaceum bulbs. Phytochemistry 31, 3365-3370.

Wu, G., Bryant, M.M., Voitle, R.A, Roland, D.A., 2005. Effects of β-mannanase in
P

corn-soy diets on commercial leghorns in second-cycle hens. Poult. Sci. 84, 894-897.
CE

Wu, M., Tang, C., Li, J., Zhang, H., Guo, J., 2011. Bimutation breeding of Aspergillus
AC

niger strain for enhancing β-mannanase production by solid-state fermentation.

Carbohydr. Res. 346, 2149-2155.

Xing, X., Cui, S.W., Nie, S., Phillips, G.O., Goff, H.D., Wang, Q., 2014. Study on

Dendrobium officinale O-acetyl-glucomannan (Dendronan®): Part I. Extraction,

purification, and partial structural characterization. Bioact. Carbohydr. Diet. Fibre

4(1), 74-83.

Xu, B., Sellos, D., Janson, J.-C., 2002. Cloning and expression in Pichia pastoris of a

blue mussel (Mytilus edulis) β-mannanase gene. Eur. J. Biochem. 269, 1753-1760.

61
ACCEPTED MANUSCRIPT

Xu, M., Zhang, R., Liu, X., Shi, J., Xu, Z., Rao, Z., 2013. Improving the acidic

stability of a β-mannanase from Bacillus subtilis by site-directed mutagenesis. Process

Biochem. 48, 1166-1173.

T
Xu, X., Zhang, Y., Meng, Q., Meng, K., Zhang, W., Zhou, X., Luo, H., Chen, R.,

IP
Yang, P., Yao, B., 2013. Overexpression of a fungal β-mannanase from Bispora sp.

R
MEY-1 in maize seeds and enzyme characterization. PLoS One 8, 1-10.

SC
Yamabhai, M., Sak-Ubol, S., Srila, W., Haltrich, D., 2016. Mannan biotechnology:

from biofuels to health. Crit. Rev. Biotechnol. 36 (1), 32-42.

NU
Yan, X.X., An, X.M., Gui, L.L., Liang, D.C., 2008. From structure to function:
MA
insights into the catalytic substrate specificity and thermostability displayed by

Bacillus subtilis mannanase BCman. J. Mol. Biol. 379, 535-544.


D

Yang, H., Shi, P., Lu, H., Wang, H., Luo, H., Huang, H., Yang, P., Yao, B., 2015. A
TE

thermophilic β -mannanase from Neosartorya fischeri P1 with broad pH stability and

significant hydrolysis ability of various mannan polymers. Food Chem. 173, 283-289.
P

Yang, P., Li, Y., Wang, Y., Meng, K., Luo, H., Yuan, T., Bai, Y., Zhan, Z., Yao, B.,
CE

2009. A novel β-mannanase with high specific activity from Bacillus circulans
AC

CGMCC1554: gene cloning, expression and enzymatic characterization. Appl.

Biochem. Biotechnol. 159, 85-94.

Yin, J.S., Liang, Q.L., Li, D.M., Sun, Z.T., 2013. Optimization of production

conditions for β-mannanase using apple pomace as raw material in solid-state

fermentation. Ann. Microbiol. 63, 101-108.

Yin, L.J., Tai, H.M., Jiang, S.T., 2012. Characterization of mannanase from a novel

mannanase-producing bacterium. J. Agric. Food Chem. 60, 6425-6431.

Yoon, K.-H., Chung, S., Lim, B.-L., 2008. Characterization of the Bacillus subtilis

WL-3 mannanase from a recombinant Escherichia coli. J. Microbiol. 46, 344-349.

62
ACCEPTED MANUSCRIPT

Yoon, S.-J., Chu, D.-C., Raj Juneja, L., 2008. Chemical and physical properties,

safety and application of partially hydrolized guar gum as dietary fiber. J. Clin.

Biochem. Nutr. 42, 1-7.

T
You, J., Liu, J., Yang, S., Mu, B., 2016. Low-temperature-active and salt-tolerant β-

IP
mannanase from a newly isolated Enterobacter sp. strain N18. J. Biosci. Bioeng.

