Microbial Enzymes in Bioconversions of Biomass

Download as pdf or txt
Download as pdf or txt
You are on page 1of 345

Biofuel and Biorefinery Technologies 3

Vijai Kumar Gupta Editor

Microbial
Enzymes in
Bioconversions
of Biomass
Biofuel and Biorefinery Technologies

Volume 3

Series editors
Vijai Kumar Gupta, Molecular Glycobiotechnology Group, Department of
Biochemistry, School of Natural Sciences, National University of Ireland
Galway, Galway, Ireland
Maria G. Tuohy, Molecular Glycobiotechnology Group, Department of
Biochemistry, School of Natural Sciences, National University of Ireland
Galway, Galway, Ireland
More information about this series at http://www.springer.com/series/11833
Vijai Kumar Gupta
Editor

Microbial Enzymes
in Bioconversions
of Biomass

123
Editor
Vijai Kumar Gupta
Department of Biochemistry, School of
Natural Sciences
National University of Ireland Galway
Galway
Ireland

ISSN 2363-7609 ISSN 2363-7617 (electronic)


Biofuel and Biorefinery Technologies
ISBN 978-3-319-43677-7 ISBN 978-3-319-43679-1 (eBook)
DOI 10.1007/978-3-319-43679-1

Library of Congress Control Number: 2016947366

© Springer International Publishing Switzerland 2016


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part
of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations,
recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission
or information storage and retrieval, electronic adaptation, computer software, or by similar or
dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are exempt
from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this
book are believed to be true and accurate at the date of publication. Neither the publisher nor the
authors or the editors give a warranty, express or implied, with respect to the material contained
herein or for any errors or omissions that may have been made.

Printed on acid-free paper

This Springer imprint is published by Springer Nature


The registered company is Springer International Publishing AG Switzerland
Preface

This book summarizes the information on different biomass converting enzymes


and their potential use in the bioconversion of biomass into simple sugar to generate
bioenergy and other value added co-/by products.
Biofuel produced from agricultural crops, e.g. cereals, maize, sugarcane, sugar
beet, and sweet sorghum, is referred to as first generation biofuels while that
produced from lignocellulosic materials is referred to as second generation biofuels.
The global production of first and second generation biofuels is dependent upon the
lignocellulosic materials available for bioconversion.
Microbes have been established as important mediators in the production of
biofuels and other valuable biorefinery products. It is prevalent that microbes have
been playing a key role by producing key enzymes for bioconversion of various
biomasses for biorefinery applications. This selection of microbes is based upon
their ability to selectively or simultaneously degrade lignocellulosic materials, the
high redox potential of their enzymes, their engineering capabilities and/or their
thermostability. The key to their degradative capabilities is the extracellular
enzymes they secrete. These microorganisms are capable of producing an array of
enzymes such as 1) Cellulase systems, comprised of endo-1,4-β-glucanase, cel-
lobiohydrolase and β-glucosidases; 2) Hemicellulases include endo-1,4-β-
xylanases, β-xylosidases, endo-1,4-β-mannanase and β-mannosidase; 3) Lignin
degrading enzymes comprising laccase, lignin peroxidase, manganese peroxidase
and versatile peroxidase. There are also accessory enzymes which aid the hemi-
cellulases in their degradation of hemicellulose polymers through the cleavage of side
chain residues; these enzymes include α-glucuronidase, α-L-arabinofuranosidase,
acetylxylan esterase, ferulic acid esterase and β-galactosidase.
This book summarizes the information on different biomass converting enzymes
and their potential use in the bioconversion of biomass into simple sugar to generate
bioenergy and other value added co-/by products. The chapters have been con-
tributed by the experts of the area across the globe and these chapters offer clear and
concise information on both standard and new technologies and will serve as an

v
vi Preface

invaluable reference for undergraduates, post-graduates, researchers and practi-


tioners studying and working in the field of microbial enzymes for biofuel and
biorefinery applications.

Galway, Ireland Vijai Kumar Gupta


Contents

1 Microbial Enzymes for Conversion of Biomass to Bioenergy . . . . . . 1


M.P. Raghavendra, S. Chandra Nayaka and Vijai Kumar Gupta

Part I Cellulases
2 Cellobiohydrolases: Role, Mechanism, and Recent
Developments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Neelamegam Annamalai, Mayavan Veeramuthu Rajeswari
and Nallusamy Sivakumar
3 Endo-1,4-β-glucanases: Role, Applications and Recent
Developments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
Neelamegam Annamalai, Mayavan Veeramuthu Rajeswari
and Thangavel Balasubramanian
4 The Role and Applications of β-Glucosidases in Biomass
Degradation and Bioconversion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
Hanlin Ouyang and Feng Xu

Part II Hemicellulases
5 Role and Applications of Feruloyl Esterases in Biomass
Bioconversion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
Constantinos Katsimpouras, Io Antonopoulou, Paul Christakopoulos
and Evangelos Topakas
6 Endo-β-1,4-xylanase: An Overview of Recent Developments . . . . . . 125
Alexandre Gomes Rodrigues
7 Microbial Xylanases: Sources, Types, and Their Applications . . . . . 151
Hesham Ali El Enshasy, Subeesh Kunhi Kandiyil, Roslinda Malek
and Nor Zalina Othman

vii
viii Contents

8 Mannanase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Suttipun Keawsompong
9 The Role and Applications of Xyloglucan Hydrolase in Biomass
Degradation/Bioconversion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
M. Saritha, Anju Arora, Jairam Choudhary, Vijaya Rani,
Surender Singh, Anamika Sharma, Shalley Sharma and Lata Nain

Part III Lignocellulose Oxidureductases


10 Role of Mushroom Mn-Oxidizing Peroxidases in Biomass
Conversion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
Mirjana Stajić, Jelena Vukojević, Ivan Milovanović,
Jasmina Ćilerdžić and Aleksandar Knežević
11 Role and Application of Versatile Peroxidase (VP) for Utilizing
Lignocellulose in Biorefineries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 271
Nadine Busse and Peter Czermak
12 Fungal Aryl-Alcohol Oxidase in Lignocellulose Degradation
and Bioconversion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
Juan Carro, Ana Serrano, Patricia Ferreira and Angel T. Martínez
13 Monosaccharide Oxygenase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323
Avnish Kumar, Monika Asthana, Hirawati Deval and Sarika Amdekar
Chapter 1
Microbial Enzymes for Conversion
of Biomass to Bioenergy

M.P. Raghavendra, S. Chandra Nayaka and Vijai Kumar Gupta

Abstract Microbial enzymes are capable of degrading a wide range of complex


substrates including carbohydrates into more useful energy source. The simple
sugars then can be converted into ethanol or other liquid biofuels by a large group
of fermentative microbes. Even though cellulose serves as an abundant source of
carbon and energy in the ecosystem, its exploitation as a source of biofuel is
hindered due to lack of effective microbial systems to break it down, including other
carbohydrates to simple sugars leading to more production of biofuels. If these
materials could be exploited, they would represent a massive new energy resource
for biofuel production. In continuous search for alternative energy sources, it is now
proven that electricity can be produced directly from the degradation of organic
matter in a microbial fuel cell and fermentation of lignocellulosic biomass to
ethanol, which is an attractive route to fuels that supplements the fossil
fuels. Studies have revealed that special group enzymes known as feruloyl esterases
produced by microorganisms are capable of breaking apart key links between the
polymers and helps in effective degradation of plant materials. This review covers
various known microbial approaches to convert different carbon sources to simple
soluble sugars en route to production of biofuels. The importance of the biofuel in
future is highlighted by the Renewable Fuel Standard of the United States Energy
Independence and Security Act (EISA) of 2007, which mandates that 36 billion
gallons of biofuels are to be produced annually by 2022, of which 16 billion gallons
are expected to come from cellulosic feed stocks. It is obvious fact that microor-

M.P. Raghavendra
Postgraduate Department of Microbiology, Maharani’s Science College for Women,
JLB Road, Mysore, Karnataka 570 005, India
S.C. Nayaka (&)
Department of Studies in Biotechnology, University of Mysore, Mysore,
Karnataka 570 005, India
e-mail: [email protected]
V.K. Gupta
Department of Biochemistry, School of Natural Sciences,
National University of Ireland, Galway, Ireland

© Springer International Publishing Switzerland 2016 1


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_1
2 M.P. Raghavendra et al.

ganisms and its array of enzymes need to be effectively screened, identified and
employed in developing effective strategies for converting biomass to biofuel.

1.1 Introduction

Due to over reliance on petrochemicals, rising oil prices, pollution associated with
its usage, world is looking forward for alternative renewable resources of energy
from different sources. Biofuel obtained from carbon sources are gaining
momentum as a source of biofuel but the concern over production of ethanol from
food crops is creating a imbalance and there is a search for a resources, which are
not being used as food and should be available plenty in the ecosystem so that the
problems associated with the starchy material can be addressed. In this connection
cellulosic feed stock is considered to be excellent source for ethanol production, but
there are problems associated with degradation of cellulose also due to its recal-
citrance to biodegradation at least at the early stages of degradation. Liquid fuel
derived from biomass feedstock has been considered as a low-carbon substitute for
fossil hydrocarbons in vehicles, but many of the existing approaches are encoun-
tering significant challenges.

1.2 Importance of Biofuel Research

Search for new energy supplies is an important agenda for several countries in the
coming years. They are developing new energy infrastructure capable of meeting
growing demands for electric power and transportation fuels. Even unrest in oil
producing countries will have great impact on national security and economic
strength of several oil dependent countries. India in particular currently imports
80 % of its oils and it is expected to increase to 91 % by 2020. This overreliance on
oil import is the greatest burden on Indian economy and its security. On the other
hand, India is an agrarian country and known for its biodiversity that can be
exploited to convert biomass to biofuel. Biofuels employ recycling of agricultural
byproducts and dedicated energy crops, which offer opportunities for mitigation of
greenhouse gas emission as growing these leads to C sequestration through
photosynthesis.
Various plants and plant-derived materials are used for biofuels manufacturing
including grains (1st generation) and lignocellulosic biomass (2nd generation). The
second generation of biofuels is more important as they are based on the cheap and
abundant lignocellulosic biomass and do not compete with food crops. Wheat straw
and bagasse are considered that best example, which are inexpensive and widely
available ligonocellulosic resources containing 75–80 % polysaccharides (cellulose
and hemicelluloses). The monosaccharides glucose and xylose obtained from
hydrolysis of these can be used as substrate for production of industrially important
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 3

Proteins and Secondary


Amino acids metabolites

Pretreatment, hydrolysis Microbial degradation

Lignocellulose and other carbohydrates

Lignin, Hydrolysates of
Cellulose hemicellulose

Enzymatic/acid hydrolysis

Xylose, mannose, glucose,


galactose, rhamnose, arabinose
Lignin Hexose
Fragmentation
Depolymerizatio Chemical conversion

Green gasoline or
diesel Fermentation

Gaseous fuels: H2, methane


Liquid fuels: methane, ethanol, dimethyl ether, butanol, alkanes

Fig. 1.1 Conversion of carbohydrate biomass into biofuel

products (Fig. 1.1). These products will have more potential economical, envi-
ronmental, and strategic advantages over traditional fossil-based products.
The era of advanced biofuels—cellulosic ethanol, biomass-based diesel,
biobutanol, bio-oil, green gasoline, and bio-based jet fuel—is also drawing nearer
and nearer. According to the International Energy Agency, biofuels have the
potential to meet more than a quarter of world demand for transportation fuels by
2050. Bioethanol is by far the most widely used biofuel for transportation world-
wide (Mohanram et al. 2013). It has been estimated that widespread use of biofuels,
biopower, and other bio-based products has the potential to conserve 1.26 billion
barrels of oil, 58 million tons of coal, and 682 million tons of carbon dioxide from
2020 to 2030.

1.3 Lignocelluloses and Its Structural Complexity

The cell walls of plants represent an enormous nutrient source, yet highly variable
in terms of amount, diversity, and botanical source. Plant carbohydrates are
chemically and structurally highly complex, and are arranged in a
three-dimensional network that has evolved to be intrinsically resistant to enzymatic
4 M.P. Raghavendra et al.

breakdown (Fontes and Gilbert 2010). Thus high molecular weight crystalline
cellulose microfibrils are intertwinned with hemicelluloses and pectins, which are a
whole range of homo- and heteropolysaccharides composed of dozens of different
monosaccharide units linked in a multitude of ways to resist degradation. Ester
substituents or non-carbohydrate, polymers, such as lignin, proteins, cutin, and
suberin, add a further layer of complexity. As a result, a single vegetable contains
hundreds of different bonds that need to be cleaved in order to unlock the assim-
ilable carbon of the cell wall constituents.
In general cell walls are intricate assemblages of celluloses, hemicelluloses (i.e.,
xyloglucans, arabinoxylans, and glucomannans), pectins (i.e., homogalacturonan,
rhamnogalacturonan I and II, and xylogalacturonans), lignins, and proteoglycans
(e.g., arabinogalactan-proteins, extensins, and proline-rich proteins). Most mass in
the plant cell wall is in the form of polysaccharides (cellulose and hemicelluloses).
The next most abundant polymer is lignin, which is composed predominantly of
phenylpropane building blocks.
Among these different carbon sources lignocellulosic biomass is the most
abundant, low-cost and unexploited biomass that is considered as a source for
industrial production of fuel ethanol. Considering its availability over the last
decades, researchers have been devoted to converting lignocellulosic materials to
bioethanol. Lignocellulosics are composed of heterogeneous complex of carbohy-
drate polymers (cellulose, hemicelluloses, and lignin). Majority of plant biomass is
locked up in 5- and 6-carbon sugars, comprised of mainly cellulose (a glucose
homopolymer, 40–60 % by wt.); less so, hemicelluloses (a sugar heteropolymer,
20–40 % by wt.); and least of all lignin (a complex aromatic polymer, 10–30 % by
wt). The major component cellulose, which is organized into microfibrils, each
containing up to 36 glucan chains having thousands of glucose residues linked by
β-(1,4) glycosidic bonds, is largely responsible for the plant cell wall’s mechanical
strength. Hemicelluloses are built up by pentoses (D-xylose, D-arabinose), hexoses
(D-mannose, D-glucose, D-galactose) and sugar acids. These include β-glucan,
xylan, xyloglucan, arabinoxylan, mannan, galactomannan, arabinan, etc. Hardwood
contains mainly xylans, while in softwood glucomannans are most common. Both
the cellulose and hemicelluloses can be broken down enzymatically into the
component sugars which may be then fermented to ethanol.
Cellulose can be classified into two types based on the arrangement of chains. In
type I cellulose chains are arranged in parallel direction of the long axis of the
microfibril, whereas in type II chains are in antiparrellel position. Among these,
type I is considered to be dominant that can be converted to type II by alkali
treatment. Studies show that cellulose I actually contain two distinct crystal lattices:
cellulose Iα, with triclinic symmetry, and cellulose Iβ, with monoclinic symmetry.
These two forms differ in the organization of their intermolecular hydrogen bonds
and lead to further complexity of cellulose structure. The percentage crystallinity of
cellulose varies from very low to almost 100 % and it depends on cellulose source.
However, it is clear that cellulose microfibrils are not uniformly crystalline;
imperfections in packing or mechanical damage result in a proportion of substrate
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 5

in which the lattice is disordered or paracrystalline (Ranjan 2014). Hence, microbes


have to produce cellulases with range of structures, activities, and stabilities.

1.4 Enzymatic Hydrolysis of Lignocelluloses

Even though lignocellulose is considered as the most abundant and renewable


natural resource available to man throughout the world, massive technological
difficulties must be overcome to exploit this for commercial purpose. In order that
cellulose hydrolysis becomes economically feasible, it is important to identify
methods that increase enzyme effectiveness and overcome barriers of enzymatic
hydrolysis (Kristensen et al. 2007). Major factors that influence enzymatic con-
version of lignocelluloses to fermentable sugars include accessible surface area of
lignocelluloses (Mansfield et al. 1998), enzyme loading, and presence of inhibitors
(Eriksson et al. 2002).
Pretreatment is a necessary step generally carried out to produce ethanol from
lignocellulosic materials by dilute sulfuric acid or enzymatically. The polysaccha-
ride chains, being tightly packed, require additional factors that would make the
substrate more accessible, as has been suggested since the 1950s (Horn et al. 2012).
The enzymatic hydrolysis is generally carried out by cellulolytic enzymes. Without
pretreatment, conversion is tedious and very slow. Enzymatic hydrolysis is con-
sidered to be effective because it happens in mild hydrolysis conditions and high
yields of hydrolysis, but it results in accumulation of inhibitory byproducts, costly,
and hydrolysis require long time and also results in product inhibition during
hydrolysis. The biological treatments can be achieved by enzymes of fungi and
actinomycetes, which results in delignification and reduction in degree of poly-
merization of hemicellulose and cellulose.
By the use of enzymatic hydrolysis, pure cellulose can be degraded to soluble
sugars, which can be fermented to form ethanol, butanol, acetone, single cell
protein, methane, and many other products. Enzymatic saccharification remains one
of the most costly steps in conversion of cellulosic biomass to ethanol and cellulase
preparations dedicated for bioethanol industry are hardly available. It has been
estimated that the greatest returns in cost savings will be realized by improving
conversions of biomass to sugars, increasing hydrolysis yields, reducing enzyme
loadings, and eliminating or reducing pretreatment (Lynd et al. 2008). A broader
suite of enzymes is required for hydrolysis of cellulose and hemicelluloses to
fermentable sugars (McMillan et al. 2011). Thus enzymatic hydrolysis can be
effectively carried out if a mixture of different cellulolytic and accessory enzymes is
used.
Enzyme hydrolysis is generally considered the most sustainable technology for
saccharification. Enzymatic hydrolysis of cellulose consists of three steps viz.,
6 M.P. Raghavendra et al.

1. Adsorption of cellulase enzymes onto the surface of the cellulose,


2. Biodegradation of cellulose to fermentable sugars, and
3. Desorption of cellulase (Sun and Cheng 2002).
Different strategies are also available for enzymatic hydrolysis and fermentation
of lignocelluloses such as separate enzymatic hydrolysis and fermentation (SHF),
simultaneous saccharification and fermentation (SSF), nonisothermal saccharifica-
tion and fermentation (NSSF), simultaneous saccarification, and co-fermentation
(SSCF) and consolidated bioprocessing (CBP) (Taherzadeh and Karimi 2007).
Among different strategies CBP is considered to posses outstanding potential,
ethanol together with all of the required enzymes is produced by single microbial
communities is also referred to as direct microbial conversion (DMC). It may be
involved single or multiple cultures of microorganism to directly convert cellulose
to ethanol in a single bioreactor (Wyman 2007).
The classical model for degradation of cellulose to glucose involves the coop-
erative action of endocellulases (EC 3.2.1.4), exocellulases (cellobiohydrolases,
CBH, EC 3.2.1.91; glucanohydrolases, EC 3.2.1.74), and betaglucosidases (EC
3.2.1.21). Endocellulases hydrolyze internal glycosidic linkages in a random
fashion, which results in a rapid decrease in polymer length and a gradual increase
in the reducing sugar concentration. Exocellulases hydrolyze cellulose chains by
removing mainly cellobiose either from the reducing or the nonreducing ends,
which lead to a rapid release of reducing sugars but little change in polymer length.
Endocellulases and exocellulases act synergistically on cellulose to produce
cello-oligosaccharides and cellobiose, which are then cleaved by beta-glucosidase
to glucose (Kumar et al. 2008).
Hydrolysis of hemicelluloses involve enzymes like glycoside hydrolases, car-
bohydrate esterases, polysaccharide lyases, endohemicellulases, and others, the
concerted action of which hydrolyze glycosidic bonds, ester bonds, and remove the
chain’s substituents or side chains (Sweeney and Xu 2012). These include endo-1,
4-β-xylanase, β,-xylosidase, β-mannanase, β mannosidase α-glucuronidase, α- L-
arabino furanosidase, acetylxylan esterase and other enzymes.
Glycohydrolases viz., cellulase and hemicellulase enzymes are involved in
hydrolysis of cellulose and hemicelluloses respectively. These are known to posses
15 protein families with other subfamilies. Hydrolysis of cellulose begins with
adsorption of it on to the surface of the enzyme then its biodegradation to glucose
with the synergistic action of three major classes of enzymes: endoglucanases,
exoglucanases and β-glucosidases which are generally represented as cellulase or
cellulolytic enzymes.
All these enzymes are available in the cellulosome complexes, which are intri-
cate multienzyme machines produced by many cellulolytic microorganisms and are
designed for efficient degradation of plant cell wall polysaccharides. In the past
several years, selected studies have addressed the conformational flexibility of the
cellulosome and its possible significance to biomass degradation. Early on, it was
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 7

evident that isolated cellulosomes could attain an overall tight or loose conforma-
tion (Mayer et al. 1987; Lamed et al. 1983a). The cellulolytic enzymes are either
secreted into the substrate or attached to the cell wall of the microorganism. The
former system is called noncomplexed and the latter is complexed cellulase system.
Noncomplexed cellulase systems are mostly found in filamentous fungi and acti-
nomycete bacteria, because they can penetrate the lignocellulosic material with their
hyphal extensions. The enzymes of noncomplexed cellulase systems are then just
released into the substrate and the free enzymes start hydrolyzing the cellulose. The
glucose and cellodextrins of a length of maximal four glucose molecules are taken
up by the microorganism and either used directly or cleaved further via intracellular
hydrolases. Organisms that produce noncomplexed cellulase systems are most often
used in the industrial production of cellulolytic enzymes, because the secreted
enzymes can easily be harvested (Lynd et al. 2002).
Cellulosome contains cellulose-binding domains, also called carbohydrate
binding modules (CBM), which bind to the cellulosic material (Fontes and Gilbert
2010; Sweeney and Xu 2012; Shoseyov et al. 2006). CBMs can also be found in
noncomplexed cellulase systems, where they are especially important for binding
and processing of exoglucanases. The main function of the cellulosome is to hold
the microorganism tight to the cellulose, and therefore reduce diffusion distances
and losses and to arrange the different cellulolytic enzymes in a favorable way.
More recently (Garcia-Alvarez et al. 2013), ultrastructural studies of a homoge-
neous mini-cellulosome containing three cohesin modules attached to three
matching cellulases suggested that the flexibility of the linkers connecting con-
secutive cohesin modules could control structural transitions and thus regulate
substrate recognition and degradation (Vazana et al. 2013).
One of the most potent cellulose degrading microorganisms is the well-studied
bacterium Clostridium thermocellum. This anaerobic thermophilic cellulolytic
bacterium secretes a multienzymatic complex called the cellulosome, first discov-
ered in 1983 (Lamed et al. 1983a, b). Later there are several reports on cellulosomal
oraganisms which possess an array of cellulosomal architectures (Belaich et al.
1997; Gal et al. 1997; Ding et al. 1999, 2000; Ohara et al. 2000; Rincon et al. 2003,
2004, 2010; Xu et al. 2003, 2004; Bayer et al. 2004, 2013; Dassa et al. 2012).

1.5 Microbial Enzymes for Degradation of Cellulose

The isolation and characterization of novel glycoside hydrolases from eubacteria


are now becoming widely exploited. There are several reasons for these shifts, for
one, bacteria often have a higher growth rate than fungi allowing for higher
recombinant production of enzymes. Secondly, bacterial glycoside hydrolases are
often more complex and are often expressed in multienzyme complexes providing
increased function and synergy. Most importantly, bacteria inhabit a wide variety of
8 M.P. Raghavendra et al.

environmental and industrial niches, which produce cellulolytic strains that are
extremely resistant to environmental stresses. These include strains that are ther-
mophilic or psychrophilic, alkaliphilic, or acidophilic, and, strains that are halo-
philic. Not only can these strains survive the harsh conditions found in the
bioconversion process, but also they can produce enzymes that are stable under
extreme conditions which may be present in the bioconversion process and this may
increase rates of enzymatic hydrolysis, fermentation, and, product recovery (Maki
et al. 2009). Along with these due to availability of novel techniques for gene
transfer, microbes can be genetically modified easily to suit the need of the
industrial processes and also the different substrates employed in the fermentation.
Considering these, decomposition of lignocelluloses by enzymes produced by
complex microbial communities represents a promising alternative for biomass
conversion. In particular, thermophilic consortia are a potential source of enzymes
adapted to adverse reaction conditions (Wongwilaiwalina et al. 2010; Gladden
et al. 2011). Members of the genus Caldicellulosiruptor can grow on and degrade
biomass containing high lignin content as well as highly crystalline cellulose
without conventional pretreatment (Blumer-Schuette et al. 2008; Yang et al. 2009)
and are also known to ferment all primary C5 and C6 sugars from plant biomass
and are the most thermophilic cellulolytic bacteria known, with growth temperature
optima between 78 and 80 °C raising the possibility of further economic
improvement of biofuel production from plant biomass by reducing or eliminating
the pretreatment step (Hamilton-Brehm et al. 2010).
In comparison with the anaerobic thermophilic bacteria few aerobic have been
described to produce cellulases and xylanses. The aerobic thermophiles R. marinus
produces a highly thermostable cellulose (Cel 2A), and three glycoside hydrolases
belonging to family GH10 of the carbohydrate active enzyme (CAZy) database
(http://www.cazy.org) (Cantarel et al. 2009). Aquifex aeolicus, isolated in the
Aeolic Islands in Sicily (Italy), represents one of the most thermophilic bacteria
since its growth temperature can reach 95 °C (Deckert et al. 1998).
Bacteria belonging to Clostridium, Ruminococcus, Bacteroides, Erwinia,
Acetovibrio, Microbispora, and Streptomyces can produce cellulases (Bisaria
1991). Cellulomonas fimi and Thermomonospora fusca have been extensively
studied for cellulase production. Similarly, other bacterial strains have the ability to
produce cellulase complexes aerobically as well as anaerobically. Cellulase pro-
ducing bacterial strains of Rhodospirillum rubrum, Cellulomonas fimi, Clostridium
stercorarium, Bacillus polymyxa, Pyrococcus furiosus, Acidothermus cellulolyticus,
and Saccharophagus degradans have been extensively reviewed (Kumar et al.
2008; Weber et al. 2001; Kato et al. 2005; Taylor et al. 2006). An extracellular
alkaline carboxy methyl cellulase (CMCase) from Bacillus subtilis strain AS3 has
been purified and characterized by Deka et al. (2013) for utilization of cellulosic
biomass. Although many cellulolytic bacteria, particularly the cellulolytic anaer-
obes such as Clostridium thermocellum and Bacteroides cellulosolvens produce
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 9

cellulases with high specific activity, they do not produce high enzyme titres (Lynd
et al. 2002).
Apart from the cellulolytic fungus T. reesei, many other fungi produce cellulases
and degrade treated cellulosic material or soluble cellulose derivatives such as
carboxymethylcellulose. However, they are not very effective on crystalline cel-
lulosic substrates. Mesophilic strains producing cellulases like Fusarium oxyspo-
rum, Piptoporus betulinus, Penicillium echinulatum, P. purpurogenum, Aspergillus
niger and A. fumigatus have also been reported (Martins et al. 2008; Valaskova and
Baldrian 2006). The cellulases from Aspergillus usually have high β-glucosidase
activity but lower endoglucanase levels, whereas, Trichoderma has high endo and
exoglucanase components with lower β-glucosidase levels, and hence has limited
efficiency in cellulose hydrolysis. Thermophillic microorganisms such as
Sporotrichum thermophile, Scytalidium thermophilum, Clostridium straminisolvens
and Thermomonospora curvata also produce the cellulase complex and can degrade
native cellulose (Kato et al. 2005; Kaur et al. 2004).
Even human gut microbiota encodes a huge diversity of enzymes for the
digestion of all components of plant cell wall polysaccharides including cellulose.
Turnbaugh et al. (2010) have shown that the distal gut microbiota of humans also
encodes dockerin-containing cellulolytic enzymes that indicate the presence of
cellulosomes (Fontes and Gilbert 2010; Bayer et al. 2004). The presence of
dockerin-containing proteins is also observed in the oral, nasal and vaginal samples
(Cantarel et al. 2012). In nondigestive sites, however, these dockerin domains may
have another role than attaching cellulases to form a cellulosome.
The interaction between loads of bacteria in gut with other bacteria has lead to
development of novel CAZymes. The recent outcome of the research by Hehemann
et al. (2010) proved the novel CAZymes added due to flexible microbial interac-
tions. They characterized the first porphyranases from a member of the marine
Bacteroidetes, Zobellia galactanivorans, active on the sulphated polysaccharide
porphyran from marine red algae of the genus Porphyra. Furthermore, they showed
that genes coding for these porphyranases, agarases and associated proteins have
been transferred to the gut bacterium Bacteroides plebeius isolated from Japanese
individuals. Their comparative gut metagenome analyses showed that por-
phyranases and agarases are frequent in the Japanese population and that they are
absent in metagenome data from North American individuals.
White Rot Fungi (WRF) are microorganisms of great interest that secrete
complex suites of nonspecific extracellular ligninolytic enzymes, i.e., lignin per-
oxidase (EC 1.11.1.14), manganese peroxidase (EC 1.11.1.13), and laccase (EC
1.10.3.2) to biodegrade lignin or digest substrates required for their proliferation
and growth. β-glucosidase is among the suite of enzymes produced by WRF such as
Pleurotus ostreatus, Auricularia auricula, Polyporus squamosus, Trametes versi-
color, Lentinula edodes, and Grifola frondosa to biodegrade plant biomass. The
β-glucosidase family (EC 3.2.1.21) is a wide-spread group of enzymes that catalyze
10 M.P. Raghavendra et al.

the hydrolysis of a broad variety of glycosides (Berrin et al. 2003). While some
organisms secrete either endoglucanase or β-glucosidase, in other organisms;
β-glucosidase is either lacking or produced in insufficient quantities (Kumar et al.
2008). When β-glucosidase secretion is low, cellobiose accumulates instead of
glucose. Cellobiose accumulation acts as a feedback-inhibitor of cellulose
depolymerization by endo- and exoglucanases (Howell and Stuck 1975; Morais
et al. 2010), which is a critical factor in the industrial scale conversion of cellulose
to glucose (Cai et al. 1997). This situation can be alleviated during industrial scale
conversion of cellulosic biomass by exogenous incorporation of β-glucosidase
enzyme.
Among known cellulolytic microorganisms, cellulases of Trichoderma reesei or
T. viride are well studied and characterized. Cellulase of T. reesei comprises of two
major cellobiohydrolases, (CBHI and CBHII), two major endoglucanases, (EGI and
EGII) and at least two low molecular weight endoglucanases, EGIII, and EGV. The
mixture of these enzymes is capable of solubilizing native cellulose. Many other
aerobic filamentous fungi, including Agaricus bisporus, Humicola spp., Irpex
lacteus, and Sclerotium roysii, also use similar cellulase systems.

1.6 Novel Enzymes for Cellulose Degradation

In addition to enzymes that act directly on polysaccharides, lignocelluloses


degrading microorganisms are also found to secrete several proteins, which modify
cellulose and enhance its hydrolysis by cellulase. These include the no-enzymatic
proteins such as ‘expansins’ and their fungal and bacterial homologs, the ‘swol-
lenins’ expressed by T. reesei with sequence homology to plant expansins and the
similar ‘loosenins’ produced by Bjerkandera adusta. These interact with cellulosic
substrates resulting in expansion, slippage, or lengthening of the components,
thereby facilitating the access of glycosyl hydrolases (Sweeney and Xu 2012;
Banerjee et al. 2010). Recently, cellulose-induced proteins (CIP1 and CIP2) have
been found in a transcriptional analysis of T. reesei, with some synergistic activity
with both GH61 and swollenin and are thought to play a role in the cleavage of
hemicellulose–lignin crosslinks (Sweeney and Xu 2012; Scott et al. 2009). There
are various hydrolytic enzymes and proteins produced by fungi and bacteria, which
help to facilitate the lignocellulose bioconversion process.
Along with the catalytic core, many of the enzymes also possess noncatalytic
domains including CBM and dockerins which, respectively, anchor the enzyme to
targeted substrate or onto scaffolding to assemble a cellulosome and disrupt the
crystalline cellulose microfibrils (Kumar et al. 2008; Sweeney and Xu 2012).
Recently, a new type of bacterial proteins currently classified as CBM33 (family 33
CBM) in the CAZy database and fungal proteins classified as GH61 (family 61
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 11

glycoside hydrolases) that catalyze oxidative cleavage of polysaccharides have been


discovered (Horn et al. 2012; Sweeney and Xu 2012). These copper dependent
monooxygenases act on the surfaces of insoluble substrates where they introduce
chain breaks in the polysaccharide chains thereby increasing substrate accessibility
and potentiating hydrolytic enzymes (Horn et al. 2012). Recent data obtained from
CBM33 and GH61 families revealed that these enzymes disrupt the more recalci-
trant portions of the cellulose and make the substrate more accessible to digestion
by cellulases. The emerging research on disruptive proteins from the CBM33 and
GH61 is exciting because these proteins appear to have novel mechanism to attack
cellulose and synergize with cellulase system (Kostylev and Wilson 2012). Studies
related to synergistic activity with high throughput advanced screening will yield
efficient method to degrade cellulose with these enzymes.
Inspired by the findings for CBP21 and earlier indications that certain CBM33s
may act synergistically with cellulases (Moser et al. 2008) studies were conducted
to prove if certain CBM33s act on cellulose like CBP21 acts on chitin. In 2011 it
was then shown that CelS2, a CBM33 protein from Streptomyces coelicolor, indeed
cleaves cellulose, producing aldonic acids (Forsberg et al. 2011). Like CBP21, the
activity of CelS2 depended on the presence of divalent metal ions as shown by the
inhibitory effect of EDTA and the ability to restore activity by adding divalent metal
ions. Again like CBP21, purified CelS2 was active without the addition of metals,
probably due to high affinity binding. Both the initial study on CBP21 and the study
on CelS2 concluded that the enzymes could use several divalent metal ions, but the
most recent work clearly shows that these enzymes in fact are copper dependent
monooxygenases.
At the same time, a series of elegant studies showed that GH61s are functionally
very similar to CBM33s (Quinlan et al. 2011; Beeson et al. 2012; Phillips et al.
2011; Westereng et al. 2011; Li et al. 2012). Quinlan et al. (2011) described the
crystal structure of a GH61 from Thermoascus aurantiacus (TaGH61A) and
showed that this protein catalyzes oxidative cleavage of cellulose in the presence of
an external electron donor such as gallic acid. These authors were the first to
convincingly show that enzyme activity is copper dependent. These findings were
confirmed by work on a GH61 from Phanerochaete chrysosporium (PcGH61D) by
Westereng et al. (2011) and work on several GH61 proteins from Neurospora
crassa (Beeson et al. 2012; Phillips et al. 2011; Li et al. 2012). Thus, GH61s are
copper dependent lytic polysaccharide monooxygenases. Recent work on a
chitin-active CBM33, using experimental conditions that ruled out possible effects
of metal ions trapped in the substrate, showed that this CBM33 was copper
dependent too (Vaaje-Kolstad et al. 2005).
The list of enzymes and noncatalytic proteins associated with degradation of
lignocellulose is increasing every year, the recent list of novel microbial enzymes
and its functions are given in Table 1.1 (Mohanram et al. 2013).
12 M.P. Raghavendra et al.

Table 1.1 Novel enzymes and other proteins involved in degradation of biomass
Enzymes Function Microorganisms
Endo-1,4 β-D- Hydrolysis of the internal Acidothermus cellulolyticus,
glucanglucanohydrolase glycosidic linkages in a Pectobacterium
(EC. 3. 2. 1.4) random fashion, generating chrysanthami, Thermobifida
oligosaccharides of varying fusca, Thermotoga maritime,
lengths Bacillus, Pseudomonas,
Ruminococcus, Fibrobacter,
Clostrdium, Halomonas,
Streptomyces, Cellulomonas,
Mycobacterium, Aspergillus,
Trichoderma, Anaeromyces,
Pestalotiopsis,
Phanerochaete, Fusarium,
Orpinomyces, Piromyce
Exoglucanase or 1,4-β-D- Hydrolysis of beta-D- Bacillus, Pseudomonas,
glucan cellobiohydrolase glucosidic linkages by Clostridium, Paenibacillus,
(EC3.2.1.91) releasing mainly cellobiose Thermobifida, Cellulomonas,
either from the reducing or Mycobacterium, Ralstonia,
nonreducing ends of the Trichoderma, Penicillium,
chains Aspergillus, Chaetomium,
Fusarium, Pestalotiopsis,
Orpinomyces, Piromyces,
Rhizopus
β-glucosidases or β-D- Hydrolysis of terminal, Clostridium, Cellulomonas,
glucoside glucohydrolase nonreducing β-D-glucosyl Aerobacter, Leuconostoc,
(EC 3.2.1.21) residues with release of β-D- Aspergillus, Monilia,
glucose Phanerochaete, Sclerotium,
Saccharomyces,
Kluyveromyces
Cellodextrin phosphorylase Catalysis of the reversible Clostridium, Cellvibrio
or (1 → 4)-β-D-glucan: phosphorolytic cleavage of
phosphate α-D- cellodextrins ranging from
glucosyltransferase (EC cellotriose to cellohexoses
2.4.1.49)
Cellobiose phosphorylase Catalysis of the reversible Cellulomonas, Clostridium,
or cellobiose: phosphate phosphorolytic cleavage of Ruminococcus, Thermotoga,
α-D-glucosyltransferase cellobiose Cellvibrio, Fomes annosus
(EC 2.4.1.20)
Endo-1,4-β-D-xylanase or Endohydrolysis of (1 → 4)- Bacillus circulans,
1,4-beta-xylan beta-D-xylosidic linkages in Thermoanaerobacterium,
xylanohydrolase xylans to release xylose Ruminococcus, Geobacillus,
(EC 3.2.1.8) Thermopolyspora,
Cellulomonas, Streptomyces,
Aspergillus, Trichoderma,
Thermomyces, Fusarium,
Anaeromyces,
Neocallimastix,
Dictyoglomus
Thermophilum, Streptomyces
halstedii
(continued)
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 13

Table 1.1 (continued)


Enzymes Function Microorganisms
Xylan β-1,4-xylosidase Hydrolysis of (1 → 4)-beta- Bacillus,
(EC 3.2.1.37) D-xylans, to remove Thermoanaerobacterium,
successive D-xylose residues Geobacillus, Aspergillus,
from the non-reducing Fusarium, Talaromyces,
termini Trichoderma
Mannan Random hydrolysis of Cellulomonas, Bacillus,
endo-1,4-β-mannosidase (1 → 4)-beta-D-mannosidic Clostridium, Rhodothermus,
(EC 3.2.1.78) linkages in mannans, Aspergillus, Trichoderma
galactomannans and
glucomannans
α-L-arabinofuranosidase Hydrolysis of terminal Bifidobacterium,
(EC 3.2.1.55) nonreducing alpha-L- Thermobacillus, Bacillus,
arabinofuranoside residues in Clostridium, Streptomyces,
alpha-L-arabinosides Aspergillus, Penicillium,
Fusarium, Trichoderma
Acetyl (xylan) esterase Hydrolysis of ester linkages Bacillus, Clostridium,
(EC 3.1.1.72) of the acetyl groups in Streptomyces, Fibrobacter,
position 2 and/or 3 of the Pseudomonas,
xylose moieties of natural Thermoanaerobacterium,
acetylated xylan Penicillium, Aspergillus,
Trichoderma,
Phanerochaete,
Chrysosporium
α-L-Fucoside fucohydrolase Hydrolysis of O-glycosyl Thermotoga,
or α-L-fucosidase bond in xyloglucan to release Bifidobacterium,
(EC 3.2.1.51) l-fucose residues Streptococcus, Bacillus,
Cellulomonas, Clostridium,
Fusarium, Beauveria,
Penicillium, Trichoderma,
Neurospora, Aspergillus
α-D-Glucosiduronate Hydrolysis of O-glycosyl Thermotoga, Cellvibrio,
glucuronohydrolase or bond to release 4-O- Bacteroides, Bacillus,
α-glucuronidase methylglucuronic acid from Aspergillus, Trichoderma,
(EC 3.2.1.139) xylan Thermoascus
Novel proteins
Swollenins Homologous to plant Bacillus, Cellulomonas,
expansins which rapidly Clostridium,
induce extension of plant cell Myceliophthora,
walls by weakening the Thermomonospora,
noncovalent interactions; Streptomyces, Fibrobacter,
contain an N-terminal Trichoderma, Aspergillus,
carbohydrate binding module Neosartorya, Humicola,
family 1domain (CBD) with Fusarium, Penicillium,
cellulose-binding function Neurospora, Gliocladium,
and a C-terminal Candida, Pichia,
expansin-like domain Rhodotorula,
Sporobolomyces,
Bjerkandera adusta
(continued)
14 M.P. Raghavendra et al.

Table 1.1 (continued)


Enzymes Function Microorganisms
Cellulose induced proteins Contain a carbohydrate Streptomyces coelicolor,
(CIP1 and CIP2) binding module (CBM); Trichoderma, Schizophyllum,
Hydrolysis of the ester Pyrenophora
linkage between 4-O-methyl-
D-glucuronic acid of
glucuronoxylan and lignin
alcohols
Lossenins Facilitate acess of glycosyl Bjerkandera adusta
hydrolases
Dockerin and Cohesin Assembly of cellulase and Anaerobic microorganisms
hemicellulase proteins to
structural scaffoldings on
microbial surface

1.7 Swollenins: A Novel Protein for Cellulose Degradation

Expansins in plants have been proposed to disrupt hydrogen bonding between


cellulose microfibrils or between cellulose and other cell wall polysaccharides
without having hydrolytic activity (McQueen-Mason and Cosgrove 1994; Whitney
et al. 2000). In this way they are thought to allow the sliding of cellulose fibers and
enlargement of the cell wall. Saloheimo et al. (2002) reported the discovery of a
novel fungal protein having significant sequence identity to plant expansins. Unlike
plant expansins, this protein has a modular structure with an N-terminal CBD. The
protein was named swollenin due to its ability to swell cotton fibers and loosening
effect of swollenin makes lignocellulosic biomass more accessible and readily
hydrolyzable by cellulase, thereby promoting the degradation of lignocellulose
during fermentation (Arantes and Saddler 2010; Chen et al. 2010) without pro-
ducing detectable amounts of reducing sugars. Swollenin was later shown to be
capable of weakening and disrupting hydrogen-bond networks in lignocellulose
(Wang et al. 2011). Swollenin has an N-terminal fungal type cellulose-binding
domain connected by a linker region to the expansin-like domain. The protein also
contains regions similar to mammalian fibronectin type III repeats, found for the
first time in a fungal protein. The swollenin gene is regulated in a largely similar
manner as the T. reesei cellulase genes.
Efforts were made by different groups to express swollenin gene heterologously
in Saccharomyces cerevisiae and Aspergillus niger (Saloheimo et al. 2002) and
Kluyveromyces lactis (Jäger et al. 2011). In continuation to improve the direct
conversion of cellulose to ethanol, yeast Saccharomyces cerevisiae co-displaying
cellulase and expansin-like protein on the cell surface were constructed and
examined for direct ethanol fermentation performance. The cellulase and
expansin-like protein co-expressing strain showed 246 mU/g-wet cell of
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 15

phosphoric acid swollen cellulose degradation activity, which corresponded to


2.9-fold higher activity than that of a cellulase-expressing strain. This result clearly
demonstrated that yeast cell surface expressed cellulase and expansin-like protein
act synergistically to breakdown cellulose. In fermentation experiments examining
direct ethanol production from PASC, the cellulase and expansin-like protein
co-expressing strain produced 3.4 g/L ethanol after 96 h of fermentation, a con-
centration that was 1.4-fold higher than that achieved by the cellulase-expressing
strain (2.5 g/L) (Nakatani et al. 2013).
Its exploitation for removal of detectable amounts of reducing sugars has been
recently reported by Kang et al. (2013). They have isolated a novel gene encoding a
swollenin-like protein, POSWOI, from the filamentous fungus Penicillium oxali-
cum by Thermal Asymmetric Interlaced PCR (TAIL-PCR). It consisted of a family
1 carbohydrate binding module (CBM1) followed by a linker connected to a family
45 endoglucanase-like domain. Using the cellobiohydrolase I promoter, recombi-
nant POSWOI was efficiently produced in T. reesei with a yield of 105 mg/L, and
showed significant disruptive activity on crystalline cellulose. Simultaneous reac-
tion with both POSWOI and cellulases enhanced the hydrolysis of crystalline
cellulose Avicel by approximately 50 %. Using a POSWOI-pretreatment proce-
dure, cellulases can produce nearly twice as many reducing sugars as without
pretreatment. The mechanism by which POSWOI facilitates the saccharification of
cellulose was also studied using a cellulase binding assay.

1.8 CIP1 and CIP2

Cellulose-induced protein 1 (Cip1) protein consists of a glycoside hydrolase family


1 carbohydrate binding module connected via a linker region to a domain.
A calcium ion binding site was identified in a sequence conserved region of Cip1
and is also seen in other proteins with the same general fold as Cip1, such as many
carbohydrate binding modules. The Cip1 structure was analyzed and a structural
homology search was performed to identify structurally related proteins. The two
published structures with highest overall structural similarity to Cip1 found were
two poly-lyases: CsGL, a glucuronan lyase from H. jecorina and vAL-1, an alginate
lyase from the Chlorella virus. This indicates that Cip1 may be a lyase. However,
initial trials did not detect significant lyase activity for Cip1 (Jacobson et al. 2013).
The function of CIP1 is not known yet. BLAST searches of CIP1 in public data-
bases show that it is similar to glycosyl hydrolases found in bacteria but that are not
yet characterized in detail, indicating that CIP1 could have a novel undescribed
enzymatic activity or mechanism. CIP2 was described to encode a CE15 glu-
curonoyl esterase, probably cleaving the ester linkage between 4-O-methyl-D-glu-
curonic acid of glucuronoxylan and lignin alcohols (Li et al. 2007).
16 M.P. Raghavendra et al.

1.9 Consolidated Bioprocessing (CBP) for Enhanced


Biofuel Production

CBP can be achieved by two ways by modifying cellulase producer to ferment


glucose or to modify glucose producer to produce celulase. CBP may result in poor
yield of alcohol and requires more incubation period and hence thermophilic strains
are considered to be best alternative and above said combination is appears to be
practically viable. It represents an alternative method with outstanding potential for
low-cost processing of lignocellulosic biomass (Lynd et al. 2008). Fujita et al.
(2004) constructed a whole-cell biocatalyst with the ability to induce synergistic
and sequential cellulose hydrolysis reaction through co display of three types of the
cellulololytic enzymes on the cell surface of S. cerevisiae. This strain achieved
successful conversion of cellulose to ethanol with good yield. In another attempt
Den et al. (2007) developed recombinant strain of S. cerevisiae which can be used
for CBP. Two cellulase-encoding genes, an endoglucanse of T. reesei and
β-glucosidase of Saccharomyces fibuligera in combination were expressed in
S. cerevisiae.
No ideal CBP microbe able to degrade efficiently lignocelluloses and at the same
time, to utilize the released sugars to produce ethanol is currently available.
A newly discovered thermophilic microorganism, Geobacillus sp. R7 (T 60 °C), is
a facultative anaerobic bacterium isolated from soil samples of the Homestake gold
mine, South Dakota. It produces a thermostable cellulose when grown on
extrusion-pretreated agricultural residues such as corn stover and prairie cord grass,
and ferments lignocellulosic substrates to ethanol in a single step (Zambare et al.
2011).
Cha et al. (2013) demonstrated recently the utility of gene deletion in the
pyruvate metabolic pathway for rational strain engineering of Caldicellulosiruptor
bescii while simultaneously creating a platform for further strain modification for
advanced production of fuels and chemicals from renewable plant feed stocks. The
method for creating a deletion of the ldh gene in the C. bescii chromosome was
efficient enough to allow targeted marker replacement using nonreplicating plas-
mids. The resulting mutant grew to a higher cell density and produced more
hydrogen than the wild-type strain. Using the tools developed by them, C. bescii
JWCB017 will serve as a platform for additional rational strain engineering for
production of fuels and chemicals from lignocellulosic feedstocks.
Efforts were also continued to transfer genes responsible for degradation of
cellulose to yeast. Cellulase genes from various kinds of microorganisms have been
expressed in the ethanol-producing yeast Saccharomyces cerevisiae with the aim of
directly producing ethanol from cellulose (Van and Van 1998; Fujita et al. 2002; Ito
et al. 2009; Wen et al. 2010; Yamada et al. 2013). In addition, yeast strains dis-
playing cellulase on the cell surface have also been developed for improving the
efficiency of direct ethanol production from cellulose (Yamada et al. 2013).
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 17

1.10 Recycling of Enzymes

A key factor that prevents the commercialization of enzymatic cellulose hydrolysis


is the high cost of cellulase enzymes. Enzyme cost is expected to account for more
than 20 % of ethanol production (Wooley et al. 1999). As much of it remains active
after hydrolysis, recycling of cellulases makes the overall conversion process more
economically feasible. Various methods have been used for recycling enzymes,
which include sedimentation followed by ultra filtration or micro centrifugation,
cation exchange chromatography, readsorption, and immobilization.

1.11 Potential Strategies for Stabilizing Cellulases

Enzyme stabilization and lifetime improvement has been achieved through point
substitution and chemical modification, of single catalytic domains, either through
rational, structure-based modeling, or through directed evolution. Another approach
to increase enzyme stability and enhance activity is insertion of protein domains
into stable scaffolds.
An effort was made to stabilize the cellulases by embedding cellulases of
extremophiles into hyperstable α-helical consensus. It was observed that catalytic
domains CelA (CA, GH8; Clostridium thermocellum) and Cel12A (C12A, GH12;
Thermotoga maritima) to be stable in the context of ankyrin scaffold, and to be
active against both soluble and insoluble substrates. The ankyrin repeats in each
fusion are folded, although it appears that for C12A catalytic domain (CD) (where
the N- and C-termini are distant in the crystal structure), the two flanking ankyrin
domains are independent, whereas for CA (where termini are close), the flanking
ankyrin domains stabilize each other. Although the activity of CA is unchanged in
the context of the ankyrin scaffold, the activity of C12A is increased between two
and sixfold (for RAC and CMC substrates) at high temperatures. For C12A, activity
increases by with the number of flanking ankyrin repeats. These results show
ankyrin arrays to be a promising scaffold for constructing designer cellulosomes,
preserving or enhancing enzymatic activity and retaining thermostability.
In this type of scaffolding strategy, the distance between the N and C-termini of
the inserted protein (cellulase catalytic domains) is an important parameter.
Cellulases catalytic domains belong to various Glycoside Hydrolase (GH) families,
as reported in the CAZY database, with different folds, and thus have variable
N-to-C distances (Cantarel et al. 2009).
Insertion into a scaffold protein requires appropriate selection of insertion sites to
avoid disruption of structure of the inserted protein or scaffold (Betton et al. 1997;
Cutler et al. 2009). Proteins with linear, repeated architectures, such as ankyrin
repeats, have structurally modular architectures that should be able to accommodate
insertions locally, without affecting more distant regions of the scaffold. Ankyrin
repeat proteins are roughly linear arrays of 33 residues repeating units (Sedgwick
18 M.P. Raghavendra et al.

and Smerdon 1999; Zweifel and Barrick 2001). Each unit consists of two short
helices connected by a short, conserved turn, and connects to neighboring repeats
through an extended beta-hairpin containing loop (Zweifel et al. 2003). Adjacent
repeats strongly stabilize each other through highly stabilizing interfaces. The
repeating unit spans about 11 Å, approximately the size of each cellobiose unit
(2-units of glucose 10.4 Å). Studies have found that ankyrin repeats are greatly
stabilized by consensus sequence substitutions (Wetzel et al. 2008; Aksel et al.
2011). For a five repeat consensus ankyrin protein, the thermal denaturation mid-
point (Tm) was around 90 °C (Aksel et al. 2011; Aksel and Barrick 2009).
Therefore, consensus ankyrin seems to be an ideal scaffold to study the effects of
inserting cellulase CDs with various N–C distances.

1.12 Enzyme Cocktails

Catalytic activity of an enzyme is defined and mediated by its 3D structure and its
mobility. It is impossible to obtain complete comprehension of the enzymatic action
without detailed molecular studies which include structural, biophysical, bio-
chemical and genetic investigations, enzymatic assays, bioinformatics, and
molecular dynamics computations. It is not possible to innovate and to consistently
produce competitive enzymatic cocktails/mixtures for biomass transformation
without profound understanding of function and activity of glycoside hydrolases.
Therefore, we need to integrate our knowledge of biodiversity, genomics and
genetics studies, structural and molecular biology, biochemistry, enzymology, and
bioinformatics, focusing on bioindustrial applications.
Enzyme cocktails that hydrolyze plant cell wall polysaccharides are a critical
component of bioprocessing configurations designed to transform lignocellulosic
biomass into biofuels (Lynd et al. 2002; Gao et al. 2011). Specific characteristics of
an enzyme (activity, thermostability, etc.) is often based on homology to known
enzymes (Schnoes et al. 2009). Although free cellulases and cellulosomes employ
different physical mechanisms to break down recalcitrant polysaccharides, when
these system combined display dramatic synergistic enzyme activity on cellulose.
Two natural enzyme systems—one produced by fungi and the other by bacteria—
break down cellulose faster if used in combination. The resulting process shows
promise for less expensive biofuels (Resch et al. 2013).
Current commercial cocktails consist of preparations of fungus derived glycoside
hydrolases, primarily cellulases and hemicellulases (Bouws et al. 2008; King et al.
2009). However, fungal enzymes are often deactivated by elevated temperatures or
by residual chemicals from pretreatment (Bouws et al. 2008). For example, ionic
liquids (ILs), such as 1-ethyl- 3-methylimidazolium acetate ([C2mim][OAc]), can
dissolve lignocellulosic biomass and dramatically improve cellulose hydrolysis
kinetics, yet multiple studies have shown that fungal endoglucanases are deacti-
vated at low levels of ILs that may persist in the biomass after pretreatment (Dadi
et al. 2006; Turner et al. 2003). In contrast, thermophilic bacterial and archaeal
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 19

endoglucanases have been shown to be active in 20 % [C2mim][OAc], suggesting


that thermophilic prokaryotes may be an important source of enzymes for the
development of more robust enzyme cocktails (Datta et al. 2010).
Although free cellulases and cellulosomes employ very different physical
mechanisms to break down recalcitrant polysaccharides, when combined these
systems display dramatic synergistic enzyme activity on cellulose. Two natural
enzyme systems—one produced by fungi and the other by bacteria—break down
cellulose faster if used in combination. The resulting process shows promise for less
expensive biofuels (Resch et al. 2013).

1.13 Enzyme Engineering

The natural diversity of enzymes could provide a large reservoir that can be further
improved by engineering enzymes and strains for increased performance (King
et al. 2009). A number of designer enzymes called glycosynthases, including cel-
lulases and hemicellulases, have been engineered by replacing nucleophilic residues
resulting in higher yields of different oligosaccharides (Kumar et al. 2008).

1.14 Feruloyl Esterases (Faes)

Feruloyl esterases (Faes) represent a subclass of carboxyl esterases that can release
phenolic acids, such as ferulic acids or other cinnamic acids, from esterified
polysaccharides, especially xylan and pectin. Feruloyl esterases (Faes) constitute a
subclass of carboxyl esterases that specifically hydrolyze the ester linkages between
ferulate and polysaccharides in plant cell walls. Recently, a comprehensive set of
enzymes essential for decomposing plant cell walls, including feruloyl esterase
activity, in a newly described in yak ruminal bacterium, Cellulosilyticum
ruminicola H1. Strain H1 grew robustly on natural plant fibers, such as corn cob,
alfalfa, and ryegrass, as the sole carbon and energy sources, as well as on a variety
of polysaccharides, including cellulose, xylan, mannan, and pectin (Li et al. 2011).
Therefore, Faes are regarded as the key enzymes to loosen the internal crosslink of
plant cell walls by acting as the important accessory enzymes in synergy with
(hemi)cellulases in plant cell wall hydrolysis.

1.15 Future Scope

Urgent need of the alternate energy resources increased the novel ideas to achieve
success to convert lignocelluloses and other carbohydrates to ethanol. In this
connection development of systems biology approaches and integrated, predictive
20 M.P. Raghavendra et al.

modeling capabilities for metabolic and regulatory networks of biomass degrading


microorganisms is considered to be very important. The characterization of
microbes and microbial communities from environments with high rates of ligno-
cellulose degradation has to be continued, because microorganisms display varied
capacity for carbohydrate degradation.
The following are the thrust area of research already initiated by several
scientists
• Designing of optimized cellulosomes by synthesizing hybrid scaffolding
molecules that contain cohesins with different binding specificity from different
organisms is another recent approach to develop more active cellulose degrading
enzymes (Wilson 2009). Synergies between purified cellulases and xylanases
from the thermophilic bacterium, Thermobifida fusca displayed on designer
cellulosomes were found to possess higher activity on wheat straw than the
corresponding free enzymes (Morais et al. 2010).
• The recent trend is developing multifunctional chimeras by construction,
cloning and sequencing the chimeric gene and its expression followed by
purification of chimeric proteins (Nair et al. 2010).
• Brunecky et al. (2013) proposed a novel mechanism of cellulose digestion by
the dominant multimodular cellulase CelA of the thermophilic bacterium
Caldicellulosiruptor bescii and compared its hydrolytic performance with that
of a binary mixture containing Cel7A fungal exoglucanase (cellobiohydrolase I
from Trichoderma reesei) and bacterial Acidothermus cellulolyticus Cel5A (E1)
endoglucanase. The major finding of the report, that CelA is able to excavate
extensive cavities into the surface of the cellulosic substrate, is very interesting
and indeed reflects a novel cellulose digestion paradigm.
• There are efforts to develop, create, and investigate artificial cellulosomes with
high activity, and systematically evaluate the fundamental principles that
underlie their structure, dynamics, and catalytic functions. Ranjan (2014) have
developed a polycatalytic system consisting of cellulases covalently linked on
the surface of colloidal polymers with a magnetic nanoparticle (MNP) core.
MNP provides a convenient handle to separate the complex, while the colloidal
polymer would serve as a benign scaffold to attach the enzymes.
Although biomass may ultimately only supply a relatively small amount of the
world’s energy requirements, it will nevertheless be of immense overall value. In
some parts of the world, such as Brazil and other countries of similar climatic
conditions, biomass will surely attain wider exploitation and utilization. There may
still be some disadvantages when comparing it with coal or oil, but the very fact that
it is renewable and they are not must be the spur to further research. In time,
biomass will become much more easily and economically used as a source of
energy for mankind. Approaches like enzyme engineering, reconstitution of
enzyme mixtures and bioprospecting for superior enzymes are gaining importance.
The current scenario, however, also warrants the need for research and development
of integrated biomass production and conversion systems.
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 21

References

Aksel T, Barrick D (2009) Analysis of repeat-protein folding using nearest-neighbor statistical


mechanical models. Methods Enzymol 455:95–125
Aksel T, Majumdar A, Barrick D (2011) The contribution of entropy, enthalpy, and hydrophobic
desolvation to cooperativity in repeat-protein folding. Structure 19:349–360
Arantes V, Saddler JN (2010) Access to cellulose limits the efficiency of enzymatic hydrolysis: the
role of amorphogenesis. Biotechnol Biofuels 3:4–15
Banerjee G, Scott-Craig JS, Walton JD (2010) Improving enzymes for biomass conversion: a basic
research perspective. Bioenerg. Res. 3:82–92
Bayer EA, Belaich JP, Shoham Y, Lamed R (2004) The cellulosomes: multi-enzyme machines for
degradation of plant cell wall polysaccharides. Annu Rev Microbiol 58:521–554
Bayer EA, Shoham Y, Lamed R (2013) The prokaryotes: lignocellulose-decomposing bacteria and
their enzyme systems. In: Rosenberg E (ed) The prokaryotes, 4th edn. Springer, Berlin,
pp 216–266
Beeson WT, Phillips CM, Cate JHD, Marletta MA (2012) Oxidative cleavage of cellulose by
fungal copper-dependent polysaccharide monooxygenases. J Am Chem Soc 134:890–892
Belaich JP, Tardif C, Belaich A, Gaudin C (1997) The cellulolytic system of Clostridium
cellulolyticum. J Biotechnol 57:3–14
Berrin JG, Czjzek M, Kroon PA, McLauchlan WR, Puigserver A, Williamson G, Juge N (2003)
Sub- strate (aglycone) specificity of human cytosolic beta-glucosidase. Biochem J 373:41–48
Betton JM, Jacob JP, Hofnung M, Broome-Smith JK (1997) Creating a bifunctional protein by
insertion of beta-lactamase into the maltodextrin-binding protein. Nat Biotechnol
15:1276–1279
Bisaria VS (1991) Bioprocessing of agro-residues to glucose and chemicals. In: Martin AM
(ed) Bioconversion of waste materials to industrial products. Elsevier, London, pp 210–213
Blumer-Schuette SE, Kataeva I, Westpheling J, Adams MW, Kelly RM (2008) Extremely
thermophilic microorganisms for biomass conversion: status and prospects. Curr Opin
Biotechnol 19:210–217
Bouws H, Wattenberg A, Zorn H (2008) Fungal secretomes: nature’s toolbox for white
biotechnology. Appl Microbiol Biotechnol 80:381–388
Brunecky R, Alahuhta M, Xu Q, Donohoe BS, Crowley MF, Kataeva IA, Yang S-J, Resch MG,
Adams MMW, Lunin VV, Michael E, Himmel ME, Yannick J, Bomble YJ (2013) Revealing
nature’s cellulase diversity: the digestion mechanism of Caldicellulosiruptor bescii CelA.
Science 342:1513–1516
Cai Z, Xing G, Yan X, Xu H, Tsuruta H, Yagi K, Minami K (1997) Methane and nitrous oxide
emissions from rice paddy fields as affected by nitrogen fertilisers and water management.
Plant Soil 196:7–14
Cantarel BL, Coutinho PM, Rancurel C, Bernard T, Lombard V, Henrissat B (2009) The
carbohydrate-active enzymes database (CAZy): an expert resource for glycogenomics. Nucleic
Acids Res 37:233–238
Cantarel BL, Lombard V, Henrissat B (2012) Complex carbohydrate utilization by the healthy
human microbiome. PLoS ONE 7(6):121–131
Cha M, Daehwan C, James GE, Adam MG, Janet W (2013) Metabolic engineering of
Caldicellulosiruptor bescii yields increased hydrogen production from lignocellulosic biomass.
Biotechnol Biofuels 6:85–93
Chen XA, Ishida N, Todaka N, Nakamura R, Maruyama J, Takahashi H, Kitamoto K (2010)
Promotion of efficient saccharification of crystalline cellulose by Aspergillus fumigatus SWO1.
Appl Environ Microbiol 76:2556–2561
Cutler TA, Mills BM, Lubin DJ, Chong LT, Loh SN (2009) Effect of interdomain linker length on
an antagonistic folding-unfolding equilibrium between two protein domains. J Mol Biol
386:854–868
22 M.P. Raghavendra et al.

Dadi AP, Varanasi S, Schall CA (2006) Enhancement of cellulose saccharification kinetics using
an ionic liquid pretreatment step. Biotech. Bioeng. 95:904–910
Dassa B, Borovok I, Lamed R, Henrissat B, Coutinho P, Hemme L, Huang Y, Zhou J, Bayer EA
(2012) Genome-wide analysis of Acetivibrio cellulolyticus provides a blueprint of an elaborate
cellulosome system. BMC Genom 13:210–223
Datta S, Holmes B, Park JI, Chen Z, Dibble DC, Hadi M, Blanch HW, Simmons BA, Sapra R
(2010) Ionic liquid tolerant hyperthermophilic cellulases for biomass pretreatment and
hydrolysis. Green Chem 12:338–345
Deckert G, Warren PV, Gaasterland T, Young WG, Lenox AL, Graham DE, Overbeek R,
Snead MA, Keller M, Aujay M, Huber R, Feldman RA, Short JM, Olsen GJ, Swanson RV
(1998) The complete genome of the hyperthermophilic bacterium Aquifex aeolicus. Nature 392
(6674):353–358
Deka D, Jawed M, Goyal A (2013) Purification and characterization of an alkaline cellulase
produced by Bacillus subtilis (AS3). Prep Biochem Biotechnol 43:256–270
Den HR, Rose SH, Lynd LR, van Zyl WH (2007) Hydrolysis and fermentation of amorphous
cellulose by recombinant Saccharomyces cerevisiae. Metab Eng 9(1):87–94
Ding SY, Bayer EA, Steiner D, Shoham Y, Lamed R (1999) A novel cellulosomal scaffoldin from
Acetivibrio cellulolyticus that contains a family 9 glycosyl hydrolase. J Bacteriol
181:6720–6729
Ding SY, Bayer EA, Steiner D, Shoham Y, Lamed R (2000) A scaffoldin of the Bacteroides
cellulosolvens cellulosome that contains 11 type II cohesins. J Bacteriol 182:4915–4925
Eriksson T, Borjesson J, Tjerneld F (2002) Mechanism of surfactant effect in enzymatic hydrolysis
of lignocelluloses. Enz Microb Technol 31:353–364
Fontes CM, Gilbert HJ (2010) Cellulosomes: highly efficient nanomachines designed to
deconstruct plant cell wall complex carbohydrates. Annu Rev Biochem 79:655–681
Forsberg Z, Vaaje-Kolstad G, Westereng B, Bunæs AC, Stenstrøm Y, MacKenzie A, Sørlie M,
Horn SJ, Eijsink VGH (2011) Cleavage of cellulose by a CBM33 protein. Prot Sci
20:1479–1483
Fujita Y, Takahashi S, Ueda M, Tanaka A, Okada H, Morikawa Y, Kawaguchi T, Arai M,
Fukuda H, Kondo A (2002) Direct and efficient production of ethanol from cellulosic material
with a yeast strain displaying cellulolytic enzymes. Appl Environ Microbiol 68:5136–5141
Fujita Y, Iro J, Ueda M, Fukuda H, Kondo A (2004) Synergistic saccharification and direct
fermentation to ethanol, of amorphous cellulose by use of an engineered yeast strain
codisplaying three types of cellulolytic enzyme. Appl Environ Microbiol 70(2):1207–1212
Gal L, Pages S, Gaudin C, Belaich A, Reverbel-Leroy C, Tardif C, Belaich JP (1997)
Characterization of the cellulolytic complex (cellulosome) produced by Clostridium cellu-
lolyticum. Appl Environ Microbiol 63:903–909
Gao D, Uppugundla N, Shishir P, Chundawat S, Yu X, Hermanson S, Gowda K, Brumm P,
Mead D, Balan V, Dale BE (2011) Hemicellulases and auxiliary enzymes for improved
conversion of lignocellulosic biomass to monosaccharides. Biotechnol Biofuels 4:5–16
Garcia-Alvarez B, Melero R, Dias FM, Prates JA, Fontes CM, Smith SP, Romao MJ, Vazana Y,
Barak Y, Unger T, Peleg Y, Shamshoum M, Ben-Yehezkel T, Mazor Y, Shapiro E, Lamed R,
Bayer EA (2013) A synthetic biology approach for evaluating the functional contribution of
designer cellulosome components to deconstruction of cellulosic substrates. Biotechnol
Biofuels 6:182–200
Gladden JM, Allgaier M, Miller CS, Hazen TC, Vander Gheynst JS, Hugenholtz P, Simmons BA,
Singer SW (2011) Glycoside hydrolase activities of thermophilic bacterial consortia adapted to
switchgrass. Appl Environ Microbiol 77:5804–5812
Hamilton-Brehm SD, Mosher JJ, Vishnivetskaya T, Podar M, Carroll S, Allman S, Phelps TJ,
Keller M, Elkins JG (2010) Caldicellulosiruptor obsidiansis sp. nov., an anaerobic, extremely
thermophilic, cellulolytic bacterium isolated from Obsidian Pool, Yellowstone National Park.
Appl Environ Microbiol 76:1014–1020
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 23

Hehemann J, Correc G, Barbeyron T, Helbert W, Czjzek M, Michel G (2010) Transfer of


carbohydrate-active enzymes from marine bacteria to Japanese gut microbiota. Nature
464:908–912
Horn SJ, Vaaje-Kolstad G, Westereng B, Eijsink VGH (2012) Novel enzymes for the degradation
of cellulose. Biotechnol Biofuels 5:45–59
Howell JA, Stuck JD (1975) Kinetics of solka floc cellulose hydrolysis by Trichoderma viride
cellulase. Biotechnol Bioeng 17:873–893
Ito J, Kosugi A, Tanaka T, Kuroda K, Shibasaki S, Ogino C, Ueda M, Fukuda H, Doi RH,
Kondo A (2009) Regulation of the display ratio of enzymes on the Saccharomyces cerevisiae
cell surface by the immunoglobulin G and cellulosomal enzyme binding domains. Appl
Environ Microbiol 75:4149–4154
Jacobson F, Karkehabadi S, Hansson H, Goedegebuur F, Wallace L, Mitchinson C, Piens K,
Stals I, Sandgren M (2013) The crystal structure of the core domain of a cellulose induced
protein (Cip1) from Hypocrea jecorina, at 1.5 Å resolutions. PLoS ONE 8(9):e70562
Jäger G, Girfoglio M, Dollo F, Rinaldi R, Bongard H, Commandeur U, Fischer R, Spiess AC,
Büchs J (2011) How recombinant swollenin from Kluyveromyces lactis affects cellulosic
substrates and accelerates their hydrolysis. Biotechnol Biofuels 4:33–49
Kang K, Wang S, Lai G, Liu G, Xing M (2013) Characterization of a novel swollenin from
Penicillium oxalicum in facilitating enzymatic saccharification of cellulose. BMC Biotechnol
13:42–51
Kato S, Haruta S, Cui ZJ, Ishii M, Igarashi Y (2005) Stable coexistence of five bacterial strains as a
cellulose-degrading community. Appl Environ Microbiol 71:7099–7106
Kaur G, Kumar S, Satyanarayana T (2004) Production, characterization and application of a
thermostable polygalacturonase of a thermophilic mould Sporotrichum thermophile Apinis.
Bioresour Technol 94:239–243
King BC, Donnelly MK, Bergstrom GC, Walker LP, Gibson DM (2009) An optimized microplate
assay system for quantitative evaluation of plant cell wall-degrading enzyme activity of fungal
culture extracts. Biotechnol Bioeng 102:1033–1044
Kostylev M, Wilson D (2012) Synergistic interactions in cellulose hydrolysis. Biofuels 3(1):61–70
Kristensen JB, Boijesson J, Brunn MH, Tjerneld F, Jorgensen H (2007) Use of surface active
additives in enzymatic hydrolysis of wheat straw lignocelluloses. Enz Micro Technol
40:888–895
Kumar R, Singh S, Singh OV (2008) Bioconversion of lignocellulosic biomass: biochemical and
molecular perspective. J Ind Microbiol Biotechnol 35:377–391
Lamed R, Setter E, Bayer EA (1983a) Characterization of a cellulose-binding, cellulase-containing
complex in Clostridium thermocellum. J Bacteriol 156:828–836
Lamed R, Setter E, Kenig R, Bayer EA (1983b) The cellulosome—a discrete cell surface organelle
of Clostridium thermocellum which exhibits separate antigenic, cellulose-binding and various
cellulolytic activities. Biotechnol Bioeng Symp 13:163–181
Li XL, Spanikova S, de Vries RP, Biely P (2007) Identification of genes encoding microbial
glucuronoyl esterases. FEBS Lett 581(21):4029–4035
Li J, Cai S, Dong X (2011) Three feruloyl esterases in Cellulosilyticum ruminicola H1 act
synergistically to hydrolyze esterified polysaccharides. Appl Environ Microbiol 77
(17):6141–6147
Li X, Beeson WT, Phillips CM, Marletta MA, Cate JHD (2012) Structural basis for substrate
targeting and catalysis by fungal polysaccharide monooxygenases. Structure 20:1051–1061
Lynd LR, Weimer PJ, van Zyl WH, Pretorius IS (2002) Microbial cellulose utilization:
fundamentals and biotechnology. Microbiol Mol Biol Rev 66:506–577
Lynd LR, Laser MS, Bransby D, Dale BE, Davidson B, Hamilton R, Himmel ME, Keller M,
McMillan JD, Sheehan J (2008) How biotech can transform biofuels. Nat Biotechnol
26:169–172
Maki M, Leung KT, Qin W (2009) The prospects of cellulase-producing bacteria for the
bioconversion of lignocellulosic biomass. Int J Biol Sci 5(5):500–516
24 M.P. Raghavendra et al.

Mansfield MC, Touhy M, Saddler J (1998) The effect of the initial pore volume and lignin content
on the enzymatic hydrolysis of softwoods. Biores Technol 64:113–119
Martins LF, Kolling D, Camassola M, Dillon AJ, Ramos LP (2008) Comparison of Penicillium
echinulatum and Trichoderma reesei cellulases in relation to their activity against various
cellulosic substrates. Bioresour Technol 99:1417–1424
Mayer F, Coughlan MP, Mori Y, Ljungdahl LG (1987) Macromolecular organization of the
cellulolytic enzyme complex of Clostridium thermocellum as revealed by electron microscopy.
Appl Environ Microbiol 53:2785–2792
McMillan JD, Jenning EW, Mohagheghi A, Zuccarello M (2011) Comparative performance of
precommercial cellulases hydrolyzing pretreated corn stover. Biotech Biofuels 4:29–46
McQueen-Mason S, Cosgrove DJ (1994) Disruption of hydrogen bonding between plant cellwall
polymers by proteins that induce wall extension. Proc Natl Acad Sci USA 91:6574–6578
Mohanram S, Amat D, Choudhary J, Arora A, Nain L (2013) Novel perspectives for evolving
enzyme cocktails for lignocellulose hydrolysis in biorefineries. Sustain Chem Process 1:15–27
Morais S, Barak Y, Caspi J, Hadar Y, Lamed R, Shoham Y, Wilson DB, Bayer EA (2010)
Cellulase-xylanase synergy in designer cellulosomes for enhanced degradation of a complex
cellulosic substrate. mBio 1:00285–00210
Moser F, Irwin D, Chen SL, Wilson DB (2008) Regulation and characterization of Thermobifida
fusca carbohydrate-binding module proteins E7 and E8. Biotechnol Bioeng 100:1066–1077
Nair NU, Denard CA, Zhao H (2010) Engineering of enzymes for selective catalysis. Curr Org
Chem 14:1870–1882
Nakatani Y, Yamada K, Ogino C, Kondo A (2013) Synergetic effect of yeast cell-surface
expression of cellulase and expansin-like protein on direct ethanol production from cellulose.
Microb Cell Fact 12:66–73
Ohara H, Karita S, Kimura T, Sakka K, Ohmiya K (2000) Characterization of the cellulolytic
complex (cellulosome) from Ruminococcus albus. Biosci Biotechnol Biochem 64:254–260
Phillips CM, Beeson WT, Cate JH, Marletta MA (2011) Cellobiose dehydrogenase and a
copper-dependent polysaccharide monooxygenase potentiate cellulose degradation by
Neurospora crassa. ACS Chem Biol 6:1399–1406
Quinlan RJ, Sweeney MD, Lo Leggio L, Otten H, Poulsen JCN, Johansen KS, Krogh K,
Jorgensen CI, Tovborg M, Anthonsen A, Tryfona CP, Dupree WP, Xu F, Davies GJ,
Walton PH (2011) Insights into the oxidative degradation of cellulose by a copper
metalloenzyme that exploits biomass components. Proc Natl Acad Sci USA 108:15079–15084
Ranjan KK (2014) Adsorption, diffusion and activity of polycatalytic cellulase-nanoparticle
conjugates. Ph.D Dissertations. University of Connecticut
Resch MG, Donohoe BS, Baker JO, Decker SR, Bayer EA, Beckham GT, Himmel ME (2013)
Fungal cellulases and complexed cellulosomal enzymes exhibit synergistic mechanisms in
cellulose deconstruction. Energy Environ Sci 6:1858–1867
Rincon MT, Ding SY, McCrae SI, Martin JC, Aurilia V, Lamed R, Shoham Y, Bayer EA, Flint HJ
(2003) Novel organization and divergent dockerin specificities in the cellulosome system of
Ruminococcus flavefaciens. J Bacteriol 185:703–713
Rincon MT, Martin JC, Aurilia V, McCrae SI, Rucklidge GJ, Reid MD, Bayer EA, Lamed R,
Flint HJ (2004) ScaC, an adaptor protein carrying a novel cohesin that expands the
dockerin-binding repertoire of the Ruminococcus flavefaciens 17 cellulosome. J Bacteriol
186:2576–2585
Rincon MT, Dassa B, Flint HJ, Travis AJ, Jindou S, Borovok I, Lamed R, Bayer EA, Henrissat B,
Coutinho PM, Antonopoulos DA, Berg Miller ME, White BA (2010) Abundance and diversity
of dockerin-containing proteins in the fiberdegrading rumen bacterium, Ruminococcus
flavefaciens FD-1. PLoS ONE 5:e12476
Saloheimo M, Paloheimo M, Hakola S, Pere J, Swanson B, Nyyssönen E, Bhatia A, Ward M,
Penttilä M (2002) Swollenin, a Trichoderma reesei protein with sequence similarity to the plant
expansins, exhibits disruption activity on cellulosic materials. Eur J Biochem 269:4202–4211
Schnoes AM, Brown SD, Dodevski I, Babbitt PC (2009) Annotation error in public databases:
misannotation of molecular function in enzyme superfamilies. PLoS Comput Biol 5:e1000605
1 Microbial Enzymes for Conversion of Biomass to Bioenergy 25

Scott BR, Hill C, Tomashek J, Liu C (2009) Enzymatic hydrolysis of lignocellulosic feedstocks
using accessory enzymes. United States Patent Application 2009/0061484, 5 Mar 2009
Sedgwick SG, Smerdon SJ (1999) The ankyrin repeat: a diversity of interactions on a common
structural framework. Trends Biochem Sci 24:311–316
Shoseyov O, Shani Z, Levy I (2006) Carbohydrate binding modules: biochemical properties and
novel applications. Microbiol Mol Biol Rev 70:283–295
Sun Y, Cheng J (2002) Hydrolysis of lignocellulosic materials for ethanol production. Bioresour
Technol 83:1–11
Sweeney MD, Xu F (2012) Biomass converting enzymes as industrial biocatalysts for fuels and
chemicals: recent developments. Catalysts 2:244–263
Taherzadeh MJ, Karimi K (2007) Enzyme-based hydrolysis processes for ethanol from
lignocellulosic materials: a review. BioResources 2(4):707–738
Taylor LE, Henrissat B, Coutinho PM, Ekborg NA, Hutcheson SW, Weiner RM (2006) Complete
cellulase system in the marine bacterium Saccharophagus degradans strain 2-40T. J Bacteriol
188:3849–3861
Turnbaugh PJ, Quince C, Faith JJ, McHardy AC, Yatsunenko T, Niazi F, Affourtit J, Egholm M,
Henrissat B, Knight R, Gordon JI (2010) Organismal genetic and transcriptional variation in
the deeply sequenced gut microbiomes of identical twins. Proc Natl Acad Sci USA
107:7503–7508
Turner MB, Spear SK, Huddleston JG, Holbrey JD, Rogers RD (2003) Ionic liquid salt-induced
inactivation and unfolding of cellulose from Trichoderma reesei. Green Chem 5:443–447
Vaaje-Kolstad G, Horn SJ, van Aalten DMF, Synstad B, Eijsinkm VGH (2005) The noncatalytic
chitin-binding protein CBP21 from Serratia marcescens is essential for chitin degradation.
J Biol Chem 280:28492–28497
Valaskova V, Baldrian P (2006) Degradation of cellulose and hemicelluloses by the brown rot
fungus Piptoporus betulinus production of extracellular enzymes and characterization of the
major cellulases. Microbiol 152:3613–3619
Van RP, Van ZWH (1998) Pretorius IS: engineering yeast for efficient cellulose degradation. Yeast
14:67–76
Vazana Y, Barak Y, Unger T, Peleg Y, Shamshoum M, Ben-Yehezkel T, Mazor Y, Shapiro E,
Lamed R, Bayer EA (2013) A synthetic biology approach for evaluating the functional
contribution of designer cellulosome components to deconstruction of cellulosic substrates.
Biotechnol Biofuels 6:182–200
Wang Y, Tang R, Tao J, Gao G, Wang X, Mu Y, Feng Y (2011) Quantitative investigation of
non-hydrolytic disruptive activity on crystalline cellulose and application to recombinant
swollenin. Appl Microbiol Biotechnol 91:353–1363
Weber S, Stubner S, Conrad R (2001) Bacterial populations colonizing and degrading rice straw in
anoxic paddy soil. Appl Environ Microbiol 67:1318–1327
Wen F, Sun J, Zhao H (2010) Yeast surface display of trifunctional minicellulosomes for
simultaneous saccharification and fermentation of cellulose to ethanol. Appl Environ Microbiol
76:1251–1260
Westereng B, Ishida T, Vaaje-Kolstad G, Wu M, Eijsink VGH, Igarashi K, Samejima M,
Ståhlberg J, Horn SJ, Sandgren M (2011) The putative endoglucanase PcGH61D from
Phanerochaete chrysosporium is a metal dependent oxidative enzyme that cleaves cellulose.
PLoS ONE 6(11):e27807
Wetzel SK, Settanni G, Kenig M, Binz HK, Pluckthun A (2008) Folding and unfolding
mechanism of highly stable full-consensus ankyrin repeat proteins. J Mol Biol 376:241–257
Whitney SE, Gidley MJ, McQueen-Mason SJ (2000) Probing expansin action using
cellulose/hemicellulose composites. Plant J 22:327–334
Wilson DB (2009) Cellulases and biofuels. Curr Opin Biotechnol 20:295–299
Wongwilaiwalina S, Rattanachomsria U, Laothanachareona T, Eurwilaichitra L, Igarashib Y,
Champredaa V (2010) Analysis of a thermophilic lignocellulose degrading microbial
consortium and multi-species lignocellulolytic enzyme system. Enzyme Microb Tech
47:283–290
26 M.P. Raghavendra et al.

Wooley R, Ruth M, Glassner D, Sheejan J (1999) Process design and costing of bioethanol
technology: a tool for determining the status and direction of research and development.
Biotechnol Prog 15:794–803
Wyman CE (2007) What is (and is not) vital to advancing cellulosic ethanol. Trends Biotechnol
25:153–157
Xu Q, Gao W, Ding SY, Kenig R, Shoham Y, Bayer EA, Lamed R (2003) The cellulosome system
of Acetivibrio cellulolyticus includes a novel type of adaptor protein and a cell surface
anchoring protein. J Bacteriol 185:4548–4557
Xu Q, Bayer EA, Goldman M, Kenig R, Shoham Y, Lamed R (2004) Architecture of the
Bacteroides cellulosolvens cellulosome: description of a cell surface anchoring scaffoldin and a
family 48 cellulase. J Bacteriol 186:968–977
Yamada R, Hasunuma T, Kondo A (2013) Endowing non-cellulolytic microorganisms with
cellulolytic activity aiming for consolidated bioprocessing. Biotechnol Adv 31(6):754–763
Yang SJ, Kataeva I, Hamilton-Brehm SD, Engle NL, Tschaplinski TJ, Doeppke C, Davis M,
Westpheling J, Adams MW (2009) Efficient degradation of lignocellulosic plant biomass,
without pretreatment, by the thermophilic anaerobe Anaerocellum thermophilum DSM 6725.
Appl Environ Microbiol 75:4762–4769
Zambare VP, Zambare A, Muthukumarappan K, Christopher LP (2011) Potential of thermostable
cellulases in the bioprocessing of switchgrass to ethanol. BioResources 6:2004–2020
Zweifel ME, Barrick D (2001) Studies of the ankyrin repeats of the Drosophila melanogaster
Notch receptor. 2. Solution stability and cooperativity of unfolding. Biochemistry
40:14357–14367
Zweifel ME, Leahy DJ, Hughson FM, Barrick D (2003) Structure and stability of the ankyrin
domain of the Drosophila Notch receptor. Protein Sci 12:2622–2632
Part I
Cellulases
Chapter 2
Cellobiohydrolases: Role, Mechanism,
and Recent Developments

Neelamegam Annamalai, Mayavan Veeramuthu Rajeswari


and Nallusamy Sivakumar

Abstract Cellobiohydrolases or exoglucanases are produced by various bacteria


and fungi with catalytic modules belonging to families 5, 6, 7, 9, 48, and 74
glycoside hydrolases, which act at the chains end of cellulose resulting in release of
glucose as well as cellobiose. The CBH I and II works processively from reducing
and nonreducing ends of the cellulose chain, respectively. The catalytic module of
CBHs is the tunnel structure formed by two surface loops that may covers entirety
or part of active site evidenced that the mode of action proceeds in a processive
manner as cellobiohydrolase progresses along the cellulose chain. CBHs are able to
work actively in the crystalline region of cellulose, probably peeling them from the
microcrystalline structure of Avicel. Although several assays have been proposed,
no specific substrate as well as assay method to measure exoglucanases has been
described till date. In recent days, several new approaches such as δ-sequence
mediated integration, SCHEMA, and FoldX and a ‘consensus’ sequence have been
developed to improve activity and stability of CBHs.

2.1 Introduction

Considering the plant biomass, the main component from plant cell wall is the
cellulose, which is the more abundant carbohydrate constituted by glucopyranose
monomers linked by β-1,4 glycosidic bonds with two distinct regions such as

N. Annamalai (&)
Hawaii Natural Energy Institute, University of Hawaii at Manoa,
1680, East-West Road, Honolulu, HI 96822, USA
e-mail: [email protected]
M.V. Rajeswari
Centre for Ocean Research, Sathyabama University, Jeppiar Nagar,
Chennai, TN 600119, India
N. Annamalai  N. Sivakumar
Department of Biology, College of Science, Sultan Qaboos University,
PO Box 36, PC 123 Muscat, Oman

© Springer International Publishing Switzerland 2016 29


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_2
30 N. Annamalai et al.

crystalline and amorphous regions (Dashtban et al. 2009). Cellulosic biomass is the
largest amount of waste generated through human activities, which is the most
attractive substrate for ‘biorefinery strategies’ to produce high-value products such
as fuels, bioplastics and enzymes through fermentation processes (Mazzoli et al.
2012). The value of cellulose as a renewable source of energy has made cellulose
hydrolysis the subject of intense research and industrial interest (Bhat 2000). In
recent days, there is an increasing interest on use of biomass for biofuel production
is pointed-out as an important alternative for reduction of energetic and environ-
ment problems.
Cellulases are classified into three major categories based on their substrate
specificities and hydrolysis mechanism as (i) 1,4 β-D-glucanases or endoglucanases
(EC 3.2.1.4), (ii) exoglucanases or cellobiohydrolases (EC 3.2.1.91), and (iii) cel-
lobiase or β-glucosidases (EC 3.2.1.21). The most efficient hydrolysis of cellulose is
the result of combined synergistic actions of cellulases, whereby the enzymatic
activity of an enzyme mixture is significantly higher than individual enzyme
activity (Wood 1992; Irwin et al. 1993; Nidetzky et al. 1994).

2.2 Exoglucanases or Cellobiohydrolases (CBH)


(EC 3.2.1.91)

Exoglucanases, also known as cellobiohydrolases (CBHs) that act at the end of


cellulose chains and releasing glucose as well as cellobiose as product in a pro-
cessive fashion (Medve et al. 1998). CBHs are the most-studied exoglucanase,
which is about 70 % of the secreted cellulases by cellulolytic fungi belongs to
glycoside hydrolases (GH) 6 and 7, as well as 48 families (Teeri 1997). Different
CBHs are produced by various bacteria and fungi, with catalytic modules belonging
to families 5, 6, 7, 9, 48, and 74 glycoside hydrolases. Aerobic fungal CBHs are
belongs to families 6 and 7; aerobic bacterial CBHs belong to families 6 and 48;
anaerobic fungal CBHs are in family 48 and anaerobic bacterial CBHs are in family
9 as well as 48. In general, family 7 CBHs originate only from fungi, whereas
family 48 CBHs from bacteria (Zhang and Zhang 2013).

2.3 CBH I and II

CBHs mainly release cellobiose from cellulose derivatives, and the presence of
cellobiose competitively inhibits the rate of hydrolysis. The fungal CBHs belong to
glycosyl hydrolase families 6 and 7 (GH-6 and 7) and act most efficiently on highly
ordered crystalline cellulose, hydrolyzing from either reducing or non-reducing end
to liberate predominantly cellobiose (C2) with a minor amount of cellotriose (C3)
(Boer et al. 2000; Takahashi et al. 2010). There are two types of cellobiohydrolase:
2 Cellobiohydrolases: Role, Mechanism, and Recent Developments 31

CBH I and II that works processively from the reducing and non-reducing end of
the cellulose chain respectively. The cellobiohydrolases Cel6A (a CBH II) and
Cel7a (a CBH I) are much studied enzymes of fungal origin and are usually
expressed by the ascomycete Trichoderma reesei, accounts for 40 % of total pro-
tein and 70 % of cellulytic activity in the industrially relevant fungus Hypocrea
jecorina (T. reesei) which is used for industrial-scale production due to its ability to
produce more sugars from biomass through hydrolysis (Suominen et al. 1993).

2.4 Structure and Mode of Action of CBHs

The most significant topological feature of CBHs catalytic module is the tunnel
structure, formed by two surface loops that may cover the entirety (family 7 CBHs)
or part of the active site (family 48 CBHs) (Koivula et al. 2002; Vocadlo and
Davies 2008; Zhang and Zhang 2013). The CBH I (TrCBH I) is the major com-
ponent of the T. reesei cellulase system that belongs to the GH-7 family and
hydrolyzes the β-1,4-linkages of a cellulose chain from its reducing end (Claeyssens
et al. 1990). The catalytic core domain (CCD) structures of TrCBH I suggested that
a central structural motif constituted mainly by an antiparallel β-sandwich motif that
bears four surface loops, which decorate the concave face of the sandwich forming
the cellulose-binding tunnel (Divne et al. 1998; Stahlberg et al. 1996). During
enzymatic catalysis, the cellulose chain slides through the substrate-binding tunnel,
and every second glycosidic bond is correctly presented to the catalytic apparatus
located at the far end of the tunnel to progressively liberate disaccharide units
(cellobiose) from cellulose (Divne et al. 1994).
The three-dimensional (3D) structures of two GH-6 family members including
CBH of T. reesei and that of Humicola insolens in complex with glucose, cel-
looligosaccharide, and a non-hydrolyzable substrate analog (Rouvinen et al. 1990;
Varrot et al. 2002). It seems that the significant amino acids associated with cat-
alytic core domain, where the catalytic site is buried inside a tunnel-shaped cavity
and an enzyme–cello-oligosaccharide hydrogen bond network. This typical struc-
ture suggests that the mode of action proceeds in a processive manner as cel-
lobiohydrolase progresses along the cellulose chain (Boisset et al. 2000; Henrissat
1998; Reverbel-Leroy et al. 1997).
Cellobiohydrolases or exoglucanases is able to work effectively on microcrys-
talline cellulose, presumably peeling cellulose chains from the microcrystalline
structure (Teeri 1997). In general, CBHs are active on the crystalline regions of
cellulose; whereas, endoglucanases (1,4-β-glucanases) are typically active on the
more soluble amorphous region of the cellulose crystal (Rouvinen et al. 1990;
Divne et al. 1998). It seems that there is a high degree of synergy observed between
exo- and endoglucanases, which is required for efficient hydrolysis of cellulose
crystals (Abdel-Shakour and Roushdy 2009).
32 N. Annamalai et al.

2.5 Substrates and Assay Methods

In general, enzymes in cellulase mixture which show relatively high activity on


Avicel and little activity on carboxymethyl cellulose (CMC) are identified as
exoglucanases (CBHs) (Maki et al. 2009). The processive activity of CBHs on
CMC is blocked or inhibited by the occurrence of derivatized glucose residues
which leads to severe substrate limitation. Avicel (microcrystalline cellulose or
hydrocellulose) is mainly used for measuring exoglucanase activity because of its
low degree of polymerization. However, Avicel contains some amorphous cellulose
and soluble cellodextrans which can act as substrates for both exo- and endoglu-
canases. Thus, there is no highly specific substrate to measure exoglucanase activity
(Wood and Bhat 1988).
There are several different assays have been proposed to measure exoglucanase
(CBH I and CBH II) activity; however, all of these assays have some sort of
limitations. CBHs acting from nonreducing end of the substrate can be assayed with
chromogenic arylcellobiosides, from which the chromogenic aryl residue, attached
to the reducing end of the substrate liberated. However, the processive enzymes
acting from the reducing end of the carbohydrate chain have been identified
recently, but chromogenic substrates to identify these enzymes are not yet com-
mercially available (Boisset et al. 1998). Measurement of the production of cel-
lobiose directly via high-pressure liquid chromatography (Gum and Brown 1976) or
via a cellobiose oxidoreductase assay (Kelleher et al. 1987) is complicated in that
the endoglucanases that are present in crude cellulase mixtures also produce cel-
lobiose. The substrate 4-methylumbelliferyl-β-D-lactoside was identified as an
effective substrate for assaying CBH I of T. reesei as lactose, phenol and
4-methylumbelliferone (a fluorescent signal molecule) are the hydrolysis products
(van Tilbeurgh et al. 1985). But, the activity of CBH II was not able to detect thus
the resulting activity is not an effective representation of true exoglucanase activity.
Alternatively, an assay for quantification of exoglucanase activity using
p-nitrophenyl-β-D-cellobioside as substrate to yield cellobiose and p-nitrophenol
has been developed; but (i) CBH II activity cannot be measured using p-
nitrophenyl-β-D-cellobioside, (ii) the specific activity of the available purified
endoglucanases may not be representative for all existing endoglucanases in the
mixture, and (iii) the product ratio from endoglucanase actions may be influenced
by the presence of exoglucanases (Deshpande et al. 1984).
A double-antibody sandwich enzyme-linked immunosorbent assay was devel-
oped for quantifying CBH I in cellulase complex of T. reesei. The antibody con-
figuration resulted in the highest specificity for the assay of CBH I employed a
monoclonal antibody to coat wells in polystyrene plates and peroxidase-labeled
polyclonal antibody to detect CBH I bound to the immobilized monoclonal anti-
body. Since there was no direct assay of CBH I activity in the presence of the other
enzymes in complex till date, it would be useful for quantifying this enzyme
between the ranges of 0.1–0.8 µg/ml (Riske et al. 1990).
2 Cellobiohydrolases: Role, Mechanism, and Recent Developments 33

2.6 Recent Developments

Recently, the cellulolytic yeast (Saccharomyces cerevisiae) with enhanced CBH


activity was developed by integrating three types of CBH-encoding genes (cbh1
from Aspergillus aculeatus, cbh1, and cbh2 from T. reesei) with a strong consti-
tutive promoter Ptpi were sequentially integrated into the S. cerevisiae W303-1A
chromosome via δ-sequence mediated integration. The three recombinant (W1, W2,
and W3) and control strains (W303-1A and AADY) produced ethanol (g/l) from
acid-and alkali-pretreated corncob was about 5.92 ± 0.51, 18.60 ± 0.81,
28.20 ± 0.84, 1.40 ± 0.12, and 2.12 ± 0.35, respectively (Hong et al. 2014).
The thermostable CBHs could offer potential benefits in the hydrolysis of pre-
treated lignocellulosic substrates because the harsh conditions often required by
several pretreatments can be harmful for conventional biocatalysts. Heinzelman
et al. (2010) investigated an efficient SCHEMA recombination-based approach for
screening homologous enzymes to identify stabilizing amino acid sequence blocks
which has been used to generate active, thermostable CBH I enzymes from the
390,625 possible chimeras that can be made by swapping eight blocks from five
fungal homologs. A total of 16 predicted thermostable chimeras, with an average of
37 mutations relative are more thermostable (>65 °C) than the most stable parent
CBH I of thermophilic fungus Talaromyces emersonii.
Likewise, Komor et al. (2012) attempted to produce thermostable chimeric
fungal CBH I by structure-guided recombination through FoldX and a ‘consensus’
sequence approach to identify individual mutations present in the five homologous
parent CBH I enzymes which further stabilize the chimeras. With an alignment of
41 CBH I sequences, effects on ΔG (Folding) was calculated using amino acid
frequencies at each candidate position and the mutations chosen using these
methods increased the T(50) of the most thermostable chimera by an additional
4.7 °C, to yield a CBH I with T(50) of 72.1 °C, which is 9.2 °C higher than that of
native CBH I, from Talaromyces emersonii.

2.7 Perspectives

The interest in enzymatic hydrolysis of cellulosic biomass for biofuel production is


increasing continuously due to availability and reutilization as cheaper source.
CBHs are one among the important enzymes which could produce more sugars
such as glucose and cellobiose from crystalline cellulose. (i) It is essential to
develop easy and suitable method with specific substrate in order to measure the
CBHs activity in the cellulose mixture. (ii) Engineering of CBHs is also needed in
order to enhance the specific activity to reduce the use of CBHs and also to increase
thermostability and reusability of exoglucanases. These approaches mentioned
above to enhance the activity and stability of CBHs would be useful in biorefinery
and biofuel industries.
34 N. Annamalai et al.

References

Abdel-Shakour EH, Roushdy MM (2009) An investigation for cellulase activity of a novel


antibiotic producing Streptomyces sp. isolate H-1 from Egyptian mangrove sediment. Acad
Arena 1(5):89–98
Bhat MK (2000) Cellulases and related enzymes in biotechnology. Biotechnol Adv 18:355–383
Boer H, Teeri TT, Koivula A (2000) Characterization of Trichoderma reesei cellobiohydrolase
Cel7A secreted from Pichia pastoris using two different promoters. Biotechnol Bioeng
5:486–494
Boisset C, Armand S, Drouillard S, Chanzy H, Driguez H, Henrissat B (1998) Structure–function
relationships in cellulases: the enzymatic degradation of insoluble cellulose. In: Claeyssens M,
Nerinckx W, Piens KM (eds) Carbohydrases from Trichoderma reesei and other microorgan-
isms. Royal Society of Chemistry, London, pp 124–132
Boisset C, Fraschini C, Schulein M, Henrissat B, Chanzy H (2000) Imaging the enzymatic
digestion of bacterial cellulose ribbons reveals the endo character of the cellobiohydrolase
Cel6A from Humicola insolens and its mode of synergy with cellobiohydrolase Cel7A. Appl
Environ Microbiol 66:1444–1452
Claeyssens S, Lavoinne A, Freselragot M, Bois-Joyeux B, Chanez M, Peret J (1990) Metabolic
changes in rats fed a low protein-diet during post-weaning growth. Metabol Clin Exp
39:676–681
Dashtban M, Schraft H, Qin W (2009) Fungal bioconversion of lignocellulosic residues:
opportunities and perspectives. Int J Biol Sci 5:578–595
Deshpande MV, Eriksson KE, Pettersson LG (1984) An assay for selective determination of
exo-1,4-beta-glucanases in a mixture of cellulolytic enzymes. Anal Biochem 138:481–487
Divne C, Stahlberg J, Reinikainen T, Ruohonen L, Petterson G, Knowles JK, Teeri TT, Jones TA
(1994) The 3-dimensional crystal-structure of the catalytic core of cellobiohydrolase-I from
Trichoderma reesei. Science 265:524–528
Divne C, Stahlberg J, Teeri TT, Alwyn JT (1998) High-resolution crystal structures reveal how a
cellulose chain is bound in the 50 angstrom long tunnel of cellobiohydrolase I from
Trichoderma reesei. J Mol Biol 275:309–325
Gum EK Jr, Brown RD Jr (1976) Structural characteristics of a glycoprotein cellulase, 1,4-β-D-
glucan cellobiohydrolase from Trichoderma viride. Biochim Biophys Acta 446:371–376
Heinzelman P, Komor R, Kanaan A, Romero P, Yu X, Mohler S, Snow C, Arnold F (2010)
Efficient screening of fungal cellobiohydrolase class I enzymes for thermostabilizing sequence
blocks by SCHEMA structure-guided recombination. Protein Eng Des Sel 23(11):871–880
Henrissat B (1998) Enzymatic cellulose degradation. Cellul Commun 5:84–90
Hong J, Yang H, Zhang K, Liu C, Zou S, Zhang M (2014) Development of a cellulolytic
Saccharomyces cerevisiae strain with enhanced cellobiohydrolase activity. World J Microbiol
Biotechnol 30:2985–2993
Irwin DC, Spezio M, Walker LP, Wilson DB (1993) Activity studies of eight purified cellulases:
specificity, synergism and binding domain effects. Biotechnol Bioeng 42:1002–1013
Kelleher TJ, Montenecourt BS, Eveleigh DE (1987) Cellobiose-quinone oxidoreductase—
application in monitoring cellobiohydrolase purification. Appl Microbiol Biotechnol
27:299–305
Koivula A, Ruohonen L, Wohlfahrt G, Reinikainen T, Teeri TT, Piens K, Claeyssens M,
Weber M, Vasella A, Becker D, Sinnott ML, Zou J-Y, Kleywegt GJ, Szardenings M,
Stahlberg J, Jones TA (2002) The active site of cellobiohydrolase Cel6A from Trichoderma
reesei: the roles of aspartic acids D221 and D175. J Am Chem Soc 124:10015–10024
Komor RS, Romero PA, Xie CB, Arnold FH (2012) Highly thermostable fungal cellobioh
ydrolase I (Cel7A) engineered using predictive methods. Protein Eng Des Sel 25(12):827–833
Takahashi M, Takahashi H, Nakano Y, Konishi T, Terauchi R, Takeda T (2010) Characterization
of a cellobiohydrolase (MoCel6A) Produced by Magnaporthe oryzae. Appl Environ
Miccrobiol 76(19):6583–6590
2 Cellobiohydrolases: Role, Mechanism, and Recent Developments 35

Maki M, Leung KT, Qin W (2009) The prospects of cellulase-producing bacteria for the
bioconversion of lignocellulosic biomass. Int J Biol Sci 5(5):500–516
Mazzoli M, Lamberti C, Pessione E (2012) Engineering new metabolic capabilities in bacteria:
lessons from recombinant cellulolytic strategies. Trends Biotechnol 30(2):111–119
Medve J, Karlsson J, Lee D, Tjerneld F (1998) Hydrolysis of microcrystalline cellulose by
cellobiohydrolase I and endoglucanase II from Trichoderma reesei: adsorption, sugar
production pattern, and synergism of the enzymes. Biotechnol Bioeng 59(5):621–634
Nidetzky B, Steiner W, Hayn M, Claeyssens M (1994) Cellulose hydrolysis by the cellulases
from Trichoderma reesei: a new model for synergistic interaction. Biochem J 298:705–710
Reverbel-Leroy C, Page S, Belaich A, Belaich JP, Tardif C (1997) The processive endocellulase
CelF, a major component of the Clostridium cellulolyticum cellulosome: purification and
characterization of the recombinant form. J Bacteriol 179:46–52
Riske FJ, Eveleigh DE, Macmillan JD (1990) Double-antibody sandwich enzyme-linked
immunosorbent assay for cellobiohydrolase I. Appl Environ Microbiol 56(11):3261–3265
Rouvinen J, Bergfors T, Teeri TT, Knowles JKC, Jones TA (1990) Three-dimensional structure of
cellobiohydrolase II from Trichoderma reesei. Science 249:380–386
Stahlberg J, Divne C, Koivula A, Piens K, Claeyssens M, Teeri TT (1996) Activity studies and
crystal structures of catalytically deficient mutants of cellobiohydrolase I from Trichoderma
reesei. J Mol Biol 264:337–349
Suominen PL, Mantyla AL, Karhunen T, Hakola S, Nevalainen H (1993) High frequency one-step
gene replacement in Trichoderma reesei. II. Effects of deletions of individual cellulase genes.
Mol Gen Genet 241(5–6):523–530
Teeri TT (1997) Crystalline cellulose degradation: new insights into the function of cellobiohy-
drolases. Trends Biotechnol 15:160–167
van Tilbeurgh H, Pettersson G, Bhikabhai R, De Boeck H, Claeyssens M (1985) Studies of the
cellulolytic system of Trichoderma reesei QM 9414. Reaction specificity and thermodynamics
of interactions of small substrates and ligands with the 1,4-beta-glucan cellobiohydrolase II.
Eur J Biochem 148:329–334
Varrot A, Frandsen TP, Driguez H, Davies GJ (2002) Structure of the Humicola insolens cel-
lobiohydrolase Cel6A D416A mutant in complex with a non-hydrolysable substrate analogue,
methyl cellobiosyl-4-thio-β-cellobioside at 1.9 Å. Acta Crystallogr D Biol Crystallogr
58:2201–2204
Vocadlo DJ, Davies GJ (2008) Mechanistic insights into glycosidase chemistry. Curr Opin Chem
Biol 12:539–555
Wood TM, Bhat KM (1988) Methods for measuring cellulase activities. Methods Enzymol
160:87–117
Wood TM (1992) Fungal cellulases. Biochem Soc Trans 20:46–53
Zhang XZ, Zhang YHP (2013) Cellulases: characteristics, sources, production, and applications.
In: Yang S-T, El-Enshasy HA, Thongchul N (eds) Bioprocessing technologies in biorefinery
for sustainable production of fuels, chemicals, and polymers. Wiley, New York, pp 131–146
Chapter 3
Endo-1,4-β-glucanases: Role, Applications
and Recent Developments

Neelamegam Annamalai, Mayavan Veeramuthu Rajeswari


and Thangavel Balasubramanian

Abstract The biomass conversion processes are highly dependent on the use of
efficient enzymes to degrade polymeric cellulose or hemicellulose into simple sac-
charides, sugars which can be fermented by microorganisms for the production of
valuable fuel and chemicals. The cellulose hydrolysis involves enzymes such as
endo-1,4-β-glucanases (EC 3.2.1.4), cellobiohydrolases (or exo-1,4-β-glucanases)
(EC 3.2.1.91) and β-glucosidases (EC 3.2.1.21). Endo-β-(1,4)-glucanases (EGs) or
β-(1,4)-D-glucan-4-glucanohydrolases (EC 3.2.1.4), which act randomly on soluble
and insoluble β-(1,4)-glucan substrates. EGs breakdown cellulose by attacking the
amorphous regions to produce more accessible new free chain ends for the action of
cellobiohydrolases. The EG-I, II, III, and V, respectively, GH7, 5, 12, and 45 are
most common in natural fungal cellulase mixes. EGs play an important role in
increasing yield of fruit juices, beer filtration, and oil extraction, as well as improving
the nutritive quality of bakery products and animal feed. Reducing sugar assay is the
most convenient and reliable method for EG estimation. However, it is very con-
ventional and time consuming. Recently, a specific and sensitive assay methods have
been developed using substrate mixture comprises of benzylidene end-blocked
2-chloro-4-nitrophenyl-β-cellotrioside (BzCNPG3) and 4,6-O-benzylidene-
4-methylumbelliferyl-β-cellotrioside (BzMUG3).

N. Annamalai (&)
Hawaii Natural Energy Institute, University of Hawaii at Manoa,
1680, East-West Road, Honolulu 96822, HI, USA
e-mail: [email protected]
M.V. Rajeswari
Centre for Ocean Research, Sathyabama University, Jeppiar Nagar,
Chennai 600119, TN, India
T. Balasubramanian
CAS in Marine Biology, Faculty of Marine Sciences, Annamalai University,
Parangipettai 608502, TN, India

© Springer International Publishing Switzerland 2016 37


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_3
38 N. Annamalai et al.

3.1 Introduction

Lignocellulosic biomass contains morphologically different cellulose, complex


structural and compositional hemicellulose, recalcitrant lignin, diverse proteins and
lipids and other substances that interact with each other (Sweeney and Xu 2012).
Cellulose is a long homogenous linear polymer of β-D-glucosyl units linked by
1,4-β-D-glucosidic bonds. Over the last few years, the growing challenge has been
the setup of industrial-scale conversion of cellulosic biomass into fermentable
sugars, which can in turn be used as raw material for the production of biofuels and
other bio based high value-added products (Adsul et al. 2011; Du et al. 2011;
Somerville et al. 2010). Efficient conversion of cellulosic biomass into fuels and
other products is a fossil fuel-saving and environment-friendly biotechnology
(Lange 2007). The use of enzymes reduces the harsh chemicals and/or heat for the
production of fermentable sugars, raising the environmental feasibility of the
process.
The primary role of biomass-converting enzymes is to degrade polymeric cel-
lulose or hemicellulose into simple saccharides, sugars, which can be fermented by
microorganisms to, or serve as platform molecules for synthesis of valuable fuel or
chemicals. As potential industrial catalysts for biomass conversion, enzymes might
provide high specificity, low energy or chemical consumption, or low environment
pollution (Sweeney and Xu 2012). Cellulases and most hemicellulases belong to a
group of enzymes known as glycoside hydrolases (GH), which can degrade cellulose
and hemicellulose to constituent hexoses and pentoses. In general, biomass-
converting enzymes have to work in concert, to benefit from synergism among their
specificity (towards different components and regions of lignocellulose) as well as
mitigation of their inhibition (by different lignocellulose components or degradation
products) (Sweeney and Xu 2012). The classical scheme for cellulose hydrolysis
involves three main categories of enzymes: endo-1,4-β-glucanases (EC 3.2.1.4),
cellobiohydrolases (or exo-1,4-β-glucanases) (EC 3.2.1.91), and β-glucosidases (EC
3.2.1.21). The complete degradation of cellulose into glucose is a complex process
due to its amorphous/crystalline structure, which requires the cooperative action of
endo and exo-β-1,4-glucanases (EC 3.2.1.91), and β-glucosidases (EC 3.2.1.21).
According to CAZy database (http://www.cazy.org), these enzymes are grouped into
15 of 132 glycoside hydrolase (GH) families, i.e., 1, 3, 5–9, 12, 27, 44, 45, 48, 51,
61 and 74 (Wang et al. 2014).
In this respect, the most important enzyme has long been realized to be an
endo-β-1-3,1-4-glucanase (EC 3.2.1.73), which hydrolyzes β-1-4 linkages on the
reducing side of β-1-3 linkages to produce primarily of oligosaccharides with
contiguous β-1-4 linkages that derive from cellulosic regions in the polymer as
substrates for an endo-β-1-4-glucanase (Kanauchi and Bamforth 2008).
3 Endo-1,4-β-glucanases: Role, Applications and Recent … 39

3.2 Endo-1,4-β-glucanases (EC 3.2.1.4, Endocellulase)

Endo-β-(1,4)-glucanases or β-(1,4)-D-glucan-4-glucanohydrolases (EC 3.2.1.4) act


randomly on soluble and insoluble β-(1,4)-glucan substrates. The endo-1,4-β-
glucanase (EGase, EC 3.2.1.4) often referred to as cellulase, hydrolyzes 1,4-β-D-
glucan chain molecules and plays a role in various physiological processes (Yu et al.
2013). Endoglucanases (EG) are also referred to as carboxymethylcellulases
(CMCase), named after the artificial substrate used to measure the enzyme activity.
The active sites of endoglucanases typically attain a cleft-like topology for random
fashion and bond cleavage can occur anywhere along the chain of the cellulose
(Himmel et al. 2010). Endoglucanase breakdown the cellulose by attacking the
amorphous regions of the cellulose to produce more accessible new free chain ends
for the action of cellobiohydrolases which has been shown by the effect of the
enzyme on carboxymethylcellulose and amorphous cellulose (Sandgren et al. 2005).
Degradation of amorphous cellulose can be carried out by endoglucanases
(EGs) (EC 3.2.1.4). Unlike cellobiohydrolases (CBH), EG hydrolyzes internal
glycosidic bonds in cellulose with a random, on-off fashion which make them
well-suited to less orderly or partially shielded cellulose parts, generating new
cellulose chain ends for CBH action. A few EGs can act “processively” on crys-
talline cellulose (Wilson 2008; Li et al. 2010). Also known as EG-I, II, III and V,
respectively, GH7, 5, 12 and 45 EG are most common in natural fungal cellulase
mixes. EGs genes are found in many prokaryotic and eukaryotic organisms,
including bacteria, fungi, slime molds, nematodes, animals and plants (Levy et al.
2002; Libertini et al. 2004). Most cellulolytic fungi and bacteria produce numerous
EGs. Although they all act on the same cellulose substrate, they do with different
mechanisms (“inverting” for GH6, 9, 45 and 48 EGs; “retaining” for GH5, 7, 12
EGs), which may relate to different EGs’ side-activities on hemicellulose in
degrading complex lignocellulose (Vlasenko et al. 2010), or synergism between
processive and conventional EGs (Wilson 2008). The active sites of most EGs are
cleft- or groove-shaped, inside which a cellodextrin or a cellulose segment may be
bound and acted on by EG. In addition to the catalytic core, EGs may possess
CBMs which is the direct host, but is not a pre-requisite, for EG’s action (Sweeney
and Xu 2012).
The fungal endoglucanases (EGs) are generally monomers with no or low
glycosylation and have an open binding cleft. They mostly have pH optima
between 4 and 5 and temperature optima from 50 to 70 °C. A typical cellulolytic
fungus secretes EGs at *20 % wt level in their secretomes (Sipos et al. 2010;
Chundawat et al. 2011). In addition, many of the fungi produce multiple EGs. For
example, T. reesei produces at least 5 EGs (EG I/Cel 7B, EG II/Cel5A, EG
III/Cel12A, EG IV/Cel61A and EG V/Cel45A) whereas three EGs were isolated
from white-rot fungus P. chrysosporium (EG28, EG34 and EG44) (Baldrian and
Valaskova 2008; Foreman et al. 2003). In addition, some EGs lack a CBM while
some other EGs with CBM have been described. For example, four of five EGs in
40 N. Annamalai et al.

T. reesei including EG I, EG II, EG IV and EG V have CBM whereas EG III does


not have a CBM (Sandgren et al. 2005).
In plants, EGases are encoded by a large gene family, belonging to the glycosyl
hydrolase gene family 9 (GH9) (Henrissat 1991). According to their sequence
domain structure, this multigene family can be grouped into three subclasses: A
subclass (GH9A), membrane-anchored GH9; B subclass (GH9B), secreted GH9;
and C subclass (GH9C), secreted proteins with a carbohydrate binding module,
CBM49 (Urbanowicz et al. 2007). Alternatively, the plant EGase gene family can
be divided into α, β and γ subfamilies based on phylogenetic analysis (Libertini
et al. 2004). In Arabidopsis, the α-subfamily mainly consists of secreted GH9
proteins (including GH9B and GH9C subclasses) except for one GH9A subclass
protein. All γ-subfamily members are transmembrane proteins, belonging to GH9A
subclass (Yu et al. 2013).

3.3 Criteria for Optimized EG Production

In general, cellulase production accounts for 40 % of cost in bioethanol synthesis


(Sateesh et al. 2012). Carbon and nitrogen sources are the important factors for
endoglucanase production and synthesis of various cell wall lipids in fungi
(Sarria-Alfonso et al. 2013). The effects of different carbon sources (avicel, cel-
lobiose, CMC, xylan, rice straw, bagasse and palm kernel) on EG production by
various microorganisms were investigated till date. Among these, rice straw was
reported to be a cheaper and potential sole carbon source and yeast extract as the
nitrogen source for higher EG production. In addition, a significant increase in
enzyme production with an increased fermentation time, which was presumed to be
due to rapid hydrolysis of cellulose in the medium.

3.4 Industrial Applications of Endo-β-1,4-Glucanase

Cellulases are used in the textile industry, in detergents, pulp and paper industry,
improving digestibility of animal feeds and food industry. They account for a
significant fraction of industrial enzyme markets. Obviously, cellulase will be the
largest industrial enzyme as compared to the current market size for all industrial
enzymes ($ *2 billion) (Zhang and Zhang 2013). Among the cellulases,
endoglucanases play a key role in increasing the yield of fruit juices, beer filtration
and oil extraction, as well as improving the nutritive quality of bakery products and
animal feed (Bhat 2000). Endo-β-1,4-glucanase has been widely used in industries
to hydrolyze cellulosic materials to sugars in the biofuel production (Wilson 2009),
to increase the rate of wort filtration and reduce the possibility of β-glucan pre-
cipitation in beer and reduce the mash viscosity and turbidity in the brewing
industry (Celestino et al. 2006), to bioremediate pulp waste in the paper industry
3 Endo-1,4-β-glucanases: Role, Applications and Recent … 41

(Ohmiya et al. 1997), and to increase β-glucan digestibility and improve feed
conversion efficiency in the feed industry (Mathlouthi et al. 2002). The
endo-1,4-β-glucanase is also used in the poultry and animal feed industry to
degrade β-glucan and thus increase the digestibility of feeds rich in barley and also
overcome the anti-nutritive effects of 1,3-1,4-β-D-glucan (Choct 2001). Further,
endo-1,4-β-glucanase is used for denim finishing and cotton softening in textile
industries, cleaning and colour care in the detergent industries and to modify fibre
and improves drainage in the paper industries (Cherry and Fidantsef 2003).
Thus, the enormous industrial applications demands endoglucanases with
varying pH and temperature optima, stabilities and substrate specificities. Although
cellulolytic enzymes of Trichoderma reesei have been investigated thoroughly
(Miettinen-Oinonen et al. 2004), the amount of cellulase secreted by this fungus is
insufficient for effective conversion of cellulose to glucose. Moreover, the industrial
demand is increasing day by day especially because of the emergence of second
generation-advanced biofuel industries, which require tremendous amounts of
various enzymes in their processes (Wilson 2009; Yeoman et al. 2010).
Thermostable endoglucanases/cellulases are more favourable for industrial
application due to their high activity and stability at high temperatures to decrease
process costs and increase the efficiencies (Yeoman et al. 2010). However, most
cellulases are not stable at high temperatures, and a number of thermostable cel-
lulases have been purified or cloned and characterized in recent years, some of them
display maximum activities even at 100 °C, such as GH 12 endoglucanase from
Pyrococcus furiosus (100 °C; Bauer et al. 1999), and endoglucanase CelB from
Thermotoga neapolitana (106 °C; Bok et al. 1998). As the main microbial source
of thermophilic endoglucanases, thermophilic fungi like Talaromyces emersonii
and Thermoascus aurantiacus have been reported to produce cellulases of GH 3, 5
and 7 families with temperature optima of 65–80 °C (Gomes et al. 2000; Grassick
et al. 2004).