R
121(2), 140-146.

SC
Zahura, U.A., Rahman, M.M., Inoue, A., Tanaka, H., Ojima, T., 2010. An endo-β-1,4-

mannanase, AkMan, from the common sea hare Aplysia kurodai. Comp. Biochem.

NU
Physiol. - B Biochem. Mol. Biol. 157, 137-143.
MA
Zang, H., Xie, S., Wu, H., Wang, W., Shao, X., Wu, L., Rajer, F.U., Gao, X., 2015. A

novel thermostable GH5_7 β-mannanase from Bacillus pumilus GBSW19 and its
D

application in manno-oligosaccharides (MOS) production. Enzyme Microb. Technol.


TE

78, 1-9

Zhang, H., Sang, Q., 2015. Production and extraction optimization of xylanase and β-
P

mannanase by Penicillium chrysogenum QML-2 and primary application in


CE

saccharification of corn cob. Biochem. Eng. J. 97, 101-110.


AC

Zhang, R., Zhou, J., Gao, Y., Guan, Y., Li, J., Tang, X., Xu, B., Ding, J., Huang, Z.,

2015. Molecular and biochemical characterizations of a new low-temperature active

mannanase. Folia Microbiol. 60, 483-492.

Zhang, X., Rogowski, A., Zhao, L., Hahn, M.G., Avci, U., Knox, J.P., Gilbert, H.J.,

2014. Understanding how the complex molecular architecture of mannan-degrading

hydrolases contributes to plant cell wall degradation. J. Biol Chem. 289(4), 2002-

2012.

Zhang, Y., Ju, J., Peng, H., Gao, F., Zhou, C., Zeng, Y., Xue, Y., Li, Y., Henrissat, B.,

Gao, G.F., Ma, Y., 2008. Biochemical and structural characterization of the

63
ACCEPTED MANUSCRIPT

intracellular mannanase AaManA of Alicyclobacillus acidocaldarius reveals a novel

glycoside hydrolase family belonging to clan GH-A. J. Biol. Chem. 283, 31551-

31558.

T
Zhou, H., Yang, Y., Nie, X., Yang, W., Wu, Y., 2013. Comparison of expression

IP
systems for the extracellular production of mannanase Man23 originated from

R
Bacillus subtilis B23. Microb. Cell Fact. 12, 78.

SC
Zhou, H.Y., Pan, H.Y., Rao, L.Q., Wu, Y.Y., 2011. Redesign the α/β fold to enhance

the stability of mannanase Man23 from Bacillus subtilis. Appl. Biochem. Biotechnol.

163, 186-194.
NU
MA
Zhou, J., Zhang, R., Gao, Y., Li, J., Tang, X., Mu, Y., Wang, F., Li, C., Dong, Y.,

Huang, Z., 2012. Novel low-temperature-active, salt-tolerant and proteases-resistant


D

endo-1,4-β-mannanase from a new Sphingomonas strain. J. Biosci. Bioeng. 113, 568-


TE

574.

Zou, X.T., Qiao, X.J., Xu, Z.R., 2006. Effect of β-mannanase (Hemicell) on growth
P

performance and immunity of broilers. Poult. Sci. 85, 2176-2179.


CE
AC

64
ACCEPTED MANUSCRIPT

Legends to figures

Figure 1 Schematic representation of various types of β, 1-4

mannan/heteromannan.

T
IP
Figure 2 Molecular modelling of schematic structures of galactomannan (F25):

R
(A) 2D model; (B) 3D model without energy minimization; (C) 3D

SC
model with energy minimization. Reprinted with permission from Guo

et al., 2016, Copyright© 2016 Elsevier.

Figure 3
NU
Spatial arrangement of endo-mannanase belonging to glycoside
MA
hydrolase family 5 (A) Schematic representation of the structure of T.

fusca β-mannanase. β Strands β1 and β0, which form the bottom of the
D

barrel, are shown as blue arrows; all other β strands are shown as red
TE

arrows. Helices are depicted as green spirals. The figure was generated

using the program MOLSCRIPT. Reprinted with permission from


P
CE

Hilge et al., 1998, Copyright© 1998 Elsevier, and family 26 (B)

Structural model of ManB-1601 built using BCman (PDB: 2QHA


AC

chain 1) as template. Superimposition of three-dimensional structure of

ManB-1601 (blue) with BCman(red). The figure was generated using

PyMOL graphics (http://www.pymol.org). Reprinted with permission

from Srivastava et al., 2016, Copyright© 2016 Elsevier.