3.5 Recent Developments on Endo-1,4-β-Glucanase Assay

The endo-1,4-β-glucanase in biological materials and microbial fermentation broths


can be specifically measured by the decrease in the viscosity of water soluble,
chemically modified cellulose derivatives such as CM-cellulose 7 M (McCleary et al.
2012). The reducing sugar assays method is most conventional and time consuming
(Somogyi 1952). The other methods such as measuring hydrolysis rate of substrates
such as cello-oligosaccharides, borohydride-reduced cello-oligosaccharides (McCleary
et al. 2012) or nitrophenyl-cello-oligosaccharides (Bhat et al. 1990; Claeyssens and
Henrissat 1992; Rahman et al. 2002) cannot be used to measure endo-1,4-β-glucanase
in the presence β-glucosidase and exo-1,4-β-glucanases. Hence, a specific and sen-
sitive substrate mixture comprises benzylidene end-blocked 2-chloro-4-nitrophenyl-β-
cellotrioside (BzCNPG3) was prepared as substrate for the assay of endo-1,4-β-
glucanase (cellulase). Hydrolysis by β-glucosidase and exo-1,4-β-glucanase is
42 N. Annamalai et al.

prevented by the presence of the benzylidene group on the non-reducing end D-


glucosyl residue (McCleary et al. 2014).
In addition to this, novel fluorometric assay for endo-1,4-β-glucanase that is
based on the use of 4,6-O-benzylidene-4-methylumbelliferyl-β-cellotrioside
(BzMUG3) in the presence of an ancillary β-glucosidase was developed for both
quantitative and qualitative measurements. The substrate BzMUG3 can be cleaved
in the presence of cellulase to generate a protected cello-oligosaccharide fragment
and a fluorogenic cello-oligosaccharide which will then be acted on by excess
β-glucosidase (EC 3.2.1.21) present in the reagent mixture to liberate
4-methylumbelliferone and 4,6-O-benzylidene protecting group prevents the action
of β-glucosidase on the parent substrate. The rate of formation of
4-methylumbelliferone is directly proportional to the cellulase activity in a given
sample (Mangan et al. 2014).

3.6 Future Prospective

Biomass saccharification is the largest technical and economic obstacle to biorefinery


and biofuels production. For the efficient conversion of cellulose from biomass and to
glucose for the subsequent production of fuel ethanol, protein engineering of
endo-1,4-β-glucanase should be focused to enhance specific activity towards pre-
treated biomass using enzyme cocktail and either rational design or directed evolu-
tion (Sathitsuksanoh et al. 2010; Zhang 2008), improve the stability for recycling and
reduce production costs ($ per kilogram of dry protein) (Zhu et al. 2009). In addition,
engineering of cellulolytic enzymes with improved catalytic efficiency and enhanced
thermostability is important to commercialize lignocelluloses biorefinery and further
improvement on cellulase performance needs the better understanding of hydrolysis
mechanisms as well as the relationship of cellulase molecular structure, function and
substrate characteristics. The consolidated bioprocessing (CBP) of microorganisms
or consortium would make the biomass hydrolysis easier and enhance the produc-
tivity. The improved simultaneous saccharification and fermentation (SSF) technol-
ogy would be useful to reduce end-product inhibition, investment costs and higher
yield efficient biofuel production.

References

Adsul MG, Singhvi MS, Gaikaiwari SA, Gokhale DV (2011) Development of biocatalysts for
production of commodity chemicals from lignocellulosic biomass. Bioresour Technol
102:4304–4312
Baldrian P, Valaskova V (2008) Degradation of cellulose by basidiomycetous fungi. FEMS
Microbiol Rev 32:501–521
Bauer MW, Driskill LE, Callen W, Snead MA, Mathur EJ, Kelly RM (1999) An endoglucanase,
EglA, from the hyperthermophilic archaeon Pyrococcus furiosus hydrolyzes β-1, 4 bonds in
mixed-linkage (1–3), (1–4)-β-d-glucans and cellulose. J Bacteriol 181:284–290
3 Endo-1,4-β-glucanases: Role, Applications and Recent … 43

Bhat KM, Hay AJ, Claeyssens M, Wood TM (1990) Study of the mode of action and site-specificity
of the endo-(1-4)-beta-D-glucanases of the fungus Penicillium pinophilum with normal,
1-3H-labelled, reduced and chromogenic cello-oligosaccharides. Biochem J 266:371–378
Bhat MK (2000) Cellulases and related enzymes in biotechnology. Biotechnol Adv 18:355–383
Bok JD, Yernool DA, Eveleigh DE (1998) Purification, characterization, and molecular analysis of
thermostable cellulases CelA and CelB from Thermotoga neapolitana. Appl Environ
Microbiol 64:4774–4781
Celestino KRS, Cunha RB, Felix CR (2006) Characterization of a β-glucanase produced by
Rhizopus microsporus var. microsporus, and its potential for application in the brewing
industry. BMC Biochem 7:23
Cherry JR, Fidantsef AL (2003) Directed evolution of industrial enzymes: an update. Curr Opin
Biotechnol 14:438–443
Choct M (2001) Enzyme supplementation of poultry diets based on viscous cereals. In:
Bedford M, Partridge G (eds) Enzymes in farm animal nutrition. CABI Publishing, Oxon,
pp 145–160
Chundawat SPS, Lipton MS, Purvine SO, Uppugundla N, Gao D, Balan V, Dale BE (2011)
Proteomics-based compositional analysis of complex cellulase-hemicellulase mixtures.
J Proteome Res 10:4365–4372
Claeyssens M, Henrissat B (1992) Specificity mapping of cellulolytic enzymes: classification into
families of structurally related proteins confirmed by biochemical analysis. Protein Sci 1:
1293–1297
Du J, Shao Z, Zhao H (2011) Engineering microbial factories for synthesis of value-added
products. J Ind Microbiol Biotechnol 38:873–890
Foreman PK, Brown D, Dankmeyer L, Dean R, Diener S, Dunn-Coleman NS, Goedegebuur F,
Houfek TD, England GJ, Kelley AS, Meerman HJ, Mitchell T, Mitchinson C, Olivares HA,
Teunissen PJ, Yao J, Ward M (2003) Transcriptional regulation of biomass-degrading enzymes
in the filamentous fungus Trichoderma reesei. J Biol Chem 278:31988–31997
Gomes I, Gomes J, Gomes DJ, Steiner W (2000) Simultaneous production of high activities of
thermostable endoglucanase and β-glucosidase by the wild thermophilic fungus Thermoascus
aurantiacus. Appl Microbiol Biotechnol 53:461–468
Grassick A, Murray PG, Thompson R, Collins CM, Byrnes L, Birrane G, Higgins TM, Tuohy MG
(2004) Three-dimensional structure of a thermostable native cellobiohydrolase, CBH IB, and
molecular characterization of the cel7 gene from the filamentous fungus, Talaromyces
emersonii. Eur J Biochem 271:4495–4506
Henrissat B (1991) A classification of glycosyl hydrolases based on amino acid sequence
similarities. Biochem J 280:309–316
Himmel ME, Xu Q, Luo Y, Ding SY, Lamed R, Bayer EA (2010) Microbial enzyme systems for
biomass conversion: emerging paradigms. Biofuels 1(2):323–341
Kanauchi M, Bamforth CW (2008) The relevance of different enzymes for the hydrolysis of
β-glucans in malting and mashing. J Inst Brew 114(3):224–229
Lange JP (2007) Lignocellulose conversion: an introduction to chemistry, process and economics.
Biofuel Bioprod Biorefin 1:39–48
Levy I, Shani Z, Shoseyov O (2002) Modification of polysaccharides and plant cell wall by
endo-1,4-beta-glucanase and cellulose-binding domains. Biomol Eng 19:17–30
Li Y, Irwin DC, Wilson DB (2010) Increased crystalline cellulose activity via combinations of
amino acid changes in the family 9 catalytic domain and family 3c cellulose binding module of
Thermobifida fusca Cel9A. Appl Environ Microbiol 76:2582–2588
Libertini E, Li Y, McQueen-Mason SJ (2004) Phylogenetic analysis of the plant
endo-beta-1,4-glucanase gene family. J Mol Evol 58:506–515
Mangan D, McCleary BV, Liadova A, Ivory R, McCormack N (2014) Quantitative fluorometric
assay for the measurement of endo-1,4- β -glucanase. Carbohydr Res 395:47–51
Mathlouthi N, Mallet S, Saulinier L, Quemener B, Larbier M (2002) Effects of xylanase and
beta-glucanase addition on performance, nutrient digestibility and physic-chemical conditions
44 N. Annamalai et al.

in the small intestine contents and caecal microflora of broiler chickens fed a wheat and
barley-based diet. Anim Res 51:395–406
McCleary BV, McKie V, Draga A (2012) Measurement of endo-1,4-β-glucanase. Methods
Enzymol 510:1–17
McCleary BV, Mangan D, Daly R, Fort S, Ivory R, McCormack N (2014) Novel substrates for the
measurement of endo-1,4-β-glucanase (endo-cellulase). Carbohydr Res 385:9–17
Miettinen-Oinonen A, Londesborough J, Joutsjoki V, Lantto R, Vehmaanpera J (2004) Three
cellulases from Melanocarpus albomyces for textile treatment at neutral pH. Enzyme Microb
Technol 34:332–341
Ohmiya K, Sakka K, Karita S, Kimura T (1997) Structure of cellulases and their applications.
Biotechnol Genet Eng Rev 14:365–414
Rahman M, Bhuiyan SH, Nirasawa S, Kitaoka M, Hayashi KJ (2002) Characterization of an
endo-β-1,4-glucanase of Thermotoga maritima expressed in Escherichia coli. Appl Glycosci
49:487–495
Sandgren M, Stahlberg J, Mitchinson C (2005) Structural and biochemical studies of GH family 12
cellulases: improved thermal stability and ligand complexes. Prog Biophys Mol Biol 89:
246–291
Sarria-Alfonso V, Sierra JS, Morales MA, Rojas IG, Sarmiento NM, Pinales P (2013) Culture
media statistical optimization for bio-mass production of a ligninolytic fungus for future rice
straw degradation. Indian J Microbiol 53(2):199–207
Sateesh L, Rodhe A, Naseeruddin S, Yadav K, Prasad Y, Rao L (2012) Simultaneous cellulase
production, saccharification and detoxification using dilute acid hydrolysate of S. spontaneum
with Trichoderma reesei NCIM 992 and Aspergillus niger. Indian J Microbiol 52:258–262
Sathitsuksanoh N, Zhu Z, Ho T-J, Bai M-D, Zhang Y-HP (2010) Bamboo saccharification through
cellulose solvent based biomass pretreatment followed by enzymatic hydrolysis at ultra-low
cellulase loadings. Bioresour Technol 101:4926–4929
Sipos B, Benko Z, Dienes D, Reczey K, Viikari L, Siika-aho M (2010) Characterisation of specific
activities and hydrolytic properties of cell-wall-degrading enzymes produced by Trichoderma
reesei Rut C30 on different carbon sources. Appl Biochem Biotechnol 161:347–364
Somerville C, Youngs H, Taylor C, Davis SC, Long SP (2010) Feedstocks for lignocellulosic
biofuels. Science 13:790–792
Somogyi MJ (1952) Notes on sugar determination. Biol Chem 195:19–23
Sweeney MD, Xu F (2012) Biomass converting enzymes as industrial biocatalysts for fuels and
chemicals: recent developments. Catalysts 2:244–263
Urbanowicz BR, Bennett AB, Del Campillo E, Catala C, Hayashi T, Henrissat B, Hofte H,
McQueen-Mason SJ, Patterson SE, Shoseyov O, Teeri TT, Rose JKC (2007) Structural
organization and a standardized nomenclature for plant endo-1,4-beta-glucanases (cellulases)
of glycosyl hydrolase family 9. Plant Physiol 144:1693–1696
Vlasenko E, Schulein M, Cherry J, Xu F (2010) Substrate specificity of family 5, 6, 7, 9, 12, and
45 endoglucanases. Bioresour Technol 101:2405–2411
Wang K, Luo H, Bai Y, Shi P, Huang H, Xue X, Yao B (2014) A thermophilic
endo-1,4-β-glucanase from Talaromyces emersonii CBS394.64 with broad substrate specificity
and great application potentials. Appl Microbiol Biotechnol 98:7051–7060
Wilson DB (2008) Three microbial strategies for plant cell wall degradation. Ann N Y Acad Sci
1125:289–297
Wilson DB (2009) Cellulases and biofuels. Curr Opin Biotechnol 20(3):295–299
Yeoman CJ, Han Y, Dodd D, Schroeder CM, Mackie RI, Cann IK (2010) Thermostable enzymes
as biocatalysts in the biofuel industry. Adv Appl Microbiol 70:1–55
Yu L, Sun J, Li L (2013) PtrCel9A6, an endo-1,4-β-glucanase, is required for cell wall formation
during xylem differentiation in populous. Mol Plant 6(6):1904–1917
Zhang Y-HP (2008) Reviving the carbohydrate economy via multi-product biorefineries. J Ind
Microbiol Biotechnol 35:367–375
3 Endo-1,4-β-glucanases: Role, Applications and Recent … 45

Zhang XZ, Zhang Y-HP (2013) Cellulases: characteristics, sources, production, and applications.
In: Yang ST, El-Enshasy HA, Thongchul N (eds) Bioprocessing technologies in biorefinery for
sustainable production of fuels, chemicals and polymers. Wiley, New York, pp 131–146
Zhu Z, Sathitsuksanoh N, Zhang P (2009) Direct quantitative determination of adsorbed cellulase
on lignocellulosic biomass with its application to study cellulase desorption for potential
recycling. Analyst 134:2267–2272
Chapter 4
The Role and Applications
of b-Glucosidases in Biomass Degradation
and Bioconversion

Hanlin Ouyang and Feng Xu

Abstract b-Glucosidases is known as the terminal enzyme in the synergy with


other cellulases for biomass degradation. It completes the final step of small
oligosaccharides (including cellobiose) conversion into glucose. However, this is
only a small part of the roles played by a subgroup of bacterial/fungal
b-glucosidases in cellulose biodegradation. To deepen our understanding about
the current challenges and limits in biomass conversion and to enlighten the future
for the industry, we take one step back and look into a broader range of
b-glucosidases that cross the life domains. b-Glucosidases from different subfam-
ilies are compared systematically in their distribution, phylogenetic relationship,
structure, in vivo function, mechanism of hydrolysis as well as substrate recogni-
tion, and kinetic profile. Protein engineering and application works on the enzymes
are discussed based on the knowledge herein.

4.1 Introduction

Biomass industry nowadays is developing and optimizing enzymatic solutions to


hydrolyze lignocellulosic plant-based substrates into monomeric sugars, which are
eventually fermented to bioethanol. Biomass substrate composition can be simplified
as cellulose, hemicellulose, and lignin in general while each component is targeted by
different enzymes. Glucan, or cellulose, consists of up to more than 40 % of biomass
substrates, and therefore cellulases are major players in the enzymatic hydrolysis of
glucan. Cellulases refer to a synergistic collection of three different groups of
enzyme: cellobiohydrolase (CBH, exo-b-1,4-glucanase), endo-b-1,4-glucanase
(EG), and b-glucosidase (BGL). While the exo- and endoglucanases cleave poly-

H. Ouyang (&)
Novozymes North America Inc., 77 Perrys Chapel Church Road, PO BOX 576, Franklinton,
NC 27525, USA
e-mail: [email protected]
F. Xu
Novozymes Inc., 1445 Drew Ave, Davis, CA 95618, USA
e-mail: [email protected]

© Springer International Publishing Switzerland 2016 47


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_4
48 H. Ouyang and F. Xu

meric cellulose into small oligosaccharides (cellodextrins), BGL is responsible to


release glucose (Glc) from these oligosaccharides, especially cellobiose. In addition,
different BGLs can have activity on many other glycosides, releasing the nonre-
ducing terminal glycosyl moiety from these substrates, which may be involved in
various physiological events (Ketudat Cairns and Esen 2010). The ‘Second
Generation’ (cellulosic) bioethanol industry locks their preference on BGLs from
microbes that feed on biomass (Rani et al. 2014; Sørensen et al. 2013; Teugjas and
Väljamäe 2013; Tiwari et al. 2013; Singhania et al. 2013; Sweeney and Xu 2012;
Krisch et al. 2010; Quinlan et al. 2010; Xu 2004, 2010a, b; Eyzaguirre et al. 2005;
Bhatia et al. 2002). The development of viable industrial BGLs has been challenging.
Native BGLs produced in many fungal strains occur in low abundance, which needs
to be greatly enhanced for the enzymes to become economically viable for industrial
application. The BGL’s product inhibition by Glc can be significant during the
industrial enzymatic cellulose hydrolysis. Besides, biomass-based ethanol producers
rely on thermochemical pretreatment of lignocellulosic substrates to improve enzyme
access to cellulose and cellulose hydrolysis often occurs at elevated temperature to
speed up the process. These two steps create harsh conditions for BGL’s ther-
mostability and inhibition resistance while the challenges have propelled active
studies on BGL.

4.2 Classification and Distribution

Historically, BGLs have been categorized into three groups based on the substrates:
aryl-BGLs, true cellobiases, and broad substrate specificity BGLs. Canonical BGLs
that catalyze the hydrolysis of b-glucopyranosides are assigned with the Enzyme
Commission (E.C.) number E.C.3.2.1.21. However, given the variety of b-linkage
terminal nonreducing glycosides in nature, many E.C. numbers have been assigned
to a larger group of enzymes that hydrolyze b-linkage glycosidic bond. For glucan
substrates, glucan 1,3-b-glucosidase (3.2.1.58) and glucan 1,4-b-glucosidase
(3.2.1.74) remove successively Glc from the nonreducing end with 1,3 and 1,4
b-linkage to the polysaccharide chain. Enzymes with hydrolytic activity on
b-glucosides with aglycon moieties also fall into E.C.3.2.1 family. These include
glucosylceramidase (3.2.1.45), steryl-b-glucosidase (3.2.1.104), 3a(S)-strictosidine
b-glucosidase (3.2.1.105), amygdalin-b-glucosidase (3.2.1.117), prunasin
b-glucosidase (3.2.1.118), vicianin b-glucosidase (3.2.1.119), raucaffricine
b-glucosidase (3.2.1.125), coniferin b-glucosidase (3.2.1.126), and b-D-glucopyr-
anosyl abscisate b-glucosidase (3.2.1.175). More complicated substrates like
thio-linkage, 7-[b-D-apiofuranosyl-(1, 6)-b-D-glucopyranosyloxy]isoflavonoid, b-D-
galactopyranose-(1, 4)-a-L-galactopyranose-6-sulfate in porphyran, or even 3-O-(N-
acetyl-b-D-glucosaminyl)-L-serine/threonine in proteins can be hydrolyzed by
thioglucosidase (myrosinases) (3.2.1.147), b-apiosyl-b-glucosidase (3.2.1.161),
b-porphyranase (3.2.1.178) and protein O-GlcNAcase (3.2.1.169) respectively.
E.C. numbering system is a classification based on the chemical reactions which
enzymes catalyze. It has little information on structure, mechanism, or phylogenetic
4 The Role and Applications of b-Glucosidases in Biomass … 49

relationship. A sequence-based classification has been developed since 1996. The


glycoside hydrolases (GH) are currently classified into 133 families in the
Carbohydrate Active enZyme, or CAZy database (www.cazy.org). Each GH family
contains proteins that are related by sequence, corollary, folding (based on the
hydrophobic cluster similarity). This allows the possibility of useful mechanis-
tic predictions as the motifs, key residues and geometry around the active sites
usually share high similarity for a conserved enzymatic mechanism, even when the
substrates (thus the E.C numbers) are different. Almost all the BGLs identified so
far belong to the GH1, GH3, GH5, GH9, GH30, and GH116 families. Based on the
CAZy as of June 2015, the GH1 family includes six BGLs from archaea, 194 from
bacteria, 210 from eukaryotes (especially fungi and plants), one from a virus and six
from unclassified organisms, along with 42 myrosinases and one glucosylcerami-
dase from eukaryotes. The GH3 family includes one BGL from an archaeon, 118
from bacteria, 141 from eukaryotes (especially fungi and plants), ten from
unclassified organisms, and several likely from animal (Gabrisko and Janecek
2015). The GH5 family includes one BGL from a virus, one from plant and four in
fungi, while the GH30 family includes one BGL from a fungus, two from animals
and one from human. The GH30 family also has two animal glucosylceramidases.
The GH9 family so far only covers two BGLs from the bacterium Vibrio cholera.
Most of these BGLs are intracellular or cytoplasmic enzymes. However, some of
them are membrane associated, such as bacterial Pyrococcus horikoshii bgl (GH1)
(Matsui et al. 2000) and Prevotella ruminicola cdxA (GH3) (Wulff-Strobel and
Wilson 1995). Some of them can reside in periplasmic space, such as bacterial
Azospirillum irakense salA, salB (GH3) (Faure et al. 1999), Bacillus subtilis bglA
(GH1) (Srivastava et al. 1999), Erwinia chrysanthemi bglx (GH3) (Vroemen et al.
1995), Erwinia herbicola bglA (GH1) (Marri et al. 1995) and fungal (mold)
Aspergillus kawachii bglA (GH3) (Iwashita et al. 1999). Some can be extracellular,
such as bacterial Ruminococcus albus bgl (GH3) (Takano et al. 1992), fungal
Aspergillus niger bgl1(GH1) (Dan et al. 2000), Trichoderma reesei (Hypocrea
jecorina) bgl1 (GH3) bgl2 (GH1) (Barnett et al. 1991; Takashima et al. 1999),
Saccharomycopsis fibuligera bgl1, bgl2 (both GH3) (Machida et al. 1988) and
barley (Hordeum vulgare) bgq60 (GH1) (Leah et al. 1995), Catharanthus roseus
strictosidine bgl (GH1) (Geerlings et al. 2000). The extracellular microbial BGLs
are of particular industrial interest because of their production costs (that can be
significantly lower than that of the intracellular counterparts).

4.3 In Vivo Function

4.3.1 Bacteria and Fungi

One of the functions for BGL is being a part of the synergistic cellulase system that
breaks down cellulose, deployed by a large population of organisms including
bacteria, fungi (molds, mushrooms, yeasts), and animals (e.g., termites). BGL is
50 H. Ouyang and F. Xu

used for energy/nutrition uptake in metabolism as polysaccharide is degraded into


Glc. During the same process, solid surfaces of substrate like plant cell walls are
penetrated to allow these organisms to establish pathogenic or symbiotic relation-
ships (Gilbert et al. 2008). There are generally two strategies for how the secreted,
cellulose-degrading enzymes work: to form a cell surface-anchored supramolecular
complex called cellulosome, or enzymes acting as cell-free entities. The first case
can been found with a collection of anaerobic cellulolytic bacteria like Bacteroides,
Clostridia, and Ruminococcus (Bayer et al. 2004) as well as some anaerobic cel-
lulolytic fungi like Neocallimastix frontalis (Wilson and Wood 1992) and
Piromyces (Ali et al. 1995). The second case can be found in most of the aerobic
cellulolytic fungi and bacteria. Along with multitudes of EGs, CBHs, and auxiliary
enzymes, extracellular BGLs are transported out of the cell and act in synergy with
the other cellulases to breakdown plant cell walls. Well-established examples are
BGL1 (GH3) and BGL2(GH1) from T. reesei and BGL1(GH1) from A. niger.
Some fungi, such as white rot Phanerochaete chrysoporium, may contain both
cytoplasmic and extracellular BGLs. Some of these may serve in the metabolism of
the organism’s own cell wall, while others may play a role in plant cell wall
metabolism (Igarashi et al. 2003; Tsukada et al. 2006).

4.3.2 Plants

Plant BGLs have been demonstrated with a wide functional diversity in defense,
symbiosis, cell wall catabolism and lignification (monolignol conversion), signaling
and secondary metabolism. The wide spectrum of biological activity is supported by a
large number of BGLs from different GH families. For instance, 48 GH1 genes are
found in Arabidopsis thaliana (Xu et al. 2004), while the number in rice is 40
(Opassiri et al. 2006). These GH1 enzymes cover a cluster of classical myrosinases
and another cluster of myrosinases and BGLs found in the endoplasmic reticulum
(ER) and peroxisome. Interestingly, GH1 enzymes from chloroplast of other plants
such as maize, sorghum and wheat are much more closely related to homologues in
thermophilic bacteria and archaea than rice and Arabidopsis enzymes (Thorlby et al.
2004). In addition, most plants also encode BGLs from GH families 3 and 5 (Opassiri
et al. 2007; Arthan et al. 2006). To defend themselves, plants have developed a wide
range of molecules. The first group is cyanogenic b-glycosides, including linamarin
from clover and cassava, dhurrin from sorghum and prunasin from cherry. These
cyanogenic glycosides can be hydrolyzed by BGL and release their a-hydroxynitrile
moiety, which then breakdown either enzymatically or spontaneously to yield cya-
nide (Poulton 1990; Morant et al. 2008a, b). The second group consists of non-
cyanogenic molecules, such as b- and c-hydroxynitriles (Morant et al. 2008a, b) and
isoflavones in legumes (Suzuki et al. 2006), flavonoids (Chuankhayan et al. 2005),
coumarins, hydroxaminic acids (e.g., 2,4-dihydroxy-7-methoxy-1,4-benzoxazin-
3-one) in maize and wheat (Esen 1992; Sue et al. 2006; Nikus et al. 2001), and
4 The Role and Applications of b-Glucosidases in Biomass … 51

saponins (Nisius 1988). These defensive molecules are also stored as b-D-glucosides
and unleashed by specific BGLs.
In the symbiosis between the endophytic fungus Piriformospora indica and
plant Arabidopsis, the fungus prevents Arabidopsis from overgrowing the roots and
ends up triggering a defense response (Sherameti et al. 2008; Nagano et al. 2005).
A special BGL in ER, AtBGLU23, is revealed to be critical in this symbiosis (Lipka
et al. 2005; Bednarek et al. 2009) by demonstrated activity on scopolin, the most
abundant coumarin glucoside in root (Ahn et al. 2010). Upon cell wall disruption,
plant defensive BGLs and myrosinases often bind to cytoplasmic aggregating
factors, which are believed to localize the otherwise soluble glucosidases at the site
and therefore generate the defense compounds on site (Nisius 1988; Esen and
Blanchard 2000; Kittur et al. 2007). In addition to the plant–microbe interaction,
BGL may also be involved in the plant–insect interaction.
BGLs also play an important role in cell wall metabolism. Several BGLs that
hydrolyze cell wall derived oligosaccharides have been identified. One BGL in
germinating barley seedlings showed activity toward b-1,3- and b-1,4-linked
oligosaccharides (Leah et al. 1995; Hrmova et al. 1998), preferentially man-
nooligosaccharides, which are found in barley endosperm cell walls (Hrmova et al.
2006). Rice seedling BGLs Os3BGlu7, Os3BGlu8, and Os3BGlu26 demonstrate an
increased activity as degree of polymerization (DP) increase from 2 to 6
(Kuntothom et al. 2009), while Os4BGlu12 (Opassiri et al. 2006) is not sensitive to
substrate DP, and Os3BGlu6 (Seshadri et al. 2009) is most efficient on disaccha-
rides. Os3BGlu7 and Os3BGlu8 are widely expressed in rice tissues and are thus
believed able to remodel cell wall by releasing Glc from oligosaccharides derived
from it. Some other BGLs play roles in lignification of secondary cell walls. Two
examples are coniferin BGL from lodgepole pine tree xylem found in differentiating
region of the xylem (Dhamawardhana et al. 1995) and BGL (AtBGLU45/46) from
Arabidopsis. The later has been found active in hydrolyzing coniferin, syringin and
coumaryl alcohol glucosides to release monolignols (Escamilla-Trevino et al.
2006).
Phytohormone activation in many cases also requires glucosides hydrolysis by
BGLs, such as the maize BGL Zm-p60.1 which hydrolyzes and activates cytokinin
b-glucosides (Brzobohaty et al. 1993), an extracellular BGL which hydrolyzes
abscisic acid glucosyl (Glc) ester transported from roots to leaves (Dietz et al.
2000), the BGL hydrolyzing auxin Glc ester 6-O-(4-O)-indole-3-ylacetyl-b-D-Glc
(Jakubowska and Kawalczyk 2005), and the Arabidopsis ER BGL AtBG1 that
hydrolyzes abscissic acid Glc ester in response to drought stress (Lee et al. 2006).
Mutation in this Arabidopsis AtBG1 leads to early germination and defective
stomata closing.
BGLs can also play metabolic roles to remove Glc protective group from
metabolic intermediates and consequently trigger the biosynthesis of various natural
products (phytochemicals) such as monoterpene alkaloids. For instance, one of the
downstream products of strictosidine is raucaffricine, a glucoside that could only be
further metabolized into ajmaline after deglucosylated by raucaffricine BGL
(Barleben et al. 2007).
52 H. Ouyang and F. Xu

Plant BGLs can also release volatile terpenols from glycosides as flower and
fruit scents (Reuveni et al. 1999) or attractants for parasitoid wasp to attack her-
bivores parasites (Mattiacci et al. 1995).

4.3.3 Animals

Some insects feeding on plants also developed GH1 enzymes in their larvae stage,
such as BGLs from Tenebrio molitor and Spodoptera frugiperda (Ferrieira et al.
2001; Marana et al. 2001) and myrosinases from cabbage aphid Brevicoryne
brassicae (Jones et al. 2001). These enzymes have hydrolysis capability on
gluco-oligosaccharides and plant cellobiose, gentiobiose and even amygdalin
(Marana et al. 2001) and share higher sequence similarity with vertebrate
lactase-phloridzin hydrolase (LPH) than with GH1 from plants, indicating their
further divergence from the same animal ancestor after their first divergence from
plants.
Mammalian cells contain BGLs for metabolism of glycolipids and dietary gluco-
sides. Human acid BGLs releases Glc ceramide; defects in this enzyme lead to gly-
coceramides accumulation and eventual Gaucher disease (Butters 2007). Human LPH
is involved in food digestion and displays activity on b-glucosides, b-galactosides and
phloridzin, a polyphenol carrying glycoside found in many fruits. Deficiency of this
enzyme ends up with the widespread lactose intolerance (Tribolo et al. 2007). Human
acid BGL GBA1 and bile acid BGL GBA2 belong to the GH30, and participate in the
metabolism of glycolipids and dietary glucosides (Berrin et al. 2002) (Fig. 4.1).

4.4 Structure

BGLs from various GH families can have amino acid sequences ranging from 420
to 840 units, some comprising signal peptides for secretion or other domains in
addition to the catalytic core. The structures of the catalytic core (Fig. 4.2) can be
divided into three groups. The first one is a (b/a)8-barrel found in GH1, GH5, and
GH30. The second one contains an (a/a)6-barrel found in GH9. The last one
consists of a (b/a)8-barrel and an a/b sandwich in which a 6-stranded b-sheet is
flanked by two sets of three a-helices, and is found in GH3. The (b/a)8-barrel group
features a catalytic domain ranging from 420 to 560 residues in length, depending
on the highly variable loop regions on the b-strands at the C terminus
(Sanz-Aparicio et al. 1998). Within the catalytic domain, two conserved Glu or Asp
from b-strands 4 and 7 respectively are identified as the catalytic acid/base and
nucleophilic residue (Ketudat Cairns and Esen 2010; Vallmitjana et al. 2001;
Henrissat et al. 1995). Monomers of GH1 enzymes can dimerize or form even
higher orders of aggregates. The GH5 BGLs have a (b/a)8-barrel core structure as
the GH1 BGLs (both belonging to the GH-A clan), and the rice GH5 BGL has a
4 The Role and Applications of b-Glucosidases in Biomass … 53

OH OH
OH OH HO OH
HO
HO HO O
O O HO O O
HO HO HO
O OH
O HO O
O OH
OH OH CN
O
CN OH
NC CN

Linamarin Dhurrin D-Amygdalin


Prunasin
OH

OH
OH
O HO
HO HO O
HO O
O
O OH
OH OH
O N OH
HO
O
HO O O
R
O O HO Daidzin R = H
OH
OH
O Genistin R = OH
O
DIMBOA beta-glucoside Apigenin 7-O-beta-glucoside

OH OH
OH
OH

OH HO
OH HO O
HO O
O HO O
HO O OH
HO
O OH
OH O O
OH
OH
Coniferin Phloridzin
HO
O HO
OH
OH
HO
HO O HO
HO O
OH O NH
OH O
O
6

OH
OH
11

OH
HO O Salicin
O
Glucosyl ceramide
HO
O H
OH OH
OH OH
O O
Abscisic acid glucosyl ester
HO
O O OH
OH HO
H O O HO
OH N HO
O OH
HO H
HO N
HO O O Cellobiose
OH
HN
O
OH
OH
OH HO
HO
Indoxyl beta-glucoside Strictosidine HO O O

O
OH OH OH

Laminaribiose

Fig. 4.1 Structures of some BGLs substrates involved in various physiological processes. 1 Plant
defense: 1a cyanogenic glucosides: linamarin (clover/cassava), dhurrin (sorghum), prunasin (cherry)
and its precursor D-amygdalin. 1b noncyanogenic glucosides: 2,4-dihydroxy-7-methoxy-1,4-
benzoxazin-3-one 2-O-b-glucoside (DIMBOA-Glc, maize/wheat), apigenin 7-O-b-glucoside,
daidzin, genistin, phloridzin. 2 Plant cell wall metabolism: coniferin, p-coumaryl alcohol 4-O-
b-glucoside. 3 Plant phytohormone activation: abscisic acid glucosyl ester, indoxyl b-glucoside,
salicin. 4 Plant secondary metabolism: strictosidine, precursor to a wide range of monoterpene
alkaloids. 5 Fungal and bacterial degradation of plant primary cell walls: cellobiose, laminaribiose. 6
Animal (structural, signaling and metabolic functions): glucosylceramide
54 H. Ouyang and F. Xu

fascin-like domain (Opassiri et al. 2007). The (a/a)6-barrel group seen in GH9 is
dominated by a large number of endoglycosidases and leave only a few BGLs like
that from V. cholera. So far there is no conclusive identification of the catalytic
residues on the BGL from this family. However, the study on another GH9 exo-
cellulase has identified a Glu residue on the loop connecting a-helices 12 and 13 as
the catalytic acid/base, while a water molecule activated by two conserved,
hydrogen bond-forming Asp acts as the nucleophilic residue (Schubot et al. 2004).
A similar situation might be expected in BGLs from the same family. For the GH3
enzymes, the interface of the (b/a)8-barrel and a/b sandwich harbors the catalytic
nucleophilic residue (a conserved Asp) and acid/base, which may be variable in
terms of position and identity among different BGLs (Gabrisko and Janecek 2015;
Pozzo et al. 2010; Varghese et al. 1999). A fibronectin type III domain is found at
the C-termini of Thermotoga neapolitana and Aspergillus aculeatus BGLs, and the
domain’s function remains unclear.

Fig. 4.2 Structures of BGLs from different GH families. GH1: T. reesei BGL 2 (PDB: 3AHY),
GH3: T. reesei BGL 1 (PDB: 3ZYZ), GH5: Saccharomyces cerevisiae flavonoid BGL (PDB:
1H4P), GH9: Vibrio parahaemolyticus BGL-like protein (PDB: 3H7L), GH30: Homo sapiens acid
BGL (PDB: 1Y7 V)
4 The Role and Applications of b-Glucosidases in Biomass … 55

4.5 Catalytic Mechanism

All BGLs mentioned so far are exoglycoside hydrolases working on the nonre-
ducing end of a cellodextrin or glucoside substrate. Glucosidases initiate
b-glucosidic bond cleavage with a nucleophilic attack on the anomeric carbon of
the Glc. This is carried out by either the side chain carboxyl of the conserved
catalytic Glu/Asp (Keresztessy et al. 1994), or a water molecule activated by a
conserved Asp. The oxygen in the b-glycosidic bond is activated by the conserved
proximal catalytic acid while the anomeric carbon and the ring oxygen form an
oxocarbenium-like ionic transition state (Fig. 4.3). When the water serves as the
nucleophile (as in the case of GH9), the transition moves forward as the aglycone
moiety leaves the anomeric carbon in concert with the hydroxyl (from the activated
water) displacement on the same carbon to release the Glc (Qi et al. 2008). This
leads to the inversion of chirality on the anomeric carbon and thus is referred as the
inverting mechanism. With the endogenous nucleophile, the first transition state
leads to the leaving of the aglycone moiety as well; however, the b-glycosidic bond
on the anomeric carbon is now displaced with the same carboxyl that initiates the
nucleophilic attack. This a-linked covalent enzyme-glycone intermediate then
undergoes another round of nucleophilic attack on the anomeric carbon. The cat-
alytic acid in the first step gets deprotonated after activating aglycone leaving group
and turns into a catalytic base. It then activates a water to attack the anomeric
carbon to release the Glc from the enzyme-glycone a-linkage. This retaining
mechanism allows the product keep the anomeric chirality. In myrosinases, the
conserved catalytic Glu in the second step is replaced by a Gln, and therefore an
ascorbic acid is required to fulfill the role of the catalytic base.
Unequivocal evidences of the covalent intermediates have been obtained through
incubation of Agrobacterium sp. GH1 BGL with 2,4-dinitrophenyl-2-deoxy-2-
fluoroglucoside (2F-DNPG) (Withers et al. 1987; Withers and Street 1988). The
electronegative property of fluoride destabilizes the transition state upon binding in
the active site, 2,4-dinitrophenyl aglycone moiety is a highly reactive leaving group;
both favor the formation of a lasting and stable enzyme-fluoroglycone intermediate.
The presence of fluoride was firstly supported by 19F-NMR (Withers and Street 1988)
and later observed in crystal structures (Hrmova et al. 2001; Chuenchor et al.
2008) and inactivated enzyme pepsin digest (Withers et al. 1990). In a similar
approach, [3H] conduritol B epoxide (CBE) forms a trapped, radioactively labeled
glycosyl-enzyme intermediate in Aspergillus wentii BGL A3. Radioactivity detected
in trypsin-cleaved fragments establishes the catalytic role of the nucleophilic Asp,
albeit CBE possesses less specificity than 2F-DNPG and labels nearby residues
(Bause and Legler 1980). Nevertheless, the majority of the catalytic residues have
been identified through homology, mutagenesis, chemical modification, and crystal
structure analyses. For instance, the catalytic acid/base residue and nucleophile
residue have been inactivated by mutagenesis, respectively, in the investigation of
retaining mechanism. Replacement of the nucleophile with a small non-nucleophilic
residue, e.g., Ala and Gly, inactivates the enzyme in attacking the anomeric carbon
56 H. Ouyang and F. Xu

(a) Inverting Mechanism

O
O O
H O
OH OH OH
OH O
RO
HO O O O
HO HO
HO OR HO HO
R OH
OH OH OH
O H O H OH

H H

O O HO

O O O

ES E+P
TS

(b) Retaining Mechanism

O O
OH
OH H O
OH
HO O
HO OR RO
HO O
OH HO
O OH O
R OH
O O

ES O
OH
O
TS
HO O H
HO O

OH H
O

O O O
OH
OH H O
OH
HO O Intermediate + P
HO OH O H
HO O
OH HO
O OH O

O O

E+P TS

Fig. 4.3 The inverting and retaining mechanisms of BGLs. The inverting mechanism in
Scheme A is found in the GH9 BGL where water-anomeric carbon bond is formed in a single step
of displacement reaction. However, in the retaining mechanism (Scheme B), water does not attack
the anomeric carbon in the first displacement step which is done in a similar way by the catalytic
nucleophile. This leads to a covalently bonded glycosyl intermediate with inverted anomeric
carbon conformation. The following hydrolysis step ends up with an invertion of anomeric carbon
conformation for the second time and a retaining glycoside product as a result. ES enzyme–
substrate complex, TS 6¼ transition state, E + P (resting) enzyme and product
4 The Role and Applications of b-Glucosidases in Biomass … 57

(Trimbur et al. 1992). However, it can be rescued by exogenous small nucleophiles,


e.g., azide and fluoride, similar to the effect of the activated water in GH9, to form an
a-D-glucoside azide, a chirality-inverted product (Wang et al. 1994; Mackenzie et al.
1998). Alternatively, using a-fluoroglucoside as an intermediate analog could cir-
cumvent the nucleophilic attack and allow the enzyme to release a regular Glc product
after hydrolysis (Ly and Withers 1999). When the catalytic acid/base is replaced by
Gly, water cannot be activated for the hydrolysis while small nucleophiles like azide
again are able to fulfill this step and yield a b-D-glucoside azide (Wang et al. 1995).

4.6 Substrate–Enzyme Interaction

Compared with the thorough characterization of the catalytic residues and their
mechanism in b-glycosidic bond cleavage, less is known about how the interaction
between BGLs and their substrates gives rise to the tremendous substrate diversity
and how the interaction determines BGLs’ substrate specificity. Crystal structures
have revealed that the binding pockets are cavities composed of several subsites. The
subsite that binds the Glc (glycone) unit is assigned subsite −1, whereas the aglycone
moiety binds to the subsites +1, +2, +3 and so on. The substrate specificity of BGL is
a combination of both Glc and aglycone specificities (Marana 2006).
Besides glucosides and cellodextrins, BGLs may act on other glycosides or
oligosacchardies. Such non-BGL activities are related to the coexistence of other
glycosidases in the GH families that BGLs belong to, a likely divergent evolution
effect. The same BGL may display different activities toward substrates with dif-
ferent glycosyls or glycones. For instance, S. frugiperda BGL hydrolyzes fucosides
40 times more quickly than galactosides (Marana et al. 2001). Aspergillus oryzae
GH3 BGL binds Glc well, but mannose (2-epimer of Glc) much more weakly, and
no allose or galactose (3- and 4-epimer) (Langston et al. 2006). GH1 BGLs might
act on b-D-mannosides, b-D-galactosides, b-D-fucosides and b-D-glucuronides,
while GH3 BGLs might act on b-D-xylosides and a-L-arabinosides. Hydrogen
bonding plays a key role in the BGL-substrate interaction (Fig. 4.4). In a set of
studies, the hydrogen bonds involving the glycone hydroxyls and subsite −1 are
studied in three different GH1 BGLs: human LPH (Fernandez et al. 1995),
Agrobacterium sp. Agbglu (Namchuck and Withers 1995) and S. frugiperda Sfbglu
(Marana et al. 2002). For LPH and Agbglu, replacement of the glycone hydroxyls
by hydrogen and/or fluorine is evaluated using enzyme kinetics. In the case of
Sfbglu, a combination of site-directed mutagenesis and enzyme kinetics is deployed
to determine the energy of isolated interaction between subsite −1 and glycone OH
in the glycosylation transition state. The analysis leads to the conclusion that the
specificity of glycone is controlled by a hydrogen bond network involving at least
five amino acid residues and four substrate hydroxyls. A Glu bridging the glycone
hydroxyls 4 and 6 are key in differentiating fucosides, glucosides, and galactosides.
In addition, interactions with the hydroxyl 2 are essential to the BGL activity. In
another set of studies, one GH3 cellodextrinase and three b-xylosidases from rumen
58 H. Ouyang and F. Xu

bacterium Prevotella bryantii B14 are examined for their gylcone specificity by
comparing kinetic parameters on different substrates (Dodd et al. 2010). Mutations
of each conserved residues in subsite −1 severely decrease the catalytic efficiency
without changing the overall structure of the enzyme variants. Similar to GH1
enzymes, replacing the conserved Glu115 (Asp120 in barley glucan b-glucosidase)
leads to 1.1-fold increase in kcat/Km for para-nitrophenyl-b-D-glucopyranoside
(pNP-Glc) and 14-fold decrease for pNP-b-D-xylopyranoside. Glu115 is therefore
believed to contribute to the discrimination between b-xylosides and b-glucosides.
In BGL, the conserved Asp forms hydrogen bond bridging the hydroxyls 4 and 6 of
the Glc unit, while in b-xylosidase, the conserved Glu forms hydrogen bond with
the hydroxyl 4 (Fig. 4.4b).

Fig. 4.4 Elements which are important in glycone (Dodd et al. 2010) and aglycone binding
subsites (Czjzek et al. 2000; Verdoucq et al. 2004). a GH3 active site residues for the b-glucan
exohydrolase (ExoI) from barley bound to Glc are shown (PDB: 1EX1). b Models for GH3
enzyme’s xylosides and glucosides recognition (Dodd et al. 2010). Asp120 from barley ExoI
forms hydrogen bond with the hydroxyls 4 and 6 of Glc. Glu115 from P. bryantii B14 Xyl3B
forms hydrogen bond with the hydroxyl 4 of xylose and may discriminate between Glc and xylose
on the basis of steric interactions. c, d Active site configurations from aglycone side of maize
BGL1 (ZMGlu1, c) and sorghum dhurrinase 1 (Dhr1, d), which are shown for the structures of the
ZMGlu1 Glu189Asp mutant in complex with DIMBOA-Glc (PDB: 1E56) and Dhr1 Glu191Asp
mutant in complex with dhurrin (PDB: 1V03). Phe261 residue narrows the active site in Dhr1 and
is also shown in front of the catalytic nucleophile Glu404
4 The Role and Applications of b-Glucosidases in Biomass … 59

The BGL’s interaction with the aglycone part of the substrate may be more
diverse or complex, as shown by various structure, mutagenesis and chimera
studies. In contrast to the highly conserved amino acid residues at the subsite −1, a
different though overlapping set of residues is involved in interacting with the
aglycone part of the substrate in BGL. An archetypical case is found in the maize
ZmGlu1, a GH1 enzyme, which is active on a wide range of aglycones including
DIMBOA. The structure of one of its inactive variant in complex with
DIMBOA-Glc, DIMBOA and the inhibitor dhurrin shows that the aglycone is
sandwiched by Phe198, Phe205 and Phe466 on one side and Trp378 on the other
(Czjzek et al. 2000). In addition, DIMBOA has a hydrophobic contact with Ala467.
All these residues, except Trp378, are variable among BGLs. For instance, sorghum
dhurrinase SbDhr1 conserves this Trp on the same side of the algycone subsite and
use Val196, Leu203 and Ser462 on the other side in replacement of the three Phe
residues for a tighter and more polar interaction (Verdoucq et al. 2004). The
mutagenesis of the aglycone binding residues in the Zm60.8 isoform of the ZmGlu1
further confirms the importance of these residues (Zouhar et al. 2001). It is thus
believed the conserved Trp is required to bind any aglycone and three or four
homologous positions on the other side would shape the pocket and therefore define
the aglycone specificity. Other factors may contribute to the aglycone binding as
well. It is discovered that the conserved Trp may also differentiate substrates like
dhurrin and strictosidine via its positional variation (Barleben et al. 2007; Verdoucq
et al. 2004; Cicek et al. 2000). Closely related wheat BGL (TaGlu) also hydrolyzes
DIMBOA-Glc, although a different set of residues are present in the binding pocket
(Sue et al. 2006). In the case of rice BGlu1, two of the three corresponding Phe
positions are replaced by Leu and Asn, which makes either no or indirect inter-
action with oligosaccharide substrates (Chuenchor et al. 2008). Instead, Asn245
plays a key role in binding to the third Glc moiety in the substrate, while the
corresponding residues in SbDhr1 and rice Os3BGlu6 appear to block the entrance
to their active sites, with no binding of a trimeric substrate (Opassiri et al. 2003;
Verdoucq et al. 2004). In general, the GH3 BGLs have fewer restraints on the
aglycone part of the substrate than the GH1 BGLs. For instance, A. oryzae
GH3 BGL has similar activities toward cellobiose, sophorose, laminaribiose, and
b-gentiobiose, in which the activated Glc unit (at the subsite −1) is linked to the
‘leaving’ Glc unit (corresponding to the aglycones of 4-glucosides) via 1,4-, 1,2-,
1,3-, and 1,6-glucosidic bond, respectively (Langston et al. 2006).
The GH1 BGLs have narrow and deep wells-like active sites, whose topology
(including space restricted subsites +1 or higher), electrostatic and hydrophobic
interaction properties impact the BGLs’ substrate specificity and product
(Glc) inhibition tolerance (de Giuseppe et al. 2014; Tribolo et al. 2007). The GH3
BGLs have crater-shaped active sites, whose depth, shape, and local amino acids
determine the BGLs’ substrate specificity. The topological difference is believed to
contribute to the difference in substrate reactivity and product inhibition between
the GH1 and GH3 BGLs (de Giuseppe et al. 2014).
60 H. Ouyang and F. Xu

4.7 Enzymatic Properties

4.7.1 Hydrolase Activity

When the substrate concentration is relatively low, BGLs’ main activity is hydro-
lytic, cleaving cellodextrins or oligosaccharides into smaller carbohydrate mole-
cules such as Glc, or glucosides into carbohydrate and aglycone subunits. Various
substrates may be used to assay BGLs’ properties. Chromogenic and fluorogenic
substrates, exemplified by pNP-Glc and 4-methylumbelliferyl-b-D-Glc, are quite
reactive and convenient for spectrophotometric detections, which are suitable for
microplate-based high-throughput or robotized studies (Perry et al. 2007). Because
the chromo- or fluorogenic aglycones are often unnatural, these substrates might not
yield data applicable for quantitative or comprehensive understanding of BGLs’
active sites, especially the subsites +1, +2, etc. Cellodextrins, other oligosaccha-
rides, and glucosides (such as that in Fig. 4.1), are able to reveal BGLs properties
under physiological or real applications conditions. BGLs’ action on these sub-
strates may be monitored with a range of detections including HPLC, mass spec-
trometry, coupled secondary chemical or enzyme reactions for the BGL reaction
products (such as a glucose oxidase or hexokinase assay on Glc), activity-overlay
electrophoresis (Saqib and Whitney 2006), or thermochemistry (Jeoh et al. 2005).
In general, BGLs’ hydrolase activity follows the Michaelis–Menten kinetics.
With cellobiose as the substrate, kcat, Km, and kcat/Km in the order of 101–102 s−1,
100–101 mM, and 101–102 mM−1 s−1 are often observed, while with pNP-Glc as
the substrate, kcat, Km, and kcat/Km in the order of 102 s−1, 100 mM, and
102 mM−1 s−1 are often observed (Ferrara et al. 2014; Uchiyama et al. 2013;
Frutuoso and Marana 2013; Sørensen et al. 2013; Teugjas and Väljamäe; 2013;
Singhania et al. 2013; Kuntothom et al. 2009; Mendonca and Marana 2008;
Eyzaguirre et al. 2005). Significant variations (tenfold or higher) may exist for these
parameters, depending on the BGL active sites (Tsukada et al. 2008) and assay
conditions (buffer, pH, temperature, enzyme and substrate concentrations), and no
significant statistical differences are observed between the GH1 and GH3 BGLs
(p = 0.19, 0.26, 0.59, 0.07 for the kcat (cellobiose), Km (cellobiose), kcat (pNP-Glc),
Km (pNP-Glc), from two-tail t Tests) (Fig. 4.5).
BGLs’ hydrolase activity has an optimal pH range from weakly acidic to neutral,
mainly determined by the actions of the catalytic Asp and/or Glu as the nucleophile
and general acid/base, but also with contributions from other components of the
active site (Tsukada et al. 2008). Most of the BGLs (from the mesophilic organisms)
have optimal temperatures at 50–70 °C, but the BGLs from extreme thermophiles
can have optimal temperatures close to or above 100 °C (Park et al. 2005).
Many molecules may inhibit or inactivate BGLs. Nonspecific, general
protein-denaturing inhibitors include ionic surfactants, heavy metal ions such as Hg+
and Ag+, and high concentration salts (including NaCl) which exert the typical
Hofmeister (salt-out) effect (on the enzymes’ apparent pKa, not the second-order rate
constant) (Bowers et al. 2007). Redox-active metal ions such as Cu2+ and Fe3+ may
4 The Role and Applications of b-Glucosidases in Biomass … 61

1.E+04 1.E+04
kcat, s-1, on cellobiose Km, mM, on cellobiose
1.E+03 1.E+03

1.E+02 1.E+02

1.E+01 1.E+01

1.E+00 1.E+00

1.E-01 1.E-01

1.E-02 1.E-02
GH1 (29) GH3 (57) All (101) GH1 (29) GH3 (59) All (105)
1.E+04 1.E+02
Km, mM, on pNP-Glc
1.E+03
1.E+01

1.E+02
1.E+00
1.E+01
1.E-01
1.E+00

1.E-02
1.E-01
kcat, s-1, on pNP-Glc
1.E-02 1.E-03
GH1 (71) GH3 (61) All (163) GH1 (72) GH3 (70) All (175)

Fig. 4.5 Box-and-whisker plots of the kcat and Km of BGLs, including those from the GH1 and
GH3 family, for the hydrolysis of cellobiose or pNP-Glc. In the parentheses are numbers of BGLs
analyzed. Data are from Brenda enzyme database (www.brenda-enzymes.org) and other literature
sources

inhibit BGL by oxidation (Tejirian and Xu 2010; Jeng et al. 2011; Sharmila et al.
1998). Tris, a commonly used ‘biological’ buffer, may also inhibit BGL by binding
to the active site (Jeng et al. 2011). BGLs may be deactivated by various protein
adsorbents that include soil minerals (Lammirato et al. 2010), oligo/polyphenols
(Tejirian and Xu 2011) and lignin, whose effect can become significant in the
enzymatic biomass hydrolysis (Haven and Jørgensen 2013; Xu et al. 2007).
Specific BGL inhibitors include its substrate, products, and the transition-state
mimics. Because of BGL active sites’ topological features, the products (Glc,
aglycone, or shortened oligosaccharide), which may be regarded as equivalent to
the subunits of a substrate, can bind to certain subsites and impede the enzymatic
reaction. Structural analogs of the products or substrates, or even competing sub-
strates with low-reactivity (e.g., fluoroglucosides), can also inhibit BGLs. The
transition-state mimics can lock up the active site with their structural features
highly similar to that of the activated substrate, and alter the enthalpy–entropy for
the binding, hence exerting competitive inhibitions more potent than the other
62 H. Ouyang and F. Xu

1.E+04 1.E+02
Ki(Glc), mM
1.E+01
1.E+03
1.E+00

1.E+02 1.E-01

1.E-02
1.E+01 1.E-03

1.E-04
1.E+00
Ki(GlcL), mM
1.E-05

1.E-01 1.E-06
GH1 (15) GH3 (44) All (76) GH1 (12) GH3 (25) All (44)

Fig. 4.6 Box-and-whisker plots of the Ki of Glc and GlcL on the hydrolysis of pNP-Glc by BGLs,
including those from the GH1 and GH3 family. In the parentheses numbers of BGLs analyzed.
Data are from Brenda enzyme database (www.brenda-enzymes.org) and other literature sources

inhibitors. The highly inhibitory transition-state mimics include D-glucono-1,5-


lactone (GlcL, the first derivative from Glc oxidation, a side reaction often
accompanying cellulose degradation), deoxynojirimycin, and other
heteroatom-substituted carbohydrate molecules (Gloster and Davies 2010; Gloster
et al. 2007). As inhibitor, Glc and GlcL often show Ki in the order of 101–102 and
10−1–100 mM, respectively, on the pNP-Glc hydrolysis by BGL (Uchiyama et al.
2013; Sørensen et al. 2013; Teugjas and Väljamäe; 2013; Mendonca and Marana
2008; Seidle et al. 2005, 2006; Seidle and Huber 2005; Eyzaguirre et al. 2005;
Marana et al. 2001). Significant variations (10- to 100-fold or higher) may exist for
the parameter, depending on the BGLs, substrates and assay conditions (buffer, pH,
temperature, enzyme, and substrate concentrations) (Fig. 4.6). Statistically, the
GH1 BGLs appear to have higher Ki for Glc (more tolerance to the Glc inhibition)
than that of the GH3 BGLs (p = 0.023), but comparable Ki for GlcL to that of the
GH3 BGLs (p = 0.76, from two-tail t Tests).

4.7.2 Glycosyltransferase/Transglycosylase Activity

When the substrate concentration is high, BGLs (except the GH9 ones) may show
significant glycosyltransferase or transglycosylase activity to synthesize oligosac-
charides or glucosidases, opposing the hydrolase activity (Uchiyama et al. 2013).
The (trans)glycosylation activity is allowed by the reversibility of the retaining
mechanism, when the water is replaced by a carbohydrate or another acceptor with
higher activity for the deacylation of the enzymatic intermediate (donor)
(Fig. 4.3b), as in the cases of other retaining glycosidases. The (trans)glycosylation
may yield sophorosyl, laminaribiosyl, or even cellobiosyl, products via 1,2-, 1,3-, or
1,4-glucosidic bond, or various glucosides (de Giuseppe et al. 2014).
4 The Role and Applications of b-Glucosidases in Biomass … 63

In general, a BGL’s hydrolase activity is controlled by thermodynamic factors


and water activity, while its glycosyltransferase activity is controlled by kinetic
factors and the glycosyl acceptors, with rates independent on pH (Seidle and Huber
2005). The (trans)glycosylation may be detected directly by the related products
formation, or indirectly by the apparent substrate or product inhibition of the
hydrolysis, based on the decrease of the hydrolysis products formation (Frutuoso
and Marana 2013; Kawai et al. 2004). The (trans)glycosylation may contribute to a
BGL’s tolerance to or stimulation by Glc, or maintain a BGL’s turnover. Many
examples of Glc stimulation for the GH1 BGLs, fewer for the GH3 BGLs, have
been reported (de Giuseppe et al. 2014).

4.7.3 Non-BGL Activities

BGLs may have significant side activities on non-Glc substrates (Brenda enzyme
database www.brenda.org, Uchiyama et al. 2013; Sørensen et al. 2013; Singhania
et al. 2013; Kuntothom et al. 2009; Harnipcharnchai et al. 2009; Mendonca and
Marana 2008; Tribolo et al. 2007; Kim et al. 2006; Seidle et al. 2005; Park et al.
2005; Eyzaguirre et al. 2005; Opassiri et al. 2004; Berrin et al. 2002; Hrmova et al.
2002; Vallmitjana et al. 2001; Marana et al. 2001; Perry et al. 2007). With pNP-b-D-
fucopyranoside as substrate, kcat, Km, and kcat/Km of 101–102 s−1, 100–101 mM, and
102 mM−1 s−1 are often observed for BGL’s b-fucosidase activity. With pNP-b-D-
xylopyranoside as substrate, kcat, Km, and kcat/Km of 100–101 s−1, 100 mM, and 100–
103 mM−1 s−1 are often observed for BGL’s b-xylosidase activity. With pNP-a-L-
arabinofuranoside as substrate, kcat, Km, and kcat/Km of 100–101 s−1, 100–101 mM,
and 100–102 mM−1 s−1 are often observed for BGL’s a-arabinosidase activity. With
pNP-b-D-galactopyranoside as substrate, kcat, Km, and kcat/Km of 100–101 s−1, 100–
102 mM, and 10−1–101 mM−1 s−1 are often observed for BGL’s b-galactosidase
activity. Close range of BGL’s activities on these substrates with different gly-
cones indicate that the interaction between the active subsite −1 and the substrate’s
nonreducing glycone subunits 4- and 6-OH (which are changed in the galactosyl,
fucosyl, xylosyl, and arabinosyl subunits of the non-Glc substrates) does not dom-
inate BGL’s activity. In GH1, BGLs coexist with b-fucosidase, b-xylosidase, and
b-galactosidase, as well as b-mannosidase, b-glucuronidase. In GH3, BGLs coexist
with b-xylosidase and a-arabinofuranosidase. Therefore, the side activities might
arise from divergent evolution.

4.7.4 Engineering

Recombinant DNA technology has been applied to BGLs. In combination with 3D


structure and enzymatic reaction studies, site-directed mutagenesis enables eluci-
dation of the structural–function relationship in BGL, especially the aspects on how
64 H. Ouyang and F. Xu

the composition and topology of the active site affect the substrate specificity, pH or
temperature profile, or substrate/product inhibition (Jeng et al. 2012; Khan et al.
2011; Liu et al. 2011 Chuenchor et al. 2011; Tsukada et al. 2008; Mendonca and
Marana 2008; Chuenchor et al. 2008; Vallmitjana et al. 2001).
BGL variants with improved activity, stability, or specificity have been made by
either rational design or directed evolution (Lee et al. 2012; Khan et al. 2011; Hong
et al. 2006; Hancock et al. 2005). Engineering the active site (especially the subsites
that bind the aglycone of a substrate) has been pursued for the abatement of the
product (Glc) inhibition; however, the increase in the Glc tolerance is often
accompanied by a decrease in the hydrolase activity, due to the contribution made
by the BGL-Glc interaction to both the catalysis and inhibition. Domain swap
between mesophilic and thermophilic BGLs may lead to improved thermal or other
properties for BGLs originated from mesophiles, such as the chimera constructed
from Agrobacterium tumefaciens or Cellvibrio gilvus and Thermotoga maritime
(Deog and Kiyoshi 2011; Kim et al. 2006).
Most BGLs have no distinct functional domains other than the catalytic core
(although some GH3 BGLs have a C-terminal FnIII domain). The enzymes may be
engineered to possess domains with targeted functions, such as the carbohydrate-binding
module potentially useful for the enzymatic biomass hydrolysis (Gundllapalli et al.
2007).
The transglycosylase activity of BGLs may be improved by engineering
(Frutuoso and Marana 2013; Jeng et al. 2012; Lundemo et al. 2013; Shim et al.
2012; Choi et al. 2008; Feng et al. 2005). For instance, the Trp49 and Trp262 of
A. niger GH3 BGL are found regulating the preference to the transglycosylase and
hydrolase activity, and substitutions of the two Trp result in BGL variants with
abated or enhanced relative transglycosylase activity (Seidle et al. 2005, 2006). The
regioselectivity of a transglycosylating BGL may also be modulated by site-directed
mutagenesis (Choi et al. 2008).

4.8 Application

4.8.1 Synthesis of Oligosaccharides and Glycosides

BGLs have the potential in biosynthesis of functional, performance, or therapeutic


oligosaccharides, glycol-conjugates, and glycosides, based on their glycosyltrans-
ferase activity. To increase the efficiency or selectivity, it is desirable to engineer
BGLs so that their hydrolase activity can be minimized. Engineering on BGL may be
carried out by site-directed mutagenesis or directed evolution (Shim et al. 2012). The
kinetic and thermodynamic determinants for the transglycosylation and ‘reverse
hydrolysis’ (formation of the compounds that would serve as the substrates for the
hydrolase activity), respectively, can be explored by the engineering or changes on
the reaction conditions (Bhatia et al. 2002). For instance, alterations of an amino acid
at the subsite +2 of T. neapolitana GH1 BGL are able to enhance the enzyme’s
4 The Role and Applications of b-Glucosidases in Biomass … 65

glycosyltransferase-to-hydrolase activity ratio, enabling the enzyme to catalyze


more efficiently the synthesis of alkyl glycosides as attractive surfactants (Lundemo
et al. 2013). The alkyl glycosides may also be synthesized by alcohol-tolerant or
stimulating BGL in solvents high in alcohol (Karnaouri et al. 2013).

4.8.2 Beverage, Brewing, Food, Feed, and Dietary

Combination of flavor precursors and b-glucosidases is widely used in beverage and


food production. BGLs may be added in different stage of production and brewing
to act on specific precursors to release the desired aglycones (e.g., terpenols, benzyl
alcohols, and phenylethyl alcohols) for flavor enhancement and stability.
A combination of a-rhamnosidase and BGL may be applied to debitter citrus fruits
by hydrolyzing bitter glucosides such as prunin and naringin, while BGL may be
applied to decolorize juices or wines by hydrolyzing various anthocyanins such as
malvidin, peonidin, and cyanidin (Eyzaguirre et al. 2005). The astringent diadzin,
genistin, or other isoflavonoid glucosides in soybean products may be hydrolyzed
by BGL to daidzein, genistein, or other isoflavones with more pleasant taste. BGLs
are also used in nutrition enrichment of feeds, foods and beverages to generate
vitamins, antioxidants and other dietary molecules from their glycoside precursors.
For instance, vitamin B6 is stored predominantly in its glycoside form and can be
released by rice BGLs (Opassiri et al. 2003). Feeds for the monogastric farm
animals like pigs and chickens are often treated with crude BGLs to improve
nutrition availability together with the native enzymes in the animal’s small
intestine. Aglycones from soybean isoflavone glycosides and phenylthiocarbamide
derived from cruciferous vegetable glycosides have antioxidant and anticarcino-
genic effect. They are also direct or derivative products from BGL and myrosinase
hydrolysis, respectively (Chuankhayan et al. 2007; Esen 2003).

4.8.3 Biomass Conversion

Biomass conversions lay the basis for the bioenergy, biorefinery, and other
biomass-sustained industries. The enzymatic hydrolysis of lignocellulose is a key
step of the processes, and its development has benefited greatly from the basic
biochemical studies on the natural lignocellulose degradation (Payne et al. 2015;
Xie et al. 2014; Sweeney and Xu 2012; van den Brink and de Vries 2011; Quinlan
et al. 2010; Xu 2004, 2010a, b; McFarland et al. 2007; Teter et al. 2006). The
industrial enzymatic biomass conversion can differ significantly from the natural
process in various aspects, especially the requirement for high efficiency: the much
shortened reaction time (days instead of years), enhanced turnover (to minimize the
cost of enzymes), and close to 100 % yield (to maximize the output), need for
enzymes to tolerate high levels of end products and insoluble substrates that are
66 H. Ouyang and F. Xu

often altered in composition or structure by pretreatments. Cellulolytic microbes


secrete consortia of enzymes that can work cooperatively and synergistically, but
they need adjustment in industrial applications, in terms of composition and
specificity. T. reesei is one of the best known cellulolytic microbes and industrial
hemicellulases and cellulases producers. Although the filamentous fungus can
secrete more than 20 lignocellulose-active enzymes, including at least one BGL
(Herpoël-Gimbert et al. 2008), its enzyme consortium cannot effectively hydrolyze
various industrial biomass feedstocks (such as dilute acid pretreated corn stover),
unless supplemented with exogenous BGLs (such as A. oryzae GH3 BGL), as
demonstrated by the development of the first generation of the industrial biomass
enzymes products (Rani et al. 2014; Sørensen et al. 2013; Teugjas and Väljamäe
2013; Tiwari et al. 2013; Singhania et al. 2013; Krisch et al. 2010; Eyzaguirre et al.
2005; Xu 2004; Bhatia et al. 2002). During an industrial biomass hydrolysis, the
CBHs and EGs of T. reesei hydrolyze cellulose and produce high levels of cel-
lobiose. The cellobiose cannot be degraded effectively by the native BGL present in
the fungus’ secretome in low abundance, and thus a severe product inhibition
builds on the CBHs and EGs, resulting in an underperformance of the overall
enzymatic hydrolysis. The inhibition can be mitigated by increasing the BGL
abundance.
Supplementing the BGL activity to an industrial hemicellulase and cellulase
consortium by co-expressing the enzyme in the same host (that produces the
hemicellulase and cellulase) is more economically viable than producing the enzyme
separately. Often, heterologous expression of a non-native BGL is selected to take
advantage of the enzyme’s superior properties (in comparison with the endogenous
BGL). The selection is initiated by multiple rounds of screening of individual BGLs,
based on activity or bioinformatics, for either wild-type or engineered variants. The
initial screening is followed by synergy study with other hemicellulase and cellulase
acting under industrial conditions and by heterologous expression trials. The pre-
ferred properties of superior BGLs include high specific activity, high thermal sta-
bility, suitable pH profile, low production inhibition, low lignin absorption, low
glycosyltransferase side activity, and high expression yield. A BGL’s stimulation by
Glc or xylose may further improve the enzyme’s action in hydrolyzing high con-
centrations of cellulosic materials (Souza et al. 2010). A BGL’s tolerance of, or
stimulation by, ethanol or other alcohol may further improve the enzyme’s action in
the biomass conversion processes where elevated levels of ethanol are present, such
as the simultaneous hydrolysis (saccharification) and fermentation (SHF, SSF) or
consolidated bioprocessing (CBP) (Karnaouri et al. 2013).
BGLs may be applied in individual, free forms as in the secretomes of T. reesei
and most of the aerobic cellulolytic fungi. The relative abundance (molar ratio) of
BGL in biomass-active enzyme consortia may be roughly adjusted by co-expression
or post-production mixing. BGLs engineered with altered properties, including that
gained through fused functional domains, such as the carbohydrate-binding-module
(Gundllapalli et al. 2007), may be used. The claimed benefits of applying BGL in
4 The Role and Applications of b-Glucosidases in Biomass … 67

free forms include the freedom a BGL has to act close to other enzymes or away
from them, pursuing diffused cellobiose (or other oligosaccharides) or avoiding
common deactivators.
BGLs may also be applied in associated, part of supramolecular assembly forms
as in the cellulosomes of Clostridium thermocellum and most of the anaerobic
cellulolytic bacteria. The molar ratio of the BGL in biomass-active cellulosomes
may be finely adjusted by specific dockerin–cohesin pairing. Non-native or non-
cellulosomal BGLs may be incorporated into designer cellulosomes (with selected
enzyme composition and molar ratio) with fused dockerins (Kellermann and
Rentmeister 2014; Hyeon et al. 2013). Working in the same supramolecular
assembly allows BGL to cooperate with other enzymes in close vicinity.
BGLs may also be applied in cell surface display forms. A BGL can be dis-
played, alone or with other biomass-active enzymes, on the surface of a fermen-
tative but noncellulolytic yeast or bacterium. The displayed BGL and other
enzymes can hydrolyze biomass to Glc and other fermentable sugars, which are
then fermented by the yeast or bacterium in situ. In the so-called consolidated
bioprocessing process, enzyme production and biomass-to-ethanol conversion are
done in a single unit process step, by engineered microbes such as the thermophilic
Kluyveromyces marxianus displaying A. aculeatus BGL and T. reesei EG
(Kellermann and Rentmeister 2014; Yamada et al. 2013; van Zyl et al. 2007).

References

Ahn YO, Shimizu B, Sakata K, Gantulga D, Zhou Z, Bevan DR, Esen A (2010)
Scopulin-hydrolyzing b-glucosidases in the roots of Arabidopsis. Plant Cell Physiol
51:131–143
Ali BR, Zhou L, Graves FM, Freedman RB, Black GW, Gilbert HJ, Hazelwood GP (1995)
Cellulases and hemicellulases of the anaerobic fungus Piromyces constitute a multiprotein
cellulose-binding complex and encoded by multigene families. FEMS Microbiol Lett
125:15–22
Arthan D, Kittakoop P, Esen A, Svasti J (2006) Furostanol glycoside 26-O-b-glucosidase from the
leaves of Solanum torvum. Phytochemistry 67:27–33
Barleben L, Panjikar S, Ruppert M, Koepke J, Stockigt J (2007) Molecular architecture of
strictosidine glucosidase: the gateway to the biosynthesis of the monoterpenoid indole alkaloid
family. Plant Cell 19:2886–2897
Barnett CC, Berka RM, Fowler T (1991) Cloning and amplification of the gene encoding and
extracellular b-glycosidase from Trichoderma reesei: evidence for improved rates of
saccharification of cellulosic substrates. Nat Biotechnol 9:552–567
Bause E, Legler G (1980) Isolation and structure of a tryptic glycopeptide from the active site of
b-glucosidase A3 from Aspergillus wentii. Biochim Biophys Acta 626:459–465
Bayer EA, Belaich JP, Shoham Y, Lamed R (2004) The cellulosomes: multienzyme machines for
degradation of plant cell wall polysaccharides. Annu Rev Microbiol 58:521–554
Bednarek P, Pislewska-Bednarek M, Svatos A, Schneider B, Doubsky J, Mansurova M,
Humphry M, Consonni C, Panstruga R, Sanchez-Vallet A, Molina A, Schulze-Lefert P (2009)
A glucosinolate metabolism pathway in living plant cells mediates broad-spectrum antifungal
defense. Science 323:101–106
68 H. Ouyang and F. Xu

Berrin JG, McLauchlan WR, Needs P, Williamson G, Puigserver A, Kroon PA, Juge N (2002)
Functional expression of human liver cytosolic b-glucosidase in Pichia pastoris. Insights into
its role in the metabolism of dietary glucosides. Eur J Biochem 269:249–258
Bhatia Y, Mishra S, Bisaria VS (2002) Microbial b-glucosidases: cloning, properties, and
applications. Crit Rev Biotechnol 22:375–407
Bowers EM, Ragland LO, Byers LD (2007) Salt effects on b-glucosidase: pH-profile narrowing.
Biochim Biophys Acta 1774:1500–1507
Brzobohaty B, Moore I, Kristoffersen P, Bako L, Campos N, Schell J, Palme K (1993) Release of
active cytokinin by a b-glucosidase localized to the maize root meristem. Science 262:
1051–1054
Butters TD (2007) Gaucher disease. Curr Opin Chem Biol 11:412–418
Choi W, Park KM, Jun SY, Park CS, Park KH, Cha J (2008) Modulation of the regioselectivity of
a Thermotoga neapolitana b-glucosidase by site-directed mutagenesis. J Microbiol Biotechnol
18:901–907
Chuankhayan P, Hua Y, Svasti J, Sakdarat S, Sullivan PA, Ketudat Cairns JR (2005) Purification
of an isoflavonoid 7-O-bapiosyl-glucoside b-glycosidase and its substrates from Dalbergia
nigrescens Kurz. Phytochemistry 66:1880–1889
Chuankhayan P, Rimlumduan T, Svasti J, Ketudat Cairns JR (2007) Hydrolysis of soybean
isoflavonoid glycosides by Dalbergia b-glucosidases. J Agric Food Chem 55:2407–2412
Chuenchor W, Pengthaisong S, Robinson RC, Yuvaniyama J, Svasti J, Ketudat Cairns JR (2011)
The structural basis of oligosaccharide binding by rice BGlu1 beta-glucosidase. J Struct Biol
173:169–179
Chuenchor W, Pengthaisong S, Robinson RC, Yuvaniyama J, Oonanant W, Bevan DR, Esen A,
Chen CJ, Opassiri R, Svasti J, Ketudat Cairns JR (2008) Structural insights into rice BGlu1
b-glucosidase oligosaccharide hydrolysis and transglycosylation. J Mol Biol 377:1200–1215
Cicek M, Blanchard D, Bevan DR, Esen A (2000) The aglycone specificity-determining sites are
different in 2,4-dihydroxy-7-methoxy-1,4-benzoxazin-3-one (DIMBOA)-glucosidase (maize
beta-glucosidase) and dhurrinase (sorghum beta-glucosidase). J Biol Chem 275:20002–20011
Czjzek M, Cicek M, Zamboni V, Bevan DR, Henrissat B, Esen A (2000) The mechanism of
substrate (aglycone) specificity in b-glucosidases is revealed by crystal structures of mutant
maize b-glucosidase-DIMBOA, -DIMBOAGlc, and dhurrin complexes. Proc Natl Acad
Sci USA 97:13555–13560
Dan S, Marton T, Dekol M, Bravdo BA, He S, Withers SG, Shoserov O (2000) Cloning,
expression, characterization and nucleophile identification of family 3 Aspergillus niger
b-glycosidase. J Biol Chem 275:4973–4980
Deog KJ, Kiyoshi H (2011) Construction and characterization of novel chimeric b-glucosidases
with Cellvibrio gilvus (CG) and Thermotoga maritima (TM) by overlapping PCR. In: Carpi A
(ed) Progress in molecular and environmental bioengineering—from analysis and modeling to
technology applications. InTech, Rijeka, pp 633–646
de Giuseppe PO, Souza Tde A, Souza FH, Zanphorlin LM, Machado CB, Ward RJ, Jorge JA,
Furriel Rdos P, Murakami MT (2014) Structural basis for glucose tolerance in GH1
b-glucosidases. Acta Crystallogr D Biol Crystallogr 70:1631–1639
Dhamawardhana DP, Ellis BE, Carlson JE (1995) A b-glucosidase from lodgepole pine xylem
specific for the lignin precursor coniferin. Plant Physiol 107:331–339
Dietz KJ, Sauter A, Wichert K, Messdaghi D, Hartung W (2000) Extracellular b-glucosidase
activity in barley involved in the hydrolysis of ABA glucose conjugate in leaves. J Exp Bot
51:937–944
Dodd D, Kiyonari S, Mackie RI, Cann IK (2010) Functional diversity of four glycoside hydrolase
family 3 enzymes from the rumen bacterium Prevotella bryantii B14. J Bacteriol 192:
2335–2345
Escamilla-Trevino LL, Chen W, Card ML, Shih MC, Cheng CL, Poulton JE (2006) Arabidopsis
thaliana b-glucosidases BGLU45 and BGLU46 hydrolyse monolignol glucosides.
Phytochemistry 67:1651–1660
4 The Role and Applications of b-Glucosidases in Biomass … 69

Esen A (1992) Purification and partial characterization of maize (Zea mays L.) b-glucosidase.
Plant Physiol 98:174–182
Esen A (2003) b-Glucosidases. In: Whitaker JR, Voragen AGJ, Wong DWS (eds) Handbook of
food enzymology. Marcel Dekker, New York, pp 791–804
Esen A, Blanchard DJ (2000) A specific b-glucosidase-aggregating factor (BGAF) is responsible
for the b-glucosidase null phenotype in maize. Plant Physiol 122:563–572
Eyzaguirre J, Hidalgo M, Leschot A (2005) b-Glucosidases from filamentous fungi: properties,
structure, and applications. In: Yarema KJ (ed) Handbook of carbohydrate engineering. Taylor
& Francis, Boca Raton, pp 645–685
Faure D, Desair J, Keijers V, Bekri MA, Proost P, Henrissat B, Vanderleyden J (1999) Growth of
Azospirillum irakense KBC 1 on the aryl b-glycosidase salicin requires either Sal A or Sal B.
J Bacteriol 181:3003–3009
Feng HY, Drone J, Hoffmann L, Tran V, Tellier C, Rabiller C, Dion M (2005) Converting a
b-glycosidase into a b-transglycosidase by directed evolution. J Biol Chem 280:37088–37097
Fernandez P, Canada FJ, Jimenez-Barbero J, Martın-Lomas M (1995) Substrate specificity of
small-intestinal lactase: study of the steric effects and hydrogen bonds involved in
enzyme-substrate interaction. Carbohydr Res 271:31–42
Ferrara MC, Cobucci-Ponzano B, Carpentieri A, Henrissat B, Rossi M, Amoresano A, Moracci M
(2014) The identification and molecular characterization of the first archaeal bifunctional
exo-beta-glucosidase/N-acetyl-beta-glucosaminidase demonstrate that family GH116 is made
of three functionally distinct subfamilies. Biochim Biophys Acta 1840:367–377
Ferrieira AHP, Marana SR, Terra WR, Ferreira C (2001) Purification, molecular cloning, and
properties of a b-glycosidase isolated from midgut lumen of Tenebrio molitor (Coleoptera)
larvae. Insect Biochem Mol Biol 31:1065–1076
Frutuoso MA, Marana SR (2013) A single amino acid residue determines the ratio of hydrolysis to
transglycosylation catalyzed by b-glucosidases. Protein Peptide Lett 20:102–106
Gabrisko M, Janecek S (2015) Novel family GH3 b-glucosidases or b-xylosidases of unknown
function found in various animal groups, including birds and reptiles. Carbohydr Res 408:
v44–50
Geerlings A, Ibanez MML, Memelinks J, Heijden RVD, Verpoorte R (2000) Molecular cloning
and analysis of strictosidine b-D-glycosidase, an enzyme in terpenoid indole alkaloid
biosynthesis in Catharanthus roseus. J Biol Chem 275:3051–3056
Gilbert HJ, Stalbrand H, Brumer H (2008) How the walls come tumbling down: recent structural
biochemistry of plant polysaccharide degradation. Curr Opin Plant Biol 11:338–348
Gloster TM, Davies GJ (2010) Glycosidase inhibition: assessing mimicry of the transition state.
Org Biomol Chem 8:305–320
Gloster TM, Meloncelli P, Stick RV, Zechel D, Vasella A, Davies GJ (2007) Glycosidase
inhibition: an assessment of the binding of 18 putative transition-state mimics. J Am Chem Soc
129:2345–2354
Gundllapalli SB, Pretorius IS, Cordero Otero RR (2007) Effect of the cellulose-binding domain on
the catalytic activity of a b-glucosidase from Saccharomycopsis fibuligera. J Ind Microbiol
Biotechnol 34:413–421
Hancock SM, Corbett K, Fordham-Skelton AP, Gatehouse JA, Davis BG (2005) Developing
promiscuous glycosidases for glycoside synthesis: residues W433 and E432 in Sulfolobus
solfataricus beta-glycosidase are important glucoside- and galactoside-specificity determinants.
ChemBioChem 6:866–875
Harnipcharnchai P, Champreda V, Sornlake W, Eurwilaichitr L (2009) A thermotolerant
b-glucosidase isolated from an endophytic fungi, Periconia sp. with a possible use for biomass
conversion to sugars. Prot Express Purif 67:61–69
Haven MØ, Jørgensen H (2013) Adsorption of b-glucosidases in two commercial preparations
onto pretreated biomass and lignin. Biotechnol Biofuels 6:165
Henrissat B, Callebaut I, Fabrega S, Lehn P, Mornon JP, Davies G (1995) Conserved catalytic
machinery and the prediction of a common fold for several families of glycosyl hydrolases.
Proc Natl Acad Sci USA 92:7090–7094
70 H. Ouyang and F. Xu

Herpoël-Gimbert I, Margeot A, Dolla A, Jan G, Mollé D, Lignon S, Mathis H, Sigoillot JC,


Monot F, Asther M (2008) Comparative secretome analyses of two Trichoderma reesei
RUT-C30 and CL847 hypersecretory strains. Biotechnol Biofuels 1:18
Hong J, Tamaki H, Kumagai H (2006) Unusual hydrophobic linker region of b-glucosidase
(BGLII) from Thermoascus aurantiacus is required for hyper-activation by organic solvents.
Appl Microbiol Biotechnol 73:80–88
Hrmova M, MacGregor EA, Biely P, Stewart RJ, Fincher GB (1998) Substrate binding and
catalytic mechanism of a barley b-D-glucosidase/(1, 4)-b-D-glucan exohydrolase. J Biol Chem
273:11134–11143
Hrmova M, Varghese JN, De Gori R, Smith BJ, Driguez H, Fincher GB (2001) Catalytic
mechanisms and reaction intermediates along the hydrolytic pathway of a plant beta-D-glucan
glucohydrolase. Structure 9:1005–1016
Hrmova M, De Gori R, Smith BJ, Fairweather JK, Driquez H, Varghese JN, Fincher GB (2002)
Structural basis for broad substrate specificity in higher plant b-D-glucan glucohydrolases.
Plant Cell 14:1033–1052
Hrmova M, Burton RA, Biely P, Lahnstein J, Fincher GB (2006) Hydrolysis of (1,4)-b-D-mannans
in barley (Hordeum vulgare L.) is mediated by the concerted action of (1,4)-b-D-mannans
endohydrolase and b-D-mannosidase. Biochem J 399:77–90
Hyeon JE, Jeon SD, Ha SO (2013) Cellulosome-based, Clostridium-derived multi-functional
enzyme complexes for advanced biotechnology tool development: advances and applications.
Biotechnol Adv 31:936–944
Igarashi K, Tani T, Kawal R, Samejima M (2003) Family 3 beta-glucosidase from
cellulose-degrading culture of the white-rot fungus Phanerochaete chrysosporium. J Biosci
Bioeng 95:572–576
Iwashita K, Nagahara T, Kimura H, Takano M, Shimoi K, Ito K (1999) The bgl gene of
Aspergillus kawachii encodes both extracellular and wall bound b-glycosidases. Appl Environ
Microbiol 65:5546–5553
Jakubowska A, Kawalczyk S (2005) A specific enzyme hydrolyzing 6-O(4-O)-
indole-3-ylacetyl-b-D-glucose in immature kernels of Zea mays. J Plant Physiol 162:207–213
Jeng WY, Wang NC, Lin CT, Chang WJ, Liu CI, Wang AHJ (2012) High-resolution structures of
Neotermes koshunensis b-glucosidase mutants provide insights into the catalytic mechanism
and the synthesis of glucoconjugates. Acta Cryst D68:829–838
Jeng WY, Wang NC, Lin MH, Lin CT, Liaw YC, Chang WJ, Liu CI, Liang PH, Wang AH (2011)
Structural and functional analysis of three b-glucosidases from bacterium Clostridium
cellulovorans, fungus Trichoderma reesei and termite Neotermes koshunensis. J Struct Biol
173:46–56
Jeoh T, Baker JO, Ali MK, Himmel ME, Adney WS (2005) b-D-Glucosidase reaction kinetics
from isothermal titration microcalorimetry. Anal Biochem 347:244–253
Jones AME, Bridges M, Bones AM, Cole R, Rossiter JT (2001) Purification and characterisation
of a non-plant myrosinase from the cabbage aphid Brevicoryne brassicae. Insect Biochem Mol
Biol 31:1–5
Karnaouri A, Topakas E, Paschos T, Taouki I, Christakopoulos P (2013) Cloning, expression and
characterization of an ethanol tolerant GH3 b-glucosidase from Myceliophthora thermophile.
PeerJ 1:e46
Kawai R, Igarashi K, Kitaoka M, Ishii T, Samejima M (2004) Kinetics of substrate
transglycosylation by glycoside hydrolase family 3 glucan (1 ! 3)-beta-glucosidase from
the white-rot fungus Phanerochaete chrysosporium. Carbohydr Res 339:2851–2857
Kellermann SJ, Rentmeister A (2014) Current developments in cellulase engineering.
ChemBioEng Rev 1:6–13
Keresztessy Z, Kiss L, Hughes MA (1994) Investigation of the active site of the cyanogenic beta-
D-glucosidase (linamarase) from Manihot esculenta Crantz (cassava). II. Identification of
Glu-198 as an active site carboxylate group with acid catalytic function. Arch Biochem
Biophys 315:323–330
Ketudat Cairns JR, Esen A (2010) b-Glucosidases. Cell Mol Life Sci 67:3389–3405
4 The Role and Applications of b-Glucosidases in Biomass … 71

Khan S, Pozzo T, Megyeri M, Lindahl S, Sundin A, Turner C, Karlsson EN (2011) Aglycone


specificity of Thermotoga neapolitana b-glucosidase 1A modified by mutagenesis, leading to
increased catalytic efficiency in quercetin-3-glucoside hydrolysis. BMC Biochem 12:11
Kim BJ, Singh SP, Hayashi K (2006) Characteristics of chimeric enzymes constructed between
Thermotoga maritima and Agrobacterium tumefaciens beta-glucosidases: role of C-terminal
domain in catalytic activity. Enzyme Microb Technol 38:952–959
Kittur FS, Lalgondar M, Yu HY, Bevan DR, Esen A (2007) Maize b-glucosidase-aggregating
factor is a polyspecific jacalin related chimeric lectin, and its lectin domain is responsible for
b-glucosidase aggregation. J Biol Chem 282:7299–7311
Krisch J, Takó M, Papp T, Vágvölgyi C (2010) Characteristics and potential use of b-glucosidases
from Zygomycetes. In: Vilas AM (ed) Current research, technology and education topics in
applied microbiology and microbial biotechnology extremadura. Formatex Research Center,
Spain, pp 891–896
Kuntothom T, Luang S, Harvey AJ, Fincher GB, Opassiri R, Hrmova M, Ketudat Cairns JR
(2009) Rice family GH1 glycosyl hydrolases with b-D-glucosidase and b-D-mannosidase
activities. Arch Biochem Biophys 491:84–95
Lammirato C, Miltner A, Wick LY, Kästner M (2010) Hydrolysis of cellobiose by b-glucosidase
in the presence of soil minerals—interactions at solid-liquid interfaces and effects on enzyme
activity levels. Soil Biol Biochem 42:2203–2210
Langston J, Sheehy N, Xu F (2006) Substrate specificity of Aspergillus oryzae family 3
b-glucosidase. Biochim Biophys Acta Proteins Proteom 1764:972–978
Leah R, Kiegel J, Suendson IB, Mundy J (1995) Biochemical and molecular characterization of a
barley seed b-glycosidase. J Biol Chem 22:15789–15796
Lee HL, Chang CK, Jeng WY, Wang AH, Liang PH (2012) Mutations in the substrate entrance
region of b-glucosidase from Trichoderma reesei improve enzyme activity and thermostability.
Protein Eng Des Sel 25:733–740
Lee KH, Piao HL, Kim HY, Choi SM, Jiang F, Hartung W, Hwang I, Kwak JM, Lee IJ (2006)
Activation of glucosidase via stress-induced polymerization rapidly increases active pools of
abscisic acid. Cell 126:1109–1120
Lipka V, Dittgen J, Bednarek P, Bhat R, Wiermer M, Stein M, Landtag J, Brandt W, Rosahl S,
Scheel D, Llorente F, Molina A, Parker J, Somerville S, Schulze-Lefert P (2005) Pre- and
postinvasion defenses both contribute to nonhost resistance in Arabidopsis. Science 310:1180–
1183
Liu J, Zhang X, Fang Z, Fang W, Peng H, Xiao Y (2011) The 184th residue of b-glucosidase
Bgl1B plays an important role in glucose tolerance. J Biosci Bioengineer 112:447–450
Lundemo P, Adlercreutz P, KarlssonImproved EN (2013) Transferase/hydrolase ratio through
rational design of a family 1 b-glucosidase from Thermotoga neapolitana. Appl Environ
Microbiol 79:3400–3405
Ly HD, Withers SG (1999) Mutagenesis of glycosidases. Annu Rev Biochem 68:487–522
Machida M, Ohtsuki I, Fukui S, Yamashita I (1988) Nucleotide sequences of Saccharomycopsis
fibuligera genes for extracellular b-glycosidases as expressed in Saccharomyces cerevisiae.
Appl Environ Microbiol 54:3147–3155
Mackenzie LF, Wang Q, Warren RAJ, Withers SG (1998) Glycosynthases: mutant glycosidases
for oligosaccharide synthesis. J Am Chem Soc 120:5583–5584
Marana SR (2006) Molecular basis of substrate specificity in family 1 glycoside hydrolases.
IUBMB Life 58:63–73
Marana SR, Jacobs-Lorena M, Terra WR, Ferrieira C (2001) Amino acid residues involved in
substrate binding and catalysis in an insect digestive b-glycosidase. Biochim Biophys Acta
1545:41–52
Marana SR, Terra WR, Ferreira C (2002) The role of amino acid residues Q39 and E451 in the
determination of substrate specificity of the Spodoptera frugiperda b-glycosidase. Eur J
Biochem 269:3705–3714
72 H. Ouyang and F. Xu

Marri L, Valentini S, Venditti D (1995) Cloning and nucleotide sequence of bglA from Erwinia
herbicola and expression of b-glycosidase activity in Escherichia coli. FEMS Microbiol Lett
128:135–138
Matsui I, Sakai Y, Matsui E, Kikuchi M, Kawarabayasi Y, Honda K (2000) Novel substrate
specificity of a membrane-bound b-glycosidase from the hyperthermophilic archeon
Pyrococcus horikoshii. FEBS Lett 467:195–200
Mattiacci L, Dicke M, Posthumus MA (1995) Beta-glucosidase: an elicitor of herbivore-induced
plant odor that attracts hostsearching parasitic wasps. Proc Natl Acad Sci USA 92:2036–2040
McFarland KC, Ding H, Teter S, Vlasenko E, Xu F, Cherry J (2007) Development of improved
cellulase mixtures in a single production organism. In: Eggleston G, Vercellotti JR
(eds) Industrial application of enzymes on carbohydrate based materials. American Chemical
Society & Oxford University Press, New York, pp 19–31
Mendonca LMF, Marana SR (2008) The role in the substrate specificity and catalysis of residues
forming the substrate aglycone-binding site of a b-glycosidase. FEBS J 275:2536–2547
Morant AV, Bjarnholt N, Kragh ME, Kjaergaard CH, Jørgensen K, Paquette SM, Piotrowski M,
Imberty A, Olsen CE, Møller BL, Bak S (2008a) The beta-glucosidases responsible for
bioactivation of hydroxynitrile glucosides in Lotus japonicus. Plant Physiol 147:1072–1091
Morant AV, Jørgensen K, Jørgensen C, Paquette SM, Sanchez-Perez R, Møller BL, Bak S (2008b)
b-Glucosidases as detonators of plant chemical defense. Phytochemistry 69:1795–1813
Nagano AJ, Matsushima R, Hara-Nishimura I (2005) Activation of an ER-body-localized
b-glucosidase via a cytosolic binding partner in damaged tissues of Arabidopsis thaliana. Plant
Cell Physiol 46:1140–1148
Namchuck NM, Withers SG (1995) Mechanism of Agrobacterium b-glucosidase: kinetic analysis
of the role of noncovalent enzyme/substrate interactions. Biochemistry 34:16194–16202
Nikus J, Daniel G, Jonsson LM (2001) Subcellular localization of beta-glucosidase in rye, maize
and wheat seedlings. Plant Physiol 111:466–472
Nisius A (1988) The stroma centre in Avena plastids: an aggregation of b-glucosidase responsible
for the activation of oat-leaf saponins. Planta 173:474–481
Opassiri R, Hua Y, Wara-Aswapati O, Akiyama T, Svasti J, Esen A, Ketudat Cairns JR (2004)
b-Glucosidase, exo-b-glucanase and pyridoxine transglucosylase activities of rice BGlu1.
Biochem J 379:125–131
Opassiri R, Ketudat Cairns JR, Akiyama T, Wara-Aswapati O, Svasti J, Esen A (2003)
Characterization of a rice b-glucosidase highly expressed in flower and germinating shoot.
Plant Sci 165:627–638
Opassiri R, Pomthong B, Okoksoong T, Akiyama T, Esen A, Ketudat Cairns JR (2006) Analysis
of rice glycosyl hydrolase family 1 and expression of Os4bglu12 b-glucosidase. BMC Plant
Biol 6:33
Opassiri R, Pomthong B, Akiyama T, Nakphaichit M, Onkoksoong T, Ketudat Cairns M, Ketudat
Cairns JR (2007) A stress-induced rice b-glucosidase represents a new subfamily of glycosyl
hydrolase family 5 containing a fascin-like domain. Biochem J 408:241–249
Park TH, Choi KW, Park CS, Lee SB, Kang HY, Shon KJ, Park JS, Cha J (2005) Substrate
specificity and transglycosylation catalyzed by a thermostable b-glucosidase from marine
hyperthermophile Thermotoga neapolitana. Appl Microbiol Biotechnol 69:411–422
Payne CM, Knott BC, Mayes HB, Hansson H, Himmel ME, Sandgren M, Ståhlberg J,
Beckham GT (2015) Fungal cellulases. Chem Rev 115:1308–1448
Perry JD, Morris KA, James AL, Oliver M, Gould FK (2007) Evaluation of novel chromogenic
substrates for the detection of bacterial b-glucosidase. J Appl Microbiol 102:410–415
Poulton JE (1990) Cyanogenesis in plants. Plant Physiol 94:401–405
Pozzo T, Pasten JL, Karlsson EN, Logan DT (2010) Structural and functional analyses of
beta-glucosidase 3B from Thermotoga neapolitana: a thermostable three-domain representative
of glycoside hydrolase 3. J Mol Biol 397:724–739
Qi M, Jun HS, Forsbert CW (2008) Cel9D, an atypical 1, 4-b-D-glucan glucohydrolase from
Fibrobacter succinogenes: characteristics, catalytic residues, and synergistic interactions with
other cellulases. J Bacteriol 109:1976–1984
4 The Role and Applications of b-Glucosidases in Biomass … 73

Quinlan RJ, Teter S, Xu F (2010) Towards the development of cellulases: approaches, obstacles
and outlook. In: Waldron K (ed) Bioalcohol production: biochemical conversion of
lignocellulosic biomass. Woodhead Publishing, CRC Press, Boca Raton, pp 178–204
Rani V, Mohanram S, Tiwari R, Nain L, Arora A (2014) Beta-glucosidase: key enzyme in
determining efficiency of cellulase and biomass hydrolysis. J Bioprocess Biotechnol 5:197
Reuveni M, Sagi Z, Evnor D, Hetzroni A (1999) b-Glucosidase activity is involved in scent
production in Narcissus flowers. Plant Sci 147:19–24
Sanz-Aparicio J, Hermoso JA, Martinez-Ripoll M, Lequerica JL, Polaina J (1998) Crystal structure
of beta-glucosidase A from Bacillus polymyxa: insights into the catalytic activity in family 1
glycosyl hydrolases. J Mol Biol 275:491–502
Saqib AAN, Whitney PJ (2006) Esculin gel diffusion assay (EGDA): a simple and sensitive
method for screening b-glucosidases. Enzyme Microb Technol 39:182–184
Schubot FD, Kataeva IA, Chang J, Shah AK, Ljungdahl LG, Rose JP, Wang BC (2004) Structural
basis for the exocellulase activity of the cellobiohydrolase CbhA from Clostridium
thermocellum. Biochemistry 43:1163–1170
Seidle HF, Allison SJ, George E, Huber RE (2006) Trp-49 of the family 3 beta-glucosidase from
Aspergillus niger is important for its transglucosidic activity: creation of novel
beta-glucosidases with low transglucosidic efficiencies. Arch Biochem Biophys 455:110–118
Seidle HF, Huber RE (2005) Transglucosidic reactions of the Aspergillus niger family 3
b-glucosidase: qualitative and quantitative analyses and evidence that the transglucosidic rate is
independent of pH. Arch Biochem Biophys 436:254–264
Seidle HF, McKenzie K, Marten I, Shoseyov O, Huber RE (2005) Trp-262 is a key residue for the
hydrolytic and transglucosidic reactivity of the Aspergillus niger family 3 b-glucosidase:
substitution results in enzymes with mainly transglucosidic activity. Arch Biochem Biophys
444:66–75
Seshadri S, Akiyama T, Opassiri R, Kuaprasert B, Ketudat Cairns J (2009) Structural and
enzymatic characterization of Os3BGlu6, a rice b-glucosidase hydrolyzing hydrophobic
glycosides and (1 ! 3)- and (1 ! 2)-linked disaccharides. Plant Physiol 151:47–58
Sharmila T, Sreeramulu G, Nand K (1998) Purification and characterization of b-1-4,-glucosidase
from Clostridium papyrosolvens. Biotechnol Appl Biochem 27:175–179
Sherameti I, Venus Y, Drzewiecki C, Tripathi W, Dan VM, Nitz I, Varma A, Grundler F,
Oelmuller R (2008) PYK10, a b-glucosidase located in the endoplasmatic reticulum, is crucial
for the beneficial interaction between Arabidopsis thanliana and the endophytic fungus
Piriformospora indica. Plant J 54:428–439
Shim JH, Chen HM, Rich JR, Goddard-Borger ED, Withers SG (2012) Directed evolution of a
b-glycosidase from Agrobacterium sp. to enhance its glycosynthase activity toward
C3-modified donor sugars. Protein Eng Des Sel 25:465–472
Singhania RR, Patel AK, Sukumaran RK, Larroche C, Pandey A (2013) Role and significance of
beta-glucosidases in the hydrolysis of cellulose for bioethanol production. Bioresour Technol
127:500–507
Sørensen A, Lübeck M, Lübeck PS, Ahring BK (2013) Fungal beta-glucosidases: a bottleneck in
industrial use of lignocellulosic materials. Biomolecules 3:612–631
Souza FHM, Nascimento CV, Rosa JC, Masui DC, Leone FA, Jorge JA, Furriel RPM (2010)
Purification and biochemical characterization of a mycelial glucose- and xylose-stimulated
b-glucosidase from the thermophilic fungus Humicola insolens. J Process Biochem 45:
272–278
Srivastava KK, Verma PK, Srivastava R (1999) A recombinant cellulolytic Escherichia coli:
cloning of the cellulase gene and characterization of a bifunctional cellulase. Biotechnol Lett
21:293–297
Sue M, Yamazaki K, Yajima S, Nomura T, Matsukawa T, Iwamura H, Miyamoto T (2006)
Molecular and structural characterization of hexameric beta-D-glucosidases in wheat and rye.
Plant Physiol 141:1237–1247
Suzuki H, Takahasi S, Watanabe R, Fukushima Y, Fujita N, Noguchi A, Yokoyama R,
Nishitani K, Nishino T, Nakayama T (2006) An isoflavone conjugate-hydrolyzing
74 H. Ouyang and F. Xu

b-glucosidase from the roots of soybean (Glycine max) seedlings. J Biol Chem 281:30251–
30259
Sweeney MD, Xu F (2012) Biomass converting enzymes as industrial biocatalysts for fuels and
chemicals: recent developments. Catalysts 2:244–263
Takano M, Moriyama R, Ohmiya K (1992) Structure of a b-glycosidase gene from Ruminococcus
albus and properties of the translated product. J Ferment Bioeng 73:79–88
Takashima S, Nakamura A, Hidaka M, Masaki H, Uozumi T (1999) Molecular cloning and
expression of the novel fungal b-glycosidase genes from Humicola grisea and Trichoderma
reesei. J Biochem 125:728–736
Tejirian A, Xu F (2010) Inhibition of cellulase-catalyzed lignocellulosic hydrolysis by iron and
oxidative metal ions and complexes. Appl Environ Microbiol 76:7673–7682
Tejirian A, Xu F (2011) Inhibition of enzymatic cellulolysis by phenolic compounds. Enzyme
Microb Technol 48:239–247
Teter S, Xu F, Nedwin GE, Cherry JR (2006) Enzymes for biorefineries. In: Kamm B, Gruber PR,
Kamm M (eds) Biorefineries—industrial processes and products: status quo and future
directions. Wiley-VCH, Weinheim, pp 357–384
Teugjas H, Väljamäe P (2013) Selecting b-glucosidases to support cellulases in cellulose
saccharification. Biotechnol Biofuels 6:105
Thorlby G, Fourier N, Warren G (2004) The SENSITIVE TO FREEZING2 gene, required for
freezing tolerance in Arabidopsis thaliana, encodes a beta-glucosidase. Plant Cell 16:
2192–2203
Tiwari R, Singh S, Nain PK, Rana S, Sharma A, Pranaw K, Nain L (2013) Harnessing the
hydrolytic potential of phytopathogenic fungus Phoma exigua ITCC 2049 for saccharification
of lignocellulosic biomass. Bioresour Technol 150:228–234
Tribolo S, Berrin J-G, Kroon PA, Czjzek M, Juge N (2007) The structure of human cytoplasmic
b-glucosidase unravels substrate aglycone specificity of a family 1 glycoside hydrolase. J Mol
Biol 370:964–975
Trimbur DE, Warren RAJ, Withers SG (1992) Region-directed mutagenesis of residues
surroundingthe active site nucleophile in b-glucosidase from Agrobacterium faecalis. J Biol
Chem 267:10248–10251
Tsukada T, Igarashi K, Fushinobu S, Samejima M (2008) Role of subsite +1 residues in pH
dependence and catalytic activity of the glycoside hydrolase family 1 b-glucosidase BGL1A
from the Basidiomycete Phanerochaete chrysosporium. Biotechnol Bioeng 99:1295–1302
Tsukada T, Igarashi K, Yoshida M, Samejima M (2006) Molecular cloning and characterization of
two intracellular b-glucosidases belonging to glycoside hydrolase family 1 from the
basidiomycete Phanerochaete chrysosporium. Appl Microbiol Biotechnol 73:807–814
Uchiyama T, Miyazaki K, Yaoi K (2013) Characterization of a novel b-glucosidase from a
compost microbial metagenome with strong transglycosylation activity. J Biol Chem
288:18325–18334
Vallmitjana M, Ferrer-Navarro M, Planell R, Abel M, Ausín C, Querol E, Planas A, Pérez-Pons JA
(2001) Mechanism of the family 1 beta-glucosidase from Streptomyces sp.: catalytic residues
and kinetic studies. Biochemistry 40:5975–5982
van den Brink J, de Vries RP (2011) Fungal enzyme sets for plant polysaccharide degradation.
Appl Microbiol Biotechnol 91:1477–1492
van Zyl WH, Lynd LR, den Haan R, McBride JE (2007) Consolidated bioprocessing for
bioethanol production using Saccharomyces cerevisiae. Adv Biochem Eng Biotechnol
108:205–235
Varghese JN, Hrmova M, Fincher GB (1999) Three-dimensional structure of a barley b-D-glucan
exohydrolase; a family 3 glycosyl hydrolase. Structure 7:179–190
Verdoucq L, Moriniere J, Bevan DR, Esen A, Vasella A, Henrissat B, Czjzek M (2004) Structural
determinants of substrate specificity in family 1 b-glucosidases: novel insights from the crystal
structure of sorghum dhurrinase-1, a plant b-glucosidase with strict specificity, in complex with
its natural substrate. J Biol Chem 279:31796–31803
4 The Role and Applications of b-Glucosidases in Biomass … 75

Vroemen S, Heldens J, Boyd C, Henrissat B, Keen NT (1995) Cloning and characterization of the
bgxa genefrom Erwinia chrysanthemi D1 that encodes a b-glycosidase/xylosidase enzyme.
Mol Gen Genet 246:465–477
Wang Q, Graham RW, Trimbur D, Warren RAJ, Withers SG (1994) Changing enzymic reaction
mechanisms by mutagenesis: conversion of a retaining glucosidase to an inverting enzyme.
J Am Chem Soc 116:11594–11595
Wang Q, Trimbur D, Graham R, Warren RA, Withers SG (1995) Identification of the acid/base
catalyst in Agrobacterium faecalis beta-glucosidase by kinetic analysis of mutants.
Biochemistry 34:14554–14562
Wilson CA, Wood TM (1992) The anaerobic fungus Neocallimastix frontalis: isolation and
properties of a cellulosome-type enzyme fraction with the capacity to solubilize
hydrogen-bond-ordered cellulose. Appl Microbiol Biotechnol 37:125–129
Withers SG, Street IP, Bird P, Dolphin DH (1987) 2-Deoxy-2-fluoroglucosides: a novel class of
mechanism based inhibitors. J Am Chem Soc 109:7530–7531
Withers SG, Street IP (1988) Identification of a covalent a-D-glucopyranosyl enzyme intermediate
formed on a b-glucosidase. J Am Chem Soc 110:8551–8553
Withers SG, Warren RAJ, Street IP, Rupitz K, Kempton JB, Aebersold R (1990) Unequivocal
demonstration of the involvement of a glutamate residue as a nucleophile in the mechanism of
a retaining glycosidase. J Am Chem Soc 112:5887–5889
Wulff-Strobel CR, Wilson DB (1995) Cloning, sequencing, and characterization of a membrane
associated Prevotella ruminicola B1 4-b-glycosidase with cellodextrinase and cyanoglycosi-
dase activities. J Bacteriol 177:5884–5890
Xie S, Syrenne R, Sun S, Yuan JS (2014) Exploration of natural biomass utilization systems
(NBUS) for advanced biofuel—from systems biology to synthetic design. Curr Opin
Biotechnol 27:195–203
Xu F (2010a) Enzymatic degradation of lignocellulosic biomass. In: Tao A, Kazlauskas R
(eds) Biocatalysis for green chemistry and chemical process development. Wiley, Hoboken,
pp 361–390
Xu F (2010) Biomass-converting enzymes and their bioenergy applications. In: Baltz RH,
Demain AL, Davies JE (ed-in-chief) Bull AT, Junker B, Katz L, Lynd LR, Masurekar P,
Reeves CD, Zhao H (eds) The manual of industrial microbiology and biotechnology, 3rd edn.
American Society for Microbiology Press, Washington, DC, pp 495–508
Xu F (2004) Enhancing biomass conversion to fermentable sugars: a progress report of a joint
government-industrial project. In: Ohmiya K, Sakka K, Karita S, Kimura T, Sakka M,
Onishi Y (eds) Biotechnology of lignocellulose degradation and biomass utilization. Uni
Publishers, Tokyo, pp 793–804
Xu F, Ding H, Osborn D, Tejirian A, Brown K, Albano W, Sheehy N, Langston J (2007) Partition
of enzymes between the solvent and insoluble substrate during the hydrolysis of lignocellulose
by cellulases. J Mol Catal B Enz 51:42–48
Xu Z, Escamilla-Trevino LL, Zeng L, Lalgondar M, Bevan DR, Winkel BSJ, Mohamed A,
Cheng C, Shih M, Poulton JE, Esen A (2004) Functional genomic analysis of Arabidopsis
thaliana glycoside hydrolase family 1. Plant Mol Biol 55:343–367
Yamada R, Hasunuma T, Kondo A (2013) Endowing non-cellulolytic microorganisms with
cellulolytic activity aiming for consolidated bioprocessing. Biotechnol Adv 31:754–763
Zouhar J, Vevodova J, Marek J, Damborsky J, Su X-D, Bryzobohaty B (2001) Insights into the
functional architecture of the catalytic center of a maize b-glucosidase Zm-p60.1. Plant Physiol
127:973–985
Part II
Hemicellulases
Chapter 5
Role and Applications of Feruloyl
Esterases in Biomass Bioconversion

Constantinos Katsimpouras, Io Antonopoulou, Paul Christakopoulos


and Evangelos Topakas

Abstract Ferulic acid esterases (FAEs) act synergistically with xylanases to


hydrolyze the feruloylated decorations of the hemicellulosic fraction of cell wall
material and therefore play a major role in the degradation of plant biomass. In this
review, their role in plant biomass degradation, their production, classification, and
structural determination are discussed. In addition, the production, physicochemical
properties, and molecular biology of the different type of FAEs are presented,
giving emphasis in their potential applications utilizing their hydrolytic and syn-
thetic activity. A detailed map of the reaction systems used to date is demonstrated,
underpinning the potential of these enzymes as biosynthetic tools in the synthesis of
bioactive compounds for use in food and cosmeceutical industries.

5.1 Introduction

The combustion of fossil fuels constitutes a great percentage of our current energy
supply, and with depleting crude oil reserves and climate change due to the
greenhouse gas (GHG) emissions, it is imperative that we focus on more renewable
energy sources such as biofuels (Damásio et al. 2013). Biofuels from lignocellu-
losic biomass, also referred to as second-generation biofuels, are considered to be
the key due to their potential to mitigate GHG emissions compared with conven-
tional fuels, reduce oil dependency, and eradicate public concerns about a trade-off
between foods and fuels.

C. Katsimpouras  E. Topakas (&)


Biotechnology Laboratory, School of Chemical Engineering,
National Technical University of Athens, 15780 Athens, Greece
e-mail: [email protected]; [email protected]
I. Antonopoulou  P. Christakopoulos
Biochemical and Chemical Process Engineering, Division of Sustainable Process
Engineering, Department of Civil, Environmental and Natural Resources Engineering,
Luleå University of Technology, SE-97187 Lulea, Sweden

© Springer International Publishing Switzerland 2016 79


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_5
80 C. Katsimpouras et al.

Production of fuel ethanol from renewable lignocellulosic materials, such as agri-


cultural residues, forest residues, and energy crops has been extensively studied in the
last few decades. Lignocellulosic raw materials, being low-cost and abundant as
residual biomass from agricultural and agroindustrial waste, provide an attractive
feedstock for the production of ethanol and other value-added products under the
biorefinery concept without competing with food and feed industry (Larsen et al. 2012).
Through enzymatic hydrolysis processes, commonly known as saccharification, the
main components of lignocellulosic biomass can be converted to sugars and subse-
quently fermented to bioethanol. However, the plant cell wall, developed in the span of
million of years of co-evolution as the plants’ defense system against pathogens,
bottlenecks the aforementioned processes and several improvements should be made in
order to produce cellulosic ethanol at competitive costs (Malinovsky et al. 2014).
A possible strategy that is being suggested requires the usage of hemicellulose-
degrading enzymes in order to generate more efficient enzyme cocktails for the sac-
charification of lignocellulosic biomass and reduce catalysts costs. In addition, the
cooperative action of enzymes could also lead to less severe, and thus more sustainable
and environmentally friendly, pretreatment processes (Couturier et al. 2011).
A set of enzymes that is considered to be a biotechnological key in cell wall
hydrolysis and in the extraction of phenolic acids is ferulic acid esterases (FAEs) or
feruloyl esterases, as they are also known. FAEs (E.C. 3.1.1.73) represent a sub-
class of the carboxylic ester hydrolases, which liberate phenolic acids such as
ferulic acid (FA) or p-coumaric acid and their dimers from naturally occurring
hemicelluloses (Fig. 5.1) and pectins, where they mainly occur as esters with
L-arabinofuranose-containing polysaccharides, such as L-arabino-D-xylans and

Fig. 5.1 Action of FAEs for the hydrolysis of ester bonds in plant cell wall arabinoxylan, as
indicated by black arrows. However, most of the characterized FAEs are acting on soluble
feruloylated fractions created by the action of endo-β-1,4-xylanases and not directly on the sugar
polymer showing strong synergistic interaction
5 Role and Applications of Feruloyl Esterases … 81

L-arabinans (Topakas et al. 2007). Besides FAEs’ role in hydrolytic reactions, they
are also applicable for the synthesis of bioactive compounds with antioxidant and/or
antimicrobial activity, and other compounds of high industrial interest. The present
review is an attempt to elucidate the role and applications of FAEs in lignocellu-
losic biomass degradation and as biosynthetic tools for the modification of plant
derived antioxidants.

5.2 Plant Cell Wall Functionality

Plant cell walls, besides their role as structural support to the plant body, also
restrict enzyme access rendering this complex polysaccharide network recalcitrant
to biochemical degradation. Cellulose, hemicellulose, lignin, and pectin are the
main components of lignocellulosic biomass along with proteins and aromatic
compounds at proportions that differ among plant species (de Oliveira et al. 2014).
In plants, two major types of cell walls could be distinguished: the primary cell
walls, which are composed of cellulose, hemicelluloses, pectins, and proteins, and
the secondary cell walls, consisting of cellulose, hemicelluloses, and lignin that are
formed after the end of cell expansion (Endler and Persson 2011). Cellulose is a
linear natural biopolymer consisting of glucose units linked by β-1,4-glycosidic
bonds forming crystalline microfibrils via hydrogen bonding and van der Waals
interactions (Horn et al. 2012).
After cellulose, hemicelluloses are the second most abundant polysaccharides on
earth with xylans to be their main representative. Hemicelluloses are a group of
heterogeneous polysaccharides consisting of a β-1,4-linked backbone of pentoses,
hexoses, and sugar acids, which is usually decorated with side branches (Saha
2003). The exact structure and abundance of hemicelluloses diversify widely
among different plant species and cell types with xyloglucans occurring mostly in
the primary walls of dicots and conifers, while arabinoxylans dominate in com-
melinid monocots (Scheller and Ulvskov 2010). Through interactions with cellu-
lose, and sometimes lignin, hemicelluloses have the ability to strengthen the cell
walls (Endler and Persson 2011).
Pectins are a group of heterogeneous and branched polysaccharides that can be
found only in the primary cell wall and consist of a backbone of α-1,4-linked D-
galacturonic acid residues that can be methyl-esterified or decorated with acetyl
groups. Homogalacturonans, xylogalacturonan, and rhamnogalacturonans (I & II)
are the main groups in which pectins could be classified according to their basic
structure (Wong 2008). There is also increasing evidence that pectin interacts
covalently with hemicelluloses providing structural and functional complexity to
the plant cell wall (Caffal and Mohnen 2009).
Cell walls are also fortified by the deposition of the phenylpropanoid polymer
lignin, which is one of the main contributors in recalcitrance of cell walls to enzy-
matic saccharification (Chen and Dixon 2007). The term lignin is employed to
describe a group of aromatic and nonsoluble heteropolymers consisting mainly of
82 C. Katsimpouras et al.

phenylpropanoid units derived from the oxidative polymerization of three hydrox-


ycinnamyl alcohols (coniferyl alcohol, sinapyl alcohol, and p-coumaryl alcohol)
derivatives (Vanholme et al. 2010; Vogt 2010). The aforementioned hydroxycin-
namyl alcohols are incorporated into the lignin polymer in the form of guaiacyl (G),
syringyl (S), and p-hydroxyphenyl (H) units. In gymnosperms G-units are mainly
present with small amounts of H-units, while lignin in angiosperm dicots consists of
G- and S-units. In grasses and softwoods H-units amounts are higher (Vanholme
et al. 2010). In lignocellulosic biomass, lignin is cross-linked with cell wall polymers
by ether or ester bonds with FA being the predominant connector as the FA that is
etherified to lignin is also esterified to polysaccharides (Iiyama et al. 1990).
Hydroxycinnamic acids such as FA, sinapic acid, and p-coumaric acid are com-
ponents of plant cell wall with the ability to esterify hemicelluloses due to a carboxylic
group at the end of their propenyl group. FA plays an important role in the plant cell
wall by cross-linking with polysaccharides by ester bonds and with lignin by ether
bonds regulating this way the cell wall extensibility and inhibiting pathogen invasion
(de Oliveira et al. 2014). Implication of FA and diferulates to function as nucleation
sites for the lignification process has also been described (Ralph et al. 1995).
FA is synthesized in the phenylpropanoid pathway from phenylalanine and in
nature exists mainly in E (trans) form since isomerization in Z (cis) form occurs
during extraction due to the light sensitivity of hydroxycinnamates (Faulds and
Williamson 1999). In monocotyledons such as grasses and cereals, the arabinofu-
ranoses are esterified by hydroxycinnamates with trans-FA, (E)-4-hydroxy-3-
methoxycinnamic acid to be the most abundant whereas dicots contain ferulated
pectic polysaccharides (Mueller-Harvey et al. 1986). FA is esterified at position O-5
to α-L-arabinofuranosyl side chains in arabinoxylans (Mueller-Harvey et al. 1986),
at position O-6 to β-D-galactopyranosyl residues in pectic rhamno-galacturonanans
(Colquhoun et al. 1994) and at position O-4 to a-D-xylopyranosyl residues in
xyloglucans (Ishii et al. 1990). In some dicots such as spinach (Fry 1983) and sugar
beet pulp (Colquhoun et al. 1994), feruloylation occurs at position O-2 to α-L-
arabinofuranosyl residues in arabinans and at position O-6 to β-D-galactopyranosyl
residues in pectic rhamno-galacturonanans. FA amount varies in the cell walls of
different plant materials ranging from 0.1 to 3.8 % (w/w, dry weight). In wheat
bran, FA (w/w, dry weight) account for 0.66 %, 3.1 % in maize bran, 1.24 % in
wheat straw, 0.9 % in rice endosperm cells, 0.14 % in barley grains, 0.32 % in
barley spent grain, 1.4 % in barley hulls, 2.2–3.8 % in oat hulls, and 0.87 % in
sugar beet pulp (Smith and Hartley 1983; Saulnier et al. 1995; Benoit et al. 2006;
Shibuya 1984; Nordkvisk et al. 1984; Bartolome et al. 1997a, b; Tenkanen et al.
1991; Garleb et al. 1988; Yu et al. 2002a, b; Kroon and Williamson 1996).
Covalent cross-linking between feruloylated polysaccharide chains or between
hemicellulose and lignin can be achieved through the formation of ferulate dimers.
Dimers of FA can be formed by radical coupling reactions or by photochemical
coupling reactions (Ralph et al. 1994; Ishii 1997). Photochemical [2+2]-cyclodi-
merization (Ford and Hartley 1990) produces cyclodimers, while peroxidase-
catalyzed coupling produces several forms of dehydrodimers such as 8-5-, 8-O-4-,
5-5-, 8-8-, and 4-O-5-coupled dehydrodimers (Bunzel et al. 2005) commonly known
5 Role and Applications of Feruloyl Esterases … 83

as diferulates. In pectins isolated from sugar beet pulp, 8.8 % of the ferulates were
mostly 8-8- and 8-O-4-coupled dehydrodimers (Oosterveld et al. 1997).
Fry et al. (2000) suggested that trimers or even larger products contribute highly
to cross-linking between polysaccharides in culture maize cells. The first FA
dehydrotrimer was isolated from maize bran insoluble fiber and found to be a 5-5/8-
O-4-coupled dehydrotrimer (Bunzel et al. 2003). Since then, more dehydrotrimers
and dehydrotetramers have been identified and characterized (Rouaou et al. 2003;
Bunzel et al. 2005, 2006; Funk et al. 2005; Barron et al. 2007; Hemery et al. 2009).

5.3 Role of FAEs in Plant Biomass Degradation and Their


Synergistic Action

The complex and extended polysaccharide networks in plant cell walls, due to the
covalent cross-linking mediated by FA, raise hurdles in utilizing lignocellulosic
biomass for biofuel production, as it contributes to biomass recalcitrance to enzy-
matic degradation (Wong et al. 2013). In order to surmount these hurdles, along
with cellulases and hemicellulases, accessory enzymes such as FAEs should be
employed, enhancing this way the fermentable sugars yield from lignocellulosic
biomass (Faulds 2010). FAEs action depends highly on the type of xylanases that is
being used with, having an effect not only on the amount but also in the form of FA
released, with family 11 xylanases being more efficient in the hydrolysis of FA
whereas family 10 xylanases present a more synergistic effect on the release of
diferulic acid (Faulds et al. 2006). The synergism of a recombinant FAE isolated
from a rumen microbial metagenome (RuFae2), in association with GH10 and
GH11 endoxylanases was investigated by Wong et al. (2013). The results obtained
from the release of FA from several natural substrates such as corn fiber, corn bran,
wheat bran, wheat-insoluble arabinoxylan, and switchgrass indicated that the GH10
xylanase showed a greater synergistic effect than that of the GH11 xylanase (Wong
et al. 2013). The synergistic action between xylanases and FAEs on the release of
FA from feruloylated polysaccharides seems to render the biomass more vulnerable
to glycoside hydrolase enzymes (Yu et al. 2003).
Tabka et al. (2006) reported a synergistic effect of a recombinant FAE from
Aspergillus niger when combined with cellulases and xylanases from Trichoderma
reesei in the hydrolysis of wheat straw. Significant increase in the release of reducing
sugars (34.8 % increase in total reducing sugars) from oat hulls was also a result of
synergistic interaction between A. niger FAE and T. reesei xylanase (Yu et al. 2003).
The enzymatic saccharification of pretreated corn stover by a cellobiohydrolase from
T. reesei was significantly enhanced by the addition of small quantities of an
endoxylanase, a FAE, and an acetyl xylan esterase. FAE addition to cellobiohydrolase
achieved a 37 % synergistic improvement in glucan conversion to cellobiose, while a
substantial increase of 85 % was shown when all enzymes were added (Selig et al.
2008). Supplementation of a commercial mixture of cellulase and β-glucosidase with
pectinase and FAE preparations resulted in higher arabinose and xylose yields from
84 C. Katsimpouras et al.

pretreated dried distiller’s grains with solubles, a co-product of corn ethanol pro-
duction (Dien et al. 2008). Kim et al. (2008) reported a 15 % increase in glucose yields
and 2–4 times enhancement for xylose yields in the enzymatic hydrolysis of pretreated
distiller’s grain when cellulases were supplemented with xylanase and FAE activities.
Two recombinant FAEs from Penicillium funiculosum expressed in Aspergillus
awamori (FaeA and FaeB) were able to enhance the cellulolytic activity of purified T.
reesei on pretreated corn stover by releasing 19 and 7 % more cellobiose, respectively
(Knoshaug et al. 2008). The synergistic action among cellulases, xylanases,
β-glucosidases, and FAEs produced by T. reesei and A. awamori was investigated for
the hydrolysis of steam-pretreated sugarcane bagasse, as reported by Gottschalk et al.
(2010). Addition of FAE and xylanase activities, produced by Aspergillus oryzae, in a
commercial enzyme preparation resulted in increased bioconversion of sugarcane
bagasse by 36 % (Braga et al. 2014).
Enzymatic release of FA from agroindustrial waste materials with the use of
FAEs raises considerable interest due to its antioxidant and anti-inflammatory
properties. FA is considered to alleviate oxidative stress in neurodegenerative
disorders such as Alzheimer’s disease (Sultana et al. 2005). Furthermore, FA could
be employed as a source for the bioconversion into value-added products such as
vanillin, which is one of the most used flavoring agents in the food, pharmaceutical,
and cosmetic industries (Priefert et al. 2001).
Two FAEs from Streptomyces sp. were tested on their ability to release FA from
corn bran, wheat bran, and defatted rice bran in combination with xylanase and α-L-
arabinofuranosidase activities from Streptomyces with the results suggesting that the
amount of FA released from biomass increased due to a synergistic effect between
these enzymes (Uraji et al. 2014). A recombinant FAE from Aspergillus usamii in
combination with a recombinant GH 11 xylanase from the same microorganism
exhibited a 27 % increase in FA release from destarched wheat bran compared to that
of the FAE alone (Gong et al. 2013). The amount of FA released from steam exploded
corn stalk, when a FAE from Aspergillus flavus (AfFaeA) combined with a GH 10
xylanase from Geobacillus stearothermophilus, was 13-fold higher than that released
by AfFaeA alone (Zhang et al. 2013). Wu et al. (2012) reported the characterization of
two FAEs of myxobacterial origin (Sorangium cellulosum) capable of releasing FA
from grass biomass, such as triticale bran, and from wheat bran yielding up to 85 % of
total alkali-extractable FA in presence of a Trichoderma viride xylanase.
A recombinant FAE (MtFae1a) from the thermophilic fungus Myceliophthora ther-
mophila (synomym Sporotrichum thermophile) was able to release up to 41 % of the
total alkali-extractable FA from wheat bran only with the presence of a xylanase,
indicating this way the synergistic interaction between MtFae1a and M3 Trichoderma
longibrachiatum xylanase (Topakas et al. 2012). Synergistic interaction between a
FAE (Tx-Est1) and a GH11 xylanase (Tx-Xyl11) both from Thermobacillus
xylanilyticus was also reported by Rakotoarivonina et al. (2011), where esterase’s
efficiency in releasing FA and diferulic acid from destarched wheat bran was seven-
to eightfold higher than that of Tx-Est1 alone. A maximum of 67 % of total FA was
released from destarched wheat bran by the combined action of a type C FAE from
Fusarium oxysporum and a M3 T. longibrachiatum xylanase (Moukouli et al. 2008).
5 Role and Applications of Feruloyl Esterases … 85

5.4 FAE Classification and Structural Determination

FAEs appear to be a very diverse set of enzymes, with little unifying sequence and
physical characteristics to link them. In order to categorize and classify these car-
bohydrate esterases into different types sharing the same substrate specificity and/or
the same structure, several attempts have been made starting in 2004. Under this
scope, the use of multiple alignments of sequences or domains demonstrating FAE
activity, as well as related sequences, helped to construct a neighborhood-joining
phylogenic tree (Crepin et al. 2004a). The outcome of this genetic comparison
supported substrate specificity data and allowed FAEs to be subclassified into 4
types: A, B, C, and D, a classification that is still used in literature by many
researchers. Thus, there does appear to be an evolutionary relationship between
FAEs, acetyl xylan esterases, tannases and certain lipases. The substrate specificity
was based on their specificity toward mono- and diferulates, on substitutions on the
phenolic ring, and on their amino acid sequence identity (Crepin et al. 2004a). An
unofficial nomenclature for describing FAEs was followed based on this classifi-
cation, following both the source of the enzyme and the type of the esterases (e.g.,
the type-A FAE produced by F. oxysporum is termed FoFaeA).
It is extremely common for esterases to act on a broad range of substrates.
Type A FAEs exhibit relatively high sequence identity with fungal lipases, however
lipase activity is not detected. For example, the Type A FAE from A. niger
(AnFaeA; Faulds et al. 1997) shows homology with lipases from Thermomyces
lanuginosus TLL (30 % sequence identity) and Rhizomucor miehei (37 % sequence
identity). This subclass show preference for the phenolic moiety of the substrate
containing methoxy substitutions, especially at meta- position(s), as occurs in fer-
ulic and sinapic acids, while type B FAEs shows complementary activity, showing
preference to substrates containing one or two hydroxyl substitutions as found in p-
coumaric or caffeic acid. Type B FAEs, such as M. thermophila MtFae1a (Topakas
et al. 2012), P. funiculosum PfFaeB (Kroon et al. 2000) and Neurospora crassa
Fae-1 (Crepin et al. 2003a), show high sequence identity with acetyl xylan esterases
and are the only type of FAEs that are members of carbohydrate esterase
(CE) family 1 of the Carbohydrate Active enZymes (CAZy) database (http://www.
cazy.org/CE1.html; Lombard et al. 2013). In contrast to type B esterases, type A
FAEs appear to prefer hydrophobic substrates with bulky substituents on the
benzene ring (Kroon et al. 1997; Topakas et al. 2005a). One of the famous Τype of
FAEs, due to their synthetic potential, is type C that exhibit high sequence identity
with fungal tannases. Together with type D FAEs, these esterases exhibit broad
specificity against synthetic hydroxycinnamic acids (ferulic, p-coumaric, caffeic and
sinapic acid) showing difference only in the ability to release 5-5′ diFA (Crepin
et al. 2004a, b; Vafiadi et al. 2006b). Lastly, Type D FAEs have been found only in
bacteria, such as XYLD from Pseudomonas fluorescens (reclassified as Cellvibrio
japonicus) (Ferreira et al. 1993) that show sequence similarity with xylanases
(Crepin et al. 2004a). These four different types of FAEs have different evolutionary
origin, as found by phylogenetic analysis resulting in five major clades I to V
86 C. Katsimpouras et al.

(Olivares-Hernández et al. 2010). Clade I contains Type A FAEs, such as the


characterized FAEs from A. niger, A. awamori and A. tubingensis that belong to
Eurotiomycetes, except one FAE sequence from Laccaria bicolor belonging to
Agaricomycetes class of basidiomycetes. Clade II contains Type B FAE sequences
from both Sordariomycetes and Eurotiomycetes; however, the clade does not reflect
any phylogenetic structure. Clade IV contains characterized Type B and C FAEs
including also putative FAE sequences from four taxonomic classes, with its basis
consisted of esterases from Magnaporthe, Pyrenophora, Phaeosphaeria, and
Fusarium.
Recently, a modification to this classification has been proposed by Benoit et al.
(2008), while a descriptor-based computational analysis with pharmacophore
modeling provided a different approach for the classification of FAEs (Udatha et al.
2011). According to them, FAEs could be classified into seven subfamilies
depending on the phylogenetic analysis of fungal genomes, whereas the
descriptor-based classification and structural analysis of experimentally verified and
putative FAEs proposed 12 families. FAEs are also members to ESTHER database,
a comprehensive and continuously updated database devoted to α/β hydrolase fold
proteins that hierarchically classifies more than 30,000 proteins by blocks and Rank
families (Lenfant et al. 2013). In this database, FAEs are members of five Rank 1
families, four in Block X (Abhydrolase_6, Antigen85c, Esterase_phb, Tannase) and
one in Block L (Lipase_3).
Relatively few studies have been performed to elucidate the functional rela-
tionships between sequence and diverse FAEs. To date, the crystal structures of
FAEs include a Type A or Lipase_3 (ESTHER) AnFaeA from A. niger (Hermoso
et al. 2004; related PDB entries: 1UWC, 1USW, 1UZA, 2BJH, 2IX9, 2HL6, 2IX9,
2HL6), a FAE (Est1E) from Butyrivibrio proteoclasticus (PDB_ID: 2WTM, 2WTN;
Goldstone et al. 2010) that belongs to Abhydrolase_6 family (ESTHER) and FAE
domains, XynY and XynZ (Prates et al. 2001; Schubot et al. 2001 with related PDB
entries: 1JJF, 1GKK, 1GKL, 1JT2, 1WB4, 1WB5, 1WB6) of the cellulosomal
enzymes included in the cellulosome complex from Clostridium thermocellum that
belongs to Antigen85c family (ESTHER). Recently, a cinnamoyl esterase (LJ0536)
from Lactobacillus johnsonii was characterized and its 3D structure was determined
that resembles the structure of the Est1E from B. proteoclasticus (PDB_IDs: 3PF8,
3PF9, 3PFB, 3PFC, 3QM1, 3S2Z; Lai et al. 2011). Unfortunately, none of the
proposed FAE classifications include and categorize LJ0536 FAE or any of its
homologs. In 2014, the first crystal structure of a type B FAE was solved, belonging
to the tannase family of the ESTHER database from A. oryzae (AoFaeB) with the
serine and histidine residues of the catalytic triad, a typical motif of serine hydro-
lases, to be directly connected by a disulfide bond of the neighboring cysteine
residues (Suzuki et al. 2014; PDB_ID: 3WMT). As reported also in the first attempt
of FAE classification by Crepin et al. (2004a), Type C FAEs show homology with
members of tannase family showing broad substrate specificity against four methyl
esters of hydroxycinnamic acids. However, only one member of Type C FAE from
Talaromyces stipitatus (TsFaeC; Vafiadi et al. 2006b) has a substrate specificity
profile that corresponds to Type C FAEs, while the rest of them show a specificity
5 Role and Applications of Feruloyl Esterases … 87

profile of Type B FAEs with weak or no activity against methyl sinapate. Such
discrepancy is well documented in literature with typical examples of Type C FAEs
with B type mode of action, such as AnFaeB (Kroon and Williamson 1996), AoFaeΒ
(Suzuki et al. 2014) and FoFaeC (Moukouli et al. 2008) from Aspergillus and
Fusarium species. Based on sequence homology, the structure determination of
AoFaeB esterase underpins the elucidation of the first structure of Type C FAEs.
These Type B and C FAEs are also members of the tannase family of ESTHER
database, where most of them do not exhibit tannase activity, however, Tan410
discovered from a soil metagenomic library exhibits both FAE and tannase activities
(Yao et al. 2013). The different Types of fungal and bacterial FAEs that have been
structural determined to date, is shown in Fig. 5.2.
The aforementioned FAEs have a common α/β-hydrolase fold that is
well-known in literature for the diverse activities of the enzymes that belong to this
structural superfamily, such as hydrolases, lipases, dehalogenases and peroxidases
(Holmquist 2000). In addition, these enzymes also share a conserved catalytic triad
(Ser–His–Asp) composed of a nucleophile (Ser, Cys or Asp), a conserved histidine

Fig. 5.2 The structures of fungal FAEs, a AnFaeA from A. niger (Hermoso et al. 2004), b AoFaeB
from A. oryzae (Suzuki et al. 2014) and bacterial FAEs c EstE1 from B. proteoclasticus (Goldstone
et al. 2010), d LJ0536 from L. johnsonii (Lai et al. 2011), domains, e XynY (Prates et al. 2001) and
f XynZ (Schubot et al. 2001) of C. thermocellum cellulosome complex. The catalytic α/β hydrolase
domain (green) and the lid domain (magenta) are shown. GlcNAc residues of N-glucans and the
catalytic triad are shown as cyan and yellow sticks, respectively. A calcium ion is represented as an
orange sphere. The 3D structures were prepared using PyMOL (http://pymol.org)
88 C. Katsimpouras et al.

and an acidic residue. These residues are sequenced in the order of nucleophile,
acidic and histidine residues, with the nucleophile to be located between the fifth
β-strand and the following helix of the nucleophile elbow (Nardini and Dijkstra
1999). The tetrahedral oxyanion intermediate formed during catalysis is stabilized
by the oxyanion hole that is formed by the contribution of the nucleophilic elbow.
The catalytic triad of the Type A FAE AnFaeA and the active-site cavity is confined
by a lid and a loop that confers plasticity to the substrate binding site in analogy
with lipases (Fig. 5.2). What is surprising is the fact that FAE’s lid exhibits a high
ratio of polar residues keeping it in an open conformation that gives the typical
preference of catalyzing the hydrolysis of hydrophilic substrates compared
to lipases. In addition, the open conformation is further stabilized by a
N-glycosylation site (Hermoso et al. 2004). These subtle aminoacid and confor-
mational changes are a result of an evolutionary divergence within the FAE family,
as further discussed by Levasseur et al. (2006). A functional shift followed by a
duplication event was suggested, which was occurred within the ancestral lipase
genes. Such a “neofunctionalization” was a result of positive selection during the
functional shift, possibly due to the drastic environmental changes happened by the
colonization of land by terrestrial plants, giving the selective advantage to
Euascomycetes (Aspergilli), as hypothesized by the authors (Levasseur et al. 2006).
The Type A FAE from A. niger together with Est1E from B. proteoclasticus exhibit
a small lid domain (16 and 46 amino acid residues, respectively) compared with the
AoFaeB (159 amino acid residues), as shown by the first solved FAE structure of
tannase family (Suzuki et al. 2014). In addition, the lid domain of BpEst1E has a
unique fold forming a flexible β-sheet structure around a small hydrophobic core,
while on the other hand, FAE domains of C. thermocellum do not possess a lid
domain (Fig. 5.2). This newly discovered lid underpins the continuing diversity of
insertions that decorate the common α/β fold of hydrolases that point toward a
dynamic mechanism for binding and release of substrates (Goldstone et al. 2010).

5.5 Production and Physicochemical Characteristics

Since 1987, when FAE activity was detected in Streptomyces olivochromogenes


cultures grown on wheat bran for the first time (MacKenzie et al. 1987), and the first
purified FAE from the same microorganism was reported (Faulds and Williamson
1991), many FAEs have been produced and characterized from fungi and bacteria,
and are considered to be a common component of hemicellulose preparations. In
the last decade, over 50 FAEs have been produced through various fermentation
processes employing different lignocellulose derived carbon sources such as maize
bran, corn bran, and destarched wheat bran but with the most interest focused on
production through heterologous expression (Table 5.1). An extended table on the
microbial production of FAEs is provided by Topakas et al. (2007) that complement
the present investigation.
Table 5.1 Microbial production of FAEs both from WT and recombinant sources
Organism Carbon source/ Cultivation conditions Assay FAE activity References
inducer (mU/mL) (mU/mg)
A. mucronatus YE505 1 mM IPTG Standard protocol MFA/EFA – 9940/7750 Qi et al. (2011)
(E. coli BL21)
A. awamori G-2 1 % WB 30 °C, 72 h 1-Naphthyl acetate 394,000 – Kanauchi et al. (2008)
A. awamori (E. coli BL21) 1 mM IPTG Standard protocol EFA – 1850 Fazary et al. (2010)
A. awamori 2B.361 U2/1 3 % WB V = 300 mL, 30 °C, EFA 170 – Gottschalk et al. (2013)
200 rpm, 7 days
A. clavatus NRRL1 0.5 mM IPTG Standard protocol MFA/MpCA/MCA – – Damásio et al. (2013)
(E. coli BL21)
A. flavipes 1 % WB 28 °C, 6 days MFA 6820 10,570 Mathew et al. (2005)
1 % MB 28 °C, 5 days MFA 33,180 6980 Mathew et al. (2005)
A. flavus CBE332.1 Methanol V = 250 mL, 30 °C, CNPF 500 – Zhang et al. (2013)
(P. pastoris X-33) 2 days
5 Role and Applications of Feruloyl Esterases …

A. nidulans (S. cerevisiae) 2 % (w/v) V = 500 mL, 30 °C, 20 h, MFA – 14,900 Shin and Chen (2007)
Galactose 250 rpm
A. nidulans (P. pastoris) Methanol 30 °C, 3 days MFA – 21,700 Debeire et al. (2012),
Bauer et al. (2006)
A. niger NRRL3 1 % CB V = 100 mL, 30 °C, MFA/CB 13.9/0 – Shin and Chen 2006
5 days
A. niger CFR 1105 WB 30 °C, 96 h/V = 100 mL, 4NPF – 12,800/11700a Hegde and Muralikrishna
30 °C, 105 rpma (2009)
A. niger (S. cerevisae 2 % Glycerol V = 1 L, 30 °C 4NPF – 8200 Wong et al. (2011)
DY150)
A. oryzae RIB40 Koseki et al. (2009)
(P. pastoris GS115)
(continued)
89
Table 5.1 (continued)
90

Organism Carbon source/ Cultivation conditions Assay FAE activity References


inducer (mU/mL) (mU/mg)
AoFaeB Methanol Manufacturer’s protocol MCA/MpCA – 3200/30,600
AoFaeC Methanol Manufacturer’s protocol MCA/MpCA – 34,900/14,300
A. oryzae 0.5 % (v/v) Multicopy Pichia MFA – 58,400 Zeng et al. (2014)
(P. pastoris GS115) Methanol Expression kit manual
A. tubingensis 1 % DSWB, 35 °C, 3 days DSWB 2440 – Tai et al. (2014)
1 % Pectin
A. usamii E001 1 % (v/v) 30 °C, 3 days MFA – 4490 Gong et al. (2013)
(P. pastoris GS115) Methanol
B. proteoclasticus IPTG Standard protocol EFA – 37,000 Goldstone et al. (2010)
(E. coli BL21)
C. ruminicola H1 Li et al. (2011)
FaeI (E. coli BL21) 1 mM IPTG Standard protocol MFA – 3780
FaeII (E. coli 1 mM IPTG Standard protocol MFA – 38,760
Rosetta-gami2)
FaeIII (E. coli Rosetta) 1 mM IPTG Standard protocol MFA – 56,880
C. japonicus IPTG Standard protocol MFA – 28 McClendon et al. (2011)
(E. coli BL21)
Chaetomium sp. CQ31 4 % CC V = 250 mL, 40 °C, MFA 2100 – Yang et al. (2013)
200 rpm, 6 days
Cotton soil metagenomic 0.5 mM IPTG V = 50 mL, 37 °C, pNFA – – Yao et al. (2013)
library (E. coli BL21)
D. dadantii 3937 Hassan and
(E. coli BL21) Hugouvieux-Cotte-Pattat
(2011)
(continued)
C. Katsimpouras et al.
Table 5.1 (continued)
Organism Carbon source/ Cultivation conditions Assay FAE activity References
inducer (mU/mL) (mU/mg)
FaeD 2 mM IPTG Standard protocol pNPA –
FaeT 2 mM IPTG Standard protocol pNPA –
F. oxysporumA 0.5 % (v/v) V = 150 mL, 30 °C, DSWB – – Moukouli et al. (2008)
(P. pastoris X-33) Methanol 6 days
Fusarium proliferatum 1 % CB V = 100 mL, 30 °C, MFA 33.46 – Shin and Chen 2006
NRRL 26517 5 days
Fusarium verticillioides 1 % CB V = 100 mL, 30 °C, MFA/CB 19.6/0 – Shin and Chen 2006
NRRL 26518 5 days
Humicola grisea var. 2 % BSG/2 % V = 100 mL, 45 °C, MFA 470/330b – Mandalari et al. (2008)
thermoidea WB 10 days
JSC-3 1 % WB 28 °C, 9 days MFA 6320 10,090 Mathew et al. (2005)
1 % MB 28 °C, 8 days MFA 7170 36,710 Mathew et al. (2005)
5 Role and Applications of Feruloyl Esterases …

L. johnsonii 1 mM IPTG Standard protocol Esters – – Kin et al. (2009)


(E. coli BL21)
L. plantarum WCFS1 4 mM IPTG Standard protocol Esteban-Torres et al.
(E. coli BL21) (2013)
M. thermophila 0.5 % (v/v) V = 150 mL, 30 °C, DSWB 930 – Topakas et al. (2012)
(P. pastoris X-33) Methanol 6 days
P. brasilianum IBT 20888 20 % BSG 30 °C, 196 h, 1542c – Panagiotou et al. (2006)
(2.5 g) Vextr = 10 mL/1 g dry
material
P. funiculosum Knoshaug et al. (2008)
(A.awamori)
(continued)
91
Table 5.1 (continued)
92

Organism Carbon source/ Cultivation conditions Assay FAE activity References


inducer (mU/mL) (mU/mg)
FaeA Glucoamylase V = 2.8 L, 32 °C, 4– MFA – –
6 days
FaeB Glucoamylase V = 2.8 L, 32 °C, 4– MFA – –
6 days
Piromyces equi 30 °C, 120 rpm, 2 days MpCA 660 20,000 Poidevin et al. (2009)
(T. reesei RutC30)
P. eryngii SNL medium, 24 °C, 13 days, 150 rpm MFA – 650 Nieter et al. (2014)
0.4 % Tween
80
P. sapidus 8266 DSMZ SNL medium, 5 days pNPB 243 – Linke et al. (2013)
0.4 % Tween
80
Rumen microbial IPTG Standard protocol WB – 10,300 Wong et al. (2013)
metagenome
(E. coli BL21)
Russula virescens MFA Wang et al. (2014)
S. cellulosum So ce56 Wu et al. (2012)
(E. coli Tuner cells)
ScFAE1 1 mM IPTG Standard protocol MFA – 12,200
ScFAE2 1 mM IPTG Standard protocol MFA – 19,300
Streptomyces ambofaciens 1 % (w/v) MpCA/MFA Kheder et al. (2009)
DSWB
S. cinnamoneus FAE activity was Uraji et al. (2014)
(E. coli BL21) expressed on a dry weight
basis
(continued)
C. Katsimpouras et al.
Table 5.1 (continued)
Organism Carbon source/ Cultivation conditions Assay FAE activity References
inducer (mU/mL) (mU/mg)
R18 IPTG Standard protocol MFA – 23.07
R43 IPTG Standard protocol MFA – 19.8
Streptomyces C-248 1 % DSWB V = 1 L, 37 °C, 3 days DSWB 80 300 Faulds et al. (2006)
Streptomyces S10 1.5 % DSWB V = 20 mL, 30 °C, 96 h DSWB 2.0 15.45 Mukherjee et al. (2007)
T. stipitatus 2 % BSG/2 % V = 100 mL, 37 °C, MFA 140/90b – Mandalari et al. (2008)
WB 10 days
Thermoanaerobacter 0.1 mM IPTG Standard protocol MFA/MpCA – 456/426 Abokitse et al. (2010)
tengcongencis (E. coli)
T. xylanilyticus (E. coli) IPTG Standard protocol MFA – 25410 Rakotoarivonina et al.
(2011)
Literature is cited after 2005, for earlier data see Topakas et al. (2007) for a review
WB wheat bran, MB maize bran, DSWB destarched wheat bran, BSG brewer’s spent grain, CB corn bran, CC corn cobs, IPTG isopropyl β-D-
5 Role and Applications of Feruloyl Esterases …

1-thiogalactopyranoside, MFA methyl ferulate, MpCA methyl p-coumarate, EFA ethyl ferulate, MCA methyl caffeate, 4NPF 4-nitrophenyl ferulate, pNFA
p-nitrophenyl ferulate, pNPA p-nitrophenyl acetate, pNPB p-nitrophenyl butyrate, CNPF 2-chloro-4-nitrophenyl ferulate, SNL standard nutrition liquid
a
ssf/smf
b
BSG/WB
c
FAE activity was expressed on a dry weight basis
93
94 C. Katsimpouras et al.

FAE production through native secretory pathways of microorganisms is mainly


performed with two types of fermentation. Submerged fermentation (SmF), where
the microorganism is in contact with the liquid growth medium, and solid-state
fermentation (SSF), where the microorganism grows on solid substrates in the near
absence of water. SSF main advantages over SmF include higher yields, simpler
technology with rare operational problems, and resemblance to microorganism’s
natural habitat (Katsimpouras et al. 2014). Among Aspergillus species, A. flavipes
(Mathew et al. 2005), A. niger (Johnson et al. 1989) and A. awamori (Kanauchi
et al. 2008) were reported as very efficient FAE producers under SmF in the
presence of lignocellulosic materials such as wheat bran and destarched wheat bran
while P. brasilianum was the more active producer under SSF conditions on
brewer’s spent grain (Panagiotou et al. 2006).
The choice of the optimal substrate stand to be crucial for the homologous
esterase expression as its role is not only to serve as a carbon and energy source but
also to provide the necessary inducing compounds for the microorganism.
Lignocellulosic carbon sources that contain high amounts of esterified FA such as
wheat bran (Mathew et al. 2005; Kanauchi et al. 2008; Mandalari et al. 2008;
Hegde and Muralikrishna 2009; Gottschalk et al. 2013), corn bran (Shin and Chen
2006), corn cobs (Yang et al. 2013), destarched wheat bran (Faulds et al. 2006;
Mukherjee et al. 2007; Kheder et al. 2009; Tai et al. 2014), maize bran (Mathew
et al. 2005) and brewer’s spent grain (Panagiotou et al. 2006; Mandalari et al.
2008), have been successfully employed for FAEs production. Furthermore, in
many cases the effect of removal of FA from these materials was investigated with
the results revealing that deesterified carbon sources lead to a decreased production
of FAE compared to untreated materials even when the media were supplemented
with free FA (Topakas et al. 2003a, b, c; Topakas and Christakopoulos 2004; Shin
and Chen 2006). In contrast to these results, production of FAE was enhanced with
the addition of free FA but also FA’s breakdown products and other aromatic
compounds could induce FAE production as well (Faulds et al. 1997; Faulds and
Williamson 1999; de Vries and Visser 1999; de Vries et al. 2002).
FAEs’ wide range of potential applications necessitates the efficient production
of novel recombinant enzymes, and the availability of more and more genome
sequences for several microorganisms constitutes a powerful tool facilitating their
discovery. Native enzyme mixtures production poses some difficulties due to their
high production cost, as there is a need for special culturing and induction condi-
tions (Lambertz et al. 2014). In order to overcome this obstacle, gene cloning and
heterologous expression have been employed, producing enzymes with superior
properties.
To date, host microorganisms such as bacteria and yeasts have been used for
FAEs expression. The most widely used bacterial expression host is Escherichia
coli and more specifically the strain BL21, which has the advantage of being
deficient in the Ion and ompT proteases. As shown in Table 5.1, E. coli BL21 cells
were employed in recombinant FAEs expression from several microorganisms such
as Streptomyces cinnamoneus, Thermobacillus xylanilyticus, C. japonicus,
L. plantarum, Aspergillus clavatus, Cellulosilyticum ruminicola, Anaeromyces
5 Role and Applications of Feruloyl Esterases … 95

mucronatus, Dickeya dadantii, Thermoanaerobacter tengcongencis, A. awamori,


B. proteoclasticus and L. johnsonii. Li et al. (2011) used E. coli Rosetta-gami 2 and
Rosetta for the expression of two FAEs (FaeII and FaeIII, respectively) from C.
ruminicola as these particular strains enable the enhanced formation of disulfide
bonds in the cytoplasm and facilitate the expression of genes that encode rare
E. coli codons. Furthermore, ScFAE1 and ScFAE2 from S. cellulosum were
overexpressed using E. coli Tuner cells, which enable precise control of protein
expression with IPTG (Wu et al. 2012).
Besides bacterial expression systems, yeasts such as Saccharomyces cerevisiae
and Pichia pastoris constitute also attractive host organisms for heterologous pro-
duction of eukaryotic proteins exhibiting several advantages such as high growth
rate and protein titers using low-cost growth media (Várnai et al. 2014). Yeasts are
also capable of protein folding and posttranslational modifications such as glyco-
sylation, with P. pastoris having the advantage over S. cerevisae of not hypergly-
cosylating secreted proteins (Grinna and Tschopp 1989). In addition, P. pastoris
secretes low levels of native proteins simplifying the purification process of the
recombinant protein (Barr et al. 1992). Moukouli et al. (2008) described first the
successful cloning and expression of a type C FAE from F. oxysporum in P. pastoris
X-33 employing a bioinformatic-assisted functional screening in order to discover
and analyze the enzyme. FAEs were also cloned and expressed in P. pastoris
from different Aspergillus species such as A. oryzae (Koseki et al. 2009; Zeng et al.
2014), A. flavus (Zhang et al. 2013), Aspergillus nidulans (Bauer et al. 2006; Debeire
et al. 2012) and A. usamii (Gong et al. 2013) and from M. thermophila (Topakas
et al. 2012).
A type B recombinant FAE was cloned from the genomic DNA of A. nidulans
and expressed in S. cerevisiae exhibiting about 86 % of total FAE activity in the
culture medium indicating an efficient protein secretion (Shin and Chen 2007).
Wong et al. (2011) cloned a FAE gene from A. niger into S. cerevisae achieving an
efficient secretion and a yield of about 2 mg/L.
Recently, there are reports describing the production, purification, and charac-
terization of FAEs from basidiomycetes. The limited knowledge on FAEs from
basidiomycetes could be explained due to their high laccase activity as it interferes
with determination of FA (Nieter et al. 2014). Schizophyllum commune and
Pleurotus sapidus were the only basidiomycetes, which were associated with FAE
activity (MacKenzie and Bilous 1988; Linke et al. 2013). However, Haase-Aschoff
et al. (2013) came up with a new efficient analytical method, using multiple sub-
strates, for FAEs detection circumventing substrate and FA polymerization. After
screening 41 basidiomycete strains, more than half of them exhibited FAE activity
with different substrate specificity. Pleurotus eryngii was cultivated in standard
nutrition liquid supplemented with Tween 80 as inducer and reached maximum
FAE activity on the 13th day (Nieter et al. 2014).
Physical characteristics such as molecular weight, isoelectric point, and optimum
hydrolytic reaction conditions differ significantly among purified FAEs (Table 5.2).
FAEs exhibit a wide range of molecular weights and isoelectric points from 11 to
210 kDa and from 3.0 to 9.9 respectively. The extracellular FAE from
Table 5.2 Physicochemical properties of purified FAEs known to date
96

Microorganism Enzyme Gene Data bank FAE MW pHopt Topt pI References


type (kDa) (°C)
A. awamori FE – 112 3.7 Wang et al. (2004)
CE – 75 4.2 Wang et al. (2004)
– – 35 5.0 45 3.8 Koseki et al. (1998)
AwFAEA AwfaeA AB032760 A 37 5.01 Koseki et al. (2005)
– 78 5.012 4012 Kanauchi et al. (2008)
AwFaeA AwfaeA AB032760 A 35 5.513 5513 4.2 Fazary et al. (2010)
A. clavatus AcFAE D 31 71 301 Damásio et al. (2013)
A. flavus AfFaeA A 40 6.02 582 Zhang et al. (2013)
A. mucronatus Fae1A fae1A – 37 7.2 37 Qi et al. (2011)
A. nidulans AnidFAE AN5267.2 – 28 6.12 372 Debeire et al. (2012), Bauer et al.
(2006)
AN1772.2 B 130 7.02 452 Shin and Chen (2007)
a
A. niger FAE-I B 63 3.0 Faulds and Williamson (1993)
FAE-II A 29 3.6 Faulds and Williamson (1993)
FAE-III or faeA Y09330 A 36 5.02 552 3.3 Faulds et al. (1997)
AnFaeA
CinnAE or faeB AJ309807 Bb 75.8a 6.03 503 4.8 Kroon and Williamson (1996)
AnFaeB
CE – 120 McCrae et al. (1994)
FAE-1 50 9.014 4014 Hegde and Muralikrishna (2009)
FAE-2 55 6.014 40–5014 Hegde and Muralikrishna (2009)
(continued)
C. Katsimpouras et al.
Table 5.2 (continued)
Microorganism Enzyme Gene Data bank FAE MW pHopt Topt pI References
type (kDa) (°C)
– faeA XP_001393337 31 6–714 5014 Wong et al. (2011)
A. oryzae FAE – 30 4.5– 3.6 Tenkanen et al. (1991)
6.04
AoFaeB AofaeB XP_001818628 B 61 6.01 Koseki et al. (2009)
AoFaeC AofaeC XP_001819091 C 75 6.01 Koseki et al. (2009)
AoFaeA AofaeA A 37 5.02 502 Zeng et al. (2014)
A. pullulans – B 210 6.75 605 6.5 Rumbold et al. (2003)
A. tubingensis FaeA faeA Y09331 – 36 Zwane et al. (2014)
A. usamii E001 AuFaeA AufaeA A 36 5.02 452 Gong et al. (2013)
Butyrivibrio – cinI or U44893 – Barbe and Dubourdieu (1998)
fibrisolvents cinA
5 Role and Applications of Feruloyl Esterases …

– cinII or U64802 – De Vries et al. (1997)


cinB
B. proteoclasticus Est1E est1E 31.6 Goldstone et al. (2010)
C. ruminicola FaeI faeI – 58 6–72 402 Li et al. (2011)
FaeII faeII – 32 82 352 Li et al. (2011)
FaeIII faeIII – 46 92 402 Li et al. (2011)
C. japonicus XLYD or xynD X58956 D 59 Ferreira et al. (1993)
CjXYLD
C. japonicus Fee1B fee1B D 61 6.52 35–402 McClendon et al. (2011)
2
Chaetomium sp. – B 30.2 7.5 602 Yang et al. (2013)
(continued)
97
Table 5.2 (continued)
98

Microorganism Enzyme Gene Data bank FAE MW pHopt Topt pI References


type (kDa) (°C)
Chrysosporium FaeA1 JF826027 A 29 6–72 452 5.5 Kühnel et al. (2012)
lucknowense
FaeA2 JF826028 A 36 7.52 402 5.2 Kühnel et al. (2012)
2
FaeB2 JF826029 B 33 7 452 6.0 Kühnel et al. (2012)
C. stercorarium – C or D 33 8.02 652 Donaghy et al. (2000)
C. thermocellum XynZ XynZ M22624 – 45 4–76 50–606 5.8 Blum et al. (2000)
– XynY X83269 – Blum et al. (2000)
D. dadantii FaeT faeT 35 Hassan and Hugouvieux-Cotte-Pattat
(2011)
FaeD faeD 31, 35 Hassan and Hugouvieux-Cotte-Pattat
(2011)
F. oxysporum FoFAE-I or B 31 7.07 557 >9.5 Topakas et al. (2003a, b, c)
FoFaeB
FAE-II or A 27 7.08 458 9.9 Topakas et al. (2003a, b, c)
FoFaeA
F. proliferatum FAE B 31 6.5– 502 Shin and Chen (2006)
7.52
Lactobacillus – – 36 5.69 379 Topakas et al. (2004)
acidophilusc
L. johnsonii Lj0536 WP_004898050.1 31 7.8 20 Kin et al. (2009)
(continued)
C. Katsimpouras et al.
Table 5.2 (continued)
Microorganism Enzyme Gene Data bank FAE MW pHopt Topt pI References
type (kDa) (°C)
Lj1228 WP_011162057.1 31 6.7 30 Kin et al. (2009)
L. plantarum Lp_0796 lp_0796 – 28 715 30–3715 Esteban-Torres et al. (2013)
M. thermophila FoFaeC-12213 Foxg- C 62 6.02 652 6.8 Moukouli et al. (2008)
12213.2
M. thermophila MtFae1a fae1a 96,478 B 39 7.02 502 Topakas et al. (2012)
a 10
Neocallimastix MC-2 pCAE – 11 7.2 4.7 Borneman et al. (1993)
FAE-I – 69 Borneman et al. (1992)
FAE-II – 24 Borneman et al. (1992)
N. crassa Fae-1 Fae-1 AJ293029 B 35 6.02 552 Crepin et al. (2003a)
NcFaeD-3.544 faeD- D 32 Crepin et al. (2004a, b)
3.544
5 Role and Applications of Feruloyl Esterases …

P. equi EstA estA AF164516 D 55 6.711 50–6011 Fillingham et al. (1999)


7
estA AF16516 30 6.5 50–607 Poidevin et al. (2009)
P. eryngii PeFaeA A 67 5.02 502 5.2 Nieter et al. (2014)
Penicillium expansum – possible B 57.5 5.62 372 Donaghy and McKay (1997)
P. funiculosum FAEB or faeB AJ291496 B 53 6.0 Kroon et al. (2000)
PfFaeB
FaeA faeA AJ312296 Knoshaug et al. (2008)
Penicillium pCAE/FAE – 57 6.02 552 4.6 Castanares et al. (1992)
pinophilum
P. sapidus A 55 6.02 502 5.7 Linke et al. (2013)
Rumen microbial RuFae2 C 29 7 50 8.5 Wong et al. (2013)
metagenome
(continued)
99
Table 5.2 (continued)
100

Microorganism Enzyme Gene Data bank FAE MW pHopt Topt pI References


type (kDa) (°C)
R. virescens – 62 5.02 502 Wang et al. (2014)
17
Soil metagenomic Tan410 tan410 – 55 7 3517 4.7 Yao et al. (2013)
library
S. cinnamoneus R18 AB921569 D 38 7.513 5013 Uraji et al. (2014)
13
R43 AB921570 D 52 7 4013 Uraji et al. (2014)
S. cellulosum ScFAE1 sce1896 D 35 72 5.07 Wu et al. (2012)
ScFAE2 sce8927 D 34 6–82 4.67 Wu et al. (2012)
a
S. thermophile StFAEA or B 33 6.02 55–602 3.5 Topakas et al. (2004)
StFaeB
StFaeC C 23a 6.02 552 <3.5 Topakas et al. (2005a, b)
2 2
S. olivochromogenes FAE – 29 5.5 30 7.9 Faulds and Williamson (1991)
T. stipitatus TsFaeA A 35 5.3 Garcia-Conesa et al. (2004)
TsFaeB B 35 3.5 Garcia-Conesa et al. (2004)
TsFaeC faeC AJ505939 C 66 6–73 603 4.6 Garcia-Conesa et al. (2004), Crepin
et al. (2003b)
T. tengcongensis TtFAE A 33 8.016 Abokitse et al. (2010)
T. xylanilyticus Tx-Est1 Tx-est1 – 37.4 8.52 652 Rakotoarivonina et al. (2011)
Numbers in superscripts indicate the substrates used for pH and temperature optimum: 1. α-naphthylbutyrate; 2. methyl ferulate (MFA); 3. methyl caffeate (MCA); 4.
wheat straw (WS); 5. 4-nitrophenyl 5-O-trans-feruloyl-α-L-arabinofuranoside (NPh-5-Fe-Araf); 6. O-{5-O-(trans-feruloyl)-[O-β-D-xylopyranosyl-(1 → 2)]-O-α-L-
arabinofuranosyl-[1 → 3]}-O-β-D-xylopyranosyl-(1 → 4)-D-xylopyranose (FAXXX); 7. methyl p-coumarate (MpCA); 8. methyl sinapate (MSA); 9. [2-O-(trans-
feruloyl)-α-L-arabinofuranosyl]-(1 → 5)-L-arabinofuranose (FAA); 10. O-[5-O-(trans-p-coumaroyl)-α-L-arabinofuranosyl]-(1 → 3)-O-β-D-xylopyranosyl-(1 → 4)-D-
xylopyranose (PAXX); 11. O-[5-O-(trans-feruloyl)-α-L-arabino-furanosyl]-(1 → 3)-O-β-D-xylopyranosyl-(1 → 4)-D-xylopyranose (FAXX); 12. 1-naphthyl acetate; 13.
Ethyl ferulate (EFA); 14. 4-nitrophenyl ferulate (4NPF); 15. p-nitrophenyl butyrate; 16. pNP caprylate; 17. p-nitrophenyl ferulate
a
Dimeric proteins (molecular weight estimated with SDS-PAGE electrophoresis)
b
Phylogenetic analysis of AnFaeB indicated that this enzyme belongs to the type C subclass (Crepin et al. 2004a)
c
Typical human intestinal bacterium
C. Katsimpouras et al.
5 Role and Applications of Feruloyl Esterases … 101

Aureobasidium pullulans with an apparent molecular weight of 210 kDa, is the


largest single-subunit esterase of its kind due to an extensive Asp-linked glyco-
sylation reaching a degree of 48 % (Rumbold et al. 2003). Optimum hydrolytic
conditions also vary with pH ranging from 4.0 to 9.0 and temperature from 20 to
65 °C depending on the source microorganism. Comparing the physicochemical
characteristics and conditions for optimal activity of purified FAEs shown in
Table 5.2, there is no pattern indicating a correlation among them.

5.6 Applications of FAEs

During the last decade, FAEs have gained increased interest acquiring considerable
role in biotechnological processes. Except for their role in biofuel industry as
accessory enzymes for the degradation of lignocellulose, FAEs’ potential applica-
tions cover a broad spectrum, including the pulp and paper industry as bleaching
agents, the cosmetic and pharmaceutical industry for the synthesis of bioactive
compounds with antioxidant and/or antimicrobial activity or for the production of
flavor and fragrance precursors, the food industry as food additives or as feed
additives in animal feeds (Fig. 5.3).

5.6.1 Exploiting the Hydrolytic Activity

The utilization of the FAEs’ hydrolytic activity lies on the release of FA from
hemicellulose present in plant cells walls. In ruminal digestion, FAEs are important
as they deesterify dietary fibers releasing hydroxycinnamates but are also take part
in colonic fermentation where their activities in gut ruminal microorganisms

Fig. 5.3 Potential applications of FAEs in Industrial Biotechnology


102 C. Katsimpouras et al.

enhance the breakdown of ester bonds in hydroxycinnamates (Esteban-Torres et al.


2013). This is why FAEs are used for the pretreatment of food and animal feeds or
even as food/feed additives, as they allow the in situ improved digestibility of
hemicellulose. In animal feed industry the advantages of their use include improved
feed utilization, control of body weight gain and higher milk yield in cattle and
sheep (Howard et al. 2003). In food industry, FAE preparations are also widely
used in the bakery industry along with glucanases and oxidases in order to solu-
bilize arabinoxylan fractions of the dough resulting to increased bread volume and
improved quality (Butt et al. 2008). Other applications of FAEs include the syn-
thesis of oligosaccharides that can be utilized as functional food additives and their
use for juice clarification. On the other part, FA is one of the major antioxidant
additives in food and beverages such as beer (Benoit et al. 2008). The pretreatment
of agricultural by-products with FAEs could offer an economic alternative for the
production of FA for these purposes. Being also a flavor precursor, FA can be
further converted to valuable compounds such as vanillin and vanillic acid. The
biotransformation of FA has been investigated using different processes and fila-
mentous fungi (Benoit et al. 2008; Walton et al. 2000). Apart from its use for
flavoring, vanillin is also a fundamental constituent for the synthesis of pharma-
ceuticals and is used extensively in the perfume and metal plating industries, while
its herbicidal activity is useful as ripening agent for the achievement of higher
yields of sucrose in sugar canes (Fazary and Ju 2008).
In paper pulp processing, environment-friendly approaches include the
replacement of chemicals with enzymes, resulting to the reduction of water pol-
lution and associated clean-up costs (Koseki et al. 2009). Xylanases and laccases
are mainly used for enzymatic delignification and biobleaching, respectively,
leading to the reduction or elimination of chlorine-based chemicals (Valls et al.
2010; Thakur et al. 2012). The use of FAEs along with other accessory enzymes
might enhance this process by removing substitutions and linkages between
polymers, resulting to the detachment of hemicellulose walls and the release of
lignin fragments. A recombinant FAEA from A. niger has been used for the first
time in 2003 in combination with laccase and xylanase for the efficient delignifi-
cation of wheat straw pulp, which yielded 78 % (Record et al. 2003). In addition, it
has been shown to boost delignification in flax pulp resulting in very low kappa
number and pulp brightness (Sigoillot et al. 2005). The commercial lipase A
“Amano” with significant FAE and other accessory activities, when tested for its
bleachability in kraft pulps, offered the first evidence that accessory enzymes from a
commercial preparation such as FAE and arabinofuranosidase can result to direct
bleaching effect (Nguyen et al. 2008). Other minor applications of the FAEs’
hydrolytic activity in paper industry is the removal of fine particles from the pulp
facilitating water removal and the enhancement of chemical and mechanical
paper-pulping methods allowing the easier solubilization of lignin–carbohydrate
complexes (Fazary and Ju 2008). A prerequisite for their use is that enzyme
preparations are free of cellulases, since cellulose degradation would result in a
reduction in the quality of the pulp.
5 Role and Applications of Feruloyl Esterases … 103

5.6.2 FAEs as Biosynthetic Tools of Plant Derived


Antioxidants

Ester-linked substituents (ferulic, p-coumaric, caffeic, sinapinic acid) on plant cell


wall polysaccharides have widespread potential due to their antimicrobial, photo-
protectant, antitumour and antioxidant activities (Faulds 2010), while some have
long been used as food preservatives in order to inhibit microbial growth (Fazary
and Ju 2008). However, their application in oil-based food and cosmetics industry
is limited due to their relatively low solubility in aprotic media. The direct esteri-
fication of phenolic, including hydroxycinnamic, acids or the transesterification of
their esters with fatty alcohols or sugars has gained increased interest as it targets to
the production of biologically active compounds with improved properties
(Fig. 5.4). The synthesized phenolic fatty esters are generally more lipophilic and
allow their application in oil-based processes; on the other hand, the phenolic acid
sugar esters are more hydrophilic nonionic biosurfactants and have clear antitumor
activity, so the potential to be used in order to formulate antimicrobial, antiviral
and/or anti-inflammatory agents is promising (Fazary and Ju 2008). Enzymatic
transesterification offers an alternative to the poor selectivity of chemical synthesis.
It is a one-step process resulting to the synthesis of one product instead of a mixture
of esters thus there is no unwanted side reactions causing darkening or the for-
mation of odors and no need of by-product and catalyst residues removal.
Complementary to the attractive environment-friendly configurations of the pro-
cess, the use of enzymes derived from thermophilic fungi allows the implementa-
tion of lower temperature (50–60 °C) than that of the chemical process (160 °C)
resulting to lower operating costs. Enzyme-catalyzed synthesis has been widely
studied in lipases including hydroxycinnamate (trans)esterification in nonconven-
tional media (organic solvents, ionic liquid mixtures). Lipases are known to be
more stable than FAEs in such reaction media, display high activity at low water

Fig. 5.4 Modification of hydroxycinnamic acids by FAEs in nonconventional reaction media. FA


ferulic acid, pCA p-coumaric acid, SA sinapic acid, CA caffeic acid
104 C. Katsimpouras et al.

content and give high yields (Zeuner et al. 2011). On the contrary, FAEs exhibit
higher substrate specificity constituting them a potential biosynthetic tool with high
efficiency.
As with lipases, a reaction medium with low or non-water content is essential in
order to boost the FAEs’ synthetic activity to the detriment of the hydrolytic one.
The first synthetic reaction catalyzed by FAE was achieved in a water-in-oil
microemulsion system for the synthesis of 1-pentyl-ferulate (Giuliani et al. 2001).
Since then, detergentless microemulsions, which are ternary systems consisted of a
hydrocarbon, a short-chained alcohol and water, have mostly been employed for the
synthesis of various alkyl and sugar esters (Table 5.3). They are low water content
media representing thermodynamically stable dispersions of aqueous microdroplets
in the hydrocarbon solvent. The spherical droplets are stabilized by alcohol
molecules adsorbed at their surface, while the enclosed in water enzyme is pro-
tected from contacting the outer organic phase and from denaturation due to a
water-rich layer (Khmelnitsky et al. 1988). An important advantage of these mix-
tures as reaction systems is that they allow the separation of reaction products and
enzyme reuse, while the solubility of relatively polar phenolic acids is high owing
to the presence of large amount of polar alcohol. In these media, transesterification
reactions using activated esters of FA as acyl donors have been proven to be faster
and more efficient compared to the direct esterifications (Olsson et al. 2011).
Another approach for efficient transesterification is the use of ionic liquid mixtures
(ILs), which comprise organic salts that remain liquid under ambient temperature
(Table 5.4). The interest in ILs as reaction media is mainly urged by their lack of
vapor pressure as they have the potential to replace volatile organic solvents
resulting to the development of greener processes. A major advantage of using ILs
is their tailorability as they exhibit increased selectivity and ability to solubilize
both polar and nonpolar substrates and products, as well as they can adjust other
physicochemical properties such as density, viscosity and solvating power through
alternating the cation and anion type to meet the needs of each system (Zeuner et al.
2011). The lack of volatility together with their chemical and thermal stability allow
simple recycling and reuse therefore could reduce significantly the cost of the
process with respect to organic-based ones. When referring to transesterification
with saccharides, the advantage of using ILs, as compared to organic solvents is the
increased substrate solubility. Furthermore higher yields in shorter reaction times
including regioselectivity may also be achieved in enzymatic acylation of saccha-
rides in ILs compared to organic solvents. Most of the research has used
second-generation ILs, which may be too expensive for commercial applications.
The availability of advanced ionic liquids- greener, inexpensive and biodegradable-
increases the likelihood that they will find commercial use in biocatalysis (Gorke
et al. 2010). Although synthesis in organic solvents has been widely studied in
lipases, their use in FAEs has been rarely reported in literature due to stability
restraints (Table 5.5). An important advantage is that they allow the implementation
of one phase reaction and the separation of reaction products, although they do not
consist a green approach for transesterification.
Table 5.3 Enzymatic synthesis of hydroxycinnamic acid esters in detergentless microemulsions reported in the literature
Hydroxycinnamate Acyl donor Enzyme Solvent system Yield (h) T (°C) References
Synthesis of aliphatic
esters
Methyl ferulate 1-Butanol StFaeC n-Hexane:1-butanol: MES-NaOH pH Up to 20 % 35 Topakas
6.0 (53.4:43.4:3.2 v/v/v) (120) et al. (2005a, b)
Methyl sinapate Up to 10 %
(*144)
Methyl p-coumarate Up to 8 %
(*144)
Methyl caffeate Up to 5.5 %
(*144)
Methyl ferulate 1-Butanol StFae-A n-Hexane:1-butanol: Up to 8 % 35 Topakas et al.
buffer (47.2:50.8:2.0 v/v/v) (144) (2004)
Methyl p-coumarate Up to 50 %
5 Role and Applications of Feruloyl Esterases …

(144)
Methyl caffeate Up to 25 %
(144)
Methyl p-coumarate 1-Butanol FoFae-I n-Hexane/1-butanol/MES-NaOH pH Up to 70 % 35 Topakas et al.
6.0 (47.2:50.8:2.0 v/v/v) (144) (2003a)
Methyl caffeate Up to 22 %
(144)
Methyl ferulate Up to 13 %
(144)
Methyl sinapinate Up to 1 %
(144)
(continued)
105
Table 5.3 (continued)
106

Hydroxycinnamate Acyl donor Enzyme Solvent system Yield (h) T (°C) References
p-hydroxyphenylacetic 1-Propanol FoFae-II n-Hexane/1-propanol/MES-NaOH pH 75 (224) 30 Topakas et al.
acid 6.6 (47.2:50.8:2.0 v/v/v) (2003b)
p- 70 (224)
Hydroxyl-phenylpropionic
acid
Methyl sinapate 1-Butanol AnFaeA+1 n-Hexane:1- or 2-butanol: 56 % (120) 35 Vafiadi et al.
MES-NaOH pH 6.0 (47.2: 50.8: 2.0 (2008a)
v/v/v)
78 %a (120)
Methyl ferulate AnFaeA 42 % (120)
Methyl p-coumarate 2 % (120)
Methyl sinapate 2-Butanol 9 % (120)
Methyl ferulate 1-Butanol Ultraflo L+1 n-Hexane/1-butanol/water 97 % (144) 37 Vafiadi et al.
(47.2:50.8:2.0 v/v/v) (2008b)
Ultraflo 3.6 % (144)
Methyl ferulate 1-Butanol Depol n-Hexane/1-butanol/water 87 % (144) 37 Vafiadi et al.
740L+1 (47.2:50.8:2.0 v/v/v) (2008b)
Depol 740L 2.6 % (144)
Methyl ferulate 1-Butanol Depol 670L+ n-Hexane/1-butanol/water 5 % (144) 37 Vafiadi et al.
(47.2:50.8:2.0 v/v/v) (2008b)
Synthesis of sugar esters
Methyl ferulate L-Arabinose StFaeC n-Hexane:t-butanol:piperazine- HCl 40 % (160) 35 Topakas et al.
pH 6.0 (53.4:43.4:3.2 v/v/v) (2005b)
Methyl ferulate Up to 50 %a Vafiadi et al.
(120) (2005)
(continued)
C. Katsimpouras et al.
Table 5.3 (continued)
Hydroxycinnamate Acyl donor Enzyme Solvent system Yield (h) T (°C) References
Ethyl ferulate 6.3 % (–)
n-Propyl ferulate 3.8 % (–)
n-Butyl ferulate 3.4 % (–)
Isopropyl ferulate 3.3 % (–)
2-Butyl ferulate 2.7 % (–)
Isobutyl ferulate 4.2 % (–)
Methyl ferulate D-Arabinose 45 % (–)
D-Glucose n.q. (120) Vafiadi et al.
(2007a)
D-Xylose n.q. (120) Vafiadi et al.
(2005)
D-Mannose n.q. (120)
5 Role and Applications of Feruloyl Esterases …

D-Fructose n.q. (120)


D-Galactose n.q. (120)
D-Ribose n.q. (120)
pNP-arabinofuranoside n.q. (120)
n.q. (120)
pNP-anabinopyranoside n.q. (–)
n.q. (–)
L-Arabinotriose n.q. (–) 37 Vafiadi et al.
(2007b)
(continued)
107
Table 5.3 (continued)
108

Hydroxycinnamate Acyl donor Enzyme Solvent system Yield (h) T (°C) References
L-Arabinotetraose n-Hexane:2-methyl-2-propanol: n.q. (–)
piperazine HCl pH 6.0 (47.2:50.8:2.0
v/v/v)
Methyl ferulate L-Arabinopentaose 24 %a (96)
Ethyl ferulate L-Arabinohexaose 3 % (96)
Propyl ferulate L-Arabinose 4 % (96) 35 Vafiadi et al.
(2006a)
Methyl ferulate L-Arabinose TsFaeC n-Hexane: t-butanol: MOPS-NaOH 21.2 %a (96) 40 Vafiadi et al.
pH 6.0 (53.4:43.4:3.2 v/v/v) (2006b)
Ethyl ferulate 1.4 % (96)
Ferulic acid D-Arabinose Multifect n-Hexane/1-butanol or 11.3– 35 Couto et al.
P 3000 2-butanone/MES-NaOH pH 6.0 36.7 % (2010)
(51:46:3 v/v/v) (144)
D-Galactose 5.6–8.6 %
(144)
D-Xylose 12.1–
30.8 %
(144)
Ferulic acid D-Arabinose Ceremix n-Hexane/1-butanol or 15.3– 35 Couto et al.
2-butanone/MES-NaOH pH 6.0 32.5 % (2010)
(51:46:3 v/v/v) (144)
Ferulic acid D-Galactose RP-1 n-Hexane/1-butanol or 10.2– 35 Couto et al.
2-butanone/MES-NaOH pH 6.0 19.8 % (2010)
(51:46:3 v/v/v) (144)
D-Xylose 4.4–16.3 %
(144)
(continued)
C. Katsimpouras et al.
Table 5.3 (continued)
Hydroxycinnamate Acyl donor Enzyme Solvent system Yield (h) T (°C) References
Ferulic acid D-Arabinose Depol 670 n-Hexane/1-butanol or 7.2–17.7 % 35 Couto et al.
2-butanone/MES-NaOH pH 6.0 (144) (2010)
(51:46:3 v/v/v)
D-Galactose 3.4–15.8 %
(144)
D-Xylose 20.9–
26.5 %
(144)
Ferulic acid D-Arabinose Flavourzyme n-Hexane/1-butanol or 21.9– 35 Couto et al.
2-butanone/MES-NaOH pH 6.0 36.7 % (2010)
(51:46:3 v/v/v) (144)
D-Galactose 36.2–
41.9 %
(144)
5 Role and Applications of Feruloyl Esterases …

D-Xylose 20.1–
21.7 %
(144)
Ferulic acid Raffinose Depol 740L n-Hexane/2-butanone/MES-NaOH pH 11.9 %a 35 Couto et al.
6.0 (51:46:3 v/v/v) (168) (2011)
Xylobiose 9.4 % (144)
Arabinose 7.9 % (144)
Galactobiose 5.4 % (144)
(continued)
109
Table 5.3 (continued)
110

Hydroxycinnamate Acyl donor Enzyme Solvent system Yield (h) T (°C) References
Sucrose 13.2 % (–)
Lactose 4.4 % (–)
XOS 2.8 % (–)
FOS n-Hexane/1,4-dioxane/MES-NaOH 9.6 % (–)
pH 6.0 (51:46:3 v/v/v)
Raffinose 3.1 % (144)
Xylobiose 4.2 % (144)
Galactobiose 26.8 %
(144)
a
After optimization of reaction conditions; +1: Immobilized with CLEAs methodology; n.q.: detected but not quantified; StFaeC: FAE type C from S.
thermophile ATCC 34628; StFaeA: FAE type B from S. thermophile ATCC 34628; FoFae-I/FoFae-II: FAE from F. oxysporum F3; AnFaeA: FAE type A
from A. niger; Ultraflo L/ Depol 740L: multienzymatic preparation from H. insolens; Depol 670L: multienzymatic preparation from T. reesei; TsFaeC: FAE
type C from T. stipitatus; Multifect P 3000: multienzymatic preparation from Bacillus amyloliquefaciens; Ceremix: multienzymatic preparation from Bacillus
spp.; RP-1: multienzymatic preparation from Bacillus subtilis; Depol 670L: multienzymatic preparation from T. reesei; Flavourzyme: multienzymatic
preparation from A. oryzae; Depol 740L: multienzymatic preparation from H. insolens. Systems with conversion <1 % were not included
C. Katsimpouras et al.
5 Role and Applications of Feruloyl Esterases … 111

Table 5.4 Enzymatic synthesis of hydroxycinnamic acid esters in ILs reported in the literature
Hydroxycinnamate Acceptor Enzyme Ionic liquid Yield (h) T (°C) References
system
Sinapic acid Glycerol AnFaeA [C2OHmim] 72.5%a (24) 50 Vafiadi et al.
[PF6] (85%), (2009)
MOPS-NaOH
pH 6.0 (15%)
[C5OHmim] 76.5%a (24)
[PF6] (85%),
MOPS-NaOH
pH 6.0 (15%)
Methyl sinapate AnFaeA+1 [ΒΜΙm][PF6] 13% (0.5) 40 Zeuner et al.
AnFaeA (85%), MOPS (2011)
pH 6.0 (15%)
[C2OHMIm] 21% (0.5)
[BF4] (85%),
MOPS pH 6.0
(15%)
[C5OHmim] 72.5%a (24) 50 Vafiadi et al.
[PF6] (85%), (2009)
MOPS-NaOH
pH 6.0 (15%)
[C2OHmim] Up to 2.5%
[PF6] (94 %), (24)
MOPS-NaOH
pH 6.0 (6%)
[C5OHmim] Up to 7%
[PF6] (94%), (24)
MOPS-NaOH
pH 6.0 (6%)
Sinapic acid Glycerol AndFaeC [ΒΜΙm][PF6] 1.1% (0.5) 40 Zeuner et al.
(85%), MOPS (2011)
pH 6.0 (15%)
Sinapic acid Glycerol Ultraflo L [ΒΜΙm][PF6] 1% (0.5) 40 Zeuner et al.
(85%), MOPS (2011)
pH 6.0 (15%)
Ferulic acid Raffinose Depol [ΒΜΙm][PF6] 2% (144) 35 Couto et al.
740L (96%), (2011)
MES-NaOH
pH 6.0 (3%)
D-xylose 4.2% (144)
D- 2.8% (144)
Arabinose
D- 1% (144)
Galactose
a
After optimization of reaction conditions; +1: Immobilized with CLEAs methodology; AnFaeA: FAE type
A from A. niger; AnFaeC: FAE type C from Aspergillus nidulans; Ultraflo L/ Depol 740L: multienzymatic
preparation from H. insolens. Systems with conversion <1 % were not included
Table 5.5 Enzymatic synthesis of hydroxycinnamic acid esters in organic solvents reported in the literature
112

Hydroxycinnamate Enzyme Solvent system Yield (h) T (°C) References


Ferulic acid Glycerol FAE-PL Glycerol/ DMSO/acetate buffer pH 4.0 or 5.0 81%a (–) 50 Tsuchiyama et al.
(85:5:10 v/v/v) (2006)
Sinapinic acid n.q. (0.25)
3,4-Dimethoxy-cinnamic n.q. (0.25)
acid
p-Coumaric acid n.q. (0.25)
Methyl ferulate n.q. (0.25)
Ethyl ferulate n.q. (0.25)
Methyl sinapate n.q. (0.25)
Methyl n.q. (0.25)
3,4-dimethoxycinnamate
Arabinose n.q. (0.5)
Fructose n.q. (0.5)
Ferulic acid Galactose n.q. (0.5)
Glucose n.q. (0.5)
Xylose n.q. (0.5)
Arabinose n.q. (0.03)
Fructose n.q. (0.03)
Ethyl ferulate Galactose n.q. (0.03)
Glucose n.q. (0.03)
Xylose n.q. (0.03)
Methyl ferulate 1-Butanol Depol 1-Butanol/MOPS pH 6.0 (92.5:7.5 v/v) Up to 90 %a 37 Thorn et al. 2011
740L+2 (*192)
Depol Up to 40 %
740L (*192)
a
C. Katsimpouras et al.

After optimization of reaction conditions; +2: Immobilized in mesoporous silica MPS-9D; n.q.: detected but not quantified; FAE-PL: FAE from A. niger (purified from
the commercial preparation Pectinase PL “Amano”); Depol 740L: multienzymatic preparation from H. insolens. Systems with conversion <1 % were not included
5 Role and Applications of Feruloyl Esterases … 113

Novel FAEs purified from various filamentous fungi such as F. oxysporum,


S. thermophile, and A. niger have been used for the synthesis of alkyl ferulates
(1-propyl ferulate, 1-butyl ferulate, 1-pentyl-ferulate, 1-glyceryl ferulate) and
feruloylated-arabino-oligosaccharides (with up to six arabinose units) showing
regioselectivity for the primary hydroxyl group of the nonreducing arabinofuranose
ring (Table 5.3). In detergentless microemulsions, FoFaeI from F. oxysporum has
achieved the synthesis of various 1-butyl hydroxycinnamates exhibiting highest
yield on 1-butyl coumarate (up to 70 %), while FoFaeII has esterified p-hydro-
xyphenylacetic and p-hydroxyl-phenylpropionic acid with 1-propanol (70–75 %
yield) (Topakas et al. 2003a, b). The thermophilic StFaeC from S. thermophile has
synthesized 1-butyl hydroxycinnamates but also a variety of hydroxycinnamic acid
sugar esters, including the feruloylation of both L- and D-arabinose, showing no
chiral selectivity. The synthesized D-enantiomer successfully inhibited the growth of
Mycobacterium bovis BCG, indicating that FAEs are novel chemoenzymatic tools
for the synthesis of new drug (Vafiadi et al. 2007a). Multienzymatic preparations
containing FAE activity such as Ultraflo L and Depol 740L from Humicola insolens
have shown high yields up to 97 % in the transesterification of methyl ferulate to
1-butyl ferulate when immobilized with Crossed Linked Enzyme Aggregates
(CLEAs) methodology (Vafiadi et al. 2008b), while immobilized Depol 740L in
mesoporous silica MPS-90 supported significantly higher butyl ferulate yield up to
90 % comparing to the free enzyme (Thörn et al. 2011). Direct esterification of FA
with monomer sugars or oligosaccharides has been achieved by FAE multienzy-
matic preparations with relatively lower yields (up to 40 %; Couto et al. 2010).
The modification of sinapic acid, which is the second strongest antioxidant in
nature after FA, has also been achieved. AnFaeA transesterified methyl sinapate to
1- and 2-butyl sinapate, while StFaeC and FoFae-I have successfully synthesized
1-butyl sinapate in detergentless microemulsions (Topakas et al. 2003a, b; Vafiadi
et al. 2008a). Although, immobilized AnFaeA with CLEAs methodology resulted to
22 % lower conversion of methyl sinapate than that of the free enzyme. ILs have
been used for the synthesis of glyceryl-sinapate from sinapic acid and methyl
sinapate, where free and CLEAs AnFaeA also resulted to 76.5 and 72.5 % con-
version yield of sinapic acid, respectively (Vafiadi et al. 2009; Table 5.4). Butyl
esters of sinapic acid exhibit higher antioxidant activity against Low Density
Lipoprotein (LDL) oxidation comparing to their free acid or 1-butyl ferulate,
showing that sinapate esters are more promising antioxidants in vitro (Vafiadi et al.
2008a). On the contrary, sinapoylated glycerol has been shown to be a slower
antioxidant, when compared to free sinapic acid in LDL oxidation (Tsuchiyama
et al. 2006). It has not been established yet why hydroxycinnamate glycerols are
poorer antioxidants compared to their starting acids, although they are more
water-soluble than FA thus have a wider range of applications in water-based
industries, such as food, beverage and cosmetic industries. Sinapic acid and its
derivatives have the potential to be used in antiaging skin cosmetics, sun screen and
agents to improve DNA repair. They have also been implicated in cosmetic
preparations for the treatment of skin and dietary diseases and stimulation of hair
growth ailments (Faulds 2010).
114 C. Katsimpouras et al.

5.7 Conclusions

A clean technology for the sustainable production of biofuels and fine chemicals is
anticipated, utilizing the agricultural and agroindustrial residual biomass. The use of
enzymes is essential for the production and bioconversion of bioactive compounds
from waste residues resulting in a competitive price of biofuels through the
biorefinery concept. FAEs will have a profound role in utilizing waste biomass for
the production of energy and high-added value compounds. Since their discovery in
1990s, novel FAEs have been isolated and characterized showing different varia-
tions in physical characteristics that result in a diverse specificity for the hydrolysis
of model and natural substrates. In addition, many attempts have been made to
elucidate the functional relationships between sequence-diverse enzymes in respect
to their corresponding structure; however, the limited number of crystal structures
available complicates this mission. In this review, FAEs are shown to be involved
not only in the degradation of residual plant biomass, but also as tools for the
synthesis of novel bioactive components for use in health and cosmeceutical
industries. The improvements in molecular biology and process engineering permit
the overproduction of FAEs in different expression hosts, opening the way for their
commercial production that is prerequisite for many industrial applications.

Acknowledgments Paul Christakopoulos and Io Antonopoulou would like to thank the EU


Framework 7 project OPTIBIOCAT for the financial support.

References

Abokitse K, Wu M, Bergeron H, Grosse S, Lau PCK (2010) Thermostable feruloyl esterase for the
bioproduction of ferulic acid from triticale bran. Appl Microbiol Biotechnol 87:195–203
Barbe C, Dubourdieu D (1998) Characterisation and purification of a cinnamate esterase from
Aspergillus niger industrial pectinase preparation. J Sci Food Agric 78:471–478
Barr KA, Hopkins SA, Sreekrishna K (1992) Protocol for efficient secretion of HAS developed
from Pichia pastoris. Pharm Eng 12:48–51
Barron C, Surget A, Rouau X (2007) Relative amounts of tissues in mature wheat (Triticum aestivum
L.) grain and their carbohydrate and phenolic acid composition. J Cereal Sci 45:88–96
Bartolome B, Faulds CB, Kroon PA, Waldron K, Gilbert HJ, Hazlewood G, Williamson G (1997a)
An Aspergillus niger esterase (ferulic acid esterase III) and a recombinant Pseudomonas
fluorescens subsp. cellulosa esterase (XylD) released a 5-5′ dihydrodimer (diferulic acid) from
barley and wheat cell walls. Appl Environ Microbiol 63:208–212
Bartolome B, Faulds CB, Williamson G (1997b) Enzymic release of ferulic acid from barley spent
grain. J Cereal Sci 25:285–288
Bauer S, Vasu P, Persson S, Mort AJ, Somerville CR (2006) Development and application of a
suite of polysaccharide-degrading enzymes for analyzing plant cell walls. Proc Natl Acad
Sci USA 103:11417–11422
Benoit I, Navarro D, Marnet N, Rakotomanomana N, Lesage-Meessen L, Sigoillot JC, Asther M
(2006) Feruloyl esterases as a tool for the release of phenolic compounds from agro-industrial
by-products. Carbohydr Res 314:1820–1827
5 Role and Applications of Feruloyl Esterases … 115

Benoit I, Danchin EGJ, Bleinchodt RJ, de Vries RP (2008) Biotechnological applications and
potential fungal feruloyl esterases based on prevalence, classification and biochemical
diversity. Biotechnol Lett 30:387–396
Blum DL, Kataeva IA, Li X-L, Ljungdahl LG (2000) Feruloyl esterase activity of the Clostridium
thermocellum cellulosome can be attributed to previously unknown domains of XynY and
XynZ. J Bacteriol 182:1346–1351
Borneman WS, Ljungdahl LG, Hartley RD, Akin DE (1992) Purification and partial character-
ization of two feruloyl esterases from the anaerobic fungus Neocallimastix strain MC-2. Appl
Environ Microbiol 58:3762–3766
Borneman WS, Ljungdahl LG, Hartley RD, Akin DE (1993) Feruloyl and p-coumaroyl esterases
from the anaerobic fungus Neocallimastix strain MC-2: properties and functions in plant cell
wall degradation. In: Coughland MP, Hazlewood GP (eds) Hemicellulose and hemicellulases.
Portland Press, London and Chapel Hill, pp 85–102
Braga CMP, da Silva Delabona P, da Silva Lima DJ, Paixão DAA, da Cruz Pradella JG,
Farinas CS (2014) Addition of feruloyl esterase and xylanase produced on-site improves
sugarcane bagasse hydrolysis. Biores Technol 170:316–324
Bunzel M, Ralph J, Funk C, Steinhart H (2003) Isolation and identification of a ferulic acid
dehydrotrimer from saponified maize bran insoluble fiber. Eur Food Res Technol 217:128–133
Bunzel M, Ralph J, Funk C, Steinhart H (2005) Structural elucidation of new ferulic
acid-containing phenolic dimers and trimmers isolated from maize bran. Tetrahedron Lett
46:5845–5850
Bunzel M, Ralph J, Brüning P, Steinhart H (2006) Structural identification of dehydrotriferulic and
dehydrotetraferulic acids isolated from insoluble maize bran fiber. J Agric Food Chem
54:6408–6418
Butt MS, Tahir-Nadeem M, Ahmad Z, Sultan MT (2008) Xylanases and their applications in
baking industry. Food Technol Biotechnol 46:22–31
Caffal KH, Mohnen D (2009) The structure, function, and biosynthesis of plant cell wall pectic
polysaccharides. Carbohydr Res 344:1879–1900
Castanares A, McCrae SI, Wood TM (1992) Purification and properties of a feruloyl/ρ -coumaroyl
esterase from the fungus Penicillium pinophilum. Enzyme Microb Technol 14:875–884
Chen F, Dixon RA (2007) Lignin modification improves fermentable sugar yields for biofuel
production. Nat Biotechnol 25:759–761
Colquhoun IJ, Ralet MC, Thibault JF, Faulds CB, Williamson G (1994) Structure identification of
feruloylated oligosaccharides from sugar-beet pulp by NMR spectroscopy. Carbohydr Res
263:243–256
Couto J, Karboune S, Mathew R (2010) Regioselective synthesis of feruloylated glycosides using
the feruloyl esterase expressed in selected commercial multi-enzymatic preparations as
biocatalysts. Biocatal Biotransform 28:235–244
Couto J, St-Louis R, Karboune S (2011) Optimization of feruloyl esterase-catalyzed synthesis of
feruloylated oligosaccharides by response face methodology. J Mol Catal B Enzym 73:53–62
Couturier M, Haon M, Coutinho PM, Henrissat B, Lesage-Meesen L, Berrin JG (2011) Podospora
anserina hemicellulases potentiate the Trichoderma reesei secretome for saccharification of
lignocellulosic biomass. Appl Env Microbiol 77:237–246
Crepin VF, Faulds CB, Connerton IF (2003a) A non-modular type B feruloyl esterase from
Neurospora crassa exhibits concentration-dependent substrate inhibition. Biochem J 370:
417–427
Crepin VF, Faulds CB, Connerton IF (2003b) Production and characterization of the Talaromyces
stipitatus feruloyl esterase FAEC in Pichia pastoris: identification of the nucleophilic serine.
Protein Expr Purif 29:176–184
Crepin VF, Faulds CB, Connerton IF (2004a) Functional classification of the microbial feruloyl
esterases. Appl Microbiol Biotechnol 63:647–652
Crepin VF, Faulds CB, Connerton IF (2004b) Identification of a type-D feruloyl esterase from
Neurospora crassa. Appl Microbiol Biotechnol 63:567–570
116 C. Katsimpouras et al.

Damásio ARL, Braga CMP, Brenelli LB, Citadini AP, Mandelli F, Cota J, de Almeida RF,
Salvador VH, Paixao DAA, Segato F, Mercadante AZ, de Oliveira Neto M, do Santos WD,
Squina FM (2013) Biomass-to-bio-products application of feruloyl esterase from Aspergillus
clavatus. Appl Microbiol Biotechnol 97:6759–6767
Debeire P, Khoune P, Jeltsch JM, Phalip V (2012) Product patterns of a feruloyl esterase from
Aspergillus nidulans on large feruloyl-arabino-xylo-oligosaccharides from wheat bran.
Bioresour Technol 119:425–428
de Oliveira DM, Finger-Texeira A, Mota TR, Salvador VH, Moreira-Vilar FC, Molinari HBC,
Mitchell RAC, Marchiosi R, Ferrarese-Filho O, dos Santos WD (2014) Ferulic acid:
a key component in grass lignocellulose recalcitrance to hydrolysis. Plant Biotechnol J 13:
1224–1232
de Vries RP, Visser J (1999) Regulation of the feruloyl esterase (fae A) gene from Aspergillus
niger. Appl Environ Microbiol 65:5500–5503
de Vries RP, Michelsen B, Poulsen CH, Kroon PA, van den Heuvel RH, Faulds CB,
Williamson G, van den Hombergh JP, Visser J (1997) The faeA genes from Aspergillus niger
and Aspergillus tubingensis encode ferulic acid esterases involved in degradation of complex
cell wall polysaccharides. Appl Environ Microbiol 63:4638–4644
de Vries RP, van Kuyk PA, Kester HCM, Visser J (2002) The Aspergillus niger faeB gene
encodes a second feruloyl esterase involved in pectin and xylan degradation and is specifically
induced in the presence of aromatic compounds. Biochem J 363:377–386
Dien BS, Ximenes EA, O’Bryan PJ, Moniruzzaman M, Li X-L, Balan V, Dale B, Cotta MA
(2008) Enzyme characterization for hydrolysis of AFEX and liquid hot-water pretreated
distiller’s grains and their conversion to ethanol. Biores Technol 99:5216–5225
Donaghy JA, Bronnenmeier K, Soto-Kelly PF, McKay AM (2000) Purification and character-
ization of an extracellular feruloyl esterase from the thermophilic anaerobe Clostridium
stercorarium. J Appl Microbiol 88:458–466
Endler A, Persson S (2011) Cellulose synthases and synthesis in Arabidopsis. Mol Plant 4:
199–211
Esteban-Torres M, Reverón I, Mancheño JM, de las Rivas B, Muñoz R (2013) Characterization of
a feruloyl esterase from Lactobacillus plantarum. Appl Environ Microbiol 79:5130–5136
Faulds CB (2010) What can feruloyl esterases do for us? Phytochem Rev 9:121–132
Faulds CB, Williamson G (1991) The purification and characterization of
4-hydroxy-3-methoxycinnamic (ferulic) acid esterase from Streptomyces olivochromogenes.
J Gen Microbiol 137:2339–2345
Faulds C, Williamson G (1993) Ferulic acid esterase from Aspergillus niger: purification and
partial characterization of two forms from a commercial source of pectinase. Biotechnol Appl
Biochem 17:349–359
Faulds CB, Williamson G (1999) Review: the role of hydroxycinnamates in the plant cell wall.
J Sci Food Agric 79:393–395
Faulds CB, Mandalari G, Lo Curto RB, Bisignano G, Christakopoulos P, Waldron KW (2006)
Synergy between xylanases from glycoside hydrolase family 10 and family 11 and a feruloyl
esterase in the release of phenolic acids from cereal arabinoxylan. Appl Microbiol Biotechnol
71:622–629
Faulds CB, de Vries RP, Kroon PA, Visser J, Williamson G (1997) Influence of ferulic acid on the
production of feruloyl esterases by Aspergillus niger. FEMS Microbiol Lett 157:239–244
Fazary AE, Ju YH (2008) The large-scale use of feruloyl esterases in industry. Biotechnol Mol
Biol Rev 3:95–110
Fazary AE, Hamad HA, Lee JC, Koskei T, Lee CK, Ju YH (2010) Expression of feruloyl esterase
from Aspergillus awamori in Escherichia coli: characterization and crystal studies of the
recombinant enzyme. Int J Biol Macromol 46:440–444
Ferreira LMA, Wood TM, Williamson G, Faulds CB, Hazlewood G, Gilbert HJ (1993) A modular
esterase from Pseudomonas fluorescens subsp. cellulosa contains a non-catalytic binding
domain. Biochem J 294:349–355
5 Role and Applications of Feruloyl Esterases … 117

Fillingham IJ, Kroon PA, Williamson G, Gilbert HJ, Hazlewood GP (1999) A modular cinnamoyl
ester hydrolase from the anaerobic fungus Piromyces equi acts synergistically with xylanase
and is part of a multiprotein cellulose-binding cellulase-hemicellulase complex. Biochem J 343
Pt 1:215–224
Ford CW, Hartley RD (1990) Cyclodimers of p-coumaric and ferulic acids in the cell walls of
tropical grasses. J Sci Food Agric 50:29–43
Fry SC (1983) Feruloylated pectins from the primary cell wall: their structure and possible
functions. Planta 157:111–123
Fry SC, Willis SC, Paterson AEJ (2000) Intraprotoplasmic and wall-localised formation of
arabinoxylan bound diferulates and larger ferulate coupling-products in maize cell-suspension
cultures. Planta 211:679–692
Funk C, Ralph J, Steinhart H, Bunzel M (2005) Isolation and structural characterization of
8-O-4/8-O4- and 8-8/8-O4-coupled dehydrotriferulic acids from maize bran. Phytochemistry
66:363–371
Garcia-Conesa M-T, Crepin VF, Goldson AJ, Williamson G, Cummings NJ, Connerton IF,
Faulds CB, Kroon PA (2004) The feruloyl esterase system of Talaromyces stipitatus:
production of three discrete feruloyl esterases, including a novel enzyme, TsFaeC, with a broad
substrate specificity. J Biotechnol 108:227–241
Garleb KA, Fahey GC Jr, Lewis SM, Kerley MS, Montgomery L (1988) Chemical composition
and digestibility of fibre fractions of certain by-product feedstuffs fed to ruminants. J Anim Sci
66:2650–2662
Giuliani S, Piana C, Setti L, Hochkoeppler A, Pifferi PG, Williamson PG, Faulds CB (2001)
Synthesis of pentylferulate by a feruloyl esterase from Aspergillus niger using water-in-oil
microemulsions. Biotechnol Lett 23:325–330
Goldstone DC, Villas-Bôas SG, Till M, Kelly WJ, Attwood GT, Arcus VL (2010) Structural and
functional characterization of a promiscuous feruloyl esterase (Est1E) from the rumen
bacterium Butyrivibrio proteoclasticus. Proteins 78:1457–1469
Gong YY, Yin X, Zhang HM, Wu MC, Tang CD, Wang JQ, Pang QF (2013) Cloning, expression
of a feruloyl esterase from Aspergillus usamii E001 and its applicability in generating ferulic
acid from wheat bran. J Ind Microbiol Biotechnol 40:1433–1441
Gorke J, Srienc F, Kazlauskas R (2010) Towards advances ionic liquids. Polar enzyme-friendly
solvents for biocatalysis. Biotechnol Bioproc Eng 15:40–53
Gottschalk LMF, Oliveira RA, da Silva Bon EP (2010) Cellulases, xylanases, β-glucosidase and
ferulic acid esterase produced by Trichoderma and Aspergillus act synergistically in the
hydrolysis of sugarcane bagasse. Biochem Eng J 51:72–78
Gottschalk LMF, de Sousa Paredes R, Teixeira RSS, da Silva ASA, da Silva Bon EP (2013)
Efficient production of lignocellulolytic enzymes xylanase, β-xylosidase, ferulic acid esterase
and β-glucosidase by the mutant strain Aspergillus awamori 2B.361 U2/1. Braz J Microbiol
44:569–576
Grinna LS, Tschopp JF (1989) Size distribution and general structural features of N-linked
oligosaccharides from the methylotrophic yeast, Pichia pastoris. Yeast 5:107–115
Haase-Aschoff P, Linke D, Berger RG (2013) Detection of feruloyl- and cinnamoyl esterases from
basidiomycetes in the presence of interfering laccase. Bioresour Technol 130:231–238
Hassan S, Hugouvieux-Cotte-Pattat N (2011) Identification of two feruloyl esterases in Dickeya
dadantii 3937 and induction of the major feruloyl esterase and of pectate lyases by ferulic acid.
J Bacteriol 193:963–970
Hegde S, Muralikrishna G (2009) Isolation and partial characterization of alkaline feruloyl
esterases from Aspergillus niger CFR 1105 grown on wheat bran. World J Microbiol
Biotechnol 25:1963–1969
Hemery Y, Lullien-Pellerin V, Rouau X, Abecassis J, Samson MF, Åman P, von Reding W,
Spoerndli C, Barron C (2009) Biochemical markers: efficient tools for the assessment of wheat
grain tissue proportions in milling fractions. J Cereal Sci 49:55–64
118 C. Katsimpouras et al.

Hermoso JA, Sanz-Aparicio J, Molina R, Juge N, Gonzalez R, Faulds CB (2004) The crystal
structure of feruloyl esterase A from Aspergillus niger suggests evolutive functional
convergence in feruloyl esterase family. J Mol Biol 338:495–506
Holmquist M (2000) Alpha/beta-hydrolase fold enzymes: structures, functions and mechanisms.
Curr Protein Pept Sci 1:209–235
Horn SJ, Vaaje-Kolstad G, Westereng B, Eijink VGH (2012) Novel enzymes for the degradation
of cellulose. Biotechnol Biofuels 5:45
Howard RL, Abotsi E, Jansen van Rensburg EL, Howard S (2003) Lignocellulose biotechnology:
issues of bioconversion and enzyme production. Afr J Biotechnol 2:602–619
Lai K-K, Stogios PJ, Vu C, Xu X, Cui H, Molloy S, Savchenko A, Yakunin A, Gonzalez CF
(2011) An inserted α/β subdomain shapes the catalytic pocket of Lactobacillus johnsonii
cinnamoyl esterase. PLoS ONE 6(8):e23269
Iiyama K, Lam TBT, Stone BA (1990) Phenolic acid bridges between polysaccharides and lignin
in wheat internodes. Phytochemistry 29:733–737
Ishii T (1997) Structure and functions of feruloylated polysaccharides. Plant Sci 127:111–127
Ishii T, Hiroi T, Thomas JR (1990) Feruloylated xyloglucan and p-coumaroyl arabinoxylan
oligosaccharides from bamboo shoot cell-walls. Phytochemistry 29:1999–2003
Johnson KG, Silva MC, MacKenzie CR, Schneider H, Fontana JD (1989) Microbial degradation
of hemicellulosic materials. Appl Biochem Biotechnol 20–21:245–258
Kanauchi M, Watanabe S, Tsukada T, Atta K, Kakuta T, Koizumi T (2008) Purification and
characteristics of feruloyl esterase from Aspergillus awamori G-2 strain. J Food Sci 73:
458–463
Katsimpouras C, Christakopoulos P, Topakas E (2014) Fermentation and enzymes. In:
Varzakas T, Tzia C (eds) Food engineering handbook. CRC Press, Boca Raton, pp 495–497
Kheder F, Delaunay S, Abo-Chameh G, Paris C, Muniglia L, Girardin M (2009) Production and
biochemical characterization of a type B ferulic acid esterase from Streptomyces ambofaciens.
Can J Microbiol 55:729–738
Khmelnitsky YL, Hilhorst R, Veeger C (1988) Detergentless microemulsions as media for
enzymatic reactions. Eur J Biochem 176:265–271
Kim Y, Hendrickson R, Mosier NS, Ladisch MR, Bals B, Balan V, Dale BE (2008) Enzyme
hydrolysis and ethanol fermentation of liquid hot water and AFEX pretreated distiller’s grains
at high-solids loadings. Biores Technol 99:5206–5215
Kin KL, Lorca GL, Gonzalez CF (2009) Biochemical properties of two cinnamoyl esterases
purified from a Lactobacillus johnsonii strain isolated from stool samples of diabetes-resistant
rats. Appl Environ Microbiol 75:5018–5024
Knoshaug EP, Selig MJ, Baker JO, Decker SR, Himmel ME, Adney WS (2008) Heterologous
expression of two ferulic acid esterases from Penicillium funiculosum. Appl Biochem
Biotechnol 146:79–87
Koseki T, Furuse S, Iwano K, Matsuzawa H (1998) Purification and characterization of a
feruloylesterase from Aspergillus awamori. Biosci Biotechnol Biochem 62:2032–2034
Koseki T, Fushinobu S, Shirakawa AH, Komai M (2009) Occurrence, properties and application
of feruloyl esterases. Appl Microbiol Biotechnol 84:803–810
Koseki T, Takahashi K, Fushinobu S, Iefuji H, Iwano K, Hashizume K, Matsuzawa H (2005)
Mutational analysis of a feruloyl esterase from Aspergillus awamori involved in substrate
discrimination and pH dependence. Biochim Biophys Acta—Gen Subj 1722:200–208
Kroon PA, Williamson G (1996) Release of ferulic acid from sugar-beet pulp by using
arabinanase, arabinofuranosidase and an esterase from Aspergillus niger. Biotechnol Appl
Biochem 23:263–267
Kroon PA, Faulds CB, Brezillon C, Williamson G (1997) Methyl phenylalkanoates as substrates to
probe the active sites of esterases. Eur J Biochem 248:245–251
Kroon PA, Williamson G, Fish NM, Archer DB, Belshaw NJ (2000) A modular esterase from
Penicillium funiculosum which releases ferulic acid from plant cell walls and binds crystalline
cellulose contains a carbohydrate binding module. Eur J Biochem 267:6740–6752
5 Role and Applications of Feruloyl Esterases … 119

Kühnel S, Pouvreau L, Appeldoorn MM, Hinz SWA, Schols HA, Gruppen H (2012) The ferulic
acid esterases of Chrysosporium lucknowense C1: purification, characterization and their
potential application in biorefinery. Enzyme Microb Tech 50:77–85
Lambertz C, Garvey M, Klinger J, Heesel D, Klose H, Fischer R, Commandeur U (2014)
Challenges and advances in the heterologous expression of cellulolytic enzymes: a review.
Biotechnol Biofuels 7:135
Larsen J, Østergaard Haven M, Thirup L (2012) Inbicon makes lignocellulosic ethanol a
commercial reality. Biomass Bioenergy 46:36–45
Lenfant N, Hotelier T, Velluet E, Bourne Y, Marchot P, Chatonnet A (2013) ESTHER, the
database of the alpha/beta-hydrolase fold superfamily of proteins: tools to explore diversity of
functions. Nucleic Acids Res 41:D423–D429
Levasseur A, Gouret P, Lesage-Meessen L, Asther M, Asther M, Record E, Pontarotti P (2006)
Tracking the connection between evolutionary and functional shifts using the fungal
lipase/feruloyl esterase A family. BMC Evol Biol 6:92
Li J, Cai S, Luo Y, Dong X (2011) Three feruloyl esterases in Cellulosilyticum ruminicola H1 act
synergistically to hydrolyze esterified polysaccharides. Appl Environ Microbiol 77:6141–6147
Linke D, Matthes R, Nimtz M, Zorn H, Bunzel M, Berger RG (2013) An esterase from the
basidiomycete Pleurotus sapidus hydrolyzes feruloylated saccharides. Appl Microbiol
Biotechnol 97:7241–7251
Lombard V, Ramulu HG, Drula E, Coutinho PM, Henrissat B (2013) The carbohydrate-active
enzymes database (CAZy) in 2013. Nucl Acids Res 42:D490–D495
MacKenzie CR, Bilous D (1988) Ferulic acid esterase activity from Schizophyllum commune.
Appl Environ Microbiol 54:1170–1173
Mackenzie CR, Bilous D, Schneider H, Johnson KG (1987) Induction of cellulolytic and
xylanolytic enzyme systems in Streptomyces spp. Appl Environ Microbiol 53:2835–2839
Malinovsky FG, Fangel JU, Willats WGT (2014) The role of the cell wall in plant immunity. Front
Plant Sci 5:1–12
Mandalari G, Bisignano G, Lo Curto RB, Waldron KW, Faulds CB (2008) Production of feruloyl
esterases and xylanases by Talaromyces stipitatus and Humicola grisea var. thermoidea on
industrial food processing by-products. Biores Technol 99:5130–5133
Mathew S, Abraham TE (2005) Studies on the production of feruloyl esterase from cereal brans
and sugar cane bagasse by microbial fermentation. Enzyme Microb Technol 36:565–570
McClendon SD, Shin HD, Chen RR (2011) Novel bacterial ferulic acid esterase from Cellvibrio
japonicus and its application in ferulic acid release and xylan hydrolysis. Biotechnol Lett
33:47–54
McCrae SI, Leith KM, Gordon AH, Wood TM (1994) Xylan-degrading enzyme system produced
by the fungus Aspergillus awamori: isolation and characterization of a feruloyl esterase and a
p-coumaroyl esterase. Enzyme Microb Technol 16:826–834
Moukouli M, Topakas E, Christakopoulos P (2008) Cloning, characterization and functional
expression of an alkalitolerant type C feruloyl esterase from Fusarium oxysporum. Appl
Microbiol Biotechnol 79:245–254
Mueller-Harvey I, Hartley RD, Harris J, Curzon EH (1986) Linkage of p-coumaroyl and feruloyl
groups to cell-wall polysaccharides of barley straw. Carbohydr Res 148:71–85
Mukherjee G, Singh RK, Mitra A, Sen SK (2007) Ferulic acid esterase production by
Streptomyces sp. Biores Technol 98:211–213
Nardini M, Dijkstra BW (1999) Alpha/beta hydrolase fold enzymes: the family keeps growing.
Curr Opin Struct Biol 9:732–737
Nguyen D, Zhang X, Jiang ZH, Audet A, Paice MG, Renaud S, Tsang A (2008) Bleaching of kraft
pulp by a commercial lipase: accessory enzymes degrade hexenuronic acids. Enz Microbial
Technol 43:130–136
Nieter A, Haase-Aschoff P, Linke D, Nimtz M, Berger RG (2014) A halotolerant type A feruloyl
esterase from Pleurotus eryngii. Fungal Biol 118:348–357
Nordkvisk E, Salomonsson AC, Amar P (1984) Distribution of insoluble bound phenolic acids in
barley grain. J Sci Food Agric 35:657–661
120 C. Katsimpouras et al.

Olivares- Hernández R, Sunner H, Frisvad JC, Olsson L, Nielsen J, Panagiotou G (2010)


Combining substrate specificity analysis with support vector classifiers reveals feruloyl esterase
as a phylogenetically informative protein group. PLoS ONE 5:e12781
Olsson L, Panagiotou G, Christakopoulos P, Topakas E, Olivares R (2011) Nutraceutical
compounds: feruloyl esterases as biosynthetic tools. Encycl Biotechnol Agric Food 1:448–453
Oosterveld A, Grabber JH, Beldman G, Ralph J, Voragen AGJ (1997) Formation of ferulic acid
dehydrodimers through oxidative cross-linking of sugar beet pectin. Carbohydr Res 300:
179–181
Panagiotou G, Granouillet P, Olsson L (2006) Production and partial characterization of
arabinoxylan-degrading enzymes by Penicillium brasilianum under solid-state fermentation.
Appl Microbiol Biotechnol 72:1117–1124
Poidevin L, Levasseur A, Paës G, Navarro D, Heiss-Blanquet S, Asther M, Record E (2009)
Heterologous production of the Piromyces equi cinnamoyl esterase in Trichoderma reesei for
biotechnological applications. Lett Appl Microbiol 49:673–678
Prates JAM, Tarbouriech N, Charnock SJ, Fontes CMGA, Ferreira LMA, Davies GJ (2001) The
structure of the feruloyl esterase module of xylanase 10B from Clostridium thermocellum
provides insights into substrate recognition. Structure 9:1183–1190
Priefert H, Rabenhorst J, Steinbüchel A (2001) Biotechnological production of vanillin. Appl
Microbiol Biotechnol 56:296–314
Qi M, Wang P, Selinger LB, Yanke LJ, Forster RJ, McAllister TA (2011) Isolation and
characterization of a ferulic acid esterase (Fae1A) from the rumen fungus Anaeromyces
mucronatus. J Appl Microbiol 110:1341–1350
Rakotoarivonina H, Hermant B, Chabbert B, Touzel JP, Remond C (2011) A thermostable
feruloyl-esterase from the hemicellulolytic bacterium Thermobacillus xylanilyticus releases
phenolic acids from non-pretreated plant cell walls. Appl Microbiol Biotechnol 90:541–552
Ralph J, Quideau S, Grabber JH, Hatfield RD (1994) Identification and synthesis of new ferulic
acid dehydrodimers present in grass cell walls. J Chem Soc Perkin Trans 1:3485–3498
Ralph J, Grabber J, Hatfield RD (1995) Lignin-ferulate crosslinks in grasses: active incorporation
of ferulate polysaccharide esters into ryegrass lignins. Carbohydr Res 275:167–178
Record E, Asther M, Sigoillot C, Pages S, Punt PJ, Delattre M, Haon M, van den Hondel CAMJJ,
Sigoillot JC, Lesage-Meessen L, Asther M (2003) Overproduction of the Aspergillus niger
feruloyl esterase for pulp bleaching application. Appl Microbiol Biotechnol 62:349–355
Rouaou X, Cheynier V, Surget A, Gloux D, Barron C, Meudec E, Louis-Montero J, Criton M
(2003) A dehydrotrimer of ferulic acid from maize bran. Phytochemistry 63:899–903
Rumbold K, Biely P, Mastihubova M, Gudelj M, Gubitz G, Robra K-H, Prior BA (2003)
Purification and properties of a feruloyl esterase involved in lignocellulose degradation by
Aureobasidium pullulans. Appl Environ Microbiol 69:5622–5626
Saha BC (2003) Hemicellulose bioconversion. J Ind Microbiol Biotechnol 30:279–291
Saulnier L, Vigouroux L, Thibault JF (1995) Isolation and partial characterization of feruloylated
oligosaccharides from maize bran. Carbohydr Res 272:241–253
Scheller HV, Ulvskov P (2010) Hemicelluloses. Annu Rev Plant Biol 61:263–289
Schubot FD, Kataeva IA, Blum DL, Shah AK, Ljungdahl LG, Rose JP, Wand BC (2001)
Structural basis for the substrate specificity of the feruloyl esterase domain of the cellulosomal
xylanase Z from Clostridium thermocellum. Biochemistry 40:12524–12532
Selig MJ, Knoshaug EP, Adney WS, Himmel ME, Decker SR (2008) Synergistic enhancement of
cellobiohydrolase performance on pretreated corn stover by addition of xylanase and esterase
activities. Biores Technol 99:4997–5005
Shibuya N (1984) Phenolic acids and their carbohydrate esters in rice endosperm cell walls.
Phytochemistry 23:2233–2237
Shin HD, Chen RR (2006) Production and characterization of a type B feruloyl esterase from
Fusarium proliferatum NRRL 26517. Enzyme Microb Technol 38:478–485
Shin HD, Chen RR (2007) A type B feruloyl esterase from Aspergillus nidulans with broad pH
applicability. Appl Microbiol Biotechnol 73:1323–1330
5 Role and Applications of Feruloyl Esterases … 121

Sigoillot C, Camareto S, Vidal T, Record E, Asther M, Boada MP, Martinze MJ, Sigoillot JC,
Asther M, Colom JF, Martinez AT (2005) Comparison of different fungal enzymes from
bleaching high-quality paper pulps. J Biotechnol 115:333–343
Smith MM, Hartley RD (1983) Occurrence and nature of ferulic acid substitution of cell-wall
polysaccharides in graminaceous plants. Carbohydr Res 118:65–80
Sultana R, Ravagna A, Mohmmad-Abdul H, Calabrese V, Butterfield DA (2005) Ferulic acid ethyl
ester protects neurons against amyloid beta-peptide (1-42)-induced oxidative stress and
neurotoxicity: relationship to antioxidant activity. J Neurochem 92:749–758
Suzuki K, Hori A, Kawamoto K, Thangudu RR, Ishida T, Igarashi K, Samejima M, Yamada C,
Arakawa T, Wakagi T, Koseki T, Fushinobu S (2014) Crystal structure of a feruloyl esterase
belonging to the tannase family: a disulfide bond near a catalytic triad. Proteins 82:2857–2867
Tabka MG, Herpoël-Gimbert I, Monod F, Asther M, Sigoillot JC (2006) Enzymatic sacchari-
fication of wheat straw for bioethanol production by a combined cellulose xylanase and
feruloyl esterase treatment. Enz Microbial Technol 39:897–902
Tai ES, Hsieh PC, Sheu SC (2014) Effect of polygalacturonase and feruloyl esterase from
Aspergillus tubingensis on demucilage and quality of coffee beans. Process Biochem 49:
1274–1280
Tenkanen M, Schuseil J, Puls J, Poutanen K (1991) Production, purification and characterisation of
an esterase liberating phenolic acids from lignocellulosics. J Biotechnol 18:69–84
Thakur VV, Jain RK, Mathur RM (2012) Studies on xylanase and laccase enzymatic prebleaching
to reduce chlorine-based chemicals during CEH and ECF bleaching. Bioresources 7:
2220–2235
Thörn C, Gustafsson H, Olsson L (2011) Immobilization of feruloyl esterases in mesoporous
materials leads to improved transesterification yield. J Mol Catal B Enzym 72:57–64
Topakas E, Christakopoulos P (2004) Production and partial characterization of alkaline feruloyl
esterases by Fusarium oxysporum during submerged batch cultivation. World J Microb Biot
20:245–250
Topakas E, Stamatis H, Mastihubova M, Biely P, Kekos D, Macris BJ, Christakopoulos P (2003a)
Purification and characterization of a Fusarium oxysporum feruloyl esterase (FoFAE-I)
catalysing transesterification of phenolic acid esters. Enz Microbial Technol 33:729–737
Topakas E, Stamatis H, Biely P, Kekos D, Macris BJ, Christakopoulos P (2003b) Purification and
characterization of a feruloyl esterase from Fusarium oxysporum catalyzing esterification of
phenolic acids in ternary water-organic solvent mixtures. J Biotechnol 102:33–44
Topakas E, Kalogeris E, Kekos D, Macris BJ, Christakopoulos P (2003c) Production and partial
characterization of feruloyl esterase by Sporotrichum thermophile under solid-state fermen-
tation. Process Biochem 38:1539–1543
Topakas E, Stamatis H, Biely P, Christakopoulos P (2004) Purification and characterization of a
type B feruloyl esterase (StFae-A) from the thermophilic fungus Sporotrichum thermophile.
Appl Microbiol Biotechnol 63:686–690
Topakas E, Christakopoulos P, Faulds CB (2005a) Comparison of mesophilic and thermophilic
feruloyl esterases: characterization of their substrate specificity for methyl phenylalkanoates.
J Biotechnol 115:355–366
Topakas E, Vafiadi C, Stamatis H, Christakopoulos P (2005b) Sporotrichum thermophile type C
feruloyl esterase (StFaeC): purification, characterization and its use for phenolic acid (sugar)
ester synthesis. Enz Microbial Technol 36:729–736
Topakas E, Vafiadi C, Christakopoulos P (2007) Microbial production, characterization and
applications of feruloyl esterases. Proc Biochem 42:497–509
Topakas E, Moukouli M, Dimarogona M, Christakopoulos P (2012) Expression, characterization
and structural modelling of a feruloyl esterase from the thermophilic fungus Myceliophthora
thermophila. Appl Microbiol Biotechnol 94:399–411
Tsuchiyama M, Sakamoto T, Fujita T, Murata S, Kawasaki H (2006) Esterification of ferulic acid
with polyols using a ferulic acid esterase from Aspergillus niger. Biochim Biophys Acta
1760:1071–1079
122 C. Katsimpouras et al.

Udatha DBRKG, Kouskoumvekaki I, Olsson L, Panagiotou G (2011) The interplay of


descriptor-based computational analysis with pharmacophore modeling builds the basis for a
novel classification scheme for feruloyl esterases. Biotechnol Adv 29:94–110
Uraji M, Arima J, Inoue Y, Harazono K, Hatanaka T (2014) Application of two newly identified
and characterized feruloyl esterases from Streptomyces sp. in the enzymatic production of
ferulic acid from agricultural biomass. PLoS ONE 9(8):e104584
Vafiadi C, Topakas E, Wong KKY, Suckling ID, Christakopoulos P (2005) Mapping the
hydrolytic and synthetic selectivity of a type C feruloyl esterase (StFaeC) from Sporotrichum
thermophile using alkyl ferulates. Tetrahydron: Asymmetry 16:373–379
Vafiadi C, Topakas E, Christakopoulos P (2006a) Regioselective esterase-catalyzed feruloylation
of L-arabinodiose. Carbohydr Res 341:1992–1997
Vafiadi C, Topakas E, Christakopoulos P, Faulds CB (2006b) The feruloyl esterase system of
Talaromyces stipitatus: determining the hydrolytic and synthetic specificity of TsFaeC.
J Biotechnol 125:210–221
Vafiadi C, Topakas E, Alderwick LJ, Besra GS, Christakopoulos P (2007a) Chemoenzymatic
synthesis of feruloyl-D-arabinose as a potential anti-mycobacterial agent. Biotechnol Lett
29:1771–1774
Vafiadi C, Topakas E, Bakx EJ, Schols HA, Christakopoulos P (2007b) Structural characterization
of ESI-MS of feruloylated arabino-oligosaccharides synthesized by chemoenzymatic esterifi-
cation. Molecules 12:1367–1375
Vafiadi C, Topakas E, Alissandratos A, Faulds CB, Christakopoulos P (2008a) Enzymatic
synthesis of butyl hydroxycinnamates and their inhibitory effect on LDL-oxidation.
J Biotechnol 133:497–504
Vafiadi C, Topakas E, Christakopoulos P (2008b) Preparation of multipurpose cross-linked
enzyme aggregates and their application to production of alkyl ferulates. J Mol Catal B Enzym
54:35–41
Vafiadi C, Topakas E, Nahmias VR, Faulds CB, Christakopoulos P (2009) Feruloyl
esterase-catalyzed synthesis of glycerol sinapate using ionic liquid mixtures. J Biotechnol
139:124–129
Valls C, Colom JF, Baffert C, Gimbert I, Roncero MB (2010) Comparing the efficiency of the
laccase-NHA and laccase-HBT systems in eucalyptus pulp bleaching. Biochem Eng J 49:
401–407
Vanholme R, Demedts B, Morreel K, Ralph J, Boerjan W (2010) Lignin biosynthesis and
structure. Plant Physiol 153:895–905
Várnai A, Tang C, Bengtsson O, Atterton A, Mathiesen G, Eijsink GH (2014) Expression of
endoglucanases in Pichia pastoris under control of the GAP promoter. Microb Cell Fact 13:57
Vogt T (2010) Phenylpropanoid Biosynthesis. Mol Plant 3:2–20
Walton NJ, Narbad A, Faulds C, Williamson G (2000) Novel approaches to the biosynthesis of
vanillin. Curr Opin Biotechnol 11:490–496
Wang X, Geng X, Egashira Y, Sanada H (2004) Purification and characterization of a feruloyl
esterase from the intestinal bacterium Lactobacillus acidophilus. Appl Environ Microbiol
70:2367–2372
Wang L, Zhang R, Ma Z, Wang H, Ng T (2014) A feruloyl esterase (FAE) characterized by
relatively high thermostability from the edible mushroom Russula virescens. Appl Biochem
Biotechnol 172:993–1003
Wong D (2008) Enzymatic deconstruction of backbone structures of the ramified regions in
pectins. Protein J 27:30–42
Wong DWS, Chan VJ, Batt SB, Sarath G, Liao H (2011) Engineering Saccharomyces cerevisiae
to produce feruloyl esterase for the release of ferulic acid from switchgrass. J Ind Microbiol
Biotechnol 38:1961–1967
Wong DWS, Chan VJ, Liao H, Zidwick MJ (2013) Cloning of a novel feruloyl esterase gene from
rumen microbial metagenome and enzyme characterization in synergism with endoxylanases.
J Ind Microbiol Biotechnol 40:287–295
5 Role and Applications of Feruloyl Esterases … 123

Wu M, Abokitse K, Grosse S, Leisch H, Lau PCK (2012) New feruloyl esterases to access
phenolic acids from grass biomass. Appl Biochem Biotechnol 168:129–143
Yang SQ, Tang L, Yan QJ, Zhou P, Xu HB, Jiang ZQ, Zhang P (2013) Biochemical characteristics
and gene cloning of a novel thermostable feruloyl esterase from Chaetomium sp. J Mol Catal B
Enzym 97:328–336
Yao J, Chen QL, Shen AX, Cao W, Liu YH (2013) A novel feruloyl esterase from a soil
metagenomic library with tannase activity. J Mol Catal B Enzym 95:55–61
Yu P, Maenz DD, McKinnon JJ, Racz VJ, Christensen DA (2002a) Release of ferulic acid from
oat hulls by Aspergillus ferulic acid esterase and Trichoderma xylanase. J Agric Food Chem
50:1625–1630
Yu P, McKinnon JJ, Maenz DD, Racz VJ, Christensen DA (2002b) The interactive effects of
enriched sources of Aspergillus ferulic acid esterase and Trichoderma xylanase on the
quantitative release of hydroxycinnamic acids from oat hulls. Can J Anim Sci 82:251–257
Yu P, McKinnon JJ, Maenz DD, Olkowski AA, Racz VJ, Christensen DA (2003) Enzymic release
of reducing sugars from oat hulls by cellulase, as influenced by Aspergillus ferulic acid esterase
and Trichoderma xylanase. J Agric Food Chem 51:218–223
Zeng Y, Yin X, Wu MC, Yu T, Feng F, Zhu TD, Pang QF (2014) Expression of a novel feruloyl
esterase from Aspergillus oryzae in Pichia pastoris with esterification activity. J Mol Catal B
Enzym 110:140–146
Zeuner B, Stahlberg T, van Buu ON, Kurov-Kruse AJ, Riisager A, Meyer AS (2011) Dependency
of the hydrogen bonding capacity of the solvent anion on the thermal stability of feruloyl
esterases in ionic liquid systems. Green Chem 13:1550–1557
Zhang SB, Zhai HC, Wang L, Yu GH (2013) Expression, purification and characterization of a
feruloyl esterase A from Aspergillus flavus. Prot Expr Pur 92:36–40
Zwane EN, Rose SH, Van Zyl WH, Rumbold K, Viljoen-Bloom M (2014) Overexpression of
Aspergillus tubingensis faeA in protease-deficient Aspergillus niger enables ferulic acid
production from plant material. J Ind Microbiol Biotechnol 41:1027–1034
Chapter 6
Endo-b-1,4-xylanase: An Overview
of Recent Developments

Alexandre Gomes Rodrigues

Abstract This chapter presents the main important aspects about the enzyme
endo-beta-1,4-xylanase. It starts with the biological grounds for the use of the
enzyme. Moving on, the advancements in the production process and optimisation
are substantially described, along with the microorganisms and a broad range of
molecular techniques used to obtain best performance enzyme. Finally, it delineates
the most common applications related to xylanase, such as in biobleaching, baking,
feedstock and the enzymatic role in juice and ink industries, among others.
Furthermore, the material brings updated research performed during the last few
years combined with essential established facts along the time.

6.1 Introduction

Since ancient times the mankind has made used of enzymes to produce goods as
wine, beer and bread. The gained knowledge in fields as chemistry, biology,
engineering and biotechnology have enabled more precise and efficient processes to
obtain and apply such biomolecules not only in the existing products but also to
create new ones, exploring fully the potential of a material and expanding its roles.
In this regard, plants serve us in many aspects, with their fundamental biological
and microbiological functions, and have been increasingly exploited to provide
valuable biomaterials, which are employed to boost advancements in daily life with
better and suitable products, while reducing the damage caused to the environment
thereby making our lives more comfortable. Hemicelluloses are the second most
present polysaccharide in plant cell wall and are made up of important polymers as
xylan (Schaedel et al. 2010). Xylanases are enzymes secreted by microorganisms,
mostly fungi and bacteria are used in a broad range of important manufacturing
pathways in the papermaking industry as well as in the food industry and in the

A.G. Rodrigues (&)


Institute of Pharmacy, Martin-Luther University Halle-Wittenberg,
Halle, Germany
e-mail: [email protected]

© Springer International Publishing Switzerland 2016 125


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_6
126 A.G. Rodrigues

production of chemicals such as xylitol, in addition to their natural function. Due to


its key role the xylanases have reached an outstanding market position. The esti-
mative was that the market in which the enzyme is present represents about 200
million dollars (Sonia et al. 2005). In 2005, xylanases along with cellulases and
pectinases sliced 20 % of the global industrial enzyme market (Polizeli et al. 2005).
The innumerous applications of xylanases have raised interest and attracted many
researches and resources towards its whole production chain (Wong et al. 1988;
Coughlan and Hazlewood 1993; Beg et al. 2001; Harris and Ramalingam 2010; Juturu
and Wu 2012). Xylanase transforms water insoluble hemicellulose into soluble form,
which binds water in the dough, therefore decreasing the dough firmness, increasing
volume and creating finer and more uniform crumbs (Butt et al. 2008). The enzymes
break down xylan into oligosaccharides, lowering the viscosity, resulting in more
appropriate processed food and drink, as dough in baking, in animal feed to increase
digestibility, wood pulp, liquid fuels, solvents and other chemicals (Bhat 2000; Butt
et al. 2008). The current technological knowledge and available facilities provide
means by which the processes regarding the obtaining of xylanases and process
optimisation are improved. A wide range of techniques from molecular biology to
analytical chemistry and biochemical engineering have been reported as enabling
researchers to progress and provide better products continuously.
This chapter comprises the fundamental aspects related to endo-b-1,4-xylanase,
namely its early appearance and applications which have encouraged much effort
and investment, along with the most recent studies conducted worldwide and their
main outcomes leading relevant improvements and achievements in the field.

6.2 Plant Cell Wall Structure

Plant cell wall contains a variable amount of different chemical compounds making
up their structures. The cell wall itself is composed of three layers. The compounds
present in a plant cell wall are cellulose and hemicellulose, pectin and lignocellu-
lose. After cellulose, hemicellulose is the most abundant natural polysaccharide in
plants and accounts to one-third of plant biomass. However, it varies according to
plant species due to environmental conditions (Caffall and Mohnen 2009; Schaedel
et al. 2010; Yin et al. 2010). Hemicellulose contributes not only to biological
processes in nature but also in our daily life, through the employment of its com-
ponents in several industrial sectors from feedstock to paper making and bread
baking (Yin et al. 2010).
Schulze (1891) first introduced the term ‘hemicellulose’ for the fractions isolated
or extracted from plant materials with diluted alkali. It is an amorphous
heteropolymer formed by aldopentoses (xylose and arabinose) and aldohexoses
(glucose, galactose, mannose), forming a short polymer chain presenting lower
molar mass, when compared to cellulose (Jacobsen 2000). It is present in hardwood
(angiosperm) as well as in softwood (gymnosperm). Hardwood hemicelluloses are
xylans, whereas softwood consists of glucomannans. The portion present in
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 127

hardwood is the most important due to its higher amount. Xylan backbone is highly
branched by side chains consisting of xylose, arabinose and galucuranic acids (Saha
2003). According to the type of sugar residue hemicelluloses are framed into four
categories: xylans, secondary cell walls of hardwood and herbaceous plants;
xyloglucans, primary cell walls of higher plants (bound to cellulose); mannans,
secondary cell walls of conifers and Leguminosae (Schaedel et al. 2010).
The degradation of hemicelluloses is mostly carried out by microorganisms that
can be found either free in nature or as part of the digestive tract in higher animals.
Such degradation is accomplished with the aid of enzymatic complexes and leads to
the production of several important industrial compounds.

6.3 Endo-b-1,4 Beta Xylanases

Xylan is the major component in hemicellulose in hardwood, but is less abundant in


softwood and accounts for one-third of all renewable organic carbon available on
earth (Prade 1996; Kulkarni et al. 1999). Yet, it is a polysaccharide formed by
monomers of 1,4-b-D-xylose with side chain components, such as arabinosyl,
glucuronosyl, methylglucuronosyl, acetyl, feruloyl and p-coumaroyl residues,
depending on the plant source (Biely 1985; Knob and Carmona 2008).
In the biodegradation of xylan the molecule is broken down into its units,
namely xylose, a pentose monosaccharide that serves as a primary source of car-
bons to bacteria and fungi, and was the first sugar isolated from wood in 1886
(Koch 1886; Sohpal et al. 2010). Xylose itself is composed by arabinofuranosyl,
glucuronyl and acetyl groups. Due to its heterogeneity and complex nature, the
complete breakdown of xylan requires the synergistic action of a large variety of
hydrolytic enzymes (Biely 1985; Coughlan and Hazlewood 1993). The sugar is
hydrolysed into its monomers by action of different enzymes, among them
endo-beta-1,4-xylanase.
Xylanases (EC 3.2.1.8) are classified into glycoside hydrolase (GH) families and
specific information can be found in the Carbohydrate-Active Enzyme (CAZy)
database (Coughlan and Hazlewood 1993; Biely et al. 1997; Pastor et al. 2007). The
protein presents more conserved folds in some families than their amino acid
sequences and these families are grouped into clans (Coughlan and Hazlewood
1993; Biely et al. 1997). Endo-xylanases are found in families 5, 7, 8, 10, 11, 26 and
43. The enzymes from families 10 (formerly known as F) and 11 (formerly known as
G) have been widely described in the literature. Family 10 endo-xylanases posses
higher molecular weight, compared to the ones of family 11, structurally composed
of a cellulose-binding domain and a catalytic domain connected by a linker peptide
(Biely et al. 1997). Family 11 endo-xylanases contain lower molecular weight
enzymes. Based on their isoelectric points, family 11 xylanases are further
sub-grouped into alkaline and acidic enzymes. It is considered that, due to their
relatively small sizes, these endo-xylanases can pass through the pores of hemi-
celluloses network performing efficient hydrolysis (Törrönen and Rouvinen 1997).
128 A.G. Rodrigues

These xylan degrading enzymes are expressed by a wide variety of fungi (Wase
et al. 1985; Bakir et al. 2001; Li et al. 2006) and bacteria (Mamo et al. 2006), but
also actinomycetes (Ball and McCarthy 1988), and yeast (Hrmova et al. 1984).
Even if there is a great variety of xylanases and their folds, mechanisms of action,
substrate specificities, hydrolytic activities and physicochemical characteristics
being diverse, mostly attentions have been driven to two of the xylanases con-
taining carbohydrolase families from families 10 and 11 and are based on their
amino acid sequence homologies (Henrissat 1991). These enzymes catalyse the
1,4-beta-D-xylosidic linkages in xylan (Collins et al. 2005). As described in the
literature (Biely et al. 1983), the enzymes not only hydrolyse the substrate but also
catalyse various biomolecular reactions, such as transglycosylation and degradation
of oligosaccharides. The authors explained the dependence of enzymatic activity on
the substrate concentration and the preference of xylanases from different sources,
as those examined from A. niger and C. albidus, for cleaving glycosidic bonds.
The thermophilic fungus Thermomyces lanuginosus, first isolated in 1899 by
Tsiklinskaya from a potato, is referred as the most suitable microorganism for the
cellulose-free xylanases production both in shake flask and bioreactor cultivations;
however, due to intraspecies variability the enzymatic expression may be affected.
According to extensive studies on the molecular, physiological and ecological
properties of T. lanuginosus strains have been carried out (Singh et al. 2003).
Furthermore, the cultivation conditions of each strain require optimisation since the
strains are influenced by nutrient and growth conditions used in their cultivation. In
addition, Aspergillus niger, Trichoderma reesei and Streptomyces have been fre-
quently explored for the production of xylanases (Pradeep et al. 2013). With the
advancements of recombinant DNA technology, the gene responsible for the
enzyme started being cloned and expressed in other microorganisms, enabling
researchers to end up with purer products, presenting higher enzymatic activity and
in increased amounts. The enzyme has been cloned in bacteria, mostly Escherichia
coli, and filamentous fungi such as Trichoderma reesei and Fusarium oxysporum
have also been employed for this purpose (He et al. 2009; Moukouli et al. 2011). In
most cases, when it comes to the heterologous expression of xylanase, the host
models used are widely applied yeast Pichia pastoris and in some cases E. coli (He
et al. 2009; Cai et al. 2011; Moukouli et al. 2011).