Figure 4 Schematic representation of the T. fusca mannanase–mannotriose

complex interactions. The positions of the four subsites are indicated

below. As the –1 mannosyl residue is not clearly visible in the

electrondensity map, it is depicted with dashed lines at the expected

position. The five water molecules are represented by spheres. The two

65
ACCEPTED MANUSCRIPT

water molecules in the –1 subsite have low B factors and may represent

the positions of HO–C(3) and HO–C(2). In the –3 subsite the double

conformation of the primary hydroxyl group is shown. Reprinted with

T
permission from Hilge et al., 1998, Copyright© 1998 Elsevier.

IP
Figure 5 GGM5 hydrolysis by StMandC and TfMandC. (A) Schematic

R
representation of GGM5 hydrolysis by mannanases. The parenthesis

SC
and numbers show the minus subsites in StMandC and TfMandC. The

NU
triangles show the cleavage site for StMandC (closed) and TfMandC

(open). Reprinted with permission from Kumagai et al., 2015,


MA
Copyright© 2015 John Wiley and Sons.

Figure 6 Conformational states of sugar during glycosylation step of cellulase


D
TE

(family-5 and -7 retaining cellulases) (Scheme 1) and mannanase

(Man26A E212A) (Scheme 2) hydrolysis. Reprinted with permission


P

from Ducros et al., 2002, Copyright© 2002 John Wiley and Sons.
CE

Figure 7 Structural model of ManB-1601 built using BCman (PDB: 2QHA


AC

chain 1) as template. Metal binding site (in golden brown) in zinc-

coordination environment involving His (1), His (23), Glu (336)

residues is shown in stick representation. The figure was generated

using PyMOL graphics (http://www.pymol.org). Reprinted with

permission from Srivastava et al., 2016, Copyright© 2016 Elsevier.

66
ACCEPTED MANUSCRIPT

T
H OH H OH H OH
H OH

IP
HO HO HO O
HO O O O
O O O O
O

R
HO H HO H H H
H H HO H H
HO
H H H H H H
H H

SC
Mannose Mannose Mannose Mannose

OH Linear mannan
Galactose HO

NU
H O
H
HO H
H OH
MA
H O H OH
H OH H H OH

HO HO HO O
HO O O O
O O O O
O
HO H HO H H H
H H HO H H
HO
D

H H H H H H
H H
Mannose Mannose Mannose Mannose
TE

Galactomannan
H OH H OH H OH
H OH
H O HO
P

O H O HO O
O O O O
HO O
CE

HO H HO HO
H OH H H H H
OH
H H H H H H
H H
Glucose Mannose Glucose Mannose
Glucomannan
AC

HO OH

Galactose H O
H
HO H
H OH
H
H OH H O H OH
H OH
H O HO O H O HO O
O O O O
HO HO HO O
H H H HO H
OH H OH H
H H H H H H
H H
Glucose Mannose Glucose Mannose
Galactoglucomannan

Figure 1

67
ACCEPTED MANUSCRIPT

T
RIP
SC
NU
MA
D
TE
P
CE
AC

Figure 2

68
ACCEPTED MANUSCRIPT

T
IP
R
SC
NU
MA
D
P TE
CE

(A) (B)
AC

Figure 3

69
ACCEPTED MANUSCRIPT

T
RIP
SC
NU
MA
D
TE
P
CE
AC

Figure 4

70
ACCEPTED MANUSCRIPT

T
RIP
SC
NU
MA
D
TE
P
CE
AC

Figure 5

71
ACCEPTED MANUSCRIPT

T
R IP
SC
NU
MA

Scheme 1. Substrate conformations along the glycosylation step of family-5 and -7 retaining cellulases: a) Michaelis complex
D

(1S3), b) transition state (4H3), c) covalent intermediate (4C1). Planes containing four atoms within the pyranoside
ring are indicated with gray solid and dashed lines. Reprinted with permission from Ducros et al. (2002). Copyright
TE

© 2002 John Wiley and Sons.