6.4 Molecular Features in Process and Process


Optimisation

As described by Harris and Ramalingam (2010) endo-xylanases cleave the glyco-


sidic bonds in the xylan backbone. The enzymes have been differentiated according
to the end products they release from the hydrolysis of xylan (for instance xylose,
xylobiose and xylotriose and arabinose). Therefore, xylanases may be classified as
non-debranching (arabinose non-liberating) or debranching (arabinose liberating)
enzymes.
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 129

Due to the absent of standardisation of the various existing sources of the


enzyme and process parameters Bailey et al. (1992) compared the measurement of
xylanase activity among twenty different laboratories. According to the authors, the
major source of variation between apparent xylanase activities was probably the
substrate chosen, although small differences in protocols were also significant. After
standardisation of substrate 260 and method, the interlaboratory standard variation
of the results decreased from 108 to 17 % from the mean.
Based on its technical and economic importance and vast range of applications,
endo-xylanases have been extensively investigated regarding its expression and
process optimisation, particularly over the last few years. Due to the current con-
ditions of operations it is desired that the employed enzyme presents thermostability
and an adequate pH range in the production process. The molecular features are of
upmost concern since detailed knowledge is required to obtain more efficient
processes and appropriate amounts of product with high quality. In order to
overcome enzyme limitations under diverse conditions and enhance stability,
among other important issues related to enzyme yield and industrial scale pro-
duction, many research groups have focused on molecular engineering techniques
in search for enzymes with superior performance, and substantial results have been
reported. Moreover, hybrid xylanases have been created and tend to be increasingly
applied in the coming years to enable such progress. Because the processes in
which endo-xylanases are applied are undertake in harsh conditions, such as the pH
in the case of pulp bleaching, there are specific requirements to improve the process
conditions (Biely et al. 1983; Nakamura et al. 1993; Stephens et al. 2007).
Therefore, it is crucial to understand the properties of the enzyme, its structure and
tune its features as possible in order to achieve optimal efficiency (van Dyk et al.
2010).
An endo-xylanase isolated from the soil showed high sequence homology with
the xylanases from Bacillus pumilus and Clostridium acetobutylicum in the
N-terminal region, with stability in alkaline pH (9.0) and optimum temperature for
the activity around 50 °C (Nakamura et al. 1993). The influence of medium
compounds in the induction and repression of xylanase has been described (Gomes
et al. 1994) demonstrating that arabinose, glucose, lactose and xylose were inef-
fective in the induction of xylanase from Thermoascus aurantiacus. The study
showed yet that while wood xylan, birch wood glucuronoxylan and methyl/3-D-
xylopyranoside (MXP) induced xylanase, glucose acted as a repressor in that case.
Purkarthofer and Steiner (1995) also studied the effect of carbohydrates on the
production of inducible b-xylanase of Thermomyces lanuginosus, concluding that
D-pentoses as well as xylan and xylobiose served as inducers, but D-lyxose, D-
arabinose, D-ribose and L-arabinose were significantly not as pronounced as xylose,
xylobiose and xylan. The crystallographic structure of endo-xylanase I from family
G, which typically exhibit acidic pH, resulting in a proposed model to serve as an
explanation of the mechanisms that govern the low pH optimum of the enzyme
(Krengel and Dijkstra 1996). By solving the structure of an endo-xylanase from
Aspergillus niger employing molecular replacement Krengel and Dijkstra
130 A.G. Rodrigues

(1996) revealed aspects of the enzyme conformation and purposed that two glu-
tamate residues would be involved in the catalysis.
De Groot et al. (1998) cloned the gene encoding xylanase from Agaricus bis-
porus and determined its regulation by heterologous screening techniques. Georis
et al. (1999) presented a recombinant xylanase from Streptomyces sp. The authors
studied the cleavage site between the signal peptide and the protein and compared
the similarities and strict identities of the enzyme with other family 11 xylanases.
By means of crystallography the group of Professor Mizuno in Japan (Fujimoto
et al. 2000) depicted the structure of the xylan-binding domain endo-beta-xylanase
family 10 from Streptomyces olivaceoviridis. Turunen et al. (2001) reported the
synergistic effect of a xylanase II from Trichoderma reesei brought about by sta-
bilising mutations to access the information about its thermal stability, catalytic
function and pH-dependent properties. Zhengqiang et al. (2001) isolated two
xylanases genes from the bacterium Thermotoga maritima. The genes were sub-
sequently cloned and expressed in E. coli and the authors found that the optimum
condition for the enzyme activity was at 90 °C and pH of 6.14. A disulphide bridge
into the N-terminal region of Trichoderma reesei endo-1,4-beta-xylanase was
engineered by Fenel et al. (2004), increasing the enzyme thermostability by about
15 °C. Subsequently Fenel et al. (2006) achieved high stability using a mutant
xylanase from Trichoderma reesei, which increased brightness in sulphate pulp
bleaching. Brutus et al. (2004) showed the cloning, purification and expression of
endo-xylanase from Penicillium funiculosum in E. coli and Pichia pastoris as hosts.
Wakiyama et al. (2008), Jun et al. (2009) and Do et al. (2013) also published studies
related to the expression of xylanase in the same host. The group of Professor
Murray contributed to the elucidation of the structure of a endo-xylanase (family
10) from Thermoascus aurantiacus, showing that the glycone subsite of the enzyme
makes extensive direct and indirect interactions with the arabinose side chain and
concluding that xylan side chains are not just accommodated but can actually
constitute significant substrate specificity determinants in the enzyme under study
(Vardakou et al. 2005).
Another study explored the function of three enzymes, namely xylosidase–ara-
binosidase from Thermoanaerobacter ethanolicus and xylanase from Thermomyces
lanuginosus, with the aim to achieve more efficiency in the decomposition of ara-
binoxylan agricultural residue for the bioconversion into fuels or industrial chemi-
cals (Xue et al. 2009). The multifunctional enzyme was expressed in E. coli and
exhibited enhanced enzymatic activity in the degradation of arabinoxylan into its
monomer constituents when compared to the corresponding free enzymes. Vieira
et al. (2009) reported the effect of temperature and binding of substrate on the
dynamics of the family 11 xylanase from Bacillus circulans and its molecular
dynamics simulations in the presence of xylobiose. The study indicates that the
thermophilicity of the 11/G xylanases might be increased by rational design based on
two variables: mutations that lead to changes in the flexibility of the thumb loop and
mutations of residues in the substrate-binding cleft that aim to optimise the xyla-
nase–substrate intermolecular interaction. With that the authors purposed that the
residues may be considered as preferential targets for site-directed mutagenesis
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 131

experiments that modulate the enzyme substrate affinity, thus increasing the catalytic
efficiency of the enzyme. The work Michaux et al. (2010), based on crystallographic
techniques, with an acidophilic xylanase from Scytalidium acidophilum, a fungi
isolated from an uranium mine in acidic waters (pH 2.0), revealed features about the
stability and acidophilic adaptation. The conclusions were that various sequence and
structure modifications may be responsible for the acidophilic characteristic of the
enzyme. They would be the presence of aspartic acid hydrogen bonded to the
acid/base catalyst; the nature of specifically conserved residues in the active site; the
negative potential at the surface and the decreased number of salt bridges and
hydrogen bonds in comparison with highly alkaline enzymes.
A xylanase cloned and expressed in E. coli by Hwang et al. (2010) had its activity
not affected by most salts, such as NaCl, LiCl, KCl, NH(4)Cl, CaCl(2), MgCl(2),
MnCl(2) and CsCl(2) at 1 mM; however, the enzymatic activity was affected by
CuSO(4), ZnSO(4) and FeCl(3). As described, xylobiose was the major product
obtained. The presence of NaCL at 12.5 proved to be not only beneficial but
also the optimum condition to an endo-xylanase from thermophilic bacterium
Thermoanaerobacterium saccharolyticum isolated from a hydrothermal vent (Hung
et al. 2011). Furthermore, the expression of endo-xylanase has been described in
homologous (Anasontzis et al. 2011) and heterologous (Damásio et al. 2011) models
and applied in the production of ethanol and functional foods, respectively. In 2012
Trevizano et al., by means of direct evolution, improved the thermostability of
endo-xylanase. The mutant enzymes generated through error-prone PCR presented a
good performance in extreme pH conditions, retaining their ability to hydrolyse
birchwood and oat spelt xylans, according to the authors. Kim et al. (2012) studied
the intra-molecular hydrophobic interactions of xylanase, applying network analysis
to interpret such events, and noticed that the enzyme half-life was increased by
78-fold, highlighting the advantages of interpreting collective hydrophobic inter-
action patterns. Verma and Satyanarayana (2012) also reported a thermostable gene
from Geobacillus thermoleovorans expressed in E. coli with enzymatic production
of 27-fold higher than the wild strain. Thermostabilisation was once more investi-
gated by Li et al. (2013). Using Dictyoglomus thermophilum as an enzymatic source
of xylanase, the researchers stabilised the enzyme by an engineered N-terminal
disulphide bridge. The stabilisation was then tested against high temperatures, being
achieved at 100–110 °C and the ionic liquid used in the study appeared to affect the
functioning of the enzyme’s active site to a greater extent than the stability of an
already thermostable protein structure. Recently, Mander et al. (2014) produced a
cellulase-free xylanase from Streptomyces sp. using an agricultural residue (wheat
bran) as a growth substrate. The outcome was that the enzyme was not only able to
hydrolyse commercially available pure beechwood xylan to xylose, xylobiose and
xylotriose, but also abundantly available lignocellulosic agricultural residues in
nature such as wheat bran to xylooligosaccharides. Other studies have also
approached the thermal and alkaline stability, as well as enzyme–substrate interac-
tion of hybrid xylanases, as the ones reported by Song et al. (2014), Stephens et al.
(2014) and Chen et al. (2014).
132 A.G. Rodrigues

Lately, the enzymatic stability was investigate by Li et al. (2015a, b) employing


a disulphide mutant at pressure and temperature as high as 500 MPa and 80 °C,
respectively. Likewise, Qian et al. (2015) found that an engineered xylanase was
stable at 55 °C in a broad active pH range (5.5–10), being purposed as a candidate
for industrial applications given its high specific activity, stability and soluble
protein yield. Huy et al. (2015) observed the synergistic action of a endo glucanase
together with endo-xylanase, with increased reducing sugar release of birchwood
xylan (6.4 %), beechwood xylan (13 %) and arabinoxylan (15.8 %) and larger
increase of reducing sugar release from pretreated barley straw that addition at the
start or by treatment with endo-xylanase alone, showing a great potential applica-
tion for hemicellulose saccharification.

6.5 Techniques Used in Xylanases Process

A specific range of techniques has been used to enable researchers and producers
transforming what nature provides us, making important and always better goods in
daily life. From molecular biology to analytical chemistry and biochemistry engi-
neering, these methods and their improvements assure better results and more
efficient processes to be conducted.
Regarding the recombinant DNA techniques used in the process of obtaining,
characterising and optimising key factors of the enzyme, polyacrylamide gel
electrophoresis (SDS–PAGE) is a widely used technique to determine molecular
mass of proteins, thus aiding the characterisation of such biomolecules (Pribowo
et al. 2012). The technique has been applied through the decades as a key tool to
identify xylanases and is part of the essential technique repertoire (Biely et al. 1983;
Coughlan and Hazlewood 1993; Takahashi et al. 2013). Polymerase chain reaction
(PCR) and gene cloning comprises the paramount employed in the analysis and
improvement of the enzyme production and analysis of its properties (Stephens
et al. 2014).
Among the analytical chemistry techniques, UV/vis spectrophotometry has been
employed to analyse the enzymatic activity of xylanases (Sonia et al. 2005).
Chromatographic methods, namely high performance liquid chromatography
(HPLC), have been used to purify the enzyme as demonstrated in a recent work
(Pribowo et al. 2012). Column chromatography is another method to purify the
enzymes from cultivation (Heck et al. 2006), as well as thin layer chromatography
(TLC) (Chanwicha et al. 2015), used in the analysis of xylanases. Along with the
above-mentioned methods, mass spectrometry finds its use in the xylanases process
as described by Jänis et al. (2007) for the determination of steady-state kinetic
parameters for a xylanase-catalysed hydrolysis of xylooligosaccharides. Puchart
and Biely (2008) carried out the production of endo-beta-1,4-xylanase using
Thermomyces lanuginosus, and the authors studied the compounds formed
employing mass spectrometry, suggesting that the fungus might be of importance
regarding the conversion of xylan into xylooligosaccharides.
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 133

6.6 Biomass Production Methods

Xylanolytic enzymes from various sources, and most importantly from microor-
ganisms, have been studied along the last decades to understand their physical and
biochemical characteristics, due to their applications in several fields (Polizeli et al.
2005).
Xylanases from natural sources are produced only to relatively low yields;
therefore, the enzyme production cannot meet the demand of our societies. In view
of this, heterologous expression approaches have been implemented to design
xylanases with the desired characteristics for the production at the industrial scale.
The expression of fungal xylanase genes in Saccharomyces cerevisiae, Pichia
pastoris and other microorganisms has been reported in the literature (Karaoglana
et al. 2014). The enzyme properties and yield somehow vary depending on the gene
source, host system and secretion signal sequences, besides the process parameters.
Until the advent of recombinant DNA technology, enzymes were produced by
fermentation of the microorganisms that express the enzymes. Cultivation of fila-
mentous fungi for large-scale protein production is very complex, often ending up
in many interfering enzymes. Purification of target enzymes from a pool of proteins
requires several purification steps thereby increasing their costs (Kormelink et al.
1993). Recombinant DNA technology allows large-scale expression of these
enzymes in both homologous and heterologous protein expression hosts (Puchart
and Biely 2008; Juturu and Wu 2012).
Regarding the methods of obtaining endo-b-1,4-xylanases, solid-state fermen-
tation (SSF) is the most present in the literature (Filho et al. 1993; Pandey 2002;
Sonia et al. 2005; Chanwicha et al. 2015). SSF is the most suitable method for
xylanase production, since it deals with low water activity environment and allows
the use of agro waste substrates like corn and sugarcane bagasse. The method
received more attention after during 1950–1970 with the steroid transformation
using fungal cultures and the production of proteins for cattle feed enrichment. Later
on SSF would gain more importance with the design of bioreactors and the microbial
production of food, feed and enzymes such as lipase, phytase, cellulase, ligninase,
amylase, glutaminase, pectinase and xylanase, besides the production of organic
acids, secondary metabolites, fatty acids and biofuel (Pandey 2002; Bhargav et al.
2008). Compared to submerged fermentation (SmF), it facilitates the recovery of the
final product, requires less energy (no need for vigorous agitation) and has high
volumetric production, being thus eco-friendly and industrially favourable (Filho
et al. 1993; Pandey 2002; Bhargav et al. 2008). The method is applied under
microbial conditions in which cultures are closer to their natural habitat, considering
that fungi grow in nature on solid substrates such as pieces of wood, seeds, stems and
roots, therefore having their activity increased. The process can still be classified on
the grounds of substrate nature, which can be pure or mixed (Bhargav et al. 2008).
Key factors have to be satisfied if appropriate production is intended to be
achieved. Among these are moisture and water contend. This is due to the fact that
water activity of the substrate influences the microbial activity, spore production
134 A.G. Rodrigues

and metabolites secreted. Another issue to be addressed is the removal of heating


since in SSF a large amount of heat if generated based on the low thermal con-
ductivity of the substrates used. As important are the biomass determination and the
mass transfer during the process, where O2 and CO2 diffusion, nutrient absorption
and metabolites formation, airflow into and out of the SSF system, types of sub-
strate, mixing of substrate, bioreactor design, space between particles, variation in
particle size and microorganisms all play an important role (Bhargav et al. 2008).
Among the aspects to be considered when working with solid-state fermentation
are the selection of the suitable microorganism and substrate, optimisation of the
process parameters and isolation and purification of the product. Studies have been
conducted in this direction as reported by Sonia et al. (2005), who investigated the
endo-xylanase production by Thermomyces lanuginosus in SSF using sorghum
straw as substrate, obtaining high levels of xylanases. In another study the method
was employed by Irfan et al. (2014), in order to obtain endo-xylanase from
Trichoderma viride. The authors evaluated the enzymatic production using different
substrates such as wheat bran, rice polish, rice husk, soybean meal, sunflower meal,
sugarcane bagasse or corn cobs and found that sugar cane was the best substrate in
that occasion.
For practical reasons, immobilisation of microorganism or enzymes on solid
materials offer several advantages, including repeated usage of the catalyst, ease of
product separation and improvement of enzyme stability (Beg et al. 2001).
Endo-xylanase has also been successfully immobilised in other microorganisms as
Trichoderma reesei (Haapala et al. 1996) and Bacillus pumilus (Kapoor et al.
2008).

6.7 Technological Developments


of Endo-Beta-1,4-xylanase

Apart from its use in the pulp and paper industry, xylanases are also used as food
additive, in wheat flour for improving dough handling and quality of baked
products, for the extraction of coffee, plant oils and starch, and in the improvement
of nutritional properties of animal feed, and in combination with pectinase and
cellulase for clarification of fruit juices (Biely 1985; Wong et al. 1988; Maat et al.
1992; Beg et al. 2001).

6.8 Biobleaching

Xylanases and other side-cleaving enzymes have been used in pulp bleaching
primarily to reduce lignin and increase the brightness of the pulp. This process is
necessary due to the presence of residual lignin and its derivatives in the pulping
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 135

process, which causes the resultant pulp to gain a characteristic brown colour. The
use of xylanase in bleaching pulp requires the use of enzymes with special char-
acteristics (Beg et al. 2001).
A key requirement is to be cellulose-free, to avoid damaging the pulp fibres, as
cellulose is the primary product in the paper industry (Srinivasan and Rele 1999;
Subramaniyan and Prema 2000). In pulp bleaching, the enzyme must tolerate high
temperature and pH after alkaline cooking to be economical (Turunen et al. 2001).
The process of lignin removal from chemical pulps to produce bright or completely
white finished pulp is called ‘bleaching’. It is necessary for aesthetic reasons and for
improvement of paper properties, because the left-over residual lignin after sulphite
pulping imparts an undesirable brown colour to the paper. The bleaching of kraft
pulp uses large amounts of chlorine-based chemicals and sodium hydrosulfite.
These bleaching chemicals cause several effluent-based problems in the pulp and
paper industries. Byproducts from using these chemicals are chlorinated organic
substances, some of which are toxic, mutagenic, persistent and bioaccumulate, and
cause numerous harmful disturbances in biological systems (Onysko 1993). In
response to public interests, paper industries are currently changing practices to
minimise the use of chlorine-based chemicals. The available options are oxygen
delignification, extended cooking and substitution of chlorine dioxide for chlorine,
hydrogen peroxide and ozone. But most of these methods involve high capital
investment for process change. Thus, an alternative and cost-effective method, i.e.,
use of enzymes, has provided a very simple and economic way to reduce the use of
chlorine and other bleaching chemicals. Biobleaching involves using microorgan-
isms and enzymes for bleaching pulp. It relies on the ability of some microor-
ganisms to depolymerize lignin directly and on the use of microorganism or
enzymes that attack hemicellulose and hence favour subsequent depolymerization
(Jimenez et al. 1997).
Several criteria are essential for choosing a microorganism to produce xylanases.
To give the desired bleaching effect, the resulting enzyme preparation must be
completely free of any cellulase activity (Srinivasan and Rele 1999; Subramaniyan
and Prema 2000; Beg et al. 2001), since any cellulase activity will have serious
economic implications in terms of cellulose loss, degraded pulp quality and
increased effluent treatment cost.
Use of hemicellulolytic enzymes was the first large-scale application of enzymes
in the pulp and paper industry (Viikari et al. 1986). Limited hydrolysis of hemi-
cellulose in pulps by hemicellulases (mainly xylanases) increased the extractability
of lignin from the kraft pulps and reduced the chlorine required in subsequent
bleaching. The xylanase from T. reesei has been reported to act uniformly on all
accessible surfaces of kraft pulp and to be effective during biobleaching (Saake
et al. 1995; Suurnakki et al. 1996; Bhat 2000).
The enzyme is already on the market and has been successfully applied in the
bleaching process as demonstrated by Shatalov and Pereira (2007), nevertheless
many efforts have been done with the aim to improve its performance.
An xylanase with highly specific activity towards xylan was reported by Li et al.
(2005), with a narrow optimum pH range (7.0–7.5) and decrease in the chlorine
136 A.G. Rodrigues

consumption of just over 28 %. Battan et al. (2007) employed Bacillus pumilus to


produce xylanase, achieving an increase of 13-fold after process optimisation. The
enzyme was used in the biobleaching of eucalyptus kraft pulp. It was observed an
improvement in the brightness as well as in the whiteness, besides a decrease in the
yellowness of the material. Other parameters such as viscosity tensile strength,
breaking length, burst factor and burstness also presented better properties.
Moreover, the reduction in the consumption of chloride used was of 20 %. Another
study, from the group of professor Hazlewood, in California, screened 200 xyla-
nases and assessed their performance in biobleaching of softwood and hardwood
from different regions in the US, Canada and South America (Esteghlalian et al.
2008). The enzyme with the best properties was a xylanase from family 11,
although alkali-tolerant enzymes from the family 10 were also identified and tested.
The later enzyme, however, presented less effective bleachability, leading to a
minor reduction of chemicals at the same enzyme activity level when compared
with the selected candidate of family 11.
Maalej-Achouri et al. (2012) reported a reduction of 40 % of chloride con-
sumption using xylanase from Talaromyces thermophilus for the bleaching of kraft
pulp. The synergism of xylanase and a commercial laccase resulted in a higher
enzymatic activity when the sample was treated for 3 h at 50 °C. The enzyme
showed optimal activity under slight basic pH (7–8). Zheng et al. (2012) obtained
50 % reduction of chloride consumption and 3.65 % increase in the brightness on
cotton stalk pulp using a recombinant xylanase. The researchers also used the
enzyme in the saccharification of paper sludge in combination with mixed cellu-
lolytic enzymes and found that the efficiency of recycled material was increased by
10 %. Besides that, the recombinant enzyme presented thermal stability in the range
of 30–80 °C and in the pH from 4 to 9 being, as proposed by the group, suitable for
industrial applications in pulp biobleaching and bioethanol production. Other
studies have demonstrated the usefulness and importance of using endo-xylanases
in the biobleaching process with enhanced brightness, whiteness and significant
reduction of chemicals in the process, contributing thereby to better environmental
practices (Saleem et al. 2009; Kumar et al. 2009; Li et al. 2010; Ko et al. 2011; Lin
et al. 2013).

6.9 Baking

Hemicellulases, especially endo-xylanases have also been used to improve the


quality of dough, bread, biscuits, cakes and other bakery products (Poutanen 1997;
Butt et al. 2008). Although the endo-xylanases are known to exhibit many bene-
ficiary effects during dough handling and baking, improving the texture and shelf
life of bread, their actual mechanism of action is not well understood. It has been
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 137

hypothesised that the ability of endo-xylanases to hydrolyse arabinoxylan present in


dough facilitates the redistribution of water in both dough and bread, and is
responsible for the observed favourable effects on dough handling, bread volume,
texture and stability.
The enzyme was first introduced in the baking industry in the 1970s. They affect
rheological properties of dough, as well as the organoleptics properties of the bread
(Qi Si and Drost-Lustenberger 2002; Collins et al. 2006). Besides that, the addition
of endo-xylanases during dough processing is expected to increase the concentra-
tion of arabino-xylo-oligosaccharides in bread, which have beneficiary effects on
human health. Recently, arabinases, a-L-arabinofuranosidases, arabinoxylan a-L-
arabinofuranohydrolases and esterases have been reported to play important roles in
improving the texture, quality and sensory attributes of bakery products (Poutanen
1997). However, a suitable combination of these enzymes is vital for achieving
maximum benefit during dough processing and baking.
The hole of endo-xylanase purified from Trichoderma reesei in rye baking was
investigated by Autio et al. (1996), using two rye varieties, Danko (Polish culti-
vated in Sweden) and Muskate (Canadian). The authors found a lesser viscosity in
the bread mass after adding endo-xylanase. In relation to the fermentation assay, the
dough in both varieties tested increased with the use of xylanase with slight dif-
ference between the samples analysed; however, the bread volume was not
increased. This fact might be related to the degradation of arabinoxylans and beta
glucans, leading to the release of the gas during the process, which plays an
important role in the bread volume. The enzyme also influenced the softening of the
dough. Besides that, it was observed that the amount of xylan present in the rye also
influences properties as the molecular weight when xylanase was used, since it
affects the amount of xylooligosaccharides generated. Collins et al. (2006) studied
the properties of endo-xylanase family 8 in the baking of Argentinean long breads
and Belgian hard rolls. The increase in the bread loaf volume was higher when
using xylanase 8 than with the compared xylanase 10. Furthermore, the cut width
was also increased with xylanase 8.

6.10 Hydrolysis of Xylan into Xylooligosaccharides

The hydrolysis of xylan by endo-xylanases has already been reported during the last
decades in order to elucidate the structure of such xylooligosaccharides and the
enzymatic activity (Gorbacheva and Rodionova 1977; Meagher et al. 1988;
Kormelink et al. 1993; Anand and Vithayathil 1996; Vardakou et al. 2005; Rantanen
et al. 2007; Yang et al. 2011; Li et al. 2012, 2014).
Recently, the manufacture of xylooligosaccharides (XOS) from lignocellulosic
materials as novel sweeteners and functional foods attracts growing interest.
Oligosaccharides are generally defined as saccharides containing between 2 and 10
sugar moieties (Wang et al. 2009; Damen et al. 2012). As one of the non-digestible
oligosaccharides (NDOs), XOS exhibit many important and interesting functional
138 A.G. Rodrigues

properties, including non-toxicity, non-metabolism by human digestive system,


promotion of bowel function, calcium absorption, lipid metabolism, and growth of
beneficial intestinal bacteria, and reduction of colon cancer by forming short-chain
fatty acids. Because xylan is stable in alkaline media, it can be obtained by dis-
solving in caustic liquors and then precipitating with organic solvents. Then
enzymatic hydrolysis was extensively employed to degrade xylan to produce XOS,
due to less undesirable byproducts and specialised equipments. During this bio-
conversion process, the branch structure of natural xylan requires the coordination
of several hydrolases to breakdown. 1,4-b-D-xylanase xylanohydrolase hydrolyses
1,4-b-D-xylosidic bonds within the b-(1,4)-linked D-xylosyl backbone of xylan
randomly and liberate b-anomeric XOS (Belkacemi and Hamoudi 2003).
Due to the fact that chemical methods are less specific and may result in removal
of side groups and considering the importance of xylooligosaccharides in xylan,
arabinoxylans have been investigated and its production has been optimised, in this
way, resulting in better process conditions. Enzymes, specific glycosidic linkages
can be split, resulting in unmodified oligosaccharides (Kormelink et al. 1993;
Vardakou et al. 2005). The arabinoxylan backbone can be randomly hydrolysed
using endo-xylanases (EC 3.2.1.8), producing a mixture of various oligosaccha-
rides. Arabinose side groups are liberated by beta-L-arabinofuranosidases (EC
3.2.1.55) and exo-xylosidases (EC 3.2.1.37) release xylose units from the
non-reducing end of xylo-oligosaccharides (Biely 1985). The fragments released by
xylanase represent usually a complex mixture of arabinoxylooligosaccharides, as
reported by Puchart and Biely (2008).
Guerfali described the synergistic action of an immobilised endo-beta-
1,4-xylanase from Talaromyces thermophilus (Guerfali et al. 2011). The potential
of the combined use of endo-xylanase and arabinofuranosidase was highlighted as a
potential catalyst source and final application in food processing. Likewise, Várnai
et al. (2011) studied the combined action of endo-xylanase and endo-b-mannanase,
leading to a higher liberation of glucose than that theoretically expected in gluco-
mannan in softwood.
The obtainment of xylooligosaccharides using a plethora of microorganisms as
endo-xylanases source as well as its engineering in order to optimise the process
and improve efficiency has been continuously explored, especially in the last few
years in order to provide the needs the modern societies demand (Gonçalves et al.
2012; Verma and Satyanarayana 2012; Zhao et al. 2012; Chen et al. 2013; Kim
et al. 2014; Bibi et al. 2015; McCleary et al. 2015; Yang et al. 2015).

6.11 Feedstock

The animal feed industry is an important sector of agro-business with an annual


production of 600 million tons of feed, worth 50 billion US dollars. Of the total feed
produced, the major share is taken by poultry, pigs and ruminants (up to 90 %),
while the pet foods and fish farming account for 10 % (Bhat 2000).
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 139

Xylans are major constituents in the non-nutritional constituent of feed in


monogastric animals since the animals do not naturally produce the enzymes
required for the breakdown of these components. Another reason for the use of the
enzyme is that, with increasing concerns associated with antibiotic resistance, there
is pressure to reduce the use of antibiotics in feed. Alternatively probiotics can be
added to feed as a replacement for antibiotics. Since probiotics are effective in
enhancing the immune system, increasing body weight gain, reducing diarrhea and
improving feed conversion efficiency. Cereals (namely barley, wheat, rye and oats)
are major feed components for monogastric animals. The cell walls of cereals are
primarily composed of carbohydrate complexes referred to as nonstarch polysac-
charides, such as b-glucans in barley and wheat and arabinoxylans in rye and oats.
Due to low digestibility of b-glucans and arabinoxylans and their propensity to
form high-molecular-weight viscous aggregates in the gastrointestinal tract, these
polymers exhibit in antinutritive effect. Therefore, addition of specific enzymes
such as xylanase or b-glucanase into wheat- or barley-based diets for nonruminant
animals decreases viscosity and consequently reduces the antinutritional effect of
nonstarch polysaccharides, leading to better production performance (Liu et al.
2005). Xylanases have been extensively used to improve the performance in animal
feed (Beachemin et al. 1999; Deng et al. 2006; Cai et al. 2011). In feed application,
the use of the enzyme requires a set of parameters. It has to be stable in high
temperature during feed preparation, but the catalytic activity is needed at the body
temperature of domestic animals. The addition of xylanase in animal feed to
degrade xylans converts the more complex polysaccharides into fermentable energy
sources. This helps to maintain a lower pH, which helps exclude potential enter-
opathogens. The enzyme has a major economic importance in developing countries,
where the potential of improved dairy of ruminants is of significant importance
(Phakachoed et al. 2012). Improvements in animal performance due to the use of
enzyme additives can be attributed mainly to improvements in ruminal fibre
digestion resulting in increased digestible energy intake (Phakachoed et al. 2012).
Grounded on role of the enzyme in the animal feed, several researchers have
concentrated their efforts in the investigation properties and optimisation process of
xylanases in this regard. Liu et al. (2005) cloned and expressed three rumen microbial
fibrolytic enzyme genes in a strain of Lactobacillus reuteri and investigated the
probiotic characteristics of these genetically modified lactobacilli. The genes of
Neocallimastix patriciarum xylanase, Fibrobacter succinogenes b-glucanase and
the Piromyces rhizinflata cellulase were cloned in a strain of L. reuteri isolated from
the gastrointestinal tract of broilers. The enzymes were expressed and secreted under
the control of the Lactococcus lactis lacA promoter. According to the authors the L.
reuteri transformed strains not only acquired the capacity to break down soluble
carboxymethyl cellulose, b-glucan, or xylan but also showed high adhesion efficiency
to mucin and mucus and resistance to bile salt and acid. Other studies followed this
pattern and investigated in more detail the performance of xylanase in the field
(Panwar et al. 2013; Pedersena et al. 2015).
140 A.G. Rodrigues

6.12 Xylitol

Xylitol is the second most abundant polyol derived from xylose (Kirilin et al.
2012). Its application hits a broad spectrum in the actual scenario and more are
promising to come true (Aranda-Barradas et al. 2010; Kaialy et al. 2014). The sugar
alcohol is metabolised in the body by insulin-independent pathways and presents
good sweetness as sucrose, however, being less caloric. For these reasons the
molecule has been applied in the food industry (present in chewing gums, sweets,
soft drinks and ice creams, for instance) as an artificial sweetener in the treatment of
diabetes and erythrocytic glucose-6-phosphate dehydrogenase deficiency (Ping
et al. 2013).
It has also been used as a starting material in organic chemistry reactions, as a
potential anticancer compound, in the biomedical field, as a dental caries inhibitor
and oral biofilm formation (Huang et al. 2011; Li et al. 2015a, b; Cardoso et al.
2014; Ma et al. 2014). The molecule has a strong market position, in the range of
90–340 million dollars a year, with a production of 20,000–40,000 tons per year,
and therefore has been extensively studied regarding its production parameters
(Granström et al. 2007; Aranda-Barradas et al. 2010; Li et al. 2015a, b).
Kirilin et al. (2012) described the aqueous phase reforming (APR) process to
produce hydrogen. The work led to interesting results about the stability of
hydrogen formed and the conversion of xylitol. Apart from that, the researchers also
obtained higher yield and selectivities of xylitol than sorbitol towards hydrogen.
Methods to improve the performance in the food industry have also been attempted
as the microencapsulation of xylitol in gum arabic by complex coacervation method
(Santos et al. 2015). Saleh et al. (2014) explored the obtainment of xylitol through
olive stones. The researchers employed response surface methodology (RSM) to
optimise the hydrolysis conditions, analysing treatment temperature and process
time as key factors. Another study (Zhang et al. 2015) conducted at high temper-
ature, using engineered Kluyveromyces marxianus, resulted in high yield, demon-
strating the potential of thermo-tolerant organisms in the biomass conversion of
xylitol, using glycerol as the best substrate in this case. The simultaneous of ethanol
and xylitol production by pinch analysis was performed by Franceschin et al.
(2011).
The group of professor Miyatake in Japan recently reported synthesis of xylitol
microcapsules to encapsulate phase-change materials (PCM), a material capable of
storing and releasing large amount of energy, thereby improving its durability while
adding the advantages of smaller particles in relation to the bulk material (Makuta
et al. 2015).
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 141

6.13 Juice and De-inking Processes

Together with pectinases, cellulases and other hemicellulases, xylanases is part of


complex macerate enzymes, used in the extraction and clarification of juices. These
enzymes improve the texture and decrease the viscosity of the juices, contributing
to a more palatable end product. These enzymes participate in two steps of juice
production: after crushing, to macerate the fruit pulp either to partial or complete
liquefaction, which not only increases the juice yield and reduces the processing
time, but also improves the extraction of valuable fruit components, and after the
juice extraction, where mainly pectinases are used for its clarification, thereby
lowering the viscosity of fruit juice prior to concentration and increasing the fil-
tration rate and stability of the final product (Bhat 2000).
The application of enzymes in de-inking has been intensively studied in both
laboratory and pilot scales, but the technique has not yet been commercialised
(Buchert et al. 1998). The two principal approaches in using enzymes for de-inking
include the hydrolysis of soy-based ink carriers by lipase and the release of ink from
fibre surfaces by cellulases, xylanases and pectinases. The main advantage of
enzymatic de-inking is the avoidance of the use of alkali. De-inking, using enzymes
at acidic pH, also prevents the alkaline yellowing, simplifies the de-inking process,
changes the ink particle size distribution and reduces the environmental pollution. In
addition, the enzymatic de-inking improves the fibre brightness, strength properties,
pulp freeness and cleanliness as well as reduces fine particles in the pulp. Xylanase
treatment has been reported to increase the strength properties, while cellulase
treatment improved the brightness and freeness of the pulp (Prasad 1993).

6.14 Final Remarks

Endo-beta-1,4-xylanases comprise a series of enzymes obtained mostly in fungi and


applied in a wide range of processes. The roles of the enzyme in the chemical, food
and paper industries position this biomolecule as an important and interesting
biomaterial. The complexity of its obtaining and production optimisation has
gathered researchers from various fields during the last decades to collaborate and
contribute to advance towards better manufacturing processes.
Efforts have been applied in order to push through advancements that may result
in a finer understanding of the functionality and behaviour of the xylanases and its
relation with other molecules. Back in 1988, Meagher et al. determined the subsite
map of these hydrolases secreted by A. niger in order to get insights in the bond
cleavages in oligosaccharides and much progress have been noticed since then.
Molecular biology has been used as an important technique to perform such tasks
as enhancing the thermo stability of the enzyme, thereby enabling the makers of
goods to obtain the best possible products with maximum efficiency and expand the
range of applications to the ever growing needs and opportunities (Zhang et al. 2010;
142 A.G. Rodrigues

Deesukon et al. 2011; Karaoglana et al. 2014). The advancements in molecular


biology have provided tools to clone and express xylanases in heterologous hosts
and produce hybrid enzymes, thereby enhancing the quality and increasing the yield
of the product by tuning specific features, key factors in its production.
In short, the function of endo-xylanase in the production of paper, chemicals and
in the food industry has proved to be one of the central points in such fields and will
indeed continue to expand, fostering technological advancements and benefiting us
the best products through the most efficient processes.

Acknowledgments The author thanks Prof. V.K. Gupta for the invitation to write this chapter.
The previous work in the field was supported by the Coordination for the Improvement of Higher
Level Personnel (CAPES) and São Paulo Research Foundation (FAPESP), Brazil.

References

Anand L, Vithayathil PJ (1996) Xylan-degrading enzymes from the thermophilic fungus Humicola
Zanuginosa (Griffon and Maublanc) Bunce: action pattern of xylanase and p-glucosidase on
xylans, xylooligomers and arabinoxylooligomers. J Ferment Bioeng 81:511–517
Anasontzis GE, Zerva A, Stathopoulou PM, Haralampidis K, Diallinas G, Karagouni AD,
Hatzinikolaou DG (2011) Homologous overexpression of xylanase in Fusarium oxysporum
increases ethanol productivity during consolidated bioprocessing (CBP) of lignocellulosics.
J Biotechnol 152:16–23
Aranda-Barradas JS, Garibay-Orijel C, Badillo-Corona JA, Salgado-Manjarrez E (2010) A
stoichiometric analysis of biological xylitol production. Biochem Eng J 50:1–9
Autio K, Harkonen H, Parkkonen T, Frigard T, Poutanen K, Siika-aho M, Aman P (1996) Effects
of purified endo-beta-xylanase and endo-beta-glucanase on the structural and baking
characteristics of rye doughs. LWT Food Sci Technol 29:18–27
Bailey MJ, Biely P, Poutanen K (1992) Interlaboratory testing of methods for assay of xylanase
activity. J Biotechnol 23:257–270
Bakir U, Yavascaoglu S, Guvenc F, Ersayin A (2001) An endo-beta-1,4-xylanase from Rhizopus
oryzae: production, partial purification and biochemical characterization. Enzyme Microb
Technol 29:328–334
Ball AS, McCarthy AJ (1988) Saccharification of straw by actinomycete enzymes. J Gen Microb
134:2139–2147
Battan B, Sharma J, Dhiman SS, Kuhad RC (2007) Enhanced production of cellulase-free
thermostable xylanase by Bacillus pumilus ASH and its potential application in paper industry.
Enzyme Microb Technol 41:733–739
Beachemin KA, Rode LM, Karren D (1999) Use of enzymes in feed lot finishing diets. Can J
Anim Sci 79:243–246
Beg QK, Kapoor M, Mahajan L, Hoondal GS (2001) Microbial xylanases and their industrial
applications: a review. Appl Microb Biotechnol 56:326–338
Belkacemi K, Hamoudi S (2003) Enzymatic hydrolysis of dissolved corn stalk hemicelluloses:
reaction kinetics and modeling. J Chem Technol Biotechnol 78:802–808
Bhargav S, Panda BP, Ali M, Javed S (2008) Solid-state fermentation: an overview. Chem
Biochem Eng Q 22:49–70
Bhat MK (2000) Cellulases and related enzymes in biotechnology. Biotechnol Adv 18:355–383
Bibi Z, Shahid F, Qader SAU, Aman A (2015) Agar–agar entrapment increases the stability of
endo-beta-1,4-xylanase for repeated biodegradation of xylan. Int J Biol Macromol 75:121–127
Biely P (1985) Microbial xylanolytic systems. Trends Biotechnol 3:286–290
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 143

Biely P, Vrsanska M, Gorbacheva IV (1983) The active site of an acidic endo-1,4-beta-xylanase of


Aspergillus niger. Biochim Biophys Acta 743:155–161
Biely P, VrSanskh M, Tenkanen M, Kluepfel D (1997) Endo-beta-1,4-xylanase families:
differences in catalytic properties. J Biotechnol 57:151–166
Brutus A, Villard C, Durand A, Tahir T, Furniss C, Puigserver A, Juge N, Giardina T (2004) The
inhibition specificity of recombinant Penicillium funiculosum xylanase B towards wheat
proteinaceous inhibitors. Biochim Biophys Acta 1701:121–128
Buchert J, Oksanen T, Pere J, Siika-aho M, Suurnakki A, Viikari L (1998) Applications of
Trichoderma reesei enzymes in the pulp and paper industry. In: Harman GF, Kubicek CP
(eds) Trichoderma & gliocladium–enzymes, biological control and commercial applications,
Taylor and Francis Ltd., London, UK, vol 2, pp 343–63
Butt MS, Tahir-Nadeem M, Ahmad Z, Sultan MT (2008) Xylanases in baking industry. Food
Technol Biotechnol 46:22–31
Caffall KH, Mohnen D (2009) The structure, function, and biosynthesis of plant cell wall pectic
polysaccharides. Carbohydr Res 344:1879–1900
Cai H, Shi P, Bai Y, Huang H, Yuan T, Yang P, Luo H, Meng K, Yao B (2011) A novel
thermoacidophilic family 10 xylanase from Penicillium pinophilum C1. Process Biochem
46:2341–2346
Cardoso CAB, de Castilho ARF, Salomao PMA, Costa EN, Magalhaes AC, Buzalaf MAR (2014)
Effect of xylitol varnishes on remineralization of artificial enamel caries lesions in vitro. J Dent
42:1495–1501
Chanwicha N, Katekaew S, Aimi T, Boonlue S (2015) Purification and characterization of alkaline
xylanase from Thermoascus aurantiacus var. levisporus KKU-PN-I2-1 cultivated by
solid-state fermentation. Mycosci 56:309–318
Chen S, Kaufman MG, Miazgowicz KL, Bagdasarian M, Walker ED (2013) Molecular
characterization of a cold-active recombinant xylanase from Flavobacterium johnsoniae and its
applicability in xylan hydrolysis. Bioresour Technol 128:145–155
Chen C-C, Luo H, Han X, Lv P, Ko T-P, Peng W, Huang C-H, Wang K (2014) Structural
perspectives of an engineered beta-1,4-xylanase with enhanced thermostability. J Biotechnol
189:175–182
Collins T, Gerday C, Feller C (2005) Xylanases, xylanase families and extremophilic xylanases.
FEMS Microb Rev 29:3–23
Collins T, Hoyoux A, Dutron A, Georis J, Genot B, Dauvrin T, Arnaut F, Gerday C, Feller G
(2006) Use of glycoside hydrolase family 8 xylanases in baking. J Cereal Sci 43:79–84
Coughlan MP, Hazlewood GP (1993) b-1,4-D-Xylan-degrading enzyme systems: biochemistry,
molecular biology and applications. Biotechnol Appl Biochem 17:259–289
Damásio ARL, Silva TM, Almeida FBR, Squina FM, Ribeiro DA, Leme AFP, Segato F (2011)
Heterologous expression of an Aspergillus niveus xylanase GH11 in Aspergillus nidulans and
its characterization and application. Process Biochem 46:1236–1242
Damen B, Pollet A, Dornez E, Broekaert WF, van Haesendonck I, Trogh FI, Arnaut F,
Delcour JA, Courtin CM (2012) Xylanase-mediated in situ production of arabinoxylan
oligosaccharides with prebiotic potential in whole meal breads and breads enriched with
arabinoxylan rich materials. Food Chem 131:111–118
de Groot PWJ, Basten DEJW, Sonnenberg ASM, Van Griensven L, Visser JLDJ, Schaap PJ
(1998) An endo-1,4-beta-xylanase-encoding gene from Agaricus bisporus is regulated by
compost-specific factors. J Mol Biol 277:273–284
Deesukon W, Nishimura Y, Harada N, Sakamoto T, Sukhumsirichart W (2011) Purification,
characterization and gene cloning of two forms of a thermostable endo-xylanase from
Streptomyces sp. SWU10. Process Biochem 46:2255–2262
Deng P, Li D, Cao Y, Lu W, Wang C (2006) Cloning of a gene encoding an acidophilic
endo-beta-1,4-xylanase obtained from Aspergillus niger CGMCC1067 and constitutive
expression in Pichia pastoris. Enzyme Microb Technol 39:1096–1102
Do TT, Quyen DT, Nguyen TN, Nguyen VT (2013) Molecular characterization of a glycosyl
hydrolase family 10 xylanase from Aspergillus niger. Protein Expr Purif 92:196–202
144 A.G. Rodrigues

Esteghlalian AR, Kazaoka MM, Lowery BA, Varvak A, Hancock B, Woodward T, Turner JO,
Blum DL, Weiner D, Hazlewood GP (2008) Prebleaching of softwood and hardwood pulps by
a high performance xylanase belonging to a novel clade of glycosyl hydrolase family 11.
Enzyme Microb Technol 42:395–403
Fenel F, Leisola M, Jänis J, Turunen O (2004) A de novo designed N-terminal disulphide bridge
stabilizes the Trichoderma reesei endo-1,4-beta-xylanase II. J Biotechnol 108:137–143
Fenel F, Zitting A-J, Kantelinen A (2006) Increased alkali stability in Trichoderma reesei
endo-1,4-beta-xylanase II by site directed mutagenesis. J Biotechnol 121:102–107
Filho EXF, Puls J, Coughlan MP (1993) Physicochemical and catalytic properties of a
low-molecular-weight endo-l,4-beta-D-xylanase from Myrothecium verrucaria. Enzyme
Microb Technol 15:535–540
Franceschin G, Sudiro M, Ingram T, Smirnova I, Brunner G, Bertucco A (2011) Conversion of rye
straw into fuel and xylitol: a technical and economical assessment based on experimental data.
Chem Eng Res Des 89:631–640
Fujimoto Z, Kuno A, Kaneko S, Kobayashi SYH, Kusakabe I, Mizuno H (2000) Crystal structure
of Streptomyces olivaceoviridis E-86 beta-xylanase containing xylan-binding domain. J Mol
Biol 300:575–585
Georis J, Giannotta F, Lamotte-Brasseur J, Devreese B, van Beeumen J, Granier B, Frere J-M
(1999) Sequence, overproduction and purification of the family 11 endob-1,4-xylanase
encoded by the xyl1 gene of Streptomyces sp. S38. Gene 237:123–133
Gomes DJ, Gomes J, Steiner W (1994) Factors influencing the induction of endo-xylanase by
Thermoascus aurantiacus. J Biotechnol 33:87–94
Gonçalves TA, Damásio ARL, Segato F, Alvarez TM, Bragatto J, Brenelli LB, Citadini APS,
Murakami MT, Ruller R, Leme AFP, Prade RA, Squina FM (2012) Functional characterization
and synergic action of fungal xylanase and arabinofuranosidase for production of
xylooligosaccharides. Bioresour Technol 119:293–299
Gorbacheva IV, Rodionova NA (1977) Studies on xylan-degrading enzymes II. Action pattern of
endo-1,4-beta-xylanase from aspergillus niger str. 14 on xylan and xylooligosaccharides.
Biochim Biophys Acta 484:94–102
Granström TB, Izumori K, Leisola M (2007) A rare sugar xylitol. Part II: biotechnological
production and future applications of xylitol. Appl Microb Biotechnol 74:273–276
Guerfali M, Gargouri A, Belghith H (2011) Catalytic properties of Talaromyces thermophilus-
l-arabinofuranosidase and its synergistic action with immobilized endo-beta-1,4-xylanase.
J Mol Catal B Enzym 68:192–199
Haapala R, Parkkinen E, Suominen P, Linko S (1996) Production of endo-1,4-beta-glucanase and
xylanase with nylon-web immobilized and free Trichoderma reesei. Enzyme Microb Technol
18:495–501
Harris AD, Ramalingam C (2010) Xylanases and its application in food industry: a review. J Exp
Sci 1:01–11
He J, Yu B, Zhang K, Ding X, Chen D (2009) Expression of endo-1,4-beta-xylanase from
Trichoderma reesei in Pichia pastoris and functional characterization of the produced enzyme.
BMC Biotechnol 9:56
Heck JX, Soares LHB, Hertz PF, Ayub MAZ (2006) Purification and properties of a xylanase
produced by Bacillus circulans BL53 on solid-state cultivation. Biochem Eng J 32:179–184
Henrissat B (1991) A classification of glycosyl hydrolases based on amino acid sequence
similarities. Biochem J 280:309–316
Hrmova M, Biely P, Vrsanska M, Petrakova E (1984) Induction of cellulose- and xylan-degrading
enzyme complex in the yeast Trichosporon cutaneum. Arch Microb 138:371–376
Huang CF, Jiang YF, Guo GL, Hwang WS (2011) Development of a yeast strain for xylitol
production without hydrolysate detoxification as part of the integration of co-product
generation within the lignocellulosic ethanol process. Bioresour Technol 102:3322–3329
Hung K-S, Liu S-M, Tzou W-S, Lin F-P, Pan C-L, Fang T-Y, Sund K-H, Tang S-J (2011)
Characterization of a novel GH10 thermostable, halophilic xylanase from the marine bacterium
Thermoanaerobacterium saccharolyticum NTOU1. Process Biochem 46:1257–1263
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 145

Huy ND, LeNguyen C, Seo J-W, Kim D-H, Park S-M (2015) Putative endoglucanase PcGH5 from
Phanerochaete chrysosporium is a b-xylosidase that cleaves xylans in synergistic action with
endo-xylanase. J Biosci Bioeng 119:416–420
Hwang T, Lim HK, Song HY, Cho SJ, Chang J-S, Park N-J (2010) Cloning and characterization of
a xylanase, KRICT PX1 from the strain Paenibacillus sp. HPL-001. Biotechnol Adv
28:594–601
Irfan M, Nadeem M, Syed Q (2014) One-factor-at-a-time (OFAT) optimization of xylanase
production from Trichoderma viride-IR05 in solid-state fermentation. J Radiat Res Appl Sci
7:317–326
Jacobsen SE, Wyman CE (2000) Cellulose and hemicellulose hydrolysis models for application to
current and novel pretreatment processes. Appl Biochem Biotechnol 84–86:81–96
Jänis J, Pulkkinen P, Rouvinen J, Vainiotalo P (2007) Determination of steady-state kinetic
parameters for a xylanase-catalyzed hydrolysis of neutral underivatized xylooligosaccharides
by mass spectrometry. Anal Biochem 365:165–173
Jimenez L, Martinez C, Perez I, Lopez F (1997) Biobleaching procedures for pulp and agricultural
residues using Phanerochaete chroysosporium and enzymes. Process Biochem 4:297–304
Jun H, Bing Y, Keying Z, Daiwen C (2009) Functional characterization of a recombinant
thermostable xylanase from Pichia pastoris: a hybrid enzyme being suitable for xylooligosac-
charides production. Biochem Eng J 48:87–92
Juturu V, Wu JC (2012) Microbial xylanases: engineering, production and industrial applications.
Biotechnol Adv 30:1219–1227
Kaialy W, Maniruzzaman M, Shojaee S, Nokhodchi A (2014) Antisolvent precipitation of novel
xylitol-additive crystals to engineer tablets with improved pharmaceutical performance. Int J
Pharm 477:282–293
Kapoor M, Nair LM, Kuhad RC (2008) Cost-effective xylanase production from free and
immobilized Bacillus pumilus strain MK001 and its application in saccharification of Prosopis
juliflora. Biochem Eng J 38:88–97
Karaoglana M, Yildiza H, Inan M (2014) Screening of signal sequences for extracellular
production of Aspergillus niger xylanase in Pichia pastoris. Biochem Eng J 92:16–21
Kim T, Jeong JC, Yoo YJ (2012) Hydrophobic interaction network analysis for thermostabiliza-
tion of a mesophilic xylanase. J Biotechnol 161:49–59
Kim HM, Lee KH, Kim KH, Lee D-S, Nguyen QA, Bae H-J (2014) Efficient function and
characterization of GH10 xylanase (Xyl10 g) from Gloeophyllum trabeum in lignocellulose
degradation. J Biotechnol 172:38–45
Kirilin AV, Tokarev AV, Kustov LM, Salmi T, Mikkola J-P, Yu DM (2012) Aqueous phase
reforming of xylitol and sorbitol: comparison and influence of substrate structure. Appl Catal A
Gen 435–436:172–180
Knob A, Carmona EC (2008) Xylanase production by Penicillium sclerotiorum and its
characterization. World Appl Sci J 4:277–283
Ko C-H, Tsai C-H, Tu J, Yang B-Y, Hsieh D-L, Jane W-N, Shih T-L (2011) Identification of
Paenibacillus sp. 2S-6 and application of its xylanase on biobleaching. Int Biodeterior
Biodegrad 65:334–339
Koch E (1886) Pharm. Z. Russland, 26, 657. Ber. them. Ges., Ref., 20, 145. 1887
Kormelink FJM, Hoffmann RA, Gruppen H, Voragen AGJ, Kamerling JP, Vliegenthart FG (1993)
Characterisation by H NMR spectroscopy of oligosaccharides derived from alkali-extractable
wheat-flour arabinoxylan by digestion with endo-(1 + 4)-P-D-xylanase III from Aspergillus
awamori. Carbohydr Res 249:369–382
Krengel U, Dijkstra BW (1996) Three-dimensional structure of endo-1,4-beta-xylanase I from
Aspergillus niger: molecular basis for its low pH optimum. J Mol Biol 263:70–78
Kulkarni N, Shendye A, Rao M (1999) Molecular and biotechnological aspects of xylanases.
FEMS Microb Rev 23:411–456
Kumar KS, Manimaran A, Permaul K, Singh S (2009) Production of b-xylanase by a
Thermomyces lanuginosus MC 134 mutant on corn cobs and its application in biobleaching of
bagasse pulp. J Biosci Bioeng 107:494–498
146 A.G. Rodrigues

Li XT, Jiang ZQ, Li LT, Yang SQ, Feng WY, Fan JY, Kusakabe I (2005) Characterization of a
cellulase-free, neutral xylanase from Thermomyces lanuginosus CBS 288.54 and its
biobleaching effect on wheat straw pulp. Bioresour Technol 96:1370–1379
Li L, Tian H, Cheng Y, Jiang Z, Yang S (2006) Purification and characterization of a thermostable
cellulase-free xylanase from the newly isolated Paecilomyces themophila. Enzyme Microb
Technol 38:780–787
Li X, She Y, Sun B, Song H, Zhu Y, Lv Y, Song H (2010) Purification and characterization of a
cellulase-free, thermostable xylanase from Streptomyces rameus L2001 and its biobleaching
effect on wheat straw pulp. Biochem Eng J 52:71–78
Li X, Li E, Zhu Y, Teng C, Sun B, Song H, Yang R (2012) A typical endo-xylanase from
Streptomyces rameus L2001 and its unique characteristics in xylooligosaccharide production.
Carbohydr Res 359:30–36
Li H, Kankaanpää A, Xiong H, Hummel M, Sixtac H, Ojamo H, Turunen O (2013)
Thermostabilization of extremophilic Dictyoglomus thermophilum GH11 xylanase by an
N-terminal disulfide bridge and the effect ofionic liquid [emim]OAc on the enzymatic
performance. Enzyme Microb Technol 53:414–419
Li J, Zhou P, Liu H, Xiong C, Lin J, Xiao W, Gong Y, Liu Z (2014) Synergism of cellulase,
xylanase, and pectinase on hydrolyzing sugarcane bagasse resulting from different pretreatment
technologies. Bioresour Technol 155:258–265
Li H, Voutilainen S, Ojamo H, Turunen O (2015a) Stability and activity of Dictyoglomus
thermophilum GH11 xylanaseand its disulphide mutant at high pressure and temperature.
Enzyme Microb Technol 70:66–71
Li Z, Guo X, Feng X, Li C (2015b) An environment friendly and efficient process for xylitol
bioconversion from enzymatic corncob hydrolysate by adapted Candida tropicalis. Chem
Eng J 263:249–256
Lin X-Q, Han S-Y, Zhang N, Hu H, Zheng S-P, Ye Y-R, Lin Y (2013) Bleach boosting effect of
xylanase A from Bacillus halodurans C-125 in ECF bleaching of wheat straw pulp. Enzyme
Microb Technol 52:91–98
Liu J-R, Yu B, Liu F-H, Cheng K-J, Zhao, X (2005) Expression of rumen microbial fibrolytic
enzyme genes in probiotic Lactobacillus reuteri. Appl Environ Microbiol 71:6769–6775
Ma P, Li T, Wu W, Shi D, Duan F, Bai H, Dong W, Chen M (2014) Novel poly(xylitol sebacate)/
hydroxyapatite bio-nanocomposites via one-step synthesis. Polym Degrad Stab 110:50–55
Maalej-Achouri I, Guerfali M, Romdhane IB-B, Gargouri A, Belghith H (2012) The effect of
Talaromyces thermophilus cellulase-free xylanase and commercial laccase on lignocellulosic
components during the bleaching of kraft pulp. Int Biodeterior Biodegrad 75:43–48
Maat J, Roza M, Verbakel J, Stam H, da Silra MJS, Egmond MR, Hagemans MLD, van
Garcom RFM, Hessing JGM, van Derhondel C, van Rotterdam C (1992) Xylanases and their
application in baking. In: Visser J, Beldman G, van Someren MAK, Voragen AGJ (eds) Xylan
and xylanases. Elsevier, Amsterdam, pp 349–360
Makuta T, Kadoya K, Izumi H, Miyatake M (2015) Synthesis of cyanoacrylate-covered xylitol
microcapsules for thermal storage. Chem Eng J 273:192–196
Mamo G, Hatti-Kaul R, Mattiasson B (2006) A thermostable alkaline active endo-beta-
1-4-xylanase from Bacillus halodurans S7: Purification and characterization. Enzyme Microb
Technol 39:1492–1498
Mander P, Choi YH, Pradeep GC, Choi YS, Hong JH, Cho SS, Yoo JC (2014) Biochemical
characterization of xylanase produced from Streptomyces sp. CS624 using an agro residue
substrate. Process Biochem 49:451–456
McCleary BV, McKie VA, Draga A, Rooney E, Mangan D, Larkin J (2015) Hydrolysis of wheat
flour arabinoxylan, acid-debranched wheat flour arabinoxylan and arabino-xylo-
oligosaccharides by b-xylanase, a-L-arabinofuranosidase and b-xylosidase. Carbohydr Res
407:79–96
Meagher MM, Tao BY, Chow JM, Reilly PJ (1988) Kinetics and subsite mapping of A
D-xylobiose- and D-XmsE producing Aspergiflus niger endo-(1+4)-/W-xylanase. Carbodydr
Res 173:273–283
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 147

Michaux C, Pouyez J, Mayard A, Vandurm P, Housen I, Wouters J (2010) Structural insights into
the acidophilic pH adaptation of a novel endo-1,4-beta-xylanase from Scytalidium aci-
dophilum. Biochimie 92:1407–1415
Moukouli M, Topakas E, Christakopoulos P (2011) Cloning and optimized expression of a GH-11
xylanase from Fusarium oxysporum in Pichia pastoris. New Biotechnol 28:369–374
Nakamura S, Wakabayashi K, Nakai R, Aono R, Horikoshi K (1993) Purification and some
properties of an alkaline xylanase from alkaliphilic Bacillus sp. Strain 41 M-1. Appl Environ
Microb 59:2311–2316
Onysko KA (1993) Biological bleaching of chemical pulps: a review. Biotechnol Adv 11:179–198
Pandey A (2002) Solid-state fermentation. Biochem Eng J 13:81–84
Panwar D, Srivastava PK, Kapoor M (2013) Production, extraction and characterization of alkaline
xylanase from Bacillus sp. PKD-9 with potential for poultry feed. Biocatal Agric Biotechnol
3:118–125
Pastor FIJ, Gallardo Ó,Sanz-Aparicio J, Díaz P (2007) Xylanases: molecular properties and
applications. In: Polaina J, MacCabe AP (eds) Industrial enzymes. Springer, Dordrecht, The
Netherlands, pp 65–82
Pedersena MB, Dalsgaarda S, Arenta S, Lorentsena R, Knudsen KEB, Yua S, Lærke HN (2015)
Xylanase and protease increase solubilization of non-starch polysaccharides and nutrient
release of corn- and wheat distillers dried grains with solubles. Biochem Eng J 98:99–106
Phakachoed N, Lounglawan P, Suksombat W (2012) Effects of xylanase supplementation on
ruminal digestibility in fistula tednon-lactating dairy cows fed rice straw. Livest Sci
149:104–108
Ping Y, Ling H-Z, Song G, Ge J-P (2013) Xylitol production from non-detoxified corncob
hemicellulose acid hydrolysate by Candida tropicalis. Biochem Eng J 75:86–91
Polizeli MLTM, Rizzatti ACS, Monti R, Terenzi HF, Jorge JA, Amorim DS (2005) Xylanases
from fungi: properties and industrial applications. Appl Microb Biotechnol 67:577–591
Poutanen K (1997) Enzymes: an important tool in the improvement of the quality of cereal foods.
Trends Food Sci Technol 8:300–306
Prade RA (1996) Xylanases: from biology to biotechnology. Biotechnol Genet Eng Rev
13:101–131
Pradeep GC, Choi YH, Choi YS, Seong CN, Cho SS, Lee HJ, Yoo JC (2013) A novel
thermostable cellulase free xylanase stable in broad range of pH from Streptomyces sp. CS428.
Process Biochem 48:1188–1196
Prasad DY (1993) Enzymatic deinking of laser and xerographic office wastes. Appita J
46:289–292
Pribowo A, Arantes J, Saddler VN (2012) The adsorption and enzyme activity profiles of specific
Trichoderma reesei cellulase/xylanase components when hydrolyzing steam pretreated corn
stover. Enzyme Microb Technol 50:195–203
Puchart V, Biely P (2008) Simultaneous production of endo-beta-1,4-xylanase and branched
xylooligosaccharides by Thermomyces lanuginosus. J Biotechnol 137:34–43
Purkarthofer H, Steiner W (1995) Induction of endo-P-xylanase in the fungus Thermomyces
lanuginosus. Enzyme Microb Technol 17:114–118
Qi Si J, Drost-Lustenberger C (2002) Enzymes for bread, pasta and noodle products. In:
Whitehurst RJ, Law BA (eds) Enzymes in food technology. Sheffield Academic Press,
Sheffield, pp 19–56
Qian C, Liu N, Yan X, Wang Q, Zhou Z, Wang Q (2015) Engineering a high-performance,
metagenomic-derived novelxylanase with improved soluble protein yield and thermostability.
Enzyme Microb Technol 70:35–41
Rantanen H, Virkki L, Tuomainen P, Kabel M, Schols H, Tenkanen M (2007) Preparation of
arabinoxylobiose from rye xylan using family 10 Aspergillus aculeatus endo-
1,4-beta-D-xylanase. Carbohydr Polym 68:350–359
Saake B, Clark T, Puls J (1995) Investigations on the reaction mechanism of xylanases and
mannanases on sprucewood chemical pulps. Holzforschung 49:60–68
148 A.G. Rodrigues

Saha BC (2003) Hemicellulose bioconversion. J Ind Microbiol Biotechnol 30:279–291


Saleem M, Tabassum MR, Yasmin R, Imran M (2009) Potential of xylanase from thermophilic
Bacillus sp. XTR-10 in biobleaching of wood kraft pulp. Int Biodeterior Biodegrad
63:1119–1124
Saleh M, Cuevas M, García JF, Sánchez S (2014) Valorization of olive stones for xylitol and
ethanol production from dilute acid pretreatment via enzymatic hydrolysis and fermentation by
Pachysolen tannophilus. Biochem Eng J 90:286–293
Santos MG, Bozza FT, Thomazini M, Favaro-Trindade CS (2015) Microencapsulation of xylitol
by double emulsion followed by complex coacervation. Food Chem 171:32–39
Schaedel C, Bloechl A, Richter A, Hoch G (2010) Quantification and monosaccharide
composition of hemicelluloses from different plant functional types. Plant Physiol Biochem
48:1–8
Schulze E (1891) Information regarding chemical composition of plant cell membrane. Ber Dtsch
Chem Ges 24:2277–2287
Shatalov AA, Pereira H (2007) Xylanase pre-treatment of giant reed organosolv pulps: direct
bleaching effect and bleach boosting. Ind Crops Prod 25:248–256
Singh S, Madlala AM, Prior BA (2003) Thermomyces lanuginosus: properties of strains and their
hemicellulases. FEMS Microb Rev 27:3–16
Sohpal VK, Dey A, Singh A (2010) Investigate of process parameters on xylanase enzyme activity
in Melanocarpus albomyces batch culture. In: WCE 2010, London, UK
Song L, Dumon C, Siguier B, André I, Eneyskaya E, Kulminskaya A, Bozonnet S, O’Donohue MJ
(2014) Impact of an N-terminal extension on the stability and activity of theGH11 xylanase
from Thermobacillus xylanilyticus. J Biotechnol 174:64–72
Sonia KG, Chadha BS, Saini HS (2005) Sorghum straw for xylanase hyper-production by
Thermomyces lanuginosus (D2W3) under solid-state fermentation. Bioresour Technol
96:1561–1569
Srinivasan MD, Rele MV (1999) Microbial xylanases for paper industry. Curr Sci 77:137–142
Stephens DE, Rumbold K, Permaul K, Prior BA, Singh S (2007) Directed evolution of the
thermostable xylanase from Thermomyces lanuginosus. J Biotechnol 127:348–354
Stephens DE, Khan FI, Singh P, Bisetty K, Singh S, Permaul K (2014) Creation of thermostable
and alkaline stable xylanase variants by DNA shuffling. J Biotechnol 187:139–146
Subramaniyan S, Prema P (2000) Cellulase-free xylanases from Bacillus and other microorgan-
isms. FEMS Microbiol Lett 183:1–7
Suurnakki A, Heijnesson A, Buchert J, Westermark U, Viikari L (1996) Effect of pulp surfaces on
enzyme-aided bleaching on kraft pulps. J Pulp Pap Sci 22:591–596
Takahashi Y, Kawabata H, Murakami S (2013) Analysis of functional xylanases in xylan
degradation by Aspergillus niger E-1 and characterization of the GH family 10 xylanase
XynVII. SpringerPlus 2:447
Törrönen A, Rouvinen J (1997) Structural and functional properties of low molecular weight
endo-1,4-b-xylanases. J Biotechnol 57:137–149
Trevizano LM, Ventorim RZ, de Rezende ST, Junior FPS, Guimaraes VM (2012) Thermostability
improvement of Orpinomyces sp. xylanase by directed evolution. J Mol Catal B Enzym
81:12–18
Turunen O, Etuaho K, Fenel F, Vehmaanpera J, Wu X, Rouvinen J, Leisola M (2001) A
combination of weakly stabilizing mutations with a disulfide bridge in the alpha-helix region of
Trichoderma reesei endo-1,4-beta-xylanase II increases the thermal stability through
synergism. J Biotechnol 88:37–46
van Dyk JS, Sakka M, Sakka K, Pletschke BI (2010) Characterisation of the multi-enzyme
complex xylanase activity from Bacillus licheniformis SVD1. Enzyme Microb Technol
47:174–177
Vardakou M, Flint J, Christakopoulos P, Lewis RJ, Gilbert HJ, Murray JW (2005) A Family 10
Thermoascus aurantiacus xylanase utilizes arabinose decorations of xylan as significant
substrate specificity determinants. J Mol Biol 352:1060–1067
6 Endo-b-1,4-xylanase: An Overview of Recent Developments 149

Várnai A, Huikko L, Pere J, Siika-aho M, Viikari L (2011) Synergistic action of xylanase and
mannanase improves the total hydrolysis of softwood. Bioresour Technol 102:9096–9104
Verma D, Satyanarayana T (2012) Cloning, expression and applicability of thermo-alkali-stable
xylanase of Geobacillus thermoleovorans in generating xylooligosaccharides from
agro-residues. Bioresour Technol 107:333–338
Vieira DS, Degrève L, Ward RJ (2009) Characterization of temperature dependent and
substrate-binding cleft movements in Bacillus circulans family 11 xylanase: a molecular
dynamics investigation. Biochim Biophys Acta 1790:1301–1306
Viikari L, Ranva M, Kantelinen A, Sandquist J, Linko M (1986) Bleaching with enzymes. In:
Third international conference in biotechnology in pulp and paper industry, Stockholm,
pp 67–69
Wakiyama M, Tanaka H, Yoshihara K, Hayashi S, Ohta K (2008) Purification and properties of
family-10 endo-1,4-b-xylanase from Penicillium citrinum and structural organization of
encoding gene. J Biosci Bioeng 105:367–374
Wang J, Sun BG, Cao YP, Tian Y, Wang CT (2009) Enzymatic preparation of wheat bran
xylooligosaccharides and their stability during pasteurization and autoclave sterilization at low
pH. Carbohydr Polym 77:816–821
Wase DAJ, Raymahasay S, Wang CW (1985) Production of/ -D-glucosidase, endo- 1,4-/ -
D-glucanase and D-xylanase from straw by Aspergillus fumigatus IMI 255091. Enzyme
Microb Technol 7:225–229
Wong KKY, Tan LUL, Saddler JN (1988) Multiplicity of b-1,4-xylanase in microorganisms:
functions and applications. Microbiol Rev 52:305–317
Xue Y, Peng J, Wang R, Song X (2009) Construction of the trifunctional enzyme associating the
Thermoanaerobacter ethanolicus xylosidase-arabinosidase with the Thermomyces lanuginosus
xylanase for degradation of arabinoxylan. Enzyme Microb Technol 45:22–27
Yang H, Wang K, Song X, Xu F (2011) Production of xylooligosaccharides by xylanase from
Pichia stipitis based on xylan preparation from triploid Populas tomentosa. Bioresour Technol
102:7171–7176
Yang Q, Gao Y, Huang Y, Xu Q, Luo X-M, Liu J-L, Feng J-X (2015) Identification of three
important amino acid residues of xylanaseAfxynA from Aspergillus fumigatus for enzyme
activity and formation of xylobiose as the major product. Process Biochem 50:571–581
Yin Y, Chen H, Hahn MG, Mohnen D, Xu Y (2010) Evolution and function of the plant cell wall
synthesis-related glycosyltransferase family 81. Plant Physiol 153:1729–1746
Zhang S, Zhang K, Chen X, Chu X, Sun F, Dong Z (2010) Five mutations in N-terminus confer
thermostability on mesophilic xylanase. Biochem Biophys Res Commun 395:200–206
Zhang J, Zhang B, Wanga D, Gao X, Hong J (2015) Improving xylitol production at elevated
temperature with engineered Kluyveromyces marxianus through over-expressing transporters.
Bioresour Technol 175:642–645
Zhao L-C, Wang Y, Lin J-F, Guo L-Q (2012) Adsorption and kinetic behavior of recombinant
multifunctional xylanase in hydrolysis of pineapple stem and bagasse and their hemicellulose
for Xylo-oligosaccharide production. Bioresour Technol 110:343–348
Zheng H, Liu Y, Liu X, Han Y, Wang J, Lu F (2012) Overexpression of a Paenibacillus
campinasensis xylanase in Bacillus megaterium and its applications to biobleaching of cotton
stalk pulp and saccharification of recycled paper sludge. Bioresour Technol 125:182–187
Zhengqiang J, Kobayash A, Ahsan MM, Lite L, Kitaoka M, Hayashi K (2001) Characterization of
a thermostable family 10 endo-xylanase (XynB) from Thermotoga maritima that cleaves
p-nitrophenyl-P-D-xyloside. J Biosci D Bioeng 92:423–428
Chapter 7
Microbial Xylanases: Sources, Types,
and Their Applications

Hesham Ali El Enshasy, Subeesh Kunhi Kandiyil, Roslinda Malek


and Nor Zalina Othman

Abstract Biomass conversion to an utilizable energy sources such as monomer


sugars using enzymatic hydrolysis has been emerged as the current technology
which promises the future energy. In nature, bioconversion process of biomass is
mediated by a group of biofunctional hydrolytic enzymes. These enzymes generally
work in cooperative synergetic action to facilitate enhanced effective degradation of
biomass. Xylanase is one of the crucial hydrolytic enzymes involved in hydrolysis
of xylan, the hemicellulose which constitutes 15–30 % of the plant biomass. This
chapter discusses in detail about the enzymatic hydrolysis of xylan by the xylolytic
enzyme endo-1,4-b-xylanase, its occurrence in nature and mode of action, structure
and classifications, current methods for its production, purification, and character-
ization. In addition, the major and recent industrial applications of this enzyme were
highlighted as well.

7.1 Introduction

Nowadays, the governments promote extensive researches for the development of


an alternative transportation fuel from a renewable energy sources, realizing the
upcoming major energy crisis due to the depletion of petroleum-derived fuels. As
an initiative, the department of energy in US has started producing biofuels with a
set target of 60 billion gallon per year by 2030. Europe also has the similar targets
by that time to replace 25 % of petroleum-based liquid transportation fuel by
biofuels (Himmel et al. 2007). However, it is a challenging target since the biofuel
production using substrates such as sugar cane and corn has limited capacity to
supply such a huge volumes. Apart from this, in many countries people protest
against the use of food crops for biofuel production and many debates that it leads
to a major food crisis. As a remedy, researchers found out that the lignocellulose

H.A. El Enshasy (&)  S.K. Kandiyil  R. Malek  N.Z. Othman


Institute of Bioproduct Development (IBD), Faculty of Chemical Engineering (FKK),
University Teknology Malaysia (UTM), 81310 Johor Bahru, Malaysia
e-mail: [email protected]

© Springer International Publishing Switzerland 2016 151


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_7
152 H.A. El Enshasy et al.

biomass which consists of 75 % polysaccharide sugars can be used as a significant


feedstock for biofuel production (Lynd et al. 1991). If the attraction is more, lig-
nocellulose biomasses are renewable energy source and exist abundant in nature
and it can be derived predominantly from agricultural wastes (Gomez et al. 2008).
Lignocelluloses are heteropolysaccharides that give structural rigidity to all kinds of
plant. They account for approximately more than 50 % of the total biomass in the
nature, synthesized (estimated 10–50  1012 tons year−1) by plants through pho-
tosynthesis (Claassen et al. 1999). Lignocelluloses are composed of three major
polymeric constituents such as cellulose (an unbranched linier polymers of long
b-1-4 glucan between 2000 and 27,000 units), hemicellulose (a heterologous
polysaccharides which are complex in nature with sugar monomeric subunits such
as D-xylose, D-mannose, D-galactose, and L-arabinose) and lignin (complex phenolic
structure act as linkage between cellulose and hemicellulose.
The name hemicellulose was first introduced by Schulze (1891). These
heteropolysaccharides are abundant in plant cell wall and have complex structures
when compared to cellulose due to the enormous number of sugar monomeric side
chains (500–3000). It creates a cross-linked network between the cellulose
microfibrils and lignin and plays a major role in structural integrity of plant cell
walls. It also regulates the rate of expansion of primary cell wall and acts as a shield
over cellulose to protect it from enzymatic hydrolysis. Hemicellulose is the second
most abundant renewable biomass that is available after cellulose within the lig-
nocellulose matrix representing about 20–35 % of total biomass (Saha 2003).
Table 7.1 summarizes the approximate composition of various biomass materials.
Classification of hemicellulose was made based on the types of sugar monomer
units present on them. As mentioned above, hemicelluloses are composed of xylan,
mannan, galactan, and arabinan polymers. Among these xylan is the most fre-
quently occurring polymer composed mainly of b-D-xylopyranosyl residues which
linked by b-1,4-glycosidic bonds (Beg et al. 2001).
In hardwoods plants, hemicellulose primarily consists of xylans and gluco-
mannans whereas in softwood plants, it is composed of arabinoglucuronoxylans

Table 7.1 Approximate composition (as a percentage) of various biomass materials


Biomass Cellulose Hemicellulose Lignin References
(%) (%) (%)
Bermuda grass 47.8 13.3 19.4 Li et al. (2010)
Reed canary straw 42.6 29.7 7.6 Bridgeman et al. (2008)
Corn cobs 35–39 38–42 4.5–6.6 Okeke and Obi (1994)
Rice straw 41 21.5 9.9 Lee (1997)
Bagasse 38.1 26.9 18.4 Lee (1997)
Coffee pulp 24.0 8.9 19.4 Dijkerman et al. (1997)
Coconut fiber 17.7 2.2 34 Dijkerman et al. (1997)
Rice hulls 24–29 12–14 11–13 Okeke and Obi (1994)
Corn stove 39 19.1 15.1 Lee (1997)
Wheat straw 36.6 24.8 14.5 Lee (1997)
Sawdust 45.0 15.1 25.3 Dijkerman et al. (1997)
7 Microbial Xylanases: Sources, Types, and Their Applications 153

Table 7.2 The hemicellulolytic enzymes and their substrates (Shallom and Shoham 2003)
Enzymes Substrates
Endo-b-1,4-xylanase b-1,4-Xylan
Exo-b-1,4-xylosidase b-1,4-Xylooligomers
a-L-Arabinofuranosidase a-Arabinofuranosyl, (1–3) xylooligomers, a-1,5-arabinan
Endo-a-1,5-arabinanase a-1,5-Arabinan
a-Glucuronidase 4-O-methyl-a-glucuronic acid, (1–2) xylooligomers
Endo-b-1,4-mannanase b-1,4-Mannan
Exo-b-1,4-mannosidase b-1,4-Mannooligomers, mannobiose
b-Galactosidase a-1,6-Galactopyranose, mannooligomers
b-Glucosidase b-1,4-Glucopyranose, mannopyranose
Endo-galactanase b-1,4-Galactan

(xylans), xyloglucans, glucomannans, and arabinogalactans. Xylans are the major


form of hemicellulose made of D-xylopyranosyl units which are linked by
b-1,4-glycosidic bonds. b-mannan-based polymers are the second major form of
cellulose which are made of b-1,4-linked mannose and/or glucose residues.
Galactoglucomannans which consist of a-1,6-linked galactose side chains are also
grouped under these polymers. They are found abundant in hardwood hemicellu-
lose, which comprised varying ratios of D-mannopyranose units a-(1–6)-substituted
galactopyranose side chains with O-acetyl side chains and b-1-4-linked-D-gluco-
pyranose units. Arabinan and arabinogalactans are also considered as hemicellu-
loses which are generally composed of a-1,5-linked L-arabinofuranosyl units.
Table 7.2 summarizes the major enzymes involved in the breakdown of
cross-linked hemicelluloses. Those groups of enzymes which are involved in
hydrolysis of xylan are generally known as xylanases. The current book focuses on
one of the crucial xylolytic enzymes known as endo-1-4-b-xylanase.

7.2 Enzymatic Hydrolysis of Xylan

In most terrestrial plant species, secondary cell wall thickening takes place by the
deposition of xylan and they are found in-between cellulosic fiber sheets and lignin
with complex binding relationship through hydrogen bonding. Xylan thus forms a
think wall over cellulose and enables the protection from degradation by different
cellulose degrading enzymes. Xylan constitutes 15–30 % of the plant biomass in
hardwood and 7–12 % of the plant biomass in softwood. Since they are the major
hemicellulose group in most of the plant species, they have been considered as one
among the renewable source of energy in the form of biomass. Structure of xylan is
complex in nature as they are composed of b-1,4 linked-D-xylopyranose units with
a-L-arabinofuranose and 4-O-methyl-a-D-glucuronopyranosyl acid side chains.
Figure 7.1 shows the major enzymes involved and their site of action during the
enzymatic hydrolysis is xylan (Polizeli et al. 2005).
154 H.A. El Enshasy et al.

Fig. 7.1 Enzymatic hydrolysis of xylan (Polizeli et al. 2005)

Hardwood xylan (e.g., birchwood xylan) consists of more than 70 b-1,4 linked-
D-xylopyranose units linked by b-1,4 glycosidic bonds and 4-O-methyl-a-D-glu-
curonopyranosyl acid side chains which are found attached at second carbon of
every tenth xylopyranose units. Acetylation rate is higher in these groups and it is
quite frequent at second and third carbon atom on xylopyranose ring. In softwood
xylan, acetylation rate is zero but instead of an acetyl group they possess a-L-
arabinofuranose linked by a-1-3 glycosidic bond at the third carbon atom on
xylopyranose ring. Based on the side chains of the xylan backbone, they are
classified into different groups such as linear homoxylan, arabinoxylan, glu-
curonoxylan, and glucuronoarabinoxylan. These four groups are heterogenic in
terms of their degree and nature of branching. In many plants, xylans are found
partially acetylated, which protect them from complete degradation by xylolytic
enzymes. This could be the reason behind the fact that the complete degradation of
xylan is accomplished only by synergetic interaction of acetyl xylan esterase and
endo-xylanase enzymes.
7 Microbial Xylanases: Sources, Types, and Their Applications 155

Due to the heterogeneity and complex chemical structure of polymer, the


complete breakdown of xylan requires the synergetic action of several hydrolytic
enzymes which are specific in their mode of actions. Generally, xylans are not
attacked randomly, but the bonds selected for hydrolysis depend on several bio-
chemical factors that determine the nature of the substrate molecules such as the
chain length, the degree of branching, and the presence of substituents.
The major xylolytic enzymes involved in xylan hydrolysis are as follows:
• Endo-1,4-b-xylanase—cleaves long xylan backbone into short
xylooligosaccharides.
• b-xylosidases—also attacks the b-1,4-glycosidic linkages to liberate xylose
from short oligosaccharides.
• a-L-arabinofuranosidases—remove L-arabinofuranose side chains.
• a-D-glucuronidases—hydrolyze the methyl glucuronate residues.
• Acetyl xylan esterases—hydrolyze acetate groups on xylan backbone.
• Ferulic acid esterases—hydrolyze the aromatic acids groups linked on arabi-
nofuranoside residues.
Endo-1,4-b-xylanase: (EC 3.2.1.8) cleaves the glycosidic bonds between the D-
xylopyranose units on the xylan backbone and as a result, short xylooligosaccharides
are formed. b-D-xylosidases: (EC 3.2.1.37) liberate xylose from short xylooligosac-
charides which are formed during xylan hydrolysis by endo-1,4-b-xylanase. These
enzymes are classified based on their relative affinity for the xylooligosaccharides into
distinct entities such as xylobiases and exo-1,4-b-xylanases. Even though they are two
different enzymes, they have been recognized as b-D-xylosidases in general, and
releases b-D-xylopyranosyl residues from small xylooligosaccharides and xylobiose.
Their affinity for the xylooligosaccharides is inversely proportional to its complexity.
Xylobiose was found to be the best substrate for these enzyme groups. These groups of
enzymes play important role in xylan hydrolysis when there is endo-1,4-b-xylanase
inhibition due to the accumulation of short oligomers of b-D-xylopyranosyl units, by
hydrolyzing these products and remove the cause of inhibition thereby enhancing the
xylan hydrolysis (Andrade et al. 2004). The molecular weights of these enzyme
groups are relatively high, between 60 and 260 kDa and their pH optima is between
4.0 and 5.0. Generally, these enzymes are thermophilic in nature and their optimum
temperature varies from 40° to 80°. However, in most cases they exhibit highest
activity around 60 °C (Rizzatti et al. 2001). a-L-arabinases: These enzymes remove L-
arabinose side chains which are substituted at second and third carbon on b-D-xylo-
pyranosyl ring of arabino xylan. They have been classified as exo-a-L-arabinofur-
anosidase (EC 3.2.1.55) and endo-1,5-a-L-arabinase (EC 3.2.1.99), based on their
distinct mode of action. The first group, which is most common, degrades branched
arabinans and p-nitrophenyl-a-L-arabinofuranosidase, whereas the second group
hydrolyzes only the linear arabinans (de Vries et al. 2000). The a-D-glucuronidases
(EC 3.2.1.131) can hydrolyze the a-1,2 linkages between glucuronic acid and xylose
residues found in glucuronoxylan. They are classified under glycoside hydrolase
family 67 and rarely found in nature. During biodegradation of glucuronoxylan, these
156 H.A. El Enshasy et al.

enzymes are able to release glucuronic acid. However, the enzyme activity is only
limited on short xylooligomers due to the partial hindrance by acetyl group present in
xylan backbone (Puls et al. 1991). Acetylxylan esterase: (EC 3.1.1.6) it acts by
removing the O-acetyl groups from second and third carbon on b-D-xylopyranosyl
ring of acetyl xylan. This enzyme plays important role in xylan hydrolysis by
removing O-acetyl side chains, since O-acetyl groups in acetyl xylan interfere the
activity of endo-1,4-b-xylanase and b-D-xylosidases on xylan backbone. Xylan
extraction from hardwoods is usually mediated by alkali treatment, resulting in acetyls
hydrolysis, and probably this could be the reason of the late discovery of this enzyme
(Shao and Wiegel 1995; Blum et al. 1999). Ferulic acid esterase: Ferulic acid esterases
(EC 3.1.1.-) act by hydrolyzing the bond between arabinose side chain and ferulic acid
group attached (Crepin et al. 2004). The cooperative functioning among the
above-mentioned xylolytic enzymes enhance the complete biodegradation of xylan. It
was also observed that acetylxylan esterase activity on xylan results in deacetylation
of xylan backbone and these deacetylated xylans are more easily hydrolyzed by
endo-xylanase. Similarly, there was a significant increase in hydrolysis efficiency
when arabinoxylan was pretreated by a-arabinofuranosidase. This was due to the
removal of arabinan side chains which act as hindrance to endo-xylanase activity (de
Vries et al. 1999).

7.3 Occurrence of Endo-1,4-b-xylanase

Endo-1,4-b-xylanase are also known as endo-(1!4)-b-xylan 4-xylanohydrolase,


endo-1,4-xylanase, xylanase, b-1,4-xylanase, endo-1,4-xylanase, endo-b-1,4-
xylanase, 1,4-b-xylan xylanohydrolase, b-xylanase, b-1,4-xylan xylanohydrolase,
endo-1,4-b-xylanase, b-D-xylanase, 4-b-D-xylan xylanohydrolase, endo-(1!4)-
beta-xylan 4-xylanohydrolase, beta-1,4-xylanase, endo-beta-1,4-xylanase, endo-1,
4-beta-D-xylanase, 1,4-beta-xylan xylanohydrolase, beta-xylanase, beta-1,4-xylan
xylanohydrolase, endo-1,4-beta-xylanase, beta-D-xylanase, and 4-beta-D-xylan
xylanohydrolase. In nature, the endo-1,4-b-xylanase enzymes are widely distributed
among eukaryotes and prokaryotes. The occurence of these enzymes was also
reported in higher eukaryotes such as plants, protozoa, small insects, and several
marine species. A Japanese pear fruit was reported producing endo-xylanase during
its over-ripening period. Endo-xylanase of molecular weight 55 kDa was also iso-
lated from wheat flour (Cleemput et al. 1997). Apart from these sources, members of
higher animals such as insects and fresh water mollusc were also reported as xylanase
producers (Yamura et al. 1997). However, bacteria, fungi, and actinomycetes were
found to be the largest source of xylanase enzyme.
There have been several reports on microbial endo-1,4-b-xylanase since 1960.
Nevertheless, most of these xylanase enzymes remained unnoticed. During 1980s
first studies on xylanase-mediated bio-bleaching in pulp industry was reported and
only since then the scientific world realized the great impact of xylanase enzymes in
industrial applications. As mentioned above, a number of microorganisms including
7 Microbial Xylanases: Sources, Types, and Their Applications 157

bacteria and fungi have the capacity for hydrolyzing xylans by synthesizing a
variety of xylolytic enzymes. Early reports reveal that many of these microbial
species are plant pathogens as they play an important role in degradation and
invasion of plant tissues. It also shows that xylanases can elicit defense mechanisms
in plants by a collective functioning with other cellulolytic enzyme groups.

7.3.1 Fungal Endo-1,4-b-xylanase

Many of the fungal species that are pathogenic to plants produce plant cell wall
polysaccharide degrading enzymes. Endo-1,4-b-xylanase is one among the major
group of such enzymes and they result in partial degradation of cell wall structures
to the region of penetration (Subramaniyan 2000). A hypothetical model of fungal
invasion on plant tissue is shown in Fig. 7.2.
A number of fungal species have been used to produce xylanase since they are
the major producer of xylanases in nature. Table 7.3 shows the major fungal species
used to produce xylanases. Phanerochaete chrysosporium, a potent plant pathogen,
reported to produce xylanase activity of 15–20 U mL−1 along with considerable
amount of cellulose activity (Copa-Patino et al. 1993). Other study also showed that
the thermophilic fungus Thermomyces lanuginosus has the capacity of high xyla-
nase production up to 3576 U mL−1 (Singh et al. 2000). Many fungal species have
been reported as xylanase producers; however, species with high xylanase activity
and negligible cellulase activity are found to be very rare in nature. It was also
observed that the pH optima of many of these fungal xylanases are between pH 5
and 6 even though they are stable at pH between 3 and 8. On the other hand,
bacterial xylanases showed slightly higher pH optima that makes them suitable for
many of the industrial applications. Thermal tolerance of the majority of fugal
xylanase reported so far has been found below 50 °C and in most cases this

Fig. 7.2 Fungal invasion on plant tissue by xylolytic degradation (Prade 1996)
158 H.A. El Enshasy et al.

Table 7.3 Major fungal xylanase and its characteristics


Source Molecular weight Optimum conditions References
(kDa) Temperature pH
(°C)
Aspergillus 39, 23, 26 45–55 4.0–5.5 Kormelink et al. (1993)
awamori
A. fumigatus 19, 8.5 55 5.5 Silva et al. (1999)
A. kawachii 35, 26, 29 60, 55, 50 5.5, 4.5 Ito et al. (1992)
A. nidulans 55 8 Taneja et al. (2002)
A. oryzae 35 60 5 Kitamoto et al. (1999)
A. nidulans 22, 34 62, 56 5.5, 6.0 Fernández-Espinar et al.
(1994)
A. sojae 32.7, 35.5 60, 50 5.0, 5.5 Kimura et al. (1998)
A. sydowii 33 50 4 Ghosh and Nanda (1994)

particular property of fungal xylanase makes them less favorable for application in
harsh industrial conditions. Reduced xylanase yield in fermenter studies is another
major problem associated with fungi xylanase. This was due to poor oxygen
transfer, shear force during fermentation, and the enzyme production process in
fungi is highly regulated by growth morphology (El Enshasy et al. 1999, 2006; El
Enshasy 2007). Generally, the fungal species has filamentous growth pattern, the
fungal growth in fermenters is restricted due to the shear stress, and eventually this
results in poor xylanase production (Dean et al. 1991).

7.3.2 Bacterial Endo-1,4-b-xylanase

Since 1980s, several bacterial strains which are capable of producing alkaline and
highly thermophilic xylanase enzymes have been reported. Among these wide
spectrum of bacteria, Bacilluss species are the most predominant producers of
endo-1,4-b-xylanase enzyme with negligible amount of cellulose activity under
optimized growth conditions. Other author showed a high yield of xylanase up to
506 IU mL−1 by Bacillus SSP-34 (Subramaniyan and Prema 2002). In general,
bacterial endo-1,4-b-xylanase is suitable for industrial application since they have a
wide pH optima and improved thermostability when compared to fungal xylanases.
Ratto et al. (1992) reported Bacillus circulans strain with xylanase activity of
400 IU mL−1. They observed the optimal enzyme activity was at pH 7 and retained
40 % of activity at high pH up to 9.2. Many other researchers reported
cellulose-free xylanase enzyme from Bacillus stearothermophilus strain T6 (Khasin
et al. 1993; Lundgren et al. 1994). A relatively high xylanase activity was reported
in Bacillus sp. strain NCL when it was grown in zeolite-induced medium
(Balakrishnan et al. 2000). Thermophile bacteria such as Rhodothermus marinus
produced approximately 1.8–4.03 IU mL−1 of thermostable xylanase along with
7 Microbial Xylanases: Sources, Types, and Their Applications 159

Table 7.4 Major bacterial xylanase and its characteristics


Source Molecular weight Optimum conditions
(kDa) Temperature pH
(°C)
Aeromonas caviae ME1 20 50 7
Bacillus amyloliquefaciens 18.5–19.6 80 9
B. circulans WL-12 85 – –
Bacillus sp. strain SPS-0 99 75 –
Bacillus sp. W1 (JCM2888) 21.5, 49.5 65, 70 4.5–10, 4.5–7
Bacillus sp. strain 41-1(36) 36 50 9
Bacillus sp. strain TAR-1 40 75 6
Bacillus stearothermophilus T-6 43 75 6.5
Streptomyces T-7 20.6 60 4.5–5.5
Streptomyces sp. No 3137 50, 25, 25 60–65 6-May
Thermotoga maritima – 85 6.5
T. thermarum – 80–100 7
Modified from Subramanya and Prema 2000

detectable amount of cellulose activity. Other strain such as Bacillus circulans


showed high yield of xylanase (400 IU mL−1) which has pH optima of 7 but with a
high stability at alkaline pH (Beg et al. 2000). The concept of TCF (total chlorine
free) bleaching of pulp was developed during this period and it was achieved with
xylanase from Bacillus stearothermophilus strain T6 which has optimum activity at
pH 6.5 (Kohli et al. 2001). A detailed description of major bacterial xylanase and
their characteristics are given in Table 7.4

7.4 Biosynthesis and Regulation of Microbial


Endo-1,4-b-xylanase

Biosynthesis of xylanase enzyme by microorganisms and the phenomenon of


xylanase induction are less investigated at molecular level due to the difficulty in
setting up a cell-free system under experimental conditions. However, Srivastava
and Srivastava (1993) introduced a hypothetical model of xylanase biosynthesis and
regulation to describe similar conditions. Xylans are comparatively complex in
structure with high molecular weight and thus it is usually hard to be utilized
directly by microbes. Xylans are converted into smaller molecules such as xylose,
xylobiose, xylotriose, or xylooligosaccharides by the constitutively produced small
amount of endo-1,4-b-xylanase enzymes. These simpler molecules can be easily
utilized by microbial cell as the carbon source (Zhao et al. 1997). These low
molecular weight molecules can act as inducer for further xylanase expression and
this has been one of the possible explanations for direct induction of xylanase gene.
Xylanases are usually secreted in the media which contains pure xylan or xylan
residues in cultures of different species such as Aspergillus awamori, Trametestrogii,
160 H.A. El Enshasy et al.

and Streptomyces sp. QG-11-3 (Beg et al. 2001). However, several exceptional cases
have also been reported such as in Cellulomonas flavigena; xylan acts as a poor
inducer of xylanase gene (Avalos et al. 1996). In some rare cases, for example, in
yeast strain Trichosporon cutaneum, xylanase biosynthesis is induced by certain
positional isomers. In many bacterial species, the xylanase induction is also possible
with various sugars such as D-xylose, D-maltose, D-glucose, and D-arabinose. On the
other hand, in many fungal species, the natural lignocelluloses such as corn cobs,
rice straw, sugarcane bagasse, and wheat bran were found to be capable of triggering
the xylanase induction (Gupta et al. 2000). Other study conducted by Kumar and
Satyanarayana (2011) reported on the wheat bran-mediated xylanase induction in
Bacillus halodurans TSEV1 strain. However, the hypothetical mechanism of direct
induction is questionable when the transportation across the cell wall can be blocked
by larger molecules. Based on the study of Gomes et al. (1994), there has been a
universally supported concept based on intracellular b-xylosidases. They explains
that the larger xylooligomers formed during xylan hydrolysis are directly transported
into the cell matrix and it was further degraded into xylose residues by b-xylosidase
which are present in the cytoplasm.
Hydrolysis of xylooligomers by hydrolytic transporter proteins during their
transportation through cell membrane is another possible explanation for above
phenomenon. There are also rare cases in which the high xylose concentration in
the media inhibits the xylanase gene expression (Strauss et al. 1995). Exclusion of
inducer transport across the cell membrane is another possible reason for poor gene
expression. In similar cases such as in E. coli it has been observed that the presence
of glucose in the media prevents the lactose transportation through the cell mem-
brane which is inducer for lac operon (Borralho et al. 2002). It was also observed
that the xylanase biosynthesis is mediated by complex metabolic pathway in which
the inducer level and the level of repressor molecule to that particular inducer vary
with the organism and their growth medium. For example, the xylanase production
by Streptomycetes sp. was increased when cellulose substrates are used in the
growth media, whereas in Cellulomonas favigena, sugarcane bagasse was found to
be the best inducer of xylanase enzyme (Alejandro et al. 2007). Addition of xylose
in fermentation media increased xylanase production to a significant amount in
Bacillus sp. (Gupta and Kar 2009). However, other studies showed that the pres-
ence of readily utilizable sugars such as glucose, xylose, and galactose can suppress
xylanase biosynthesis in other strains such as in Streptomyces sp. (Bajaj and Singh
2010) and in T. reesei (Mach-Aigner et al. 2010).

7.5 Classification and Structures of Xylanase

Xylanases are considered as one of the major hydrolyzing enzyme groups as they
mediate the xylan degradation to simpler utilizable units. Activity of these enzymes
depends on the substrate specificity as well as substrate complexity. Wong et al.
(1988) classified xylanases into two major groups based on the end product of
7 Microbial Xylanases: Sources, Types, and Their Applications 161

hydrolysis reaction. They were debranching enzymes which liberate arabinose and
non-debranching enzymes which do not liberate arabinose residues from a,1,3,L-
arabinofuranosyl. A few years later, another classification system was introduced
based on the physiological properties of xylanase enzymes such as isoelectric point
(PI) and molecular weight (MW). Therefore, xylanases were classified into two
major groups: end xylanase enzymes with molecular weight lower than 30 kDa and
basic pH and those with molecular weight higher than 30 kDa and acidic pH.
However, few years later, this classification system was found incorrect since it
matches for only a few xylanase enzymes.
Henrissat and Bairoch (1993) proposed a broad classification system based on
the similarities in amino acid sequence and catalytic module. The glycoside
hydrolases are further classified into different families. This classification system
has got wide acceptance since it gives information regarding the structural prop-
erties, the catalytic mechanisms of the enzyme, and their evolutionary relationship
within the group. Xylanases were grouped under glycoside hydrolases (GH) 5, 8,
10, and 11 families. However, the enzymes with multi-domain that exhibit
detectable amount of xylanase activity are grouped into GH7, 16, 43, and 62
families (Cantarel et al. 2009). Table 7.5 comprises major xylolytic enzymes, their
classification into CE, GH families, and their current crystallographic structure.
Some of these GH families are further classified into super families that represent a
more distant common evolutionary ancestor. Major GH family xylanase and their
general structures are shown in Fig. 7.3 (Table 7.6).

Table 7.5 Xylolytic enzymes, their classification into CE, GH families, and the their current
crystallographic structure status
Enzyme Enzyme family Structure status
EC GH
Endo-b-1,4-xylanase 3.2.1.8 5, 8, 10, 11 and GH5-Cryst. (1BQC)
43 GH8-Cryst. (1IS9)
GH10-1FH7,
1CLX, 1XYZ
GH11-1H4G, 1BCX,
1IGO
GH43-(1GYD)
Exo-b-1,4-xylosidase/b-xylosidases 3.2.1.37 3, 39, 43, 52 GH3-Cryst. (1EX1)
and 54 GH39-Cryst.
GH43-(1GYD)
a-L-Arabinofuranosidase 3.2.1.55 3, 43 and 51 GH3-(1EX1)
GH43-(1GYD)
GH51-Cryst
Endo-a-1,5-arabinanase 3.2.1.99 43 1GYD
a-Glucuronidase 3.2.1.139 67 1K9D, 1GQI
Acetyl xylan esterase 3.1.1.72 1 (1JJF)
5 1QOZ, 1G66
Ferulic acid esterases 3.1.1.73 1 1JJF, 1GKK
Source Shallom and Shoham 2003
162 H.A. El Enshasy et al.

Fig. 7.3 Structures of xylanase major GH family xylanase (Collins et al. 2005a, b)

Table 7.6 CBMs of known structure linked to xylanases (Berrin and Juge 2008)
CBMs family Source of xylanase GH family PDB code
CBM-2 Cellulomonas fimi 10A, 11A 1EXG, 2XBD, 1HEH
CBM-4 Rhodothermus marinus 10A 1K45
CBM-6 Clostridium thermocellum 11A 1UXX, 1NAE, 1UY4
CBM-9 Thermotoga maritima 10A 1I8A
CBM-10 Cellvibrio japonicus 10A 1QLD
CBM-13 Streptomyces olivaceovidris 10A 1XYF
CBM-15 Cellvibrio japonicus 10C 1GNY
CBM-22 Clostridium thermocellum 10B 1DYO
CBM-36 Paenibacillus polymyxa 43A 1UX7

7.5.1 GH10 Xylanases

Most of the glycoside hydrolases which are classified under GH10 are
endo-b-1,4-xylanases. Apart from these enzyme, G10 group also consists of a small
number of endo-b-1,3-xylanases (EC 3.2.1.32), which cleave b-1,3-glycosidic
linkages randomly in b-1,3-xylans backbone. A typical GH10 xylanases has a wide
spectrum of substrate specificity as they are able to hydrolyze various forms of xylans
in nature. These enzymes are capable to attack not only the linear forms of xylan but
also the highly branched heteroxylans and xylosomes. Kolenova et al. (2006)
7 Microbial Xylanases: Sources, Types, and Their Applications 163

analyzed the hydrolysis product from glucuronoxylan and confirmed that GH10
xylanases can attack xylan at its non-reducing end if there are two or more unbran-
ched xylose residues which are present on backbone. It was also found that GH10
xylanases can even hydrolyse the highly branched arabinoxylan into small fragments.
Plant xylanases which have been identified so far are grouped under GH10 family. In
recent years many studies were carried out to characterize these plant xylanases and it
was observed that these enzymes have limited substrate specificity (Chithra and
Muralikrishna 2008). The study of Van Campenhout et al. (2007) was focused on the
characterization of GH10 xylanase isolated from barley and they reported that the
enzyme can release small xylose units from various substrates. GH10 xylanases, in
addition to their xylolytic activity, are also active against glucose-derived substrates
like aryl-cello-oligosaccharides (Charnock et al. 1997; Andrews et al. 2000).
Three-dimensional structure of xylan was first described by Atkins (1992).
Subsequently, more studies were carried out using NMR spectrometry and X-ray
crystallography techniques to obtain more carbohydrate structure details. It was
found that under crystallized condition, the xylan backbone exhibits a threefold
left-handed confirmation and the geometry showed that there is no effect on b-1,4
glycosidic bonds by the side chains of those attached. Electron diffraction pattern
study confirmed the structure and the hexagonal morphology. The hydrogen at the
fifth carbon atom on xylose ring has binding properties either intra- or interchain.
The study also suggested that the structural confirmation of D-xylose ring indicated
the di-equatorial binding property of both 1–4 and 1–3 glycosidic linkages.
The X-ray crystallographic image of endo-b-1,4-xylanases enzyme isolated from
Thermoascus aurantiacus has been used to explain the general structure of GH
family 10 xylanase (Fig. 7.4). It was found that the 32.5 kDa long polypeptide
chain with a b/a-fold TIM barrel structure consists of eight major b strands which
are arranged side by side and parallel, forming a cylinder in the center followed by
eight major a helixes. In addition, around six short helixes are also found attached
with the polypeptide chain. The catalytic domains of family 10 xylanases are found
in cylindrical shape and the overall side view resembles a ‘Salad bowl’ (Fig. 7.5).
In this configuration, the catalytic sites are seen relatively closer to the carboxyl
terminus end of xylan backbone. Since the molecular weight of these xylanase is
higher, they always exhibit low degree of oligosaccharide polymerization. Both the
disulfide bond and salt bridges present on the xylan structure improve its ther-
mostability. In the overall structure, the top phase of the molecule which is b-barrel
side has larger than the bottom face, the a-b turns. This was due to the elaborate
architecture of b–a loops (Natesh et al. 1999).

7.5.2 GH11 Xylanases

GH11 xylanase unlike the other xylanase families, they only consist of
endo-b-1,4-xylanases that are exclusive enzymes cleaves b-1,4-xylosidic bonds
between xylose monomers. These enzymes are often sub-classified based on their
164 H.A. El Enshasy et al.

Fig. 7.4 a A front view


showing the (a/b) 8 TIM
barrel fold, b salad bowl view
exposing the substrate
binding cleft of
endo-1,4-b-xylanase from T.
aurantiacus (Natesh et al.
1999)

Fig. 7.5 Three-dimensional


structure of NpXyn11A. The
protein schematic is color
ramped from the N terminus
(blue) to the C terminus (red)
(Vardakou et al. 2005)
7 Microbial Xylanases: Sources, Types, and Their Applications 165

PI values into acidic and alkaline xylanases (Joshi et al. 1997). Many studies
demonstrated the correlation between the pH optima and the amino acid residues
which are present adjacent to the catalytic site of these enzymes. In many cases, it
was observed that the acidic xylanase (pH < 5) consists of asparagine, whereas in
alkaline xylanase has arginine (Krengel and Dijkstra 1996; Fushinobu et al. 1998).
This was also confirmed by other study which reported that arginine introduction to
bacterial xylanase shifts the pH optimum to acidic range (Pokhrel et al. 2013).
Heteroxylan, xylobiose, and xylotriose are the major subunits formed during GH11
xylanase-mediated xylan hydrolysis. Further hydrolytic activities on these subunits
were found negligible; however, hydrolysis products such as xylotetrose,
xylopentose, and xylohexose are further hydrolyzed by GH11 xylanases
(Cervera-Tison et al. 2009). These enzymes cleave the xylose backbone at
unsubstituted regions which are quite away from the branched xylose present at
non-reducing end. It was also found that GH11 xylanases require minimum of three
unsubstituted consecutive xylose residues for the primary binding and initiation of
hydrolysis (Biely et al. 1997; Katapodis and Christakopoulos 2008).
Ko et al. (1992) reported the first structural description of a GH11 xylanase that
was isolated from Bacillus pumilus. But his analysis was not very precise and he
failed to deposit the 3D structure in protein database. To date, more than 100 GH11
xylanase 3D structures from different fungal and microbial species have been
solved and made available in CAZy database. Xylanases isolated from a ther-
mophilic fungi Thermomyces lanuginosus are widely used to explain the general
structure of GH11 xylanases. The structure shows a globular protein composed of
two b-sheets (b-a, b-b) with a molecular weight of 25 kDa. The outer b-sheet (b-a)
is composed of five antiparallel b strands with polar and uncharged amino acids
such as threonine and serine. The inner b-b sheet is made up of nine antiparallel b
strands of which the outer sides are developed to catalytic sites and the inner sides
are found attached with b-a forming a hydrophilic core (Torronen et al. 1994). In
general, most of the GH11 xylanases are made of only a single catalytic domain and
found in b-jelly roll structure. It also resembles the shape of partially closed right
hand in which two b-sheets represent the ‘fingers,’ the loops between b-strands of
b-b such as B7 and B8 represent the ‘thumb,’ and the twisted inner sides of b-b and
a-helix represent the ‘palm’ of the hand (Gruber et al. 1998).
While comparing these two major GH families, the GH11 family consists only
of ‘true xylanases’ displaying substrate specificity toward D-xylose containing xylan
substrates. Their catalytic versatility is lower when compared to GH10 xylanases
and the products from its action such as xylobiose and xylotriose usually required
further hydrolysis by other xylanase enzymes. The GH10 xylanases can mediate
hydrolysis until the release of xylose as end product. Another interesting feature of
GH10 xylanases is their ability to tolerate glucose-derived to a certain extend in
addition to their xylanolytic activity that exhibits in addition to their xylanolytic
activity (Collins et al. 2005a). It is generally accepted that GH10 can cleave dec-
orated regions on AX backbone and its activity is less hampered by a-L-arabino-
furanosyl and acetyl or 4-O-methyl-D-glucuronate side chains present on xylan
166 H.A. El Enshasy et al.

backbone, whereas GH11 xylanases are very specific in their action as they cleave
only at the unsubstituted regions (Biely et al. 1997). This property is also reflected
in the shape of their active binding sites, where GH10 xylanase has a shallow
groove active site which has less affinity toward the unsubstituted regions and
GH11 xylanases possess a cleft-shaped active site which has higher affinity toward
unsubstituted consecutive xylose.

7.5.3 GH8 and GH5 Xylanases

In addition to the above-mentioned GH families, GH8 family xylanase is another


most studied xylanase which consists of both endo- and exo-xylanases enzymes.
Exo-xylanases are those enzymes which reduce xylooligosaccharides (XOS) and
release xylose subunits. A very few number of endo-xylanase have been identified
so far that belong to GH8 family such as xylanases isolated from
Pseudoalteromonas haloplanktis (Collins et al. 2005a); GH8 exo-oligoxylanases
was also isolated from Bifidobacterium adolescentis (Lagaert et al. 2007) and from
Bacillus halodurans (Honda and Kitaoka 2004). In general, the diversity in sub-
strate specificity of these enzymes is large in nature and rXyn8 isolated from
P. haloplanktis is the best example which shows highest activity on mixed linkage
(b-1,3- and b-1,4-bonds) homoxylan (Pollet et al. 2010). Studies on
three-dimensional structure of GH8 xylanase isolated from P. haloplanktis xylanase
show that it is a single-domain molecule with six (a/a)-fold barrel which is found
similar to GH48 enzymes (De Vos et al. 2006). Glycoside hydrolases 5 (GH5) is
another largest enzyme family which consists of more than 1800 entries in the
CAZy database. Under this family, several enzymes with apparent activity on xylan
have been reported. Endo-xylanases from Aeromonas punctuate (Suzuki et al.
1997) and Meloidogyne incognita (Mitreva-Dautova et al. 2006) are the major
reported GH5 xylanases so far. However, certain enzymes in this group such as
xylanase from H. jecorina also exhibit exo-activity. Larson et al. (2003) was the
first one who reported the structure of GH5 xylanase (XynA) isolated from E.
chrysanthemi. In recent years, many other X-ray analysis of GH5 xylanase has been
reported. St John et al. (2009) reported X-ray crystallographic analysis of XynC
from B. subtilis, which shows a multidomain protein (consist of a catalytic domain
along with a C-terminal CBM) with (a/b) 8-fold barrel.

7.6 Catalytic Mechanisms of Endo-1,4-b-xylanase

Hydrolysis of glycosidic bonds is carried out mainly by two different catalytic


residues that are present in the active site of the enzyme. Hydrolysis takes place at
the anomeric center of the substrate molecules either by inversion or retention of the
configuration and this has been depended on factors such as the structure and
7 Microbial Xylanases: Sources, Types, and Their Applications 167

Fig. 7.6 A hypothetical model of catalytic mechanism of GH xylanase. The double-displacement


reaction mechanism for retaining glycosidases (Koshland mechanism). Modified from Paës et al.
(2012)

position of catalytic residues on the enzymes (Collins et al. 2005b). Many of the
endo-1,4-b-xylanase exhibit a double-displacement mechanism in their operation
on specific substrate molecules (Fig. 7.6). In most cases, the first acid/base catalytic
residue initiate the hydrolysis by protonating the xylopyranosyl linkages between
the xylan monomers and this process is termed as glycosylation. The second cat-
alytic residue which acts as a nucleophile attacks the same linkage and results in the
formation of enzyme–glycosyl intermediate while passing through an
oxocarbenium-ion-like transition state. In consecutive steps which are also termed
as deglycosylation, the first catalytic residue exhibits basic properties by activating
the incoming water molecule and abstracting a proton from it. The activated water
molecule readily attacks the anomeric carbon of the enzyme–glycosyl complex
formed in the previous step and release the product with an a-configuration at the
anomeric carbon (Sidhu et al. 1999; Chiku et al. 2008).

7.7 Factors Affecting Endo-1,4-b-xylanase Functionality

7.7.1 GH Family Origin

Among the above-mentioned families, endo-xylanases GH10 and GH11 are the
most studied. Endo-xylanase enzymes with relatively higher molecular weight are
grouped into family F/10 and they are found with cellulose-binding domain and a
catalytic domain that are usually connected by linker peptide. Enzyme belonging to
this family also found to have a (b/a)-fold TIM barrel structure (Biely et al. 1997),
whereas the family G/11 xylanase is generally of low molecular weight and is further
168 H.A. El Enshasy et al.

classified into two major groups based on their PI value. Similarly, b-xylosidases,
another major xylolytic enzyme is grouped under GH classification system into
various families such as 3, 39, 43, 52, and 54. Studies showed that b-xylosidases
from the families 3, 39, 52, and 54 use a retaining mechanism to hydrolyze
xylooligomers, while those from family 43 mediate the hydrolysis by inverting the
anomeric configuration. Advanced bioinformatics tools such as Basic Local
Alignment Search (BLAST) and pair-wise alignments of the protein sequences have
been used to identify the xylanase enzymes and compared with closely related
enzymes within the families. The sequences of family 10 xylanase are generally used
to identify mutually exclusive enzymes using BLAST search. Further confirmations
are also made using X-ray crystallographic studies. A continuous update to all these
GH family classifications is provided by the carbohydrate-active enzyme database
(CAZy) since there is a direct relationship between the protein folding and the
sequence. This system is universally accepted as it can provide structural features of
enzymes that help to predict their sole substrate specificity; it helps to identify and
reveal the evolutionary relationships between the enzyme groups, and also act as a
convenient tool to derive mechanistic information.

7.7.2 Carbohydrate-Binding Modules (CBMs)

The X-ray crystallographic studies on the xylanase structure reveal that most of
them have a modular structure composed of a catalytic site and one or more
carbohydrate-binding modules (CBMs) that are interconnected by flexible linkages
(Kulkarni et al. 1999; Subramaniyan and Prema 2002; Collins et al. 2005b).
A CBM is defined as a specially arranged amino acid sequence with high affinity to
carbohydrate molecules within a carbohydrate-active hydrolytic enzyme. These
unique sites are designed to bind with specific polysaccharides on plant tissue and
mediate the structural damage by enzymatic hydrolysis (Bolam et al. 2001;
Boraston et al. 2002). Similar to the catalytic modules of glycoside hydrolases,
CBMs are classified into 64 families according to their amino acid sequence sim-
ilarity and this classification is available in CAZy database (Tomme et al. 1994).
A study of Boraston and his group reported a detailed overview about the structure
and functions of CBMs (Boraston et al. 2000). However, a very few members of
GH11 xylanases carry CMB. Among these only a few of them have been experi-
mentally demonstrated, but others are hypothetically explained based on their
sequential similarities with well-studied ones. It have been also reported that certain
CBMs from Xyl-11 are specific for cellulose and this characteristic feature helps the
xylanase to localize the substrates indirectly even when they are in close association
with cellulose. Major CBMs of known structure which are linked to xylanases are
presented in Table 7.7. Xylanases isolated from fungi such as Penicillium funicu-
losum XynB (Brutus et al. 2004) and Neocallimastix patriciarum XynS20 (Liu
et al. 2008) are found to have CMB1 modules. It was also reported that the
7 Microbial Xylanases: Sources, Types, and Their Applications 169

Table 7.7 Sensitivity of Xylanase source XIP-1 TAXI-1 TLXI


GH11 xylanases to
proteinaceous inhibitors Fungi
(XIP-1, TAXI-1 and TLXI) Penicillium purpurogenum − + −
(Gebruers et al. 2008) Fusarium graminearum + − −
P. funiculosum + + +
P. funiculosum + + +
Trichoderma + + −
longibrachiatum
T. viride + + +
Bacteria
Bacillus agaradhaerens − − −
B. subtilis − + −
Fibrobacter succinogenes − − −

Fig. 7.7 Spatial


conservation of residues
paving the catalytic cleft of
Xyl-11 (Paës et al. 2012)

occurrence of a C-terminal CBM1 in Xyl-11 isolated from Podospora anserina


found to have significantly improved ability to reduce the sugar during the
hydrolysis of wheat straw (Couturier et al. 2010). Figure 7.7 shows the
three-dimensional structure of NpXyn11A which belongs to GH11. Some GH11
xylanases, apart from the CBMs, also consist of dockerin domains that are key
elements which help in the formation of cellulose-binding multi-enzyme complex
known as cellulosomes. These enzyme complexes are originally observed in
Clostridium thermocellum (Lamed et al. 1983). In addition to cellulolytic enzymes,
cellulosomes are also found with hemicellulase such as xylosidases, xylanases, and
arabinofuranosidases. Other research showed also the similar results in XynC
isolated from Fibrobacter succinogenes S85 (Marrone et al. 2000).
170 H.A. El Enshasy et al.

7.7.3 Xylanase Enzyme Inhibitors

Almost all the plant species have developed a self-defense mechanism as response to
the pathogenic microbial attack by hydrolases enzymes. Xylanase inhibition by the
proteinaceous inhibitors is one among those defense response widely seen in both
soft- and hardwood plants. A numerous studies have been carried out to understand
this defense mechanism since 1990s and it was found that Triticum aestivum
xylanase inhibitor (TAXI) and xylanase inhibitor protein (XIP) are the two major
groups of proteins responsible for Xyl-11 in specific (Debyser et al. 1999; Rouau
et al. 2006). Apart from the above two groups, the third group of inhibitor known as
thaumatin-like xylanase inhibitor (TLXI) also been isolated from wheat. It was also
found that none of these xylanase inhibitors are effective against plant origin xyla-
nase enzymes (Dornez et al. 2010a, b). Occurrence of these inhibitors seems to be a
significant hindrance in xylanase-mediated industrial process such as bread making
and brewing as they inhibit effective hydrolysis of substrates (Sorensen and Sibbesen
2006). Therefore, it has been an important topic of research to understand the mode
of action of these inhibitors. Sensitivity of GH11 xylanases to proteinaceous inhi-
bitors (XIP-1, TAXI-1, and TLXI) are as given in Table 7.7 (Fig. 7.8).

Fig. 7.8 a Structure of the inhibition complex between P. funiculosum Xyl-11 (in green) and
XIP-I inhibitor (in blue). XIP-I inhibiting loop is in red, catalytic residues are in yellow.
b Structure of the inhibition complex between B. subtilis Xyl 11 (in green) and TAXI-IA inhibitor
(in blue). His374 residue is in red, catalytic residues are in yellow (Paës et al. 2012)
7 Microbial Xylanases: Sources, Types, and Their Applications 171

7.7.3.1 Triticum Aestivum Endo-xylanase Inhibitor (TAXI)

These are proteins with pI values of  8.0, which occur in two different molecular
forms known as A and B (Gebruers et al. 2008). Form A is seen as full-length
proteins with molecular mass near to 40 kDa, whereas the form B consists of two
polypeptide chains (a longer chain and a shorter chain of size 28–30 and 10–
12 kDa, respectively) that bound together by disulfide bonds (Croes et al. 2009).
These inhibitors are seen largely in wheat grains during the early stages of grain
development and specifically inhibit GH11 xylanase disrespect to their origin
(Tables 7.8, 7.9). Further analysis of originally identified TAXI inhibitor proteins
revealed that it is a mixture of two different proteins of different pI values. They are
named as TAXI-I and TAXI-II and have different inhibition activities on GH11
endo-xylanase isolated from A. niger (Gebruers et al. 2004). Other research carried
out by Raedschelders et al. (2004) identified and isolated the genes coding for these
two inhibitor proteins from ray and barley grains and based on this study Fierens
et al. (2004) developed a Pichia pastoris-based expression system for the
large-scale production. By 2005, two more new TAXI class named TAXI-III and
TAXI-IV were also identified (Igawa et al. 2005). TAXI-type inhibitors are found
to be very specific in their inhibitory characteristics. The available research data
shows that they inhibit only GH11 endo-xylanases of bacterial and fungal origin.
However, the subclass TAXI-II was turned to be an exception, as they are unable to
inhibit GH11 endo-xylanases such as XynBc1 and ExlA isolated from B. cinerea
and A. niger, respectively (Brutus et al. 2005).

Table 7.8 Occurrence of TAXI-type inhibitors in different cereals (Raedschelders et al. 2004)
Cereal Inhibitor proteins
TAXI-I TAXI-II Inhibitor level (ppm)
Wheat + + ca. 38
Durum wheat + + ca. 11
Rye + − ca. 21–37
Barley + − ca. 4–5
Maize, rice, oats, buckwheat − − −

Table 7.9 Occurrence of Cereal Inhibitor proteins


XIP-type inhibitors in
XIP-I Inhibitor level (ppm)
different cereals
(Raedschelders et al. 2004) Wheat + ca. 80
Durum wheat + ca. 14
Rye + ca. 95
Barley + ca. 3
Maize + ca. 2
Rice, oats, buckwheat − −
Sorghum − −
172 H.A. El Enshasy et al.

7.7.3.2 Endo-xylanase Inhibiting Proteins (XIP)

Basically, these are glycosylated single-chained proteins with molecular weight of


30 kDa and its pI value falls between 8.0 and 9.0. The crystallography study shows
that it has similar structural features of GH18 with typical (a/b) 8-fold TIM barrel
shape (Payan et al. 2003). However, they are found to be passive on GH18 enzymes
such as chitinases due to the high degree of mismatch at its enzyme biding site. In
general, these inhibitors consist of two independent enzyme-binding sites that
mediate binding with almost all GH10 and GH11 endo-xylanases that are fungal
origin. However, GH11 endo-xylanase from Neocallimastix patriciarum (Payan
et al. 2004) and GH10 endo-xylanase from Aspergillus aculeatus (Flatman et al.
2002) are found to be insensitive to XIP. Similar to TAXI-type inhibitors, XIP-type
inhibitors are also categorized into different polymorphic families based on the
recent proteomics studies. Followed by identification of Xip-I (the most common
inhibitor in the XIP) class, Durand et al. (2005) identified Xip-II class in T. tur-
gidum ssp. In addition to this, Xip-III type inhibitor encoding gene was discovered
in T. aestivum and was observed that these genes are transcribed very rarely under
stressed leaves. Later on, several other XIP-type genes were also identified known
as Xip-R genes, OsXip that are of plant origin (Takahashi-Ando et al. 2007). Biely
et al. (2008) confirmed the existence of XIP-type inhibitors in maize leaves and
roots during the first week after germination.

7.7.3.3 Thaumatin-like Xylanase Inhibitor (TLXI)

Thaumatin-like endo-xylanase inhibitors (TLXI) consist of a single amino acid


chain with molecular weight of 18 kDa. This type of inhibitor was first discovered
in wheat by Fierens et al. (2007). These proteins have high pI and usually exhibit
glycosylation at varying extents that found to have a very high stability (no
denaturation was observed at 100 °C at extreme pH ranges even after 120 min).
These inhibitors are grouped into class 5 pathogenesis-related proteins as they show
very high sequential similarity with certain antifungal proteins (Trudel et al. 1998).
Even though there are no crystallographic studies that have been reported on
structure of TLXI inhibitor, the structure has been predicted and shows the exis-
tence of five disulfide bridges and three b-sheets (Fierens et al. 2009). The inhi-
bitory mechanism of these proteins was found to be specific against Xyl-11. The
xylanase inhibitors were recently reviewed by Dornez et al. (2010a, b).

7.8 Recombinant Endo-1,4-b-xylanase

The advances in molecular biology and genetic engineering opened up several new
applications of recombinant DNA technology. The new opportunity for the con-
struction of GM microbial strains with selected enzyme machinery was one of the
7 Microbial Xylanases: Sources, Types, and Their Applications 173

greatest breakthroughs. On the basis of this, more efficient endo-1,4-b-


xylanase-producing microorganisms were constructed to improve the biodegrada-
tion of hemicellulosic residues and thus find many industrial applications. In this
section, we will highlight in detail the recent advance in recombinant xylanase
production. Moreover, studies on xylanase gene regulation and biosynthesis at
molecular level and various tools applied to enhance the novel characteristic of
xylanase enzymes will be discussed.
Several xylanases have been isolated and identified from various sources.
However, very limited numbers of these enzymes are found to be suitable for
industrial applications due to their poor performance in harsh process conditions.
Poor thermostability is the biggest issue associated with most of the xylanases.
Enzymes with higher thermostability can enhance the efficiency of industrial pro-
cess with an increased residual time. Therefore, there has been a huge demand for
thermostable xylanases in pulp and paper industry as they are applied during pulp
bleaching process which usually carried out at temperatures more than 90 °C. Most
of the xylanases we have today are not able to withstand this high temperature.
However, highly thermophilic xylanases have been reported from thermophilic
bacteria such as G. thermoleovorans and Thermotoga sp. (Sharma et al. 2007).
Alkali stability is also another major problem associated with industrial applica-
tions. It was observed that most of the highly thermophilic xylanases are less stable
under alkaline conditions which limit their potential use in industries. Most of the
xylanases reported so far have optimal activity at either acidic or neutral pH (Wang
et al. 2010). Exceptional cases are also reported, such as xylanases from Bacillus
halodurans which exhibit high alkali tolerance (Mamo et al. 2006), but these
enzymes are found to have poor thermostability. Cellulose-free xylanase is another
desirable characteristics required in pulp bleaching industries to mediate efficient
hydrolysis without disturbing the cellulosic content of the pulp. Studies also shows
that low molecular weight xylanases are more efficient in pulp bleaching as they can
easily diffuse into pulp fibers. In practical and economical point of view, growing
these extremophilic microorganisms in large scale is not feasible. Moreover, it is
difficult to have a single enzyme with all desired characteristics. Conventional
enzyme stabilization techniques such as helix capping and salt bridging (Puchkaev
et al. 2003) have been practiced to improve the enzyme characteristics. However,
these methods are found to be ineffective when the enzyme is required in large
quantities. To overcome these problems and enhance the large-scale production for
the industrial application, recombinant xylanases were introduced using gene
cloning and expression techniques. rDNA technology has been widely used
nowadays to make alteration or modifications in existing wild-type xylanase to
improve their specific characteristics such as thermostability and alkaline stability.
A number of xylanase coding genes from different microbial species have been
isolated and expressed successfully in suitable expression hosts. The expression
level of xylanase gene in host cell usually depends on several factors such as
cloning strategies used, host cell, biosynthesis mechanism, gene regulation, fer-
mentation media, and many bioprocessing parameters. Bacterial and fungal species
are generally used to express xylanases in large scale. The cloning vector selection
174 H.A. El Enshasy et al.

is usually carried out based on the expression host and a wide variety vectors have
been introduced in last few decades. Even though the expression rate is higher in
recombinant strain, the enzyme activity is usually less than in the native producer
strain. Several reasons have been identified for this activity loss and the intercellular
accumulation of the enzyme due to the lack of post-translational modification is
suggested to be the key reason (Schlacher et al. 1996).

7.8.1 Cloning and Expression of Fungal


Endo-1,4-b-xylanase Gene

It has been more than three decades since filamentous fungi are used in the pro-
duction of xylanase enzyme. Fungal cells are the widely used biofactories in
large-scale production of xylanases and have been considered as more potent
xylanase producers as they are capable of secreting much higher amounts of xyla-
nolytic enzymes in fermentation media than bacteria or yeast (Bergquist et al. 2002;
Fang et al. 2008). Large numbers of fungal strains are widely used for xylanases
production such as many species from Penicillium, Aspergillus, Fusarium,
Trichoderma, and Disporotrichum. However, in many of these fungi, the difficulty
in isolating pure form of xylanase was the major problem since they generally
exhibit a low amount of cellulase activity. Recombinant DNA technology has been
widely applied to overcome this drawback and production of xylanase expression
systems which are free of cellulase activity which improved its acceptability in
industrial applications. Apart from the bacterial expression hosts, filamentous fungi
are also attractive hosts for xylanase expression. It is because of their ability to
secrete the proteins into the fermentation media in large quantities. Aspergillus and
Trichoderma species are the widely used expression hosts in homolog expression
system. Kitamoto et al. (1999) successfully overexpressed XynF1 gene in A. oryzae
under a strong TEF1 promoter in glucose-based submerged fermentation. XynF1,
XynF3, and XynG2 are another important xylanase genes expressed efficiently in
A. oryzae under stronger promoter called P-No 8142 in similar fermentation con-
ditions. Aspergillus niger is another attractive host for recombinant xylanases
production due to its ability to produce and secrete protein in high capacity up to
30 g L−1 in the fermentation media (Hessing et al. 1994). Xylanase coding gene
from T. reesei was successfully expressed in A. niger cD15 stain under
G6P-dehydrogenase promoter and glaA terminator which is isolated from
Aspergillus awamori (Rose and van Zyl 2002). This recombinant xylanase retained
its 75 % activity even after 3 h of incubation at 50 °C. The optimal condition of the
enzyme activity was observed at temperature between 50 and 60 °C and pH 5–6.
Levasseur et al. (2005) also reported on the potential use of Aspergillus niger as host
to express xynB under promoter sequence isolated from Aspergillus nidulans. They
showed that, A. nidulans can be used as efficient host for xylanase expression and
production. Kimura et al. (1998) expressed xynG1 in A. nidulans under its own
promoter showed that the xylanase expression was induced in the presence of
7 Microbial Xylanases: Sources, Types, and Their Applications 175

Table 7.10 Major fungal xylanase expression system


Source of xylanase gene Expression host Vector MWt References
(kDa)
Heterologous expression host
Aspergillus niger P. pastoris GS115 pHIL-D2 30 Berrin et al.
(2000)
A. usamii E001 E. coli BL21 pET-28a 20 Zhou et al.
(+) (2008a)
Thermobifida fusca NTU22 P. pastoris KM71H pPICZa A 36 Cheng et al.
(2005)
Cochliobolus sativus E. coli SOLR Lambda 25 Emami and Hack
ZAP (2001)
A. niger BCC14405 P. pastoris KM71 pPICZaA 21 Ruanglek et al.
(2007)
Aureobasidium pullulans E. coli pYES2 – Ohta et al. (2001)
Penicillium sp. 40 E. coli DH5a pUC119 20.7 Kimura et al.
(2000)
Trichoderma reesei Rut E. coli BL21 (DE3) pET-28a 24 Jun et al. (2008)
C-30
T. reesei PC-3-7 E. coli JM109 pT7Blue-T 33.1 Ogasawara et al.
(2006)
Homologs expression host
A. oryzae KBN 616 Aspergillus oryzae pNAN8142 32 Kimura et al.
KBN616 (2000)
A. niger BRFM281 Aspergillus niger pAN52.3 23 Levasseur et al.
D15#26 (2005)
Acrophialophora nainiana Trichoderma reesei pHEN11 19 Salles et al. (2007)
Rut C-30 exp
Orpinomyces sp. PC-2 Hypocrea jecorina pT3C 28 Li et al. (2007)

glucose in the media. Filamentous fungi enable extracellular secretion of heterolo-


gous protein which facilitates the downstream processes and ease the purification
and characterization of the produced enzyme. Moreover, most genes from fungi have
introns and filamentous fungi are able to recognize them and express with minimal
miscoding. In addition, fungal proteins are glycosylated and the protein expression
in another filamentous fungi results in similar glycosylation pattern. For expressing
highly hemophilic xylanase, many researches have been selected filamentous fungi
because of above-mentioned advantages when compared to other expression hosts
like bacteria. Table 7.10 shows major fungal xylanase expression system.
Yeast-based heterologous gene expression is considered as an excellent alter-
native system when based on their ease scaling up compared to fungi. The presence
of eukaryotic post-translational modification mechanism is one of the main reasons
behind the wide application of yeast expression systems in industry (Cheng et al.
2005). S. cerevisiae is one of the important hosts for xylanase production by cloning
of xylanase cDNA obtained by RT-PCR through crossing over. The presence of
intron sequence (lake of proper splicing of introns) that hinders the xylanase gene
expression is the major disadvantage of these expression systems (Moreau et al.
176 H.A. El Enshasy et al.

1992). It has been also observed that a significant increase in xylanase expression in
yeast transformed with the xynA gene without intron (26.2 U mL−1) when compared
to the expression from the gene with intron (16.7 U mL−1) (Li and Ljungdahl 1996).
It is also worthy to note that Chavez et al. (2004) made the first successful attempt to
clone and express the xynA gene which is isolated from P. purpurogenum in S.
cerevisiae. Isolated xynA gene that consists of eight introns regions was spliced
correctly and was integrated on yeast chromosome. The Xln A gene was then
expressed under transcriptional control (XlnR and CreA) where xylose or xylan acts
as inducer and glucose acts as repressor (van Peij et al. 1997). The methanotropic
yeast Pichia pastoris is another widely used xylanase expression host which has
many advantages over S. cerevisiae such as improved secretion efficiency, ease to
attain high cell density culture in inexpensive media, and relative ease in scaled up
fermentation process (Cereghino and Cregg 2000). Moreover, the presence of strong
and regulatory alcohol oxidase (AOX I and AOX 11) promoters which are involved in
methanol metabolism promotes the overexpression of xylanase gene upon the
addition of methanol in the fermentation media (Tsai and Huang 2008). Table 7.11
summarizes some examples of successful cloning of fungal xylanase in yeast.

7.8.2 Cloning and Expression of Bacterial


Endo-1,4-b-xylanase Gene

La Grange et al. (2000) successfully co-expressed xylanase genes (xln B from


Bacillus pumilus along with xyn2 isolated from T. reesei) in yeast S. cerevisiae. E
coli-based expression systems are widely used to express the xylanase.
Endo-1,4-b-xylanase coding genes isolated from A. usamii were cloned in E. coli
BL-21 with the help of PET-28a plasmid vector and expressed successfully and a
maximum xylanase activity of 49.4 U mg−1 was reported. The gene expression was
induced by IPTG and the protein was expressed with 6-X histidine tags that
facilitate the purification of expressed protein (Zhou et al. 2008a). Similarly, for the
xylanase gene from B. circulans Teri-42 when cloned in E. coli DH5-alpha using

Table 7.11 Cloning of different fungal xylanase genes in yeast


Xylanase source Expression host Vector MWt (kDa) References
Aspergillus niger P. pastoris GS115 pPIC9K 20 Liu et al. (2006)
A. niger P. pastoris GS115 pHIL-D2 30 Berrin et al.
(2000)
A. niger BCC14405 P. pastoris KM71 pPICZaA 21 Ruanglek et al.
(2007)
A. sulphureus P. pastoris X33 pGAPZaA – Cao et al. (2007)
Aureobasidium pullulans P. pastoris GS115 pPIC3.5 39 Tanaka et al.
(2004)
A. pullulans S. cerevisiae INVSc1 pYES2 – Ohta et al. (2001)
Thermobifida fusca NTU22 P. pastoris KM71H pPICZa A 36 Cheng et al.
(2005)
T. fusca P. pastoris GS115 pPIC9K 31 Sun et al. (2007)
7 Microbial Xylanases: Sources, Types, and Their Applications 177

pUC19 plasmid vector, the xylanase expression was found to be reduced. However,
when the same gene is expressed in B. subtilis using plasmid pBA7, 14-fold
increase in expression was observed. Therefore, Bacillus became more attractive
hosts compared to E. coli and thus used widely in heterogeneous protein produc-
tion. Bacillus sp. used in industrial fermentation are non-pathogenic in nature and in
bacillus-based expression, the proteins are usually secreted out of the cell.
Moreover, they are gram-positive bacteria and therefore, they do not produce
endotoxins (lipopolysaccharides). These features could be of great advantage by
reducing some purification steps in terms of breaking the cells and extraction from
large number of intracellular proteins.

7.8.3 Overexpression in Escherichia coli

For the past several decades, E. coli has been recognized as the ideal platform for
expression of recombinant proteins. Factors such as rapid growth on inexpensive
media, non-pathogenicity, easy to make gene modification without effecting the cell
growth, easy methods required for isolation and purification of expressed proteins
made E. coli the most successful expression host for industrial use. In spite of these
advantages, due to the lake of specific post-translational modification such as
glycosylation and disulfide bond formation, many heterologous proteins were not
successfully expressed in E. coli. Xylanase was one of those which require
N-glycosylation during post-translation stage, whereas E. coli only can perform
O-glycosylation. However, many exceptional cases such as successful expression
of a glycosylated b-xylosidase gene isolated from thermophilic fungi
P. thermophile have been reported (Teng et al. 2011). It was also observed that the
endo-1,4-b-xylanase expression levels are significantly higher in gram-positive
hosts such as Lactobacillus species and B. subtilis compared to E. coli as they can
perform N-glycosylation.
In general, the recombinant xylanases expressed in E. coli are accumulated in the
cytoplasm or in periplasmic place in the absence of a proper secretory channel on cell
wall (Schlacher et al. 1996). However, in many cases, xylanase activity has also been
detected outside the cell (Karlsson et al. 1997; Ebanks et al. 2000). Xylanase
expression rate in E. coli mainly depends on the efficiency of transcription of
xylanase gene which is cloned under promoter sequence. Even though there are
several cloning vectors and host strains have been introduced, eukaryotic gene
expression in E. coli is unable to carryout due to the absence of a functional promoter
sequence. Basaran et al. (2001) isolated b-xylanase gene from Pichia stipites and
successfully cloned and expressed in E. coli. The recombinant enzyme activity was
low (4 U mg−1) when compared to the activity original enzyme activity (30 U mg−1)
in wild stain. Although a variety of cloning vectors have been used, pET expression
vector systems were reported to be the most efficient for protein expression in E. coli.
Xylanase genes isolated from different sources are cloned under the control of a
strong T7 promoter which is isolated from bacteriophage genome. This promoter
178 H.A. El Enshasy et al.

Table 7.12 Major cloning vectors used in E.coli-based expression system


Source of xylanase gene Cloning vector References
Bacteria
AeromoA75:C93nas sp. 212 pBR 322 Kudo et al. (1985)
Bacillus circulans pUC 19 Yang et al. (1988)
B. polymyxa pUC 13, pBR 322 Yang et al. (1988)
B. ruminicola pUC 18 Whitehead and Hespell (1989)
Butyrivibrio fibrosolvens pUC 19 Mannarelli et al. (1990)
Caldocellum saccharolyticum k1059, pBR 322 Luthi et al. (1990)
Cellulomonas sp. pUC 18 Bhalerao et al. (1990)
Chainia sp. pUC 8, kgt 10 Chauthaiwale and Deshpande (1992)
Clostridium stercorarium F9 pBR 322 Sakka et al. (1991)
C. thermocellum pUC 8 Hu et al. (1991)
Neocallimastix patriciarum kZAP II Lee et al. (1993)
Fungi
Aspergillus oryzae KBN 616 pNAN-d Kimura et al. (2000)
A. usamii E001 pET-28a(+) Zhou et al. (2008b)
Aureobasidium pullulans pCRII, k ZapII Li and Ljungdahl (1996)
Y-2311-1
Cochliobolus sativus k ZAP Emami and Hack (2001)
Helminthosporium turcicum H-2 pBluescript SK(+) Degefu et al. (2004)
Neocallimastix patriciarum pBTac2 Xue et al. (1992)
Penicillium sp. 40 pUC119 Kimura et al. (2000)
Trichoderma reesei Rut C-30 pET-28a Jun et al. (2008)
T. reesei PC-3-7 pT7Blue-T Ogasawara et al. (2006)
T. reesei C30 pGEM5Z(+) Torronen et al. (1994)

remains silent until T7 RNA polymerase is produced by the host cell genome.
Table 7.12 shows the major cloning vectors used in E. coli-based expression system.
Ogasawara et al. (2006) amplified xynIII gene from T. reesei genomic DNA
using a RT-PCR and was cloned and expressed in E. coli JM109 using pAG9-3
plasmid vector. When the xylanase gene expression was induced by IPTG, they
could produce 26 mU mL−1 of xylanase activity in cellular extract, which was very
low. However, the XynIII activity increased by about 500-fold (13.2 U mL−1)
compared to the activity in soluble supernatant, when the xylanase inclusion bodies
were refolded in 8 mM urea solution. Zhou et al. (2008a) reported the maximum
xylanase activity up to 49.4 U mg−1 when they cloned and expressed xynII cDNA
isolated from A. usamii in E. coli BL21-Codon plus (DE3) RIL strain using
pET-28a (+) expression vector. The xylanase protein expressed is attached to
6X-His-tag which ease protein purification by affinity chromatography. Jun et al.
(2008) also used the same pET vector to clone and express a b-xylanase gene, xyn2,
isolated from T. reesei in E. coli and reported the enhanced xylanase activity of 650
U mg−1. In some cases, the heterologous cloning and expression results in the
7 Microbial Xylanases: Sources, Types, and Their Applications 179

change in original characteristics of xylanase gene. For example, a


non-glycosylated XynA gene when expressed in E. coli exhibited significant loss in
its substrate specificity when compared to the glycosylated xylanase from the wild
strain P. stipites. The stability of the enzyme also decreased due to the change in
optimum temperature rage. However, improved thermostability and pH stability
have been reported when Xyn2 from T. reesei was expressed in E. coli. Table 7.13
shows the properties of major endo-b-1,4-xylanase enzyme expressed in E. coli.

Table 7.13 Properties of major endo-b-1,4-xylanase enzyme expressed in E. coli


Source of xylanase gene MWt Optimum Optimum temperature References
(kDa) pH (°C)
Geobacillus 45 8.5 80 Verma and
thermoleovorans Satyanarayana
(2012)
Paenibacillus macerans 205 4.5 60 Dheeran et al.
IIPSP3 (2012)
Thermoanaerobacterium 50 6.5 63 Hung et al.
saccharolyticum NTOU1 (2011)
Clostridium sp. TCW1 53, 60 6 75 Lo et al. (2010)
Actinomadura sp. S14 21, 30 6 80 Sriyapai et al.
and 40 (2011)
Alicyclobacillus sp. A4 42.5 7 55 Bai et al. (2010)
Anoxybacillus sp. E2 38.8 7.8 65 Wang et al.
(2010)
Nesterenkonia 22 7 55 Kui et al. (2010)
xinjiangensis CCTCC
AA001025
Bacillus sp. JB 99 20 8 70 Shrinivas et al.
(2010)
Paenibacillus sp. DG-22 20 6 60 Lee et al. (2007)
Bacillus licheniformis 77-2 40 8 75 Damiano et al.
(2003)
Enterobacter 43 9 100 Khandeparkar
sp. MTCC-5112 and Bhosle
(2006)
Geobacillus sp. MT-1 36 7 70 Wu et al. (2006)
Bacillus sp. 44 6.5, 8.5 50 Sapre et al.
and 10.5 (2005)
Bacillus thermantarcticus 45 5, 6 80 Lama et al.
(2004)
Bacillus thermoleovorans
K-3d 40, 69 7 70, 80 Sunna et al.
(1997)
Bacillus flavothermus 80– 7 70 Sunna et al.
LB3A 130 (1997)
Thermotoga sp. FjSS3-B. 31 5.5 80 Simpson et al.
1 (1991)
180 H.A. El Enshasy et al.

7.9 Improvement of Endo-1,4-b-xylanase Characteristics

In general, enzymes are classified based on their conserved regions on them.


Xylanases are specific in their identity due to the random variations in amino acid
sequence and it was found that some very specific stretches of amino acids defines
the structural and functional behavior of enzyme. Existing xylanases can be
improved in terms of their activities and other novel characteristics such as substrate
specificity, thermostability, and pH stability using protein engineering approaches.
Critical amino acid exchanges are usually practiced in order to enhance the char-
acteristics of existing xylanase enzymes. Therefore, before amino acid sequence
modification a detailed studies on the native xylanase amino acid sequences are
required to fully understand the structural and functional behaviors such as the
presence of catalytic domains, specificity of substrate-binding sites, occurrence of
thermo-stabilizing domains, and the presence of covalent/non-covalent interactions.
The significance of a particular acid or a set of amino acids for novel enzyme
characteristics can be identified easily using specific amino acid inhibitors such as
N-bromosuccinimide (tryptophan), phenylmethylsulfonyl fluoride (serine), and iodo
acetate (cysteine). It was also found that cysteine plays critical role in the formation
of covalent glycosyl enzyme intermediate during most bacterial xylanase activity.
Many of the family 10 xylanases possess three different amino acids such as Phe4,
Trp6, and Tyr 343. They were identified as crucial in protein folding and enzyme
stability even under extreme conditions. Torronen and Rouvinen (1997) studied the
significance of asparagines and aspartic acid on the pH optima of family 11 xyla-
nases. The studies conducted on xylanase isolated from B. circulans revealed that
the presence of asparagine at 52nd position of amino acid sequence increased the
thermal fluctuations at unstable regions. However, when it is substituted with tyr-
osine, the overall thermostability of the enzyme exhibited significant increased (Joo
et al. 2011). It was also reported that the catalytic activity of B. circulans xylanase
increased when the Asn at position 35 is mutated to Asp (Li and Wang 2011). Other
study showed also a significant increase in thermostability (10–15 °C) as a result of
point mutation (aspartic acid to asparagine) at position 56 (Yin et al. 2010).
However, in many other cases, the point mutation was reported to have negative
effects such as loss in substrate-binding affinity, alkaline stability, and thermosta-
bility. Simpson et al. (1999) reported the complete loss of carbohydrate-binding
module affinity toward the xylan when the arginine was mutated to glycine. In case
of B. circulans xylanase, change in pH optima was reported when R73V and
Q167M were mutated (Yang et al. 2008). Table 7.14 comprises significant
achievements made in xylanase protein engineering to increase enzyme stability.
7 Microbial Xylanases: Sources, Types, and Their Applications 181

Table 7.14 Significant achievements made in xylanase protein engineering (Verma and
Satyanarayana 2012)
Xylanase source Approaches/alterations Significant inferences References
Bacillus sp. NG-27 Deletion of Phe4 and Complete loss in activity, Bhardwaj
Trp6 while Trp6 and Tyr343 et al.
affected folding (2010)
B. circulans Amino acid Slight increase in half-life Joo et al.
modification (N52Y) and Tm as compared to (2011)
wild type
B. circulans Asn35Asp Improvement in catalytic Li and
activity Wang
(2011)
N. patriciarum D57N Enhancement in You et al.
thermostability by 10– (2010)
15 °C
G. stearothermophilus Substitution with Significant improvement Sun et al.
N-terminal of T. fusca in thermostability (2007)
T. reesei Improvement of Half-life of the mutant Turunen
C-terminal packaging xylanase was increased et al.
by 63 min (2001)
T. lanuginosus Introduction of Improvement in Gruber
DSM5726 disulfide bond thermostability et al.
between Cys100 and (1998)
Cys154
B. stearothermophilus Introduction of Enhancement in Jeong
No. 236 between Ser and Cys) thermostability by 5 °C et al.
100 and (Asn to Cys) (2007)
150
A. niger BCC144505 Substitution of Enhancement in the Sriprang
arginine in place of thermostability et al.
Ser/Thr (2006)
Thermobifida fusca Error prone PCR to Improved stability at Wang and
obtain a mutant having alkaline pH, 4.5-fold Xia (2008)
T21A, G25P, V87P, increase in Km and
I91T, and G217L 12-fold enhancement in
Kcat/Km
Thermomyces Error prone PCR Alkali-stable mutant Stephens
lanuginosus A54T et al.
(2009)

7.10 Industrial Production of Endo-1,4-b-xylanase

Although many microbial species such as bacteria, fungi, and actinomycetes have
been reported as xylanase producers, a few number of strains are acceptable by the
industrial society. To meet the current and forecasting market demands, xylanase
production need to be increased by many folds through efficient production
182 H.A. El Enshasy et al.

Table 7.15 Major commercial xylanases produced by SmF techniques (Polizeli et al. 2005)
Commercial Distributors Microorganism Application
name
Allzym PT Alltech Aspergillus Animal feed
niger improvement
Bio-Feed Plus Novo Nordisk Humicola Animal feed
insolens
EcopulpX-200 Primalco Trichoderma Cellulose pulp bleaching
reesei
Ecosane Biotec Trichoderma Animal feed
reesei
Grindazym GP, Danisco ingredients A. niger Bird and pig feed
GV
Irgazyme 40 Nalco-Genencor, Ciba, T. Paper industry and
Geigy longibrachiatum animal feed
Solvay Solvay Enzymes T. reesei Starch and bread making
pentonase
Xylanase Seikagaku Trichoderma sp. Carbohydrate structural
studies
Xylanase GS35 Iogen T. reesei Pulp bleaching

strategies. Therefore, optimizations of production and purification processes are the


key to obtain maximum xylanase yield either by solid-state fermentation (SSF) or
submerged fermentation (SmF) (Narang and Satyanarayana 2001). Nowadays, the
most of commercial xylanases production processes are carried out by SmF as
shown in Table 7.15. Cultivation media plays important role in the production of
xylanases in industrial scale. A suitable and economically viable media formulation
which supports the maximum xylanase production is usually required for
large-scale production. Therefore, abundantly available natural agriculture residues
are widely used for xylanase production. Agricultural wastes such as corn cob, con
stover, and wheat straw were successfully used as suitable substrates for fungal
xylanases production (Damiano et al. 2003; Gupta et al. 2000).

7.10.1 Physical and Nutritional Parameters Affecting


Endo-1,4-b-xylanase Production

In SSF, biomass and xylanase productions depend on several factors such as the
type of microorganism, inoculums size, temperature, oxygen diffusion, the period
of cultivation, the substrate used and its pretreatment, size, water activity, relative
humidity, pH, and many other factors. On the other hand, for SmF, the temperature,
pH, agitation, and dissolved O2 are the most critical physical parameters affecting
xylanase production in SmF. Most of the xylanase-producing microorganisms
reported so far are either mesophilic or thermophilic (Coughlan and Hazlewood
7 Microbial Xylanases: Sources, Types, and Their Applications 183

1993). It was observed that these microbes show highest xylanase activity and
optimum growth at temperature between 40 and 70 °C. However, those extremo-
philic such as Thermotoga sp., Dictyoglomus, and Geobacillus exhibit even higher
optimum temperature range of up to 80 °C (Ko et al. 2011; Sharma et al. 2007;
Qureshy et al. 2002). Xylanases of fungal origin usually are found less thermophilic
in nature. Even though some exceptional cases have been reported, the optimum
temperature for most fungal xylanases is below 55 °C. This has been one of the
major drawbacks of fungal xylanase in industrial application. On the other hand,
bacterial xylanases exhibit higher thermostability. The pH of the fermentation
media is another major factor effecting the xylanase production as well as the
enzyme stability. Fungal xylanases usually are highly alkaliphilic as they can be
active at wide ranges of pH, usually between 5.0 and 10.0, whereas the bacterial
xylanases have quite narrow pH spectrum (Dhillon et al. 2000). Aeration and
agitation rates during fermentation are the other crucial factors in case of xylanase
production process. Studies show that xylanase production by Bacillus sp. is
enhanced at an agitation rate of 200–250 rpm and 30 % and above dissolved O2
saturation (Anuradha et al. 2007; Sanghi et al. 2009). In cretin fungal species which
exhibit mycelia growth pattern, higher agitation rate results in poor growth as well
as less xylanase production due to the high sheer force. Studies made by Archana
and Satyanarayana (1998) and Sharma et al. (2007) deal with the optimization of
physical parameters effecting xylanase production.
In addition, carbon and nitrogen sources and their concentration in the fer-
mentation media are found to be not only effecting the xylanase production but also
the localization of enzymes. The fungal xylanase producers are able to utilize wide
verity lignocellulosic-derived substrates such as corn cob, corn straw, wheat bran,
rice straw, and sugarcane bagasse. For production in SSF system, these substrates
are used as they are cheaper and easily available raw materials (Chauhan et al.
2007). However, in bacterial cells in SmF, simpler and readily utilizable nutrients
are used. Organic nitrogen sources such as tryptone, peptone, yeast extract, and
soybean meal are widely used as they enhance the xylanase production as well as
biomass. Kapoor et al. (2008) observed a significant increase in xylanase produc-
tion which was observed when the Bacillus sp. SSP-34 was provided with yeast
extract in combination with peptone. In similar study, xylanase production by
Bacillus sam-3 was increased up to 25-folds higher when the inorganic nitrogen
source was replaced by soybean meal. Use of corn steep powder also significantly
enhanced xylanase production by T. reesei (Lappalainen et al. 2000). Apart from
these major nutrients, the incorporation of micronutrients such as trace elements,
vitamins, and amino acids improved the xylanase production to great extend in
cases of thermo-anaerobe-mediated xylanase fermentation. In cases of recombinant
strains, the xylanase expression greatly depends on the gene inducers present in the
fermentation media. IPTG is the most widely used chemical inducer for ‘lac’-based
expression system (Mamo et al. 2007). However, in large-scale production, IPTG is
not applicable due to its high cost.
In cases of recombinant xylanase, the expression of xylanase genes usually
depends on several aspects such as biosynthesis mechanism, gene regulation, vector
184 H.A. El Enshasy et al.

construction, expression host used, expression media, and other growth parameters
during the fermentation. For commercial applications, xylanases have to be ideally
produced quickly and in large quantities. The conventional production strategies
have been followed are found to be less effective to meet the growing need in the
present xylanase market. Therefore, many researches are going on for developing
recombinant strains for xylanase production. However, a very limited number of
these recombinant strains are studied further for the scaling up of xylanase pro-
duction in pilot scale. Hence, more investigations on this area are required and it is
very significant to have more efficient and economically viable process to reduce
the cost of xylanase production.

7.10.2 Optimization of Endo-1,4-b-xylanase Production

Media screening and medium composition optimization are usually the preliminary
steps involved in bioprocess optimization which are always carried out in shake
flask level to select the most suitable medium formulation. Both physical and
chemical parameters effecting the cell growth as well as xylanases production are
monitored and optimized at this stage with a number of experiments. Basar et al.
(2010) observed that the recombinant xylanase production in E. coli is a
growth-associated process. They observed a rapid growth of E. coli, reaching the
stationary phase after 16 h and the maximal xylanase production up to 324.72
U mL−1 was obtained in defined medium. They also reported the effects of different
concentrations of glucose in growth media on cell growth and xylanase production.
Farliahati et al. (2009) reported the xylanase expression in recombinant E. coli
DH5a in different media compositions (defined, semi-defined, and complex).
Optimization study of pH and agitation ranges also carried out in shake flask
cultivation mode. From these studies, they observed the highest xylanase produc-
tion (up to 2122 U mL−1) in defined media at initial pH 7 and agitation speed of
200 rpm. A suitable cultivation strategy for the production of two truncated ther-
mostable recombinant xylanases (XynlAN and XynlANC), isolated from
Rhodothermus marinas, was developed by Karlsson et al. (1998). They found that
the fed-batch cultivations of E. coli strain BL21 (DE3) with a controlled expo-
nential glucose feed led to high specific production of the recombinant proteins and
addition of complex nutrients such as Tryptone Soya Broth (TSB) to the media
exhibits positive impact on cell growth rate and cell productivity. Single addition of
either lactose or isopropyl-thio-go-galactoside (IPTG) was used for induction. In
lactose-induced cells, the production of recombinant xylanase was delayed for
approximately 30 min compared to those induced with IPTG, but the specific
product levels were comparable at 3 h after induction. Huang et al. (2006) cloned a
xylanase gene from B. subtilis into E. coli and found that the xylanase distributions
in extracellular, intracellular, and periplasmic fractions were in ratio 22.4, 28.0, and
49.6 %, respectively, with an optimal enzyme production at pH 6.0 and 50 °C. The
7 Microbial Xylanases: Sources, Types, and Their Applications 185

superiority of defined medium for xylanase production using recombinant bacteria


was also reported by Farliahati et al. (2009).
Catabolic repression of the xylanases biosynthesis is found to be a common
phenomenon usually in the presence of glucose in culture medium. When glucose is
abundant in the grown medium, the repression of catabolic enzyme synthesis takes
place due to the gene regulation. When the microbe can easily absorb the readily
available glucose in the medium, they will not utilize for other complex nutrients by
spending energy to metabolize complex nutrients. Xylanase production reported to
be reduced due to the catabolic repression that makes the xylanase coding gene in
inactive mode. In another study by Basar et al. (2010) they enhanced xylanase
production using recombinant E. coli by optimizing the media composition and
other growth factors. Two basal mediums (defined media and complex media) have
been used and found out that the defined media gives the highest growth as well as
the xylanase production under experimental conditions. At 20 % enhanced dis-
solved oxygen tension (DOT) in lab scale bioreactor, they increased xylanase
production up to 1784 mL−1.

7.10.3 Application of Response Surface Methodology


(RSM) in Optimization of Xylanase Production

The conventional media optimization method based on step-by-step process vary-


ing one variable at a time (OFAT) while keeping the other variables constant is a
time-consuming process and less reliable in many cases. To overcome this problem,
statistical approach using experimental designs was applied. Response surface
methodology (RSM) is the most reliable statistical application that has been used
for optimization of fermentation media. Optimizations of physiological and nutri-
tional growth factors are found to be very important procedures to reduce the
production cost of xylanases in industrial scale. Many researches have been carried
out based on RSM which used to understand and evaluate the significant interaction
between the physiological and nutritional factors effecting the cell growth and
product formation. A prior knowledge about these growth factors and their inter-
actions is necessary for the designing of a more realistic model. Hence, many of the
RSM studies are found to be based on the results obtained from OFAT method.
Selection of these growth parameters has been done by checking their significance
level using a 2FFD Pareto chart before proposing an experimental design. Most
significant factors were then used as variables in central composite design
(CCD) (Farliahati et al. 2010). The effect of various growth parameters in pro-
duction of xylanase enzyme was studied by Mullai et al. (2010). In his study,
statistical experimental designs (CCD) were constructed with the help of statistical
application MINITAB 14. Plackett–Burman (PB) design for seven different vari-
ables, NH4NO3, KH2PO4, CaCl2, MgSO47H2O, FeSO47H2O, MnSO47H2O, and
NaCl at two levels +1 and −1 (higher and lower level, respectively) and the
186 H.A. El Enshasy et al.

statistical significance were studied by determining the F-value. The optimum


levels of selected variables from the Plackett–Burman design were investigated
using central composite design (CCD). The response was measured in terms of
xylanase activity. Counter plots and response surface 3D graphical representations
generated could show the interactions of different variables under experimental
conditions. Garai and Kumar (2012) optimized the xylanase production by A.
candidus in SSF. They studied the effect of various nitrogen sources in the pro-
duction media and they found that xylanase production was enhanced when the
ammonium nitrate was used as sole nitrogen source. In the same study, applying
RSM-based BoxBehnken design, the researcher also optimized the physical
parameters such as time of incubation, temperature, and moisture content which
effects xylanase activity. The crude xylanase recovered at the end of solid-state
fermentation was used for the scarification of ammonia-treated lignocellulose
materials to study the substrate-binding assay. Among the various substrates used,
corn comb was found releasing the maximum reduced sugar (438.47 mg g−1) when
it was treated with crude xylanase enzyme. In RSM-based optimization, xylanase
production was optimized by Box–Behnken design. Initially, Planket–Berman
design was employed to identify the key ingredients and process conditions which
exhibit the most significant effect on the xylanase expression. In the later stage,
using Box–Behnken design, the optimal level of the identified key ingredients in
production media as well as the optimum range of process parameters such as
cultivation time, pH, temperature, and agitation are identified. Using this approach
6.83-fold increase in xylanase production was obtained under optimized conditions
when compared to that of initial un-optimized conditions.
Solid-state fermentation (SSF) is also widely used for large-scale production of
xylanase enzymes. It is basically designed to mimic the natural habitat of microbes
that are less likely to grow in submerged fermentation (SMF) conditions. In SSF,
the solid bed is used as nutrient source as well as the mechanical support for cell
growth. A. niger, T. reesei, and T. koningii are the major fungal species used to
produce xylanase under SSF conditions (Dhiman et al. 2008; Garg et al. 1998).
However, xylanase production by bacteria is commonly carried out in submerged
fermentation conditions where the physical parameters such as temperature, pH,
and dissolved O2 level involved are controlled to the optimum levels with more
ease, and therefore an enhanced production can be achieved. Esteban et al. (1982)
reported that the large-scale production of xylanase in SmF is the growth-associated
process. However, several exceptional cases were also been reported such as
xylanase production using Bacillus sp. (Dey et al. 1992). Later study by Bocchini
et al. (2002) reported the production of highly thermostable cellulase-free xylanase
in SMF using B. circulans. Similar study was also carried out by Sharma et al.
(2007) who used Geobacillus thermoleovorans AP7 to produce highly
thermo-alkali-stable xylanase. The switch to xylanase-producing recombinant
E. coli is seen as an economic strategy toward higher productivity and easier
downstream purification. Sandhu and Kennedy (1984) reported a large-scale pro-
duction of xylanase by transforming E. coli with recombinant plasmids carrying a
1,4-b,D-xylanase gene from B. polymyxa. In another study, xylanase gene isolated
7 Microbial Xylanases: Sources, Types, and Their Applications 187

from A. usmani was cloned in pET-28a expression vector and when it is expressed
in E. coli BL21, the maximum xylanase activity was found to be 49.4 U mg−1. The
gene expression was induced by IPTG and the xylanase was found expressed with
6-X histidine tag that facilitates an easy purification of expressed protein (Zhou
et al. 2008b). A very successful cultivation strategy for the production of two
truncated thermostable recombinant xylanases (XynIAN and XynIANC), isolated
from Rhodothermus marinas, was developed by Karlsson et al. (1999). They found
that the fed-batch cultivations of E. coli strain BL21 (DE3) with a controlled
exponential glucose feed led to high specific production of the recombinant proteins
with additional complex medium. Highest xylanase production in defined medium
was achieved by Farliahati et al. (2009). In this study, xylanase production reached
up to 2122.5 U mL−1 at pH 7.4 and when (NH4)2HPO4 was used as the main
nitrogen source. They also reported that the xylanase production was a
growth-associated process as the enzyme secretion was greatly dependent on bio-
mass concentration for strain E. coli DH5a.
Biswas et al. (2010) used mutant IITD3A of Melanocarpus albomyces for the
production of high titer cellulose-free xylanase in 14-L stirred tank bioreactor where
an optimized (RSM based) production media and process parameters are used. The
authors reported the xylanase activity reached up to 415 U mL−1 after 24 h by pH
recycling strategy (between 7.8 and 8.2) during the production phase. They also
reported that the xylanase production is significantly influenced by aeration and
agitation. A 5.2-fold increase (overall volumetric productivity of 22,000 U L−1 h−1)
in activity was observed when the cells exhibited pellet form at an agitation speed
of 600 RPM.
Kumar et al. (2009) optimized the xylanase production in lab scale stirred tank
bioreactor using mycelial fungal strain T. lanuginosus MC 134. Fermentation media
contains corn cob as a sole source of carbon which was inoculated with T.
lanuginosus MC 134 and observed the production in terms of xylanase activity over
a period of 9 days at various process parameters. A highest xylanase activity, 2009
U mL−1, was obtained after 6 days of fermentation. However, a significant decrease
(40 %) in productivity was observed in bioreactors when compared to shake flask
culture. Similarly, xylanase production by several microbial strains using corn cob
was reported and from these studies it was clear that the production largely depends
on rheological factors in bioreactors and the type of extraction methods used.
Ruanglek et al. (2007) successfully cloned and expressed XylB (564 bp) encoding
endo-1,4-b-xylanase obtained from A. niger BCC14405 in Pichia pastoris under
AOX1 promoter. A maximum of 7352 U mg−1 of xylanase-specific activity was
obtained in 2 L lab scale bioreactor with BMGY/BSM medium under exponential
feeding strategy. During the batch phase which was aimed to achieve HCDC, the
temperature and pH were maintained at 30 °C and 5.0, respectively. Agitation was
maintained in the range of 600–1000 rpm. Fed-batch phase was started after 16–
20 h by feeding 67 % (v/v) glycerol plus 1 % (v/v) trace mineral solution in
exponential manner. Table 7.16 shows examples for xylanase production in lab and
pilot-scale submerged fermentation by different fungal strains.
188 H.A. El Enshasy et al.

Table 7.16 A comparison of xylanase production in lab and pilot-scale submerged fermentation
using fungal strains
Name of the Substrate Mode of Xylanase Productivity References
organism used operation activity (IU L−1 h−1)
(IU L−1)
Aspergillus niger Xylan (from Batch (20 L) 290,000 3820 Yuan et al.
corncob) (2005)
Aspergillus oryzae Spent sulfite Batch (15 L) 199,000 13,100 Chipeta
liquor et al.
(2008)
Thermomyces Oat husk Batch (2 L) 210,000 4380 Xiong
lanuginosus DSM hydrolysate et al.
10635 (2004)
Thermomyces Coarse Batch (5 L) 2,010,000 13,950 Kumar
lanuginosus MC corncob et al.
134 (2009)
Trichoderma Oat husk Fed-batch (5 1,350,000 14,060 Xiong
reesei hydrolysate L) et al.
(2005)
Melanocarpus Soluble Batch with 550,000 22,000 Biswas
albomyces wheat straw pH cycling et al.
extract (14 L) (2010)

7.10.4 pH Optima and Stability of Endo-1,4-b-xylanase


Enzymes

In general, endo-1,4-b-xylanases are active at wide pH ranges and their pH optima


depend on the producer organism and the physiochemical properties of the substrate
molecules. Fungal xylanases are usually acidophilic in nature. However, the opti-
mal pH for bacterial xylanases is usually above 5.5. Endo-1,4-b-xylanase from A.
kawachii reported as the most acidophilic xylanase which has a very low pH
optimum of 2. On the other hand, xylanase from Bacillus sp. 41 M1 reported as the
most alkalophilic xylanase which has pH optimum 9.0 (Nakamura et al. 1993). In
certain cases, different pH optimums were observed in different xylanases secreted
by same organism. For example, XynI and XynII produced by H. jecorina were
reported to have pH optimums 3–4 and 5–5.5, respectively. This showed that the pI
values of the enzymes are correlated with the amino acid residues at various
positions on xylanase. However, among Xyl-11 family, those are acidophilic in
nature possess an Asp residues at position 33 even though there are very rare
exceptions such as A. kawachii IFO 4308 xylanase which has pH optimum close to
5.0 (Zhu et al. 1994). However, many alkalophilic xylanases such as Xyn11 from A.
versicolor (Carmona et al. 1998), XynC from Uccinogenes sp. (Zhu et al. 1994)
also displayed an Asp at the same position. Similarly, studies on P. griseofulvum
Xyl-11 reported that it displays a Ser residue at position 44 and mutating this
7 Microbial Xylanases: Sources, Types, and Their Applications 189

residue resulted significant changes in pH optimum of the enzyme (Andre-Leroux


et al. 2008).
There have been many different explanations for the factors determining the
optimal pH value of xylanases. However, it believes that certain amino acid resi-
dues of xylanase structure play key role in the determination of the optimal pH
value of the enzyme. In case of acidic pH optima, the Asp/Glu residues near the
catalytic cleft become protonated and it decreases the electrostatic repulsion
between the substrate and enzyme, whereas in alkaline xylanases, instead of acidic
residues more alkaline residues such as arginine are seen. de Lemos Esteves et al.
(2005) experimented various mutations in Streptomyces sp. S38, and observed a pH
optimum shift from 6.0 to 7.5, which enhanced the xylanase enzyme activity
several folds higher than the enzyme from the wild type. The pI values of catalytic
domain present on xylanase play a key role in its pH activity profile. Many recent
studies reveals that, among GH11 xylanase, the residues those contribute positive
charges and hydrogen bonds to the catalytic domain help in increasing or
decreasing the pI values depending upon the electrostatic interaction between the
substrate molecules (Joshi et al. 1997). Apart from these residues, factors such as
occurrence of salt bridges are also reported to have significant effects on pH optima
(Hakulinen et al. 1998).

7.10.5 Endo-1,4-b-xylanase of Extremophilic Origin

Considerable progress has been made in the isolation of extremophilic microor-


ganisms and their successful cultivation in the laboratory. Commercial applications
of the xylanases demand highly thermostable enzyme which can work effectively
under elevated temperature. Many advantages such as reduced contamination risk
and faster reaction rates have been proposed for the use of thermophiles in
biotechnology processes. In general, parameters such as temperature, pH, and
chemical and enzymatic stability are important for the industrial applicability of any
enzyme. Recently, a simple model, based on the thermodynamic and kinetic
parameters for inactivation of the enzyme, has been presented for predicting
(1) thermostabilities in the presence of substrate and (2) residual activities of
enzymes in harsh processing environments. The study of extremophiles and their
enzymes can extend the present understanding of protein chemistry in addition to
expanding the potential applications of biocatalysts. Studies of alkaliphiles have led
to the discovery of a variety of enzymes which exhibit some unique properties.
Biological detergents contain enzymes such as alkaline proteases and/or alkaline
cellulases from alkaliphiles. Commercial production became economical only after
the discovery of alkaliphilic Bacillus-producing CGT with enhanced pH stability.
Alkaline xylanases have gained importance due to their application for the devel-
opment of eco-friendly technologies used in paper and pulp industries. The
enzymes are able to hydrolyze xylan which is soluble in alkaline solutions.
190 H.A. El Enshasy et al.

7.10.5.1 Alkaliphilic and Acidophilic Endo-1,4-b-xylanase

The pH tolerance of microbial endo-1,4-b-xylanase enzymes depends on the natural


growing environment of the microbial species. Endo-1,4-b-xylanase-producing
alkalophilic microbes grow optimally at higher pH above 9.0 and those produce
acidophilic xylanases grow between pH 1.0–5.0 (Kimura et al. 2000). These
organisms are usually isolated from various natural environments such as soil,
decomposing organic matters, paper pulp industry wastes, and many other sources.
Bacillus sp. C-59-2 was the first reported xylanase-producing alkaliphilic
microorganism (Nakamura et al. 1993). Since then, there have been a number of
reports on xylanases production using alkaliphilic and acidophilic microbes.
However, the studies indicated that the intracellular xylanase produced by many of
these microbes may not be adapted to extreme environmental growth conditions of
their hosts. A large number of alkaliphilic microbes found to produce xylanase at
optimal pH close to neutral although their activity can be retained in alkaline
conditions. Many exceptional cases are also reported such as XylB from Bacillus
sp. which has pH optima of 9.0–10 (Gessesse and Mamo 1998). Many acidophilic
xylanases of fungal and bacterial origin have also been reported in last two decades.
Cryptococcus sp. S-2 and Penicillium sp. 40 (Kimura et al. 2000) are the most
studied extremely acidophilic (pH optima 2–3) xylanase producers.

7.10.5.2 Psychrophilic Endo-1,4-b-xylanase

Even though the xylanase-producing microorganisms are abundant in nature, only a


very less number of endo-1,4-b-xylanase-producing psychrophilies have been
identified so far. Gram-negative bacteria such as Pseudoalteromonas haloplanktis
TAH3a (Collins et al. 2005a) and Flavobacterium frigidarium (Humphry et al.
2001); gram-positive Clostridium sp. PXYL1 (Akila and Chandra 2002); fungal
species such as Alternaria alternate, Penicillium sp., and Phoma sp.; and yeast sp
Cryptococcus adeliae (Petrescu et al. 2000) are the major psychrophilic organisms
reported to be endo-1,4-b-xylanase producers. Almost all these microbial species
are isolated from Antarctic regions. The best studied two psychrophilic xylanases,
which are from Pseudoalteromonas haloplanktis and Cryptococcus adeliae, were
classified under GH 8 and GH10, respectively. The (a/a) 6 barrel structure of these
structures is described by Van Petegem et al. (2003). The limited studies revealed
that these enzyme groups have very poor stability due to the reduced number of salt
bridges present on it but higher catalytic activity even at very low temperature.
Georlette et al. (2004) also observed that psychrophilic xylanases are characterized
by up to 10-folds higher activity even at mild cool temperature of 5 °C when
compared to mesophilic xylanase activity. Moreover, the psychrophilic xylanase
displayed higher catalytic and retained 60 % of maximum activity at very low
temperature. However, the psychrophilic xylanases are totally unstable at high
temperature (temperature 55 °C) and exhibited 12 times shorter half-life compared
to mesophilic xylanases (Collins et al. 2005b).
7 Microbial Xylanases: Sources, Types, and Their Applications 191

7.10.5.3 Thermophilic Endo-1,4-b-xylanase

A large number of thermophilic and hyper-thermophilic (optimal growth temper-


ature 50–80 °C and more than 80 °C, respectively) microbes which are able to
produce xylanases have been isolated from various sources such as thermal hot
springs, deep marine fields, self-heating organic debris, etc. Almost all of these
enzymes were grouped under families 10 and 11. Extremophile microbial species
such as Caldicellulosiruptor sp. (Luthi et al. 1990), Rhodothermus marinus (Abou
Hachem et al. 2000), and Bacillus stearothermophilus (Khasin et al. 1993) are the
major producers of thermophilic family 10 xylanases, whereas family 11 ther-
mophilic xylanases are less common and have been isolated from extremophiles
such as Bacillus strain D3 (Connerton et al. 1999), Chaetomium thermophilum
growing medicinal mushrooms in Egypt as a new strategy for cancer treatment,
dictyoglomus thermophilum (McCarthy et al. 2000), paecilomyces variotii (Kumar
et al. 2000), and thermomyces lanuginosus (Schlacher et al. 1996).
Optimum activity range of these thermophilic xylanases was found between 65
and 80 °C, whereas the xylanases from Thermotoga sp. exhibit optimal activity at
105 °C and pH 5.5. X-ray crystallographic structure and amino acid sequential
analysis of many of these thermophilic xylanase indicated very close similarities
with mesophilic xylanase and probable reason for the higher thermostability could
be the minor changes in amino acid alignment. Many mutagenic studies also have
indicated that minor modifications in mesophilic aminoacid array enhanced the
thermostability to many folds (Winterhalter and Liebl 1995).

7.10.5.4 Features Responsible for Endo-1,4-b-xylanase


Thermostability

Thermostability comparison made between the enzymes are usually misleading


since in most cases, the enzymes compared do not share the same experimental
conditions such as incubation time, concentration of protein, nature, and concen-
tration of the substrate. Many researchers believe that the high thermostability of
xylanases is due to the presence of a network of certain charged amino acid residues
near to the catalytic sites. These residues are usually found coupled with the
disulfide bridges between b-strand and the a-helix. However, many recombinant
thermostable xylanases are expressed in E. coli and are found to have thermosta-
bility ranging from 70 to 80 °C. When analyzing their protein structure, compar-
atively longer N-terminal ends are observed which may account for its higher
thermostability (Morris et al. 1996). Many other analyzes also propose that the
thermostability is due to the presence of highly flexible a-helix and C-terminal
regions where the protein unfolding is generally initiated (Kumar et al. 2000).
However, several other factors have also been identified that explain the xylanase
thermostability such as follows:
192 H.A. El Enshasy et al.

• Presence of disulfide bridges which may strengthen the whole protein frame-
work would give more stability to the enzyme.
• Presence of ion and aromatic pairs and their interactions may favor to more
stable framework.
• Strategic position of water molecules on the framework, since the readily
available water bindings are required for the catalytic activity even at higher
temperature.
• Complex folding nature of enzyme. The higher the folding complexity, the
higher the stability.
• Occurrence of oligomerization and the presence of certain aromatic residues
result in the hydrophobic patches which allows stronger monomeric interactions
enhancing the enzyme stability.
Certain GH11 endo-1,4-b-xylanases display disulfide bridges at position
between b-strand and a-helix. However, these enzymes do not exhibit higher
thermostability, whereas endo-1,4-b-xylanase produced by Dictyoglomus ther-
mophilum exhibits thermostability even though that do not have any disulfide
bridges on their structure. Therefore, the presence of disulfide bridges does not
necessarily account for the thermophilic characters of the enzymes (Table 7.17).

Table 7.17 Techniques used for purification of endo-1,4-b-xylanase enzymes


Source Purification Buffer solution References
Acrophialophora Gel filtration and ion 50 mM acetate Salles et al.
nainiana exchange (2000)
chromatographies
Bacteroides Metal affinity resin and 50 mM phosphate Mirande
xylanisolvens batch/gravity-flow et al. (2010)
XB1A column
Sulfolobus Ultrafiltration and 50 mM Tris–HCl Cannio et al.
solfataricus AKTA Fast Protein (2004)
Liquid Chromatography
Streptomyces Filtration and overnight 50 mM sodium citrate Nascimento
sp. AMT-3 dialysis et al. (2002)
Glaciecola Precipitation with 60 % 50 mM citrate Guo et al.
mesophila KMM saturation of ammonium (2009)
241 sulfate and His-Tag
Ni-affinity column
Bacillus sp. YJ6 Sephacryl S-100 h 20 mM citrate (pH 3.0– Yin et al.
chromatography 6.0), sodium phosphate (2010)
(pH 6.0–8.0), Tris–HCl
(pH 8.0–9.0), sodium
carbonate (pH 8.0–11.0)
Paenibacillus Precipitation with 20 % 50 mM phosphate Valenzuela
barcinonensis ammonium sulfate and et al. (2010)
cation exchange
chromatography
Aspergillus DEAE–Sephadex and McIlvaine (pH 4.0–8.0), Carmona
versicolor HPLC GF-510 gel Tris–HCl (pH 8.0–9.0), et al. (2005)
filtration glycine–NaOH (9.0–9.5)
7 Microbial Xylanases: Sources, Types, and Their Applications 193

7.10.5.5 Improvement of Endo-1,4-b-xylanase Thermostability

In last two decades, thermostability of xylanase enzymes was continuously


improved through the protein engineering approaches. In most of these studies, the
researches considered to modify the mesophilic xylanase enzyme structure to
design a new structural model similar to the thermophilic xylanase. Arase et al.
(1993) used chemical random mutagenesis in XYNA from B. pumilus and this
strategy was successful to increasing the enzymes’ half-life from 4 to 12 min at
57 °C. In a similar study, the specific activity and optimum temperature of Xyl1
from Streptomyces sp. S38 was enhanced from 2100 to 2500 IU mg−1 with the shift
of the optimal temperature from 57 to 66 °C (Georis et al. 2000). Other study
showed that the replacement of the N-terminal end of mesophilic xylanase xlnB
from Streptomyces lividans with those from thermostable xylanases TfxA from T.
fusca by random gene shuffling and observed a significant increase in thermosta-
bility (Shibuya et al. 2000). Introduction of two disulfide bridges, one between B9
strand and the a-helix and another between N and C-terminal ends, also enhanced
the thermostability of Tx-xyl (Paës and O’Donohue 2006). The authors also
reported that the occurrence of residues that met at N-terminal end brought negative
impact on xylanase thermostability and temperature optimum which are reduced by
10-folds and 5 °C, respectively. From these engineering strategies applied in
xylanases protein engineering, researchers concluded that:
• The a-helix on xylanase structure seems very sensitive to thermal unfolding—
making these are more rigid and may enhance the thermostability of the enzyme.
However, it may also diminish the catalytic activity (Muilu et al. 1998).
• N-terminal end plays key role in thermostabilization in GH11 xylanase—linking
the N-terminal extremity to A2 strand or C-terminal extremity enhanced the
thermostability of xylanase. Introduction of an additional complete b strand by
mutagenesis also beneficial for improved thermostability.
• Presence of disulfide bridges does not necessarily enhance the thermophilic
behavior of the enzymes. Therefore, addition of these bridges does not always
bring positive impact on enzyme thermostability.

7.11 Industrial Applications of Endo-1,4-b-xylanase

Industrial production of xylanases has been grown significantly during last few
decades based on their increased application. Xylan can be converted into many
useful products such as xylitol and furfurol through enzymatic hydrolysis
(Subramaniyan and Prema 2002). The hydrolysis could be also carried out through
acid treatment process which is relatively easy, fast, and cheap. However, the
application of this method in industries is limited due to the formation of toxic end
194 H.A. El Enshasy et al.

substances that may have cross reactions with product of interest (Beg et al. 2001).
Therefore, enzymatic hydrolysis method is widely applied even though it is a slow
process but more efficient and eco-friendly. During industrial application, the
xylolytic enzymes are not commonly applied in pure form but in combination with
other hydrolytic enzymes to achieve better results. The industrial applications of
xylanases cover different areas and the list of applications increased by time and
thus increases the market demand of xylanases accordingly.

7.11.1 Animal Feed Industry

Xylanases are widely used in animal feed manufacturing. They are usually applied
in combination of other hydrolytic enzymes such as pectinase, protease, cellulose,
lipase, glucanase, alginase, and phytase. These enzymes are used to reduce the
viscosity of raw materials used in animal feeds. The complex arabinoxylan that is
present on the cell wall of most grains may reduce the effective digestion and
absorption of nutrients in poultry. Studies on this area have reported that the
xylanase-treated feed has better digestibility and nutrient absorption rate when
compared to raw feeds (Bedford and Classen 1992). In animal feed industries, the
feed conversion rate (FCR) is defined as the ratio of feed consumed to the body
weight gained by the animal. A lower FCR means improved feed digestibility and
nutrient absorption. Xylanase enzymes are widely incorporated in poultry feed to
reduce the FCR. It have been reported that the addition of endo-xylanase is isolated
from Acidothermus cellulolyticus in poultry feed and observed a significant
decrease in FCR, indicating an improved digestibility of feed (Clarkson et al. 2001).
Similar results were observed when endo-xylanase is isolated from Neocallimastix
patriciarum added to canola seed after oil extraction and applied for cattle feeding.
The digestibility and stability of nutritive value of silage for cattle are found to be
enhanced when it was treated with xylanase along with probiotic lactobacillus. The
xylose formed by the de-polymerization of hemicellulose was fermented by the
probiotic lactobacillus and thus enhanced the feed nutritive value. It was also
reported that xylanase supplementation in the animal diet improved the animal
growth through the better nutrition absorbability and enhancement of immune
response as well as the gut micro-flora (Gao et al. 2008). In other study, Vandeplas
et al. (2010) reported that when T. viride xylanase is incorporated in poultry feed, it
increased nutrient digestion by 60 % and resulted in a significant increase in
chicken weight. Pretreatment of forage crops with xylanase leads to better digestion
in cattle and these enzymes are often found in synergy with other cellulolytic
enzymes. The assimilation of ruminant feed was also improved many folds by
xylanases addition (Yang and Xie 2010).
7 Microbial Xylanases: Sources, Types, and Their Applications 195

7.11.2 Food and Beverages Industry

Food and beverages industries use also xylanases in wide range of applications.
Xylanases are capable of hydrolyzing the hemicellulose in wheat that enhances the
water absorption of dough at leavening level in bread making process. This could
possibly enhance the fermentation rate and as a result the softness of the bread and
volume was increased (Rouau 1993). Xylanase cocktails are often used to reduce
the stickiness of the dough thus enhance the crumb (Goesaert et al. 2005; Butt et al.
2008). Other study showed also that under specific process condition, incorporation
of xylanase in the dough increased the shelf life of bread and also improved the
volume up to 24 % (Verjans et al. 2010). However, enzyme selection is critical in
these conditions since the cereals used for bread making often exhibit the xylanase
inhibition by TAXI, XIP, or TLXI (Gebruers et al. 2004). Pasta processing is
another area where xylanases are widely used as solubilizing agent of durum wheat
semolina without changing its rheological properties (Ingelbrecht et al. 2001; Brijs
et al. 2004). It was also reported that xylanase enzymes can improve the separation
of wheat flour into starch and gluten (Frederix et al. 2004). However, in certain
cases, action of Xyl-11 does not favor the separation due to high viscosity caused
by the un-extractable arabino xylanase hydrolysis. In such conditions, synergetic
dosages of Xyl-10 and Xyl-11 are used as it can mediate-improved agglomeration
of gluten protein even though the wheat quality is very poor (Van Der Borght et al.
2005). Table 7.18 shows some examples of commonly used xylanase preparations
for the baking industry. Xylanases are widely used also in juice, beer, and wine
making industries. The enzymatic breakdown of fruits and vegetables hemicellulose
can increase the clarity and stability of juice and wine. Xylanases can be used along
with several other enzymes such as pectinase and amylase to improve the quality of
beverage products in terms of aroma, taste, clarity, and stability (Wong et al. 1988).
It was reported that up to 27 % decrease in insoluble fibers was achieved in juice
clarification by addition of xylanase enzyme isolated from Sclerotinia sclerotiorum
(Olfa et al. 2007). In beer production, partial hydrolysis of arabinoxylan found in
the barley cell wall gives a middy viscous appearance to the fermented beer. In such

Table 7.18 Examples of commonly used xylanase preparations for the baking industry (Collins
et al. 2005a, b)
Supplier Brand Enzyme source
Beldem-Puratos Bel’ase B210 Bacillus subtilis
Bel’ase F25 Aspergillus niger
Genencor Intl. Multifectw Trichoderma reesei
Danisco Grindamyle H Aspergillus niger
DSM Bakezymew HS, BXP Aspergillus niger
Novozymes Pentopan Mono Thermomyces lanuginosus
AB enzymes Veronw 191 Aspergillus niger
Veronw Special Bacillus subtilis
196 H.A. El Enshasy et al.

cases, various xylanase enzyme groups are used to increase the filtration perfor-
mance of beer wort by hydrolysis of viscous AXs fractions. Xylanase inhibitors
present in the malt are the major hinderance to xylanase action. The xylanase
activity is retained with the help of other synergetic enzymes used in the process
(Debyser et al. 1997).

7.11.3 Paper and Pulp Industries

Endo-1,4-b-xylanase enzymes play significant role in paper and pulp industry since
they facilitate an eco-friendly bio-bleaching of wood pulp by reducing the use of
chemical bleaching agents such as chlorine and chlorine dioxide. For the past few
decades, kraft pulp bleaching industry is recognized as the biggest area of xylanase
application. Several studies reported that the removal of lignin content can be
achieved by xylanase enzymes far better than by other lignin-degrading enzymes
(Lundgren et al. 1994). Use of chemical bleaching agents in conventional pulp
bleaching methods cause serious environmental pollution issues and this drove the
researcher’s attention on xylanase enzymes (Subramaniyam and Prema 2002).
Table 7.19 shows major endo-1,4-b-xylanase supplier in pulp bleaching industry.
Application of endo-1,4-b-xylanase enzymes in pulp bleaching can reduce using
chlorine as well as the total energy requirement for this process. Kulkarni et al.
(1999) observed that xylanase pretreatment of kraft pulp lowers the usage of
chemicals by 10–20 %. The concept of TCF (total chlorine free) bleaching tech-
nologies was also recently introduced to make the process more eco-friendly.
However, cellulase-free endo-1,4-b-xylanase is required in this process since the
cellulose enzyme may degrade the cellulose which is the main target of the industry

Table 7.19 Major xylanase supplier in pulp bleaching industry (Beg et al. 2001)
Supplier Product trade name
Alko Rajamaki, Finland Ecopulp
Sandoz, Charlotte, N.C. and Basle, Switzerland Cartazyme
Clarient, UK Cartazyme HS, HT and SR 10
Genercor, Finland; Ciba Giegy, Switzerland Irgazyme 40–4X/Albazyme 40–4X
Novo Nordisk, Denmark Pulpzyme HA, HB and HC
Biocon India, Bangalore Bleachzyme F
Rohn Enzyme 0Y; Primalco, Finland Ecopulp X-100, X-200, and X-200/4
Solvay Interox, USA Optipulp L-8000
Thomas Swan, UK Ecozyme
Iogen, Canada GS-35, HS70
7 Microbial Xylanases: Sources, Types, and Their Applications 197

(Bajpai and Bajpai 2001). Relatively lower molecular weight of xylanase enzymes
helps them to penetrate more deep into the lignocelluloses substrates and facilitate
better hydrolytic actions (Beaugrand et al. 2005; Beg et al. 2001). Higher ther-
mostability and alkaline stability are other key features that make xylanase more
favorable for bio-bleaching industry (Buchert et al. 1995). The mechanisms of pulp
bleaching by xylanase have been studied and many researchers believe that xyla-
nase hydrolysis takes place at lignin xylan interaction and thus results in a loosely
packed cellulosic framework (Shatalov and Pereira 2007). However, it was recently
reported the potential use of Aspergillus oryzae MDU-4-based fungal xylanase for
effective deinking of newspaper pulp where a significant improvement in optical
properties such as brightness up to 57.9 % ISO with decreased (211 ppm) residual
ink concentration (Chutani and Sharma 2015).

7.11.4 Biofuel Production

The next-generation fuel production is based on renewable sources such as ligno-


cellulosic biomass. As first step, the complex lignocellulosic biomass needs to be
breakdown to a mixture of relatively smaller units made of hexoses and pentoses or
their monosaccharides that are further fermented to ethanol or other biofuels
(Margeot et al. 2009). The process of converting the lignocellulosic feedstocks into
biofuels usually involves the following steps:
• Enzyme hydrolysis: the critical step involved where the hydrolysis of ligno-
celluloses into various sugar monomers by the action of cocktail of hydrolytic
enzymes.
• Fermentation: production of biofuel by the utilization of sugar monomers
formed by various microbial species, usually carried out by different bacterial
and yeast strains depends on the desired type of biofuel.
• Distillation: separation and purification of biofuel produced.
The controlled hydrolysis of lignocelluloses using enzymatic cocktail to produce
the desirable products is the challenge involved in this process. Xylolytic enzymes,
such as Endo-1,4-b-xylanase in particular, are very essential in biomass hydrolysis
as they initiate progressive cell wall in the assembly and thus mediate the cell wall
penetration by other cellulolytic enzymes (Beaugrand et al. 2005).

7.11.5 Other Applications

Apart from the traditional applications in pulp beaching industry, xylanases have
been used in several other areas as well. The broad spectrum of xylanase application
includes the saccharification of xylan to xylose, plant cell protoplasting, coffee
198 H.A. El Enshasy et al.

mucilage liquefaction, vegetable softening, extraction of plant pigments and oils,


etc. (Kulkarni et al. 1999). In laundry industries, endo-1,4-b-xylanase was effec-
tively used to remove the plant-based stains. The stain-removing capacity of family
11 endo-xylanase was improved when it chemically linked with cellulose-binding
domain of cellulases. In textile industries, xylanase enzymes can be used to process
the plant fiber as they can decrease the lignin content and improve the color and
quality of fibers (Csiszár et al. 2001; Battan et al. 2012). Until now, xylanase
applications in pharmaceutical industries are very limited. However, it is used in
some dietary supplements in combination with other enzymes (Polizeli et al. 2005).
Commercial products such as chewing gum contain xylitol and it can be easily
produced by hydrolysis of xylan using xylanase enzymes.

7.11.6 Future Perspectives

The current list of highly diversified applications of xylanases clearly demonstrates


the increased demand of this type of enzymes. In addition, the continuous
improvement of the enzyme catalytic properties increases the potential uses of this
enzyme by adding many new industrial applications. The improvement of catalytic
properties of the xylanases by shifting the optimal operation conditions of the
enzyme and by isolation of new potential xylanases from extremophilic microbes
are further expressed in suitable mesophilic host to industrialize the process. In
addition, with the increased knowledge about protein engineering, it become pos-
sible also to increase enzyme stability and kinetic properties using different
approaches such as changing of amino acid sequence, creation of sulfide bridge
within the molecule, and changing the enzyme 3D configuration. All these will help
to increase the potential application of xylanases under extreme operation
conditions.
However, some recent studies were skeptic about the future application of
xylanases in biofuel sector based on the current dramatic drop of oil price which
make biofuel non-competitive alternative to fossil fuel in economic point of view.
However, xylan biorefinery is not limited to fuel market but the biohydrolysis
products of xylan such as pentose oligomers and monomers are also used as
intermediate in the production of many biochemicals. Moreover, the increased
public awareness about the negative impacts of chemicals on the environment,
ecosystem, and human and animal health increased the pressure on chemical
industries to replace the current chemical-based catalytic processes to a biobased
and eco-friendly processes especially in paper/pulp, leather, and detergent indus-
tries. In addition, there is significant increased demand for xylanases application in
food and feed industries for the production of safe and high-quality product. These
all together support our current estimation of the continuous increase of xylanases
demand in many industrial applications.
7 Microbial Xylanases: Sources, Types, and Their Applications 199

References

Abou Hachem M, Karlsson EN, Bartonek-Roxa E, Raghothama S, Simpson PJ, Gilbert HJ,
Williamson MP, Holst O (2000) Carbohydrate-binding modules from a thermostable
Rhodothermus marinus xylanase: cloning, expression and binding studies. Biochem J
345:53–60
Alejandro SH, Jesús VE, del María CMH, María EHL (2007) Purifcation and characterization of
two sugarcane bagasse-absorbable thermophilic xylanases from the mesophilic Cellulomonas
Xavigena. Microbiol Biotechnol 34:331–338
Akila G, Chandra TS (2002) A novel cold-tolerant Clostridium strain PXYL1 isolated from a
psychrophilic cattle manure digester that secretes thermolabile xylanase and cellulase. FEMS
Microbiol Lett 219:63–67
Andrade SV, Polizeli MLTM, Terenzi HF, Jorge JA (2004) Effect of carbon source on the
biochemical properties of b-xylosidases produced by Aspergillus versicolor. Process Biochem
39:931–938
Andre-Leroux G, Berrin JG, Georis J, Arnaut V, Juge V (2008) Structure-based mutagenesis of
Penicillium griseofulvum xylanase using computational design. Proteins 72(4):1298–1307
Andrews SR, Charnock SJ, Lakey JH, Davies GJ, Claeyssens M, Nerinckx V, Underwood M,
Sinnott ML, Warren RA, Gilbert HJ (2000) Substrate specificity in glycoside hydrolase family
10. Tyrosine 87 and leucine 314 play a pivotal role in discriminating between glucose and
xylose binding in the proximal active site of Pseudomonas cellulosa xylanase 10A. J Biol
Chem 275(30):23027–23033
Anuradha P, Vijayalakshmi K, Prasanna ND, Sridevi K (2007) Production and properties of
alkaline xylanases from Bacillus sp. isolated from sugarcane fields. Curr Sci 92(9):1283–1286
Arase A, Yomo T, Urabe I, Hata Y, Katsube Y, Okada H (1993) Stabilization of xylanase by
random mutagenesis. FEBS Lett 316(2):123–127
Archana A, Satyanarayana T (1998) Cellulase-free xylanase production by thermophilic Bacillus
licheniformis A99. Indian J Microbiol 38:135–139
Atkins EDT (1992) Three-dimensional structure, interactions and properties of xylans. Xylanas
Xylanases 7:39–50
Avalos OP, Noyola TP, Plaza IM, Torre M (1996) Induction of xylanase and b-xylosidase in
Cellulomonas flavigena growing on different carbon sources. Appl Microbiol Biotchnol
46:405–409
Bai Y, Wang J, Zhang Z, Yang P, Shi P, Luo H, Meng K, Huang H, Yao B (2010) A new xylanase
from thermoacidophilic Alicyclobacillus sp. A4 with broad-range pH activity and pH stability.
J Ind Microbiol Biotechnol 37(2):187–194
Bajpai P, Bajpai PK (2001) Development of a process for the production of dissolving kraft pulp
using xylanase enzyme. Appita J 54(4):381–384
Bajaj BK, Singh NP (2010) Production of xylanase from an alkali tolerant Streptomyces sp. 7b
under solid-state fermentation, its purification, and characterization. Appl Biochem Biotechnol
162(6):1804–1818
Balakrishnan H, Srinivasan MC, Rele MV, Chaudhari K, Chandwadkar AJ (2000) Effect of
synthetic zeolites on xylanase production from an alkalophilic Bacillus sp. Curr Sci 79:95–97
Basar B, Shamzi MM, Rosfarizan M, Puspaningsih NNT, Ariff AB (2010) Enhanced production
of thermophilic xylanase by recombinant Escherichia coli DH5a through optimization of
medium and dissolved oxygen level. Int J Agric Biotechnol 12:321–328
Basaran P, Hang YD, Basaran N, Worobo RW (2001) Cloning and heterologous expression of
xylanase from Pichia stipitis in Escherichia coli. J Appl Microbiol 90(2):248–255
Battan B, Dhiman SS, Ahlawat S, Mahajan R, Sharma J (2012) Application of thermostable
xylanase of Bacillus pumilus in textile processing. Indian J Microbiol 52(2):222–229
Beaugrand J, Paës G, Reis D, Takahashi M, Debeire P, O’Donohue MJ, Chabbert B (2005)
Probing the cell wall heterogeneity of micro-dissected wheat caryopsis using both active and
inactive forms of a GH11 xylanase. Planta 222:246–257
200 H.A. El Enshasy et al.

Bedford MR, Classen HL (1992) Reduction of intestinal viscosity through manipulation of dietary
rye and pentosanase concentration is effected through changes in the carbohydrate composition
of the intestinal aqueous phase and results in improved growth rate and food conversion
efficiency of broiler chicks. J Nutr 122:560–569
Beg QK, Bhushan B, Kappor M, Hoondal G (2000) Production and characterization of
thermostable xylanase and pectinase from Streptomyces sp. QG-11-3. J Ind Microbiol
Biotechnol 24:396–402
Beg QK, Kapoor M, Mahajan L, Hoondal GS (2001) Microbial xylanases and their industrial
applications: a review. Appl Microbiol Biotechnol 56(3–4):326–338
Bergquist P, Te’o V, Gibbs M, Cziferszky A, de Faria FP, Azevedo M, Nevalainen H (2002)
Expression of xylanase enzymes from thermophilic microorganisms in fungal hosts.
Extremophiles 6(3):177–184
Berrin JG, Juge N (2008) Factors affecting xylanase functionality in the degradation of
arabinoxylans. Biotechnol Lett 30(7):1139–1150
Berrin JG, Williamson G, Puigserver A, Chaix JC, McLauchlan WR, Juge N (2000) High-level
production of recombinant fungal endo-beta-1,4-xylanase in the methylotrophic yeast Pichia
pastoris. Protein Expr Purif 19(1):179–187
Bhalerao J, Patki AH, Bhave V, Khurana I, Deobagkar DN (1990) Molecular-cloning and
expression of a xylanase gene from Cellulomonas sp. into Escherichia coli. Appl Microbiol
Biotechnol 34(1):71–76
Bhardwaj A, Leelavathi S, Mazumdar-Leighton S, Ghosh A, Ramakumar S, Reddy VS (2010) The
critical role of N- and C-terminal contact in protein stability and folding of a family 10
xylanase under extreme conditions. PLoS ONE 5(6):e11347
Biely P, Vrsanska M, Kluepfel M, Tenkanen D (1997) Endo-beta-1,4-xylanase families:
differences in catalytic properties. J Biotechnol 57(1–3):151–166
Biely P, Leathers TD, Cziszarova M, Vrsanska M, Cotta MA (2008) Endo-beta-1,4-xylanase
inhibitors in leaves and roots of germinated maize. J Cereal Sci 48(1):27–32
Biswas R, Sahai V, Mishra S, Bisaria VS (2010) Bioprocess strategies for enhanced production of
xylanase by Melanocarpus albomyces IITD3A on agro-residual extract. J Biosci Bioeng
110:702–708
Blum DL, Li XL, Chen H, Ljungdahl LG (1999) Characterization of an acetyl xylan esterase from
the anaerobic fungus Orpinomyces sp. strain PC-2. Appl Environ Microbiol 65(9):3990–3995
Bocchini DA, Alves-Prado HF, Baida LC, Roberto IC, Gomes E, Da-Silva R (2002) Optimization
of xylanase production by Bacillus circulans D1 in submerged fermentation using response
surface methodology. Process Biochem 38:727–731
Bolam DN, Xie HF, White P, Simpson PJ, Hancock SM, Williamson MP, Gilbert HJ (2001)
Evidence for synergy between family 2b carbohydrate binding modules in Cellulomonas fimi
xylanase 11A. Biochemistry 40(8):2468–2477
Boraston AB, Tomme P, Amandoron EA, Kilburn DG (2000) A novel mechanism of xylan
binding by a lectin-like module from Streptomyces lividans xylanase 10A. Biochem J 350(Pt
3):933–941
Boraston AB, McLean BW, Chen V, Li A, Warren RA, Kilburn DG (2002) Co-operative binding
of triplicate carbohydrate-binding modules from a thermophilic xylanase. Mol Microbiol 43
(1):187–194
Borralho T, Chang Y, Jain P, Lalani M, Parghi K (2002) Lactose induction of the lac operon in
Escherichia coli B23 and its effect on the o-nitrophenyl b-galactoside assay. J Exp Microbiol
Immunol 2:117–123
Bridgeman TG, Jones JM, Shield I, Williams PT (2008) Torrefaction of reed canary grass, wheat
straw and willow to enhance solid fuel qualities and combustion properties. Fuel 87
(6):844–856
Brijs K, Ingelbrecht JA, Courtin CM, Schlichting L, Marchylo BA, Delcour JA (2004) Combined
effects of endoxylanases and reduced water levels in pasta production. Cereal Chem
81:361–368
7 Microbial Xylanases: Sources, Types, and Their Applications 201

Brutus A, Villard C, Durand A, Tahir T, Furniss C, Puigserver A, Juge N, Giardina T (2004) The
inhibition specificity of recombinant Penicillium funiculosum xylanase B towards wheat
proteinaceous inhibitors. Biochim Biophys Acta 1701(1–2):121–128
Brutus A, Reca IB, Herga S, Mattei B, Puigserver A, Chaix JC, Juge V, Bellincampi D, Giardina T
(2005) A family 11 xylanase from the pathogen Botrytis cinerea is inhibited by plant
endoxylanase inhibitors XIP-I and TAXI-I. Biochem Biophys Res Commun 337(1):160–166
Buchert J, Teleman A, Harjunpa V, Tenkanen M, Viikari L, Vuorinen T (1995) Effect of cooking
and bleaching on the structure of xylan in conventional pine kraft pulp. Tappi J 78
(11):125–130
Butt MS, Nadeem MT, Ahmad Z, Sultan MT (2008) Xylanases and their applications in baking
industry. Food Technol Biotechnol 46(1):22–31
Cannio R, Di Prizito N, Rossi M, Morana A (2004) A xylan-degrading strain of Sulfolobus
solfataricus: isolation and characterization of the xylanase activity. Extremophiles 8
(2):117–124
Cantarel BL, Coutinho PM, Rancurel C, Bernard T, Lombard V, Henrissat B (2009) The
carbohydrate-active enzymes database (CAZy): an expert resource for glycogenomics. Nucleic
Acids Res 37:D233–D238
Cao Y, Qiao J, Li Y, Lu W (2007) De novo synthesis, constitutive expression of Aspergillus
sulphureus beta-xylanase gene in Pichia pastoris and partial enzymic characterization. Appl
Microbiol Biotechnol 76(3):579–585
Carmona EC, Brochetto-braga MR, Pizzirani-kleiner AA, Jorge JA (1998) Purification and
biochemical characterization of an endoxylanase from Aspergillus versicolor. FEMS Microbiol
Lett 166:311–315
Carmona EC, Fialho MB, Buchgnani EB, Coelho GD, Brocheto-Braga MR, Jorge JA (2005)
Production, purification and characterization of a minor form of xylanase from Aspergillus
versicolor. Process Biochem 40(1):359–364
Cereghino JL, Cregg JM (2000) Heterologous protein expression in the methylotrophic yeast
Pichia pastoris. FEMS Microbiol Rev 24(1):45–66
Cervera-Tison MC, Andre-Leroux G, Lafond M, Georis J, Juge N, Berrin J-G (2009) Molecular
determinants of substrate and inhibitor specificities of the Penicillium griseofulvum family 11
xylanases. Biochim Biophys Acta 1794:438–445
Charnock SJ, Lakey JH, Virden R, Hughes N, Sinnott ML, Hazlewood GP, Pickersgill R,
Gilbert HJ (1997) Key residues in subsite F play a critical role in the activity of Pseudomonas
fluorescens subspecies cellulosa xylanase A against xylooligosaccharides but not against
highly polymeric substrates such as xylan. J Biol Chem 272(5):2942–2951
Chauhan K, Trivedi U, Patel KC (2007) Statistical screening of medium components by Plackett–
Burman design for lactic acid production by Lactobacillus sp. KCP01 using date juice.
Bioresour Technol 98:98–103
Chauthaiwale VM, Deshpande VV (1992) Molecular cloning and expression of the xylanase gene
from Chainia in Escherichia coli. FEMS Microbiol Lett 99(2–3):265–270
Chavez R, Schachter K, Navarro C, Peirano A, Bull P, Eyzaguirre J (2004) The acetyl xylan
esterase II gene from Penicillium purpurogenum is differentially expressed in several carbon
sources, and tightly regulated by pH. Biol Res 37(1):107–113
Chutani P and Sharma KK (2015) Biochemical evaluation of xylanases from various filamentous
fungi and their application for the deinking of ozone treated newspaper pulp. Carbohydrate
Polymers 127:54–63
Cheng YF, Yang CH, Liu WH (2005) Cloning and expression of Thermobifida xylanase gene in
the methylotrophic yeast Pichia pastoris. Enz Microb Technol 37(5):541–546
Chiku K, Uzawa J, Seki H, Amachi S, Fujii T, Shinoyama H (2008) Characterization of a novel
polyphenol-specific oligoxyloside transfer reaction by a family 11 xylanase from Bacillus
sp. KT12. Biosci Biotechnol Biochem 72(9):2285–2293
Chipeta ZA, Du-Preez JC, Christopher L (2008) Effect of cultivation pH and agitation rate on
growth and xylanase production by Aspergillus oryzae in spent sulphite liquor. J Ind Microbiol
Biotechnol 35:587–594
202 H.A. El Enshasy et al.

Chithra M, Muralikrishna G (2008) Characterization of purified xylanase from finger millet


(Eleusine coracana-Indaf 15) malt. Eur Food Res Technol 227(2):587–597
Claassen PAM, van Lier JB, Lopez Contreras AM, van Niel EJW, Sijtsma L, Stams AJM, de
Vries SS, Weusthuis RA (1999) Utilisation of biomass for the supply of energy carriers. Appl
Microbiol Biotechnol 52(1999):741–755
Clarkson K, Siika-aho M, Tenkanen M, Bower BS, Penttilä ME, Saloheimo MLA (2001)
Trichoderma reesei xylanase. Patent WO 0149859
Cleemput G, Hessing M, van Oort M, Deconynck M, Delcour JA (1997) Purification and
characterization of a [beta]-D-xylosidase and an endo-xylanase from wheat flour. Plant Physiol
113(2):377–386
Collins T, Gerday C, Feller G (2005a) Xylanases, xylanases families and extremophilic xylanases.
FEMS Microbiol Rev 29:3–23
Collins T, De Vos D, Hoyoux A, Savvides SN, Gerday C, Van Beeumen J, Feller G (2005b) Study
of the active site residues of a glycoside hydrolase family 8 xylanase. J Mol Biol 354
(2):425–435
Connerton I, Cummings N, Harris GW, Debeire P, Breton C (1999) A single domain thermophilic
xylanase can bind insoluble xylan: evidence for surface aromatic clusters. Biochim Biophys
Acta 1433:110–121
Copa-Patino JL, Broda YG, Broda P (1993) Production and initial characterization of the
xylan-degrading system of Phanerochaete chrysosporium. Appl Microbiol Biotechnol
40:69–76
Coughlan MP, Hazlewood GP (1993) Beta-1,4-D-xylan-degrading enzyme-systems-biochemistry,
molecular-biology and applications. Biotechnol Appl Biochem 4:259–289
Couturier M, Haon M, Coutinho PM, Henrissat B, Lesage-Meessen L, Berrin JG (2010)
Podospora anserina hemicellulases potentiate the Trichoderma reesei secretome for saccha-
rification of lignocellulosic biomass. Appl Environ Microbiol 77:237–246
Crepin VF, Faulds CB, Connerton IF (2004) Functional classification of the microbial feruloyl
esterases. Appl Microbiol Biotechnol 63:647–652
Croes E, Gebruers K, Carpentier S, Swennen R, Robben J, Laukens K, Witters E, Delcour JA,
Courtin CM (2009) A quantitative portrait of three xylanase inhibiting protein families in
different wheat cultivars using 2D-DIGE and multivariate statistical tools. J Proteomics 72
(3):484–500
Csiszár E, Urbánszki K, Szakás G (2001) Biotreatment of desized cotton fabric by commercial
cellulase and xylanase enzymes. J Mol Catal B Enzym 11:1065–1072
Damiano VB, Bocchini DA, Gomes E, Silva RD (2003) Application of crude xylanase from
Bacillus licheniformis 77-2 to the bleaching of Eucalyptus kraft pulp. World J Microbiol
Biotechnol 19:139–144
Dautova M, Roze E, Overmars H, de Graaff L, Schots A, Helder J, Goverse A, Bakker J, Smant G
(2006) A symbiont-independent endo-1,4-beta-xylanase from the plant-parasitic nematode
Meloidogyne incognita. Mol Plant Microbe Interact 19(5):521–529
De Lemos Esteves F, Gouders T, Lamotte-Brasseur J, Rigali S, Frere JM (2005) Improving the
alkalophilic performances of the Xyl1 xylanase from Streptomyces sp. S38: structural
comparison and mutational analysis. Protein Sci 14(2):292–302
De Vos D, Collins T, Nerinckx W, Savvides SN, Claeyssens M, Gerday C, Feller G, Van
Beeumen J (2006) Oligosaccharide binding in family 8 glycosidases: crystal structures of
active-site mutants of the beta-1,4-xylanase pXyl from Pseudoaltermonas haloplanktis TAH3a
in complex with substrate and product. Biochemistry 45(15):4797–4807
De Vries RP, Visser J, de Graaff LH (1999) CreA modulates the XlnR-induced expression on
xylose of Aspergillus niger genes involved in xylan degradation. Res Microbiol 150
(4):281–285
De Vries RP, Kester HC, Poulsen CH, Benen JA, Visser J (2000) Synergy between enzymes from
Aspergillus involved in the degradation of plant cell wall polysaccharides. Carbohydr Res 327
(4):401–410
7 Microbial Xylanases: Sources, Types, and Their Applications 203

Dean JFD, Gross KC, Anderson JD (1991) Ethylene biosynthesis-inducing xylanase. 3. Product
characterization. Plant Physiol 96(2):571–576
Debyser W, Peumans WJ, Van Damme EJM, Delcour JA (1999) Triticum aestivum xylanase
inhibitor (TAXI), a new class of enzyme inhibitor affecting breadmaking performance. J Cereal
Sci 30(1):39–43
Debyser W, Derdelinckx G, Delcour JA (1997) Arabinoxylan solubilization and inhibition of the
barley malt xylanolytic system by wheat during mashing with wheat whole meal adjunct:
evidence for a new class of enzyme inhibitors in wheat. Am Soc Brew Chem 55:153–156
Degefu Y, Lohtander K, Paulin L (2004) Expression patterns and phylogenetic analysis of two
xylanase genes (htxyl1 and htxyl2) from Helminthosporium turcicum, the cause of northern
leaf blight of maize. Biochimie 86(2):83–90
Dey D, Hinge J, Shendye A, Rao M (1992) Purification and properties of extracellular
endoxylanases from alkalophilic thermophilic Bacillus sp. Can J Microbiol 38:436–442
Dheeran P, Nandhagopal N, Kumar S, Jaiswal YK, Adhikari DK (2012) A novel thermostable
xylanase of Paenibacillus macerans IIPSP3 isolated from the termite gut. J Ind Microbiol
Biotechnol 39(6):851–860
Dhillon A, Gupta JK, Khanna S (2000) Enhanced production, purification and characterisation of a
novel cellulase-poor thermostable, alkalitolerant xylanase from Bacillus circulans AB 16.
Process Biochem 35(8):849–856
Dhiman SS, Sharma J, Battan B (2008) Industrial applications and future prospects of microbial
xylanases: a review. BioResources 3(4):1377–1402
Dijkerman R, Ledeboer J, Op den Camp HJM, Prins RA, van der Drift C (1997) The anaerobic
fungus Neocallimastix sp. strain L2: growth and production of (Hemi) cellulolytic enzymes on
a range of carbohydrate substrates. Curr Microbiol 34(2):91–96
Dornez E, Croes E, Gebruers K, De Coninck B, Cammue BPA, Delcour JA, Courtin CM (2010a)
Accumulated evidence substantiates a role for three classes of wheat xylanase inhibitors in
plant defense. Crit Rev Plant Sci 29(4):244–264
Dornez E, Croes E, Gebruers K, Carpentier S, Swennen R, Laukens K, Witters E, Urban M,
Delcour JA, Courtin CM (2010b) 2-D DIGE reveals changes in wheat xylanase inhibitor
protein families due to Fusarium graminearum DeltaTri5 infection and grain development.
Proteomics 10(12):2303–2319
Durand A, Hughes R, Roussel A, Flatman R, Henrissat B, Juge N (2005) Emergence of a
subfamily of xylanase inhibitors within glycoside hydrolase family 18. FEBS J 272
(7):1745–1755
Ebanks R, Dupont M, Shareck F, Morosoli R, Kluepfel D, Dupont C (2000) Development of an
Escherichia coli expression system and thermostability screening assay for libraries of mutant
xylanase. J Ind Microbiol Biotechnol 25(6):310–314
El Enshasy H, Hellmuth K, Rinas U (1999) Fungal morphology in submerged cultures and its
relation to glucose oxidase excretion by recombinant Aspergillus niger. Appl Biochem
Biotechnol 81:1–11
El Enshasy H, Kleine J, Rinas U (2006) Agitation effects on morphology and protein productive
fractions of filamentous and pelleted growth forms of recombinant Aspergillus niger. Process
Biochem 41:2103–2112
El Enshasy H (2007) Filamentous fungal culture—process characteristics, products, and
applications. In: Yang ST (ed) Bioprocessing for value-added products from renewable
resources. Elsevier Press, Amsterdam. ISBN-10: 0-444-52114-3
Emami K, Hack E (2001) Characterisation of a xylanase gene from Cochliobolus sativus and its
expression. Mycol Res 105:352–359
Esteban R, Villanueva JR, Villa TG (1982) b-D-xylanases of Bacillus circulans WL-12. Can J
Microbiol 28:733–739
Fang HY, Chang SM, Lan CH, Fang TJ (2008) Purification and characterization of a xylanase
from Aspergillus carneus M34 and its potential use in photoprotectant preparation. Process
Biochem 43(1):49–55
204 H.A. El Enshasy et al.

Farliahati MR, Mohamad R, Puspaningsih NNT, Ariff AB (2009) Kinetics of xylanase


fermentation by recombinant Escherichia coli DH5a in shake flask culture. Am J Biochem
Biotechnol 5(3):110–118
Farliahati MR, Ramanan RN, Mohamad R, Puspaningsih NNTM, Ariff AB (2010) Enhanced
production of xylanase by recombinant Escherichia coli DH5 alpha through optimization of
medium composition using response surface methodology. Ann Microbiol 60(2):279–285
Fernández-Espinar M, Pinaga F, Degraaff L, Visser J, Ramon D, Valles S (1994) Purification,
characterization and regulation of the synthesis of an Aspergillus nidulans acidic xylanase.
Appl Microbiol Biotechnol 42(4):555–562
Fierens E, Gebruers K, Voet AR, De Maeyer M, Courtin CM, Delcour JA (2009) Biochemical and
structural characterization of TLXI, the Triticum aestivum L. thaumatin-like xylanase inhibitor.
J Enzyme Inhib Med Chem 24(3):646–654
Fierens E, Rombouts S, Gebruers K, Goesaert H, Brijs K, Beaugrand J, Volckaert G, Van
Campenhout S, Proost P, Courtin CM, Delcour JA (2007) TLXI, a novel type of xylanase
inhibitor from wheat (Triticum aestivum) belonging to the thaumatin family. Biochem J 403
(3):583–591
Fierens K, Geudens N, Brijs K, Courtin CM, Gebruers K, Robben J, Van Campenhout S,
Volckaert G, Delcour JA (2004) High-level expression, purification, and characterization of
recombinant wheat xylanase inhibitor TAXI-I secreted by the yeast Pichia pastoris. Protein
Expr Purif 37(1):39–46
Flatman R, McLauchlan WR, Juge N, Furniss C, Berrin JG, Hughes RK, Manzanares P,
Ladbury JE, O’Brien R, Williamson G (2002) Interactions defining the specificity between
fungal xylanases and the xylanase-inhibiting protein XIP-I from wheat. Biochem J 365(Pt
3):773–781
Frederix SA, Courtin CM, Delcour JA (2004) Substrate selectivity and inhibitor sensitivity affect
xylanase functionality in wheat flour gluten–starch separation. J Cer Sci 40:41–49
Fushinobu S, Ito K, Konno M, Wakagi T, Matsuzawa H (1998) Crystallographic and mutational
analyses of an extremely acidophilic and acid-stable xylanase: biased distribution of acidic
residues and importance of Asp37 for catalysis at low pH. Protein Eng 11(12):1121–1128
Gao J, Zhang HJ, Yu SH, Wu SG, Yoon I, Quigley J, Gao YP, Qi GH (2008) Effects of yeast
culture in broiler diets on performance and immunomodulatory functions. Poult Sci
87:1377–1384
Garai D, Kumar V (2012) Response surface optimization for xylanase with high volumetric
productivity by indigenous alkali tolerant Aspergillus candidus under submerged cultivation.
Biotechnology 3:127–136
Garg AP, Roberts JC, McCarthy AJ (1998) Bleach boosting effect of cellulase-free xylanase of
Streptomyces thermoviolaceus and its comparison with two commercial enzyme preparations
on Birchwood kraft pulp. Enzyme Microb Technol 22:594–598
Gebruers K, Brijs K, Courtin CM, Fierens K, Goesaert H, Raedschelders G, Robben J,
Sorensen JF, Van Campenhout S, Delcour JA (2004) Properties of TAXI-type endoxylanase
inhibitors. Biochim Biophys Acta 1696:213–221
Gebruers K, Beaugrand J, Croes E, Dornez E, Courtin CM, Delcour JA (2008) Quantification of
wheat TAXI and XIP type xylanase inhibitors: a comparison of analytical techniques. Cer
Chem 85(5):586–590
Georlette D, Blaise V, Collins T, D’Amico S (2004) Some like it cold: biocatalysis at low
temperatures. FEMS Microbiol Rev 28:25–42
Gessesse A, Mamo G (1998) Purification and characterization of an alkaline xylanase from
alkaliphilic Micrococcus sp AR-135. J Ind Microbiol Biotechnol 20(3–4):210–214
Ghosh M, Nanda G (1994) Purification and some properties of a xylanase from Aspergillus
sydowii MG49. Appl Environ Microbiol 60(12):4620–4623
Goesaert H, Gebruers K, Courtin CM, Delcour JA (2005) Purification and characterization of a
XIP-type endoxylanase inhibitor from rice (Oryza sativa). J Enz Inhib 20:95–101
7 Microbial Xylanases: Sources, Types, and Their Applications 205

Georis J, Giannotta F, De Buyl E, Granier B, Frère J (2000) Purification and properties of three
endo-b-1,4-xylanases produced by Streptomyces sp. strain S38 which differ in their ability to
enhance the bleaching of kraft pulps. Enzyme Microb Technol 26:178–186
Gomes DJ, Gomes J, Steiner W (1994) Factors influencing the induction of endo-xylanase by
Thermoascus aurantiacus. J Biotechnol 33(1):87–94
Gomez LD, Steele-King CG, McQueen-Mason SJ (2008) Sustainable liquid biofuels from
biomass: the writing’s on the walls. New Phytol 178(3):473–485
Gruber K, Klintschar G, Hayn M, Schlacher V, Steiner W, Kratky C (1998) Thermophilic
xylanase from Thermomyces lanuginosus: high-resolution X-ray structure and modeling
studies. Biochemistry 37(39):13475–13485
Guo B, Chen XL, Sun CY, Zhou BC, Zhang YZ (2009) Gene cloning, expression and
characterization of a new cold-active and salt-tolerant endo-beta-1,4-xylanase from marine
Glaciecola mesophila KMM 241. Appl Microbiol Biotechnol 84(6):1107–1115
Gupta S, Bhushan B, Hoondal GS (2000) Isolation, purification and characterization of xylanase
from Staphylococcus sp. SG-13 and its application in biobleaching of kraft pulp. J Appl
Microbiol 88:325–334
Gupta U, Kar R (2009) Xylanase production by a thermos-tolerant Bacillus species under
solid-state and submerged fermentation. Braz Arch Biol Technol 52:1363–1371
Hakulinen N, Tenkanen M, Rouvinen J (1998) Crystallization and preliminary X-ray diffraction
studies of the catalytic core of acetyl xylan esterase from Trichoderma reesei. Acta
Crystallogr D Biol Crystallogr 54(Pt 3):430–443
Henrissat B, Bairoch A (1993) New families in the classification of glycosyl hydrolases based on
amino acid sequence similarities. Biochem J 293(3):781–788
Hessing JGM, van Rotterdam C, Verbakel JMA, Roza M, Maat J, van Gorcom RFM, van den
Hondel CAMJJ (1994) Isolation and characterization of a 1,4-b-endoxylanase gene of A.
awamori. Curr Genet 26:228–232
Himmel ME, Ding SY, Johnson DK, Adney WS, Nimlos MR, Brady JW, Foust TD (2007)
Biomass recalcitrance: engineering plants and enzymes for biofuels production. Science 315
(5813):804–807
Honda Y and Kitaoka M (2004) A family 8 glycoside hydrolase from Bacillus halodurans C-125
(BH2105) is a reducing end xylose-releasing exo-oligoxylanase. J Biol Chem 279:55097–55103
Hu YJ, Smith DC, Cheng KJ, Foresberg CW (1991) Cloning of a xylanase gene from Fibrobacter
succinogenes 135 and its expression in Escherichia coli. Can J Microbiol 37(7):554–561
Huang J, Wang G, Xiao L (2006) Cloning, sequencing and expression of the xylanase gene from a
Bacillus subtilis strain B10 in Escherichia coli, Bioresour Technol 97:802–808
Humphry DR, GeorgeA Black GW, Cummings SP (2001) Flavobacterium frigidarium sp. an
aerobic, psychrophilic, xylanolytic and laminarinolytic bacterium from Antarctica. Int J Syst
Evol Microbiol 51:1235–1243
Hung KS, Liu SM, Fang TY, Tzou WS, Lin FP, Sun KH, Tang SJ (2011) Characterization of a
salt-tolerant xylanase from Thermoanaerobacterium saccharolyticum NTOU1. Biotechnol Lett
33(7):1441–1447
Igawa T, Tokai T, Kudo T, Yamaguchi I, Kimura M (2005) A wheat xylanase inhibitor gene,
Xip-I, but not Taxi-I, is significantly induced by biotic and abiotic signals that trigger plant
defense. Biosci Biotechnol Biochem 69(5):1058–1063
Ingelbrecht JA, Moers K, Abecassis J, Rouau X, Delcour JA (2001) Influence of arabinoxylans
and endoxylanases on pasta processing and quality. Production of high-quality pasta with
increased levels of soluble fiber. Cereal Chem 78:721–729
Ito K, Ikemasu T, Ishikawa T (1992) Cloning and sequencing of the xynA gene encoding xylanase
A of Aspergillus kawachii. Biosci Biotechnol Biochem 56(6):906–912
Jeong MY, Kim S, Yun CW, Choi YJ, Cho SG (2007) Engineering a de novo internal disulfide
bridge to improve the thermal stability of xylanase from Bacillus stearothermophilus No. 236.
J Biotechnol 127(2):300–309
206 H.A. El Enshasy et al.

Joo JC, Pack SP, Kim YH, Yoo YJ (2011) Thermostabilization of Bacillus circulans xylanase:
computational optimization of unstable residues based on thermal fluctuation analysis.
J Biotechnol 151(1):56–65
Joshi MD, Hedberg A, McIntosh LP (1997) Complete measurement of the pKa values of the
carboxyl and imidazole groups in Bacillus circulans xylanase. Protein Sci 6(12):2667–2670
Jun H, Bing Y, Zhang K, Ding X, Daiwen C (2008) Expression of a Trichoderma reesei
B-xylanase gene in Escherichia coli and activity of the enzyme on fiber-bound substrates.
Protein Expr Purif 67:1–6
Kapoor M, Nair LM, Kuhad RC (2008) Cost-effective xylanase production from free and
immobilized Bacillus pumilus strain MK001 and its application in saccharification of Prosopis
juliflora. Biochem Eng J 38:88–97
Karlsson EN, BartonekRoxa E, Holst O (1997) Cloning and sequence of a thermostable
multidomain xylanase from the bacterium Rhodothermus marinus. Biophys Acta Gene Struct
Expr 1353(2):118–124
Karlsson EN, Bartonek Roxa E, Holst O (1998) Evidence for substrate binding of a recombinant
thermostable xylanase originating from Rhodothermus marinus. FEMS Microbiol Lett 168
(1):1–7
Karlsson EN, Holst O, Tocaj A (1999) Efficient production of truncated thermostable xylanases
from Rhodothermus marinus in Escherichia coli fed-batch cultures. J Biosci Bioeng
87:598–606
Katapodis P, Christakopoulos P (2008) Enzymatic production of feruloyl xylo-oligosaccharides
from corn cobs by a family 10 xylanase from Thermoascus aurantiacus. LWT Food Sci
Technol 41(7):1239–1243
Khandeparkar R, Bhosle NB (2006) Purification and characterization of thermoalkalophilic
xylanase isolated from the Enterobacter sp. MTCC 5112. Res Microbiol 157(4):315–325
Khasin A, Alchanati I, Shoham Y (1993) Purification and characterization of a thermostable
xylanase from Bacillus stearothermophilus T-6. Appl Environ Microbiol 59(6):1725–1730
Kimura T, Kitamoto N, Kito Y, Karita S, Sakka K, Ohmiya K (1998) Molecular cloning of
xylanase gene xynG1 from Aspergillus oryzae KBN 616, a shoyu koji mold, and analysis of its
expression. J Ferm Bioeng 85(1):10–16
Kimura T, Ito J, Kawano A, Makino T, Kondo H, Karita S, Sakka K, Ohmiya K (2000)
Purification, characterization, and molecular cloning of acidophilic xylanase from Penicillium
sp. 40. Biosci Biotechnol Biochem 64(6):1230–1237
Kitamoto N, Yoshino S, Ohmiya K, Tsukagoshi N (1999) Purification and characterization of the
over expressed Aspergillus oryzae xylanase, XynF1. Biosci Biotechnol Biochem 63
(10):1791–1794
Ko EP, Akatsuka H, Moriyama H, Shinmyo A, Hata Y, Katsube Y, Urabe I, Okada H (1992)
Site-directed mutagenesis at aspartate and glutamate residues of xylanase from Bacillus
pumilus. Biochem J 288(Pt 1):117–121
Ko CH, Tsai CH, Tu J, Yang BY, Hsieh DL, Jane WN, Shih TL (2011) Identification of
Paenibacillus sp. 2S-6 and application of its xylanase on biobleaching. Int Biodeterior
Biodegrad 65:334–339
Kohli U, Nigam P, Singh D, Chaudhary K (2001) Thermostable, alkalophilic and cellulase free
xylanase production by Thermoactinomyces thalophilus subgroup C. Enzyme Microb Technol
28(7–8):606–610
Kolenova K, Vrsanska M, Biely P (2006) Mode of action of endo-beta-1,4-xylanases of families
10 and 11 on acidic xylooligosaccharides. J Biotechnol 121:338–345
Kormelink FJ, Gruppen H, Vietor RJ, Voragen AG (1993) Mode of action of the xylan-degrading
enzymes from Aspergillus awamori on alkali-extractable cereal arabinoxylans. Carbohydr Res
249(2):355–367
Krengel U, Dijkstra BW (1996) Three-dimensional structure of Endo-1,4-beta-xylanase I from
Aspergillus niger: molecular basis for its low pH optimum. J Mol Biol 263(1):70–78
Kudo T, Ohkoshi A, Horikoshi K (1985) Molecular cloning and expression of a xylanase gene of
alkalophilic Aeromonas sp. no. 212 in Escherichia coli. J Gen Microbiol 131(10):2825–2830
7 Microbial Xylanases: Sources, Types, and Their Applications 207

Kui H, Luo H, Shi P, Bai Y, Yuan T, Wang Y, Yang P, Dong S, Yao B (2010) Gene cloning,
expression, and characterization of a thermostable xylanase from Nesterenkonia xinjiangensis
CCTCC AA001025. Appl Biochem Biotechnol 162(4):953–965
Kulkarni N, Lakshmikumaran M, Rao M (1999) Xylanase II from an alkaliphilic thermophilic
Bacillus with a distinctly different structure from other xylanases: evolutionary relationship to
alkaliphilic xylanases. Biochem Biophys Res Commun 263(3):640–645
Kumar KS, Manimaran A, Permaul K, Singh S (2009) Production of b-xylanase by a
Thermomyces lanuginosus MC 134 mutant on corn cobs and its application in biobleaching of
bagasse pulp. J Biosci Bioeng 107(5):494–498
Kumar PR, Eswaramoorthy S, Vithayathil PJ, Viswamitra MA (2000) The tertiary structure at 1.59
Å resolution and the proposed amino acid sequence of a family-11 xylanase from the
thermophilic fungus Paecilomyces varioti Bainier. J Mol Biol 295(3):581–593
Kumar V, Satyanarayana T (2011) Applicability of thermo-alkali-stable and cellulase-free
xylanase from a novel thermo-halo-alkaliphilic Bacillus haloduransin producing xylooligosac-
charides. Biotechnol Lett 33:2279–2285
La Grange DC, Claeyssens M, Pretorius IS, Van Zyl WH (2000) Coexpression of the Bacillus
pumilus beta-xylosidase (xynB) gene with the Trichoderma reesei beta xylanase 2 (xyn2) gene
in the yeast Saccharomyces cerevisiae. Appl Microbiol Biotechnol 54(2):195–200
Lagaert S, Van Campenhout S, Pollet A, Bourgois TM, Delcour JA, Courtin CM, Volckaert G
(2007) Recombinant expression and characterization of a reducing-end xylose-releasing
exo-oligoxylanase from Bifidobacterium adolescentis. Appl Environ Microbiol 73:5374–5377
Lama L, Calandrelli V, Gambacorta A, Nicolaus B (2004) Purification and characterization of
thermostable xylanase and beta-xylosidase by the thermophilic bacterium Bacillus ther-
mantarcticus. Res Microbiol 155(4):283–289
Lamed R, Setter E, Bayer EA (1983) Characterization of a cellulose-binding, cellulase-containing
complex in Clostridium thermocellum. J Bacteriol 156(2):828–836
Lappalainen A, Siika-aho M, Kalkkinen N, Fagerstrom R, Tenkanen M (2000) Endoxylanase II
from Trichoderma reesei has several isoforms with different isoelectric points. Biotechnol Appl
Biochem 31:61e68
Larson SB, Day J, Barba de la Rosa AP, Keen NT, McPherson A (2003) First crystallographic
structure of a xylanase from glycoside hydrolase family 5: implications for catalysis.
Biochemistry 42(28):8411–8422
Lee J (1997) Biological conversion of lignocellulosic biomass to ethanol. J Biotechnol 56(1):1–24
Lee JM, Hu Y, Zhu H, Cheng KJ, Krell PJ, Forsberg CW (1993) Cloning of a xylanase gene from
the ruminal fungus Neocallimastix patriciarum 27 and its expression in Escherichia coli. Can J
Microbiol 39(1):134–139
Lee TH, Lim PO, Lee YE (2007) Cloning, characterization, and expression of xylanase A gene
from Paenibacillus sp. DG-22 in Escherichia coli. J Microbiol Biotechnol 17(1):29–36
Levasseur A, Asther M, Record E (2005) Overproduction and characterization of xylanase B from
Aspergillus niger. Can J Microbiol 51(2):177–183
Li XL, Ljungdahl LG (1996) Expression of Aureobasidium pullulans xynA in, and secretion of the
xylanase from Saccharomyces cerevisiae. Appl Environ Microbiol 62(1):209–213
Li JH, Wang LS (2011) Why substituting the asparagine at position 35 in Bacillus circulans
xylanase with an aspartic acid remarkably improves the enzymatic catalytic activity? A
quantum chemistry-based calculation study. Polym Degrad Stab 96(5):1009–1014
Li C, Knierim B, Manisseri C, Arora R, Scheller HV, Auer M, Vogel KP, Simmons BA, Singh S
(2010) Comparison of dilute acid and ionic liquid pretreatment of switchgrass: biomass
recalcitrance, delignification and enzymatic saccharification. Bioresour Technol 101(13):4900–
4906
Li XL, Skory CD, Ximenes EA, Jordan DB, Dien BS, Hughes SR, Cotta MA (2007) Expression of
an AT-rich xylanase gene from the anaerobic fungus Orpinomyces sp. strain PC-2 in and
secretion of the heterologous enzyme by Hypocrea jecorina. Appl Microbiol Biotechnol
74:1264–1275
208 H.A. El Enshasy et al.