P
CE

Scheme 2. Substrate conformations along the glycosylation step of Man26A E212A. a) Michaelis complex (1S5), b)
AC

transition state (B2,5), c) covalent intermediate (OS2). Gray lines indicate planes containing four atoms within
the pyranoside ring. . Reprinted with permission from Ducros et al. (2002). Copyright © 2002 John Wiley and
Sons.

Figure 6

72
ACCEPTED MANUSCRIPT

T
RIP
SC
NU
MA
D
TE
P
CE
AC

Figure 7

73
ACCEPTED MANUSCRIPT

Table 1 Sources of mannanase§

Sources Reference
Bacteria
Bacillus licheniformis DSM13 Songsiriritthigul et al., 2010

T
Bacillus nealsonii PN-11 Chauhan et al., 2014a, 2014b
Bacillus subtilis B23 Zhou et al., 2011

IP
Bacillus pumilus GBSW19 Zang et al., 2015

R
Bacillus subtilis YH12 Liu et al., 2015

SC
Caldicellulosiruptor strain Rt8B.4 Sunna, 2010
Fungi

NU
Aspergillus niger BK01 Huang et al., 2014
Aspergillus niger strain BCC4525 Sornlake et al., 2013
Chrysonilia sitophila Gonçalves et al., 2012
MA
Neosartorya fischeri P1 Yang et al., 2015

Penicillium chrysogenum QML-2 Zhang and Sang, 2015


Penicillium occitanis Pol6 Blibech et al., 2010
D

Talaromyces leycettanus JCM12802 Wang et al., 2015


Other microorganisms
TE

Streptomyces sp. Shi et al., 2011

Symbiotic protist community in termite gut Tsukagoshi et al., 2014a, 2014b


P

(Reticulitermes speratus)
CE

Plants
Arabidopsis thaliana Wang et al., 2015
Germinating Coffea arabica grains Marraccini et al., 2001
AC

Germinating Lilium testaceum bulbs Wozniewski et al., 1992


Lactuca sativa Dulson & Bewley, 1989; Halmer, 1989
Oryza sativa L. cv. Taichung 65 Ren et al., 2007

Ripening fruits of tomato Sozzi et al., 1996; Wang et al., 2009


(Lycopersicon lycopersicum)
Trigonellafoenum-graecum Dirk et al., 1999
Animals
Antarctic springtail (Cryptopygus antarcticus) Kim et al., 2013; Song et al., 2008
Blue mussel (Mytilus edulis) Larsson et al., 2006; Xu et al., 2002
Common sea hare (Aplysia kurodai) Zahura et al., 2010
Edible snail (Helix lucorum) Flari et al., 1995
§ Only representative references published after 2010 are included with respect to microbial
sources. For more information, the reader is advised to refer to Chauhan et al., 2012, Dhawan
and Kaur, 2006, Zyl et al., 2010 and Moreira and Filho, 2008.

74
ACCEPTED MANUSCRIPT

Table 2 Optimized conditions for endo-mannanase production from


microorganisms under fermentative process§

Organism Incubation Incubation Initial SmF/ Agitation Reference


time Temperature pH SSF (rpm)
0
(h) ( C)

T
Bacillus 48 40 11 SmF 200 Vijayalaxmi et

IP
halodurans al., 2013
strain PPKS-

R
2

SC
Bacillus 96 37 8 SmF 150 Chauhan et al.,
nealsonii 2014a

NU
PN-11
Bacillus 36 50 - SmF 180 Summpunn et
MA
subtilis BCC al., 2011
41051
Bacillus sp. 24 45 6 SmF 200 Srivastava and
D