Liu JR, Duan CH, Zhao X, Tzen JT, Cheng KJ, Pai CK (2008) Cloning of a rumen fungal xylanase
gene and purification of the recombinant enzyme via artificial oil bodies. Appl Microbiol
Biotechnol 79(2):225–233
Liu MQ, Weng XY, Sun JY (2006) Expression of recombinant Aspergillus niger xylanase A in
Pichia pastoris and its action on xylan. Protein Expr Purif 48(2):292–299
Lo YC, Lu WC, Chen CY, Chen WM, Chang JS (2010) Characterization and high-level
production of xylanase from an indigenous cellulolytic bacterium Acinetobacter junii F6-02
from southern Taiwan soil. Biochem Eng J 53(1):77–84
Lundgren KR, Bergkvist L, Hogman S, Joves H, Eriksson G, Bartfai T, Vanderlaan J,
Rosenberg E, Shoham Y (1994) TCF mill trial on softwood pulp with korsnas thermostable
and alkaline stable xylanase T6. Fems Microbiology Reviews 13(2–3):365–368
Luthi E, Love DR, McAnulty J, Wallace C, Caughey PA, Saul D, Bergquist PL (1990) Cloning,
sequence analysis, and expression of genes encoding xylan-degrading enzymes from the
thermophile Caldocellum saccharolyticum. Appl Environ Microbiol 56(4):1017–1024
Lynd LR, Cushman JH, Nichols RJ, Wyman CE (1991) Fuel ethanol from cellulosic biomass.
Science 251(4999):1318–1323
Mach-Aigner A, Pucher ME, Mach RL (2010) D-Xylose as a repressor or inducer of xylanase
expression in Hypocrea jecorina (Trichoderma reesei). Appl Environ Microbiol 76:1770–1776
Mamo G, Hatti-Kaul R, Mattiasson B (2006) A thermostable alkaline active endo-b-1-4-xylanase
from Bacillus halodurans S7: Purification and characterization. Enzyme Microb Technol
39:1492–1498
Mamo G, Hatti-Kaul R, Mattiasson B (2007) Fusion of carbohydrate binding modules from
Thermotoga neapolitana with a family 10 xylanase from Bacillus halodurans S7.
Extremophiles 11:169–177
Mannarelli BM, Evans S, Lee D (1990) Cloning, sequencing, and expression of a xylanase gene
from the anaerobic ruminal bacterium Butyrivibrio fibrisolvens. J Bacteriol 172(8):4247–4254
Margeot A, Hahn-Hagerdal B, Edlund M, Slade R, Monot F (2009) New improvements for
lignocellulosic ethanol. Curr Opin Biotechnol 20:372–380
Marrone L, McAllister KA, Clarke AJ (2000) Characterization of function and activity of domains
A, B and C of xylanase C from Fibrobacter succinogenes S85. Protein Eng 13(8):593–601
McCarthy AA, Morris DD, Bergquist PL, Baker EN (2000) Structure of XynB, a highly
thermostable beta 1,4-xylanase from Dictyoglomus thermophilum Rt46B.1, at 1.8 Å resolution.
Acta Crystallogr D56:1367–1375
Mirande C, Mosoni P, Bera-Maillet C, Bernalier-Donadille A, Forano E (2010) Characterization of
Xyn10A, a highly active xylanase from the human gut bacterium Bacteroides xylanisolvens
XB1A. Appl Microbiol Biotechnol 87(6):2097–2105
Moreau A, Durand S, Morosoli R (1992) Secretion of a Cryptococcus albidus xylanase in
Saccharomyces cerevisiae. Gene 116(1):109–113
Morris DD, Gibbs MD, Bergquist PL (1996) Cloning of a family G xylanase gene (XYNB) from
the extremely thermophilic bacterium Dictyoglomus thermophilum and action of the gene
product on kraft pulp. Am Chem Soc 211:234
Muilu J, Torronen A, Perakyla M, Rouvinen J (1998) Functional conformational changes of
endo-1,4-xylanase II from Trichoderma reesei: a molecular dynamics study. Proteins
31:434–444
Mullai P, Fathima NSA, Rene ER (2010) Statistical analysis of main and interaction effects to
optimize xylanase production under submerged cultivation conditions. J Agric Sci 2
(1):144–153
Nakamura S, Wakabayashi K, Nakai R, Aono R, Horikoshi K (1993) Production of alkaline
xylanase by a newly isolated alkaliphilic Bacillus sp strain 41 m-1. World J Microbiol
Biotechnol 9(2):221–224
Narang S, Satyanarayana T (2001) Thermostable alpha-amylase production by an extreme
thermophile Bacillus thermooleovorans. Lett Appl Microbiol 32:31–35
7 Microbial Xylanases: Sources, Types, and Their Applications 209

Nascimento RP, Coelho RRR, Marques S, Alves L, Girio FM, Bon EPS, Amaral-Collaco MT
(2002) Production and partial characterisation of xylanase from Streptomyces sp strain AMT-3
isolated from Brazilian cerrado soil. Enzyme Microb Technol 31(4):549–555
Natesh R, Bhanumoorthy P, Vithayathil PJ, Sekar K, Ramakumar S, Viswamitra MA (1999)
Crystal structure at 1.8 angstrom resolution and proposed amino acid sequence of a
thermostable xylanase from Thermoascus aurantiacus. J Mol Biol 288(5):999–1012
Ogasawara W, Shida Y, Furukawa T, Shimada R, Nakagawa S, Kawamura M, Yagyu T,
Kosuge A, Xu J, Nogawa M, Okada H, Morikawa Y (2006) Cloning, functional expression and
promoter analysis of xylanase III gene from Trichoderma reesei. Appl Microbiol Biotechnol
72(5):995–1003
Ohta K, Moriyama S, Tanaka H, Shige T, Akimoto H (2001) Purification and characterization of
an acidophilic xylanase from Aureobasidium pullulans var. melanigenum and sequence
analysis of the encoding gene. J Biosci Bioeng 92(3):262–270
Okeke BC, Obi SKC (1994) Lignocellulose and sugar compositions of some agro-waste materials.
Bioresour Technol 47(3):283–284
Olfa E, Mondher M, Issam S, Ferid L, Nejib NM (2007) Induction, properties and application of
xylanase activity from Sclerotinia sclerotiorum S2 fungus. J Food Chem 31:96–107
Paës G, Berrin J-G, Beaugrand J (2012) GH11 xylanases: structure/function/properties relation-
ships and applications. Biotechnol Adv 30:564–592
Paës G, O’Donohue MJJ (2006) Engineering increased thermostability in the thermostable GH-11
xylanase from Thermobacillus xylanilyticus. J Biotechnol 125:338–350
Payan F, Leone P, Porciero S, Furniss C, Tahir T, Williamson G, Durand A, Manzanares P,
Gilbert HJ, Juge N, Roussel A (2004) The dual nature of the wheat xylanase protein inhibitor
XIP-I: structural basis for the inhibition of family 10 and family 11 xylanases. J Biol Chem 279
(34):36029–36037
Payan F, Flatman R, Porciero S, Williamson G, Juge N, Roussel A (2003) Structural analysis of
xylanase inhibitor protein I (XIP-I), a proteinaceous xylanase inhibitor from wheat (Triticum
aestivum var soisson). Biochem J 372(Pt 2):399–405
Petrescu I, Lamotte-Brasseur J, Chessa JP, Ntarima P, Claeyssens M, Devreese B, Marino G,
Gerday C (2000) Xylanase from the psychrophilic yeast Cryptococcus adeliae. Extremophiles
4(3):137–144
Pokhrel S, Joo JC, Yoo YJ (2013) Shifting the optimum pH of Bacillus circuulans xylanase
towards acidic side by introducing arginine. Biotechnol Bioprocess Eng 18:35–42
Polizeli MLTM, Rizzatti ACS, Monti R, Terenzi HF, Jorge JA, Amorim DS (2005) Xylanases
from fungi: properties and industrial applications. Appl Microbiol Biotechnol 67:577–591
Pollet A, Schoepe J, Dornez E, Strelkov SV, Delcour JA, Courtin CM (2010) Functional analysis
of glycosidehydrolase family 8 xylanases shows narrow but distinct substrate specificities and
biotechnological potential. Appl Microbiol Biotechnol 87:2125–2135
Prade RA (1996) Xylanases: from biology to biotechnology. Biotechnol Genet Eng Rev 13
(1):101–132
Puchkaev AV, Koo LS, Ortiz de Montellano PR (2003) Aromatic stacking as a determinant of the
thermal stability of CYP119 from Sulfolobus solfataricus. Arch Biochem Biophys 409:52–58
Puls J, Tenkanen M, Korte HE, Poutanen K (1991) Products of hydrolysis of beechwood
acetyl-4-O-methylglucuronoxylan by a xylanase and an acetyl xylan esterase. Enzyme Microb
Technol 13(6):483–486
Qureshy AF, Khan LA, Khanna S (2002) Cloning, regulation and purification of cellulase-free
xylanase from Bacillus circulans Teri-42. Ind J Microbiol 42:35–41
Raedschelders G, Debefve C, Goesaert H, Delcour JA, Volckaert G, Van Campenhout S (2004)
Molecular identification and chromosomal localization of genes encoding Triticum aestivum
xylanase inhibitor I-like proteins in cereals. Theor Appl Genet 109(1):112–121
Ratto M, Poutanen K, Viikari L (1992) Production of xylanolytic enzymes by an alkalitolernt
Bacillus circulans stain. Appl Microbiol Biotechnol 37:470–473
210 H.A. El Enshasy et al.

Rizzatti ACS, Jorge JA, Terenzi HF, Rechia CGV, Polizeli MLTM (2001) Purification and
properties of a thermostable extracellular b-xylosidase produced by a thermotolerant
Aspergillus phoenicis. J Ind Microbiol Biotechnol 26:156–160
Rose SH, van Zyl WH (2002) Constitutive expression of the Trichoderma reesei beta-1,4-xylanase
gene (xyn2) and the beta-1,4-endoglucanase gene (egl) in Aspergillus niger in molasses and
defined glucose media. Appl Microbiol Biotechnol 58(4):461–468
Rouau X (1993) Investigations into the effects of an enzyme preparation for baking on what flour
dough pentosans. J Cereal Sic 18:145–157
Rouau X, Daviet S, Tahir T, Cherel B, Saulnier L (2006) Effect of the proteinaceous wheat
xylanase inhibitor XIP-I on the performances of an Aspergillus niger xylanase in bread
making. J Sci Food Agric 86(11):1604–1609
Ruanglek V, Sriprang R, Ratanaphan N, Tirawongsaroj P, Chantasigh D, Tanapongpipat S,
Pootanakit K, Eurwilaichitr L (2007) Cloning, expression, characterization, and high
cell-density production of recombinant endo-1,4-beta-xylanase from Aspergillus niger in
Pichia pastoris. Enzyme Microb Technol 41(1–2):19–25
Saha BC (2003) Hemicellulose bioconversion. J Ind Microbiol Biotechnol 30(5):279–291
Sakka K, Maeda Y, Hakamada Y, Takahashi N, Shimada K (1991) Purification and some
properties of xylanase from Clostridium stercorarium strain Hx-1. Agric Biol Chem 55
(1):247–248
Salles BC, Cunha RB, Fontes W, Sousa MV, Filho EXF (2000) Purification and characterization
of a new xylanase from Acrophialophora nainiana. J Biotechnol 81(2–3):199–204
Salles BC, Te’o VS, Gibbs MD, Bergquist PL, Filho EX, Ximenes EA, Nevalainen KM (2007)
Identification of two novel xylanase-encoding genes (xyn5 and xyn6) from Acrophialophora
nainiana and heterologous expression of xyn6 in Trichoderma reesei. Biotechnol Lett 29
(8):1195–1201
Sanghi A, Garg N, Kuhar K, Kuhad RC, Gupta VK (2009) Enhanced production of cellulase-free
xylanase by alkalophilic Bacillus subtilis ASH and its application in biobleaching of kraft
pulp. BioResources 4:1109–1129
Sandhu JS, Kennedy JF (1984) Molecular cloning of Bacillus polymyxa (1–4)-P-D-xylanase gene
in Escherichia coli. Enzyme Microb Technol 6:271–274
Sapre MP, Jha H, Patil MB (2005) Purification and characterization of a thermoalkalophilic
xylanase from Bacillus sp. World J Microbiol Biotechnol 21(5):649–654
Schlacher A, Holzmann K, Hayn M, Steiner W, Schwab H (1996) Cloning and characterization of
the gene for the thermostable xylanase XynA from Thermomyces lanuginosus. J Biotechnol 49
(1–3):211–218
Schulze E (1891) Amphibia Europaea, annual report and treatises society of natural scientists in
Magdeburg 1890:163–178
Shallom D, Shoham Y (2003) Microbial hemicellulases. Curr Opin Microbiol 6(3):219–228
Shao W, Wiegel J (1995) Purification and characterization of two thermostable acetyl xylan
esterases from Thermoanaerobacterium sp. strain JW/SL-YS485. Appl Environ Microbiol 61
(2):729–733
Sharma A, Adahikari S, Styanarayana T (2007) Alkali-thermostable and cellulase-free xylanase
production by an extreme thermophile Geobacillus thermoleovorans. World J Microbiol
Biotechnol 23:483–490
Shatalov AA, Pereira AA (2007) Xylanase pre-treatment of giant reed organosolv pulps: direct
bleaching effect and bleach boosting. Ind Crops Prod 25:248–256
Shibuya H, Kaneko S, Hayanshi K (2000) Enhancement of thermostability and hydrolytic activity
of xylananse by random gene shuffling. Biochem J 349:651–656
Shrinivas D, Savitha G, Raviranjan K, Naik GR (2010) A highly thermostable alkaline
cellulase-free xylanase from thermoalkalophilic Bacillus sp. JB 99 suitable for paper and pulp
industry: purification and characterization. Appl Biochem Biotechnol 162(7):2049–2057
Sidhu G, Withers SG, Nguyen NT, McIntosh LP, Ziser L, Brayer GD (1999) Sugar ring distortion
in the glycosyl-enzyme intermediate of a family G/11 xylanase. Biochemistry 38
(17):5346–5354
7 Microbial Xylanases: Sources, Types, and Their Applications 211

Silva CHC, Puls J, de Sousa MV, Ferreira EX (1999) Purification and characterization of a low
molecular weight xylanase from solid-state cultures of Aspergillus fumigatus Fresenius. Rev
Microbiol 30(2):114–119
Simpson HD, Haufler UR, Daniel RM (1991) An extremely thermostable xylanase from the
thermophilic bacterium Thermotoga. Biochem J 277(2):413–417
Simpson PJ, Bolam DN, Cooper A, Ciruela A, Hazlewood GP, Gilbert HJ, Williamson MP (1999)
A family IIb xylan-binding domain has a similar secondary structure to a homologous family
IIa cellulose-binding domain but different ligand specificity. Structure 7(1999):853–864
Singh S, Pillay B, Dilsook V, Prior BA (2000) Production and properties of hemicellulases by a
Thermomyces lanuginosus strain. J Appl Microbiol 88:975–982
Sorensen JF, Sibbesen O (2006) Mapping of residues involved in the interaction between the
Bacillus subtilis xylanase A and proteinaceous wheat xylanase inhibitors. Protein Eng Des Sel
19(5):205–210
Sriprang R, Asano K, Gobsuk J, Tanapongpipat S, Champreda V, Eurwilaichitr L (2006)
Improvement of thermostability of fungal xylanase by using site-directed mutagenesis.
J Biotechnol 126(4):454–462
Srivastava R, Srivastava AK (1993) Characterization of a bacterial xylanase resistant to repression
by glucose and xylose. Biotechnol Lett 15(8):847–852
Sriyapai T, Somyoonsap P, Matsui K, Kawai F, Chansiri K (2011) Cloning of a thermostable
xylanase from Actinomadura sp. S14 and its expression in Escherichia coli and Pichia
pastoris. J Biosci Bioeng 111(5):528–536
St John FJ, Godwin DK, Preston JF, Pozharski E, Hurlbert JC (2009) Crystallization and
crystallographic analysis of Bacillus subtilis xylanase C. Acta Crystallogr Sect F Struct Biol
Crystallogr Commun 65(Pt 5):499–503
Strauss J, Mach RL, Zeilinger S, Hartler G, Stoffler G, Wolschek M, Kubicek CP (1995) Cre1, the
carbon catabolite repressor protein from Trichoderma reesei. FEBS Lett 376:103–107
Stephens DE, Singh S, Permaul K (2009) Error-prone PCR of a fungal xylanase for improvement
of its alkaline and thermal stability. FEMS Microbiol Lett 293(1):42–47
Subramaniyan S (2000) Studies on the production of bacterial xylanases. Ph.D. Thesis, Cochin
University of Science and Technology, India
Subramaniyan S, Prema P (2002) Biotechnology of microbial xylanases: enzymology, molecular
biology and application. Crit Rev Biotechnol 22:33–46
Sun JY, Liu MQ, Weng XY, Qian LC, Gu SH (2007) Expression of recombinant
Thermomonospora fusca xylanase A in Pichia pastoris and xylooligosaccharides released
from xylans by it. Food Chem 104(3):1055–1064
Sunna A, Prowe SG, Stoffregen T, Antranikian G (1997) Characterization of the xylanases from
the new isolated thermophilic xylan-degrading Bacillus thermoleovorans strain K-3d and
Bacillus flavothermus strain LB3A. FEMS Microbiol Lett 148(2):209–216
Suzuki T, Ibata K, Hatsu M, Takamizawa K, Kawai K (1997) Cloning and expression of a 58-kDa
xylanase VI gene (xynD) of Aeromonas caviae ME-1 in Escherichia coli which is not
categorized as a family F or family G xylanase. J Ferm Bioeng 84(1):86–89
Takahashi-Ando N, Inaba M, Ohsato S, Igawa T, Usami R, Kimura M (2007) Identification of
multiple highly similar XIP-type xylanase inhibitor genes in hexaploid wheat. Biochem
Biophys Res Commun 360(4):880–884
Tanaka H, Okuno T, Moriyama S, Muguruma M, Ohta K (2004) Acidophilic xylanase from
Aureobasidium pullulans: efficient expression and secretion in Pichia pastoris and mutational
analysis. J Biosci Bioeng 98(5):338–343
Taneja K, Gupta S, Kuhad RC (2002) Properties and application of a partially purified alkaline
xylanase from an alkalophilic fungus Aspergillus nidulans KK-99. Bioresour Technol 85
(1):39–42
Teng C, Jia H, Yan Q, Zhou P, Jiang Z (2011) High-level expression of extracellular secretion of a
b-Xylosidase gene from Paecilomyces thermophila in Escherichia coli. Bioresour Technol
102:1822–1830
212 H.A. El Enshasy et al.

Tomme P, Gilkes NR, Miller RC, Warren AJ, Kilburn DG (1994) An internal cellulose-binding
domain mediates adsorption of an engineered bifunctional xylanase/cellulase. Protein Eng 7
(1):117–123
Torronen A, Harkki A, Rouvinen J (1994) 3-Dimensional structure of endo-1,4-beta-xylanase-II
from Trichoderma reesei-2 conformational states in the active-site. EMBO J 13
(11):2493–2501
Torronen A and Rouvinen J (1997) Structural and functional properties of low molecular weight
endo-1,4-beta-xylanases. J Biotechnol 57:137–149
Trudel J, Grenier J, Potvin C, Asselin A (1998) Several thaumatin-like proteins bind to
beta-1,3-glucans. Plant Physiology 118:1431–1438
Tsai CT, Huang CT (2008) Overexpression of the Neocallimastix frontalis xylanase gene in the
methylotrophic yeasts Pichia pastoris and Pichia methanolica. Enzyme Microb Technol 42
(6):459–465
Turunen O, Etuaho K, Fenel F, Vehmaanpera J, Wu XY, Rouvinen J, Leisola M (2001) A
combination of weakly stabilizing mutations with a disulfide bridge in the alpha-helix region of
Trichoderma reesei endo-1,4-beta-xylanase II increases the thermal stability through
synergism. J Biotechnol 88(1):37–46
Valenzuela SV, Diaz P, Javier Pastor FI (2010) Recombinant expression of an alkali stable GH10
xylanase from Paenibacillus barcinonensis. J Agric Food Chem 58(8):4814–4818
Van Campenhout S, Pollet A, Bourgois TM, Rombouts S, Beaugrand J, Gebruers K, De Backer E,
Courtin CM, Delcour JA, Volckaert G (2007) Unprocessed barley aleurone
endo-beta-1,4-xylanase X-I is an active enzyme. Biochem Biophys Res Commun 356
(3):799–804
Van Der Borght A, Goesaert H, Veraverbeke WS, Delcour JA (2005) Fractionation of wheat and
wheat flour into starch and gluten: overview of the main processes and the factors involved.
J Cereal Sci 41:221–237
Van Peij NN, Brinkmann J, Vrsanska M, Visser J, de Graaff LH (1997) Beta-xylosidase activity,
encoded by xlnD, is essential for complete hydrolysis of xylan by Aspergillus niger but not for
induction of the xylanolytic enzyme spectrum. Eur J Biochem 245(1):164–173
Van Petegem F, Collins T, Meuwis MA, Gerday C, Feller G, Van Beeumen J (2003) The structure
of a cold-adapted family 8 xylanase at 1.3 Å resolution. Structural adaptations to cold and
investigation of the active site. J Biol Chem 278(9):7531–7539
Vandeplas S, Dauphin RD, Thonart P, Thewis A, Beckers Y (2010) Effect of the bacterial or
fungal origin of exogenous xylanases supplemented to a wheat-based diet on performance of
broiler chickens and nutrient digestibility of the diet. Can J Anim Sci 90:221–228
Vardakou M, Flint J, Christakopoulos P, Lewis RJ, Gilbert HJ, Murray JW (2005) A family 10
Thermoascus aurantiacus xylanase utilizes arabinose decorations of xylan as significant
substrate specificity determinants. J Mol Biol 352:1060–1067
Verjans P, Dornez E, Delcour JA, Courtin CM (2010) Selectivity for water-unextractable
arabinoxylan and inhibition sensitivity govern the strong bread improving potential of an
acidophilic GH11 Aureobasidium pullulans xylanase. Food Chem 123:331–337
Verma D, Satyanarayana T (2012) Cloning, expression and applicability of thermo-alkali-stable
xylanase of Geobacillus thermoleovorans in generating xylooligosaccharides from
agro-residues. Bioresour Technol 107:333–338
Wang JS, Bai YG, Yang PL, Shi PJ, Luo HY, Meng K, Huang HQ, Yin J, Yao B (2010) A new
xylanase from thermoalkaline Anoxybacillus sp E2 with high activity and stability over a broad
pH range. World J Microbiol Biotechnol 26(5):917–924
Wang Q, Xia T (2008) Enhancement of the activity and alkaline pH stability of Thermobifida fusca
xylanase A by directed evolution. Biotechnol Lett 30(5):937–944
Whitehead TR, Hespell RB (1989) Cloning and expression in Escherichia coli of a xylanase gene
from Bacteroides ruminicola 23. Appl Environ Microbiol 55(4):893–896
Winterhalter C, Liebl W (1995) Two extremely thermostable xylanases of the hyperthermophilic
bacterium Thermotoga maritima MSB8. Appl Environ Microbiol 61:1810–1815
7 Microbial Xylanases: Sources, Types, and Their Applications 213

Wong KK, Tan LU, Saddler JN (1988) Multiplicity of beta-1,4-xylanase in microorganisms:


functions and applications. Microbiol Rev 52(3):305–317
Wu SJ, Liu B, Zhang XB (2006) Characterization of a recombinant thermostable xylanase from
deep-sea thermophilic Geobacillus sp MT-1 in East Pacific. Appl Microbiol Biotechnol 72
(6):1210–1216
Xiong H, Turunen O, Pastinen O, Leisola M, von Weymarn N (2004) Improved xylanase
production by Trichoderma reesei grown on L-arabinose and lactose or D-glucose mixtures.
Appl Microbiol Biotechnol 64:353–358
Xiong H, Weymarn N, Turunen O, Leisola M, Pastinen O (2005) Xylanase production by
Trichoderma reesei Rut C-30 grown on L-arabinose-rich plant hydrolysates. Bioresour
Technol 96:753–759
Xue GP, Gobius KS, Orpin CG (1992) A novel polysaccharide hydrolase cDNA (celD) from
Neocallimastix patriciarum encoding three multi-functional catalytic domains with high
endoglucanase, cellobiohydrolase and xylanase activities. J Gen Microbiol 138(11):2397–2403
Yang HJ, Xie CY (2010) Assessment of fibrolytic activities of 18 commercial enzyme products
and their abilities to degrade the cell wall fraction of corn stalks in in vitro enzymatic and
ruminal batch cultures. Anim Feed Scie Technol 159:110–121
Yang JH, Park JY, Kim SH, Yoo YJ (2008) Shifting pH optimum of Bacillus circulans xylanase
based on molecular modeling. J Biotechnol 133(3):294–300
Yang RC, MacKenzie CR, Narang SA (1988) Nucleotide sequence of a Bacillus circulans
xylanase gene. Nucleic Acids Res 16(14B):7187
Yamura I, Koga T, Matsumoto T, Kato T (1997) Purification and some properties of endo-1,4-[5-
D-xylanase from a fresh water mollusc, Pomacea insularus (de Ordigny). Biosci Biotechol
Biochem 61(4):615–620
Yin LJ, Lin HH, Chiang YI, Jiang ST (2010) Bioproperties and purification of xylanase from
Bacillus sp. YJ6. J Agric Food Chem 58(1):557–562
You C, Yuan H, Huang Q, Lu H (2010) Substrate molecule enhances the thermostability of a
mutant of a family 11 xylanase from Neocallimastix patriciarum. African J Biotechnol
9:1288–1294
Yuan Q, Wang J, Zhang H, Qian Z (2005) Effect of temperature shift on production of xylanase by
Aspergillus niger. Process Biochem 40:3255–3257
Zhao Y, Chany CJ, Sims PF, Sinnott ML (1997) Definition of the substrate specificity of the
‘sensing’ xylanase of Streptomyces cyaneus using xylooligosaccharide and cellooligosaccha-
ride glycosides of 3,4-dinitrophenol. J Biotechnol 57(1–3):181–190
Zhou CY, Bai JY, Deng SS, Wang J, Zhu J, Wu MC, Wang W (2008a) Cloning of a xylanase gene
from Aspergillus usamii and its expression in Escherichia coli. Bioresour Technol 99:831–838
Zhou CY, Li DF, Wu MC, Wang W (2008b) Optimized expression of an acid xylanase from
Aspergillus usamii in Pichia pastoris and its biochemical characterization. World J Microbiol
Biotechnol 24(8):1393–1401
Zhu H, Paradis FW, Krell PJ, Phillips JP, Forsberg CW (1994) Enzymatic specificities and modes
of action of the two catalytic domains of the XynC xylanase from Fibrobacter succinogenes
S85. J Bacteriol 176(13):3885–3894
Chapter 8
Mannanase

Suttipun Keawsompong

Abstract β-Mannanases (endo-1,4-β-D-mannanase) is endohydrolase that catalyze


the random hydrolysis of the β-1,4-D-mannopyranosyl linkage within the main
chain of various mannan-based polysaccharides to yield mannooligosaccharides
products. β-Mannanase have been isolated and characterized from different sources
including bacteria, fungi, higher plants, and animals. However, microbial man-
nanases are wildly used in the industrial application. β-Mannanases was classified
based on the amino acid sequence similarity into glycoside hydrolase (GH) families
5 and 26 and a few member of family 113. These enzymes from different organisms
have different properties such as enzyme activity, optimal pH, and optimal tem-
perature. So, β-mannanases with high specific activity and remarkable enzymatic
properties are required for the application. β-Mannanase is very useful enzyme that
has been used in several industrial applications including food, feed, pulp, and
paper industries. This enzyme can be used to improve the bleaching of pulp by
facilitating the release of lignin from paper pulp leading to the reduction of
chemical reagents. It can be used to reduce the viscosity of instant coffee and to
clarify fruit juices and wines in food industry. β-Mannanase also have been used as
animal feed additive enzyme to increase the nutritional value of animal feed
components. Moreover, there is increasing interest in using β-mannanase to pro-
duce mannooligosaccharides (MOS) which have the prebiotic properties from
natural mannan-based substrates. This enzyme is also used for the pretreatment of
biomass in the bioethanol production.

S. Keawsompong (&)
Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University,
Bangkok, Thailand
e-mail: [email protected]

© Springer International Publishing Switzerland 2016 215


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_8
216 S. Keawsompong

8.1 Mannans

Mannans and heteromannans are widely distributed in nature as part of the hemi-
cellulose fraction in hardwoods and softwoods, seeds of leguminous plants
(Handford et al. 2003; Buckeridge et al. 2000), and in beans (Lundqvist et al. 2003).
Hemicelluloses are copolymers of both hexose and pentose sugars. The branched
structure allows hemicellulose to exist in an amorphous form that is more sus-
ceptible to hydrolysis. Within biomass, mannans or the hemicelluloses are situated
between the lignin and the collection of cellulose fibers underneath. Consistent with
their structure and side group substitutions, mannans seem to be interspersed and
covalently linked with lignins at various points while producing a coat around
underlying cellulose strands via hydrogen bonds, but as few H-bonds are involved
they are much more easily broken down than cellulose. The mannan layer with its
covalent linkage to lignin and its non-covalent interaction with cellulose maybe
important in maintaining the integrity of the cellulose in situ and in helping to
protect the fibers against degradation to cellulases (Puls and Schuseil 1993).
Mannan is the predominant hemicellulosic polysaccharide in softwoods from
gymnosperms, but is the minor hemicellulose in hardwood from angiosperms (Puls
and Schuseil 1993). Unsubstituted β-1,4-mannan, composed of a main chain of
β-mannose residues (Fig. 8.1), is an important structural component of some marine
algae and terrestrial plants such as ivory nut (Chanzy et al. 2004) and coffee bean
(Nunes et al. 2006). It resembles cellulose in the conformation of the individual
polysaccharide chains, and is water insoluble.
The galactomannans are mainly found in the seeds of the family of
Leguminoseae and are located in the endospermic part of the seeds (Dea and
Morrison 1975). Galactomannans consist of water-soluble 1,4-linked β-D-manno-
pyranosyl residues with side chains of single 1,6-linked α-D-galactopyranosyl
groups (Shobha et al. 2005).
Glucomannan is a water-soluble polysaccharide that is considered a dietary fiber.
It is a hemicellulose component in the cell walls of some plant species (Gonzáles
et al. 2004). Glucomannan is a food additive used as an emulsifier and thickener.
The polysaccharide consists of glucose and mannose in a proportion of 1:3 joined
by β-(1,4) linkages (Popa and Spiridon 1998). Hardwoods consist of glucomannan
with a glucose/mannose ratio of 1:1.5–2 (Hongshu et al. 2002).
Galactoglucomannan is a water-soluble hemicellulose, consisting of galactose,
glucose, and mannose. Many softwood species, e.g., Norway spruce are rich of
galactoglucomannans and can contain it up to 10–20 %. Galactoglucomannan
consists of a backbone of randomly distributed (1 → 4)-linked mannose and glu-
cose units with (1 → 6)-linked galactose units attached to mannose units. The
hydroxyl groups in locations C2 and C3 in mannose are partially substituted by
acetyl groups (Willför et al. 2007).
8 Mannanase 217

Fig. 8.1 Structure of a mannan, b galactomannan, c glucomannan, and d galactoglucomannan.


Source: Dhawan and Kaur (2007)

8.2 Mannan-Degrading Enzymes

The mannan-degrading enzymes, a variety of hydrolytic enzymes involves in the


degradation of mannan polysaccharides, are consisted of β-mannanase,
β-mannosidase, β-glucosidase, and other addition enzymes such as acetyl mannan
esterase and α-galactosidase (Moreira and Filho 2008). The overall mechanism of
mannan-degrading enzymes is shown in Fig. 8.2. β-Mannanase, endo acting enzyme,
randomly hydrolyzes the β-1,4-linked internal linkage of the mannan backbone to
produce mannooligosacharides and new chain ends. β-Mannosidase, exo acting
218 S. Keawsompong

Fig. 8.2 Mechanism of mannan-degrading enzymes in the galactoglucomannan degradation.


Source Puls and Schuseil (1993)

enzyme, cleaves β-1,4-linked mannosides and releases mannose from the nonre-
ducing ends of mannan and mannooligosacharides. β-Glucosidases, exo acting
enzyme, hydrolyzes β-1,4-glucopyranose at the nonreducing ends of oligosaccharides
that released from glucomannan and galactoglucomannan degradation. In addition,
the side chain sugars that attached at various points on mannan are removed by
α-galactosidase and acetyl mannan esterase. α-Galactosidase hydrolyzes α-1,6-linked
D-galactopyranose at side chain of galactomannan and galactoglucomannan. Acetyl
mannan esterase releases acetyl groups from galactoglucomannan.
However, mannan degradation by these enzymes is affected by the degree and
pattern of substitution of glucose and glucose residues in backbone of these
polysaccharides (Van Zyl et al. 2010). In addition, pattern of distribution of
O-acetyl group in glucomannan also affects the hydrolysis by the enzymes.

8.2.1 β-Mannanase

β-Mannanase or endo-1,4-β-D-mannanases (EC 3.2.1.78) is the endo hydrolase


enzyme that catalyze the random hydrolysis of internal β-1,4-mannosidic linkages
in backbone chain of mannan polysaccharides, producing various size of man-
nooligosacharides (Puls 1997). β-Mannanase requires at least four sugar residues of
backbone chain for enzyme binding and efficient hydrolysis (Davies et al. 1997).
β-Mannanase hydrolyzes mannooligosaccharides up to a degree of polymerization
(DP) of 4 (Chauhan et al. 2012).
8 Mannanase 219

β-Mannanase is classified into glycoside hydrolases (GH) based on the sequence


similarity. Glycoside hydrolase is a group of enzymes that hydrolyze the glycosidic
bond between molecules in polysaccharides. Based on the amino acid sequence
similarity, glycoside hydrolases are classified into 131 families. This classification
is available on The Carbohydrate-Active Enzymes database (CAZy). Moreover,
each glycoside hydrolase families can be classified into clan based on the catalytic
domains and the protein structure. The different glycoside hydrolase families have
been found to have different folds but, some enzymes from different families have
related folds. Based on these criteria, β-mannanase is classified into glycoside
hydrolase family 5, 26, and a few members in 113 (Dhawan and Kaur 2007; Zhang
et al. 2008). Both of these families are classified into clan A which have a typical
structure of (β/α)8-barrel fold (TIM barrel fold). The crystal structure of
β-mannanases from both of GH family 5 and 26 showed that glutamic acid is
conserved catalytic modules (nucleophiles and acid/base) that are positioned on the
C-terminal of β4 and β7 strands, respectively. In addition, β-mannanase also has an
open cleft with at least four subsites on the active site (Hogg et al. 2003; Zhang
et al. 2008; Chauhan et al. 2012).
Glycosyl hydrolases consist of two hydrolysis mechanisms, retaining and
inverting, leading to either overall retention or inversion of the anomeric configu-
ration at the hydrolysis site (Henrissat et al. 1995). β-Mannanase hydrolyzes the
mannan substrates by retaining mechanism by double displacement reaction
(Fig. 8.3). For the retaining mechanism, two carboxylic acid residues on the active

Fig. 8.3 The retaining mechanism of β-mannanase. Source Chauhan et al. (2012)
220 S. Keawsompong

site of enzyme involve in the hydrolysis as nucleophiles and acid/base catalytic


module. In the double displacement reaction, β-mannanase enzyme attacks by a
nucleophilic carboxylate on the anomeric carbon with concomitant releases of the
aglycon, resulting in a covalent intermediate. Then, the covalent intermediate is
attacked by a nucleophilic water lead to releasing of the glycoside from the enzyme
(Chauhan et al. 2012).
Although, β-mannanases have the similar structure including active site, but
there are difference in the presence and position of catalytic carbohydrate binding
modules (CBMs) that may contain in the structure (Ximenes et al. 2005). CBMs is
use for enhance β-mannanase activity for degrading of cellulose-conjugated man-
nan polysaccharide (Benech et al. 2007). β-Mannanases from GH family 5 contain
CBMs that bind crystalline polysaccharides. For example, β-mannanase from
Cellulomonas fimi, GH family 5, has a mannan-binding module that binds in a
reversible way to soluble mannan, cellulose, chitin, and xylan (Stoll et al. 2000).
Moreover, overexpression of mannanase gene that contains CBMs in Escherichia
coli showed the activity of enzyme for several cellulose substrates (Ximenes et al.
2005). But, β-mannanases from GH family 26 lack of CBMs, so there are no
enzyme activity in crystalline polysaccharides substrate (Hogg et al. 2003). So, this
refers to the different substrate target of β-mannanase in GH family 5 and 26 in
nature.
β-Mannanse enzymes have been isolated and characterized from several different
sources such as bacteria, fungi, plants, and animals. Although the β-mannanases are
widely distributed in microorganisms, higher plants, and animals but the microbial
mannanases are regarded to be useful for their applications. Numerous bacterial and
fungal β-mannanases have been identified and characterized from various strains in
20 different genuses. Among bacteria, most of β-mannanases producing strains are
gram positive bacteria such as Bacillus species and Clostridia species (Dhawan and
Kaur 2007). However, some gram negative bacterial strains have been reported to
produce β-mannanases such as Klebiella sp. (Titapoka et al. 2008) and Vibrio
sp. (Tamamura et al. 1995). The most of β-mannanases producing strains among
fungi are in the genus Aspergillus while Penicillium sp. and Trichoderma sp. have
also been reported (Chauhan et al. 2012). Moreover, some actinomycetes such as
Streptomyces sp. and Cellulomonas sp. have been reported to be β-mannanases
producing strains (Stoll et al. 2000; Bhoria et al. 2009).
Microbial manannanase are extracellular enzyme which could be induced by
mannan polysaccharide (Moreira and Filho 2008; Chauhan et al. 2012). Many
nutritional and physiochemical factors involve and influence in the β-mannanase
production such as carbon and nitrogen sources, inorganic salts, temperature, pH,
time, and dissolved oxygen concentration (Moreira and Filho 2008; Chauhan et al.
2012). There are differences in the incubation time and optimum temperature and
pH for microbial β-mannanase production. For the incubation time, it ranges from
24 to 96 h in bacteria and 3–11 days in fungi. The optimum temperature for
β-mannanase production is in the mesophilic range that corresponds with temper-
ature for microbial growth. For the optimum pH, neutral to alkaline pH is the best
8 Mannanase 221

condition for bacterial growth, and production and acidic pH for fungi (Chauhan
et al. 2012). However, bacterial β-mannanases are more thermostable than fungal
β-mannanases that are important for industrial application, such as β-mannanases
from Bacillus species have the advantages of high activity and convenient isolation
and thus have been used in research and industry (Araujo and Ward 1990).

8.3 Application of β-Mannanase

β-Mannanase has been used in several industrial applications because of its broad
substrate specificity.

8.3.1 Pulp and Paper Industry

β-Mannanase can be used in the enzymatic bleaching of softwood pulps to digest


the mannan component without affecting the cellulose component. The use of
β-mannanase with other enzymes is the alternative method that can equally facil-
itate lignin removal in pulp bleaching and give results comparable to alkaline
pretreatment without the environmental pollution problems (Dhawan and Kaur
2007).

8.3.2 Detergent Industry

Alkaline β-mannanase with stable in detergents has been used in laundry segments
as stain removal boosters. Mannans are generally used as thickening agents in
several products such as hair gel, shampoo, conditioner, and toothpaste. The stains
containing mannan are difficult to remove so this enzyme can cleave into smaller
carbohydrate fragments that can reduce the stain cleaning process and remove
during the washing. Moreover, β-mannanase can be formulated as sanitization
products, contact lens cleanser and hard surface cleansers (Chauhan et al. 2012).

8.3.3 Food Industry

β-Mannanase has been used in the viscosity reduction of coffee bean extracts by
hydrolyzing the mannan component in the coffee extract (Chauhan et al. 2012). It
has been used in the maceration of fruit and vegetable materials and clarification of
222 S. Keawsompong

fruit juices (Moreira and Filho 2008). In addition, β-mannanase can be used in
enzymatic oil extraction of coconut. The enzymatic process can eliminate the
problems of alfatoxin contamination and oxidative rancidity of products (Chauhan
et al. 2012).

8.3.4 Feed Industry

β-Mannanase can be used to improve the nutritional value of animal feed especially
poultry. Mannan polysaccharides are commonly found in feed ingredients such as
soyabean meal, guar meal, copra meal, palm kernel meal, and sesame meal. All
these meals have some common properties such as high fiber content, low
palatability, lack of several essential amino acids, and high viscosity coupled with
several anti-nutritional factors such as mannan, galactomannan, xylan, and arabi-
noxylan that limit the utilization in the animal intestine (Chauhan et al. 2012).
Moreover, they have been found to be highly deleterious to animal performance,
severely compromising weight gain and feed conversion as well as glucose and
water absorption (Dhawan and Kaur 2007). Therefore, incorporation of
β-mannanase in feed can help to cleave mannan and release of nutrients results in
increased villus height in duodenum and jejunum that leads to increase in surface
area and adsorption and decreased intestinal viscosity. So, it can improve both the
weight gain and feed conversion efficiency (Adibmoradi and Mehri 2007). Hemicell
supplied by ChemGen, USA is a fermentation product of B. lentus containing high
amount of β mannanases that degrade mannan in feed (Daskiran et al. 2004).

8.3.5 Mannooligosaccharide Production

Mannooligosaccharide (MOS) is non-oligosaccharides digestibles (NODs) that can


be produced from mannan polysaccharides. MOS is widely used in nutrition as a
natural additive which can improve gastrointestinal health and also overall health
by supporting the microflora in the digestive system.
MOS affects bacterial attachment in the intestinal tract leading to the reduction in
the prevalence and concentration of different strains of Salmonella, as well as
E. coli. Moreover, it effects on promoting beneficial bacteria, such as Lactobacillus
and Bifidobacteria (Spring et al. 2000). MOS helps to limit the pathogenic
microorganisms bacteria by blocking the colonization on the intestinal mucosa lead
so they cannot physically reach or adhere to the intestinal cells and are eliminated
from body.
Large surface area is a key for optimal digestive function of small intestine so the
surface of the small intestine should be covered with long healthy villi. Several
studies of MOS in poultry have looked at the intestinal structure and discovered
8 Mannanase 223

longer villi and a more shallow crypt (Yang et al. 2008; Baurhoo et al. 2009). These
studies showed the results that MOS could increase the energy of digestion and
production of digestive enzymes such as alkaline phosphatase, maltase, and leucine
aminopeptidase. Moreover, MOS could increase the goblet cells, mucus-producing
cells so the villi and intestinal surface could be more protected.

8.3.6 Pharmaceutical Applications

Mannose has been used as a component of medicine because of its properties such
as fast dissolving and structure forming (Chauhan et al. 2012). Mannose also has
been used as a remedy for urinary tract infection (Van Zyl et al. 2010). There is a
significant increase in using this sugar, so β-mannanase and other enzymes can be
used for the economical production of mannose from low-cost mannan substrates
such as palm kernel cake and copra meal. Guar gum has the positive effects on
some physiological functions like reducing plasma cholesterol and body fat without
reducing protein utilization and increased fecal excretion volume (Takeno et al.
1990). Therefore, a partially hydrolyzed guar gum (PHGG) with β-mannanase is
used in beverage form for treatment of several diseases such as irritable bowel
syndrome (IBS). PHGG can increase stool weight and decrease colon transit time
by providing non-digestible bulk, retaining water, and serving as a substrate for
microbial growth in the colon (Parisi et al. 2002). In addition, PHGG supplemented
with oral rehydration solution is also used for the treatment of acute diarrhea in
children by providing short chain fatty acids in large intestine and maintaining the
balance of salt and water (Alam et al. 2000).

8.3.7 Pretreatment of Biomass in the Bioethanol Production

The molecules in wood and soft wood are linked together in complexes known as
cellulose and hemicelluloses and together can represent up to 75 % of dried wood
weight. Moreover, sugar and nutrient is found in wood tissue and soft wood.
Getting access to these complexes is not easy. Surrounding the cellulose and
hemicelluloses is lignin which serves as the glue and protects the polysaccharides
from enzyme and microbial deconstruction. The lignin, cellulose, and hemicellulose
form strong bonds and their combined structure is named lignocellulose. The
process to overcome lignocellulosic recalcitrance and expose the cellulose and
hemicellulose, so that individual sugars can be released, is called pretreatment
shown in Fig. 8.4.
Pretreatment represents the necessary steps to convert the raw substrate into a
suitable form and includes size reduction by grinding, physical, chemical, or
enzymatic hydrolysis to increase substrate availability to release nutrient and sugar.
224 S. Keawsompong

Fig. 8.4 Effect of pretreatment on lignocellulose. Source Mosier et al. 2005

8.3.7.1 Chemical Treatment

Acid Pretreatment

Concentrated acid hydrolysis disrupts the hydrogen bonding of the cellulose chains,
converting the cellulose from a crystalline state to an amorphous state with little
glucose liberation (Kosaric and Vardar-Sukan 2001; Orozco et al. 2007). In 1965,
Oshima showed that crystalline cellulose achieves complete solubility at 72 %
H2SO4 or 42 % HCl at the relatively low temperatures (10–45 °C) (Oshima 1965;
Kosaric and Vardar-Sukan 2001). In the process of concentrated acidic hydrolysis,
dilute acid is used initially in the prehydrolysis digestion column for hemicellulose
removal prior to passing to the secondary counter current column to contact a strong
acidic solution With the concentrated acid method, glucose yields greater than 90 %
have been achieved with a rapid production rates whereas the increased acid
concentration of HCl, a reduction of xylose yield has been showed by the work of
Orozco et al. (2007).

Alkaline Hydrolysis

Alkali is seen as a swelling agent where the alkali acts indirectly with water being
the lysis agent (Kosaric and Vardar-Sukan 2001). By swelling the biomass, the
surface area is increased opening up the structure for water to migrate into the
material. Once inside the biomass, the water disrupts the hydrogen bonding
between the hemicellulose and the lignin carbohydrates (Balat et al. 2008). The
effect is the decrease in crystal and lignin disruption. The advantages of using alkali
over acidic methods are the removal of the lignin fraction without the degradation
8 Mannanase 225

of the other major constituents. The compromise is the increased length of reaction
times [hours or days in comparison to minutes for other processing methods (Balat
et al. 2008)].
Balat et al. (2008) pretreated corn stover with NaOH (2 %) and combined with
irradiation (500 gGy). They found that the glucose yields to increase from 20 % for
NaOH pretreatment to 43 % in combination (Balat et al. 2008). Although NaOH is
the dominant alkali approach, lime [calcium hydroxide, Ca(OH)2] can also be
employed as an alternative method. Ca(OH)2 has been used in the pretreatment of
several feedstocks in pilot scale applications, such as wheat straw (358 K for 3 h
and corn stover (373 K for 13 h). The attraction of lime/alkali pretreatment is the
removal of acetyl and various uronic acid substitutions on hemicellulose that lowers
the accessibility of the enzymes to the hemicellulose and cellulose surface (Sun and
Cheng 2002; Balat et al. 2008).

8.3.7.2 Physical Treatment

Steam Pretreatment

Steam pretreatment, also known as “steam explosion,” has been extensively


investigated and tested in several pilot plants and demo plants worldwide (Galbe
and Zacchi 2012). An additional acid catalyst can be used to increase the effec-
tiveness of the steam pretreatment, in which case hemicellulose recovery and the
enzymatic hydrolysis of the solids both increase (Galbe et al. 2005). The effects of
initial moisture content on steam consumption, mechanism and rate of heat transfer,
pentosan solubilization, and subsequent glucose yield were summarized. Treatment
at 190 °C with complete bleed-down (no explosion), when compared with that at
240 °C with explosion from full pressure, showed at least as good solubilization of
pentosan and enzymatic hydrolysis but showed greater pentosan destruction for the
same degree of pentosan removal. Neither the explosion nor the high temperatures
(above 190 °C) are necessary (Brownell and Saddle 1987).

8.3.7.3 Enzymatic Hydrolysis

During hydrolysis, the polymers of hemicellulose are degraded into their mono-
meric subunits. Complete hemicellulose hydrolysis results in several hexoses and
pentoses. Main sugar in hemicellulose derived from soft wood is mannose
(Taherzadeh and Karimi 2007). The enzyme hydrolysis is catalyzed by hemicel-
lulose enzymes; without any pretreatment the conversion of native hemicellulose to
sugar is extremely slow, since hemicellulose is well protected by the lignin.
Therefore, pretreatment of these materials is necessary to increase the rate of
hydrolysis of hemicellulose (Galbe et al. 2007).
Efficiently releasing all sugars in biomass therefore requires the combined action
of a large number of different enzymes. Furthermore, some of these enzymes could
226 S. Keawsompong

work synergistically and increase overall amount of monosaccharides released.


Cervero et al. (2010) suggested that binary mixtures of commercial enzymes, 1:1
mixture of Mannaway and Gammanase, were used to increase the conversion of
cell-wall material present in biomass to obtain monosaccharides. The binary mix-
tures of commercial enzymes showed good synergistic effect releasing 30 % more
mannose than the sum obtained using these enzymes individually.

8.3.8 Other Applications

β-Mannanase can use as slime control agent in water purification system, waste
water treatment, and cooling water treatment system. It has been used in the
enzymatic hydrolysis of galactomannan to enhance the flow of oil and gas in
drilling operation in the oil and gas industries (Chauhan et al. 2012).

8.4 Conclusions

β-Mannanase (endo-1,4-β-D-mannanase) is an endohydrolase that catalyzes the


random hydrolysis of the β-1,4-D-mannopyranosyl linkage within the main chain of
mannans and various mannan-based polysaccharides to yield mannooligosaccha-
rides products. It can effectively remove mannan (hemicellulose) from biomass
structure in pretreatment, resulting in better digestion of cellulosic materials.
β-mannanases enzyme has been isolated and characterized from several different
sources including bacteria, fungi, higher plants, and animals. It can be widely
applied in industry, especially in the pretreatment of biomass conversion to bio-
sugar for fermentation.

References

Adibmoradi M, Mehri M (2007) Effects of β-mannanase on broiler performance and gut


morphology. In: 16th European symposium on poultry nutrition, Stasburg, France, pp 471–47
Alam NH, Meier R, Schneider H, Sarker SA, Bardhan PK, Mahalanabis D, Fuchs GJ, Gyr N
(2000) Partially hydrolyzed guar gum supplemented oral rehydration solution in the treatment
of acute diarrhea in children. J Pediatr Gastroenterol Nutr 31:503–507
Araujo A, Ward OP (1990) Hemicellulases of Bacillus species: preliminary comparative studies on
production and properties of mannanases and galactanases. J Appl Bacteriol 8:253–261
Balat M, Balat H, Ȍz C (2008) Progress in bioethanol processing. Prog Energy Combust Sci 34
(5):551–573
Baurhoo B, Ferket PR, Zhao X (2009) Effects of diets containing different concentrations of
mannanoligosaccharide or antibiotics on growth performance, intestinal development, cecal
and litter microbial populations, and carcass parameters of broilers. Poult Sci 88
(11):2262–2272
8 Mannanase 227

Benech RO, Li X, Patton D, Powlowski J, Storms R, Bourbonnais R, Paice M, Tsang A (2007)


Recombinant expression, characterization, and pulp prebleaching property of a Phanerochaete
chrysosporium endo-β-1,4-mannanase. Enzyme Microb Technol 41:740–747
Bhoria P, Singh G, Hoondal GS (2009) Optimization of mannanase production from Streptomyces
sp. PG-08-03 in submerged fermentation. Bioresources 4(3):1130–1138
Brownell HH, Saddle JN (1987) Steam pretreatment of lignocellulosic material for enhanced
enzymatic hydrolysis. Biotechnol Bioeng 29(2):228–235
Buckeridge MS, Dietrich SMC, Lima DU (2000) Galactomannans as the reserve carbohydrate of
legume seeds. In: Gupta AK, Kaur N (eds) Developments in crop science, vol 26. Elsevier
Science B.V., Amsterdam, pp283–316
Cerveró JM, Skovgaard PA, Hanne CF, Sørensenc R, Jørgensen H (2010) Enzymatic hydrolysis
and fermentation of palm kernel press cake for production of bioethanol. Enzyme Microbial
Technol 46:177–184
Chanzy H, Dube M, Marchessault RH (2004) Single crystals and oriented crystallization of ivory
nut mannan. Biopolymers 18:887–898
Chauhan PS, Puri N, Sharma P, Gupta N (2012) Mannanases: microbial sources, production,
properties and potential biotechnological applications. Appl Microbiol Biotechnol
93:1817–1830
Daskiran MRG, Teeter DW, Fodge D, Hsiao HY (2004) An evaluation of endo-ß-D-mannanase
(Hemicell) effects on broiler performance and energy use in diets varying in β-mannan content.
Poult Sci 83:662–668
Davies GJKS, Wilson KS, Henrissat B (1997) Nomenclature for sugar binding subsites in glycosyl
hydrolases. Biochemistry 321:557–559
Dea ICM, Morrison A (1975) Chemistry and interactions of seed galactomannans. Adv Carbohydr
Chem Bio-Chem 31:241–242
Dhawan S, Kaur J (2007) Microbial mannanases: an overview of production and applications. Crit
Rev Biotechnol 27:197–216
Galbe M, Zacchi G (2012) Pretreatment: the key to efficient utilization of lignocellulosic materials.
Biomass Bioenergy 70:70–78
Galbe M, Liden G, Zacchi G (2005) Production of ethanol from biomass-research in Sweden. J Sci
Ind Res 64:905–919
Galbe M, Sassner P, Wingren A, Zacchi G (2007) Process engineering economics of bioethanol
production. Adv Biochem Eng Biotechnol 108:303–327
Gonzáles CA, Fernández MN, Sahagún AM, García VJJ, Díez LMJ, Calle PAP, Castro RLJ,
Sierra VM (2004) Glucomannan: properties and therapeutic applications. Nutr Hosp 19
(1):45–50
Handford MG, Baldwin TC, Goubet F, Prime TA, Miles J, Yu X, Dupree P (2003) Localisation
and characterisation of cell wall mannan polysaccharides in Arabidopsis thaliana. Planta
218:27–36
Henrissat B, Callebaut I, Fabrega S, Lehn P, Mornon J, Davie G (1995) Conserved catalytic
machinery and the prediction of a common fold for several families of glycosyl hydrolases.
Proc Natl Acad Sci 92:7090–7094
Hogg D, Pell G, Dupree P, Goubet F, Martin-oure SM, Armand S, Gilbert HJ (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
Hongshu Z, Jinggan Y, Yan Z (2002) The glucomannan from ramie. Carbohydr Polym 47:83–86
Kosaric N, Vardar-Sukan F (2001) Potential source of energy and chemical products. The
biotechnology of ethanol. M. Roehr. Wiley, Weinheim
228 S. Keawsompong

Lundqvist J, Jacobs A, Palm M, Zacchi G, Dahlman O, Stalbrand H (2003) Characterization of


glactoglucomannan extracted from spruce (Picea abies) by heatfractionation at different
conditions. Carbohydr Polym 51:203–211
Moreira LRS, Filho EXF (2008) An overview of mannan structure and mannan-degrading enzyme
systems. Appl Microbiol Biotechnol 79:165–178
Mosier NS, Wyman C, Dale B, Elander R, Lee YY, Holtzapple M, Ladisch M (2005) Features of
promising technologies for pretreatment of lignocellulosic biomass. Bioresour Technol
96:673–686
Nunes MDS, Santos RAM, Ferreira SM, Vieira J, Vieira CP (2006) Variability patterns and
positively selected sites at the gametophytic self-incompatibility pollen SFB gene in a wild
self-incompatible Prunus spinosa (Rosaceae) population. New Phytol 172:577–587
Orozco A, Ahmad M, Rooney D, Walker G (2007) Dilute acid hydrolysis of cellulose and
cellulosic bio-waste using a microwave reactor system. Process Saf Environ Prot 85:446–449
Oshima M (1965) Wood chemistry process engineering apsects. Noyes Development Corporation,
New York
Parisi GC, Zilli M, Miani MP, Carrara M, Bottona E, Verdianelli G, Battaglia G, Desideri S,
Faedo A, Marzolino C, Tonon A, Ermani M, Leandro G (2002) High-fibre diet supplemen-
tation in patients with irritable bowel syndrome (IBS): a multicenter, randomized, open trial
comparison between wheat bran diet and partially hydrolyzed guar gum (PHGG). Dig Dis Sci
47(8):1697–1704
Popa VI, Spiridon J (1998) Hemicelluloses: structure, properties, in polysaccharides. In:
Dumitriu S (ed) Polysaccharides: structural diversity and functional versality. Marcel
Dekker, New York, pp 297–311
Puls J (1997) Chemistry and biochemistry of hemicelluloses: relationship between hemicellulose
structure and enzymes required for hydrolysis. Macromol Symp 120:183–196
Puls J, Schuseil J (1993) Chemistry of hemicellulose: relationship between hemicellulose structure
and enzyme required for hydrolysis. In Coughlan MP, Hazlewood GP (eds) Hemicellulose and
hemicellulases. Portland, London, pp 1–27
Shobha MS, Kumar ABV, Tharanathan RN, Koka R, Gaonkar AK (2005) Modification of guar
galactomannan with the aid of Aspergillus niger pectinase. Carbohydr Polym 62:267–273
Spring PC, Wenk KA, Dawson KA, Newman KE (2000) The effects of dietary mannaoligosac-
charides on cecal parameters and the concentrations of enteric bacteria in the ceca of
salmonella-challenged broiler chicks. Poult Sci 79(2):205–211
Sun Y, Cheng J (2002) Hydrolysis of lignocellulosic materials for ethanol production: a review.
Bioresour Technol 83(1):1–11
Stoll D, Boraston A, Stålbrand H, McLean BW, Kilburn DG, Warren AJ (2000) Mannanase
Man26A from Cellulomonas fimi has a mannan-binding module. FEMS Microbiol Lett
183:265–269
Taherzadeh MJ, Karimi K (2007) Bioethanol review. Bioresources 2(3):472–499
Takeno F, Yamada H, Sekiya K, Fujitani B, Ohtsu K (1990) Effect of partially decomposed guar
gum on high-cholesterol-fed rats and non-dietary fiber-fed rats. J Jpn Soc Nutr Food Sci.
43:421–425
Tamamura Y, Araki T, Amagoi H, Mori H, Morishita T (1995) Purification and characterization of
an extracellular beta-1,4-mannanase from a marine bacterium, Vibrio sp. strain MA-138. Appl
Environ Microbiol 61(12):4454–4458
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
Van Zyl WH, Rose SH, Trollope K, Gorgens JF (2010) Fungal β-mannanases: mannan hydrolysis,
heterologous production and biotechnological applications. Process Biochem 45:203–1213
8 Mannanase 229

Willför S, Sundberg K, Tenkanen M, Holmbom B (2007) Spruce-derived mannans—a potential


raw material for hydrocolloids and novel advanced natural materials. Carbohydr Polym 72
(2):197–210
Ximenes EA, Chen H, Kataeva IA, Cotta MA, Felix CR, Ljungdahl LG, Li XL (2005) A
mannanase, ManA, of the polycentric anaerobic fungus Orpinomyces sp. strain PC-2 has
carbohydrate binding and docking modules. Can J Microbiol 51:559–568
Yang Y, Iji PA, Kocher A, Mikkelsen LL, Choct M (2008) Effects of mannanoligosaccharide and
fructooligosaccharide on the response of broilers to pathogenic Escherichia coli challenge. Br
Poult Sci 49(5):550–559
Zhang Y, Ju J, Peng H, Gao F, Zhou C, Zeng Y, Xue Y, Li Y, Henrissat B, Gao GF, Ma Y (2008)
Biochemical and structural characterization of the intracellular mannanase AaManA of
Alicyclobacillus acidocaldarius reveals a novel glycoside hydrolase family belonging to clan
GH-A. J Biol Chem 283(46):31551–31558
Chapter 9
The Role and Applications of Xyloglucan
Hydrolase in Biomass
Degradation/Bioconversion

M. Saritha, Anju Arora, Jairam Choudhary, Vijaya Rani,


Surender Singh, Anamika Sharma, Shalley Sharma and Lata Nain

Abstract Lignocellulosic biomass is currently the most promising alternative


energy source for realizing sustainable demands of agrarian economies. Its natural
recalcitrance to degradation necessitates a detailed study on the complex bio-
chemistry involved in bioconversion of this lignin–carbohydrate complex.
A comprehension of the enzymology and role of principal and accessory glycosyl
hydrolases involved in biomass degradation are, hence, noteworthy in this context
and the xyloglucan-active hydrolases warrant special mention. These are enzymes
which carry out hydrolysis and transglucosylation of xyloglucan, the major hemi-
cellulosic polysaccharide in plant biomass. The structurally complex xyloglucans
cover and cross-link the cellulosic microfibrils in plant cell walls and make cellu-
lose inaccessible to saccharification by cellulases. Solubilisation of biomass
polysaccharides and release of sugars are central to the biomass-to-bioethanol
process. Complete conversion of biomass carbohydrates requires a suite of
hydrolytic enzymes, which may be designed specifically to accommodate the
predominant and subsidiary biomass-cleaving enzymes. Xyloglucan hydrolases
which are known to act synergistically with cellulases and xylanases in loosening
the plant cell wall are vital enzymes to be deployed for successful bioconversion
processes. This chapter is an insight into the capacity of these accessory, but
indispensable, hydrolytic enzymes in unlocking the inaccessible biomass polysac-
charides for increased sugar recovery and thereby, in drafting the fuels of future.

9.1 Introduction

Biofuel production employs renewable agricultural waste and dedicated energy


crops, which also provide opportunity for alleviation of green house gas release.
Various plants and parts of plants are used for biofuel production such as grains for

M. Saritha  A. Arora  J. Choudhary  V. Rani  S. Singh  A. Sharma  S. Sharma 


L. Nain (&)
Division of Microbiology, Indian Agricultural Research Institute,
New Delhi 110012, India
e-mail: [email protected]

© Springer International Publishing Switzerland 2016 231


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_9
232 M. Saritha et al.

25

Ethanol production (Billion Gallons)


20
Rest of World

15 Canada
China
10 Europe
Brazil
5 USA

0
2007 2008 2009 2010 2011 2012 2013

Fig. 9.1 Country-wise global ethanol production (2007–2013). Adapted from www.afdc.energy.
gov/data/

the first-generation and lignocellulosic biomass for the second-generation of bio-


fuels. The United States is the world’s largest producer of bioethanol, having
produced over 13 billion gallons in 2013 alone (Ethanol Industry Outlook Report
2013). Together, the U.S. and Brazil produce 84 % of the world’s ethanol, which
amounted to 23,429 million gallons in 2013 (Fig. 9.1). Driven by the growth in use
of biofuels and natural gas, nonpetroleum energy makes up the highest percentage
of total fuel consumption for transport since 1954. According to a new report of the
U.S. Energy Information Administration (EIA), 8.5 % of the fuel used by trans-
portation sector in 2014 came from nonpetroleum sources (AEO 2015). Today,
most of the gasoline available throughout the United States is a blend of 90 %
gasoline and up to 10 % ethanol, known as ‘E10’.
The vast majority of the U.S. ethanol is produced from corn, while Brazil
primarily uses sugarcane. The current production of first-generation bioethanol from
sugars or starch has, but raised a worldwide “food vs fuel” debate putting a question
mark on food security. The diversion of food crops for biofuel production also leads
to food price hikes. Advanced generation biofuels are, therefore, imperative as they
are based on inexpensive and plentiful resources which do not compete with food
crops and do not threaten food security.
As a result, the center of attention in development of technology for ethanol
production has shifted toward the use of nonedible residual lignocellulosic materials
as well as to lower the cost of production. Lignocellulosic biomass is the most
abundant source of renewable energy on the planet. The sugars present in the
lignocellulosic biomass can be converted to biofuel molecules such as ethanol and
butanol. Production of biofuels from lignocellulosic biomass requires the efficient
deconstruction of carbohydrate polymers to fermentable sugars by a cocktail of
9 The Role and Applications of Xyloglucan Hydrolase … 233

enzymes containing cellulases, xylanases, and other glycosyl hydrolases. The


economic feasibility of production of bioethanol from lignocellulose and its com-
petition with petrol is, but, a main challenge. The main bottlenecks in this tech-
nology are low efficiency due to the natural recalcitrance of lignocellulose to
deconstruction and high cost of enzymatic conversion (Mohanram et al. 2013). To
address these, significant research has to be directed toward the identification of
efficient enzyme systems and process conditions for the improved enzymatic uti-
lization of biomass feedstocks. This warrants a detailed understanding of the
enzymology involved in biomass bioconversion.

9.2 Polysaccharides of Lignocellulosic Biomass

Lignocellulose is the main structural constituent of woody plants, herbs such as


grasses and crop biomass. It is the most abundant biopolymer on the planet and
comprises about 50 % of world’s biomass (Claassen et al. 1999). About 686 MT of
crop residues are generated every year in India, as predicted by The Ministry of
Agriculture, Govt. of India (Hiloidhari et al. 2014), out of which 234 MT can be
used as substrate for production of many value-added products like biofuel.
Lignocellulose consists of 40–60 % cellulose, 20–40 % hemicellulose, and
10–25 % lignin (Wyman 1996). Lignocellulosic biomass is a substrate of massive
biotechnological value because of the chemical attributes of its components
(Malherbe and Cloete 2003). The average composition of some lignocellulosic
materials that can be used as substrates for biofuel production is given in Table 9.1.
Plant biomass is mainly composed of the polysaccharides, cellulose, and
hemicellulose with pentose and hexose sugars as the main building blocks.
Cellulose is a D-glucose polymer with organized microfibrillar forms, with each
microfibril containing up to 36 glucan chains having thousands of glucose residues
joined by β-1,4-glycosidic bonds. Hemicelluloses are complex homopolysaccha-
rides (glucan, xylan, mannan) or heteropolysaccharides (xyloglucan, galactoglu-
comannans, arabinoxylan, glucomannan) with both hexose (glucose, mannose,
galactose) and pentose sugars (arabinose, xylose) along with their uronic acid
derivatives (glucouronic acid, galatouronic acid, mannuronic acid) (Mohanram
et al. 2013).
Thus, plant cell walls are built from a mixture of polysaccharides, proteins, and
lignin, whereas the noncrystalline matrix of plant cell walls is built of one or more
of the different types of hemicelluloses such as galactoglucomannan, glucomannan,
arabinoglucuronoxylan, glucuronoxylan, and xyloglucan (Timell 1967). Some of
the common hemicelluloses of plant origin are given in Table 9.2.
Among the hemicelluloses, xyloglucans are widely distributed throughout the
plant kingdom as a major structural and storage polysaccharide. The Type I plant
cell wall, predominant in dicotyledonous plants and non-commelinoid mono-
cotyledons, is characterized by equal proportions of cellulose and xyloglucans. On
234 M. Saritha et al.

Table 9.1 Average composition of biomass expressed as percentage on dry weight basis
Biomass Cellulose (%) Hemicellulose (%) Lignin (%)
Barley wood 40 20 15
Bermuda grass 25 35.7 6.4
Birch wood 40 33 21
Corn cobs 42 39 14
Corn stalks 35 15 19
Corn stover 38 26 19
Cotton seed hair 80–95 5–20 0–5
Flax sheaves 35 24 22
Forage sorghum 34 17 16
Grasses 25–40 35–50 10–30
Groundnut shells 38 36 16
Hardwood stem 40–55 34–40 18–25
Leaves 15–20 80–85 0–5
Miscanthus 43 24 19
Oat straw 41 16 11
Paper 85–99 0–5 0–15
Pine 41 10 27
Rice straw 32 24 13
Rice husk 36 15 19
Rye straw 31 25 7
Saw dust 55 14 21
Sorghum straw 33 18 15
Soybean stalks 34 25 20
Sugarcane 33 30 29
Sugarcane bagasse 42 25 20
Sweet sorghum 23 14 11
Switch grass 37 29 19
Salix 41.5 22–25 25
Softwood stem 45–50 25–35 25–35
Spruce 45 26 28
Wheat straw 30 24 18
Adapted from Jørgensen et al. (2007) and Kumar et al. (2009)

the other hand, Type II cell wall, present in commelinoid monocots like rice,
contains less of xyloglucans and shows predominance of glucuronoarabinoxylan
along with cellulose microfibrils (Yokoyama et al. 2004). Whatever the plant type,
the considerable amount of xyloglucan present is always around 20–25 % of dry
weight in dicots and 2–5 % in grasses (Hayashi 1989; Benko et al. 2008).
9 The Role and Applications of Xyloglucan Hydrolase … 235

Table 9.2 Common hemicelluloses of plant origin


Polysaccharide type Biological Amount Type of linkage between monomeric
origin (%) units
Glucuronoxylans Hardwood 15–35 β-D-xylopyranosyl units linked by β-
(1,4) glycosidic bonds
Galactoglucomannans Softwood 20–25 β-D-glucopyranosyl and β-D-
mannopyranosyl units, linked by β-
(1,4) glycosidic bonds
Arabinoglucuronoxylan Agricultural 5–10 β-(1,4)-D-xylopyranose backbone
crops containing 4-O-methyl-α-D-
(nonwoody glucopyranosyluronic acid and α-L-
material) arabinofuranosyl linked by α-(1,2) and
α-(1,3) glycosidic bonds
Xyloglucan Hardwood 2–25 β-1,4-linked D-glucose backbone with
(mainly 75 % of these residues substituted at
dicotyledons) O-6 with D-xylose. L-arabinose and D-
galactose residues can be attached to
the xylose residues forming di-, or
triglycosyl side chains.
Glucomannan Softwood and 2–5 β-(1 → 4)-linked D-mannose and D-
hard woods glucose
Galactoglucomannan Softwood 10–25 β-(1 → 4)-linked mannose and
glucose units with α-(1 → 6)-linked
galactose units attached to mannose
units
Arabinoxylan Cereal grain 0.15–30 β-(1,4)-D-xylopyranose backbone is
cell walls substituted by a-L-arabinofuranosyl
units in the positions 2-O and/or 3-
O and by α-D-glucopyranosyl uronic
unit or its 4-O-methyl derivative in the
position 2-O
Adapted from Girio et al. (2010)

9.2.1 Xyloglucan: A Predominant Cell Wall Hemicellulose

Xyloglucan is one of the most widespread hemicellulosic polysaccharides in a


lignocellulosic biomass. Xyloglucans form a sheath around the cellulose
microfibrils preventing them from direct aggregation and cross-link different cel-
lulose microfibrils via hydrogen bonds and other non-covalent interactions, thereby
providing structural strength to the plant cell wall (Fry 1989; Hayashi 1989;
Baumann 2007a). These polymers consist of a linear backbone of β-1,4-glucan
linkages and are structurally related to cellulose, but are distinguished by having up
to 75 % of β-D-glucopyranose (β-D-Glcp) residues covalently linked to α-D-xylo-
pyranose (α-D-Xylp) at the O-6 position (Carpita and McCann 2000). Some of the
xylosyl residues are further substituted by galactosyl, arabinosyl, or fucosyl resi-
dues, forming a complex structure (Enkhbaatar et al. 2012; Damásio et al. 2012).
Different xyloglucan side chains are depicted using a single-letter nomenclature, as
236 M. Saritha et al.

Fig. 9.2 The typical structures of the subunits in XXXG-type xyloglucans. Adapted from http://
www.ccrc.uga.edu/

proposed by Fry et al. (1993), in order to overcome the complexity in their


structural description. According to it, the unsubstituted and substituted
Glcp residues are represented as G and X, respectively. The substituted
Glcp residue may be further denoted as L/F/U/S, depending upon the substituted
saccharide residue. For example, F represents a Glcp residue that is substituted with
a fucose-containing trisaccharide. Most xyloglucans consist of repeating units of
either XXXG (XXXG-type), or XXGG (XXGG-type) (Vincken et al. 1997). The
primary walls of a wide range of dicots, non-graminaceous monocots, and gym-
nosperms contain fucosylated xyloglucans with an XXXG-type structure, the typ-
ical subunit structures of which are shown in the Fig. 9.2.
The side chains confer several properties on the polymer. The high solubility of
xyloglucan in water and its rheological properties make it a useful food additive,
thickener, stabilizer, and gelling agent (Yamatoya and Shirakawa 2003; Baumann
2007b). Xyloglucan can also replace starch and galactomanan used in papermaking
(Shankaracharya 1998). However, it cannot form ordered crystalline microfibrils
like cellulose (Fry 1989). Pauly et al. (1999) described three structural domains of
xyloglucans: the xyloglucan cross-links that are susceptible to enzymatic cleavage,
a xyloglucan fraction that is tightly bound to the microfibril surface and a third
component that is trapped within the microfibril periphery. This structural com-
plexity suggests the importance of understanding the mechanism of xyloglucan
metabolism and biochemical characterization of the enzymes involved.

9.3 Enzymology of the Biomass Bioconversion Process

Due to high carbohydrate content, lignocellulose biomass like agricultural waste


and plant residues, dedicated energy crops, municipal, and industrial wastes hold
great potential for large scale production of bioethanol (Farrell et al. 2006). The
cellulose and hemicellulose components of biomass can be converted into sugar
monomers from which a range of biomaterials viz. ethanol or other important
chemical intermediates can be synthesized via fermentation or other processes.
The classical model for hydrolysis of cellulose to glucose involves the synergistic
action of endocellulases (EC 3.2.1.4), exocellulases (cellobiohydrolases, CBH, EC
3.2.1.91; glucanohydrolases, EC 3.2.1.74), and β-glucosidases (EC 3.2.1.21).
9 The Role and Applications of Xyloglucan Hydrolase … 237

Endocellulases cleave glucan chains internally in a random fashion, which results in


a faster decrease in length of polymer and a gradual increase in the reducing sugar
concentration. Exocellulases hydrolyze cellulose mainly by removing cellobiose
either from the reducing or non reducing ends, which leads to a rapid release of
reducing sugars but a small change in length of polymer. By cooperative action of
endocellulases and exocellulases on cellulose, cello-oligosaccharides and cellobiose
are produced, which are further hydrolyzed by β-glucosidase to glucose (Mohanram
et al. 2013). Though cellulases are the most important hydrolytic enzymes for sac-
charification of complex polymers, they can only unlock the sugars entrapped in
cellulose leaving behind the unused hemicellulose. Thus, the hemicellulose portion
of biomass is the largest polysaccharide fraction that gets wasted in most lignocel-
lulosic biomass-associated ethanol production processes (Girio et al. 2010). As the
cellulose polymers are entangled with hemicellulose which gives them structural
strength, the accessibility of the cellulose polymers to hydrolytic enzymes is hin-
dered, which thereby interferes with the saccharification process.
Hemicelluloses are cleaved by enzymes like carbohydrate esterases, glycoside
hydrolases, polysaccharide lyases, xyloglucan hydrolases, endohemicellulases, and
others, the concerted action of which hydrolyze ester bonds, glycosidic bonds, and
remove the chain substituents or side chains (van den Brink and de Vries 2011;
Sweeney and Xu 2012). The various hydrolytic enzymes which target the linkages
present in the hemicellulose polymers are detailed in Table 9.3.

Table 9.3 List of hemicellulases that can be supplemented to cellulase enzyme to enhance
saccharification efficiency of lignocellulosic biomass
Enzyme GH family Enzyme Site of action
classification
number (EC)
β-Xylanase GH 10, GH EC 3.2.1.8 Hydrolyses β-1,4 glycosidic bond
11 in xylan
β-Xylosidase GH 52 EC 3.2.1.37 Hydrolyses xylobioses
Glucanase GH 16 EC 3.2.1.- Hydrolyses β-1,3; β-1,6; α-1,4; α-1,6
glucans
Mannanase GH 26 EC 3.2.1.78 Hydrolyses β-1,4-mannosidic linkage in
the main chain of mannan,
galactomannan, glucomannan and
galactoglucomannan
Xyloglucan GH 12, GH EC Hydrolyses xyloglucan (β-1,4 glucan
hydrolase 16 3.2.1.150,151,155 with α-1,6 linked xylose)
Arabinofuranosidase GH 43, GH EC 3.2.1.55 Removes arabinose substituents from
51, GH 54, α-arabinofuranoside, arabinoxylans,
GH 127 arabinogalactans
Glucouronidase GH 67, GH EC 3.2.1.139 Removes α-1,2 linked glucuronoyl or its
79 methyl ester in xylan
Acetyl xylan CE 1, CE 5, EC 3.1.1.72 Removes acetyl group from xylans or
esterase CE 16 other xylooligosaccharides
Feruloyl esterase CE 10 EC 3.1.1.73 Hydrolyses feruloyl esters
Glucuronoyl CE 15 EC 3.1.1.- Demethylates methyl glucuronoyl α-1,2
esterase linked to backbone xylose
Adapted from Sweeney and Xu (2012)
238 M. Saritha et al.

Depending upon the type and abundance of hemicellulose polymer present in the
biomass used as substrate, different combinations of hydrolytic enzymes listed in
Table 9.3 can be supplemented to cellulases. Presence of hemicellulases in
hydrolytic enzyme cocktail has been reported to enhance the saccharification yield
by releasing the sugars entrapped in hemicelluloses and also by increasing the
accessibility of the cellulase enzyme to the cellulose complex (Singh et al. 2014).

9.4 Xyloglucan-Active Enzymes in Modification of Cell


Wall Polysaccharides

The hydrogen bonding present between cellulose and xyloglucan makes the
extraction of latter difficult. In order to access and modify the cellulose fibers
embedded and cross-linked by xyloglucans in the plant cell walls, remodeling of
xyloglucan is required. In nature, the enzymatic hydrolysis of xyloglucan and the
resulting xyloglucan solubilization is important in controlling wall strength during
cell growth (Takahashi et al. 2015). The enzymatic machinery comprising of
specific xyloglucanases that can digest xyloglucans to xyloglucan-oligosaccharides
is, thus, of utmost importance in improving accessibility of cellulose to hydrolytic
enzymes and in enhancing the conversion rate of polymers to sugars in order to
economize the saccharification process. In contrast to the cell wall degrading
enzymes, they cause cell wall modifications that are usually associated with the
weakening of extracellular barriers.
Xyloglucan depolymerization was earlier thought to be due to the action of
endoglucanases and other undefined hydrolases. Later, xyloglucan endotransgly-
colases (XETs) and xyloglucan-specific endo-β-1,4-glucanase were identified from
germinating Nasturtium seeds, where it catalyzed the seed storage xyloglucan
depolymerization (Rose et al. 2003). Subsequently, several proteins belonging to
the same class and with sufficient sequence homology were discovered. Presently,
xyloglucan hydrolases which carry out hydrolysis of xyloglucan are classified in the
xyloglucan endotransglucosylase (XET)/hydrolase (XEH) superfamily, known as
the XTH superfamily (Rose et al. 2003). The modes of action of these enzymes are
illustrated in the Fig. 9.3.
Several xyloglucan-active degrading enzymes of the XTH superfamily have been
reported by various researchers. These include xyloglucan-specific
endo-β-1,4-glucanase (EC 3.2.1.151), xyloglucan-specific exo-β-1,4-glucanase (EC
3.2.1.155), oligoxyloglucan reducing-end-specific cellobiohydrolase (EC 3.2.1.150),
xyloglucan endotransferase (EC 2.4.1.207), xyloglucan 4-glucosyltransferase (EC
2.4.1.168), xyloglucan 6-xylosyltransferase (EC 2.4.2.39), and xyloglucan
2-galactose transferase (EC 2.4.1.-), belonging to several glycosyl hydrolase
(GH) families: GH5, GH12, GH16, GH 26, GH44, and GH74 (Hayashi et al. 1980;
Fry et al. 1992; Pauly et al. 1999; Faik et al. 2002; Madson et al. 2003; Grishutin et al.
9 The Role and Applications of Xyloglucan Hydrolase … 239

Fig. 9.3 Modes of xyloglucan depolymerization by the XTH family of enzymes

Table 9.4 Nomenclature and enzyme characteristics of xyloglucan hydrolase


Particulars Xyloglucan hydrolase
Oligoxyloglucan β-glycosidase Xyloglucosyl transferase
EC number 3.2.1.120 2.4.1.207
Systemic name Oligoxyloglucanxyloglucohydrolase Xyloglucan:
Xyloglucosyltransferase
Reaction type Hydrolysis of O-glycosyl bond Transglycosylation
pH optimaa 4.5–5.0 5.0–6.0
Temp. optimaa 55–60 °C 25–30 °C
Km value Varies according to substrate 1.4–2.2 (xyloglucan)
Tamarind xyloglucan: 0.0024
Xyoglucan oligosaccharide: 0.966
Thermostabilitya Stable below 35 °C Stable below 35 °C
Inhibitors Cu2+,Fe2+,Fe3+, Hg2+, SDS Ammonium sulfate, 4-O-methyl
glucuronoxylan, Ag+, AlCl3,
alginate, ATP, CaCl2, La3+,
NaN3, etc.
a
Varies according to the organism from which enzyme is produced
Adapted from BRENDA enzyme database (http://www.brenda-enzymes.org/)

2004; Yaoi et al. 2004; Baumann 2007b). The particulars of a few enzymes involved
in xyloglucan hydrolysis are given in Table 9.4.
As stated previously, xyloglucan hydrolases are believed to be cell
wall-loosening enzymes that are responsible for cell elongation by continuous
splicing of existing polymers and linking of new xyloglucan residues (Fry et al.
1992; Miedes et al. 2011). High expression of the enzyme in growing parts like
elongating tissues in Cicer arientinum (Romo et al., 2005), hypocotyls in pine
(Lorences 2004), germinating Nasturtium seeds (Stratilova et al. 2010), fiber
elongation in cotton (Michailidis et al. 2009), and even ripening of tomatoes
240 M. Saritha et al.

(Miedes and Lorences 2009) have been reported. Enhanced expression of the
enzyme in plants has also been reported on contact with pathogenic or beneficial
microorganisms. Mycorrhizal association in Medicago truncatula caused enhanced
expression of enzyme in roots both intimately and distantly associated with the
fungal partner (Maldonado-Mendoza et al. 2005) and Botryosphaeria dothidea
caused enhanced expression of the enzyme in apple tree (Bai et al. 2015).
However, due to their role in maintaining the cell wall structure, xyloglucan-
active enzymes may also be considered potential hemicellulose-repairing enzymes.
The XTHs produced during infection of apple fruit by Penicillium expansum has a
dual role of integrating newly secreted xyloglucan chains into an existing
wall-bound xyloglucan and restructuring existing cell wall material by catalyzing
transglucosylation between previously wall-bound xyloglucan molecules
(Muñoz-Bertomeu and Lorences 2014). Their activity gets inhibited as the infection
progresses, leading to hemicellulose degradation.

9.4.1 Mechanism of Action of Xyloglucan Hydrolases

All glycosyl hydrolase enzymes act by causing splits in glycosidic bonds and
presumably, utilize either the inverting or the retaining mechanism. The inverting
mechanism is a direct displacement mechanism, resulting in the inverted configu-
ration on the anomeric carbon. Enzymes employing this reaction mechanism belong
to either GH 44 or GH 74 family and generally exhibit a distance of approximately
11 Å between the catalytic residues (Baumann 2007b). The cellulosomes of
Clostridium thermocellum are found to produce extracellular cellulases, along with
an inverting xyloglucanase CtXGH74A (Martinez-Fleites et al. 2006). Retaining
xyloglucan hydrolases utilize the double displacement mechanism for catalysis. The
cell wall active xyloglucan hydrolases belonging to the GH 16 family employ this
mechanism that involves the formation of a covalent glycosyl-enzyme intermediate
(Baumann 2007b). The only xyloglucan hydrolyzing enzyme from GH16 charac-
terized in detail is NXG1, originally isolated from Nasturtium cotyledons (Edwards
et al. 1985). Recently, close homologs of NXG1 analyzed by expression profiling
have been shown to be expressed during different stages of plant development.
These include the OsXTH19 from rice leaves (Yokoyama et al. 2004) and the
SlXTH6 from tomato shoot, seeds, and roots (Saladie et al. 2006). The GH16
transglycosylases which catalyze endotransglycosylation of xyloglucan that enables
cell expansion also employ the retaining mechanism (Sinnott 1990). The modes of
action of xyloglucanases have also been variously classified into the endo-mode
involving cleavage at multiple sites along the backbone, and the exo-mode
involving reducing end-specific cleavage (Feng et al. 2014).
9 The Role and Applications of Xyloglucan Hydrolase … 241

9.4.2 Role of Xyloglucan-Active Enzymes in Biomass


Saccharification for Biofuel Production

Xyloglucan hydrolase is a ubiquitous enzyme produced by both plants and


microorganisms. Plants, being more complex systems, express a great diversity of
these hydrolytic enzymes, but cannot be exploited for their commercial production.
Microorganisms offer more congenial culturing conditions for greater production in
less space, while allowing for manipulation of culture conditions or gene expression
to a greater extent.
The first described xyloglucanase of fungal origin was the xyloglucan-specific
endo-β-1,4-glucanase of Aspergillus aculeatus (XEG; 34 kDa, pI 3.4, GH12)
(Pauly et al. 1999). Since then, several enzymes involved in the transformation of
xyloglucan have been identified from microbial sources. Table 9.5 gives a list of
various microorganisms known to produce xyloglucan-hydrolysing enzymes.
There have also been several reports on gene cloning of xyloglucanases from
bacteria and fungi. Two GH74 genes from Streptomyces avermitilis (sav_1856 and
sav_2574 genes) have been cloned and expressed using a Streptomyces expression
system (Ichinose et al. 2012). The C-terminal of sav_1856 gene product
(SaGH74A) was found to be similar to CBM2, which showed similarity with the
sequences of CBMs (Carbohydrate Binding Molecules) from Cellulomonas fimi
and Clostridium cellulovorans, while the sav_2574 gene product (SaGH74B), in
contrast showed relatively low similarity with the other GH74 enzymes. Song et al.
(2013) cloned two GH family 12 xyloglucanase genes (designated RmXEG12A and
RmXEG12B) from the thermophilic fungus Rhizomucor miehei and expressed in
Escherichia coli. The xyloglucanases had the ability to hydrolyze tamarind
xyloglucan into oligomers, mainly XXXG, XXLG or XLXG, and XLLG, and
showed no activity toward other linear polymers. In another study, Aspergillus
nidulans A773 (pyrG89) recombinant secretion of a xyloglucan-specific
endo-β-1,4-glucanohydrolase (XegA) cloned from Aspergillus niveus was studied
(Damásio et al. 2012). XegA was found to generate a xyloglucan-oligosaccharides
pattern similar to that observed for cellulases from family GH12, demonstrating that
its mode of action includes hydrolysis of the glycosidic linkages between glucosyl
residues that are not branched with xylose.
Various endoglucanases (non-xyloglucan-specific endo-β-1,4-glucanases) have
been reported to hydrolyze xyloglucan as a substrate analogue (Vlasenko et al.
2010). The Trichoderma reesei endoglucanase (Cel12) hydrolyzes 1,4-β-glucans
such as cellulose, 1,3-1,4-β-glucan and xyloglucan (Yuan et al. 2001). However,
xyloglucan-specific endo-β-1,4-glucanases which display high activity toward
xyloglucan, with little or no activity toward cellulose or cellulose derivatives, have
also been reported (Song et al. 2013). Also, enzymes annotated as 1,4-β-D-glucan
glucohydrolases or β-glucosidases of GH3 family have greater activities toward
xyloglucan oligosaccharide than toward cello-oligosaccharide (Yaoi and Miyazaki
2012). Xyloglucanases from the GH74 family have been frequently reported to
have high specificity toward xyloglucan (Enkhbaatar et al. 2012; Feng et al. 2014).
242

Table 9.5 Characteristics of xyloglucan hydrolases produced by different microorganisms


Microorganism Molecular weight Isoelectric point Optimum Optimum Km for xyloglucan References
(kDa) (pI) temperature pH (mg/ml)
Bacteria
Ruminococcus 78 – 25 5.0 2.79 Warner et al. (2011)
flavefaciens
Bacillus licheniformis 26 – – – – Gloster et al. (2007)
Paenibacillus pabuli 40 – – – – Gloster et al. (2007)
Fungi
Aspergillus japonicus 32 2.8 – 5.0 0.67 Grishutin et al.
(2004)
Aspergillus niger – – – – 0.54 Powlowski et al.
(2009)
Chrysosporium 78 3.8 – 6.0 0.31 Grishutin et al.
lucknowense (2004)
Trichoderma reesei 75–105 4.1–4.3 – 5.3 0.30 Grishutin et al.
(2004)
Geotrichum sp. 80 4.8 55 °C 5.5 – Yaoi and Mitsuishi
(2004)
M. Saritha et al.
9 The Role and Applications of Xyloglucan Hydrolase … 243

These cleave the ‘X–X’ and ‘X–G’ motifs and have been characterized as having
endo-processive modes of action, i.e., the enzymes act by sliding along the back-
bone during the catalytic action (Matsuzawa et al. 2014). These include the
endo-xyloglucanase (XcXGHA) from Xanthomonas citri pv. mangiferaeindicae
(Feng et al. 2014), and SaGH74A and SaGH74B from Streptomyces avermitilis
(Ichinose et al. 2012). In contrast, the OXG-RCBH from Geotrichum sp. M128
recognizes the reducing end of various xyloglucan-derived oligosaccharides and
releases two glycosyl residues from this site in the exo-mode (Ichinose et al. 2012).
A growing interest in xyloglucanases is mainly due to the possibility of their
application in a number of biotechnological processes, such as conversion of plant
waste, modification of xyloglucans for use in food and feed industries as well as
pulp and paper industry, production of novel surfactants from oligoxyloglucans,
and thermally reversible xyloglucan gels for drug delivery (Sinitsyna et al. 2010;
Damásio et al. 2012). During the conversion of biomass to value-added products, a
suitable cocktail of various hydrolytic enzymes is required to release all the sugars
present in the biomass. Enzymatic saccharification has renewed and centralized the
focus on different aspects of hemicellulases as they play an important role in
improving the economics of the overall process. The barriers, which include
development of robust enzyme formulations containing all accessory enzymes in
commercial cellulases, have to be alleviated for successful commercialization of
biofuels. An economically feasible enzyme technology for complete hydrolysis of
biomass polysaccharides into sugar monomers is very important for cost-effective
biofuel production. To achieve this, there is need to search for organisms with
hypercellulase activity for developing better quality cellulase formulations with
superior attributes such as higher efficiencies, thermostability, and lowered feed-
back inhibition and tolerance to inhibition by toxic byproducts from pretreatments,
using sophisticated biotechnological tools (Bon and Ferrara 2007).
Another feasible option is to supplement cellulase preparations with hemicel-
lulases and other accessory enzymes to enhance the sugar recovery from pretreated
lignocellulosic biomass. It has been reported that Trichoderma reesei, a widely
known cellulolytic filamentous fungus and the main industrial source of commer-
cially available cellulases has only one GH74 gene that codes for xyloglucanase,
while several other fungi have more than two genes for the same (Ichinose et al.
2012). In this context, microbial xyloglucan hydrolase preparations may be
important for designing enzyme cocktails for industrial biomass conversion,
because the degradation of xyloglucan could enhance cellulase accessibility to
cellulose (Kaida et al. 2009). There are several reports on improvement in sugar
yields during biomass bioconversion when cellulases were supplemented with
xyloglucan hydrolase. Supplementation of α xylosidase with commercial cellulases
enhanced the release of free glucose (82–88 %) and xylose (55–60 %) from
hydrogen peroxide pretreated corn stover (Jabbour et al. 2013). Xyloglucanase
addition has also been shown to enhance the hydrolysability of various lignocel-
lulosic substrates when added to a cellulase mixture (Nishitani 1995; Benko et al.
2008). Hydrolysis of glycosidic linkage of xyloglucans resulted in swelling of
cellulose microfibrils which enhanced cellulose accessibility and, subsequently,
244 M. Saritha et al.

efficiency of enzymatic hydrolysis (Vincken et al. 1995; Chanliaud et al. 2004).


Hydrolysis of various steam pretreated lignocellulosic substrates was greatly
enhanced by the supplementation of family 10 and 11 endo-xylanases (GH10 EX
and GH11 EX) and family 5 xyloglucanase (GH5 XG) with cellulose monocom-
ponents (Hu 2014). However, the extent of enhancement caused by xyloglucan
hydrolase depends on the relative concentration of hemicellulose in the biomass
(Benko et al. 2008). In other words, the accessibility of polymers to hydrolytic
enzymes is affected by the method of pretreatment adopted in the conversion
process. Higher conversion rate is observed when large amount of hemicellulose
remains in the raw material after pretreatment signifying the need of xyloglucan
hydrolases to loosen the complex network of sugars (Benko et al. 2008).
Supplementations of other hemicellulases like xylanase (Choudhary et al. 2014) and
xylosidase (Jabbour et al. 2013) has also caused significant enhancement in sugar
production, increasing the efficiency of the overall bioconversion process. It is quite
evident from the above reports that supplementation of hemicellulases to enzymatic
cocktails can cause significant enhancement in conversion rate of polymers to
sugars but its role in economy of the overall bioconversion process cannot be
clearly envisaged. It is well known that enzyme production and purification is a
costly affair. It cannot be clearly said whether enhancement in sugar yield would
compensate the cost involved in the production of the hydrolytic enzyme.
Development of transgenic microbes capable of producing the entire hydrolytic
enzyme required for biomass conversion in appropriate ratio is one way to save the
cost incurred in the process (Rani et al. 2014). Hyper-xyloglucan hydrolase pro-
ducing microorganisms may be developed to produce xyloglucan-active enzyme
preparation to design cellulase cocktails of the future. Another recent approach has
been the creation of engineered chimeras by fusing xyloglucan-specific CBMs to
xyloglucanases to assist polysaccharide recognition and binding, increasing the
enzyme concentration on the substrate surface and thereby improving the hydrol-
ysis rate (Guillen et al. 2010). Recently, the efficiency of a xyloglucan-specific
endo-β-1,4-glucanase from Aspergillus niveus (XegA) was enhanced by fusion of
the xyloglucan-specific CBM44 on XegA leading to the creation of an engineered
CBM44-XegA chimera (Furtado et al. 2015). This fusion conferred superior cat-
alytic properties and thermal stability on the XegA. Further detailed enzymatic
studies at the molecular level are required to provide the fundamental understanding
required to engineer these proteins toward new applications.

9.5 Conclusion

Breakdown of lignocellulosic biomass by microorganism is a highly complex


process, requiring multiple types of synergistic catalytic activities concurrently
acting upon a variety of both soluble and insoluble polymeric substrates. Industrial
biomass utilization processes will also require a combination of hydrolytic enzymes
in order to degrade lignocellulosic feedstocks. Structural complexity of
9 The Role and Applications of Xyloglucan Hydrolase … 245

lignocellulosic biomass makes the enzymatic saccharification of polysaccharides


present in biomass more difficult and costlier. As xyloglucan is a major hemicel-
lulosic polysaccharide present in the cell wall of dicotyledonous plants which
provides structural rigidity to cell wall, xyloglucan hydrolases are of special
interest. Commercial cellulases in combination with accessory enzymes like xyla-
nases and xyloglucanases enhance the yields of glucose and xylose from biomass,
and may prove beneficial in industrial biofuel production. This is because cellulases
interact synergistically with endo-xylanases and xyloglucanases to improve sugar
recovery from various pretreated lignocellulosic substrates. The future efforts will
have to focus on detailed enzyme structure–function analysis of the various
xyloglucanases used by microorganisms during plant cell wall degradation to be
used as accessory enzymes for effective saccharification of lignocellulosic biomass.

References

Annual Energy Outlook (AEO) (2015) U.S. Energy Information Administration, U.S. Department
of Energy, Washington, DC. http://www.eia.gov/forecasts/aeo
Bai S, Dong C, Zhu J, Zhang Y, Dai H (2015) Identification of a xyloglucan-specific endo-(1-4)-
beta-D-glucanase inhibitor protein from apple (Malus × domestica Borkh.) as a potential
defense gene against Botryosphaeria dothidea. Plant Sci 231:11–19
Baumann MJ (2007a) Structural evidence for the evolution of xyloglucanase activity from
xyloglucanendo-transglycosylases: biological implications for cell wall metabolism. Plant Cell
19:1947–1963
Baumann MJ (2007b) Xyloglucan-active enzymes: properties, structures and applications.
Doctoral thesis submitted to School of Biotechnology, Royal Institute of Technology,
Stockholm, pp 10–32
Benko Z, Siika-aho M, Viikari L, Reczey K (2008) Evaluation of the role of xyloglucanase in the
enzymatic hydrolysis of lignocellulosic substrates. Enzyme Microb Technol 43:109–114
Bon EPS, Ferrara MA (2007) Bioethanol production via enzymatic hydrolysis of cellulosic
biomass. In: The role of agricultural biotechnologies for production of bioenergy in developing
countries. FAO. http://www.fao.org/biotech/seminaroct2007.html
Carpita NC, McCann M (2000) The cell wall. In: Buchanan B, Gruissem W, Jones RL
(eds) Biochemistry and molecular biology of plants. American Society of Plant Physiologists,
Rockville, pp 52–108
Chanliaud E, de Silva J, Strongitharm B, Jeronimidis G, Gidley M (2004) Mechanical effects of
plant cell wall enzymes on cellulose/xyloglucan composites RID A-7266-2011. Plant J 38:27–
37
Choudhary J, Saritha M, Nain L, Arora A (2014) Enhanced saccharification of steam-pretreated
rice straw by commercial cellulases supplemented with xylanase. J Bioprocess Biotech 4
(7):188–194
Claassen PAM, van Lier JB, Contreras LAM, van Niel EWJ, Sijtsma L, Stams AJM, de Vries SS,
Weusthuis RA (1999) Utilisation of biomass for the supply of energy carriers. Appl Microbiol
Biotechnol 52:741–755
Damásio ARL, Ribeiro LFC, Ribeiro LF, Furtado GP, Segato F, Almeida FBR, Crivellari AC,
Buckeridge MS, Souza TACB, Murakamie MT, Ward RJ, Prade RA, Polizeli MLTM (2012)
Functional characterization and oligomerization of a recombinant xyloglucan-specific
endo-β-1,4-glucanase (GH12) from Aspergillus niveus. Biochim Biophys Acta 1824:461–467
246 M. Saritha et al.

Edwards M, Dea ICM, Bulpin PV, Reid JSG (1985) Xyloglucan (amyloid) mobilization in the
cotyledons of Tropaeolum majus L seeds following germination. Planta 163:133–140
Enkhbaatar B, Temuujin U, Lim J-H, Chi W-J, Chang Y-K, Hong S-K (2012) Identification and
characterization of a xyloglucan-specific family 74 glycosyl hydrolase from Streptomyces
coelicolor A3(2). Appl Environ Microbiol 78(2):607–611
Ethanol Industry Outlook Report-Battling for the Barrel (2013) Renewable Fuels Association.
http://www.ethanolrfa.org
Faik A, Price NJ, Raikhel NV, Keegstra K (2002) An Arabidopsis gene encoding an
α-xylosyltransferase involved in xyloglucan biosynthesis. Proc Natl Acad Sci USA
99:7797–7802
Farrell AE, Plevin RJ, Turner BT, Jones AD, O’Hare M, Kammen DM (2006) Ethanol can
contribute to energy and environmental goals. Science 113:506–508
Feng T, Yan K-P, Mikkelsen MD, Meyer AS, Schols HA, Westereng B, Mikkelsen JD (2014)
Characterisation of a novelendo-xyloglucanase (XcXGHA) from Xanthomonas that accom-
modates a xylosyl-substituted glucose at subsite-1. Appl Microbiol Biotechnol 98:9667–9679
Fry SC (1989) The structure and functions of xyloglucan. J Exp Bot 40:1–11
Fry SC, Smith RC, Renwick KF, Martin DJ, Hodge SK, Matthews KJ (1992)
Xyloglucanendotransglycosylase, a new wall-loosening enzyme activity from plants.
Biochem J 282:821–828
Fry SC, York WS, Albersheim P, Darvill A, Hayashi T, Joseleau JP, Kato Y, Lorences EP,
Maclachlan GA, McNeil M, Mort AJ, Reid JSG, Seitz HU, Selvendran RR, Voragen AGJ,
White AR (1993) An unambiguous nomenclature for xyloglucan-derived oligosaccharides.
Physiol Plant 89:1–3
Furtado GP, Santos CR, Cordeiro RL, Ribeiro LF, de Moraes LAB, Damásio ARL,
Polizeli MLTM, Lourenzoni MR, Murakami MT, Ward RJ (2015) Enhanced
xyloglucan-specific endo-β-1,4-glucanase efficiency in an engineered CBM44-XegA chimera.
Appl Microbiol Biotechnol 99(12):5095–5107
Girio FM, Fonseca C, Carvalheiro F, Duarte LC, Marques S, Bogel-Lukasik R (2010)
Hemicelluloses for fuel ethanol: a review. Bioresour Technol 101:4775–4800
Gloster TM, Ibatullin FM, Macauley K, Eklof JM, Roberts S, Turkenburg JP, Bjornvad K,
Jorgensen PL, Danielsen S, Joha KS, Borchert TV, Wilson KS, Brumer H, Davies GJ (2007)
Characterization and three-dimensional structures of two Distinct bacterial xyloglucanases
from Families GH5 and GH12. J Biol Chem 282(26):19177–19189
Grishutin SG, Gusakov AV, Markov AV, Ustinov BB, Semenova MV, Sinitsyn AP (2004)
Specific xyloglucanases as a new class of polysaccharide-degrading enzymes. Biochim
Biophys Acta 1674:268–281
Guillen D, Sanchez S, Rodriguez-Sanoja R (2010) Carbohydrate-binding domains: multiplicity of
biological roles. Appl Microbiol Biotechnol 85(5):1241–1249
Hayashi T (1989) Xyloglucans in the primary-cell wall. Annu Rev Plant Physiol Plant Molec Biol
40:139–168
Hayashi T, Kato Y, Matsuda K (1980) Biosynthesis of xyloglucan in suspension-cultured soybean
cells. Plant Cell Physiol 21:1405–1418
Hiloidhari M, Das D, Baruah DC (2014) Bioenergy potential from crop residue biomass in India.
Renew Sust Energ Rev 32:504–512
Hu J (2014) The role of accessory enzymes in enhancing the effective hydrolysis of the cellulosic
component of pretreated biomass. Thesis submitted to The University of British Columbia,
Vancouver, pp 98–106
Ichinose H, Araki Y, Michikawa M, Harazono K, Yaoi K, Karita S, Kaneko S (2012)
Characterization of an endo-processive-type xyloglucanase having a β-1,4-glucan-binding
module and an endo-type xyloglucanase from Streptomyces avermitilis. Appl Environ
Microbiol 78(22):7939–7945
Jabbour D, Borrusch MS, Banerjee G, Walton JD (2013) Enhancement of fermentable sugar yields
by α-xylosidase supplementation of commercial cellulases. Biotechnol Biofuels 6:58–66
9 The Role and Applications of Xyloglucan Hydrolase … 247

Jørgensen H, Kristensen JB, Felby C (2007) Enzymatic conversion of lignocellulose into


fermentable sugars: challenges and opportunities. Biofuels Bioprod Bior 1:119–134
Kaida R, Kaku T, Baba M, Oyadomari M, Watanabe T, Nishida K, Kanaya T, Shani Z,
Shoseyov O, Hayashi T (2009) Loosening xyloglucan accelerates the enzymatic degradation of
cellulose in wood. Mol Plant 2(5):904–909
Kumar P, Barrett DM, Delwiche MJ, Stroeve P (2009) Methods for pretreatment of lignocellulosic
biomass for efficient hydrolysis and biofuel production. Ind Eng Chem Res 48(8):3713–3729
Lorences EP (2004) Cell wall xyloglucan incorporation by
xyloglucanendotransglucosylase/hydrolase in pine hypocotyls. Plant Sci 166(5):1269–1274
Madson M, Dunand C, Li XM, Verma R, Vanzin GF, Calplan J, Shoue DA, Carpita NC,
Reiter WD (2003) The MUR3 gene of Arabidopsis encodes a xyloglucan galactosyl transferase
that is evolutionarily related to animal exostosins. Plant Cell 15:1662–1670
Maldonado-Mendoza I, Dewbre GR, Blaylock L, Harrison MJ (2005) Expression of a
xyloglucanendotransglucosylase/hydrolase gene, Mt-XTH1, from Medicagotruncatula is
induced systemically in mycorrhizal roots. Gene 345(2):191–197
Malherbe S, Cloete TE (2003) Lignocellulosic biodegradation: fundamentals and applications: a
review. Rev Environ Sci Biotechnol 1:105–114
Martinez-Fleites C, Guerreiro CIPD, Baumann MJ, Taylor EJ, Prates JAM, Ferreira LMA,
Fontes CMGA, Brumer H, Davies GJ (2006) Crystal structures of Clostridium thermocellum
xyloglucanase, XGH74A, reveal the structural basis for xyloglucan recognition and
degradation. J Biol Chem 281:24922–24933
Matsuzawa T, Saito Y, Yaoi K (2014) Key amino acid residues for the endo-processive activity of
GH74 xyloglucanase. FEBS Lett 588:1731–1738
Michailidis G, Argiriou A, Darzentas N, Tsaftaris A (2009) Analysis of
xyloglucanendotransglycosylase/hydrolase (XTH) genes from allotetraploid (Gossypium
hirsutum) cotton and its diploid progenitors expressed during fiber elongation. J Plant
Physiol 166(4):403–416
Miedes E, Lorences EP (2009) Xyloglucanendotransglucosylase/hydrolases (XTHs) during tomato
fruit growth and ripening. J Plant Physiol 166(5):489–498
Miedes E, Zarra I, Hoson T, Herbers K, Sonnewald U, Lorences EP (2011)
Xyloglucanendotransglucosylase and cell wall extensibility. J Plant Physiol 168(3):196–203
Mohanram S, Amat D, Choudhary J, Arora A, Nain L (2013) Novel perspectives for evolving
enzyme cocktails for lignocellulose hydrolysis in biorefineries. Sustain Chem Process 1:15
Muñoz-Bertomeu J, Lorences EP (2014) Changes in xyloglucan endotransglucosylase/hydrolase
(XTHs) expression and XET activity during apple fruit infection by Penicillium expansum
Link. A. Eur J Plant Pathol 138:273–282
Nishitani K (1995) Endo-xyloglucan transferase, a new class of transferase involved in cell-wall
construction. J Plant Res 108:137–148
Pauly M, Albersheim P, Darvill A, York WS (1999) Molecular domains of the
cellulose/xyloglucan network in the cell walls of higher plants. Plant J 20:629–639
Powlowski J, Mahajan S, Schapira M, Master ER (2009) Substrate recognition and hydrolysis by a
fungal xyloglucan-specific family 12 hydrolase. Carbohydr Res 344(10):1175–1179
Rani V, Saritha M, Tiwari R, Nain L, Arora A (2014) Beta-glucosidase: key enzyme in
determining efficiency of cellulase and biomass hydrolysis. J Bioprocess Biotech 5(1):197–205
Romo S, Jimenez T, Labrador E, Dopico B (2005) The gene for a
xyloglucanendotransglucosylase/hydrolase from Cicer arietinum is strongly expressed in
elongating tissues. Plant Physiol Biochem 43(2):169–176
Rose J, Catala C, Gonzalez-Carrana Z, Roberts J (2003) Cell wall disassembly. In: Rose JK
(ed) The plant cell wall. Annual Plant Reviews 8. CRC Press, Boca Raton, pp 265–324
Saladie M, Rose JKC, Cosgrove DJ, Catala C (2006) Characterization of a new xyloglucan
endotransglucosylase/hydrolase (XTH) from ripening tomato fruit and implications for the
diverse modes of enzymic action. Plant J 47:282–295
Shankaracharya NB (1998) Tamarind-chemistry, technology and uses—a critical appraisal. J Food
Sci Technol 35:193–208
248 M. Saritha et al.

Singh S, Pranaw K, Singh B, Tiwari R, Nain L (2014) Production, optimization and evaluation of
multicomponent holocellulase produced by Streptomyces sp. ssr-198. J Taiwan Inst Chem E
45:2379–2386
Sinitsyna OA, Fedorova EA, Pravilnikov AG, Rozhkova AM, Skomarovsky AA, Matys VYu,
Bubnova TM, Okunev ON, Vinetsky YuP, Sinitsyn AP (2010) Isolation and properties of
xyloglucanases of Penicillium sp. Biochemistry (Moscow) 75(1):41–49
Sinnott ML (1990) Catalytic mechanisms of enzymic glycosyl transfer. Chem Rev 90:1171–1202
Song S, Tang Y, Yang S, Yan Q, Zhou P, Jiang Z (2013) Characterization of two novel family 12
xyloglucanases from the thermophilic Rhizomucor miehei. Appl Microbiol Biotechnol 97
(23):10013–10024
Stratilova E, Ait-Mohand F, Rehulka P, Garajova S, Flodrova D, Rehulkova H, Farkas V (2010)
Xyloglucanendotransglycosylases (XETs) from germinating nasturtium (Tropaeolum majus)
seeds: isolation and characterization of the major form. Plant Physiol Biochem 48(4):207–215
Sweeney MD, Xu F (2012) Biomass converting enzymes as industrial biocatalysts for fuels and
chemicals: recent developments. Catalysts 2:244–263
Takahashi M, Yamamoto R, Sakurai N, Nakano Y, Takeda T (2015) Fungal
hemicellulose-degrading enzymes cause physical property changes concomitant with solubi-
lization of cell wall polysaccharides. Planta 241:359–370
Timell TE (1967) Recent progress in the chemistry of wood hemicelluloses. Wood Sci Technol
1:45–70
van den Brink J, de Vries RP (2011) Fungal enzyme sets for plant polysaccharide degradation.
Appl Microbiol Biotechnol 6:1477–1492
Vincken JP, Dekeizer A, Beldman G, Voragen AGJ (1995) Fractionation of xyloglucan fragments
and their interaction with cellulose. Plant Physiol 108:1579–1585
Vincken J-P, York WS, Beldman C, Voragen AGJ (1997) Two general branching patterns of
xyloglucan, XXXG and XXGC. Plant Physiol 114:9–13
Vlasenko E, Schülein M, Cherry J, Xu F (2010) Substrate specificity of family 5, 6, 7, 9, 12, and
45 endoglucanases. Bioresour Technol 101:2405–2411
Warner CD, Go RM, García-Salinas C, Ford C, Reilly PJ (2011) Kinetic characterization of a
glycoside hydrolase family 44 xyloglucanase/endoglucanase from Ruminococcus flavefaciens
FD-1. Enzyme Microb Technol 48(1):27–32
Wyman CE (1996) Handbook on bioethanol: production and utilization. Taylor Francis,
Washington, p 417
Yamatoya K, Shirakawa M (2003) Xyloglucan: structure, rheological properties, biological
functions and enzymatic modification. Current Trends Polym Sci 8:27–72
Yaoi K, Kondo H, Noro N, Suzuki M, Tsuda S, Mitsuishi Y (2004) Tandem repeat of a
seven-bladed β-propeller domain in oligoxyloglucan reducing-end-specific cellobiohydrolase.
Structure 12:1209–1217
Yaoi K, Mitsuishi Y (2004) Purification, characterization, cDNA cloning, and expression of a
xyloglucan endoglucanase from Geotrichum sp. M128. FEBS Lett 560:45–50
Yaoi K, Miyazaki K (2012) Cloning and Expression of isoprimeverose-producing oligoxyloglucan
hydrolase from actinomycetes species, Oerskovia sp. Y1. J Appl Glycosci 59(2):83–88
Yokoyama R, Rose JKC, Nishitani K (2004) A surprising diversity and abundance of
xyloglucanendotransglucosylase/hydrolases in rice. Classification and expression analysis.
Plant Physiol 134:1088–1099
Yuan S, Wu Y, Cosgrove DJ (2001) A Fungal endoglucanase with plant cell wall extension
activity. Plant Physiol 127(1):324–333
Part III
Lignocellulose Oxidureductases
Chapter 10
Role of Mushroom Mn-Oxidizing
Peroxidases in Biomass Conversion

Mirjana Stajić, Jelena Vukojević, Ivan Milovanović,


Jasmina Ćilerdžić and Aleksandar Knežević

Abstract Intensive agricultural and industrial production followed by increased


production of lignocellulosic wastes, disruption of environment, and depletion of
natural resources are features of the modern society. However, these wastes present
sustainable resources of fibers and energy and can be useful raw materials for many
industries. Therefore, development of the optimal ways for their environmental and
economical friendly biological pretreatment where the main participants will be
fungi, owing to their ability to produce lignocellulolytic enzymes, preoccupies
scientists. Mn-oxidizing peroxidases play crucial role in the process and based on
the substrate specificity, this group is divided into Mn-dependent- and versatile
peroxidases. However, delignification capacity depends on fungal species and
strain, namely on their potential of lignocellulolytic enzyme production and
degradation selectivity, type and composition of lignocellulosic wastes, and fer-
mentation conditions. Species which predominantly degrade lignin and significantly
weaker cellulose could have important role in processes of food, feed, paper, and
biofuel production.

10.1 Introduction

Steady growth of the world population accompanied by increase of agricultural and


industrial production leads to enhancement of types and amount of plant residues,
on the one hand, and to disruption of environment and rapid depletion of natural
energy and fiber sources, on the other hand. Dominant crops are different at the
continents, for example rice is the most common in Asia, maize in America, and
wheat in Europe, and result of that is accumulation of different lignocellulosic
wastes. In 2010, rice residues were the most abundant, with annual production of
104,507 × 104 tons, and wheat and maize ones followed them with production of
98,104 × 104 tons and 84,031 × 104 tons, respectively (Gupta and Verma 2015).

M. Stajić (&)  J. Vukojević  I. Milovanović  J. Ćilerdžić  A. Knežević


Faculty of Biology, University of Belgrade, Takovska 43, Belgrade 11000, Serbia
e-mail: [email protected]

© Springer International Publishing Switzerland 2016 251


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_10
252 M. Stajić et al.

According to Asim et al. (2015), world production of agricultural waste in 2013


was 150 billion tons. The municipal waste also takes significant place in total
annual waste production, approximately 10 %, and it also contains certain amount
of lignocellulosic residues in the form of paper products, food scraps, and garden
waste (Blumenthal 2011). Nowadays, management of the wastes presents real
challenge because major part is improperly deposited and fewer amounts is burned
producing not only CO2 but also other pollutants. However, lignocellulosic biomass
can be useful raw materials for many industries as well as potential source of
renewable energy. Currently, special attention is given to regular disposal and
recycling of the enormous quantity of plant wastes, energy recovering, as well as to
raising awareness of the importance of these processes.
Plant cell wall is lignocellulosic in nature and composed of cellulose (40–50 %)
and hemicellulose (20–30 %) impregnated with lignin (10–25 %) (Kirk and Cullen
1998; Anwar et al. 2014). Cellulose is the most abundant insoluble and resistant
carbohydrate in biosphere with crystalline and amorphous regions. It is composed
of chains built of (1,4)-D-glucopyranose units connected by β-1,4 linkages that have
tendency of forming intra- and intermolecular hydrogen bounds and in such a way
micro-fibrils (Iqbal et al. 2011). This polymer links by non-covalent linkages with
second complex and heterogeneous polymer, i.e., hemicellulose. The heterogeneity
originates from the presence of numerous sugars, among which xylan is the most
frequent in grasses and straws and mannan in hard- and softwood (Anwar et al.
2014).
Lignin is the most recalcitrant natural aromatic compound on Earth, which
provides strength and rigidity to plant cell walls, protection of structural polysac-
charides from hydrolytic enzymes, and barrier against microbial attack. This
heterogeneous polymer is composed of oxygenated phenylpropanoid units (con-
iferyl, sinapyl, and p-coumaryl alcohols), which are bounded by aryl–ether, aryl–
aryl, and carbon–carbon bonds (Guerriero et al. 2015). However, composition,
branching level, type of linkages with carbohydrates, as well as content of lignin
vary from species to species and also depend on plant development stage. Thus,
composition variation depending on plant species was demonstrated by Lapierre
(1993) who showed that ratio among lignin units, p-hydroxyphenyl, guaiacyl, and
syringyl, in spruce was 2:98:0, in birch 0:24:76 and in wheat straw 5:49:46.

10.2 Lignocellulose Degradation

Nowadays, special attention is given to possibility of degradation of lignin from


lignocellulosic biomass and making access to cellulose and hemicellulose that can
be converted into feeds, paper, biofuels, and other valued products. Regarding the
complex structure, degradation of lignocellulose is very demanding process that can
be realized in four ways: physical, chemical, physico-chemical, and biological,
which differ one from another in efficiency and drawbacks (Yang and Wyman
2008; Asgher et al. 2013). Although special equipments and industrial processes are
10 Role of Mushroom Mn-Oxidizing Peroxidases … 253

needed for physical treatments, they are useful because they lead to reduction of
particle size, commonly by grinding, pelleting or heating, and in such a way
improve digestibility of lignocellulosic biomass (Kuijk et al. 2015). Chemical
hydrolysis is based on usage of either acids (hydrochloric or sulphuric acid in high
concentrations) or bases (sodium or ammonium hydroxide) or not selective
chemicals (H2O2), and physico-chemical hydrolysis combine heat, moisture,
pressure, and chemicals (Hendriks and Zeeman 2009). However, numerous studies
have shown that these degradation processes are inefficient, very expensive, and
hazardous for the environment. Therefore preference is given to an alternative
environmental and economical friendly biological delignification, which unique
participants are bacteria and fungi owing to their ability to produce lignocellulolytic
enzymes. The main advantages of the process are energy saving and environmental
protection (Kuijk et al. 2015). Nevertheless, the process has specific demands and
some flaws. Namely, it can be realized only under aerobic conditions and effec-
tiveness depends on lignin structure, i.e. type and ratio of its constituents, branching
level, as well as lignin amount. Grabber (2005) showed that highly branched lignin
is less degradable than mainly linear lignin, Skyba et al. (2013) that syringyl-rich
lignin is more resistant to degradation by fungal enzymes, and Arora and Sharma
(2009) that negative correlation exists between lignin amount and dry matter
digestibility. Loss of dry matter by consumption of cellulose and hemicellulose by
fungi, slowness, and requirement for sterile conditions are the main drawbacks of
the process (Kuijk et al. 2015).
Filamentous fungi, among which Basidiomycetes stand out specially, are good
degraders of lignocellulose. According to lignocellulose decay pattern, Eriksson
et al. (1990) separated three groups of fungi: white rot, brown rot and soft rot, and
their activities depend on environmental conditions, plant species, and cell type.
White rot fungi are common in hardwood forests and include several hundred
species of Basidiomycetes and some Ascomycetes, which breakdown lignin
especially during the early phase of colonization, make access to the cellulose and
hemicellulose, and depolimerize them in fruiting stage (Hammel 1997; Kirk and
Cullen 1998; Sánchez 2009; Kuijk et al. 2015). Ginterová and Lazarová (1987)
showed that ratio among degraded lignin, cellulose, and hemicellulose at the end of
wheat straw colonization by Pleurotus ostreatus was 37.4:20:32.3 % and at the end
of fructification 41.8: 62.1: 60.8 %. Some species, such as Phanerochaete
chrysosporium, degrade lignin, cellulose, and hemicellulose simultaneously and
other species, for example Ceriporiopsis subvermispora, attack lignin first. White
rot fungi are the most efficient lignin mineralizators which have ability to cleave
ether bounds in non-phenolic lignin substructures whose proportion in the woody
biomass is 90 %. Contrary to these fungi, brown-rot species that are mainly present
in coniferous forests only modify lignin and their main activity is cellulose and
hemicellulose depolymerisation, while soft rot species degrade only cellulose under
conditions of high moisture and low lignin content (Sánchez 2009).
According to Leonowicz et al. (1999) and Sánchez (2009), white rot fungi
produce three groups of enzymes: (A) lignocellulolytic enzymes, which include
ligninases (lignin peroxidase, Mn-oxidizing peroxidases, laccase, horseradish
254 M. Stajić et al.

peroxidase, protocatechuate 3,4-dioxygenase, catechol 1,2-dioxygenase and


1,2,4-trihydroxybenzene 1,2-dioxygenase), celullases (endo-1,4-β-glucanase, cel-
lobiohydrolase, β-glucosidase), and hemicellulases (endo-1,4-β-xylanase, xylan
1,4-β-xylosidase, endo-1,4-β-mannanase, β-mannosidase, β-glucosidase,
α-galactosidase); (B) enzymes which cooperate with the first group of enzymes but
never attack wood independently (superoxide dismutase and glyoxal oxidase);
(C) feedback type enzymes that include glucose 1-oxidase, aryl-alcohol oxidase,
pyranose 2-oxidase, cellobiose dehydrogenase, and cellobiose quinone oxidore-
ductase, which are involved both in lignin depolymerization (indirectly) and cel-
lulose degradation.
Due to unique lignin properties, such as molecule size and branching level,
specific bounds and insolubility in water, delignification should be extracellular,
oxidative, less specific, and slow process where non-specific oxidoreductases have
the main role (Hammel 1997). Essential enzymes for delignification beginning are
fungal peroxidases, which produce certain radicals that can penetrate cell wall, and
enzymes producing H2O2 (aryl-alcohol oxidases, glucose 1-oxidase, pyranose
2-oxidase, glyoxal oxidase, and cellobiose quinone oxidoreductase) that is required
for peroxidase activity (Hammel and Cullen 2008). Additionally, in that stage of
lignin mineralization superoxide dismutase also has important role because of its
ability to catalyze conversion of superoxide anion into O2 and H2O2 and in such a
way protects fungal degrader from radicals.
Lignin peroxidases (EC 1.11.1.14) are H2O2 dependent heme-containing
enzymes which oxidize aromatic compounds, such as phenols and anilines that
cannot react directly with heme. These enzymes are strong oxidants because iron
ion from porphyrin ring is electron-deficient, and the presence of tryptophan
enables direct oxidation of non-phenolic substructures contrary to Mn-dependent
peroxidases, which indirectly degrade them by production of free radicals (Hammel
and Cullen 2008). Laccases (EC 1.10.3.2) are polyphenol oxidases that contain one
to four copper ions (Cu2+) and catalyze the one-electron oxidation of a broad
number of substrates (phenols, methoxyphenols, some non-phenolic aromatic
compounds, aromatic amines, some aliphatic alcohols, and lignin) using molecular
oxygen as the electron acceptor. During the process, Cu2+ reduces to Cu+ and
oxygen to water (Thurston 1994; Muñoz et al. 1997a, b; Medeiros et al. 1999).
Three more enzymes are important for lignin degradation process. First of them is
aryl-alcohol oxidases (EC 1.1.3.7), flavoenzymes that oxidize various aromatic
compounds as well as aliphatic polyunsaturated alcohols providing H2O2 required
for activity of lignin- and Mn-oxidizing peroxidases, and generates OH that ini-
tiates attack on lignocellulose (Evans et al. 1991; Gutiérrez et al. 1994; Varela et al.
2001). These enzymes act synergistically with laccases and prevent repolymer-
ization of products generated during degradation of lignin and other aromatic
compounds (Guillén et al. 2000). Aryl-alcohol dehydrogenases (EC 1.1.1.90),
which catalyze conversion of aromatic alcohol to aromatic aldehyde, and quinone
reductases (EC 1.6.5.5) that catalyze obtaining semiquinone from quinone, are also
involved in delignification.
10 Role of Mushroom Mn-Oxidizing Peroxidases … 255

Degradation of hemocellulose requires the presence of more enzymes, known as


hemicellulases, due to more complex structure of the polysaccharide. Among
numerous hemicellulases, xylan 1,4-β-xylosidases and endo-1,4-β-xylanases have
the main role in depolymerization. Cleavage of xylan to oligosaccharides is cat-
alyzed by endo-1,4-β-xylanases (EC 3.2.1.8) and hemicellulose mineralization is
finished by action of 1,4-β-xylosidases (EC 3.2.1.37), which hydrolyze oligosac-
charides to xylose (Sánchez 2009). However, some accessory enzymes (xylan
esterases, ferulic and p-coumaric esterases, α-1-arabinofuranosidases, and α-4-O-
methyl glucuronosidases) are necessary for completing the reaction.
Three cellulases catalyze synchronously hydrolysis of cellulose β-1,4-glycosidic
linkages. First, endo-1,4-β-glucanases (EC 3.2.1.4) randomly attacks numerous
sites inside the molecule. Further, cellobiohydrolases (EC 3.2.1.91), also known as
exoglucanases, hydrolyze removing monomers and dimers from the end of the
chain and act synergistically with the first enzymes. At the end, β-glucosidases (EC
3.2.1.21) cleave bounds inside glucose dimers, sometimes within cellulose
oligosaccharides, releasing glucose (Sánchez 2009).

10.2.1 Mn-Oxidizing Peroxidases

Wood-decaying and soil litter decomposing Basidiomycetes (members of families


Meruliaceae, Coriolaceae, Polyporaceae, Strophariaceae, and Tricholomataceae)
have ability to synthesize Mn-oxidizing peroxidases that belong to class II of
superfamily of plant-fungal-prokaryotic peroxidases, and which divided into the
Mn-dependent peroxidases (EC 1.11.1.13; MnPs) and versatile peroxidases (EC
1.11.1.16; VPs) based on the substrate specificity (Fernández-Fueyo et al. 2014).
MnPs posses a Mn2+ oxidation site that binds the cations, and VPs both Mn2+
oxidation site and site for direct oxidation of low- and high-redox-potential
lignin-related compounds (Perez-Boada et al. 2005). These enzymes are produced
in numerous isoforms characterized by certain structure and properties, especially
by isoelectric points and molecular weights (Martinez et al. 1996; Camarero et al.
1999; Hofrichter 2002; Stajic et al. 2006a, 2010; Lanfermann et al. 2015). Thus,
Ganoderma applanatum, Ceriporiopsis subvermispora, Phanerochaete
chrysosporium, and Phlebia chrysocreas synthetise 2, 4, 5, and 7 MnP isoforms,
respectively, which are encoded by separate genes (Martinez et al. 2004; Salame
et al. 2012; Lanfermann et al. 2015). Pleurotus ostreatus besides 6 MnPs also
produce 3 VPs, which structures, stability, and catalytic properties were charac-
terized by Fernández-Fueyo et al. (2014). However, expression of the genes
depends on cultivation conditions, substrate composition, and nutrient availability,
as well as concentration of Mn2+ ions (Urzúa et al. 1995; Ruiz-Dueñas et al. 1999;
Stajić et al. 2006a, b; 2009; Salame et al. 2012). Thus, Urzúa et al. (1995) obtained
7 isoforms of C. subvermispora MnP by cultivation in salt medium and only 4
isoenzymes on wood chips. In the case of Ph. chrysosporium, Datta et al. (1991)
noted expression of mnp5 gene during its cultivation on wood pulp, mnp4 gene was
256 M. Stajić et al.

transcribed in growth on wood-containing soil (Stuardo et al. 2004), while nitrogen


poor medium but enriched with Mn2+ expressed mnp1 and mnp2 genes (Hammel
and Cullen 2008). Letter, Salame et al. (2012) showed that Mn2+-deficient medium
caused increase of expression of mnp3 and mnp9 P. ostreatus genes and in such a
way activity of the MnP izoforms raised in 200-fold, while expression of mnp4
gene, responsible for VP synthesis, was down-regulated in the medium enriched
with this ions. Repression of VP activities in the Mn2+ presence, though the
enzymes have ability to oxidize the ions, Salame et al. (2012) explained by the fact
that VPs are expressed during the later stages of lignin degradation when majority
of Mn2+ presented in the biomass are exhausted. However, Urzúa et al. (1995)
showed that requirement for Mn2+ also depends on nature of the aromatic substrate.
The main environmental factors which affect Mn-oxidizing peroxidase activities
are pH and temperature. According to Fernández-Fueyo et al. (2014), the most
appropriate pH for wood lignin degradation by P. ostreatus is 3.0 due to numerous
lysine and other alkaline residues that increase positive charge and stability at acidic
pH values. However, pH values below 3.0 as well as pH 9.0 inactivate all perox-
idases in very short time. The peroxidase instability in alkali environment the
authors explained by loss of the Ca2+ ions. P. ostreatus VPs are more stable on high
temperature than MnPs, and some of these enzymes can retain about 80 % of
activity after incubation at 60 °C for 10 min.

10.2.1.1 Structure

Mn-oxidizing peroxidases are H2O2-dependent heme glycoproteins with an iron


protoporphyrin IX prosthetic group in the active site, which generate Mn3+ that
initiates delignification (Wariishi et al. 1988). Heme iron is Fe3+ located at an
internal cavity and connects with the protein through two histidine residues which
situate proximally and distally to the iron ion. The central cavity is delimited with
two approximately equal domains built of 10–12 predominantly α-helices com-
posed of conserved amino acid residues and located proximally and distally to the
heme (Martinez 2002; Fernández-Fueyo et al. 2014). Heme pocket bounds with two
Ca2+ ions (one above and one below the heme) which link with oxygen of amino
acid residues and form Ca2+ binding sites that stabilize the protein structure. Amino
acid residues in distal Ca2+ binding sites are conserved in all fungal Mn-oxidizing
peroxidases, while proximal site differs among fungal species by two residues. Four
or five disulfide bridges, composed of cysteine residues, also stabilize the protein
structure (Martinez 2002). Heme connects with enzyme surface by two channels.
One of them is located in front of heme propionate and delimited by three acidic
residues which form Mn2+ binding site. When Mn2+ enters in the channel it oxi-
dizes to Mn3+ which is released from the active site in the presence of chelators that
are extracellular metabolites of white rot fungi. Second, narrow, channel is the place
where H2O2 reacts with heme Fe3+ and has important role in protection of the
enzyme from substrate radicals which could inactivate it (Ruiz-Dueñas et al. 2009).
10 Role of Mushroom Mn-Oxidizing Peroxidases … 257

Mn-oxidizing peroxidases posses multiple substrate sites at both sides of the heme
pocket and could oxidize different substrates.
However, Fernández-Fueyo et al. (2014) demonstrated significant differences
between Pleurotus ostreatus MnPs and VPs in amino acid sequence length, amino
acid composition and distribution, as well as in substrate specificity and catalytic
efficiency on Mn2+. Thus, MnPs contains sequences composed of 331 and VPs of
339 residues mainly located in C-terminal region. In MnPs number of proline
residues varies from 25 to 26 and in VPs from 30 to 31, while number of lysine
residues is ranged from 7 to 10 in all isoforms except MnP4 where the number is
20. In the case of substrate and ability of Mn2+ oxidation, VPs have wider speci-
ficity and higher efficiency than MnPs. This could be explained by the fact that VPs,
besides Mn2+ oxidation site composing of three conserved acidic residues near the
internal heme propionate, also posses lignin oxidation site near tryptophan that is
substituted by an alanine or aspartic acid in MnPs. Exceptions are P. ostreatus
MnP1 and P. pulmonarius peroxidase which posses tryptophan but not activity
characterized for VPs, that Fernández-Fueyo et al. (2014) explained by change of
shape and charge of surface around tryptophan and in such a way substrate binding.
Analyzing the structures of P. ostreatus MnP4 and VP1 these authors noted a few
differences: (i) in domain close to the Ca2+ ion; (ii) in the main heme access
channel, which is wider in VP1 than in MnP4 due to the presence of three substrate
oxidation sites; (iii) in shape and charge of lower channel lip, as wide reservoir with
negative charge in VP1 and as narrow groove of less charged surface in MnP4;
(iv) in number of lysine residues, MnP4 has the minimum one unit more than VP1,
which influences isoelectic points, and (v) in number of hydrogen bonds and salt
bridges that are more numerous in MnP4 than in VP1.

10.2.1.2 Catalytic Properties

Mn-dependent peroxidases play a crucial role in the process of delignification and


characterized by classical peroxidase cycle and inability to oxidize directly
non-phenolic lignin-related structures (Hammel and Cullen 2008). H2O2 or organic
peroxide donate one electron to MnP heme producing Compound I (Fe4+-
oxo-porphyrin-radical complex) and one water molecule is expelled (Fig. 10.1).
Compound I is reduced to Compound II (Fe4+-oxo-porphyrin complex) by per-
oxidase substrate, phenols or Mn2+. However, efficient completion of MnP catalytic
cycle, i.e. reduction of Compound II to the native enzyme requires the presence of
only Mn2+ that is commonly presented in lignocellulose and oxidizes to Mn3+ with
release of the second water molecule (Martinez 2002). Mn3+ is small quite unstable
oxidizer, which is stabilized by organic acids such as oxalate, malonate, tartrate,
and lactate, and can diffuse into the plant cell wall and catalyze cleavage of bounds
in phenolic lignin substructures producing a phenoxyl radical intermediate
(Fig. 10.1). Chelates of Mn3+ with the acids can also oxidize other phenolic, var-
ious non-phenolic aromatic, amino-aromatic, thiol compounds, and unsaturated
fatty acids to phenoxyl, aryl, amino, thiyl, and peroxyl radicals, respectively. These
258 M. Stajić et al.

Fig. 10.1 Mechanism of action of Mn-oxidizing peroxidases

radicals can cleave Cα–Cβ and β-aryl ether bonds in abundant non-phenolic lignin
substructures. Contrary to fatty acids, which are normally presented in the fungal
environment, thiols are not produced by fungi and SH-groups are released in small
amount by protein degradation during cell lyses (Hofrichter 2002). Likewise, Mn3+
can oxidize the oxalate from chelate generating CO2 and anion radical, which in the
presence of O2 forms another CO2 molecule and superoxide radical (O– 2 ) that at low
wood pH values transforms into perhydroxyl radical (HOO) leading to lipid per-
oxidation and releasing of ligninolytic peroxyl radicals (Fig. 10.1). However, more
efficient mechanism of mineralization of non-phenolic lignin moieties means their
modification by cellobiose dehydrogenase to structures which are accessible to
MnPs. Chelates of Mn3+ with the acids can also react with each other and convert to
alkyl radicals which then react spontaneously with oxygen and form some other
radicals, for example superoxide, that could be used by native enzyme in the
absence of H2O2 (Hofrichter 2002). This author also emphasized that high con-
centrations of H2O2 or O2 lead to formation of Compound III, catalytically inactive
form, which cause reversible inactivation of MnPs.
Martinez et al. (1996) first purified and characterized two VPs after submerged
cultivation of Pleurotus eryngii in glucose/peptone/yeast extract medium. Later
Martinez (2002) showed that these enzymes can oxidize both Mn2+ and aromatic
compounds, and that present hybrids of MnPs and lignin peroxidases. These
enzymes occur naturally and until now it is shown that various species of the genera
Pleurotus and Bjerkandera, Lentinus edodes, Panus tigrinus, etc. can synthesize
them (Hofrichter 2002; Hammel and Cullen 2008).
10 Role of Mushroom Mn-Oxidizing Peroxidases … 259

10.3 Plant Residue Conversion

At the beginning of twenty-first century, humankind faces with high rate of world
population growth and in such a way with food insufficient, environmental pollu-
tion, and climate change. It should be emphasized that 70 % of agricultural prod-
ucts are not utilized and become ballast and potential pollutant of environment.
Numerous fungi have essential role in conversion of this enormous amount of
lignocellulosic waste in food, feed, paper pulp, biofuels, and various chemicals.
However, all of the conversions require delignification without or with minimal loss
of carbohydrates, which could be achieved by selection of the most efficient fungal
species and strains, the optimal lignocellulosic biomass, as well as by optimization
of cultivation conditions (Kuijk et al. 2015). Nutritional value and availability are
the main criteria for selection of lignocellulosic residues. However, composition of
plant waste of the same origin and consequently its degradation can vary because of
existence of numerous cultivars of the same crop, for example number of wheat
cultivars is still grown. It was demonstrated by Arora and Sharma (2009) who noted
that Phlebia floridensis more efficiently delignificated wheat straw from north-
eastern India than straw from other climatic regions. As efficiency of fungal
enzymatic system depends on substrate type, selection of degrader is conditioned
by lignocellulosic residue. Thus, Ceriporiopsis subvermispora, Pleurotus eryngii,
P. ostreatus, Lentinus edodes, Hericium clathroides, and Trametes versicolor are
excellent selective degraders, namely they mineralize more than 20 % of lignin and
less than 5 % of cellulose in wheat straw (Tuyen et al. 2012; Knežević et al. 2013a,
b; Stajić et al. 2013; Kuijk et al. 2015). Numerous studies confirmed that C.
subvermispora and P. eryngii are the most effective mineralizers of wheat straw
lignin and showed that Mn-oxidizing peroxidases have the essential role in the
process of delignification (Fernandez-Fueyo et al. 2012; Tuyen et al. 2012).
However, Asiegbu et al. (1996) and Chi et al. (2007) demonstrated that combi-
nation of few fungal species can be more effective in depolymerization of lignin in
some plant wastes than their monocultures. Thus, extent of spruce sawdust delig-
nification by co-culture of Pleurotus sajor-caju, T. versicolor, and Phanerochaete
chrysosporium was significantly higher than by each monoculture separately (16
and 0–5 %, respectively), and co-culture of C. subvermispora and P. ostreatus
caused higher loss of lignin from aspen wood than in its colonization with the
monocultures. However, due to divergence in genotype, significant differences in
lignocellulolytic enzyme production and selectivity of lignocellulose degradation
exist among strains of the same mushroom species (Simonić et al. 2010; Knežević
et al. 2013b). For example, percents of degraded wheat straw lignin and cellulose
by some P. ostreatus strains were almost the same (Salvachua et al. 2011), while
other strains predominantly depolymerized lignin (Adamović et al. 1998). Solid
state fermentation, small lignocellulosic particles of suitable shape, high oxygen
level, moderate water content, certain inoculum type (spores or spawn) in moderate
amount, and the presence of copper, manganese, linoleic acid, and veratryl alcohol
260 M. Stajić et al.

are good conditions for biological treatment of lignocellulose by white rot fungi
(Kuijk et al. 2015).

10.3.1 Food

Ability of mushrooms to produce lignocellulolytic enzymes has double benefit,


degradation of plant residues, and production of high valued food. Therefore,
numerous species are cultivated commercially and cheap and quality food, rich in
proteins, fibers, vitamins, minerals as well as biologically active compounds, is
obtained. Their consumption could compensate insufficient proteins in regions
where famine presents serious problem and enhance immune system and resistance
to diseases.
According to enzymatic system, mushrooms are divided into two groups, pri-
mary degraders-species with strong system and high degradation ability, and sec-
ondary degraders-species with poor enzymatic system that need help of other
degraders. For example, species of the genera Pleurotus, Lentinus, Grifola,
Auricularia, Flammulina, etc. belong to the first group, while Agaricus species
make the second group.
Frequently cultivation in the plastic bags enables usage of various lignocellulosic
wastes from agriculture, forestry, and food industries for compost preparation. If it
bears in mind that production of cereal residues in Europe is high (55,585 × 104
tons in 2010) and if wastes from sugar crops and tuber vegetables are added to the
value, it comes to 61,484 × 104 tons and it can be concluded that amount of
potential substrates for mushroom production is enormous (Gupta and Verma
2015). Nowadays, mushroom producers could obtain high biological efficiency,
from 60 to 75 %, depending on species. Additionally, considering that only third of
world cereal straw production is used in the processes, it can be estimated potential
of mushroom production.
Nowadays, three species of the genus Agaricus (A. bisporus, A. blazei, and A.
bitorquis) are produced in the highest amounts and the first of them is leader
(Chang and Miles 2004). Based on annual world production, Lentinus edodes takes
the second place and numerous members of the genus Pleurotus (P. ostreatus,
P. ostreatus var. florida, P. eryngii, P. eryngii var. nebrodensis, P. sajor-caju,
P. cystidiosus, P. citrinopileatus, P. djamor, P. ferulae, P. tuber-regium, and
P. sapidus) take third place. Efficiently usage of wide range of plant residues (straw,
sawdust, maize, sugar cane pulp, wastes from cotton production, etc.), without any
treatment and enrichment, by Pleurotus species (they can convert 100 g of dry
lignocellulosic wastes in 50–70 g of fresh fruiting bodies in only few weeks) is the
result of possession of strong ligninolytic enzymes (especially Mn-oxidizing per-
oxidases) characterized by well selectivity (Ginterová and Lazarová 1987).
However, biological efficiency depends on substrate composition and strain. Thus,
depending on wheat cultivar, content of reducing sugar differs considerably,
between 13.1 and 40.7 mg/g and yield of various P. ostreatus strains can vary
10 Role of Mushroom Mn-Oxidizing Peroxidases … 261

between 123 and 262 kg of fruiting bodies per ton of wheat straw (Kuijk et al.
2015).
Nowadays, number of cultivated mushroom species is relatively small, 100
species are cultivated economically, 60 commercially, and only 10 in industrial
scale, but current trend is growth of world production in favor of new species, such
as Agaricus blazei, Agrocybe aegerita, Armillaria mellea, Auricularia fuscosuc-
cinea, Coprinus comatus, Dictyophora duplicata, Lepista nuda, Stropharia
rugosaannulata, Tremella cinnabarina, T. aurantialba, and Tricholoma giganteum
(Chang and Miles 2004).

10.3.2 Feed

Most agricultural and industrial wastes that are used for animal feeding are rich in
low digestible fibers and poor in nutrients, especially in proteins and vitamins.
Besides high nutritionally valued food, result of mushroom cultivation is also
production of enormous amount of spent substrate that can be an important feed
ingredient. Mushroom mycelium is rich in protein, essential amino acids, and chitin
and could be important nitrogen sources, while β-glucans and other extra- and
intracellular polysaccharides could be an additional glucose sources and
immunostimulators for ruminants (Reis et al. 2012; Cheung 2013). Thus, protein
content in wheat straw after P. ostreatus cultivation can be increased by 89 % that
leads to overcoming of the main limitation of straw feed, i.e. nitrogen lack (Kuijk
et al. 2015). The authors also emphasized that the protein increases rumen
microorganism population and in such a way cellulose and hemicellulose
digestibility and energy releasing. Spend mushroom compost is also rich in vita-
mins, minerals, antimicrobial agents, probiotics, and other biologically active
compounds that modify animal metabolism, enhance growth, improve immune
system, and increase resistance to various diseases (Enshasy and Hatti-Kaul 2013).
Mushroom contribution to feed production is also transformation of natural
recalcitrant lignin into digestible compounds owing to well developed lignocellu-
lolytic enzyme system, especially Mn-oxidizing peroxidases. Although ruminants
posses cellulolytic microorganisms in rumen that can depolymerize cellulose and
hemicellulose from plant cell wall, connection of these carbohydrates for lignin by
covalent or other linkages is negatively correlated with their digestibility under
rumen anaerobic conditions. Rodrigues et al. (2008) showed that cultivation of
various white rot mushrooms on wheat straw could increase degradability of neutral
detergent fibers in rumen in even 13 % due to their strong ligninolytic enzymes
which act to lignin. Contrary to wheat straw that was not biologically treated, spent
wheat straw composts of Pleurotus ostreatus and P. sajor-caju were well consumed
and digested by sheep (Calzada et al. 1987; Fazaeli et al. 2006). Similar efficient
consumption and decomposition of crude protein, hemicellulose, and cellulose from
spend Coprinus fimetarius rice straw compost and spent Ganoderma sp. wheat
straw compost by goats were reported by Rai et al. (1989) and Shrivastava et al.
262 M. Stajić et al.

(2012). However, cows are more selective and their feed can be enriched with
maximum 17 % of spend P. ostreatus wheat straw substrate (Adamović et al.
1998).
Kuijk et al. (2015) reviewed results of numerous studies and also reported that
addition of selected microelements and inducers (copper, manganese, linoleic acid,
di-rhamnolipid, and veratryl alcohol) to lignocellulosic residues enhance activity of
fungal Mn-oxidizing peroxidases and indirectly fiber digestibility by animals.
However, some mushroom metabolites could be toxic for animals and humans and
special attention has to be given to that aspect of plant waste bioconversion by
fungi.

10.3.3 Paper Pulp

Since nowadays forest preservation is compulsory, plant residues that are produced
in enormous amount, but not used, can be important natural sources for paper-
making. Good sample for extraordinary production of some agricultural wastes was
given by González Alriols et al. (2009). Namely, they noted huge quantity of
unutilized Elaeis guineensis residues after oil extraction in Malaysia, even 15
million tons per year, which present serious ballast. In a few last decades, white rot
fungi or their enzymes have the main role in the process, known as biopulping. The
main reasons for that are its healthy, environmental, and economical friendly
relationships contrary to traditional usage of alkaline sulfide, as well as improve-
ment of pulp quality and quantity (Bajpai et al. 2001). Yang et al. (2008) showed
that pretreatment of eucalyptus woody biomass with Trametes hirsuta led to
increasing fiber internal bond strength in 32 %. Fungal ligninolytic enzymes, pri-
marily peroxidases, catalyze one-electron oxidation of lignin phenolic groups
producing phenoxy radicals on fiber surfaces and in such a way enhancing inter
fiber adhesion (Singh and Singh 2014). Thus, this modified lignin could replace
traditionally harmful adhesives that is current trend and one more possible appli-
cation of fungal enzymatic system.
Besides enhancement of the bond strength and fiber adhesion, wood pretreat-
ment with white rot fungi removes pitches rich in lipophylic compounds (resin and
fatty acids and triglycerides), which can reduce paper and effluent quality and cause
technical problems (Martinez-Inigo et al. 2000; Dorado et al. 2001; Gutiérrez et al.
2006; Van Beek et al. 2007). Martinez-Inigo et al. (2000) reported that Phlebia
radiate and Poria subvermispora were the most active in the removal of lipophylic
extractives and in such a way pitch from eucalypt wood, and Dorado et al. (2001)
that Bjerkandera sp. and Trametes versicolor eliminated up to 90 % of these
compounds from Pinus sylvestris wooden biomass. Letter Van Beek et al. (2007)
showed that T. versicolor was also effective in removal of resin acids and
triglycerides from spruce wood chips, which content was reduced in 40 and 100 %,
respectively, and that Pycnoporus cinnabarinus caused losses of sterols, resin acids,
and triglycerides from various pulp systems in 65–100 %. Further study showed
10 Role of Mushroom Mn-Oxidizing Peroxidases … 263

that this ability of white rot fungi is based on production of lipase, resinase,
lipoxigenases, and laccases (Gutiérrez et al. 2009). As pitch control is combined
with lignin removal from the pulp, white rot fungi play important role in pulp
bleaching (bio-bleaching) and thus in improving paper brightness, environment
protection, and energy saving (Bajpai et al. 2001; Jerusik 2010). However, the
process should be controlled in order to avoid cellulose degradation.
Besides papermaking, white rot fungi and their enzymes have one more role,
bioremediation of waste water. Namely, huge amount of water, approximately 45–
227 × 103 litters, is used for production of a ton of product resulting in the
releasing of the same quantity of effluent and further in zooplankton and fish dying,
slime and scum formation, esthetic problems, and disruption of the environment
(Pokhrel and Viraraghavan 2004). Fungi are characterized by better survival in
effluents and production of strong extracellular ligninolytic enzymes, especially
lignin- and Mn-oxidizing peroxidases that degrade various phenolic compounds
among which colors are the most resistant. Thus, numerous studies showed high
effectiveness of Trametes pubescens, Phanerochaete chrysosporium, and Pleurotus
ostreatus in the process (Prasad and Gupta 1997; Choudhury et al. 1998; González
et al. 2010; Zhang et al. 2012). Prasad and Gupta (1997), Choudhury et al. (1998),
and Saxena and Gupta (1998) noted high level of color degradation, even 80–
93.8 %, in effluents from paper industry treated by T. versicolor, Ph. chrysosporium
(sole or in combination with Pycnoporus sanguineus), Pleurotus ostreatus, and
Heterobasidion annosum.

10.3.4 Biofuels

Intensive world industrialization and motorization result in enormous rise of


demands for fuels, and nowadays 80 % needed energy is met by use of fossil fuels
that have numerous harmful effects on environment. If rapid depletion of the fuels is
added to the negative effects, current trend of finding new, alternative, renewable,
and sustainable energy sources is justified. One of the most environmental friendly
are biofuels, the consumption of which in 2020 should be about 20 % of overall
energy usage (Nigam and Singh 2011). Biofuels are classified into two groups,
primary (wood, wood chips and pellets, etc.) and secondary which are produced by
biomass processing (ethanol, biodiesel, etc.), and secondary ones are further clas-
sified based on sources (forest, agricultural or fishery products, municipal wastes
and wastes from food industry and services) and type (solid, liquid or gaseous). The
main advantages of biofuel utilization are usage of broad distributed sustainable
natural bioresources that could be serious environmental ballast, as well as lower
CO2 and polycyclic aromatic hydrocarbons emissions (Demirbas 2009).
Although crops rich in sugars or compounds that can be converted into the
sugars (starch or cellulose) present an essential bioresources for bioethanol pro-
duction, current trend is development of techniques for its production from inedible
fast-growing biomass (certain trees and grasses), for example from grass
264 M. Stajić et al.

Miscanthus sinensis, which production is 5–6 times higher than cereal one, or from
dried citrus waste that amount is about 800,000 tons only in USA (Zechendorf
1999; Tabka et al. 2006). However, in order to cellulose to be broken down to the
glucose and further ferment, depolymerization of recalcitrant network around cel-
lulose fibers, built of lignin, is necessary. Thus, it comes again to white rot fungi
and their peroxidases which are the main participants in the first step of bioethanol
production known as pretreatment, i.e. the essential catalyzers of beginning phase
of lignin degradation. This pretreatment also depends on fungal species and strain,
cultivation conditions, enzyme production and activity, and oxidative mechanisms
(Dias et al. 2010; Wan and Li 2010; Salvachua et al. 2011). Dias et al. (2010)
reported that various white rot fungi degraded between 2 and 65 % of wheat straw
lignin during its solid state fermentation and in such a way increased
cellulose/lignin ratio. However, Salvachua et al. (2011) emphasized that mode of
lignocellulosic mineralization is different from species to species, for example
contrary to Panus tigrinus and Phlebia radiata that degraded lignin and polysac-
charides simultaneously, Pleurotus eryngii and Phellinus robustus removed lignin
selectively and faster. Likewise, according to these authors, delignification extent is
not always in correlation with ligninolytic enzymes production, as well as with fiber
digestibility and sugar production. Thus, contrary to P. radiata and P. robustus,
characterized both by high Mn-oxidizing peroxidase activities and effective lignin
removal, good lignin degraders Bjerkandera adusta and Coriolopsis rigida had low
active enzymes. In the case of effectiveness of cellulose and hemicellulose enzy-
matic hydrolysis in the second step of bioethanol production, Pycnoporus coccineus
was high effective hemicellulose and weak cellulose degrader (98 and 31 %,
respectively), Bjerkandera adusta was almost equal depolymerizer of these poly-
mers (43 and 54 %, respectively), Stereum hirsutum mineralized only significant
portion of cellulose (43 %), the highest glucose yield was obtained by cultivation of
Poria subvermispora (69 %) and Irpex lacteus (66 %) and the lowest by P. eryngii
and P. robustus.

10.4 Future Trends

Importance of fungi and their enzymes in various biotechnological processes


stimulates serious investment for study of genomes of numerous fungal species, i.e.
sequencing, characterization of ligninolytic enzyme genes, their expression in
appropriate organisms, improvement of product properties, and production of
recombinants in significant amount under non-natural conditions. As Mn-oxidizing
peroxidases are secreted under conditions characterized by high moisture level and
acidic environment (pH about 3.0), which are not typical in biotechnological pro-
cesses, it needs to adapt the enzymes to the conditions. According to Guerriero et al.
(2015), the adaptation could be reached either by directed evolution of certain
ancestral enzyme, i.e. mutation of its gene, or by enzyme engineering, and result in
the both cases is similar enzyme form but with improved and desired property.
10 Role of Mushroom Mn-Oxidizing Peroxidases … 265

Namely, one enzyme rarely possesses all catalytic properties required for effective
lignocellulosic conversion. Therefore recombination can be used for unification of
several beneficial characteristics from a few enzymes into superior one with high
stability and broad substrate promiscuity. Finding of appropriate organism for
expression of the gene leads to obtaining of significant amount of the recombinant
(Alcalde 2015). Thus, Ogawa et al. (1998) showed that expression of mnp genes of
Pleurotus ostreatus, excellent Mn-oxidizing peroxidase producer, in Coprinus
cinereus, characterized with fast growth, led to increase of lignin degradation level
in only 16 days, and Pérez-Boada et al. (2002) and Salame et al. (2012) obtained
active versatile peroxidase by expression of mnp4 gene in Escherichia coli but in
lower yield. Considering that lignocellulolytic enzymes act synergistically, genetic
engineering could be used for fusion of certain enzymes and also for generation of
strain deficient in some enzyme. With this aim, Chalamcherla et al. (2010) getting
cellulase-deficient P. ostreatus strain characterized by low cellulose and high lignin
degradation, as well as by high capacity of application in various biotechnological
processes. Nowadays, special attention is also given to optimization of cultivation
conditions for production of certain lignocellulolytic enzyme and study of enzy-
matic system of fungal species living in extreme environment as well as their genes
encoding proteins with unknown function among which some can be lignocellu-
lolytic enzyme (Guerriero et al. 2015). Everything mentioned show great possi-
bilities of usage and managing of fungi and their enzymes with the aim of biomass
conversion into high-valued products and environment protection.

References

Adamović M, Grubić G, Milenković I, Jovanović R, Protić R, Lj Sretenović, Lj Stoićević (1998)


The biodegradation of wheat straw by Pleurotus ostreatus mushrooms and its use in cattle
feeding. Anim Feed Sci Tech 71:357–362
Alcalde M (2015) Engineering the ligninolytic enzyme. Trends Biotechnol 33:55–162
Anwar Z, Gulfraz M, Irshad M (2014) Agro-industrial lignocellulosic biomass a key to unlack the
future bio-energy: a brief review. J Radiat Res Appl Sci 7:163–173
Arora DS, Sharma RK (2009) Enhancement in in vitro digestibility of wheat straw obtained from
different geographical regions during solid state fermentation by white rot fungi. Bioresources
4:909–920
Asgher M, Ahmed Z, Iqbal HMN (2013) Alkali and enzymatic delignification of sugarcane
bagasse of expose cellulose polymers for saccharification and bio-ethanol production. Ind Crop
Prod 44:488–495
Asiegbu FO, Paterson A, Smith JE (1996) The effects of co-fungal cultures and supplementation
with carbohydrate adjuncts on lignin biodegradation and substrate digestibility. World J
Microb Biot 12:273–279
Asim N, Emdadi Z, Mohammad M, Yarmo MA, Sopian K (2015) Agricultural solid wastes for
green desiccant applications: an overview of research achievements, opportunities and
perspectives. J Clean Prod 91:26–35
Bajpai P, Bajpai PK, Akhtar M, Jauhari MB (2001) Biokraft pulping of eucalyptus with selected
lignin-degrading fungi. J Pulp Pap Sci 27:235–242
Blumenthal K (2011) Generation and treatment of municipal waste. Stat Focus 31:1–12
266 M. Stajić et al.

Calzada JF, Franco LF, de Arriola MC, Rolz C, Ortiz MA (1987) Acceptability, body weight
changes and digestibility of spent wheat straw after harvesting of Pleurotus sajor-caju. Biol
Waste 22:303–309
Camarero S, Sarkar S, Ruiz-Dueñas FJ, Martinez MJ, Martinez AT (1999) Description of a
versatile peroxidase involved in natural degradation of lignin that has both Mn-peroxidase and
lignin-peroxidase substrate binding sites. J Biol Chem 274:10324–10330
Chalamcherla VL, Singaracharya MA, Lakshmi MV (2010) Amino acids profile of the
lignocellulosic feed treated with cellulase-free lignolytic mutants of Pleurotus ostreatus.
Bioresources 5:259–267
Chang ST, Miles PG (2004) Mushrooms. Cultivation, nutritional value, medicinal effect, and
environmental impact. CRC Press LLC, London
Cheung PCK (2013) Mini-review on edible mushrooms as source of dietary fiber: preparation and
health benefits. Food Sci Hum Well 2:162–166
Chi YJ, Hatakka A, Maijala P (2007) Can co-culturing of two white-rot fungi increase lignin
degradation and the production of lignin-degrading enzymes? Int Biodeter Biodegr 59:32–39
Choudhury S, Sahoo N, Manthan M, Rohela RS (1998) Fungal treatment of pulp and paper mill
effluents for pollution control. J Ind Pollut Control 14:1–13
Datta A, Bettermann A, Kirk TK (1991) Identification of a specific manganese peroxidase among
ligninolytic enzymes secreted by Phanerochaete chrysosporium during wood decay. Appl
Environ Microbiol 57:1453–1460
Demirbas A (2009) Progress and recent trends in biodisel fuels. Energ Convers Manag 50:14–34
Dias AA, Freitas GS, Marques GSM, Sampaio A, Fraga IS, Rodrigues MAM, Evtuguin DV,
Bezerra RMF (2010) Enzymatic saccharification of biologically pre-treated wheat straw with
white-rot fungi. Bioresource Technol 101:6045–6050
Dorado J, van Beek TA, Claassen FW, Sierra-Alvarez R (2001) Degradation of lipophilic wood
extractive constituents in Pinus sylvestris by the white rot fungi Bjerkandera sp. and Trametes
versicolor. Wood Sci Technol 35:117–125
Enshasy HAE, Hatti-Kaul R (2013) Mushroom immunomodulators: unique molecules with
unlimited applications. Trends Biotechnol 31:668–677
Eriksson KEL, Blanchette RA, Ander P (1990) Microbial and enzymatic degradation of wood and
wood components. Springer, Berlin
Evans CS, Gallagher IM, Atkey PT, Wood DA (1991) Localisation of degradative enzymes in
white-rot decay of lignocellulose. Biodegradation 2:93–106
Fazaeli H, Azizi A, Amile M (2006) Nutritive value index of treated wheat straw with Pleurotus
fungi fed to sheep. Pakistan J Biol Sci 9:2444–2449
Fernandez-Fueyo E, Ruiz-Dueñas FJ, Ferreira P, Floudas D, Hibbett DS, Canessa P et al (2012)
Comparative genomics of Ceriporiopsis subvermispora and Phanerochaete chrysosporium
provide insight into selective ligninolysis. Proc Nat Acad Sci USA 109:5458–5463
Fernández-Fueyo E, Ruiz-Dueñas FJ, Martínez MJ, Romero A, Hammel KE, Medrano FJ,
Martínez AT (2014) Ligninolytic peroxidase genes in the oyster mushroom genome:
heterologous expression, molecular structure, catalytic and stability properties, and
lignin-degrading ability. Biotechnol Biofuels 7:2
Ginterová A, Lazarová A (1987) Degradation dynamics of lignocellulose materials by
wood-rotting Pleurotus fungi. Folia Microbiol 32:434–437
González Alriols M, Tejado A, Blanco M, Mondragon I, Labidi J (2009) Agricultural palm oil tree
residues as raw material for cellulose, lignin and hemicelluloses production by ethylene glycol
pulping process. Chem Eng J 148:106–114
González LF, Sarria V, Sánchez OF (2010) Degradation of chlorophenols by sequential
biological-advanced oxidative process using Trametes pubescens and TiO2/UV.
BioresourceTechnol 101:3493–3499
Grabber JH (2005) How do lignin composition, structure, and cross-linking affect degradability? A
review of cell wall model studies. Crop Sci 45:820–831
Guerriero G, Hausman JF, Strauss J, Ertan H (2015) Destructuring plant biomass: focus on fungal
and extremophilic cell wall hydrolases. Plant Sci 234:180–193
10 Role of Mushroom Mn-Oxidizing Peroxidases … 267

Guillén F, Gómez-Toribio V, Martinez MJ, Martinez AT (2000) Production of hydroxyl radical by


the synergistic action of fungal laccase and aryl-alcohol oxidase. Arch Biochem Biophys
383:142–147
Gupta A, Verma JP (2015) Sustainable bio-ethanol production from agro-residues: a review.
Renew Sust Energ Rev 41:550–567
Gutiérrez A, Caramelo L, Prieto A, Martinez MJ, Martinez AT (1994) Anisaldehyde production
and aryl-alcohol oxidase and dehydrogenase activities in ligninolytic fungi from the genus
Pleurotus. Appl Environ Microbiol 60:1783–1788
Gutiérrez A, del Rio JC, Rencoret J, Ibarra D, Martinez AT (2006) Main lipophilic extractives in
different paper pulp types can be removed using the laccase-mediator system. Appl Microbiol
Biotechnol 72:845–851
Gutiérrez A, del Rio JC, Martinez AT (2009) Microbial and enzymatic control of pitch in the pulp
and paper industry. Appl Microbiol Biotechnol 82:1005–1018
Hammel EK (1997) Fungal degradation of lignin. In: Cadisch G, Giller KE (eds) Driven by nature:
plant litter quality and decomposition. CAB International, Wallingford, pp 33–45
Hammel KE, Cullen D (2008) Role of fungal peroxidases in biological ligninolysis. Curr Opin
Plant Biol 11:349–355
Hendriks ATWM, Zeeman G (2009) Pretreatments to enhance the digestibility of lignocellulosic
biomass. Bioresource Technol 100:10–18
Hofrichter M (2002) Review: lignin conversion by manganese peroxidase (MnP). Enzyme Microb
Tech 30:454–466
Iqbal HMN, Ahmed I, Zia MA, Irfan M (2011) Purification and characterization of the kinetic
parameters of cellulose produced from wheat straw by Trichoderma viride under SSF and its
detergent compatibility. Adv Biosci Biotechnol 2:149–156
Jerusik RJ (2010) Fungi and paper manufacture. Fungal Biol Rev 24:68–72
Kirk TK, Cullen D (1998) Enzymology and molecular genetics of wood degradation by white-rot
fungi. In: Young RA, Akhtar M (eds) Environmentally friendly technologies for the pulp and
paper industry. Wiley, New York, pp 273–308
Knežević A, Milovanović I, Stajić M, Lončar N, Brčeski I, Vukojević J, Ćilerdžić J (2013a) Lignin
degradation by selected fungal species. Bioresource Technol 138:117–123
Knežević A, Milovanović I, Stajić M, Vukojević J (2013b) Potential of Trametes species to
degrade lignin. Int Biodeter Biodegr 85:52–56
Kuijk SJA, Sonnenberg ASM, Baars JJP, Hendriks WH, Cone JW (2015) Fungal treated
lignocellulosic biomass as ruminant feed ingredient: a review. Biotechnol Adv 33:191–202
Lanfermann I, Linke D, Nimtz M, Berger RG (2015) Manganese peroxidases from Ganoderma
applanatum degrade β-carotene under alkaline conditions. Appl Biochem Biotechnol
175:3800–3812
Lapierre C (1993) Application of new methods for the investigation of lignin structure. In:
Jung HG, Buxton DR, Hatfield RD, Ralph J (eds) Forage cell wall structure and digestibility.
American Society of Agronomy, Madison, pp 133–166
Leonowicz A, Matuszewska A, Luterek J, Ziegenhagen D, Wojtaś-Wasilewska M, Cho NS,
Hofrichter M, Rogalski J (1999) Biodegradation of lignin by white rot fungi. Fungal Gen Biol
27:175–185
Martinez AT (2002) Molecular biology and structure-function of lignin-degrading heme
peroxidases. Enzyme Microb Tech 30:425–432
Martinez MJ, Ruiz-Dueñas FJ, Guillén F, Martinez AT (1996) Purification and catalytic properties
of two manganese peroxidase isoenzymes from Pleurotus eryngii. Eur J Biochem 237:424–432
Martinez D, Larrondo LF, Putnam N, Gelpke MDS, Huang K, Chapman J, Helfenbein KG,
Ramaiya P, Detter JC, Larimer F et al (2004) Genome sequence of the lignocellulose degrading
fungus Phanerochaete chrysosporium strain RP78. Nat Biotechnol 22:695–700
Martinez-Inigo MJ, Gutiérrez A, del Rio JC, Martinez MJ, Martinez AT (2000) Time course of
fungal removal of lipophilic extractives from Eucalyptus globulus wood. J Biotechnol 84:119–
126
268 M. Stajić et al.

Medeiros BM, Bento VA, Nunes LLA, Oliveira CS (1999) Optimization of some variables that
affect the synthesis of laccase by Pleurotus ostreatus. Bioprocess Eng 21:483–487
Muñoz C, Guillen F, Martínez TA, Martínez JM (1997a) Induction and characterization of laccase
in the ligninolytic fungus Pleurotus eryngii. Cur Microbiol 34:1–5
Muñoz C, Guillen F, Martínez TA, Martínez JM (1997b) Laccase isoenzymes of Pleurotus
eryngii: characterization, catalytic properties, and participation in activation of molecular
oxygen and Mn2+ oxidation. Appl Environ Microbiol 63:2166–2174
Nigam PS, Singh A (2011) Production of liquid biofuels from renewable resources. Prog Energ
Combust 37:52–68
Ogawa K, Yamazaki T, Kajiwara S, Watanabe A, Asada Y, Shishido K (1998) Molecular breeding
of the basidiomycete Coprinus cinereus strains with high lignin-decolorization and -
degradation activities using novel heterologous protein expression vectors. Appl Microbiol
Biotechnol 49:285–289
Pérez-Boada M, Doyle WA, Ruiz-Dueñas FJ, Martínez MJ, Martínez AT, Smith AT (2002)
Expression of Pleurotus eryngii versatile peroxidase in Escherichia coli and optimisation of
in vitro folding. Enzyme Microb Tech 30:518–524
Perez-Boada M, Ruiz-Dueñas FJ, Pogni R, Basosi R, Choinowski T, Martinez MJ, Piontek K,
Martinez AT (2005) Versatile peroxidase oxidation of high redox potential aromatic
compounds: site-directed mutagenesis, spectroscopic and crystallographic investigation of
three long-range electron transfer pathways. J Mol Biol 354:385–402
Pokhrel D, Viraraghavan T (2004) Treatment of pulp and paper mill wastewater—a review. Sci
Total Environ 333:37–58
Prasad GK, Gupta RK (1997) Decolourization of pulp and paper mill effluent by two white-rot
fungi. Indian J Environ Health 39:89–96
Rai SN, Walli TK, Gupta BN (1989) The chemical composition and nutritive value of rice straw
after treatment with urea or Coprinus fimetarius in a solid state fermentation system. Anim
Feed Sci Tech 26:81–92
Reis FS, Barros L, Martins A, Ferreira ICFR (2012) Chemical composition and nutritional value of
the most widely appreciated cultivated mushrooms: an inter-species comparative study. Food
Chem Toxicol 50:191–197
Rodrigues MAM, Pinto P, Bezerra RMF, Dias AA, Guedes CVM, Cardoso VMG et al (2008)
Effect of enzyme extracts isolated from white-rot fungi on chemical composition and in vitro
digestibility of wheat straw. Anim Feed Sci Technol 141:326–338
Ruiz-Dueñas FJ, Guillen F, Camarero S, Perez-Boada M, Martinez MJ, Martinez AT (1999)
Regulation of peroxidase transcript levels in liquid cultures of the ligninolytic fungus Pleurotus
eryngii. Appl Environ Microbiol 65:4458–4463
Ruiz-Dueñas FJ, Morales M, Garcia EMY, Martinez MJ, Martinez AT (2009) Substrate oxidation
sites in versatile peroxidase and other basidiomycetes peroxidases. J Exp Bot 60:441–452
Salame TM, Knop D, Levinson D, Mabjeesh SJ, YardenO Hadar Y (2012) Release of Pleurotus
ostreatus versatile-peroxidase from Mn2+ repression enhances anthropogenic and natural
substrate degradation. PLoS ONE 7:e52446
Salvachua D, Prieto A, Lopez-Abelairas M, Lu-Chau T, Martinez AT, Martinez MJ (2011) Fungal
pretreatment: an alternative in second-generation ethanol from wheat straw. Bioresource
Technol 102:7500–7506
Sánchez C (2009) Lignocellulosic residues: biodegradation and bioconversion by fungi.
Biotechnol Adv 27:185–194
Saxena N, Gupta RK (1998) Decolourization and delignification of pulp and paper mill effluent by
white rot fungi. Indian J Exp Biol 36:1049–1051
Shrivastava B, Nandal P, Sharma A, Jain KK, Khasa YP, Das TK, Mani V, Kewalramani NJ,
Kundu SS, Kuhad RC (2012) Solid state bioconversion of wheat straw into digestible and
nutritive ruminant feed by Ganoderma sp. rckk02. Bioresource Technol 107:347–351
Simonić J, Vukojević J, Stajić M, Glamočlija J (2010) Intraspecific diversity within Ganoderma
lucidum in laccase and Mn-dependent peroxidases production during plant residues
fermentation. Appl Biochem Biotechnol 162:408–415
10 Role of Mushroom Mn-Oxidizing Peroxidases … 269

Singh AP, Singh T (2014) Biotechnological applications of wood-rotting fungi: a review. Biomass
Bioenerg 62:198–206
Skyba O, Douglas CJ, Mansfield SD (2013) Syringyl-rich lignin renders poplars more resistant to
degradation by wood decay fungi. Appl Environ Microbiol 79:2560–2571
Stajić M, Persky L, Friesem D, Hadar Y, Wasser SP, Nevo E, Vukojević J (2006a) Effect of
different carbon and nitrogen sources on laccase and peroxidases activity by selected Pleurotus
species. Enzyme Microb Tech 38:65–73
Stajić M, Persky L, Hadar Y, Friesem D, Duletić-Laušević S, Wasser SP, Nevo E (2006b) Effect of
copper and manganese ions on activities of laccase and peroxidases in three Pleurotus species
grown on agricultural wastes. Appl Biochem Biotechnol 128:87–96
Stajić M, Vukojević J, Duletić-Laušević S (2009) Biology of Pleurotus eryngii and the role in
biotechnological processes: a review. Crit Rev Biotechnol 29:55–66
Stajić M, Kukavica B, Vukojević J, Simonić J, Veljović-Jovanović S, Duletić-Laušević S (2010)
Wheat straw conversion by enzymatic system of Ganoderma lucidum. Bioresources 5:2362–
2373
Stajić M, Vukojević J, Knežević A, Milovanović I (2013) Influence of trace elements on
ligninolytic enzyme activity of Pleurotus ostreatus and P. pulmonarius. Bioresources 8:3027–
3037
Stuardo M, Vasquez M, Vicuna R, Gonzalez B (2004) Molecular approach for analysis of model
fungal genes encoding ligninolytic peroxidases in wood-decaying soil system. Lett Appl
Microbiol 38:43–49
Tabka MG, Herpoel-Gimbert I, Monod F, Asther M, Sigoillot JC (2006) Enzymatic sacchari-
fication of wheat straw for bioethanol production by a combined cellulose xylanase and
feruloyl esterase treatment. Enzyme Microb Tech 39:897–902
Thurston C (1994) The structure and function of fungal laccase. Microbiol 140:19–26
Tuyen VD, Cone JW, Baars JJP, Sonnenberg ASM, Hendriks WH (2012) Fungal strain and
incubation period affect chemical composition and nutrient availability of wheat straw for
rumen fermentation. Bioresource Technol 111:336–342
Urzúa U, Larrondo LF, Lobos S, Larrain J, Vicuña R (1995) Oxidation reactions catalyzed by
manganese peroxidase isoenzymes from Ceriporiopsis subvermispora. FEBS Lett 371:132–
136
Van Beek TA, Kuster B, Chassen FW, Tienvieri T, Bertaud F, Lenon G, Petit-Conil M,
Sierra-Alvarez R (2007) Fungal biotreatment of spruce wood with Trametes versicolor for
pitch control: influence of extractive contents, pulping process parameters, paper quality and
effluent toxicity. Bioresource Technol 98:302–311
Varela E, Guillén F, Martínez AT, Martínez MJ (2001) Expression of Pleurotus eryngii
aryl-alcohol oxidase in Aspergillus nidulans: purification and characterization of the
recombinant enzyme. Biochim Biophys Acta 1546:107–113
Wan C, Li Y (2010) Microbial pretreatment of corn stover with Ceriporiopsis subvermispora for
enzymatic hydrolysis and ethanol production. Bioresource Technol 101:6398–6403
Wariishi H, Akilswaran L, Gold MH (1988) Manganese peroxidase from the basidiomycetes
Phanerochaete chrysosporium: spectral characterization of the oxidized states and the catalytic
cycle. Biochemistry 27:5365–5370
Yang Q, Zhan H, Wang S, Fu S, Li K (2008) Modification of Eucalyptus CTMP fibers with white
rot fungus Trametes hirsute—effects on fiber morphology and paper physical strengths.
Bioresource Technol 99:8118–8124
Yang B, Wyman C (2008) Pre-treatment: the key to unlocking low-cost cellulosic ethanol.
Biofuels Bioprod Bioref 2:26–40
Zechendorf B (1999) Sustainable development: How can biotechnology contribute? Feature
17:219–225
Zhang S, Jiang M, Zhou Z, Zhao M, Li Y (2012) Selective removal of lignin in steam-exploded
rice straw by Phanerochaete chrysosporium. Int Biodeter Biodegr 75:89–95
Chapter 11
Role and Application of Versatile
Peroxidase (VP) for Utilizing
Lignocellulose in Biorefineries

Nadine Busse and Peter Czermak

Abstract Within the last few years, the relatively new heme peroxidase, versatile
peroxidase (VP), attracted some attention as a model enzyme and as an industrial
biocatalyst. VPs are interesting for efficient delignification processes due to their
ability to degrade a broad spectrum of recalcitrant substrates without using medi-
ators. Lignin is very complex, highly aromatic, and one of the major compounds of
lignocellulose, which is an important feedstock for biorefineries, particularly when
originated from wood. In utilizing lignocellulose (e.g., second generation biofuels),
lignin modification, and removal must first be addressed. Moreover, lignin degra-
dation for value-added bio-products is also of great interest. An industrial imple-
mentation of VPs would be beneficial in this context, but involves several scientific
and technical challenges.

11.1 Introduction

Lignin modification and removal plays a key role in utilizing lignocellulose/


lignocellulosic biomass for second generation biofuels, and other bio-based prod-
ucts, and therefore must be addressed first. Lignocellulose from wood is of partic-
ularly high interest for this process due to the following reasons: Wood is
inexpensive, available in large amounts (Stöcker 2008), CO2 neutral (Lange 2007),
and rich in lignin (18–35 %), as well as the carbohydrates cellulose (40–55 %) and
hemicellulose (24–40 %) (Sun and Cheng 2002; Howard et al. 2003).
Due to the fact that lignin is the only naturally synthesized aromatic biopolymer
(Dashtban et al. 2010) its effective use and degradation to value-added macro-
molecules (e.g., low cost carbon fibers, polymer modifiers) and/or aromatic
chemicals (e.g., phenols, vanillin, quinone, guaiacol, BTX chemicals) is of crucial

N. Busse  P. Czermak (&)


Institute of Bioprocess Engineering and Pharmaceutical Technology,
Mittelhessen University of Applied Sciences, Wiesenstrasse 14,
35390 Giessen, Germany
e-mail: [email protected]; [email protected]

© Springer International Publishing Switzerland 2016 271


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_11
272 N. Busse and P. Czermak

importance, with the latter of these being of high economic potential and therefore
particularly interesting (Holladay et al. 2007; Huang and Ramaswamy 2013).
However, the contributing factors affecting the probability of their successful
implementation include the structural complexity of native lignin, its recalcitrance,
and its uniqueness (the woody plant’s fingerprint) leading to inconsistent, poly-
disperse product streams. These facts still constitute a major scientific and technical
challenge (Holladay et al. 2007). Consequently, further fundamental research and
development is needed for lignin degradation in both general and targeted usage.
Prerequisites for efficient lignin modification and degradation in biorefineries
are:
(i) Conservation of important lignin related aromatic structures.
(ii) Subsequent processes like saccharification of cellulose by cellulases should
not be negatively influenced.
(iii) Mild conditions.
(iv) Moderate waste streams/effluent treatment and moderate operating costs.
Ligninolytic heme peroxidases (POXs) have gained increased importance in this
context, with particular attention on the use of versatile peroxidases (VPs). VPs are
relatively new POXs and were first identified as MnPs in the late nineties. Within
the last few years they have attracted attention “as a model enzyme and as a source
of industrial/environmental biocatalyst” (Ruiz-Dueñas et al. 2009a). The main
reason for this is their capability to digest/convert a broad spectrum of complex
substrates, ranging from azo dyes like RB5 to (non)phenolic lignin, without the
necessity of mediators (Pogni et al. 2005; Martínez et al. 2009). Nonetheless,
several factors hinder their short-term commercialization and industrial imple-
mentation. This chapter outlines the main issues for industrial applications, starting
first with a brief overview of the involvement of POXs in natural delignification.

11.2 Involvement of Heme Peroxidases


in the Lignocellulose Degradation Mechanism

In nature, lignocellulose (LC) degradation involves several factors as follows:


(i) Lignocellulose composition (e.g., lignin content, extractables).
(ii) Environmental conditions (e.g., T, pH, moisture, N2, O2) (Blanchette 1991;
Wong 2009).
(iii) Biological community.
Biological community includes microbial population, fungi, and insects, and
determines the degradation process. In order to enable the use of LC, lignin modi-
fication and removal must first be addressed. Basidiomycetes, white-rot fungi, are
well known to be the most efficient lignin degraders due to their extracellular
ligninolytic enzymes (Kirk and Farrell 1987), the H2O2-dependent heme peroxidases
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 273

Fig. 11.1 Summary of the natural degradation process of lignocellulosic biomass and the
importance of ligninolytic heme peroxidases (modified based on Dashtban et al. 2010). Here, the
reactions of the first POX intermediates are demonstrated. Lac laccase, LiP lignin peroxidase, MnP
manganese peroxidase, VP versatile peroxidase; Lac-I, LiP-I, MnP-I, and VP-I are corresponding
protein radicals, generally known as compound I. S stands for an appropriate substrate of low
molecular weight resulting in relatively reactive radicals which ultimately mediate the cleavage of
lignin bonds and lignin modifications

(POX) and O2-dependent phenol oxidases lacasses (Lac, EC 1.10.3.2) as seen in


Fig. 11.1. Figure 11.1 demonstrates a part of the full enzymatic reaction mechanism
for better clarity (specifically the reactions of the first enzyme intermediate). More
details on this will follow in subsequent chapters. The ligninolytic enzyme system
varies individually in composition. In addition, it must be further differentiated into
selective and nonselective lignin degraders (Table 11.1). The former selectively
remove lignin without considerable cellulose degradation, whereas the latter also
attack cellulose (Blanchette 1991; Hatakka 2005; Hatakka and Hammel 2011).
Some can however produce both types, such as Heterobasidion annosum (in
Table 11.1) (Eriksson et al. 1990).
Concerning lignin degradation, two pathways must be differentiated (Fig. 11.1):
(i) Direct (black dashed lines).
(ii) Indirect (black solid lines).
274 N. Busse and P. Czermak

Table 11.1 A selection of Species Secreted enzymes


POX and Lac producing
white-rot fungi (modified Lignin-selective
based on Hatakka (1994), Bjerkandera adusta LiP, MnP, VPa
Ayala et al. (2008), Sánchez Bjerkandera species VPb
(2009) and Lundell et al. Ceriporiopsis subvermispora MnP, Lac
(2010))
Dichomitus squalens MnP, Lac
Heterobasidion annosum MnP
Phanerochaete chrysosporium LiP, MnP
Phellinus pini LiP, MnP
Phlebia radiata LiP, MnP, Lac
Phlebia subserialis MnP
Phlebia tremellosus LiP, MnP, Lac
Pleurotus eryngii MnP, VPc, Lac
Pleurotus ostreatus MnP, VPd, Lac
Pleurotus sapidus VPe
Pycnoporus cinnabarinus LiP, MnP, Lac
Nonselective
Ganoderma applanatum LiP
Heterobasidion annosum MnP
Trametes versicolor LiP, MnP, VPf, Lac
Bold indicates VP producing white-rot fungi
a
Wang et al. (2003) and Busse et al. (2013)
b
Mester and Field (1998) and Moreira et al. (2006)
c
Martínez et al. (1996) and Camarero et al. (1999)
d
Hatakka and Hammel (2011)
e
Zorn et al. (2005) and Schüttmann et al. (2014)
f
Carabajal et al. (2013)

Direct degradation refers to when the ligninolytic enzyme is in its active state
(characterized by an appended suffix-I) and directly attacks native lignin linkages.
However, when low molecular weight (LMW) substrate (S) is present/provided,
this interposes an intermediate step where enzyme action results first in
substrate-related diffusible radicals mediating nonenzymatic reactions, which then
cause the cleavage of lignin bonds. This is called indirect lignin degradation,
because the LMW substrates serve as so-called mediators. Lac rely on phenolic
mediators for promoting their oxidation capabilities “towards more recalcitrant
non-phenolic lignin compounds” (Cañas and Camarero 2010). For oxidative ability,
the enzyme redox potential is a limited criterion (Ayala 2010). Lac exhibits lower
redox potential properties (≤ 0.8 V) than POX (> 1 V) (Bourbonnais and Paice
1990; Cañas and Camarero 2010). Lignin comprises 80–90 % non-phenolic moi-
eties, where just 10–20 % are phenolics (Kawai et al. 1999). The cleavage of
non-phenolics is thus essential for efficient lignin modification and degradation. The
redox potential of non-phenolic lignin compounds is also > 1 V (Cañas and
Camarero 2010). Consequently, POX have recently gained increased attention for
delignification purposes (Cañas and Camarero 2010) and received higher interest as
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 275

industrial biocatalysts (Martínez 2007). Nevertheless, manganese peroxidases


(MnP, EC 1.11.1.13) principally need manganese (Mn2+) as a substrate and
mediator to form Mn3+ ions. These ions will be subsequently chelated by organic
acids (e.g., oxalate, malonate, lactate) to powerful oxidants. As a result, MnPs are
able to oxidize certain non-phenolic aromatics (e.g., carboxylic acids, methoxy-
benzenes, lipids) and are not restricted to phenolics as usual. Recalcitrant
non-phenolic lignin units still remain unaffected. Therefore, it is supposed that MnP
activity rather causes phenoxyl radical formation (as in case of Lac) which in turn is
responsible for subsequent bond cleavages in the biopolymer lignin (Hatakka
2005). Contrary to this, lignin peroxidases (LiP, EC 1.11.1.14) prefer to react with
non-phenolic lignin substructures, interestingly, without the need for mediators.
White-rot fungi, however, simultaneously produces the non-phenolic monomer
veratryl alcohol (VA) as a second metabolite (Schoemaker and Piontek 1996). VA
is well known to be an essential substrate S for LiP, elevating its activity, as well as
increasing lignin degradation. Reaction with VA again causes radical formation
(veratryl alcohol radicals VA•), which additionally enables phenolic compound
oxidation.
Versatile peroxidases (VP, EC 1.11.1.16) exemplify catalytic properties of both
LiP and MnP, and thus initiate similar reaction pathways. VPs, however, are less
specific in facilitating degradation among a broad spectrum of substrates, irre-
spective of phenolic or non-phenolic, and do not require the presence of mediators
(Pogni et al. 2005).
In conclusion, lignin degradation in nature is not performed by just one enzyme,
but rather a synergistic process of an array of ligninolytic enzymes and their iso-
zymes, accessory enzymes (e.g., H2O2-generating oxidases (aryl-alcohol oxidase,
glyoxal oxidase)), aryl-alcohol oxidases, and quinone reductases) and
non-enzymatic reactions (Fig. 11.1). The latter mainly produces hydroxyl radicals
(•OH) which support lignin modification and degradation, as well as the degradation
of cellulose and hemicellulose in a non-specific manner. The involvement of such
radicals results in easier penetration of:
(i) POX, since they are unable to penetrate through the lignified cell wall and
therefore first erosion is necessary (Hammel et al. 1993).
(ii) Lignocellulolytic enzymes, such as (hemi)cellulases for subsequent (hemi)-
cellulose digestion (Dashtban et al. 2010).

11.3 Catalytic Mechanism of VP

As initially mentioned, VPs are relative new POXs and are attractive (model)
biocatalysts (Ruiz-Dueñas et al. 2009a). One key advantage is their independence
from mediators. For industrial application, mediators are used in high amounts.
Therefore, mediators must be (i) cheap, (ii) non-toxic, (iii) readily available,
276 N. Busse and P. Czermak

(iv) in large quantities, and/or (v) easy recyclable (Rodríguez Couto and Toca
Herrera 2006; Cañas and Camarero 2010).
Research work concerning delignification processes by VPs are limited, particu-
larly in kinetic studies with representative lignin model compounds (LMCs)
like adlerol (1-(3,4-dimethoxyphenyl)-2-(2-methoxy-phenoxy)-1,3-propanediol), a
non-phenolic β-O-4 lignin model dimer. The reason for this is the underlying, highly
complex, POX reaction mechanism which will be demonstrated in the following, prior
to focusing on mediator-independent adlerol degradation.

11.3.1 The General Peroxidase Reaction Mechanism

The catalytic peroxidase cycle is expected to follow a ping–pong mechanism.


Contrary to the classical mechanism, peroxidase reaction schemes are further
assumed to be irreversible, as sketched in Fig. 11.2 in simplified terms. Enzyme–
substrate complex formation (e.g., ES) prior to reaction product generation can thus
be neglected, since they are considered of fleeting non-detectable existence
(Dunford 1991). This is valid for most heme peroxidases, (Dunford 2010) with iron
protoporphyrin (P) IX and iron/heme (Fe3+) as the catalytic center and the pros-
thetic group, respectively (de Montellano 2010).
During the catalytic cycle (Fig. 11.2), the enzyme undergoes two fundamental
structural changes. First (pathway 1), the reaction is initiated by H2O2 through a
pH-independent, two-electron oxidation (pH range is *2–7.5 for LiP (Andrawis
et al. 1988) or 3.5 and 7 for VP (Pérez-Boada et al. 2005)). As a result, the enzyme
will be converted from its native resting ferric/ground state (E0) to the so-called
compound I (EI, first intermediate) (Eq. 11.1), a protein cation radical
(de Montellano 2010) of strong oxidative power (Arnao et al. 1990b).

! E ½Fe
k1
E0 ½Fe3 þ  þ H2 O2 I 4þ
¼ OP þ  þ H2 O ð11:1Þ

with k as the reaction rate constant and the index for describing the pathway
number.

Fig. 11.2 Simplified “POX ping–pong” mechanism. E0–EII represents the three main peroxidase
oxidation states. The symbols S and S•+ stand for an appropriate substrate (e.g., lignin, lignin
model compounds such as adlerol) and its corresponding radical cation. The reaction will be
initiated by H2O2 (pathway 1), and followed by two consecutive, one-electron reduction steps
(pathway 2–3). Reprinted from Busse et al. (2013) with slight modification
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 277

Subsequently, EI returns back to E0 via a second enzyme intermediate (EII,


compound II) due to two consecutive one-electron reduction steps triggered by
suitable reducing substrates (S, the actual electron donor “preferentially
electron-rich aromatic compounds” (Lundell et al. 1993a)). Both reactions are
pH-dependent (Pérez-Boada et al. 2005; Wong 2009) resulting in the release of
radical cation intermediates (S•+) (Palmer et al. 1987) (pathway 2 and 3 in
Fig. 11.2; Eqs. 11.2, 11.3):

! E ½Fe
k2
EI ½Fe4 þ ¼ OP þ  þ S II 4þ
¼ O þ S þ ð11:2Þ

! E ½Fe
k3
EII ½Fe4 þ ¼ O þ S 0 3þ
 þ S þ þ H2 O ð11:3Þ

In Eq. (11.2), substrates causing EI reduction can also reduce EII (Eq. 11.3) as
described by Dunford (2010) for horseradish peroxidase (HRP). The same applies
for ligninolytic enzymes, such as LiP, and phenolic substrates (Schoemaker 1990).
Here, the overall net enzyme reaction can be simply expressed by Eq. (11.4).

! 2S
r

2S + H2 O2 þ 2H2 O ð11:4Þ

Corresponding consumption rates r for S and H2O2 result in Eq. (11.5).

1 d[S] d[H2 O2  1 d[S þ 


r¼ ¼ ¼ ð11:5Þ
2 dt dt 2 dt

Assuming the POX cycle is not inhibited, and considering k3 is rate limiting
(k3 = kcat because of k2 ≫ k3), r results in Eq. (11.6) (Rasmussen et al. 1995).

2k3 ½ET ½S2 ½S1 


r¼ ð11:6Þ
k1 ½S2  þ ½S1 
k3

X
II
½ET  ¼ ½Ei  ð11:7Þ
i¼0

ET means total enzyme.


When non-phenolic substrates are involved, caution should be exercised
regarding generalization. EII of LiP, for instance, can be reduced just by
di-methoxylated (minimum amount of alkoxy substituents), non-phenolic aromatic
compounds, whereas EI also accepts mono-methoxylated aromatic components
(Schoemaker 1990).
All produced radical cation intermediates (Fig. 11.2) will further pass through
diffusion-controlled, non-enzymatic reactions as schematically illustrated in
Fig. 11.3. Such radical reactions yield complex product mixtures. Depending on the
278 N. Busse and P. Czermak

Fig. 11.3 Schematic drawing of nonenzymatic radical reactions–radical chain reactions. The
reaction is initiated by the cation radical intermediate S•+, a product of enzyme activity

nature of S•+, subsequent non-enzymatic reactions are the following (recently


summarized by Busse et al. (2013) based on Palmer et al. (1987) and Schoemaker
(1990)):
(a) One-electron oxidation of another appropriate substrate molecule RH (R also
stands for aromatic residuals, e.g., lignin or lignin derivatives), which in turn is
transferred to R•+ (Fig. 11.3). S•+ is reduced back to its ground state (Palmer
et al. 1987). This kind of reaction is referred to as mediator behavior.
(b) S•+ can also immediately be degraded to free radicals R•1 (R1 stands again for
aromatic residual) (Palmer et al. 1987), which is caused by rapid deprotonation
processes (Schoemaker et al. 1994a) (Fig. 11.3). Note, index 1 is just used for
differentiation purposes.
(c) Side-chain cleavage, e.g., Cα–Cβ bond cleavage (Fig. 11.4, section 11.3.2),
and C–H bond cleavage.
(d) Demethoxylation.
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 279

(e) Ether-bond cleavage [e.g., addition of solvent (H2O)] (Palmer et al. 1987).
(f) Hydroxylation of benzylic methylene groups, e.g., through oxygen incorpo-
ration via O2 or H2O in the solvent (Fig. 11.3).
(g) Phenol formation by nucleophilic attack, e.g., the addition of a solvent such as
H2O (Palmer et al. 1987).
(h) Aromatic ring cleavage caused by reactions with perhydroxy radicals HOO•/
HO•2 (Fig. 11.3); details will follow in the next section (Palmer et al. 1987).
The highly reactive free-radical species (in Fig. 11.3 denoted as R• and R•1) can bind
to molecular oxygen (O2), forming final products as well as superoxide anion
radicals (O•−2 ) (Schoemaker 1990) via degradation of organic peroxy radical
intermediates (ROO• or R1OO• in Fig. 11.3) (Palmer et al. 1987). O•− 2 ultimately
undergoes rapid disproportionation forming H2O2 and O2 (Harman et al. 1986;
Schoemaker 1990). Here, the O•− 2 is in a pH-dependent equilibrium with its pro-
tonated counterpart HOO• (Bielski et al. 1985). At low pH conditions, HOO•
predominates, existing as a powerful oxidant (Halliwell and Gutteridge 1985) and
thus contributing to the substrate degradation processes. This occurs because
additional bond cleavage is initiated as a consequence of the substrate (S) oxidizing
to its radical cation, where HOO• is reduced to H2O2 (Palmer et al. 1987).
Alternatively, R1OO• can further react with substrate molecules RH, resulting in
new free radical R• and organic peroxide R1OOR′ formation. If R′ represents
hydrogen, organic hydroperoxide (R1OOH) will instead be produced. As shown in
Fig. 11.3, R1OOH competes with the co-substrate H2O2 for reaction with E0–EI and
with S for EI conversion, depending on their relative concentrations. Reactions of
R1OOH with EI lead to undesired enzyme inactivations. In a low-oxygen atmo-
sphere, R1OOH formation diminishes (Acosta et al. 1988), but polymerization
increases (Palmer et al. 1987). R• products may also attack the peroxidase, again
leading to inactivation (Nicell et al. 1993). However, these radicals can also
undergo dimerization and polymerization processes (e.g., reaction with other free
radicals such as R•1) (Palmer et al. 1987), or can act as suitable substrates S for
maintaining the catalytic cycle. The latter is, however, limited in applicability for
radical cation intermediates S•+, as demonstrated later.
With consideration of further enzymatic reactions, both O•− •
2 and HOO (de-
pending on pH) can additionally serve as electron donors, instead of reducing
substrates (competitive reaction). Consequently, all three enzyme states, EI, EII, and
EIII may be, for instance, reduced by O•−2 under O2 production (Bielski et al. 1985;
Kettle et al. 2007) according to Eqs. (11.8)–(11.11), based on Kettle et al. (2007).
The reaction in Eq. (11.9) is supposed to prevent enzyme inhibition caused by poor
reducing substrates, in order to maintain activity (Kettle et al. 2007).

EI þ O
2 ! E þ O2
II
ð11:8Þ

EII þ O
2 þH
þ
! E0 þ O2 þ H2 O ð11:9Þ
280 N. Busse and P. Czermak

EIII þ O
2 þ 2H
þ
! E0 þ H2 O2 þ O2 ð11:10Þ

or

EIII þ O
2 þ 2H
þ
! EI þ H2 O þ O2 ð11:11Þ

At this point, it becomes obvious that peroxidase activity at least leads to radical
cation formation by reacting with a suitable substrate, which triggers several
non-enzymatic reactions, similar to a “domino effect.” Moreover, the complexity of
the peroxidase reaction mechanism is evident, which explains why even the
degradation of simple aromatic monomers (e.g., VA), except pathway I in Fig. 11.2
(Dunford 2010), is not fully understood, and that further fundamental research is
still needed. Indeed, in a recent review, Busse et al. (2013) found that the mech-
anism is far more complex and strongly depends on (i) reaction conditions, (ii) POX
type, (iii) the presence of inhibitors, (iv) the used substrate.

11.3.2 Adlerol Degradation

Adlerol is a non-phenolic lignin model dimer with a β-O-4 linkage (structure I in


Fig. 11.4). Such LMCs are the most abundant units in lignin polymers, with con-
centrations ranging from 50 to 65 % (Adler 1977). However, the question arises if
adlerol is a representative model compound, since this kind of systems does not
include linkages to wood polysaccharides. Studying fungal lignin-degrading ability
based on 14C-lignins, by contrast, is suggested as the most reliable method (Hatakka
and Hammel 2011). From this perspective, and due to the fact that natural lignin is a
very complex, water-insoluble biopolymer, low molecular weight LMCs like
adlerol, which is also less soluble in water, do not initially seem to appropriately
mimic reality. Not even industrial/technical lignins (e.g., high polydisperse
ligninsulfonates) or dimeric LMCs attached to PEG (polyethylene glycol) would
ever result in realistic test scenarios. Woody biomass is exclusively the most rep-
resentative substrate, which complicates the situation even more. Major drawbacks
include: (i) variations in quality from batch to batch, and the fact that (ii) high
enzyme loads are required just to achieve satisfactory reaction rates.
Alternatively, the choice of an appropriate model system depends on the research
subject(s). Upon the examination of the fundamental issues, including studying
reaction mechanisms, enzyme (deactivation) kinetics, and establishing monitoring
systems for process control, it can be determined that LMCs are in fact very useful.
β-O-4 cleavage, for instance, is considered as a substantial indicator for both enzy-
matic lignin depolymerization and modification (Tien 1987; Wong 2009), making
adlerol a suitable LMC for studying delignification mechanisms. Moreover, the β-O-4
bond is stable at an acidic pH (Glasser et al. 1993) simulating the most challenging
scenario, because a low pH is generally needed for optimal POX activity.
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 281

Fig. 11.4 Graphical illustration of the adlerol (β-O-4 model compound, structure I) biocatalysis
based on LiP. Several degradation variants (partly in a summarized manner) are shown derived
from proposed reaction schemes by Tien and Kirk (1984), Kirk and Farrell (1987), Lundell et al.
(1993b) and Schoemaker et al. (1994b). I adlerol, II adlerol-derived radical cation, III veratryl
alcohol radical (VA), IV veratraldehyde (VAld) with 60–70 % major product, V Cβ-cation, VI
glycoaldehyde, VII guaiacol, VIII Cβ-centered radical, IX hemiacetal intermediate (unstable),
X keton, a second major product (15 %). Reprinted from Busse et al. (2013) with slight
modifications
282 N. Busse and P. Czermak

In Fig. 11.4, the basic adlerol degradation steps, induced by (fungal) heme per-
oxidases, are schematically explained in conjunction to the previously described,
(non)enzymatic reactions. The catalytic cycle is initiated by H2O2 to form EI as usual.
EI further reacts with adlerol (structure I) to EII, resulting in the production of an
adlerol-derived cation radical intermediate (structure II) (Lundell et al. 1993b). Once
the radical cation is generated, it will be rapidly fragmented, non-enzymatically
(Hatakka et al. 1991). For this, various theories have been under consideration, as
summarized in Fig. 11.4. The reason for this is most likely due to both (i) S•+
chemistry (as already mentioned above), and (ii) pH-dependent product distribution
(Schoemaker 1990). Despite the non-enzymatic reaction, veratraldehyde (VAld,
structure IV) is the main fission product in the end. This same procedure is repeated
when another adlerol molecule is oxidized by EII (refer to pathway 3, Fig. 11.4).
In case adlerol will not serve as the reducing substrate for some reason, com-
ponent II (S•+), III (VA•), and VII (guaiacol) could act as substrates for driving the
cycle back to E0. The latter causes polymerization of its radical products (Chance
and Maehly 1955), which are phenoxy radicals (Lundell et al. 1993b). A reduction
of EII–E0 by component III also results in veratraldehyde (VAld) formation
(Schoemaker 1990; Schoemaker et al. 1994b). However, this is only an occurance
under low-oxygen conditions (Schoemaker et al. 1994b), since the existence of III
is supposed to be short-lived by reacting with O2 immediately. It is important here
to remember that the mediator function of radical products of component III can
cause co-oxidation (thus degradation) of adlerol as well. It is believed that efficient
mediation can only be realized if the degrading substrate will bind close to the
active site of the enzyme (Schoemaker et al. 1994b).
Under substrate (adlerol) limited conditions, Tien and Kirk (1984) demonstrated
a non-stereospecifity of their used LiP, due to complete cleavage of the β-O-4 lignin
model compound. This is an important feature, because lignin has a highly irregular
structure (Schoemaker 1990). Conversely, the involvement of nonenzymatic radical
(chain) reactions is obvious in assisting enzymatic degradation.

11.3.3 VP Reaction and Deactivation Kinetics

Up until now, the normal POX cycle has been introduced (pathway 1–3 in
Figs. 11.2, 11.4). In Fig. 11.5, the supposed VP reaction mechanism is summa-
rized. EI and EII in Fig. 11.5 are expected to be tryptophan radical (Trp•) containing
proteins. Furthermore, this tryptophan radical is suggested to be located on the
enzyme surface (≈10–11 Å to the heme center (Pogni et al. 2005; Ruiz-Dueñas
et al. 2009a)) for substrate oxidation purposes. This means there is no direct contact
between the heme (Fe3+) co-factor and the reducing aromatic substrates (e.g., VA
(non-phenolic monomer) or complex lignin-derived polymers (macromolecules))
avoiding steric hindrance. Interactions between substrate and heme group are
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 283

Fig. 11.5 Supposed H2O2-dependent reaction mechanism of the crude VP from B. adusta for
adlerol degradation. E0–EIII are enzyme intermediates. The symbols S and S•+ stand for an
appropriate substrate (e.g., adlerol) and the corresponding radical cation. O•−
2 denotes superoxide
radical anions. Pathway 1–3 depicts the usual POX reaction cycle with reversible steps, as
described by equation (a) and (b) (Ruiz-Dueñas et al. 2009b; Busse et al. 2013). The cycle will be
initiated by H2O2 (pathway 1). Pathway 4, 5, and 7 show important side reactions with H2O2. The
latter is in competition with pathway 6. Enzyme deactivation (Ei: inactivated enzyme) is found to
occur via EIII, as sketched in pathway 7. Pathway 8 illustrates a spontaneous unimolecular decay of
EIII. Reprinted from Busse et al. (2013)

enabled via long-range electron transfer (LRET) (Ruiz-Dueñas et al. 2009a). The
involvement of LRET in lignin degradation processes through ligninolytic enzymes
is generally accepted (Busse et al. 2013).
Pathway 1 is the only reaction which has been understood in the past (Dunford
2010), and therefore particular evidence for pathway 3 is still pending (Busse et al.
2013). Thus, for the adlerol bioconversion by a VP type from B. adusta, pathway 3
in Fig. 11.5 was assumed based on transient state kinetics made through VA oxi-
dation studies using a recombinant VP from E. coli (Ruiz-Dueñas et al. 2009b).
Moreover, reversible binary enzyme–substrate (E–S) complex (precursor complex
(Dunford 2010)) formation that occur before irreversible product release are cer-
tainly possible during the catalytic cycle of VP (concerns pathway 2 and 3,
Fig. 11.5) (Ruiz-Dueñas et al. 2009b). Both EI and EII exhibit reactions described
by Eqs. (11.12, 11.13) (Ruiz-Dueñas and Martínez 2009; Ruiz-Dueñas et al.
2009b).
284 N. Busse and P. Czermak

$ E  S ! E ½Fe
Km2 k2
EI ½Fe4 þ ¼ OTrp  þ S I II 3þ
Trp  þ S þ ð11:12Þ

Trp  þ S $ E  S ! E ½Fe
Km3 k3
EII ½Fe3 þ  II 0 3þ
 þ S þ ð11:13Þ

Kmi and ki (i = 2, 3) are the corresponding equilibrium dissociation constant and


rate constant, respectively. As a result, the kinetics of VP form a two substrate
ping–pong bi bi mechanism with the reaction rate r explained in Eq. (11.14) (Busse
et al. 2013).

vmax ½S1 ½S2 


r¼ ð11:14Þ
KmS1 ½S2  þ KmS2 ½S1  þ ½S1 ½S2 

vmax, S1, S2, KmS1 , and KmS2 stand for maximum reaction velocity, co-substrate H2O2,
substrate S (e.g., adlerol), dissociation constant for S1 and dissociation constant
for S2.
Maintaining constant S2 concentration, while varying S1, Eq. (11.14) can be
simplified to Eq. (11.15).

max ½S1 
vapp
r¼ ð11:15Þ
Kmapp þ ½S1 

vmax ½S2 
max ¼
vapp ð11:16Þ
KmS2 þ ½S2 

KmS1 ½S2 
Kmapp ¼ ð11:17Þ
KmS2 þ ½S2 

vapp app
max and Km are the apparent kinetic parameters of maximum reaction velocity and
dissociation constant, respectively. Equations (11.14)–(11.17) are valid in the
absence of inhibitions/deactivation phenomena and product(s) (Bisswanger 2008;
Cornish-Bowden 2012). Due to the well-known H2O2-sensitivity of POX,
inactivation/deactivation reactions are substantial side effects of crucial importance.
Figure 11.5 concerns the major reaction pathways that have a co-substrate H2O2 in
addition to substrate S. Both substrates compete for reactions with EI and EII (refer
to Fig. 11.5 pathway 2, 4, and 3, 5, respectively). When a certain S/H2O2/E ratio is
unbalanced, POX deviate from their normal catalytic cycle, and instead promote
reactions via compound III (EIII), diminishing enzyme activity (pathway 5, 7)
(Arnao et al. 1990a, b; Wariishi and Gold 1990; Busse et al. 2013). EIII is another
third enzyme intermediate and exhibits non, or sometimes barley any catalytic
activity (Schoemaker 1990). Subsequently, EIII can react further with H2O2,
resulting in an enzyme deactivation Ei (pathway 7 (Wariishi and Gold 1990;
Goodwin et al. 1994; Busse et al. 2013)), also known as suicide inactivation
(Valderrama et al. 2002). Research investigations have shown that VP deactivation
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 285

follows a time-dependent irrversible mechanism (Vlasits et al. 2010; Busse et al.


2013). Assuming first order kinetics (Eq. 11.18), and that no substrate will be
present, the observed deactivation constant kd(obs) can be determined according to
Eq. (11.19) (Busse et al. 2013).

!E
kdðobsÞ
E i ð11:18Þ

kiapp ½S1 
kdðobsÞ ¼ ð11:19Þ
Kiapp ½S1 

kiapp and Kiapp denote the apparent maximal rate of enzyme inactivation and the
apparent concentration of inhibitor for reaching half-maximal rate of inactivation,
respectively.
In the presence of substrate S, another competition with H2O2 occurs, since EIII
may also react with S by returning back to E0 (pathway 6, Fig. 11.5) under radical
cation intermediate and H2O2 formation (assumed based on Yokota and Yamazaki
(1965) and Acosta et al. (1988)). Alternatively, radical cation products S•+ are able to
revert EIII–E0. However, it does not necessarily mean that all potential substrates are
suitable for such EIII conversion. On the contrary, for a LiP H2 isozyme, for instance,
neither phenolics nor their corresponding phenoxyl radical products nor VA were
capable of returning EIII back to the native state E0, but VA•+ worked fine (Chung
and Aust 1995). The validity of VP and adlerol and its radical cation products still
needs to be clarified. Whatever the case, once an appropriate substrate like adlerol is
added, H2O2-dependent enzyme deactivation is competitively inhibited, and
Eq. (11.19) yields Eq. (11.20). This implies that the substrate does in fact possess
protective properties (Busse et al. 2013). For illustration refer to Fig. 11.6.

kapp ½S1 
kdðobsÞ ¼  i  ð11:20Þ
Kiapp 1 þ ½SS22 þ ½S1 
Km

Depending on the environmental circumstances, e.g., the used buffer system, the
right term in Eqs. (11.19) and (11.20) must be extended by summation of k0i
(Copeland 2002). Thus, k0i denotes the enzyme inactivation rate caused by other
factors apart from H2O2. Furthermore, reaction pathway 5 and 7 (Fig. 11.5), and thus
enzyme deactivation, are pH-dependent and will be favored by an acidic environ-
ment (Cai and Tien 1992; Busse et al. 2013). VPs, and all other POX, commonly
have their reaction optimum around pH 3–4 (Wong 2009; Hofrichter et al. 2010;
Liers et al. 2010; Busse et al. 2013) and are consequently quite susceptible to H2O2
(catalytic amounts of > 30–50 µM (Goodwin et al. 1994; Busse et al. 2013)). This is
a major drawback of POX compared to Lac, particularly when attempting to reach
industrial applications, as will be discussed next. Spontaneous decay of EI (is
unstable (Dunford 2010)) to EII contributes to additional loss in activity (reaction
path is omitted in Fig. 11.5). The unimolecular reaction described by pathway 8
286 N. Busse and P. Czermak

Fig. 11.6 H2O2-dependent deactivation kinetics in the absence of the substrate adlerol
(experimental data (squares), computer simulation (red solid line)) and in the presence of
4 mM adlerol (experimental data (triangles), computer simulation (black solid line)). Reaction
conditions: T: 30 ± 1 °C, pH: 4.0 (100 mM sodium tartrate buffer), n: 375 rpm, enzyme
concentration: *0.06 mg mL−1, V: 10 mL or 50 mL. Each examination was carried out in
duplicates (parallel) with a maximal standard deviation of 10 % from the mean. The inset plot
illustrates the corresponding half-life (t1/2) course as a function of initial H2O2 concentration.
Reprinted from Busse et al. (2013)

(Fig. 11.5), EIII decay to E0 along with O•−2 release, (Nakajima and Yamazaki 1987;
Arnao et al. 1990b) would imply enzyme regeneration or loss of activity.
Finally, it is obvious that balancing the S/E/H2O2 ratio is an important task for
minimizing enzyme deactivations, and thus avoiding excessive H2O2 concentra-
tions. It is in fact impossible to fully eliminate enzyme deactivation, but it can be
delayed. Maintaining enzyme consumption as low as possible is therefore a major
tasks, since POX are currently still costly.

11.4 Technical Challenges for VP Applicability on Large


Scale

11.4.1 Ensuring Cost-Effective Availability and Stability

As already stated, POX and especially VP have received a lot of attention as


biocatalysts. Unfortunately, an industrial implementation has not yet be realized.
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 287

This progress is hampered by (i) low availability of active POX in sufficient


amounts, (ii) H2O2 related instabilities (Ayala et al. 2008), and (iii) high enzyme
costs (Casella et al. 2010). Therefore, major efforts are directed toward protein
engineering for improving enzyme performance, particularly stability with respect
to H2O2, catalytic activity by redox potential enhancement, and ensuring avail-
ability by efficient homologous and heterologous (over)production (Conesa et al.
2000; Ayala et al. 2008; Longoria et al. 2010). As potential expression systems,
bacteria (E. coli), yeast (S. cerevisiae, Pichia) and fungi (white-rot fungi,
Aspergillus) are all under consideration (Conesa et al. 2000; de Weert and Lokman
2010). Moreover, active POX production in insect cells using recombinant bac-
ulovirus has been examined (Pease et al. 1991; Johnson et al. 1992). However, to
date, none of the investigated homologous and heterologous production approaches
have matched industrial requirements (de Weert and Lokman 2010). Although
homologous expression shows promising results, the most frequent cause of failure
is the instability in productivity, which is not yet fully understood (Singh and Chen
2008). Further research and development is still needed. An efficient POX-specific
production is dependent on (i) choice of production strain (Conesa et al. 2000; de
Weert and Lokman 2010), (ii) vector system (de Weert and Lokman 2010), and
(iii) culture/operating conditions (Conesa et al. 2000; Rühl et al. 2008) and inclu-
sive production plant configuration (Rühl et al. 2007).
MnP overproduction in heterologous systems might generally be easier than for
LiP and VP. It is conceivable that due to their high reactivity to a broader substrate
spectrum, production of active LiP and VP, is comparatively more harmful to host
organisms (Tsukihara et al. 2006). Table 11.2 lists host organisms for VP pro-
duction that have been studied. Here, the homologous expression system is believed
to provide a promising platform for large-scale production, resulting in recombinant
enzymes exhibiting structural and kinetic properties identical to the native one
(Tsukihara et al. 2006). In general, P. ostreatus strains are well suited for industrial
production of ligninolytic enzymes (Rühl et al. 2008).

Table 11.2 Hosts for homologous and heterologous VP production (modified based on de Weert
and Lokman (2010))
Organism Host strain Yield References
Homologous
P. ostreatus P. ostreatus TM2-18 21 mg L-1 Tsukihara et al.
7300 U L-1 (2006)
Heterologous
B. adusta E. coli strain BL21 *12 mg L−1 with max. Mohorčič et al.
(DE3)pLysS 0.25 mU mg−1a (2009)
P. eryngii Aspergillus nidulans 466 U L−1 Eibes et al.
(2009)
a
Specific activity was determined based on VA degradation
288 N. Busse and P. Czermak

11.4.2 Supply of Lignocellulosic Biomass

In using lignocellulosic biomass as a substrate, an appropriate pretreatment is


mandatory before an enzymatic bioconversion can be addressed. For reaching
satisfactory reaction rates with the smallest possible enzyme amount, or in other
words, for reaching high turnover numbers, wood substrates with high substrate
specific surfaces (high surface to volume ratios) are favored. The minimal
requirement is that native lignin must be readily accessible for enzymatic attack.
Ground wood/wood flour provides the best preconditions, although a relative high
energy demand is required depending on the particle size. Particle size is limiting
for mass transfer (Viamajala et al. 2010). Furthermore, native lignin structures have
to be conserved. Since lignin is an amorphous thermoplastic polymer (Saake and
Lehnen 2012), the grinding temperature should be maintained below its glass
transition temperature (Tg) (range hard-/soft-wood: 65–105 °C (Saake and Lehnen
2012)). Melting lignin would result in undesired and uncontrolled modifications
(structural changes, lignin relocalization) influencing subsequent enzymatic con-
version (Donohoe et al. 2008; Viamajala et al. 2010). The same is also valid for
pretreating methods reaching temperatures above Tg.
Inconsistency of wood substrate, particularly lignocellulose composition, is
another variable that interferes with enzyme activity and product quantity as well as
quality. Hybrid poplar (e.g., made of Populus maximowiczii and Populus nigra)
from short-rotation forestry is a future based raw material since it is fast-growing.
Its use means variations in wood composition will be severely minimized.

11.4.3 Supply of Lignin-Containing Product Streams

Liquid lignin product streams (technical lignins) generated via chemical pretreat-
ment of lignocellulosic biomass, for instance, is an alternative substrate for further
processing and finishing by POX. The initial chemical oxidation should preferably
lead to lower molecular weight and more biodegradable intermediates (Mantzavinos
and Psillakis 2004). However, it has to be taken into account that such a pretreatment
can also generate inhibitory products. Moreover, the pH may have to be adjusted.
This might be difficult to achieve. Depending on the isolation method, the resulting
technical lignin may not be soluble at an acidic pH. In general, most technical lignins
are soluble in organic solvents (e.g., dimethyl sulfoxide (DMSO), dioxane,
dimethylformamide, and pyridine) (Saake and Lehnen 2012). VP remains active in
organic solvents like DMSO, at least in the presence of an inorganic substrate
(Rodakiewicz-Nowak et al. 2006). Nonetheless, with the goal of reaching industrial
application, further addition of organic solvents may be uneconomic and in fact
problematic, relating to the recovery of the organic solvent as well as resulting
ecological aspects.
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 289

11.4.4 Bioprocess Engineering for Delignification

For the provision of optimal operating conditions (e.g., T, buffer system, S/H2O2/E
ratio, presence of additives, operation mode), the choice of an appropriate reactor
design is an important issue of bioprocess engineering in order to guarantee the
economic implementation of VP and POX for delignification purposes.

11.4.4.1 Reactor Design

For a rational reactor design, several aspects must be taken into account. Due to
complex and rather uncontrollable radical reactions as seen above, such aspects
include first and foremost an efficient maximization of catalytic turnover numbers
(kcat = vmax/[ET]) in order to avoid an excess of disruptive factors like increased H2O2
and phenol concentrations. Phenols cause polymerization and formation of phenoxyl
radicals affecting the catalytic cycle. A continuous removal of such compounds is
therefore highly recommended. Moreover, reuse of the enzyme is also beneficial.
Combining the enzymatic conversion process by connecting an external cross-
flow ultrafiltration (UF) unit to an enzyme membrane reactor system, as sketched in
Fig. 11.7, represents a promising technological approach for delignification
(Flaschel et al. 1983; Lema et al. 2010; Huang and Ramaswamy 2013). Such
configurations provide several important advantages:
(i) Continuous operation mode.
(ii) Complete biocatalyst retention and recycling.
(iii) Application of free/native enzyme is possible (homogeneous catalysis),
avoiding time-consuming enzyme immobilization accompanied by mass
transfer limitations (Busse et al. 2016; López et al. 2007).
(iv) Replacement of fresh active enzyme depending on deactivation in order to
maintain specified substrate conversion levels (Busse et al. 2016).
(v) Physical biocatalyst immobilization.
(vi) Rapid fractionation of biocatalyst and unreacted high molecular weight
substrates from desired product.
(vii) Continuous removal of undesired by-products and enzyme inhibitors, shift-
ing reaction toward product yield maximization (Busse et al. 2016; López
et al. 2007).
Membrane filtration processes like tubular UF membranes are already well estab-
lished for lignin recovery and separation in biorefineries (Jönsson 2013) and in the
pulp and paper industry (Ebrahimi et al. 2009). This is due to the following eco-
nomic aspects (Huang and Ramaswamy 2013; Jönsson 2013):
(i) Low chemical consumption.
(ii) Fractionation is controllable by pore size.
(iii) Feed volume reduction for further processing (Jönsson and Wimmerstedt 1985).
(iv) Any T and pH adjustments are superfluous.
290 N. Busse and P. Czermak

Despite the fact that tubular UF membranes exhibit low packing densities and high
energy demands (Leiknes 2009), they have “the largest foot-print” (Jönsson 2013).
Moreover, these membranes require low pretreatment (Jönsson 2013), tolerate feed
streams of high fouling potential and of high pollution (e.g., solids) and are easy to
clean (Melin and Rautenbach 2007; Leiknes 2009). Nevertheless, proteins are
serious membrane foulants (Blatt et al. 1970; Porter 1972; Field 1996), besides
lignin-containing feed streams (Sridhar and Bhattacharya 1991; Pfromm and
Watkins 1997; Ebrahimi et al. 2016; Busse et al. 2016). Consequently, constant
enzyme addition, and/or an increased amount of lignin-derived fragments, result in
secondary layer formation on the membrane surface (membrane fouling) limiting
filtration performance, e.g., permeability or throughput, solute retention (Busse
et al. 2016), and membrane service life. Fouling is a common issue in membrane
filtration processes and is mainly affected by (i) membrane properties (e.g., selected
pore size, material, configuration), (ii) hydrodynamic conditions, and (iii) feed
composition (Mulder 1996; Cheryan 1998; Melin and Rautenbach 2007). Filtration
performance is a crucial parameter, since it determines the efficiency of enzymatic
conversion, protein load, and reactor size, along with hydraulic retention time

Fig. 11.7 Schematic illustration of a promising enzyme membrane reactor configuration


(continuous stirred tank reactor (CSTR) equipped with an external ultrafiltration (UF) module)
for delignification purposes. Enzymatic reaction can be assumed to take place primarily in the
CSTR at constant reaction volume VR. F volumetric flow rate; c, ci concentration, concentration of
component i; VR reaction volume; t process time; ri reaction rate; J(t) time-dependent permeate
flux; AM membrane surface; ΔPTM transmembrane pressure; η dynamic viscosity of the permeate
depending on temperature T and concentration c; RT(t) total hydraulic resistance as a function of
time t due to membrane fouling
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 291

(refer to Fig. 11.7) (Busse et al. 2016). Thus, the maintenance of constant high
volumetric flow rates (or fluxes) is a major task throughout filtration processes.
Using ceramic membranes is advantageous in comparison to polymer mem-
branes. Major aspects are:
(i) High thermal and chemical stability which is beneficial for cleaning.
(ii) High mechanical stability, particularly when filtering abrasive media.
(iii) Less susceptible to fouling and low adhesion potential for molecular organic
substances (Ebrahimi et al. 2015; Fan et al. 2015).
(iv) High membrane service life/lifetime (6 years for ceramic membranes and 1.5
for polymeric) (Arkell et al. 2014).
Conversely, polymeric membranes are less expensive, easy to configure and scal-
able (Van der Bruggen 2009).
Several applications have already been reported using POX in membrane biore-
actor systems. However, these have just been for treating xenobiotics and pollutions
(e.g., azo dye Orange II by MnP, nonylphenol by VP), which use mostly polymeric
UF membranes (López et al. 2007; Méndez-Hernández et al. 2015). The choice for
the optimal material ultimately depends on individual needs (Arkell et al. 2014).

11.4.4.2 Process Control

Key prerequisite for implementing an effective process control, and thus ensuring
optimal operation condition, is an efficient and reliable online monitoring system
(except T and pH, since they are standard). Although H2O2 concentration is such a
crucial parameter, the availability of commercial H2O2 online sensors, with suffi-
cient sensitivity over a wide pH and T range, is limited. Contrarily, there are several
fluorescence probes, such as some based on oxidation of N-
acetyl-3,7-dihydroxy-phenoxazine (e.g., Amplex Red®) by HRP, for measuring
H2O2 within a range of 10 nM to 100 µM (Gomes et al. 2005). Disadvantages of
using such probes are: (i) sample preparation is required, (ii) HRP inactivation at c
(H2O2) > 100 µM (Towne et al. 2004), thus first requiring the finding of an
appropriate pre-dilution, (iii) incubation times of 15–30 min before receiving first
results, and (iv) pH is also limited, ranging from 7.5 to 8.5. Similar conditions are
true concerning any HRP/POX-related biosensor for H2O2 measurements. In the
current state, amperometric sensors equipped with platinum electrodes appear rather
suitable, since they are not susceptible to H2O2. Sample preparations are also
superfluous, among other things. Particularly, the membrane protected PEROX
H2O2 online sensors from ProMinent Dosiertechnik in Heidelberg, Germany, meet
useful requirements, including aspects such as a wide pH-/T- and c(H2O2)-range
with a comparatively low response time. First successful applications of this have
already been reported for chloroperoxidase catalyzed oxidations (Seelbach et al.
1997; van Deurzen et al. 1997).
Another important parameter is the actual enzyme activity, which could be
detected by simply monitoring the production of certain fission products and/or
292 N. Busse and P. Czermak

substrate consumption. When complex substrates such as lignin are involved and
product formation is hardly manageable, as already illustrated above, lignin degra-
dation is usually monitored by UV absorption measurements at λ = 280 nm. Highly
concentrated lignin solutions appear dark brown. The color intensity is an indirect
indicator of lignin amount (Kinstre 1993). Conversely, the release of lignin-derived
high molecular weight fragments from LC biomass would result in an intensification
of color in the culture medium. A precise assignment of these fragments is not
possible, and further analytical methods will be required which are limited in
applicability, partly very costly, and not yet sufficiently standardized. An extensive
overview will be given by Lin and Dence (1992). Moreover, a fundamental expertise
is mandatory for process optimization and data interpretation. At present, there is no
simple, standalone analytical method. Based on these facts, alternative (in)direct
measurement techniques must be found. Such techniques should be (i) cheap,
(ii) easy to implement, online, (iii) robust, (iv) quick and precise and (v) low main-
tenance. Measuring dissolved oxygen (DO), especially via optical sensors, is cur-
rently the most promising parameter for process monitoring due to pseudo-catalase
activity of POX at H2O2 excess (or low c(S)/c(H2O2) ratios) resulting in O2 formation
(Acosta et al. 1988; Wariishi and Gold 1989; Vlasits et al. 2010). Excessive c(H2O2)
causes considerable enzyme deactivation (Busse et al. 2013). On the contrary, O2
consumption is an indicator for high c(S)/c(H2O2) ratios (López et al. 2007). Thus, O2
consumption is also indicative for substrate degradation processes as seen in
Figs. 11.3 and 11.4, whereas O2 can also be generated simultaneously concerning
HO•2/O•− 2 release (Figs. 11.3 and 11.5). Recent studies by López et al. (2007), treating
xenobiotic compounds such as Organe II by MnP, have shown that DO could be
valuable for implementing process control strategies (e.g., feedback control).
However, based on the variety of radical reactions, O2 formation/consumption is not
trivial (Palmer et al. 1987). Further research is therefore needed for a better (process)
understanding, particularly in the field of delignification. Moreover, the presence of
substrates (e.g., VA and adlerol) capable of bond cleavage seems to be a precondition
for significant O2 consumption (Palmer et al. 1987).

11.5 Conclusion and Outlook

The high susceptibility to H2O2 of VP (POX in general) is an important issue which


needs to be overcome, as they are currently making wild type less attractive for
industrial use. In vivo relative low H2O2 levels are rather assumed due to H2O2
assimilation by the fungus avoiding unfavorable high concentrations (Hammel et al.
1993; Böckle et al. 1999). Moreover, VPs are regrettably limited in availability. Thus,
numerous efforts are being carried out for recombinant VP production, in order to
elevate H2O2 tolerance and availability. Alternatively, enzyme immobilization could
be another technique to overcome premature inactivation (Ei formation). However,
immobilization may not prevent EIII formations at H2O2 excess. Enzyme recondi-
tioning (EIII → E0, pathway 6 in Fig. 11.5) in turn is again influenced by the
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 293

underlying reaction conditions and not by the immobilization method. Despite sev-
eral studies, the proof-of-concept is still outstanding. Additionally, in the case of
woody biomass, good enzyme penetration must be further guaranteed, affected by
porosity and the pore size, of the substrate to be treated.
Another promising/future based strategy may be enzymes from insects and
insect-associated microorganisms. Insects are the most diverse taxonomic animal
class, colonizing almost every ecological niche. To succeed in these sometimes
extreme niches, insects have established diverse biological and chemical systems,
e.g., the production of defense molecules (Gross et al. 2008; Schlipalius et al. 2012),
protein stabilization (Bale 2002), or lytic enzymes (Landureau and Jolles 1970; Mika
et al. 2013). Insects also house symbiotic microorganisms as digestive helpers,
fodder, or both (Scharf et al. 2011). This is possible due to the special metabolic
systems, or exogenous enzymes, of the associated organisms. For instance, insects
like bark beetles, ambrosia beetles and termites, are able to feed on wood, making
them xylophagous, and are well known as wood pests. The function of their
digestive systems is not yet completely clarified. The enzymatic apparatus for the
oxidation of lignin and the hydrolysis of cellulose is only marginally understood.
One reason might be the diverse sources of the key enzymes, which may be pro-
duced either by gut-inhabiting microorganisms (Morrison et al. 2009), by symbiotic
fungi cultivated by the insects (Currie 2001), or by the insects themselves as
endogenous enzymes (Pauchet et al. 2010). Nevertheless, a lot of work has been
done in attempts to understand the lignocellulolytic system of the insects and the
degree of participation by symbionts (Geib et al. 2008; Scharf et al. 2011).
Currently, POX are still costly for industrial use. Nonetheless, there is no doubt
that once limitations are resolved, particularly H2O2 instability and efficient pro-
duction, the potential for large-scale application of VPs raises significantly (Casella
et al. 2010). However, many questions with respect to the enzymatic delignification
mechanism still remain in place, i.e., substrate preparation/pretreatment, E0
recovery, control of inhibitory reactions/factors besides H2O2, involvement of
accessory factors and enzymes, etc. Moreover, for better process control, devel-
opments focusing on simple and fast online monitoring systems will also be nec-
essary. For these purposes, further fundamental research is needed.

Acknowledgments The researchers would like to thank the Hessen State Ministry of Higher
Education, Research and the Arts for the financial support within the Hessen initiative for scientific
and economic excellence (LOEWE).