CFR1601 Kapoor, 2014


TE

Bacillus sp. 72 37 6 SSF - Srivastava and


CFR1601 Kapoor, 2013
P

Bacillus 96 50 - SmF 200 Jiang et al.,


CE

subtilis 2006
WY34
Aspergillus 96 32 - SSF - Wu et al., 2011
AC

niger LW-1
Aspergillus 48 30 5.5 SSF - Yin et al.,
niger SN-09. 2013
Penicillium 144 30 4.5 SSF - Zhang and
chrysogenum Sang, 2015
QML-2
Streptomyces 48 28 6.5 SmF 130 Pradeep et al.,
sp. CS428 2016
§ Only references published after 2010 or not covered in earlier reviews are

included. For earlier references, the reader is advised to refer to Chauhan et al.,

2012, Dhawan and Kaur, 2006, Zyl et al., 2010 and Moreira and Filho, 2008.

75
ACCEPTED MANUSCRIPT

Table 3 Heterologous cloning and expression of endo-mannanase gene from

various organisms§

T
Ge Endo-
Amino Molecu

IP
ne mannan
Source Heterolog acids lar
Vector size pI ase Reference
organism ous host (numb weight
(bp product

R
er) (kDa)
) ion

SC
GH 5 endo-mannanases
Bacteria
Bacillus E. coli pET- 113 377 45 NR NR Zang et al.,
(28)*

NU
pumilus BL21 30a- 4 2015
GBSW19 c(+)
Bacillus E. coli pH6E 208 694 76.089 NR NR Ethier et
stearothermop BL21 X3 5 al., 1998
MA
hilus
Bacillus E. coli pSMA 110 369 41 5.3 5.5 Chauhan et
nealsonii PN- DH10β RT 0 5 IU/ml al., 2015
11
D

Clostridium E. coli pET NR NR NR NR NR Malgas et


cellulovorans BL21(DE3 29b al., 2015b
TE

)
Fungi
P

Aspergillus P. pastoris pPICZ - - 56 - NR Dilokpimol


nidulans X33 α et al., 2011
CE

FGSC A4 A
Aspergillus P. pastoris pPICZ 115 383 37.5 4.1 29 U/ml Li et al.,
niger LW-1 GS115 α 2 (21)*[1 5 2012
7]#
AC

A
Chaetomium P. pastoris pPIC9 125 451 50 4.5 50030 Katrolia et
sp. CQ31 GS115 K 1 U/ml§ al., 2012
Neosartorya P. pastoris pPIC9 118 373 39.5 5.9 101.5 Yang et al.,
fischeri P1 GS115 7 3 U/ml 2015
Penicillium P. pastoris pPIC9 115 384 39 NR 2.5 g/l§ Cai et al.,
sp. C6 GS115 5 (26)* 2011
Penicillium P. pastoris pPICZ 138 426 45 5.0 NR Liao et al
oxalicum GZ- GS115 α 0 9 2015
2 A
Rhizomucor E. coli pET- 133 378 44 NR NR Katrolia et
miehei BL21 2 0 al., 2013
8
a

(
+
)

76
ACCEPTED MANUSCRIPT

Others
Cryptopygus E. coli pET- 114 382 39.5 5.9 436.6 Song et al.,
antarcticus strain 3 9 9 U/l 2008
Rosetta- 2
gami2 a
(DE3) (

T
+

IP
)
Haliotis discus - - 113 377 39.6 NR NR Ootsuka et
(17)*

R
hannai 4 al., 2006
Mytilus edulis P. pastoris pPICZ 110 368 40 6.8 900 Xu et al.,

SC
with a 4 5- mg/ml 2002
Saccharom 8.1
yces B 5

NU
cerevisiae
α-factor
GH 26 endo-mannanases
MA
Bacteria
Bacillus E. coli pFLA 108 336 45 NR 54,266 Songsiriritt
licheniformis BL21 G-CTS 0 U/l higul et al.,
DSM 13 2010
Bacillus sp. E. coli pUC18 299 997 130 NR NR Hatada et
D

strain HB101 4 al., 2005


TE

JAMB-750
Bacillus Brevibacill pHY- 110 NR 40 NR NR Zhou et al.,
subtilis B 23 us brevis p43 0 2013
P

Bacillus Bacillus pMA- 108 360 40.14 NR 220 Liu et al.,


subtilis YH12 subtilis 5 3 U/ml 2015
CE

strain
WB600
Bacillus sp. E. coli pRSE 108 336 40 5.0 666 Srivastava
AC

CFR1601 BL21(DE3 T-A 3 7 U/ml et al., 2016


)
Bacillus sp. E. coli BL pRSE - - - - 8406 Kaira et al.,
CFR1601 21(DE3)Hi T-A U/ml 2016
-control
cells
Bacillus Pichia pPIC9 108 362 41 5.8 5156.74 Li et al.,
subtilis WD23 pastoris K 9 1 2 U/ml 2015
GS115
Clostridium E. coli pET- 144 478 53 NR NR Ghosh et
thermocellum BL21(DE3 28a 9 al., 2013
ATCC 27405 ) (+)
Sphingobacter E. coli pET- 111 371 42.2 NR 65.3 Zhang et
ium sp. GN25 BL21 2 6 mg/l al., 2015
(DE3) 8
a
(
+

77
ACCEPTED MANUSCRIPT

)
Reticulitermes P. pastoris pPICZ 390 NR 40 NR 0.67 g/l Tsukagoshi

speratus a et al.,
- 2014a,
A 2014b
Fungi

T
Aspergillus A. oryzae NR NR NR 35.2 NR NR Freiesleben

IP
nidulans et al., 2016
NR: Not reported; *Amino acids present for signal peptide; # Amino acids present for

R
pro-peptide; § high cell density fermentation; ₸ Symbiotic microflora of Reticulitermes

SC
speratus; § Only references published after 2010 or not covered in earlier reviews are
included. For earlier references, the reader is advised to refer to Chauhan et al., 2012,

NU
Dhawan and Kaur, 2006, Zyl et al., 2010 and Moreira and Filho, 2008.
MA
D
P TE
CE
AC

78
ACCEPTED MANUSCRIPT

Table 4 Protocols employed for purifying wild-type endo-mannanases from


different organisms.

Specific
Purification Number Yield Purification
Organism activity Reference
protocol of steps (%) fold
(IU/mg)

T
Aplysia (NH4)2SO4(40- 4 3.3 14.5 27.4 Zahura et

IP
kurodai 60%), al., 2010
Toyopearl
Phenyl-650 M,

R
Toyopearl CM-

SC
650 M, and
Mono-S
Bacillus (NH4)2SO4(60– 3 8.92 38.96 2280.9 Chauhan
nealsonii 80%), Sephadex et al.,

NU
PN11 G-150, and 2014b
DEAE-
Cellulose
MA
Bacillus (NH4)2SO4(40- 3 20.3 5.4 8302.4 Jiang et
subtilis 80%), al., 2006
WY34 SuperdexTM75,
and
D

Q-Sepharose
fast flow
TE

Bacillus sp. (NH4)2SO4 (50– 4 21.3 50.7 10461.5 Srivastava


CFR1601 80%), DEAE- et al.,
Cellulose, 2016
P

DEAE-
CE

Sepharose and
Phenyl-
Sepharose
Coffea (NH4)2SO4 4 10 7000 1400 Marraccini
AC

arabica L. (35%), Phenyl- et al.,


Sepharose, 2001
Mono Q, and
Superdex 75 HR
10/30
Helix Hydroxyapetite, 3 13 26.4 29 Flari et al.,
lucorum L. DEAE- 1995
Sepharose, and
QA-Trisacryl
Paenibacillus (NH4)2SO4(40- 4 6.4 90.2 635.4 Yin et al.,
cookie 60%), 2012
DEAE-
Sepharose and
Sephacryl S-100
HR column
(two times)
Penicillium (NH4)2SO4 4 5.3 9.4 160 Kurakake
oxalicum SO (90%), et al.,

79
ACCEPTED MANUSCRIPT

Sephadex G-25, 2006


Super Q
Toyopearl, and
TSKgel
G3000SW

T
RIP
SC
NU
MA
D
P TE
CE
AC

80
ACCEPTED MANUSCRIPT

Table 5 Positioning of catalytic acid base and nucleophile in mannanases


from different organisms.

Organism Catalytic acid Catalytic Reference


base residue nucleophile

T
residue
A symbiotic protist from termite Glu191 Glu288 Tsukagoshi et al.,

IP
gut 2014a, 2014b

R
Alicyclobacillus acidocaldarius Glu151 Glu231 Zhang et al., 2008

SC
Tc-12-31
Aplysia kurodai (sea hare) Glu162 Glu293 Mizutani et al.,

NU
2012
Bacillus subtilis z2 Glu167 Glu266 Yan et al., 2008
Bacillus nealsonii PN11 Glu152 Glu247 Chauhan et al.,
MA
2015
Cellulomonas fimi Glu175 Glu282 Hekmat et al.,
2010
D

Cellvibrio japonicas Glu212 Glu320 Tailford et al.,


TE

2009
Cellvibrio mixtus Glu429 Glu519 Dias et al., 2004
P

Chrysonilia sitophila Glu181 Glu301 Gonçalves et al.,


CE

2012
Lycopersicon esculentum Glu204 Glu318 Bourgault et al.,
AC

(Tomato) 2005
Penicillium sp. C6 Glu203 Glu314 Cai et al., 2011
Pseudomonas fluorescens ssp. Glu212 Glu320 Bolam et al.,
Cellulose 1996; Hogg et al.,
2001
Thermomonospora fusca Glu128 Glu225 Hilge et al., 1998
Trichoderma reesei Glu169 Glu276 Sabini et al., 2000

81
ACCEPTED MANUSCRIPT

Table 6 Biochemical properties of endo-mannanase from various sources§

Mole
cular
Source/Fa Temperatur Kinetic Refer
pH weig pI
mily e (0C) parameters ence
ht

T
(kDa)
Classified Opti Stab Opti Stab Km Vmax

IP
mum ility mum ility
GH 5

R
Bact

SC
eria
Bacil 8.8 5-10 65 t1/2 50 N 7.22 750 Chau
lus 3h at R (LBG) (LBG), han et

NU
neals 700C , 344 al.,
onii 11.59 (GG) 2014b
PN1 (GG), and
MA
1 and 256
18.73 (CM)
(CM) µmol/
mg/ml ml/min
Clost 5.5– 5.5– 40 90 47 N NR NR Malga
D

ridiu 7.0 7.0 min R s et


TE

m at al.,
cellul 370C 2015b
ovor , 30
P

ans min
at
CE

500C
Fung
i
AC

Gloe 2.5 2.0 600C 35- 38.6 5. 3.71 2525 Wang


ophyl to 650C kDa 1 mg/ml U/min/ et al.,
lum 10.0 3 (LBG) mg 2016
trabe
um
CBS
900.7
3
Neos 4 3.0– 80 t1/2 39.5 5. 0.83 1937.2 Yang
artor 7.0 6h at 9 mg/ml µmol/ et al.,
ya 600C 3 (LBG) min/m 2015
fisch g
eri (LBG)
P1
GH
26
Bact

82
ACCEPTED MANUSCRIPT

eria
Bacil 6 5-10 55 NR NR N 5 5000 Srivas
lus R mg/ml tava
sp. and
CFR Kapo
1601 or,

T
2015

IP
Dicty 5 4-7 80 16h 40 N NR NR Gibbs
oglo at R et al.,

R
mus 80° 1999
ther C

SC
moph and
ilum t1/2
Rt46 5.4
B.1 min

NU
at
90°
C
MA
Fung
i
Aspe 5.5 - - Tm 35.2 N NR NR Freies
rgillu at R leben
D

s 53◦C et al.,
nidul 2016
TE

ans
Retic 5 5.0– 40 80% 40 N NR NR Tsuka
uliter 6.5 < R goshi
P

mes 300C et al.,


CE

spera , 2014a
tus₸ 400C
activ
AC

ity
lost
Uncl
assifi
ed
Bacil 6.5 4.5– 55 95% 40.14 N 30 16666. Liu et
lus 7.5 activ R mg/ml 7 U/mg al.,
subtil ity at (LBG) (LBG) 2015
is 550C
YH1 , 3h
2
Paen 5 5-7 50 Stab 68 N 0.009 556 Yin et
ibaci le R 5 and and al.,
llus belo 0.013 484 2012
cooki w 6 mg/ U/min/
e 400C ml for mg, for
, t1/2 LBG LBG
30 and and

83
ACCEPTED MANUSCRIPT

min KGM, KGM,


at respec respect
620C tively ively
NR: Not reported; LBG: Locust bean gum; KGM: Konjac glucomannan; GG: Guar
gum; CM: Copra mannan; GH: Glycosyl hydrolase; ₸ Symbiotic protist of

T
Reticulitermes speratus, § only references published after 2012 or not covered in earlier

IP
reviews are included. For earlier references, the reader is advised to refer to Chauhan
et al., 2012, Dhawan and Kaur, 2006, Zyl et al., 2010 and Moreira and Filho, 2008.

R
SC
NU
MA
D
P TE
CE
AC

84
ACCEPTED MANUSCRIPT

Table 7 Production conditions, yield, type and prebiotic role of


mannooligosaccharides (MOS)

Organism Hydrolys Substra Yield Degree of Prebiotic role Reference


is te polymerizati s
condition on

T
s (DP)
Bacillus 550C, 6 h LBG 11% 2-5 Stimulated Chauhan

IP
nealsonii Mannobiose Lactobacillus et al.,
PN-11 , 23% casei and 2014b

R
Mannotrios inhibited
e, 20% Salmonella

SC
Mannotetro enterica
se, 18%
Mannopent

NU
ose
Bacillus 500C, LBG 1.19 mg/ml 1-6 NR Zang et
pumilus 24h,10 al., 2015
GBSW19 mg/ml,
MA
LBG 10
U/mg,
endo-
mannana
D

se
TE

Bacillus sp. pH 6, GG 14% 2 and 4 Lactobacillus Srivastav


CFR1601 500C, salivarius a and
300 min, and L. Kapoor,
10 IU/ml Plantarum 2014
P

Bacillus sp. pH 6, LBG 30% 2, 3 and 5 Several Srivastav


CE

CFR1601 500C, Lactobacilli a et al.,


270 min, 2015b
10 IU/ml
AC

Bacillus pH 6.5, LBG, 1–7 (LBG), NR Liu et al.,


subtilis 550C, 8h KGM 2–7 (KGM) 2015
YH12 and XG and 1–2
(XG)
Bifidobacteri pH 5.3, KGM, - 2, 3 and 4 - Kulcinsk
um 300C, carob, aja et al.
adolescentis 0.2-1 GG 2013
µM
enzyme
Bifidobacteri pH 6, LBG - 2, 3, 4, 5 - Morrill et
um animalis 370C, 5.3 and al., 2015
subsp. lactis nM INM
Bl-04 enzyme
Clostridium pH 6.9, Defatte 40% 2-6 Lactobacillus Ghosh et
thermocellu 600C, 10 d copra Mannobiose acidophilus al., 2014
m min meal , NRRL B-
18% 4496 and
Mannotrios Bifidobacteri

85
ACCEPTED MANUSCRIPT

e um infantis

Penicillium pH 4.0, LBG 33%*, 3-4 NR Blibech


occitanis 700C, 40%# et al.,

T
10h 2011

IP
Penicillium pH 5, GG NR 2-7 NR Kurakake
oxalicum SO 400C, et al.,

R
24h 2006
Trichoderma 48h KGM 50% 2-5 NR Mikkelso

SC
reesei n et al.,
2013
NR: Not reported; * Free enzyme, # Immobilized enzyme; LBG: Locust bean gum; KGM: Konjac

NU
glucomannan; XG: Xanthan gum, GG: Guar gum, INM: Ivory nut mannan
MA
D
P TE
CE
AC

86
ACCEPTED MANUSCRIPT

Highlights

 Production, purification, heterologous expression and biochemical

characteristics of endo-β-mannanases

T
 The molecular and structural features of endo-β-mannanases responsible for

IP
its activity and thermal stability

R
 The use of rational design and genetic engineering approaches for improving

SC
endo-β-mannanases

NU
 Biotechnological applications of endo-β-mannanases
MA
D
P TE
CE
AC

87

You might also like