References

Acosta M, del Río JA, Arnao MB, Sánchez-Bravo J, Sabater F, García-Carmona F,


García-Cánovas F (1988) Oxygen consumption and enzyme inactivation in the
indolyl-3-acetic acid oxidation catalyzed by peroxidase. Biochim Biophys Acta
(BBA) Protein Struct Mol Enzymol 955(2):194–202. doi:10.1016/0167-4838(88)90193-8
Adler E (1977) Lignin chemistry—past, present and future. Wood Sci Technol 11(3):169–218.
doi:10.1007/BF00365615
294 N. Busse and P. Czermak

Andrawis A, Johnson KA, Tien M (1988) Studies on compound I formation of the lignin
peroxidase from Phanerochaete chrysosporium. J Biol Chem 263(3):1195–1198
Arkell A, Olsson J, Wallberg O (2014) Process performance in lignin separation from softwood
black liquor by membrane filtration. Chem Eng Res Des 92(9):1792–1800. doi:10.1016/j.
cherd.2013.12.018
Arnao MB, Acosta M, del Rio JA (1038) García-Cánovas F (1990a) Inactivation of peroxidase by
hydrogen peroxide and its protection by a reductant agent. Biochim Biophys Acta
(BBA) Protein Struct Mol Enzymol 1:85–89. doi:10.1016/0167-4838(90)90014-7
Arnao MB, Acosta M, del Río JA, Varón R (1041) García-Cánovas F (1990b) A kinetic study on
the suicide inactivation of peroxidase by hydrogen peroxide. Biochim Biophys Acta Protein
(BBA) Struct Mol Enzymol 1:43–47. doi:10.1016/0167-4838(90)90120-5
Ayala M (2010) Redox potential of peroxidases. In: Torres E, Ayala M (eds) Biocatalysis based on
heme peroxidases. Springer, Berlin, pp 61–77. doi:10.1007/978-3-642-12627-7_4. ISBN
978-3-642-12626-0
Ayala M, Pickard MA, Vazquez-Duhalt R (2008) Fungal enzymes for environmental purposes, a
molecular biology challenge. J Mol Microbiol Biotechnol 15(2–3):172–180. doi:10.1159/
000121328
Bale JS (2002) Insects and low temperatures: from molecular biology to distributions and
abundance. Philos Trans B 357(1423):849–862. doi:10.1098/rstb.2002.1074
Bielski BH, Cabelli DE, Ravindra LA, Ross AB (1985) Reactivity of HO2/O2-radicals in aqueous
solution. J Phys Chem Ref Data 14(4):1041–1100
Bisswanger H (2008) Enzyme kinetics: section 2. In: Enzyme kinetics. Wiley-VCH Verlag GmbH
& Co. KGaA, pp 124–193. ISBN 9783527319572
Blanchette RA (1991) Delignification by wood-decay fungi. Annu Rev Phytopathol 29(1):
381–403. doi:10.1146/annurev.py.29.090191.002121
Blatt WF, Dravid A, Michaels AS, Nelsen L (1970) Solute polarization and cake formation in
membrane ultrafiltration: causes, consequences, and control techniques. In: Flinn JE
(ed) Membrane science and technology. Springer, USA, pp 47–97. doi:10.1007/978-1-4684-
1851-4_4. ISBN 978-1-4684-1853-8 (print), 978-1-4684-1851-4 (online)
Böckle B, Martínez MJ, Guillén F, Martínez ÁT (1999) Mechanism of peroxidase inactivation in
liquid cultures of the ligninolytic fungus Pleurotus pulmonarius. Appl Environ Microbiol 65
(3):923–928
Bourbonnais R, Paice MG (1990) Oxidation of non-phenolic substrates: an expanded role for laccase
in lignin biodegradation. FEBS Lett 267(1):99–102. doi:10.1016/0014-5793(90)80298-W
Busse N, Wagner D, Kraume M, Czermak P (2013) Reaction kinetics of versatile peroxidase for
the degradation of lignin compounds. Am J Biochem Biotechnol 9(4):365–394. doi:10.3844/
ajbbsp.2013.365.394
Busse N, Fuchs F, Kraume M, Czermak P (2016) Treatment of enzyme initiated delignification
reaction mixtures with ceramic ultrafiltration membranes: Experimental investigations and
modeling approach. Sep Sci Technol 51(9):1546–1565. doi:10.1080/01496395.2016.1167739
Cai D, Tien M (1992) Kinetic studies on the formation and decomposition of compounds II and
III. Reactions of lignin peroxidase with H2O2. J Biol Chem 267(16):11149–11155
Camarero S, Sarkar S, Ruiz-Dueñas FJ, Martınez ́ MJ, Martínez ÁT (1999) Description of a
versatile peroxidase involved in the natural degradation of lignin that has both manganese
peroxidase and lignin peroxidase substrate interaction sites. J Biol Chem 274(15):10324–
10330. doi:10.1074/jbc.274.15.10324
Cañas AI, Camarero S (2010) Laccases and their natural mediators: biotechnological tools for
sustainable eco-friendly processes. Biotechnol Adv 28(6):694–705. doi:10.1016/j.biotechadv.
2010.05.002
Carabajal M, Kellner H, Levin L, Jehmlich N, Hofrichter M, Ullrich R (2013) The secretome of
Trametes versicolor grown on tomato juice medium and purification of the secreted
oxidoreductases including a versatile peroxidase. J Biotechnol 168(1):15–23. doi:10.1016/j.
jbiotec.2013.08.007
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 295

Casella L, Monzani E, Nicolis S (2010) Potential applications of peroxidases in the fine chemical
industries. In: Torres E, Ayala M (eds) Biocatalysis based on heme peroxidases. Springer,
Berlin, pp 111–153. doi:10.1007/978-3-642-12627-7_6. ISBN 978-3-642-12626-0
Chance B, Maehly AC (1955) Assay of catalases and peroxidases. In: Methods in enzymology, vol
2. Academic Press, London, pp 764–775. doi: 10.1016/S0076-6879(55)02300-8
Cheryan M (1998) Ultrafiltration and microfiltration handbook. CRC Press, London. ISBN
978-1-56676-598-5
Chung N, Aust SD (1995) Inactivation of lignin peroxidase by hydrogen peroxide during the
oxidation of phenols. Arch Biochem Biophys 316(2):851–855. doi:10.1006/abbi.1995.1114
Conesa A, van den Hondel CAMJJ, Punt PJ (2000) Studies on the production of fungal
peroxidases in Aspergillus niger. Appl Environ Microbiol 66(7):3016–3023. doi:10.1128/aem.
66.7.3016-3023.2000
Copeland RA (2002) Enzymes: a practical introduction to structure, mechanism, and data analysis.
Wiley-VCH, London. ISBN 0-471-35929-7, 0-471-22063-9
Cornish-Bowden A (2012) Fundamentals of enzyme kinetics. Wiley-Blackwell, London. ISBN
9783527330744
Currie CR (2001) A community of ants, fungi, and bacteria: a multilateral approach to studying
symbiosis. Annu Rev Microbiol 55(1):357–380. doi:10.1146/annurev.micro.55.1.357
Dashtban M, Schraft H, Syed TA, Qin W (2010) Fungal biodegradation and enzymatic
modification of lignin. Int J Biochem Mol Biol 1(1):36–50
de Montellano PRO (2010) Catalytic mechanisms of heme peroxidases. In: Torres E, Ayala M
(eds) Biocatalysis based on heme peroxidases. Springer, Berlin, pp 79–107. doi:10.1007/978-
3-642-12627-7_5. ISBN 978-3-642-12626-0
de Weert S, Lokman BC (2010) Heterologous expression of peroxidases. In: Torres E, Ayala M
(eds) Biocatalysis based on heme peroxidases. Springer, Berlin, pp 315–333. doi:10.1007/978-
3-642-12627-7_12. ISBN 978-3-642-12626-0
Donohoe BS, Decker SR, Tucker MP, Himmel ME, Vinzant TB (2008) Visualizing lignin
coalescence and migration through maize cell walls following thermochemical pretreatment.
Biotechnol Bioeng 101(5):913–925. doi:10.1002/bit.21959
Dunford HB (1991) Horsradish peroxidase: structure and kinetic properties. In: Everse J
(ed) Peroxidases in chemistry and biology, vol 2. CRC Press, London, pp 1–24. ISBN
0849369649
Dunford HB (2010) Peroxidases and catalases: biochemistry, biophysics, biotechnology and
physiology. Wiley, London. ISBN 9780470224762
Ebrahimi M, Kerker S, Wienold A, Neul H, Hilpert M, Mund P, Czermak P (2009) Processing and
characterization of ceramic membranes for the efficient removal of COD and residual lignin
from bleaching effluents. International conference and exhibition for filtration and separation
technology. Filtech Wiesbaden, Germany
Ebrahimi M, Busse N, Kerker S, Schmitz O, Hilpert M, Czermak P (2016) Treatment of
the bleaching effluent from sulfite pulp production by ceramic membrane filtration. Membranes
6(1). doi:10.3390/membranes6010007
Ebrahimi M, Kerker S, Daume S, Geile M, Ehlen F, Unger I, Schütz S, Czermak P (2015)
Innovative ceramic hollow fiber membranes for recycling/reuse of oilfield produced water.
Desalination Water Treat 55(13):3554–3567. doi:10.1080/19443994.2014.947780
Eibes GM, Lú-Chau TA, Ruiz-Dueñas FJ, Feijoo G, Martínez MJ, Martínez AT, Lema JM (2009)
Effect of culture temperature on the heterologous expression of Pleurotus eryngii versatile
peroxidase in Aspergillus hosts. Bioprocess Biosyst Eng 32(1):129–134. doi:10.1007/s00449-
008-0231-7
Eriksson K-EL, Blanchette RA, Ander P (1990) Microbial and enzymatic degradation of wood and
wood components. Springer, Berlin. ISBN 354051600X
Fan R, Ebrahimi M, Quitmann H, Czermak P (2015) Lactic acid production in a membrane
bioreactor system with thermophilic Bacillus coagulans: fouling analysis of the used ceramic
membranes. Sep Sci Technol 50(14):2177–2189. doi:10.1080/01496395.2015.1031401
296 N. Busse and P. Czermak

Field RW (1996) Mass transport and the design of membrane systems. In: Scott K, Hughes R
(eds) Industrial membrane separation technology. Springer, Netherlands, pp 67–113. doi:10.
1007/978-94-011-0627-6_4
Flaschel E, Wandrey C, Kula M-R (1983) Ultrafiltration for the separation of biocatalysts.
Downstream processing. Springer, Berlin, pp 73–142. doi:10.1007/978-3-662-39694-0_2
Geib SM, Filley TR, Hatcher PG, Hoover K, Carlson JE, Jimenez-Gasco MdM, Nakagawa-Izumi
A, Sleighter RL, Tien M (2008) Lignin degradation in wood-feeding insects. Proc Natl Acad
Sci 105(35):12932–12937. doi:10.1073/pnas.0805257105
Glasser WG, Davé V, Frazier CE (1993) Molecular weight distribution of (semi-) commercial
lignin derivatives. J Wood Chem Technol 13(4):545–559. doi:10.1080/02773819308020533
Gomes A, Fernandes E, Lima JLFC (2005) Fluorescence probes used for detection of reactive
oxygen species. J Biochem Biophys Methods 65(2–3):45–80. doi:10.1016/j.jbbm.2005.10.003
Goodwin DC, Barr DP, Aust SD, Grover TA (1994) The role of oxalate in lignin
peroxidase-catalyzed reduction: protection from compound III accumulation. Arch Biochem
Biophys 315(2):267–272. doi:10.1006/abbi.1994.1499
Gross J, Schumacher K, Schmidtberg H, Vilcinskas A (2008) Protected by fumigants: beetle
perfumes in antimicrobial defense. J Chem Ecol 34(2):179–188. doi:10.1007/s10886-007-
9416-9
Halliwell B, Gutteridge JMC (1985) The importance of free radicals and catalytic metal ions in
human diseases. Mol Aspects Med 8(2):89–193. doi:10.1016/0098-2997(85)90001-9
Hammel KE, Jensen KA, Mozuch MD, Landucci LL, Tien M, Pease EA (1993) Ligninolysis by a
purified lignin peroxidase. J Biol Chem 268(17):12274–12281
Harman LS, Carver DK, Schreiber J, Mason RP (1986) One- and two-electron oxidation of
reduced glutathione by peroxidases. J Biol Chem 261(4):1642–1648
Hatakka A (1994) Lignin-modifying enzymes from selected white-rot fungi: production and role
from in lignin degradation. FEMS Microbiol Rev 13(2–3):125–135. doi:10.1111/j.1574-6976.
1994.tb00039.x
Hatakka A (2005) Biodegradation of lignin. In: Biopolymers online. Wiley-VCH Verlag GmbH &
Co. KGaA. doi:10.1002/3527600035.bpol1005. ISBN 9783527600038
Hatakka A, Hammel K (2011) Fungal biodegradation of lignocelluloses. In: Hofrichter M
(ed) Industrial applications, vol 10. Springer, Berlin, pp 319–340. doi:10.1007/978-3-642-
11458-8_15. ISBN 978-3-642-11457-1
Hatakka A, Lundell T, Tervilä-Wilo ALM, Brunow G (1991) Metabolism of non-phenolic β-O-4
lignin model compounds by the white-rot fungus Phlebia radiata. Appl Microbiol Biotechnol
36(2):270–277. doi:10.1007/BF00164433
Hofrichter M, Ullrich R, Pecyna M, Liers C, Lundell T (2010) New and classic families of secreted
fungal heme peroxidases. Appl Microbiol Biotechnol 87(3):871–897. doi:10.1007/s00253-
010-2633-0
Holladay J, White J, Bozell J, Johnson D (2007) Top value-added chemicals from biomass—
volume II—results of screening for potential candidates from biorefinery lignin Pacific
Northwest National Laboratory. Richland, WA, U.S. Department of Energy, 87
Howard RL, Abotsi E, Jansen van Rensburg EL, Howard S (2003) Lignocellulose biotechnology:
issues of bioconversion and enzyme production. Afr J Biotechnol 2(12):602–619
Huang H-J, Ramaswamy S (2013) Overview of biomass conversion processes and separation and
purification technologies in biorefineries. In: Ramaswamy S, Huang H-J, Ramarao BV
(eds) Separation and purification technologies in biorefineries. Wiley, London, pp 3–36. ISBN
978-0-470-97796-5 (Hardcover)
Johnson TM, Pease EA, Li JKK, Tien M (1992) Production and characterization of recombinant
lignin peroxidase isozyme H2 from Phanerochaete chrysosporium using recombinant
baculovirus. Arch Biochem Biophys 296(2):660–666. doi:10.1016/0003-9861(92)90624-6
Jönsson A-S (2013) Microfiltration, ultrafiltration and diafiltration. In: Ramaswamy S, Huang H-J,
Ramarao BV (eds) Separation and purification technologies in biorefineries. Wiley, London,
pp 205–231. ISBN 978-0-470-97796-5 (Hardcover)
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 297

Jönsson AS, Wimmerstedt R (1985) The application of membrane technology in the pulp and
paper industry. Desalination 53(1–3):181–196. doi:10.1016/0011-9164(85)85060-8
Kawai S, Asukai M, Ohya N, Okita K, Ito T, Ohashi H (1999) Degradation of a non-phenolic
β-O-4 substructure and of polymeric lignin model compounds by laccase of Coriolus
versicolor in the presence of 1-hydroxybenzotriazole. FEMS Microbiol Lett 170(1):51–57.
doi:10.1111/j.1574-6968.1999.tb13354.x
Kettle AJ, Anderson RF, Hampton MB, Winterbourn CC (2007) Reactions of superoxide with
myeloperoxidase. Biochemistry 46(16):4888–4897. doi:10.1021/bi602587k
Kinstre RB (1993) An overview of strategies for reducing the environmental impact of
bleach-plant effluents. Tappi J 76(5):105–113
Kirk TK, Farrell RL (1987) Enzymatic “combustion”: the microbial degradation of lignin. Annu
Rev Microbiol 41:465–505
Landureau JC, Jolles P (1970) Lytic enzyme produced in vitro by insect cells: lysozymelor
chitinase? Nature 225(5236):968–969. doi:10.1038/225968a0
Lange J-P (2007) Lignocellulose conversion: an introduction to chemistry, process and economics.
Biofuels Bioprod Biorefin 1(1):39–48. doi:10.1002/9783527621118.ch2
Leiknes T (2009) Wastewater treatment by membrane bioreactors. In: Membrane operations.
Wiley-VCH Verlag GmbH & Co. KGaA, pp 363–395. doi: 10.1002/9783527626779.ch16.
ISBN 9783527626779
Lema J, López C, Eibes G, Taboada-Puig R, Moreira MT, Feijoo G (2010) Reactor
engineering. In: Torres E, Ayala M (eds) Biocatalysis based on heme peroxidases. Springer,
Berlin, pp 245–290. doi:10.1007/978-3-642-12627-7_10. ISBN 978-3-642-12626-0
Liers C, Bobeth C, Pecyna M, Ullrich R, Hofrichter M (2010) DyP-like peroxidases of the jelly
fungus Auricularia auricula-judae oxidize nonphenolic lignin model compounds and
high-redox potential dyes. Appl Microbiol Biotechnol 85(6):1869–1879. doi:10.1007/
s00253-009-2173-7
Lin SY, Dence CW (1992) Methods in lignin chemistry. Springer, Berlin. doi:10.1007/978-3-642-
74065-7. ISBN 9783642740671
Longoria A, Tinoco R, Torres E (2010) Enzyme technology of peroxidases: immobilization,
chemical and genetic modification. In: Torres E, Ayala M (eds) Biocatalysis based on heme
peroxidases. Springer, Berlin, pp 209–243. doi:10.1007/978-3-642-12627-7_9. ISBN
978-3-642-12626-0
López C, Moreira MT, Feijoo G, Lema JM (2007) Dynamic modeling of an enzymatic membrane
reactor for the treatment of xenobiotic compounds. Biotechnol Bioeng 97(5):1128–1137.
doi:10.1002/bit.21311
Lundell T, Schoemaker H, Hatakka A, Brunow G (1993a) New mechanism of the Ca–Cb cleavage
in non-phenolic arylglycerol b-aryl ether lignin substructures catalyzed by lignin peroxidase.
Holzforschung Int J Biol Chem Phys Technol Wood 47:219. doi:10.1515/hfsg.1993.47.3.219
Lundell T, Wever R, Floris R, Harvey P, Hatakka A, Brunow G, Schoemaker H (1993b) Lignin
peroxidase L3 from Phlebia radiata. Pre-steady-state and steady-state studies with veratryl
alcohol and a non-phenolic lignin model compound 1-(3,4-dimethoxyphenyl)-2-
(2-methoxyphenoxy)propane-1,3-diol. Eur J Biochem 211(3):391–402
Lundell TK, Mäkelä MR, Hildén K (2010) Lignin-modifying enzymes in filamentous
basidiomycetes—ecological, functional and phylogenetic review. J Basic Microbiol 50(1):
5–20. doi:10.1002/jobm.200900338
Mantzavinos D, Psillakis E (2004) Enhancement of biodegradability of industrial wastewaters by
chemical oxidation pre-treatment. J Chem Technol Biotechnol 79(5):431–454. doi:10.1002/
jctb.1020
Martínez ÁT (2007) High redox potential peroxidases. In: Polaina J, MacCabe A (eds) Industrial
enzymes. Springer, Netherlands, pp 477–488. doi:10.1007/1-4020-5377-0_27. ISBN
9781402053771
Martínez MJ, Ruiz-Dueñas FJ, Guillén F, Martínez ÁT (1996) Purification and catalytic properties
of two manganese peroxidase isoenzymes from Pleurotus eryngii. Eur J Biochem 237(2):
424–432. doi:10.1111/j.1432-1033.1996.0424k.x
298 N. Busse and P. Czermak

Martínez ÁT, Ruiz-Dueñas FJ, Martínez MJ, del Río JC, Gutiérrez A (2009) Enzymatic
delignification of plant cell wall: from nature to mill. Curr Opin Biotechnol 20(3):348–357.
doi:10.1016/j.copbio.2009.05.002
Melin T, Rautenbach R (2007) Membranverfahren Grundlagen der Modul- und
Anlagenauslegung. Springer, Berlin. ISBN 978-3-540-34327-1
Méndez-Hernández JE, Eibes G, Arca-Ramos A, Lú-Chau TA, Feijoo G, Moreira MT, Lema JM
(2015) Continuous removal of nonylphenol by versatile peroxidase in a two-stage membrane
bioreactor. Appl Biochem Biotechnol 175(6):3038–3047. doi:10.1007/s12010-014-1474-1
Mester T, Field JA (1998) Characterization of a novel manganese peroxidase-lignin peroxidase
hybrid isozyme produced by Bjerkandera species strain BOS55 in the absence of manganese.
J Biol Chem 273(25):15412–15417. doi:10.1074/jbc.273.25.15412
Mika N, Zorn H, Rühl M (2013) Insect-derived enzymes: a treasure for industrial biotechnology
and food biotechnology. In: Vilcinskas A (ed) Yellow biotechnology II. Springer, Berlin,
pp 1–17. doi:10.1007/10_2013_204. ISBN 3642399010
Mohorčič M, Benčina M, Friedrich J, Jerala R (2009) Expression of soluble versatile peroxidase of
Bjerkandera adusta in Escherichia coli. Bioresour Technol 100(2):851–858. doi:10.1016/
j.biortech.2008.07.005
Moreira PR, Bouillenne F, Almeida-Vara E, Xavier Malcata F, Frère JM, Duarte JC (2006)
Purification, kinetics and spectral characterisation of a new versatile peroxidase from a
Bjerkandera sp. isolate. Enzyme Microb Technol 38(1–2):28–33. doi:10.1016/j.enzmictec.
2004.12.035
Morrison M, Pope PB, Denman SE, McSweeney CS (2009) Plant biomass degradation by gut
microbiomes: more of the same or something new? Curr Opin Biotechnol 20(3):358–363.
doi:10.1016/j.copbio.2009.05.004
Mulder MHV (1996) Basic principles of membrane technology. Kluwer, The Netherlands. ISBN
0-7923-4247-X (HB)
Nakajima R, Yamazaki I (1987) The mechanism of oxyperoxidase formation from ferryl
peroxidase and hydrogen peroxide. J Biol Chem 262(6):2576–2581
Nicell JA, Bewtra JK, Biswas N, Taylor E (1993) Reactor development for peroxidase catalyzed
polymerization and precipitation of phenols from wastewater. Water Res 27(11):1629–1639.
doi:10.1016/0043-1354(93)90127-4
Palmer JM, Harvey PJ, Schoemaker HE (1987) The role of peroxidases, radical cations and
oxygen in the degradation of lignin [and discussion]. Philos Trans R Soc Lond A 321
(1561):495–505. doi:10.1098/rsta.1987.0027
Pauchet Y, Wilkinson P, Chauhan R, French-Constant RH (2010) Diversity of beetle genes
encoding novel plant cell wall degrading enzymes. PLoS One 5(12):e15635. doi:10.1371/
journal.pone.0015635
Pease EA, Aust SD, Tien M (1991) Heterologous expression of active manganese peroxidase from
Phanerochaete chrysosporium using the baculovirus expression system. Biochem Biophys Res
Commun 179(2):897–903. doi:10.1016/0006-291X(91)91903-P
Pérez-Boada M, Ruiz-Dueñas FJ, Pogni R, Basosi R, Choinowski T, Martínez MJ, Piontek K,
Martínez ÁT (2005) Versatile peroxidase oxidation of high redox potential aromatic
compounds: site-directed mutagenesis, spectroscopic and crystallographic investigation of
three long-range electron transfer pathways. J Mol Biol 354(2):385–402. doi:10.1016/j.jmb.
2005.09.047
Pfromm PH, Watkins EJ (1997) Fouling of electrodialysis membranes by organic macromolecules.
IPST technical paper series number 676 12
Pogni R, Baratto MC, Giansanti S, Teutloff C, Verdin J, Valderrama B, Lendzian F, Lubitz W,
Vazquez-Duhalt R, Basosi R (2005) Tryptophan-based radical in the catalytic mechanism of
versatile peroxidase from Bjerkandera adusta. Biochemistry 44(11):4267–4274. doi:10.1021/
bi047474l
Porter MC (1972) Concentration polarization with membrane ultrafiltration. Product R&D 11
(3):234–248. doi:10.1021/i360043a002
11 Role and Application of Versatile Peroxidase (VP) for Utilizing… 299

Rasmussen CB, Dunford HB, Welinder KG (1995) Rate enhancement of compound I formation of
barley peroxidase by ferulic acid, caffeic acid and coniferyl alcohol. Biochemistry 34
(12):4022–4029. doi:10.1021/bi00012a021
Rodakiewicz-Nowak J, Jarosz-Wilkołazka A, Luterek J (2006) Catalytic activity of versatile
peroxidase from Bjerkandera fumosa in aqueous solutions of water-miscible organic solvents.
Appl Catal A 308:56–61. doi:10.1016/j.apcata.2006.04.009
Rodríguez Couto S, Toca Herrera JL (2006) Industrial and biotechnological applications of
laccases: a review. Biotechnol Adv 24(5):500–513. doi:10.1016/j.biotechadv.2006.04.003
Rühl M, Kilaru S, Navarro-Gonzaléz M, Hoegger P, Kharazipour A, Kües U (2007) Production
of laccase and other enzymes for the wood industry. In: Kües U (ed) Wood production,
wood technology, and biotechnological impacts. Universitätsverlag Göttingen, Göttingen,
pp 469–507. ISBN 0849369649
Rühl M, Fischer C, Kües U (2008) Ligninolytic enzyme activities alternate with mushroom
production during industrial cultivation of Pleurotus ostreatus on wheatstraw-based substrate.
Curr Trends Biotechnol Pharm 2(4):478–492. ISSN 0973-8916
Ruiz-Dueñas FJ, Martínez ÁT (2009) Microbial degradation of lignin: how a bulky recalcitrant
polymer is efficiently recycled in nature and how we can take advantage of this. Microb
Biotechnol 2(2):164–177. doi:10.1111/j.1751-7915.2008.00078.x
Ruiz-Dueñas FJ, Morales M, García E, Miki Y, Martínez MJ, Martínez ÁT (2009a) Substrate
oxidation sites in versatile peroxidase and other basidiomycete peroxidases. J Exp Bot 60
(2):441–452. doi:10.1093/jxb/ern261
Ruiz-Dueñas FJ, Pogni R, Morales M, Giansanti S, Mate MJ, Romero A, Martínez MJ, Basosi R,
Martínez ÁT (2009b) Protein radicals in fungal versatile peroxidase: catalytic tryptophan
radical in both compound I and compound II and studies on W164Y, W164H, and W164S
variants. J Biol Chem 284(12):7986–7994. doi:10.1074/jbc.M808069200
Saake B, Lehnen R (2012) Lignin. In: Ullmann’s encyclopedia of industrial chemistry.
Wiley-VCH Verlag GmbH & Co. KGaA. doi: 10.1002/14356007.a15_305.pub3
Sánchez C (2009) Lignocellulosic residues: biodegradation and bioconversion by fungi.
Biotechnol Adv 27(2):185–194. doi:10.1016/j.biotechadv.2008.11.001
Scharf ME, Karl ZJ, Sethi A, Boucias DG (2011) Multiple levels of synergistic collaboration in
termite lignocellulose digestion. PLoS One 6(7):e21709. doi:10.1371/journal.pone.0021709
Schlipalius DI, Valmas N, Tuck AG, Jagadeesan R, Ma L, Kaur R, Goldinger A, Anderson C,
Kuang J, Zuryn S, Mau YS, Cheng Q, Collins PJ, Nayak MK, Schirra HJ, Hilliard MA,
Ebert PR (2012) A core metabolic enzyme mediates resistance to phosphine gas. Science 338
(6108):807–810. doi:10.1126/science.1224951
Schoemaker HE (1990) On the chemistry of lignin biodegradation. Recl Trav Chim Pays-Bas 109
(4):255–272. doi:10.1002/recl.19901090402
Schoemaker HE, Piontek K (1996) On the interaction of lignin peroxidase with lignin. Pure Appl
Chem 68:2089–2096. doi:10.1351/pac199668112089
Schoemaker HE, Lundell TK, Floris R, Glumoff T, Winterhalter KH, Piontek K (1994a) Do
carbohydrates play a role in the lignin peroxidase cycle? Redox catalysis in the endergonic
region of the driving force. Bioorganic Med Chem 2(6):509–519. doi:10.1016/0968-0896(94)
80021-9
Schoemaker HE, Lundell TK, Hatakka AI, Piontek K (1994b) The oxidation of veratryl alcohol,
dimeric lignin models and lignin by lignin peroxidase: the redox cycle revisited. FEMS
Microbiol Rev 13(2–3):321–331. doi:10.1111/j.1574-6976.1994.tb00052.x
Schüttmann I, Bouws H, Szweda RT, Suckow M, Czermak P, Zorn H (2014) Induction,
characterization, and heterologous expression of a carotenoid degrading versatile peroxidase
from Pleurotus sapidus. J Mol Catal B Enzym 103:79–84. doi:10.1016/j.molcatb.2013.08.007
Seelbach K, van Deurzen MPJ, van Rantwijk F, Sheldon RA, Kragl U (1997) Improvement of the
total turnover number and space-time yield for chloroperoxidase catalyzed oxidation.
Biotechnol Bioeng 55(2):283–288. doi:10.1002/(sici)1097-0290(19970720)55:2<283:aid-
bit6>3.0.co;2-e
300 N. Busse and P. Czermak

Singh D, Chen S (2008) The white-rot fungus Phanerochaete chrysosporium: conditions for the
production of lignin-degrading enzymes. Appl Microbiol Biotechnol 81(3):399–417. doi:10.
1007/s00253-008-1706-9
Sridhar S, Bhattacharya PK (1991) Limiting flux phenomena in ultrafiltration of kraft black liquor.
J Membr Sci 57(2–3):187–206. doi:10.1016/S0376-7388(00)80678-X
Stöcker M (2008) Bio- und BTL-Kraftstoffe in der Bioraffinerie: katalytische Umwandlung
Lignocellulose-reicher Biomasse mit porösen Stoffen. Angew Chem 120(48):9340–9351.
doi:10.1002/ange.200801476
Sun Y, Cheng J (2002) Hydrolysis of lignocellulosic materials for ethanol production: a review.
Bioresour Technol 83(1):1–11. doi:10.1016/S0960-8524(01)00212-7
Tien M (1987) Properties of ligninase from Phanerochaete chrysosporium and their possible
applications. Crit Rev Microbiol 15(2):141–168. doi:10.3109/10408418709104456
Tien M, Kirk TK (1984) Lignin-degrading enzyme from Phanerochaete chrysosporium:
Purification,characterization, and catalytic properties of a unique H2O2-requiring oxygenase.
PNAS 81(8):2280–2284.doi:10.1073/pnas.81.8.2280
Towne V, Will M, Oswald B, Zhao Q (2004) Complexities in horseradish peroxidase-catalyzed
oxidation of dihydroxyphenoxazine derivatives: appropriate ranges for pH values and
hydrogen peroxide concentrations in quantitative analysis. Anal Biochem 334(2):290–296.
doi:10.1016/j.ab.2004.07.037
Tsukihara T, Honda Y, Sakai R, Watanabe T, Watanabe T (2006) Exclusive overproduction of
recombinant versatile peroxidase MnP2 by genetically modified white rot fungus, Pleurotus
ostreatus. J Biotechnol 126(4):431–439. doi:10.1016/j.jbiotec.2006.05.013
Valderrama B, Ayala M, Vazquez-Duhalt R (2002) Suicide inactivation of peroxidases and the
challenge of engineering more robust enzymes. Chem Biol 9(5):555–565. doi:10.1016/S1074-
5521(02)00149-7
Van der Bruggen B (2009) Fundamentals of membrane solvent separation and pervaporation. In:
Membrane operations. Wiley-VCH Verlag GmbH & Co. KGaA, pp 45–61. doi: 10.1002/
9783527626779.ch3. ISBN 9783527626779
van Deurzen MPJ, Seelbach K, van Rantwijk F, Kragl U, Sheldon RA (1997) Chloroperoxidase:
use of a hydrogen peroxide-stat for controlling reactions and improving enzyme performance.
Biocatal Biotransform 15(1):1–16. doi:10.3109/10242429709003606
Viamajala S, Donohoe B, Decker S, Vinzant T, Selig M, Himmel M, Tucker M (2010) Heat and
mass transport in processing of lignocellulosic biomass for fuels and chemicals. In: Singh OV,
Harvey SP (eds) Sustainable biotechnology. Springer, Netherlands, pp 1–18. doi:10.1007/978-
90-481-3295-9_1. ISBN 978-90-481-3294-2
Vlasits J, Jakopitsch C, Bernroitner M, Zamocky M, Furtmüller PG, Obinger C (2010)
Mechanisms of catalase activity of heme peroxidases. Arch Biochem Biophys 500(1):74–81.
doi:10.1016/j.abb.2010.04.018
Wang Y, Vazquez-Duhalt R, Pickard MA (2003) Manganese-lignin peroxidase hybrid from
Bjerkandera adusta oxidizes polycyclic aromatic hydrocarbons more actively in the absence of
manganese. Can J Microbiol 49(11):675–682. doi:10.1139/w03-091
Wariishi H, Gold MH (1989) Lignin peroxidase compound III: formation, inactivation, and
conversion to the native enzyme. FEBS Lett 243(2):165–168. doi:10.1016/0014-5793(89)
80122-X
Wariishi H, Gold MH (1990) Lignin peroxidase compound III. Mechanism of formation and
decomposition. J Biol Chem 265(4):2070–2077
Wong DW (2009) Structure and action mechanism of ligninolytic enzymes. Appl Biochem
Biotechnol 157(2):174–209. doi:10.1007/s12010-008-8279-z
Yokota K, Yamazaki I (1965) The activity of the horseradish peroxidase compound 3. Biochem
Biophys Res Commun 18:48–53. doi:10.1016/0006-291X(65)90880-6
Zorn H, Peters T, Nimtz M, Berger RG (2005) The secretome of Pleurotus sapidus.
PROTEOMICS 5(18):4832–4838. doi:10.1002/pmic.200500015
Chapter 12
Fungal Aryl-Alcohol Oxidase
in Lignocellulose Degradation
and Bioconversion

Juan Carro, Ana Serrano, Patricia Ferreira and Angel T. Martínez

Abstract Bioconversion of lignocellulosic materials draws much interest as they


are regarded as a renewable source of energy and platform chemicals. The enzyme
aryl-alcohol oxidase (AAO) has been extensively studied, revealing its involvement
in biodegradation of lignocellulose by several well-known white-rot fungi. Its
physiological role is to supply hydrogen peroxide from the oxidation of aromatic
substrates derived from fungal secondary metabolism or lignin degradation, which
can: (i) be used by peroxidases to oxidise lignin; or (ii) give rise, through Fenton
reaction, to hydroxyl radical that is able (itself) to depolymerise cellulose and
oxidise lignin. Several features make this enzyme an appealing biocatalyst that has
shown its potential for industrial applications: AAO has a broad range of substrates
that it oxidises by stereoselective hydride transfer reaction mechanism, and reduces
atmospheric molecular oxygen as a co-substrate producing hydrogen peroxide.

Abbreviations
AAO Aryl-alcohol oxidase
DFF 2,5-Diformylfuran
FDCA 2,5-Furandicarboxylic acid
FFCA 5-Formylfurancarboxylic acid
GMC Glucose-methanol-choline oxidase/dehydrogenase superfamily
HMF 5-Hydroxymethylfurfural
HMFCA 5-Hydroxymethylfurancarboxylic acid
LiP Lignin peroxidase

MnP Manganese peroxidase

J. Carro  A. Serrano  A.T. Martínez (&)


Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
e-mail: [email protected]
P. Ferreira
Department of Biochemistry and BIFI, University of Zaragoza, Pedro Cerbuna 12, 50009
Zaragoza, Spain

© Springer International Publishing Switzerland 2016 301


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_12
302 J. Carro et al.

PEF Poly(ethylene furandicarbolylate)


UPO Unspecific peroxygenase
VP Versatile peroxidase

12.1 Introduction to AAO

Aryl-alcohol oxidase (AAO) is a fungal enzyme secreted by different fungi,


including basidiomycetes involved in the degradation of lignocellulose. It belongs
to the glucose-methanol-choline oxidase/dehydrogenase (GMC) protein super-
family (Cavener 1992) that includes oxidoreductases, although there are exceptions
such as hydroxynitrile lyase, with different substrate specificities. They all share a
common fold in two subunits (the cofactor-binding and substrate-binding ones) and
the presence of FAD as a cofactor.
AAO activity was reported during the 1960s in cultures of the lignicolous fungus
Trametes versicolor by Farmer et al. (1960), although this enzyme was not further
characterised. It was during the late 1980s and early 1990s that AAO was isolated
from several Agaricales species, such as Pleurotus sajor-caju (Bourbonnais and
Paice 1988), Pleurotus eryngii (Guillén et al. 1990) and Pleurotus ostreatus (Sannia
et al. 1991), and identified as the source of the H2O2 (Guillén et al. 1994) and
aromatic aldehydes found in cultures of the above fungi (Guillén and Evans 1994;
Gutiérrez et al. 1994). During subsequent years, its involvement in the ligninolytic
process was demonstrated. AAO is responsible, along with other GMC oxidore-
ductases and copper-radical oxidases, for the H2O2 production that, as described
below, is required by: (i) the activity of ligninolytic peroxidases; and (ii) the for-
mation of oxygen radicals that exert oxidative and depolymerising activities on
polysaccharides and also on lignin.
Some AAOs have been exhaustively characterized, being those of P. eryngii
(Ferreira et al. 2005; Guillén et al. 1992) and Bjerkandera adusta (de Jong et al.
1994; Romero et al. 2009) two remarkable examples. Their substrate specificity
showed to be broad, probably due to its extracellular and ligninolytic nature, since it
proved to use secondary fungal metabolites, p-methoxybenzyl alcohol and other
benzylic alcohols, as substrates. In fact, it is currently known that AAO can oxidise
both phenolic and non-phenolic aryl-alcohols, together with other polyunsaturated
(aliphatic) primary alcohols, to their corresponding aldehydes, as well as aromatic
secondary alcohols, albeit with much lower efficiency (Table 12.1). Moreover, the
presence of low amounts of acids attributed to its activity in some fungal cultures
(and in vitro reactions) paved the way for the demonstration of its activity on aryl
aldehydes (Ferreira et al. 2010).
This plethora of AAO substrates (Table 12.1), among which several reduced
species derived from lignin decay products are noticeable, makes it a promising
biocatalyst. The lignocellulosic materials are the main renewable resource on Earth
12 Fungal Aryl-Alcohol Oxidase … 303

Table 12.1 Comparison of the catalytic efficiencies of P. eryngii and B. adusta AAO oxidising
representative alcohol and aldehyde substrates
kcat/Km (s−1 mM−1)
P. eryngii B. adusta
Benzyl alcohol 47 ± 9 18 ± 1
OH

p-Fluorobenzyl alcohol F 59 ± 6 22 ± 2
OH

m-Fluorobenzyl alcohol 13 ± 1 47 ± 2
OH
F

p-Chlorobenzyl alcohol Cl 398 ± 32 361 ± 15


OH

m-Chlorobenzyl alcohol 203 ± 4 1050 ± 30


OH
Cl

p-Methoxybenzyl alcohol O 5230 ± 620 646 ± 45


OH

m-Methoxybenzyl alcohol 65 ± 24 349 ± 8


OH
O

Veratryl alcohol O 210 ± 5 22 ± 1


OH
O

Isovanillyl alcohol O 152 ± 5 51 ± 1


OH
HO

Vanillyl alcohol HO 0 31 ± 1
OH
O

3-Chloro-p-methoxybenzyl O 4090 ± 200 1480 ± 110


alcohol OH
Cl

2-4-Hexadien-1-ol HO 1270 ± 60 186 ± 7


Cinnamyl alcohol 78 ± 11 305 ± 11
OH

Coniferyl alcohol HO 0 5 ± 0.2


OH
O

m-Chlorobenzaldehyde 0.64 ± 0.05 1 ± 0.09


O
Cl

3-Chloro-p-methoxy-benzaldehyde O 0.085 ± 0.006 0.050 ± 0.002


O
Cl

m-Fluorobenzaldehyde 0.40 ± 0.02 0.05 ± 0.005


O
F

p-Nitrobenzaldehyde O O 0.31 ± 0.006 0.010 ± 0.001


N+
-
O
(continued)
304 J. Carro et al.

Table 12.1 (continued)


kcat/Km (s−1 mM−1)
P. eryngii B. adusta
5-Hydroxymethyl-2-furfural HO O 0.21 ± 0.02 n.d.
O

2,5-Diformylfuran O O 0.150 ± 0.008 n.d.


O

n.d. not determined

because they are widespread and abundant (forests cover 27 % of world’s area) and,
thus, they are cheap and can be easily stored. As a consequence, the conversion of
these materials into biofuels in biorefineries is of great interest. However, not only
biofuels, that is heat and power, are important, but other by-products obtained
during the biorefinery processes are also being carefully examined aiming at using
them for the production of valorised chemicals (Bozell and Petersen 2010). AAO is
a candidate for the enzymatic delignification of plant biomass (in synergy with
other oxidoreductases) and for the production of aromatic aldehydes and acids that
can originate from this renewable resource.
In this chapter, AAO involvement in lignocellulose decay owing to its capacity
to produce H2O2, as well as its potential applications in different industrial pro-
cesses, are discussed.

12.2 AAO and Lignocellulose Decay

Lignocellulosic biomass (in woody and nonwoody vascular plants) accounts for the
60 % of the total carbon fixed by land photosynthesis, and is constituted of three
main polymers: cellulose, hemicellulose and lignin (Higuchi 1997). Both cellulose
and lignin are the two most abundant polymers on Earth. Lignin is located in the
middle lamella, where it attains its highest concentration, and the secondary wall of
vascular plants, together with the above polysaccharides. Its main functions are
conferring rigidity to the plants, waterproofing vessels and providing protection
against desiccation, pathogens and irradiation (Gellerstedt and Henriksson 2008).
Since lignin is very recalcitrant, it constitutes the main sink of carbon in land
ecosystems and, as a consequence, its degradation is a key step to complete the
carbon cycle. In fact, before it started to be biodegraded and mineralised, at the end
of the Carboniferous period (*300 million year ago), the carbon that the plants
fixed accumulated and therefore gave rise to the coal deposits we currently use as
source of fossil fuels (Floudas et al. 2012).
The only organisms capable of degrading wood are some saprotrophic agari-
comycetes, feeding on the simple sugars produced when cellulose and
12 Fungal Aryl-Alcohol Oxidase … 305

hemicelluloses are hydrolysed. However, to accomplish this task, fungi must


overcome a very recalcitrant barrier: lignin. Lignin is an amorphous polymer
including a variety of bonds established among the phenylpropanoid units that form
its structure. Although these fungi do not use lignin as a nutrient, they have to
remove or modify lignin to gain access to carbohydrates. The development of an
extracellular system capable of altering the structure of lignin was, thus, a great
achievement in evolutionary and ecological terms, since the polymer is mineralised
and the carbon “sequestered” in it may go back to the atmosphere as CO2. It is
generally accepted that the first lignin-degrading organism must have been a
basidiomycete (Floudas et al. 2012), which developed some strategies to act on the
recalcitrant lignin polymer oxidising its subunits, causing bond breakages and
releasing the carbohydrates embedded within its matrix.
The role of H2O2 in lignocellulose decay was studied as it proved to be produced
simultaneously with the ligninolytic system. It was seen that the addition of cata-
lase, an enzyme that degrades H2O2, to cultures of the white-rot fungus
Phanerochaete chrysosporium diminished its lignin-degrading ability (Faison and
Kirk 1983). Two families of oxidoreductases include oxidases being able to pro-
duce this strong oxidant: copper-radical oxidases and GMC oxidoreductases.
The copper-radical oxidases are proteins that have one copper ion and a protein
radical involved in catalysis, such as extracellular glyoxal oxidase (Kersten and
Kirk 1987) and the related enzymes recently identified from sequenced genomes
(Kersten and Cullen 2014), among others. GMC oxidoreductases are a superfamily
of enzymes that share common structural patterns, including the FAD cofactor and
a histidine catalytic base, although they differ in their substrate specificities.
Extracellular GMC enzymes involved in the lignocellulose-degradation process are
cellobiose dehydrogenase (Ayers et al. 1978; Bao et al. 1993), pyranose 2-oxidase
(Daniel et al. 1994), aryl-alcohol oxidase (Guillén et al. 1990) and methanol oxi-
dase (Nishida and Eriksson 1987). The latter enzyme lacks a signal peptide to
transport the protein to the extracellular space, but its presence out of the hyphae
has been revealed, and it is supposed to be secreted by alternative secretion path-
ways not known yet (Daniel et al. 2007). Glucose oxidase (Eriksson et al. 1986) is
an intracellular enzyme, so its role in lignocellulose degradation is controversial,
since a transport system for the H2O2 formed would be required in order that it
could carry out functions in the extracellular medium.
AAO activity has been reported in different fungi including T. versicolor (Farmer
et al. 1960), Fusarium solani (Iwahara et al. 1980), Rigidoporus microporus
(Waldner et al. 1988), Pleurotus species (Bourbonnais and Paice 1988; Guillén et al.
1992; Sannia et al. 1991), B. adusta (Kimura et al. 1990; Muheim et al. 1990b;
Romero et al. 2009, 2010) and Botrytis cinerea (Goetghebeur et al. 1993); and the
corresponding genes have been identified in many basidiomycete genomes (Ferreira
et al. 2015; Floudas et al. 2012; Hernández-Ortega et al. 2012a), as described below.
Further evidence of the simultaneous expression of AAO along with enzymes
typically expressed under ligninolytic condition such as lignin peroxidase (LiP) in B.
adusta (Muheim et al. 1990a) and manganese-oxidising peroxidases in Pleurotus
cultures (Camarero et al. 1996), supported the involvement of this oxidase in lignin
306 J. Carro et al.

degradation. Moreover, the enzyme was located in the hyphal sheath (which is
formed by secreted fungal polysaccharide) during lignocellulose degradation
(Barrasa et al. 1998), as it had been previously reported for ligninolytic peroxidases
and laccases (Gallagher et al. 1989; Green et al. 1992).
The activity of AAO is supported by both non-phenolic and phenolic aryl
alcohols that can derive from fungal metabolism (de Jong et al. 1994; Gutiérrez
et al. 1994) and from lignin degradation (Kirk and Farrell 1987; Shimada and
Higuchi 1991) and are substrates for this enzyme in cooperation with related
dehydrogenases. It was seen that H2O2 is produced when adding aromatic alcohols,
as well as the corresponding aldehydes and acids, to the mycelium of P. eryngii
(Guillén et al. 1994). Thus, it was postulated that the redox cycling of these
compounds results in a continuous supply of H2O2. The product of the reaction with
AAO (aldehyde or acid) needs to be reduced in order to be re-used by the enzyme.
Therefore, it is hypothesised that the oxidised species is transported to the intra-
cellular space, where aryl-alcohol or aryl-aldehyde dehydrogenases, both
NADPH-dependent enzymes, convert it into alcohol again (de Jong et al. 1994;
Guillén and Evans 1994). Supply of reduced NADPH must be supported by the
high carbon availability for a fungus under ligninolytic conditions (Fig. 12.1).
P. eryngii AAO’s preferential substrate is p-methoxybenzyl alcohol
(Table 12.1), which is a secondary metabolite detected in fungal cultures grown

+
Fungal NADP NADPH
hypha Secondary Aryl-alcohol Catabolism
metabolism
t b li dehydrogenase

p-methoxybenzyl
alcohol H2O
+
glucose CO2
p-anisaldehyde AAO +

O2
H2O2 + Fe2+ vanillin
H2O Fenton reaction

Peroxidase OH·

Lignin Lignin Cellulose


OH Breakdown
O of linkages
OH
O Anabolism
Plant
lignin subunit lignin cation radical
cell-wall

Fig. 12.1 Scheme of the natural role of AAO in decay of plant cell-wall producing H2O2 for:
(i) activation of lignin-degrading peroxidases; and (ii) formation of cellulose-depolymerising
hydroxyl radical (also causing lignin oxidation)
12 Fungal Aryl-Alcohol Oxidase … 307

both in glucose and lignin media (Gutiérrez et al. 1994). It is known that aromatic
alcohols, aldehydes and acids are derived from the shikimic acid pathway of fungal
secondary metabolism (Turner and Aldridge 1983). It was seen that the oxidised
product, p-methoxybenzaldehyde (also known as p-anisaldehyde), was much more
abundant in the Pleurotus cultures than its alcoholic counterpart (Gutiérrez et al.
1994). High levels of related compounds, such as 3-chloro-p-anisaldehyde, were
also found to be abundant in B. adusta cultures (de Jong et al. 1992) and agree with
catalytic efficiencies towards the corresponding alcohol for B. adusta AAO, for
which seems to be the preferential substrate (Table 12.1). These chlorinated
derivatives are also minor extracellular metabolites in P. eryngii and P. ostreatus
(Gutiérrez et al. 1994; Okamoto et al. 2002).
These findings further supported the hypothesis postulating that AAO was acting
in the extracellular space involved in the said redox-cycling process for H2O2
production.
Based on macroscopic and chemical composition features, the wood-degrading
processes were split into two different types involving different decay mechanisms.
Some fungi leave a whitish residue, and thus were named white-rot fungi, while
others produce a brown residue and were called brown-rot fungi (Martínez et al.
2005; Schwarze et al. 2000; Zabel and Morrell 1992). Owing to genomic and
enzymatic studies, it is now thought that AAO could act as an auxiliary enzyme in
both processes due to its H2O2-producing activity and presence in the sequenced
genomes of Agaricomycotina responsible for the two types of wood-decay
processes.

12.2.1 White-Rot Decay

White-rot fungi developed an enzymatic machinery to degrade lignin and, hence, be


able to use cellulose and hemicellulose as a source of carbon and energy. The main
enzymes involved are metalloproteins including LiP, manganese peroxidase
(MnP) and versatile peroxidase (VP), as well as laccases (Ruiz-Dueñas and
Martínez 2009). Ligninolytic peroxidases are heme proteins that use H2O2 as the
electron-accepting substrate to oxidise the lignin units. In contrast, laccases have
copper as a cofactor, use O2 as electron acceptor, and are thought to often act
through small intermediate compounds, redox mediators, which in turn oxidise
lignin. Since most enzymes cannot penetrate the intricate and compact structure of
sound wood, small chemical oxidisers, activated oxygen species (as hydroxyl
radical), metal cations (as Mn3+ produced by MnP and VP) and aromatic radicals
(formed by different oxidoreductases) are probably responsible for the first stages of
lignin decay (Evans et al. 1994). In white-rot decay there is a need of a continuous
H2O2 flow in the extracellular environment in order that peroxidases be able to act
on lignin. Moreover, hydroxyl radical can be produced through Fenton reaction
between Fe2+ and H2O2, and participate in the oxidative modification of cellulose
308 J. Carro et al.

(as described below for brown-rot decay) and also of lignin (Fig. 12.1) (Bes et al.
1983; Forney et al. 1982; Gómez-Toribio et al. 2009).
White-rot fungi can be classified into two groups according to their gross
degradation patterns. Some of them degrade lignin and cellulose simultaneously as
it is the case for P. chrysosporium, one of the most studied lignin-degrading
organisms. Instead, others, such as Ceriporiopsis subvermispora, degrade lignin
before cellulose (Otjen and Blanchette 1986). The main differences between the
sequenced genomes of these two fungi appear to be related to (i) the peroxidase
repertoire, and (ii) the genes involved in lipid metabolism (Fernández-Fueyo et al.
2012). The latter is related to the fact that free radicals from unsaturated lipids are
supposed to also play a role in lignin attack (Bao et al. 1994). Selective white-rot
fungi are the most interesting for industrial (biotechnological) applications in which
carbohydrates are the raw material, since they release cellulose from the lignin
matrix without significantly consuming it.
The analysis of genomes of white-rot fungi in which AAO appeared to be
produced, along with peroxidases, further confirmed its involvement in the
white-rot process as an auxiliary enzyme producing H2O2. For instance, Floudas
et al. (2012) analysed 24 basidiomycete genomes to search for enzymes involved in
the degradation of lignin. On the one hand, all white-rot genomes studied possessed
AAO genes with the only exception of Auricularia delicata. On the other hand,
AAO appears to be the most common H2O2-producing GMC, since methanol
oxidase genes are not as abundant, glucose oxidase genes are absent, and pyranose
2-oxidase genes are only found in two of the genomes studied (Table 12.2).

Table 12.2 Inventory of peroxidase and GMC genes in eleven white-rot Agaricomycotina
genomes
AD PST FM DS TV SH BA PB PC GS CS
Peroxidases 21 21 33 21 39 11 34 16 18 14 25
GMCs AAO 0 4 2 9 3 14 11 3 3 9 4
CDH 1 1 1 1 1 1 1 1 1 1 1
MOX 3 3 2 4 4 7 5 6 3 4 1
P2O 2 0 0 0 2 0 1 1 1 0 0
From Floudas et al. (2012) and Ruiz-Dueñas et al. (2013) and Ferreira et al. (2015)
AAO, aryl-alcohol oxidase; CDH, cellobiose dehydrogenase; MOX, methanol oxidase and P2O,
pyranose 2-oxidase
AD, Auricularia delicata; PST, Punctularia strigosozonata; FM, Fomitiporia mediterranea; DS,
Dichomitus squalens; TV, Trametes versicolor; SH, Stereum hirsutum; BA, Bjerkandera adusta;
PB, Phlebia brevispora; PC, Phanerochaete chrysosporium; GS, Ganoderma sp. in the
Ganoderma lucidum complex and CS, Ceriporiopsis subvermispora
12 Fungal Aryl-Alcohol Oxidase … 309

12.2.2 Brown-Rot Decay

The brown-rot decay is based on a non-enzymatic attack on wood that leads to


lignin modification, instead of degradation/mineralisation and cellulose depoly-
merisation. On having no longer the machinery for degrading lignocellulose
(Table 12.3), brown-rot fungi are the only organisms known to be able to nearly
completely remove all polysaccharides from wood without causing substantial
lignin degradation (Arantes et al. 2012). Nevertheless, it has been demonstrated that
they do alter lignin in order to gain access to the carbohydrates leaving it partially
modified but still polymeric and with a recognisable chemical structure (Martínez
et al. 2011). Genomic studies have proved that these fungi stem from the white-rot
lineage, and that they have lost most of the energy-consuming apparatus their
white-rot ancestors used to have, which means that peroxidases are absent or
residual in their genomes (only one generic peroxidase gene in some species)
(Ruiz-Dueñas et al. 2013). They have originated independently in several lineages
and have representatives in five basidiomycete orders (Floudas et al. 2012).
The initial oxidative step alters the plant cell-wall structure in order to make it
more accessible to enzymes involved in subsequent decay (Goodell et al. 1997).
Reactive oxygen species, such as hydroxyl radical, generated by Fenton chemistry,
play a key role in this oxidative process (Fig. 12.1) (Baldrian and Valaskova 2008;
Halliwell 1965). Fenton chemistry is based on reactions among H2O2 and Fe2+ ions
giving rise to hydroxyl free radical. This is a radical-chain mechanism where Fe2+ is
generated from Fe3+ in a cycle that involves fungal enzymes. The hydroxyl radical
is the most powerful (non-specific) oxidant in biological systems but it has an
extremely short half-life (10−9 s) and, as a consequence, Fenton reactions need
occur immediately adjacent to the site of oxidative action within the plant cell wall
because of spatial diffusion limitations (Arantes et al. 2012).
Since one of the most remarkable chemical alterations that brown-rot decay
cause to lignin is demethoxylation (Niemenmaa et al. 2008; Yelle et al. 2008),
which ultimately gives rise to methanol, methanol oxidase is thought to be the most
important H2O2-producing enzyme in brown-rot decay. Nevertheless, genomic

Table 12.3 Inventory of peroxidase and GMC genes (see Table 12.2 for enzyme abbreviations)
in six brown-rot Agaricomycotina genomes
CP GT FP WC DSP RP
Peroxidases 0 0 1 1 0 1
GMCs AAO 0 2 1 0 0 2
CDH 1 1 0 0 0 0
MOX 2 1 4 4 1 4
P2O 0 0 0 0 0 0
From Floudas et al. (2012) and Ferreira et al. (2015)
CP, Coniophora puteana; GT, Gloeophyllum trabeum; FP, Fomitopsis pinicola; WC, Wolfiporia
cocos; DSP, Dacryopinax sp. and RP, Rhodonia placenta
310 J. Carro et al.

studies have shown that brown-rot fungi have AAO too (Martinez et al. 2009),
suggesting that the latter oxidase might play a role in Fenton chemistry. Floudas
et al. (2012) analysed seven brown-rot fungal genomes and found AAO genes in
several of them, although they were not as abundant as those of methanol oxidases
(Table 12.3).

12.3 Paper Pulp Industry: Biopulping and Biobleaching

One of the main steps of pulp and paper industry is the treatment of wood chips to
separate cellulosic fibres (the actual raw material used) from the lignin forming the
middle lamella, a process called pulping. The concern about environment and
energy wasting has stimulated studies on the use of microorganisms to accomplish
this task. The resulting biopulping process is thought to be energetically and
environmentally more favourable than chemical and/or mechanical treatments
(Blanchette et al. 1992; Rasmussen et al. 2010).
The biopulping process starts with the colonisation of wood xylem and par-
enchyma by the fungi. After hyphae have grown on the substrate, the fungus will
start producing its ligninolytic enzymatic system in order to degrade the middle
lamellae and separate fibres (Breen and Singleton 1999). Since the loss of cellulose
is undesirable, the organisms of choice are white-rot fungi showing preference for
lignin degradation rather than cellulose degradation, whose task is to make cellu-
losic fibres accessible for the papermaking process (Scott and Swaney 1998).
Several selective ligninolytic fungi, such as C. subvermispora and P. eryngii,
together with the model white-rot fungus P. chrysosporium, and brown-rot fungi
such as Postia placenta, among others, have been investigated for biopulping of
wood and annual plants (Akhtar et al. 1997; Camarero et al. 1998; Ferraz et al.
2008; Giles et al. 2014; Masarin et al. 2009; Vicentim et al. 2009). AAO is sup-
posed to act as an auxiliary enzyme providing H2O2 in these processes, as it is the
case of natural lignin degradation.
Pulp bleaching, which is the removal of chromophores in order to obtain white
paper pulp, is another process in which AAO has shown to contribute. In a study in
which two flax pulps were treated with fungal enzymes—laccases, peroxidases and
feruroyl esterases—the ability of AAO from Pleurotus pulmonarius CBS 507.85,
which is a natural hyperproducer of this enzyme, to aid in this process was tested
(Sigoillot et al. 2005). The results showed that the presence of AAO along with
laccase improved the bleaching process probably due to the ability of AAO to
prevent the repolymerisation of the phenoxy radicals released by laccases by using
them as electron acceptors (as an alternative for O2). In a similar way, AAO can be
combined with ligninolytic peroxidases, in the presence of a substrate enabling it to
release the H2O2 required by the former enzymes.
12 Fungal Aryl-Alcohol Oxidase … 311

12.4 Bioconversions

12.4.1 Flavour Synthesis

White-rot fungi are among the most versatile flavour and aroma producers in nature
(Fraatz and Zorn 2011; Lapadatescu et al. 2000). These compounds are mainly of
aromatic nature and synthesised through biotransformations by plant, enzymatic or
microbial processes (Serra et al. 2005). Since there exists demand for naturally
produced compounds, the biotechnological production of flavours and aromas
attracts much attention (Krings and Berger 1998) due to the great economic
importance this industry has.
One of the most important flavours is vanillin, naturally produced by orchids of
the Vanilla genus, but this source only represents the 1 % of the commercial vanilla
flavour. As a consequence, several methods of obtaining vanillin have been
developed (Priefert et al. 2001) that use bioconversion of lignin and phenyl-
propanoids, such as eugenol (Overhage et al. 2003). The ability of B. adusta AAO
to oxidise vanillyl alcohol reported by Romero et al. (2009), could be exploited in
the biotechnological production of this flavour. It has been suggested that AAO
may be used to avoid formation of vanillyl alcohol as a by-product diminishing the
yield of vanillin formation (Fig. 12.2a) by the fungi of the genus Pycnoporus
(Lomascolo et al. 2011).

Fig. 12.2 Catalytic (a)


efficiencies of P. eryngii and
B. adusta AAOs in B. adusta AAO
B
bioconversion of some 31±1 s-1 mM-1
flavours and aromas.
a Oxidation of vanillyl
alcohol into vanillin.
b Oxidation of benzyl alcohol
into benzaldehyde.
c Oxidation of (b)
p-methoxybenzyl alcohol into
p-methoxybenzaldehyde B. adusta AAO
18±1 s-11 mM-11
P. eryngii AAO
47±9 s-1 mM-1

(c) HO O

d t AAO
B adusta
B.
646±45 s-1 mM-1
y g AAO
P. eryngii
5230±620 s-1 mM-1
O O
312 J. Carro et al.

Another very remarkable commercial aromatic compound is benzaldehyde.


Some research has been carried out using the fungus B. adusta for the production of
this chemical from L-phenylalanine (Lapadatescu et al. 2000; Lapadatescu and
Bonnarme 1999). In this reaction of L-phenylalanine for flavour production,
aryl-alcohols are the main products obtained, as reported for B. adusta
(Lapadatescu et al. 2000) and P. chrysosporium (Jensen et al. 1994). Consequently,
the application of AAO to such biotransformations could result in the obtention of
higher levels of aromatic aldehydes, since their alcohol counterparts are substrates
of the enzyme (Fig. 12.2b). Finally, using AAO plus an unspecific peroxygenase
(UPO) from the fungus Agrocybe aegerita (Ullrich et al. 2004), an oxidoreductase
cascade can be used for toluene conversion into benzaldehyde, with the second
enzyme using the peroxide generated by AAO.
Another flavour that AAO produces as a consequence of its auxiliary role in
lignin degradation is p-anisaldehyde (4-methoxybenzaldehyde) (Fig. 12.2c). It was
shown that the corresponding alcohol is the physiological and preferential substrate
of the P. eryngii enzyme (Ferreira et al. 2005; Guillén et al. 1992) and that AAO
and mycelium-associated aromatic dehydrogenases establish a concerted anisalde-
hyde redox cycle to produce H2O2 continuously as described above (Fig. 12.1).
Therefore, AAO can be used for biotransformations aiming at the production of
vanillin, benzaldehyde, anisaldehyde and other aromas.

12.4.2 Deracemization of Chiral Secondary Alcohols

Many of the drugs and potential drug candidates possess chiral centres and most of
them need to be commercialised as enantiomers rather than racemates given that
enantiomers often carry out different activities within biological systems (Carey
et al. 2006). Therefore, chiral intermediates for pharmaceuticals are synthesised
through enantioselective asymmetric reactions (Patel 2013). As an alternative,
deracemization of chiral mixtures is used by the pharmaceutical industry to obtain
pure enantiomers. Among chiral compounds, some secondary alcohols are used as
chiral intermediates and analytical reagents, and the development of synthesis
procedures for the production of enantiomerically enriched (enantioenriched)
alcohols has gained importance in the pharmaceutical industry.
Biological systems are generally chiral and, as a consequence, many enzymes
are regio- and enantioselective. These properties are regarded to be a consequence
of the active sites’ architecture and the enzyme’s mechanism. Therefore, many
microorganisms and enzymes offer attractive alternatives for easy production
(asymmetrical synthesis or deracemization) of enantiomeric compounds of interest
in the fine chemicals and pharmaceutical sectors (de Albuquerque et al. 2015;
Matsuda et al. 2009). The AAO catalytic mechanism consists in a hydride
abstraction from the benzylic position of the alcohol by the oxidised flavin, in a
reaction aided by an active-site histidine acting as a catalytic base to form the
alkoxide intermediate (Hernández-Ortega et al. 2012a). Due to active site
12 Fungal Aryl-Alcohol Oxidase … 313

architecture, and the simultaneous nature of the hydride and proton abstractions,
hydride transfer by AAO is stereoselective (only takes place from the pro-
R position) as shown using the two α-monodeuterated enantiomers of p-methox-
ybenzyl alcohol (Hernández-Ortega et al. 2012b). Taking advantage of this infor-
mation, Hernández-Ortega et al. (2012b) assayed the transformation of racemic
secondary alcohols using P. eryngii AAO. They saw that the enzyme was able to
oxidise the racemic 1-(p-methoxyphenyl)-ethanol, although it showed an apparent
efficiency orders of magnitude smaller than the one for p-methoxybenzyl alcohol. In
this reaction, AAO enantioselectivity towards the (S) isomer was shown using
chiral HPLC, which allows for the isolation of (R) isomer from chiral mixtures
(Fig. 12.3). Moreover, the AAO enantioselectivity towards another secondary
alcohol 1-(p-fluorophenyl)-ethanol was estimated as a S/R ratio of 21, in reactions
using the individual enantiomers.
Therefore, it is plausible that AAO could be used for the isolation of isomers
from racemates taking advantage of its kinetic behaviour. However, AAO activity
on secondary alcohols is low, due to some hindrances among such substrates and
the residues forming the active site (Fernández et al. 2009). A mutated variant
(F501A), in which the side chain of a bulky aromatic residue was removed to make
room in the cavity, was created with the purpose of facilitating oxidation of sec-
ondary alcohols. The removal of this side chain resulted in a stereoselectivity S/
R ratio on 1-(p-fluorophenyl)-ethanol three-fold higher than that of the wild-type
enzyme. Hence, improved variants by directed evolution, as it has been done with
galactose oxidase for these purposes (Escalettes and Turner 2008), or further
site-directed mutagenesis would result in better deracemization reactions.

(a) (b)
(R)-isomer (S)-isomer (R)-isomer
A225

A225

30 35 40 45 50 55 30 35 40 45 50 55
Retention time (min) Retention time (min)

Fig. 12.3 Chiral HPLC chromatograms showing the deracemization of 1-(p-methoxyphenyl)-


ethanol by P. eryngii AAO. a Chromatogram of the untreated racemate. b Chromatogram after
24-h reaction with AAO, where only the peak of the (R) isomer is detected (the peak of the p-
methoxyacetophenone, formed from oxidation of the (S) isomer, eluted in a different region of the
chromatogram)
314 J. Carro et al.

12.4.3 Oxidation of Furfurals

Another remarkable class of chemicals derived from lignocellulosics, which are


substrates of AAO, is furfurals. They are formed as by-products of the pretreatment
of lignocellulosics in the paper pulp and the bioethanol industries. In spite of the
fact that they imply the loss of raw material in such processes, they are now
regarded as valorised by-products in biorefinery processes. Moreover,
5-hydroxymethylfurfural (HMF hereinafter) is commercially produced from the
dehydration of hexoses, mainly fructose, (Karinen et al. 2011) but direct production
from glucose is also possible (Zhao et al. 2007).
HMF is regarded as a chemical building block due to the presence of several
functionalities within the molecule (furan ring and hydroxyl and carbonyl groups)
(Rosatella et al. 2011), which may undergo chemical reactions that give rise to a
bunch of compounds: phenolic resins, bioplastics, Schiff bases, polyurethane foams
of epoxy resins, as well as disubstituted furan derivatives (Moreau et al. 2004). The
obtention of 2,5-furandicarboxylic acid (FDCA hereinafter) from HMF (Fig. 12.4)
is of great interest since FDCA can polymerise to give rise to polyesters that may
substitute for those produced from fossil fuels. Such polymers are called poly
(ethylene furandicarbolylate) polyesters (PEFs) and show good mechanical and
barrier properties, are biodegradable and come from renewable resources
(Papageorgiou et al. 2014).
Several processes involving enzymes and organisms have been reported aiming
at the obtention of FDCA from HMF (Carro et al. 2015; Dijkman et al. 2014, 2015;
Dijkman and Fraaije 2014; Hanke 2012; Koopman et al. 2010; van Deurzen et al.
1997). Regarding the use of AAO with this purpose, Hanke (2012) screened for
available AAOs capable of oxidising HMF with variable results. With the same
purpose, Carro et al. (2015) analysed the ability of P. eryngii AAO to oxidise HMF.

Fig. 12.4 Oxidative pathways from HMF (1) to FDCA (5). FFCA (4) formation can take place
through two alternative intermediate compounds: DFF (2) or HMFCA (3)
12 Fungal Aryl-Alcohol Oxidase … 315

In this way, it proved to be catalytically active towards HMF and to be able to


catalyse two subsequent oxidations in its molecule giving rise to
5-formylfurancarboxylic acid (FFCA, hereinafter; 84 %) and FDCA (6 %) in 24 h.
When they tested the ability to oxidise the intermediates between HMF and FDCA,
AAO proved to be catalytically active on diformylfuran (DFF), producing 86 % of
conversion into FFCA and a 14 % into FDCA. However, it did not react either with
5-hydroxymethylfurancarboxylic acid (HMFCA) or with FFCA. NMR studies of
the compounds involved in this chemical pathway demonstrated that carbonyl
groups of both DFF and FFCA were hydrated in aqueous medium giving rise to
diols in 53 and 8 % of abundance, respectively. These gem-diols resulting from the
hydration of aldehydes had been reported to be substrates of the P. eryngii AAO
(Ferreira et al. 2010). However, HMF underwent no hydration, which agrees with
AAO being active towards DFF (formed by oxidation of the HMF hydroxyl group),
but not towards HMFCA, which would be formed by a prior oxidation of the HMF
carbonyl group. Such results suggest that the oxidative pathway for the oxidation of
HMF to FFCA catalysed by this AAO proceeded via DFF. Although this enzyme
was not able to oxidise FFCA, this constraint was overcome through the application
of a sequential enzymatic cascade together with UPO (Ullrich et al. 2004). This
enzyme used the H2O2 produced by AAO to introduce a hydroxyl group in the
formyl group of FFCA, thus producing FDCA, attaining 91 % of conversion after
120 h (Fig. 12.5).
These findings paved the way to implement AAO together with the peroxyge-
nase to obtain a valorised by-product originated from lignocellulosic biomass.
Furthermore, they widened the spectrum of its aromatic substrates, which is now
thought to range from benzylic carbocycles to heterocycles, as HMF and DFF.

AAO
13±1 s-1 mM-1
1 2
O2 H2O2 47%

AAO
9±1 s-1 mM-1 UPO
2’ 4 5
53% O2 H2O2 H2O2 H2O

Fig. 12.5 Sequential enzymatic cascade for the production of FDCA (5) from HMF (1) using
P. eryngii AAO and A. aegerita UPO via DFF (2) and its hydrated gem-diol counterpart (2′)
(abundance of DFF and its gem-diol at equilibrium shown as percentage), and FFCA (4). AAO
catalytic efficiencies are indicated
316 J. Carro et al.

12.5 Conclusions

The finite character of fossil fuels makes it necessary to find new resources,
preferentially renewable ones, which allow for the substitution of the classical
resources. In this way, lignocelluloses are a remarkable material due to their
ubiquitous and renewable character. Furthermore, the concern about the environ-
ment impels us to search for new catalytic procedures that take advantage from
natural processes, instead of the chemical environmentally polluting and
energy-wasting ones. Hence, the use of organisms or enzymes to carry out catalytic
industrial processes is gaining importance.
AAO is a very promiscuous enzyme that catalyses the oxidation of a great deal
of polyunsaturated alcohols (and hydrated aldehydes). Its applicability on some
biotechnology-based industrial processes has been reviewed here and it has showed
to be promising as a biocatalyst in several conversions. AAO, because of its
involvement in the lignocellulose decay process, has potential to be applied to
lignocellulose biorefineries. AAO has so far shown its potential applications in
paper pulp manufacture and bioconversion of lignocellulose-derived compounds
(such as HMF), synthesis of flavours, and deracemization of chiral alcohols
(Fig. 12.6). In spite of the fact that the use of enzymes (among which AAO) for
these purposes is not yet sufficiently developed, much progress has been done the
recent years in this field as one can infer from the huge amount of publications
available on enzymatic biocatalysis.

Oxidation
O id ti off D
Deracemization
i ti Fl
Flavour and
d C ll l
Cellulose
furfurals of chiral alcohols aroma synthesis industries

Applications
pp cat o s
AAO
Physiology

H2O2 production

Substrate of Production of
peroxidases hydroxyl radical

Cellulose
depolymerisation
and lignin oxidation

Fig. 12.6 Scheme depicting the physiological role of AAO in lignocellulose degradation and its
potential applications in the cellulose and other chemical sectors
12 Fungal Aryl-Alcohol Oxidase … 317

Acknowledgments This work was supported by the HIPOP (BIO2011-26694) and the NOESIS
(BIO2014-56388-R) projects of the Spanish Ministry of Economy and Competitiveness, and the
INDOX (KBBE-2013-7-613549) and EnzOx2 (H2020-BBI-PPP-2015-RIA-720297) European
projects. J.C. acknowledges a FPU fellowship of the Spanish Ministry of Education, Culture and
Sport.

References

Akhtar M, Blanchette RA, Kirk TK (1997) Fungal delignification and biomechanical pulping of
wood. In: Scheper T (ed) Advances in biochemical engineering/biotechnology. Springer,
Berlin, pp 160–195
Arantes V, Jellison J, Goodell B (2012) Peculiarities of brown-rot fungi and biochemical Fenton
reaction with regard to their potential as a model for bioprocessing biomass. Appl Microbiol
Biotechnol 94:323–338
Ayers AR, Ayers SB, Eriksson K-E (1978) Cellobiose oxidase, purification and partial
characterization of a hemoprotein from Sporotrichum pulverulentum. Eur J Biochem
90:171–181
Baldrian P, Valaskova V (2008) Degradation of cellulose by basidiomycetous fungi. FEMS
Microbiol Rev 32:501–521
Bao WJ, Usha SN, Renganathan V (1993) Purification and characterization of cellobiose
dehydrogenase, a novel extracellular hemoflavoenzyme from the white-rot fungus
Phanerochaete chrysosporium. Arch Biochem Biophys 300:705–713
Bao WL, Fukushima Y, Jensen KA, Moen MA, Hammel KE (1994) Oxidative degradation of
non-phenolic lignin during lipid peroxidation by fungal manganese peroxidase. FEBS Lett
354:297–300
Barrasa JM, Gutiérrez A, Escaso V, Guillén F, Martínez MJ, Martínez AT (1998) Electron and
fluorescence microscopy of extracellular glucan and aryl-alcohol oxidase during wheat-straw
degradation by Pleurotus eryngii. Appl Environ Microbiol 64:325–332
Bes B, Ranjeva R, Boudet AM (1983) Evidence for the involvement of activated oxygen in fungal
degradation of lignocellulose. Biochimie 65:283–289
Blanchette RA, Burnes TA, Eerdmans MM, Akhtar M (1992) Evaluating isolates of
Phanerochaete chrysosporium and Ceriporiopsis subvermispora for use in biological pulping
processes. Holzforschung 46:109–115
Bourbonnais R, Paice MG (1988) Veratryl alcohol oxidases from the lignin degrading
basidiomycete Pleurotus sajor-caju. Biochem J 255:445–450
Bozell JJ, Petersen GR (2010) Technology development for the production of biobased products
from biorefinery carbohydrates—the US Department of Energy’s “Top 10” revisited. Green
Chem 12:539–554
Breen A, Singleton FL (1999) Fungi in lignocellulose breakdown and biopulping. Curr Opin
Biotechnol 10:252–258
Camarero S, Barrasa JM, Pelayo M, Martínez AT (1998) Evaluation of Pleurotus species for
wheat-straw biopulping. J Pulp Pap Sci 24:197–203
Camarero S, Böckle B, Martínez MJ, Martínez AT (1996) Manganese-mediated lignin degradation
by Pleurotus pulmonarius. Appl Environ Microbiol 62:1070–1072
Carey JS, Laffan D, Thomson C, Williams MT (2006) Analysis of the reactions used for the
preparation of drug candidate molecules. Org Biomol Chem 4:2337–2347
Carro J, Ferreira P, Rodríguez L, Prieto A, Serrano A, Balcells B, Ardá A, Jiménez-Barbero J,
Gutiérrez A, Ullrich R, Hofrichter M, Martínez AT (2015) 5-Hydroxymethylfurfural
conversion by fungal aryl-alcohol oxidase and unspecific peroxygenase. FEBS J 282
(16):3218–3229
318 J. Carro et al.

Cavener DR (1992) GMC oxidoreductases. A newly defined family of homologous proteins with
diverse catalytic activities. J Mol Biol 223:811–814
Daniel G, Volc J, Filonova L, Plihal O, Kubátová E, Halada P (2007) Characteristics of
Gloeophyllum trabeum alcohol oxidase, an extracellular source of H2O2 in brown rot decay of
wood. Appl Environ Microbiol 73:6241–6253
Daniel G, Volc J, Kubátová E (1994) Pyranose oxidase, a major source of H2O2 during wood
degradation by Phanerochaete chrysosporium, Trametes versicolor, and Oudemansiella
mucida. Appl Environ Microbiol 60:2524–2532
de Albuquerque NCP, de Gaitani CM, de Oliveira ARM (2015) A new and fast DLLME-CE
method for the enantioselective analysis of zopiclone and its active metabolite after fungal
biotransformation. J Pharm Biomed Anal 109:192–201
de Jong E, Cazemier AE, Field JA, de Bont JAM (1994) Physiological role of chlorinated aryl
alcohols biosynthesized de novo by the white rot fungus Bjerkandera sp. strain BOS55. Appl
Environ Microbiol 60:271–277
de Jong E, Field JA, Dings JAFM, Wijnberg JBPA, de Bont JAM (1992) De novo biosynthesis of
chlorinated aromatics by the white-rot fungus Bjerkandera sp. BOS55. Formation of
3-chloro-anisaldehyde from glucose. FEBS Lett 305:220–224
Dijkman WP, Fraaije MW (2014) Discovery and characterization of a 5-hydroxymethylfurfural
oxidase from Methylovorus sp strain MP688. Appl Environ Microbiol 80:1082–1090
Dijkman WP, Groothuis DE, Fraaije MW (2014) Enzyme-catalyzed oxidation of
5-hydroxymethylfurfural to furan-2,5-dicarboxylic acid. Angew Chem 126:6633–6636
Dijkman WP, Binda C, Fraaije MW, Mattevi A (2015) Structure-based enzyme tailoring of
5-hydroxymethylfurfural oxidase. ACS Catal 5:1833–1839
Eriksson K-E, Pettersson B, Volc J, Musílek V (1986) Formation and partial characterization of
glucose-2-oxidase, a H2O2 producing enzyme in Phanerochaete chrysosporium. Appl
Microbiol Biotechnol 23:257–262
Escalettes F, Turner NJ (2008) Directed evolution of galactose oxidase: generation of
enantioselective secondary alcohol oxidases. ChemBioChem 9:857–860
Evans CS, Dutton MV, Guillén F, Veness RG (1994) Enzymes and small molecular mass agents
involved with lignocellulose degradation. FEMS Microbiol Rev 13:235–240
Faison BD, Kirk TK (1983) Relationship between lignin degradation and production of reduced
oxygen species by Phanerochaete chrysosporium. Appl Environ Microbiol 46:1140–1145
Farmer VC, Henderson MEK, Russell JD (1960) Aromatic-alcohol-oxidase activity in the growth
medium of Polystictus versicolor. Biochem J 74:257–262
Fernández IS, Ruiz-Dueñas FJ, Santillana E, Ferreira P, Martínez MJ, Martínez AT, Romero A
(2009) Novel structural features in the GMC family of oxidoreductases revealed by the crystal
structure of fungal aryl-alcohol oxidase. Acta Crystallogr D Biol Crystallogr 65:1196–1205
Fernández-Fueyo E, Ruiz-Dueñas FJ, Ferreira P, Floudas D, Hibbett DS, Canessa P, Larrondo L,
James TY, Seelenfreund D, Lobos S, Polanco R, Tello M, Honda Y, Watanabe T, Watanabe T,
Ryu JS, Kubicek CP, Schmoll M, Gaskell J, Hammel KE, St. John FJ, Vanden
Wymelenberg A, Sabat G, Bondurant SS, Syed K, Yadav J, Doddapaneni H,
Subramanian V, Lavín JL, Oguiza JA, Perez G, Pisabarro AG, Ramírez L, Santoyo F,
Master E, Coutinho PM, Henrissat B, Lombard V, Magnuson JK, Kües U, Hori C, Igarashi K,
Samejima M, Held BW, Barry K, LaButti K, Lapidus A, Lindquist E, Lucas S, Riley R,
Salamov A, Hoffmeister D, Schwenk D, Hadar Y, Yarden O, de Vries RP, Wiebenga A,
Stenlid J, Eastwood DC, Grigoriev IV, Berka R, Blanchette RA, Kersten P, Martínez AT,
Vicuña R, Cullen D (2012) Comparative genomics of Ceriporiopisis subvermispora and
Phanerochaete chrysosporium provide insight into selective ligninolysis. Proc Natl Acad
Sci USA 109:5458–5463
Ferraz A, Guerra A, Mendonça R, Masarin F, Vicentim MP, Aguiar A, Pavan PC (2008)
Technological advances and mechanistic basis for fungal biopulping. Enzyme Microb Technol
43:178–185
Ferreira P, Carro J, Serrano A, Martínez AT (2015) A survey of genes encoding H2O2-producing
GMC oxidoreductases in 10 Polyporales genomes. Mycologia 107:1105–1119
12 Fungal Aryl-Alcohol Oxidase … 319

Ferreira P, Hernández-Ortega A, Herguedas B, Rencoret J, Gutiérrez A, Martínez MJ,


Jiménez-Barbero J, Medina M, Martínez AT (2010) Kinetic and chemical characterization
of aldehyde oxidation by fungal aryl-alcohol oxidase. Biochem J 425:585–593
Ferreira P, Medina M, Guillén F, Martínez MJ, van Berkel WJH, Martínez AT (2005) Spectral and
catalytic properties of aryl-alcohol oxidase, a fungal flavoenzyme acting on polyunsaturated
alcohols. Biochem J 389:731–738
Floudas D, Binder M, Riley R, Barry K, Blanchette RA, Henrissat B, Martínez AT, Otillar R,
Spatafora JW, Yadav JS, Aerts A, Benoit I, Boyd A, Carlson A, Copeland A, Coutinho PM, de
Vries RP, Ferreira P, Findley K, Foster B, Gaskell J, Glotzer D, Górecki P, Heitman J,
Hesse C, Hori C, Igarashi K, Jurgens JA, Kallen N, Kersten P, Kohler A, Kües U,
Kumar TKA, Kuo A, LaButti K, Larrondo LF, Lindquist E, Ling A, Lombard V, Lucas S,
Lundell T, Martin R, McLaughlin DJ, Morgenstern I, Morin E, Murat C, Nolan M, Ohm RA,
Patyshakuliyeva A, Rokas A, Ruiz-Dueñas FJ, Sabat G, Salamov A, Samejima M, Schmutz J,
Slot JC, St. John F, Stenlid J, Sun H, Sun S, Syed K, Tsang A, Wiebenga A, Young D,
Pisabarro A, Eastwood DC, Martin F, Cullen D, Grigoriev IV, Hibbett DS (2012) The
Paleozoic origin of enzymatic lignin decomposition reconstructed from 31 fungal genomes.
Science 336:1715–1719
Forney LJ, Reddy CA, Tien M, Aust SD (1982) The involvement of hydroxyl radical derived from
hydrogen peroxide in lignin degradation by the white rot fungus Phanerochaete chrysospo-
rium. J Biol Chem 257:11455–11462
Fraatz MA, Zorn H (2011) Fungal flavours. In: Hofrichter M (ed) The mycota. X industrial
applications. Springer, Berlin, pp 249–268
Gallagher IM, Fraser MA, Evans CS, Atkey PT (1989) Ultrastructural localization of
lignocellulose-degrading enzymes. In: Lewis NG, Paice MG (eds) ACS symposium “plant
cell-wall polymers: biogenesis and biodegradation”, vol 399, American Chemical Society,
pp 426–442
Gellerstedt G, Henriksson G (2008) Lignins: major sources, structure and properties. In:
Belgacem M, Gandini A (eds) Monomers, polymers and composites from renewable resources.
Elsevier, Amsterdam, pp 201–224
Giles RL, Galloway ER, Zackeru JC, Naithani V, Parrow MW (2014) Two stage fungal
biopulping solubilizes lignocellulosic carbohydrates without supplemental enzymatic hydrol-
ysis. Int Biodeterior Biodegradation 86:265–271
Goetghebeur M, Brun S, Galzy P, Nicolas M (1993) Benzyl alcohol oxidase and laccase synthesis
in Botrytis cinerea. Biosci Biotechnol Biochem 57:1380–1381
Gómez-Toribio V, García-Martín AB, Martínez MJ, Martínez AT, Guillén F (2009) Induction of
extracellular hydroxyl radical production by white-rot fungi through quinone redox cycling.
Appl Environ Microbiol 75:3944–3953
Goodell B, Jellison J, Liu J, Daniel G, Paszczynski A, Fekete F, Krishnamurthy S, Jun L, Xu G
(1997) Low molecular weight chelators and phenolic compounds isolated from wood decay
fungi and their role in the fungal biodegradation of wood. J Biotechnol 53:133–162
Green F III, Clausen CA, Larsen MJ, Highley TL (1992) Immuno-scanning electron microscopic
localization of extracellular wood-degrading enzymes within the fibrillar sheath of the
brown-rot fungus Postia placenta. Can J Microbiol 38:898–904
Guillén F, Evans CS (1994) Anisaldehyde and veratraldehyde acting as redox cycling agents for
H2O2 production by Pleurotus eryngii. Appl Environ Microbiol 60:2811–2817
Guillén F, Martínez AT, Martínez MJ (1990) Production of hydrogen peroxide by aryl-alcohol
oxidase from the ligninolytic fungus Pleurotus eryngii. Appl Microbiol Biotechnol 32:465–469
Guillén F, Martínez AT, Martínez MJ (1992) Substrate specificity and properties of the
aryl-alcohol oxidase from the ligninolytic fungus Pleurotus eryngii. Eur J Biochem 209:
603–611
Guillén F, Martínez AT, Martínez MJ, Evans CS (1994) Hydrogen peroxide-producing system of
Pleurotus eryngii involving the extracellular enzyme aryl-alcohol oxidase. Appl Microbiol
Biotechnol 41:465–470
320 J. Carro et al.

Gutiérrez A, Caramelo L, Prieto A, Martínez MJ, Martínez AT (1994) Anisaldehyde production


and aryl-alcohol oxidase and dehydrogenase activities in ligninolytic fungi from the genus
Pleurotus. Appl Environ Microbiol 60:1783–1788
Halliwell G (1965) Catalytic decomposition of cellulose under biological conditions. Biochem J
95:35–40
Hanke PD (2012) Enzymatic oxidation of hydroxymethylfurfural. Patent (USA) 8,183,020 B2
Hernández-Ortega A, Ferreira P, Martínez AT (2012a) Fungal aryl-alcohol oxidase: a
peroxide-producing flavoenzyme involved in lignin degradation. Appl Microbiol Biotechnol
93:1395–1410
Hernández-Ortega A, Ferreira P, Merino P, Medina M, Guallar V, Martínez AT (2012b)
Stereoselective hydride transfer by aryl-alcohol oxidase, a member of the GMC superfamily.
ChemBioChem 13:427–435
Higuchi T (1997) Biochemistry and molecular biology of wood. Springer, London
Iwahara S, Nishihira T, Jomori T, Kuwahara M, Higuchi T (1980) Enzymic oxidation of α,
β-unsaturated alcohols in the side chains of lignin-related aromatic compounds. J Ferment
Technol 58:183–188
Jensen KA Jr, Evans KMC, Kirk TK, Hammel KE (1994) Biosynthetic pathway for veratryl
alcohol in the ligninolytic fungus Phanerochaete chrysosporium. Appl Environ Microbiol
60:709–714
Karinen R, Vilonen K, Niemelä M (2011) Biorefining: heterogeneously catalyzed reactions of
carbohydrates for the production of furfural and hydroxymethylfurfural. Chemsuschem
4:1002–1016
Kersten P, Cullen D (2014) Copper radical oxidases and related extracellular oxidoreductases of
wood-decay Agaricomycetes. Fungal Genet Biol 72:124–130
Kersten PJ, Kirk TK (1987) Involvement of a new enzyme, glyoxal oxidase, in extracellular H2O2
production by Phanerochaete chrysosporium. J Bacteriol 169:2195–2201
Kimura Y, Asada Y, Kuwahara M (1990) Screening of basidiomycetes for lignin peroxidase genes
using a DNA probe. Appl Microbiol Biotechnol 32:436–442
Kirk TK, Farrell RL (1987) Enzymatic “combustion”: the microbial degradation of lignin. Annu
Rev Microbiol 41:465–505
Koopman F, Wierckx N, de Winde JH, Ruijssenaars HJ (2010) Efficient whole-cell biotransfor-
mation of 5-(hydroxymethyl)furfural into FDCA, 2,5-furandicarboxylic acid. Bioresour
Technol 101:6291–6296
Krings U, Berger RG (1998) Biotechnological production of flavours and fragrances. Appl
Microbiol Biotechnol 49:1–8
Lapadatescu C, Bonnarme P (1999) Production of aryl metabolites in solid-state fermentations of
the white-rot fungus Bjerkandera adusta. Biotechnol Lett 21:763–769
Lapadatescu C, Giniès C, Le Quéré J-L, Bonnarme P (2000) Novel scheme for biosynthesis of aryl
metabolites from L-phenylalanine in the fungus Bjerkandera adusta. Appl Environ Microbiol
66:1517–1522
Lomascolo A, Uzan-Boukhris E, Herpoel-Gimbert I, Sigoillot JC, Lesage-Meessen L (2011)
Peculiarities of Pycnoporus species for applications in biotechnology. Appl Microbiol
Biotechnol 92:1129–1149
Martínez AT, Rencoret J, Nieto L, Jiménez-Barbero J, Gutiérrez A, del Río JC (2011) Selective
lignin and polysaccharide removal in natural fungal decay of wood as evidenced by in situ
structural analyses. Environ Microbiol 13:96–107
Martínez AT, Speranza M, Ruiz-Dueñas FJ, Ferreira P, Camarero S, Guillén F, Martínez MJ,
Gutiérrez A, del Río JC (2005) Biodegradation of lignocellulosics: microbiological, chemical
and enzymatic aspects of fungal attack to lignin. Int Microbiol 8:195–204
Martinez D, Challacombe J, Morgenstern I, Hibbett DS, Schmoll M, Kubicek CP, Ferreira P,
Ruiz-Dueñas FJ, Martínez AT, Kersten P, Hammel KE, Vanden Wymelenberg A, Gaskell J,
Lindquist E, Sabat G, Bondurant SS, Larrondo LF, Canessa P, Vicuña R, Yadav J,
Doddapaneni H, Subramanian V, Pisabarro AG, Lavín JL, Oguiza JA, Master E, Henrissat B,
Coutinho PM, Harris P, Magnuson JK, Baker SE, Bruno K, Kenealy W, Hoegger PJ, Kues U,
12 Fungal Aryl-Alcohol Oxidase … 321

Ramaiya P, Lucas S, Salamov A, Shapiro H, Tu H, Chee CL, Misra M, Xie G, Teter S,


Yaver D, James T, Mokrejs M, Pospisek M, Grigoriev IV, Brettin T, Rokhsar D, Berka R,
Cullen D (2009) Genome, transcriptome, and secretome analysis of wood decay fungus Postia
placenta supports unique mechanisms of lignocellulose conversion. Proc Natl Acad Sci USA
106:1954–1959
Masarin F, Pavan PC, Vicentim MP, Souza-Cruz PB, Loguercio-Leite C, Ferraz A (2009)
Laboratory and mill scale evaluation of biopulping of Eucalyptus grandis Hill ex Maiden with
Phanerochaete chrysosporium RP-78 under non-aseptic conditions. Holzforschung 63:259–
263
Matsuda T, Yamanaka R, Nakamura K (2009) Recent progress in biocatalysis for asymmetric
oxidation and reduction. Tetrahedron-Asymmetry 20:513–557
Moreau C, Belgacem MN, Gandini A (2004) Recent catalytic advances in the chemistry of
substituted furans from carbohydrates and in the ensuing polymers. Top Catal 27:11–30
Muheim A, Leisola MSA, Schoemaker HE (1990a) Aryl-alcohol oxidase and lignin peroxidase
from the white-rot fungus Bjerkandera adusta. J Biotechnol 13:159–167
Muheim A, Waldner R, Leisola MSA, Fiechter A (1990b) An extracellular aryl-alcohol oxidase
from the white-rot fungus Bjerkandera adusta. Enzyme Microb Technol 12:204–209
Niemenmaa O, Uusi-Rauva A, Hatakka A (2008) Demethoxylation of [O14CH3]-labelled lignin
model compounds by the brown-rot fungi Gloeophyllum trabeum and Poria (Postia) placenta.
Biodegradation 19:555–565
Nishida A, Eriksson K-E (1987) Formation, purification, and partial characterization of methanol
oxidase, a H2O2-producing enzyme in Phanerochaete chrysosporium. Biotechnol Appl
Biochem 9:325–338
Okamoto K, Narayama S, Katsuo A, Shigematsu I, Yanase H (2002) Biosynthesis of p-
anisaldehyde by the white-rot basidiomycete Pleurotus ostreatus. J Biosci Bioeng 93:207–210
Otjen L, Blanchette RA (1986) A discussion of microstructural changes in wood during
decomposition by white rot basidiomycetes. Can J Bot 64:905–911
Overhage J, Steinbüchel A, Priefert H (2003) Highly efficient biotransformation of eugenol to
ferulic acid and further conversion to vanillin in recombinant strains of Escherichia coli. Appl
Environ Microbiol 69:6569–6576
Papageorgiou GZ, Tsanaktsis V, Bikiaris DN (2014) Synthesis of poly(ethylene furandicarboxy-
late) polyester using monomers derived from renewable resources: thermal behavior
comparison with PET and PEN. Phys Chem Chem Phys 16:7946–7958
Patel RN (2013) Biocatalytic synthesis of chiral alcohols and amino acids for development of
pharmaceuticals. Biomolecules 3:741–777
Priefert H, Rabenhorst J, Steinbüchel A (2001) Biotechnological production of vanillin. Appl
Microbiol Biotechnol 56:296–314
Rasmussen ML, Shrestha P, Khanal SK, Pometto AL, van Leeuwen J (2010) Sequential
saccharification of corn fiber and ethanol production by the brown rot fungus Gloeophyllum
trabeum. Bioresour Technol 101:3526–3533
Romero E, Ferreira P, Martínez AT, Martínez MJ (2009) New oxidase from Bjerkandera
arthroconidial anamorph that oxidizes both phenolic and nonphenolic benzyl alcohols.
Biochim Biophys Acta 1794:689–697
Romero E, Martínez AT, Martínez MJ (2010) Manganese peroxidase and aryl-alcohol oxidase: a
ligninolytic tandem in Bjerkandera arthroconidial anamorph. In: Proc OxiZymes in Leipzig,
14–16 June
Rosatella AA, Simeonov SP, Frade RFM, Afonso CAM (2011) 5-Hydroxymethylfurfural
(HMF) as a building block platform: Biological properties, synthesis and synthetic
applications. Green Chem 13:754–793
Ruiz-Dueñas FJ, Lundell T, Floudas D, Nagy LG, Barrasa JM, Hibbett DS, Martínez AT (2013)
Lignin-degrading peroxidases in Polyporales: an evolutionary survey based on ten sequenced
genomes. Mycologia 105:1428–1444
322 J. Carro et al.

Ruiz-Dueñas FJ, Martínez AT (2009) Microbial degradation of lignin: how a bulky recalcitrant
polymer is efficiently recycled in nature and how we can take advantage of this. Microb
Biotechnol 2:164–177
Sannia G, Limongi P, Cocca E, Buonocore F, Nitti G, Giardina P (1991) Purification and
characterization of a veratryl alcohol oxidase enzyme from the lignin degrading basidiomycete
Pleurotus ostreatus. Biochim Biophys Acta 1073:114–119
Schwarze FWMR, Baum S, Fink S (2000) Dual modes of degradation by Fistulina hepatica in
xylem cell walls of Quercus robur. Mycol Res 104:846–852
Scott GM, Swaney R (1998) New technologies for papermaking: biopulping economics. Tappi J
81:153–157
Serra S, Fuganti C, Brenna E (2005) Biocatalytic preparation of natural flavours and fragrances.
Trends Biotechnol 23:193–198
Shimada M, Higuchi T (1991) Microbial, enzymatic and biomimetic degradation of lignin. In:
Hon DNS, Shiraishi N (eds) Wood and cellulosic chemistry. Marcel Dekker, New York,
pp 557–619
Sigoillot C, Camarero S, Vidal T, Record E, Asther M, Pérez-Boada M, Martínez MJ, Sigoillot
J-C, Asther M, Colom J, Martínez AT (2005) Comparison of different fungal enzymes for
bleaching high-quality paper pulps. J Biotechnol 115:333–343
Turner WB, Aldridge DC (1983) Fungal metabolites II. Academic Press, London
Ullrich R, Nuske J, Scheibner K, Spantzel J, Hofrichter M (2004) Novel haloperoxidase from the
agaric basidiomycete Agrocybe aegerita oxidizes aryl alcohols and aldehydes. Appl Environ
Microbiol 70:4575–4581
van Deurzen MPJ, van Rantwijk F, Sheldon RA (1997) Chloroperoxidase-catalyzed oxidation of
5-hydroxymethylfurfural. J Carbohydr Chem 16:299–309
Vicentim MP, Faria RD, Ferraz A (2009) High-yield kraft pulping of Eucalyptus grandis Hill ex
Maiden biotreated by Ceriporiopsis subvermispora under two different culture conditions.
Holzforschung 63:408–413
Waldner R, Leisola MSA, Fiechter A (1988) Comparison of ligninolytic activities of selected
white-rot fungi. Appl Microbiol Biotechnol 29:400–407
Yelle DJ, Ralph J, Lu F, Hammel KE (2008) Evidence for cleavage of lignin by a brown rot
basidiomycete. Environ Microbiol 10:1844–1849
Zabel R, Morrell J (1992) Wood microbiology: decay and its prevention. Academic Press, London
Zhao H, Holladay JE, Brown H, Zhang ZC (2007) Metal chlorides in ionic liquid solvents convert
sugars to 5-hydroxymethylfurfural. Science 316:1597–1600
Chapter 13
Monosaccharide Oxygenase

Avnish Kumar, Monika Asthana, Hirawati Deval


and Sarika Amdekar

Abstract Oxygenases are enzymes that catalyze the insertion of oxygen atom into
an organic substrate. With the intention to perform this kind of reactions, those
enzymes want to prompt molecular oxygen to conquer its spin-forbidden response
with the organic substrate. In most cases, oxygenases make use of organic cofactors
to transfer electron to molecular oxygen for its activation. Due to chemo-, regio-,
and/or enantio-selective activity both mono- or dioxygenases are attractive bio-
catalysts. Oxygenases having many organic substrates, of these monosaccharide
reactive oxygenases are focused in this chapter. Most abundant protein on earth
known as Rubisco monooxygenase and 5-carbon monosaccharide, Sialic acid
monooxygenase is well studied but any dioxygenase capable to incorporate O2
atom in monosachharide is still unknown.

13.1 Introduction of Oxygenases and Monosaccharides

Biochemical reactions in which electrons are exchanged starting with one molecule
then onto the next are catalyzed by a wide assortment of enzymes, alluded to as
oxidoreductases or redox catalysts (EC 1.x.x.x) (Dixon and Webb 1979). Taking
into account the kind of reactions they catalyze, oxidoreductases have been sepa-
rated into 22 diverse EC-subclasses. A more broad order was proposed by Xu
(2005), in which these proteins were separated into four subgroups: (i) oxidases,

A. Kumar  M. Asthana (&)


Department of Biotechnology, School of Life Sciences,
Dr. Bhim Rao Ambedkar University, Agra, U.P. 282004, India
e-mail: [email protected]
H. Deval
National Institute of Virology (NIV), Indian Council of Medical Research,
Ministry of Health and Family Welfare, Govt. of India, Gorakhpur Unit,
B.R.D. Medical College Campus, Gorakhpur 273013, India
S. Amdekar
Department of Microbiology, Barkatullah University, Bhopal, M.P 462026, India

© Springer International Publishing Switzerland 2016 323


V.K. Gupta (ed.), Microbial Enzymes in Bioconversions of Biomass,
Biofuel and Biorefinery Technologies 3, DOI 10.1007/978-3-319-43679-1_13
324 A. Kumar et al.

Table 13.1 Dioxygenases and their cofactors


Enzyme Type of reaction Cofactors
Metapyrocatechase Ring cleavage Fe2+
Pyrocatechase Ring cleavage Fe3+
Prostaglandin Formation of a cyclic Heme, Tryptophane,
cyclooxygenase peroxide glutathione
Lipoxygenase Formation of an open Fe2+
peroxide
Cysteamine dioxygenase Oxygenation of sulfur Fe2+, sulfides
Tryptophane Ring cleavage Heme
2,3-dioxygenase
4-Hydroxyphenyl-pyruvate Hydroxylation, oxidative Fe2+, ascorbic acid, required
dioxygenase decarboxylation a-ketoglutarate
Monosaccharide dioxgenases are not found to be explained so far in our search study

Oxydizing enzymes
(Oxygen serve as electron acceptor)

Oxygenase Oxidases Peroxidases Dehydrogenase/


Use O2 as acceptor Use H2O2 to Reductases
produce H 2 O or H 2O2 produce H2O

Di-oxygenase
Mono-Oxygenase

Diverse Iron, Copper, or


flavin dependent

Cytochrome P- Flavin dependent Other


450 dependent Mono-oxygenases

Phenol oxygenase Lipoxygenase

Fig. 13.1 Classification of oxidative enzymes


13 Monosaccharide Oxygenase 325

(ii) peroxidases, (iii) oxygenases/hydroxylases and (iv) dehydrogenases/reductases


(Table 13.1; Fig. 13.1).

13.1.1 Oxygenases

Catalyze the incorporation of oxygen atom into the substrate. They are classified
into two groups: monooxygenase and dioxygenases. Dioxygenases consolidate
both oxygen atoms into the substrate, while monooxygenases incorporate a single
oxygen atom as a hydroxyl group into the substrate and the second oxygen atom is
reduced to water. So as to do this sort of reactions, these catalysts need to actuate
molecular oxygen to overcome its spin-forbidden reaction with the organic sub-
strate. Mostly, monooxygenases use (in)organic cofactors to exchange electrons to
molecular oxygen for its activation.

13.1.2 Monosaccharides

Monosaccharides are a class of carbohydrates that cannot be separated to less


complex sugars by hydrolysis and that constitute the building blocks of oligosac-
charides and polysaccharides. Monosaccharide comprise of no less than three
carbon molecules, one of which is connected to an oxygen atom to shape an
aldehyde group (CHO) or a ketone, and the others of which are each appended to a
hydroxyl group (OH), e.g., glucose, fructose, mannose, glucosamine, man-
nosamine, gluconic acid, sialic acid, uronic acid, N-acetyl glucosamine, N-acetyl
muramic acid, and so forth. Monosaccharide can be occurred as chains or rings.
Six-carbon sugars (hexoses) and five-carbon sugars (pentoses) are the most fre-
quently encountered monosaccharide in nature.

13.2 Monooxygenase Reactions

Oxidoreductases are getting more attention these days for selective oxygenation of
aromatic compounds and a wide range of reactions have been documented using
these enzymes from various microbial sources. Monooxygenases are an emerging
class of biotechnologically relevant enzymes due to their chemo-, regio-, and/or
enantio-selective, making those appealing biocatalysts. Monooxygenases incorpo-
rate one atom from O2 into a substrate that accepts oxygen and using another
substrate that furnishes the two H atoms to reduce the other oxygen to water.
Monooxygenases responds with O2 and supplement one atom of oxygen into the
substrate and second oxygen atom forms water with a reduced co-substrate.
326 A. Kumar et al.

S þ O2 þ RH2 ! SO þ H2 O þ R

Here, RH2 is reduced co-substrate and R is oxidized co-substrate.


If monooxygenase split hydrogen from the substrate itself then it is known as
internal monooxygenase otherwise it is called external monooxygenase.
In monooxygenation reactions, monooxygenases require electrons rather than
O2. Reactions in which organic compounds are oxygenated or hydroxylated are of
great value for organic synthesis. In any case, specific oxyfunctionalization of
organic substrates can be a specific issue in organic synthesis. These reactions are
frequently completed with solid oxidizing operators and they additionally happen
with little chemo-, regio-, and enantio selectivity (Loughlin 2000). These issues are
taken away by managing the use of oxygenases. Under optimum conditions of pH
scale and temperature, aqueous solvent, etc. these enzymes catalyze a specific
oxyfunctionalization of organic substrates. For instance, styrene monooxygenase
was demonstrated enantio specifically catalyze the epoxidation of styrene subor-
dinates, yielding the comparing (S)-epoxides with >99 % e.e. (Panke et al. 1998;
van Hellemond et al. 2007).
Chemoselectivity refers to the selective reactivity of one functional group in the
presence of others; often this process in convoluted and protecting groups are on
the molecular connectivity alone.
Regio selectivity is the preference of one direction of chemical bond making or
breaking of over all other possible directions, e.g., selectively generation of one
constitutional isomer rather than the other.
Enantio selectivity is the production of a compound by a method that selectively
preferred the formation of a specific enantiomer or diastereomer.
All styrene monooxygenases (Fig. 13.2) perform enantio-selective epoxidations
of styrene and chemically analogous compounds, which makes them interesting for
biotechnological applications (Montersino et al. 2011).
In the reaction, monooxygenases (EC 1.13.x.x and EC1.14.x.x) need to enact
molecular oxygen, as no reaction will proceed without actuation due to the spin
state of O2. This actuation endless supply of electrons to sub-atomic oxygen, after
which oxygenation of the natural substrate can happen. The formation of reactive
oxygen intermediate could be identified according to the cofactor present in the

+ +
NAD(P)H+H +O2 NAD (P) + H2O

(Styrene monooxygenase)

Styrene Styrene oxide

Styrene + FADH2 + O2 (S)-2-phenyloxirane + FAD + H2O

Fig. 13.2 Styrene oxygenase reaction


13 Monosaccharide Oxygenase 327

monooxygenase. Sometimes, no cofactor at all is available. internal monooxyge-


nases get these electrons from the substrate itself, though outside monooxygenases
are subject to outer electron contributors, e.g., coenzyme NAD(P)H (van Berkel
et al. 2006a). Torres et al. (2010) has reviewed a brief discourses of the distinctive
groups of monooxygenases, in light of the types of cofactor they require for
catalysis are as take after. Monooxygenases is presented, based on the type of
cofactor that these enzymes require. This includes not only the cytochrome P450
and flavin-dependent monooxygenases, but also enzymes that utilize pterin, metal
ions (copper or iron) or no cofactor at all. As most of these monooxygenases require
nicotinamide coenzymes as electron donors.
Heme-dependent Monooxygenases Additionally alluded to as cytochrome P450
monooxygenases or CYPs (EC 1.14.13.x, EC 1.14.14.x and EC 1.14.15.x) can be
found in numerous life shapes: eukaryotes (warm blooded animals, plants and
organisms) and microbes express a wide assortment of these catalysts (Nelson et al.
1996). CYPs are most inexhaustible in eukaryotes. They are b-sort
heme-subordinate outer monooxygenases and they owe their name to the excep-
tional light assimilation at 450 nm of the lessened CO-bound heme-complex. This
high retention is utilized to assess the centralization of the monooxygenase (Omura
and Sato 1964).
Rather than CYPs, qualities encoding Flavin-dependent monooxygenases have
all the earmarks of being moderately bottomless in prokaryotic genomes. Likewise
eukaryotes have been appeared to contain large portions of these qualities (e.g., the
genome of Arabidopsis thaliana contains 29 quality homologs of purported
‘flavin-containing monooxygenases’) (Schlaich 2007). The flavins used by these
catalysts are either FMN or FAD and are either bound firmly to the chemical
(prosthetic gathering) or capacity as a substrate (coenzyme) (van Berkel et al.
2006b). Flavin-subordinate monooxygenases can catalyze different responses, for
instance epoxidations, Baeyer–Villiger oxidations (Fig. 13.3) and halogenations.
Keeping in mind the end goal to perform these responses, flavin-subordinate
monooxygenases create a receptive transitional by shaping a covalent bond between
atomic oxygen and the C4a of the flavin (Ghisla and Massey 1989). Depending
upon the protonation state, this intermediate is proposed to lead to either

Fig. 13.3 Asymmetric chemical synthetic routes that catalyze selective oxygen insertion are
usually mediated by heavy metals with complex ligands and/or explosive peroxidic oxidizing
agents. A way to circumvent the use of these hazardous materials is by applying the
flavin-dependent Type I Baeyer–Villiger monooxygenases. These enzymes are able to catalyze
a broad range of oxidative reactions such as Baeyer–Villiger and heteroatom oxidations
328 A. Kumar et al.

nucleophilic or electrophilic oxygenations. Although this intermediate is well sta-


blized within the enzyme. (Entsch and van Berkel 1995), it will rot to the oxidized
flavin without a suitable natural substrate or halide, yielding hydrogen peroxide
(H2O2) as an item (Massey 1994).
Copper-dependent Monooxygenases (EC 1.14.17.x) constitute a comparatively
tiny family of enzymes that need copper ions for hydroxylation of their substrates.
Enzymes belonging to this family have primarily been found in organism organ-
isms. The best-studied member of this family is dopamine-monooxygenase (DbM),
a protein that hydroxylates the carbon of Dopastat at the expense of one equivalent
of molecular element and 2 equivalents of ascorbate, thereby yielding nore-
pinephrine as the main product (Levin et al. 1960). In Drosophila melanogaster, the
reaction of Dopastat is administered by aminoalkanoic acid monooxygenase (TbM)
(Monastirioti et al. 1996). Besides DbM or TbM, these organisms contain a pep-
tidylglycine–hydroxylating monooxygenase (PHM) and a monooxygenase X
(MOX) (Xin et al. 2004). The latter monooxygenase was identified for the 1st time
throughout a hunt for genes whose expression is altered in aging human fibroblasts
(Chambers et al. 1998). Furthermore, copper-dependent monooxygenases have
been identified that do not need ascorbate as molecule. An example of these
monooxygenases is that the membrane-associated methane series monooxygenase
(pMMO) from Methylococcus capsulatus. The monooxygenase utilizes two
ascorbate molecules to scale back each copper cofactor from Cu2+ to Cu+.
Thereafter, the enzyme binds the organic substrate and the reduced copper particle
reacts with molecular element to create a copper-peroxide (Aboelella et al. 2004;
Prigge et al. 2004). This reactive intermediate then oxygenates the substrate,
yielding water and the hydroxylated substrate.
Non-heme Iron-dependent Monooxygenases Utilize two iron atoms as chemical
compound for his or her aerophilic activity (Fig. 13.4). These di-iron-dependent
enzymes are conjointly stated as microorganism multicomponent monooxygenases

Fig. 13.4 Proposed catalytic cycle of non-heme iron-dependent monooxygenases (Valentine et al.
1999)
13 Monosaccharide Oxygenase 329

(BMMs), catalyze hydroxylation and epoxidation reactions and consist of 3 com-


ponents; a monooxygenase, a reductase and a tiny restrictive macromolecule
(Wallar and Lipscomb 1996; Leahy et al. 2003). The best-characterized member of
this family is the soluble methane monooxygenase (sMMO) from M. capsulatus
(Murrell et al. 2000).
Other members of this family include alkene monooxygenase (Gallagher et al.
1997), phenol hydroxylases (Whited and Gibson 1991), membrane-bound alkane
hydroxylases (van Beilen and Funhoff 2007) and four-component alkene/aromatic
monooxygenases (e.g., toluene 4-monooxygenase).
Pterin-dependent Monooxygenases (EC 1.14.16.x) is a family of monooxyge-
nases known to hydroxylate the amino acids essential amino acid, tyrosine and
tryptophane at their aromatic ring. Although some of these enzymes are found in
bacterium, they are primarily from organism origin. These monooxygenases bind
one iron atom in their situation with two histidines and a salt and utilize tetrahy-
drobiopterin (BH4) as a molecule. Upon hydroxylation of the substrate, this
coenzyme is regenerate to 4-hydroxybiopterin that, after dehydration, forms qui-
nonoid dihydrobiopterin. In vivo, a NAD(P)H-dependent dihydropteridine reduc-
tase recycles its latter product to BH4 (Fitzpatrick 1999). One of the best-studied
members of this monooxygenase-family is phenylalanine 4-monooxygenase (PheH,
EC 1.14.16.1). However, during chemical change no accumulation of the ferrous
protein has been determined. Therefore, the ferrous protein is thought-about to be
the resting type in absence of substrates. In this state, the enzyme is projected to
bind molecular element in its active site. It has been suggested that O2 is activated
by each the ferrous-ion and therefore the sure reduced tetrahydrobiopterin, after that
the side-product 4-hydroxybiopterin is fashioned additionally to the reactive
FeIVO-intermediate. This intermediate then hydroxylates the substrate, yielding the
product and ferrous protein. However, it is important to notice that the existence of
the reactive intermediates has not been confirmed, however (Fitzpatrick 2003).
Cofactor-independent Monooxygenases These are in majority of monooxyge-
nases. It requires an organic or bronze chemical compound for their chemical
change activity. However, several monooxygenases have been found that do not
need any cofactors. In 1993, tetracenomycin F1 monooxygenase (TcmH) from
Streptomyces glauscens was refined and characterized. This enzyme was shown to
be an internal cofactor-independent monooxygenase, as it required solely molecular
element for its activity and used the substrate as chemical agent. Furthermore,
inhibition studies suggested that sulfhydryl teams and essential amino acid residues
were essential for its aerophilic activity (Shen and Hutchinson 1993). The finding of
new members of this monooxygenase-family showed that a lot of those enzymes
are primarily gift in streptomycetes. For example, ActVA-orf6 from Streptomyces
coelicolorA3(2) is a monooxygenase that is thought to turn the formation of
dihydrokalafungin, an actinorhodin precursor. An exception to these findings could
be a recently known quinolmonooxygenase (YgiN) from E. coli (Adams and Jia
2005). In streptomycetes, the cofactor-independent enzymes appear to be concerned
330 A. Kumar et al.

in the synthesis of polyketide antibiotics, such as the antitumor antibiotic tetra-


cenomycin C. Another role was suggested for a theoretic monooxygenase from
eubacteria T.B. that features a similar crystal structure as ActVA-orf6. This protein
is steered to neutralize reactive element and chemical element species, which are
generated by host cells that are attacked by M. tuberculosis (Lemieux et al. 2005).
Other cofactor-dependent monooxygenases—In 2005, a methyltransferase
homolog was found to hydroxylate the C-10 carbon atom of 15-demethoxy–
rhodomycin. This aclacinomycin 10-hydroxylase from Streptomyces purpurascens
was found to insert an element atom from molecular element into the substrate.
Surprisingly, this enzyme needed S-adenosyl-L-methionine as a cofactor (Jansson
et al. 2003, 2005). It is proposed that the charge of the chemical compound
facilitates the delocalization of electrons upon chemical change of the substrate.
Thereafter, molecular oxygen is activated by the substrate, yielding a hydrox-
yperoxide intermediate. Thus far, this is the sole monooxygenase that has been
identified that does not belong to at least one of the antecedently mentioned
monooxygenase-families.

13.3 Dioxygenase Reactions

Dioxygenases introduce both gas atom of associate gas molecule into a substrate. In
most instances, the two oxygen atoms react with one substrate molecule (in-
tramolecular dioxygenase)

S þ O2 ! SO2

Some dioxygenase, however, incorporate one atom into every of the oxygen
molecule into completely different molecules of the same substrate.

S2 þ O2 ! 2SO

Or into two completely different substrate (intermolecular dioxygenase)

S þ S0 þ O2 ! SO þ S0 O

Dioxygenases include two major classes: haem-dependent iron sulfur dioxyge-


nases and Rieske iron–sulfur non-haem dioxygenases the majority of that is NADH
dependent (Wackett and Hershberger 2001; Burton 2003). The reactions catalyzed
by these enzymes include aromatic ring cleavage and hydroperoxidation or cis-
dihydroxylation (Fig. 13.5). Rieske non-heme iron-dependent oxygenases are
important enzymes that catalyze a wide variety of reactions in the biodegradation of
xenobiotics and the biosynthesis of bioactive natural products.
13 Monosaccharide Oxygenase 331

Pseudomonas putida UV4


Toluene Dioxygenase

Toluene
Industrial synthesis of
Toluene 2,3-cis-dihydrodiol

07 steps to end product from cis-dihydrodiol

6C-Methyl-D-mannose

Fig. 13.5 Toluene Dioxygenase reaction

In result of an expansion in the accessibility of catalysts that can perform par-


ticular oxygenations; an increasing enthusiasm is demonstrated by the enzyme
industries for oxidative biocatalysts.
Rieske oxygenases were first identified as enzymes involved in the degradation
of aromatic compounds by Pseudomonas putida (Fig. 13.6) (Axcell and Geary
1975; Gibson et al. 1968). These enzymes were ultimately characterized as the three
component systems naphthalene dioxygenase and toluene dioxygenase (Yeh et al.
1977; Ensley and Gibson 1983; Ensley et al. 1982). Nowadays, such enzymes are
best known as catalysts of the first step in the oxidative degradation of aromatic
compounds via regio—and stereospecific cis-dihydroxylation to produce dihydro-
diols (Jerina et al. 1971). As regards their substrate specificity some compounds
such as arenes (toluene, naphthalene) and carboxylates (benzoate, phthalate) are
substrate of this type of enzymes.
Nitropropane Dioxygenase FMN Dependent Reaction
2-Nitropropane dioxygenase from Hansenula mrakii is a flavin-dependent enzyme
that catalyzes the oxidation of anionic nitroalkanes into the corresponding carbonyl
compounds and nitrite, with oxygen as the electron acceptor (Fig. 13.6). Although
nitroalkanes are anticipated to be toxic and carcinogenic, they are used widely in
chemical industry for a quick and effective way of synthesizing common reagents.
Consequently, the biochemical and biophysical analysis of 2-nitropropane dioxy-
ganase has a potential for bioremediation purposes.
332 A. Kumar et al.

Fig. 13.6 Nitropropane


Dioxygenase reaction

Nearly all dioxygenases contains iron or copper, which in most instances is a


right away constituent of the enzyme protein. Iron will also be component or
ferroprotoporphyrin IX (Heme). The metallic turns on oxygens connected to the
enzyme, a manner that is associated with polarization of the complex. Inside the
case of the iron containing enzyme this will be formulated as below:

EFe2 þ þ O2 , EFe2 þ  OO

The enzyme oxygen complex adds to the substrate with the elimination of a
proton and the E−Fe2+ complex is regenerated.

13.4 Monosaccharides Monooxygenase

Monooxygenases are much more common than dioxygenases. The oxygenases if


utilize monosaccharides to produce reactive oxygen-intermediate are termed here as
monosacharride oxygenases.
Ribulose 1,5-Bisphosphate (RuBP) Carboxylase/Oxygenase (RubisCO)
Rubisco, the most abundant enzyme in the biosphere (Ellis 1979), fixes CO2 into
organic carbon that supports nearly all life on Earth. In today’s atmosphere, O2
competes with CO2 at Rubisco’s catalytic site, producing the toxic compound
phosphoglycolate (Somerville and Ogren 1979). Phosphoglycolate must be
metabolized at the expense of energy and loss of fixed carbon and nitrogen (Bauwe
et al. 2010). To overcome Rubisco’s limitations, many photosynthetic organisms
have evolved carbon-concentrating mechanisms (CCMs) (Sage et al. 2012;
Giordano et al. 2005). CCMs increase the CO2 concentration around Rubisco,
decreasing O2 competition and enhancing carbon fixation. Ribulose is a ketopen-
tose (monosaccharide) that comprise 5 carbon atoms, and together with a ketone
functional group. Ribulose 1,5-bisphosphate (RuBP) carboxylase/oxygenase
(RubisCO) is one of the precise enzymes of the reductive pentose phosphate
13 Monosaccharide Oxygenase 333

pathway, or Calvin–Bassham–Benson cycle on this pathway, RubisCO capabilities


to catalyze the real CO2 assimilatory step to convert RuBP and CO2 into two
molecules of 3-phosphoglyceric acid via a six-carbon carboxylated intermediate.
The unique activity of oxygenase relative to carboxylase became extraordinarily
small and the most beneficial pH changed into about 9, at which pH the carboxylase
activity was nil. Kinetics and immunological studies confirmed that the large
subunit moiety share the catalytic web site for both carboxylase and oxygenase
response (Nishimura and Akazawa 1974; Takabe and Akazawa 1975). As a con-
sequence RuBP carboxylase is unique in its bifunctional nature and additionally
due to the fact, in contrast to most different oxygenase it lacks characteristics
oxygenase prosthetic groups, i.e., flavin, Cu, and Fe (Chollet et al. 1975); this
activity of the enzyme may be characterized as an internal monooxygenase.
Under conditions of high O2 and low CO2, however, rubisco has oxygenase
activity, initiating the phenomenon called photorespiration (Fig. 13.7).
Rubisco-carboxylase-oxygenase: within the oxygenase reaction, O2 reacts with
rubisco’s 2nd substrate, RuBP, to shape 3 PG and 2 phosphoglycolate.
The charge of carboxylase reaction is four times that of the oxygenase reaction
below everyday atmospheric situations at 25 °C; the stromal awareness of carbon
dioxide is then 10 µM and that of oxygen is 250 µM. The oxygenase response, just
like the carboxylase reaction, requires that lysine 201 be in the carbamate shape.
Due to the fact this carbamate forms only inside the presence of carbon dioxide, this
assets might save you rubisco from catalyzing the oxygenase reaction exclusively
whilst carbon dioxide is absent.

Fig. 13.7 The likely mechanism of oxygenase reactions catalyzed by way of RuBP carboxylase–
oxygenase reactions
334 A. Kumar et al.

CO2 is likewise the sole carbon source for the predominant life-forms on this
planet, and its efficient incorporation into organic matter is directly related to the
productivity of important ecosystems, including agriculturally significant plants.
For maximum organisms, ribulose 1,5-bisphosphate (RuBP)
carboxylase/oxygenase (RubisCO) catalyzes the number one step of CO2 fixation
(Gutteridge and Gatenby 1995; Hartman and Harpel 1994; Tabita 1995); in spite of
the reality that it’s miles the most considerable protein located on earth (Ellis 1979),
RubisCO’s catalytic performance is seriously limited by way of the ability to
catalyze a competing O2 fixation response. This ends in inefficient CO2 fixation and
occasional productiveness. Right now, the molecular foundation for CO2/O2 dis-
crimination is not completely understood; however, the relative capacity of this
enzyme to choose either carboxylation or oxygenation is not immutable, but, varies
for different source of RubisCO (Jordan and Ogren 1981; Read and Tabita 1994).
The pyrenoid is a spherical structure in the chloroplast stroma, discovered more
than 130 years ago (Schmitz 1882; Vaucher 1803; Brown 1967). Pyrenoids have
been found in nearly all of the major oceanic eukaryotic primary producers and
mediate *28–44 % of global carbon fixation (Field et al. 1998; Behrenfeld et al.
2001; Rousseaux and Gregg 2013; Mann 1996; Thierstein and Young 2004; Meyer
and Griffiths 2013). A pyrenoid typically consists of a matrix surrounded by a
starch sheath and traversed by membrane tubules continuous with the photosyn-
thetic thylakoid membranes (Engel et al. 2015). This matrix is thought to consist
primarily of tightly packed Rubisco and its chaperone, Rubisco activase (McKay
and Gibbs 1991). In higher plants and non-pyrenoid-containing photosynthetic
eukaryotes, Rubisco is instead soluble throughout the chloroplast stroma. The
molecular mechanism by which Rubisco aggregates to form the pyrenoid matrix
remains enigmatic.
Two mechanisms for Rubisco accumulation in the pyrenoid have been proposed:
(i) Rubisco holoenzymes could bind each other directly through hydrophobic
residues (Meyer et al. 2012), or (ii) a linker protein may link Rubisco holoenzymes
together (Engel et al. 2015; Meyer et al. 2012). The second model is based on
analogy to the well-characterized prokaryotic carbon-concentrating organelle, the
b-carboxysome, where Rubisco aggregation is mediated by a linker protein con-
sisting of repeats of a domain resembling the Rubisco small subunit (Long et al.
2007).
The reaction, which occurs in C4 plants and bacteria, uses the high energy
phosphate of PEP to drive the incorporation of a carboxyl group in order to form
oxaloacetate. In C4 plants, PEP carboxylase is located in the mesophyll cells on the
external surfaces of the plant. Incorporation of CO2 in this manner helps to shuttle
CO2 to the bundle sheath cells, where the Calvin cycle enzymes are concentrated. It
also helps to avoid the wasteful photorespiration cycle due to the oxygenase activity
of the CO2 fixing enzyme, rubisco.
By catalyzing carbon dioxide fixation during photosynthesis, this enzyme is
responsible for virtually all of the reduced carbon found in living organisms.
Rubisco couples the inorganic and organic carbon pools on earth and is the most
significant route for linking these pools together by the synthesis of carbohydrate. It
13 Monosaccharide Oxygenase 335

Photosynthesis :
CO2 + H2O + ribulose-1,5-P2 2(3-phospho-D-glycerate) -----(Eq.1)

Photorespiration :
O2 + ribulose-1,5-P2 3-phospho-D-glycerate + 2- phosphoglycolate
-----(Eq.2)

Fig. 13.8 Equation of rubisco carboxylase/oxygenase catalytic mechanism during photosynthesis


and photorespiration

is, however, a poor catalyst, having both a low affinity for carbon dioxide and a
small turnover number. Autotrophic organisms must devote a major part of their
synthetic capacity to produce sufficient enzyme to sustain life.
Rubisco catalyzes the first reaction in the pathways of both photosynthesis and
photorespiration, which are, respectively, its carboxylase and oxygenase activities
(Fig. 13.8).
Photosynthesis and photorespiration are interlocking metabolic cycles, with
Rubisco determining the relative rates of carbon flow through the two pathways
(Fig. 13.9). In the dark, plants are carrying out mitochondrial respiration by the
oxidation of substrates to CO2 and the conversion of O2 to H2O. On top of that,
there is another process in plants that, like mitochondrial respiration, consumes O2
and produces CO2 and, like photosynthesis, is driven by light. This process is called
photorespiration and is a costly side reaction of photosynthesis. Rubisco requires
both CO2 and Mg as obligatory cofactors, resulting in carbamylation of an active
site lysine residue by CO2 and coordination of Mg2+.
In the Oxygenase Reaction (Eq. 2) of Photorespiration However, the enediol
intermediate reacts with molecular oxygen to form a hydroperoxy derivative that
breaks down to 2-phosphoglycolate and 3-phosphoglycerate. Photorespiration
consumes ribulose bisphosphate, which results in no net gain of carbon, and also
requires the consumption of energy to recycle the lost carbon. Partitioning of
ribulose bisphosphate between the two reactions of photosynthesis and photores-
piration can vary significantly between different photosynthetic organisms, which
has stimulated interest in improving the carboxylation efficiency of crop plants.
Rubisco activity is modulated in plants by an inhibitor and an activator. The
inhibitor 2′-carboxy arabinitol 1-phosphate (2CAIP) accumulates in some plants
during darkness and binds to the active site of Rubisco. 2CAIP is degraded by a
specific phosphatase, which presumably allows Rubisco to function during pho-
tosynthesis in the light. Rubisco can be severely inhibited by a range of sugar
bisphosphates, including substrate analogs. The enzyme Rubisco activase has the
ability to relieve the inhibition caused by sugar bisphosphates, possibly by inter-
acting with Rubisco and altering the affinity of the enzyme for bisphosphates.
336 A. Kumar et al.

Pyruvate
+
Pi – elimination
3P D-Glycerate

2P-Glycolate

Addition &
Hydration

H2O

abstraction

cis – enediol Xylulose 1, 5 – P2


Ribulose 1, 5 – P2

3-keto arabinitol 1,5 – P2

Fig. 13.9 The reactions catalyzed by Rubisco. The first intermediate of catalysis is the C2, C3 cis-
enediol form of ribulose bisphosphate (ribulose 1,5-P2) after abstraction of the C3 proton. The
enediol can partition a number of ways, the majority into the products of carboxylation (upper
reactions) or oxygenation (lower reactions). However, a number of misprotonated isomers of
ribulose 1,5-P2, for example, xylulose-bisphosphate, have been detected with the wild-type
enzyme that are produced in quantity by mutations of specific amino acid residues involved in
proton transfer. Phosphate elimination of the carbanion forms of intermediates are also produced
by some mutants. R,—CHOH—CH20P02 v 3P D-glycerate, 3-phospho glycerate; 2P glycolate,
2-phosphoglycolate. Five discrete partial reactions have been described for the carboxylation
reaction (Eq. 1) of ribulose bisphosphate. The initial formation of a C2, C3-enediol intermediate of
the ribulose bisphosphate substrate is followed by its carboxylation, where the enediol reacts with
CO2 at the C2 position. The resulting six-carbon intermediate is hydrolytically cleaved to two
molecules of 3-phosphoglycerate, resulting in an overall gain of carbon during photosynthesis
13 Monosaccharide Oxygenase 337

Regulation of Its Enzymatic Activity


RuBisCO is usually only active throughout the day as ribulose 1,5-bisphosphate is
not regenerated within the dark. This can be as a result of the regulation of many
alternative enzymes within the Calvin cycle. Additionally, the activity of RuBisCO is
coordinated thereupon of the opposite enzymes of the Calvin cycle in several ways.
Regulation by Ions
Upon illumination of the chloroplasts, the pH of the stroma rises from 7.0 to 8.0 as
a result of the proton (hydrogen particle, H+) gradient created across the thylakoid
membrane. Simultaneously, magnesium ions (Mg2+) transported outside the thy-
lakoids to raise the concentration level of magnesium within the stroma of the
chloroplasts. RuBisCO includes a high optimum hydrogen ion concentration (can
be >9.0, counting on the metal particle concentration) and, thus, becomes “acti-
vated” by the addition of CO2 and magnesium to the active sites as delineated
above.
Regulation by RuBisCO Activase
In plants and a few alga, another enzyme, RuBisCO activase, is needed to permit the
speedy formation of the crucial carbamate within the site of RuBisCO. RuBisCO
Activase is needed as a result of the ribulose 1,5-bisphosphate (RuBP) substrate
binds more powerfully to the active sites lacking the carbamate and markedly slows
down the “activation” method. During Day time light, RuBisCO activase promotes
the discharge of the repressive, or—in some views—storage RuBP from the catalytic
sites. Activase is also needed in some plants (e.g., tobacco and lots of beans) as a
result of, darkly, RuBisCO is suppressed (or protected against hydrolysis) by a
competitive inhibitor synthesized by these plants, a substrate analog 2-carboxy-D-
arabitinol 1-phosphate (CA1P) (Andralojc et al. 1994). CA1P binds tightly to the site
of carbamylated RuBisCO and inhibits catalytic activity. Within the light, RuBisCO
Activase conjointly promotes the release of CA1P from the catalytic sites. Once the
CA1P is released from RuBisCO, it is speedily converted to a non-inhibitory type by
a light-activated CA1P-phosphatase. Finally, once each many hundred reactions, the
traditional reactions with CO2 or oxygen are not completed, and other repressive
substrate analogs square measure shaped within the site. Once again, RuBisCO
Activase will promote the discharge of those analogs from the chemical action sites
and maintain the catalyst in an exceedingly catalytically active type. The properties
of Activase limit the chemical action potential of plants at high temperatures
(Crafts-Brandner and Salvucci 2000). CA1P has conjointly been shown to stay
RuBisCO in an exceedingly conformation that is protected against chemical action
(Khan et al. 1999). At high temperatures, RuBisCO Activase aggregates and might
now not activate RuBisCO. This contributes to the attenuated carboxylating capa-
bility discovered throughout heat stress (Salvucci et al. 2001).
Regulation by ATP/ADP and Stromal Reduction/Oxidation State Through the
Activase
The removal of the inhibitory RuBP, CA1P, and the other inhibitory substrate
analogs by activase requires the consumption of ATP. This reaction is inhibited by
338 A. Kumar et al.

the presence of ADP, and, thus, activase activity depends on the ratio of these
compounds in the chloroplast stroma. Furthermore, in most plants, the sensitivity of
activase to the ratio of ATP/ADP is modified by the stromal reduction/oxidation
(redox) state through another small regulatory protein, thioredoxin. In this manner,
the activity of activase and the activation state of RuBisCO can be modulated in
response to light intensity and, thus, the rate of formation of the ribulose
1,5-bisphosphate substrate (Zhang et al. 2002).
Regulation by Phosphate
In cyanobacteria, inorganic phosphate (Pi) participates in the coordinated regulation
of photosynthesis. Pi binds to the RuBisCO active site and to another site on the
large chain where it can influence transitions between activated and less active
conformations of the enzyme. Activation of bacterial RuBisCO might be particu-
larly sensitive to Pi levels, which can act in the same way as RuBisCO activase in
higher plants (Marcus and Gurevitz 2000).
Regulation by Carbon Dioxide
Since carbon dioxide and oxygen compete at the active site of RuBisCO, carbon
fixation by RuBisCO can be enhanced by increasing the carbon dioxide level in the
compartment containing RuBisCO (chloroplast stroma). Several times during the
evolution of plants, mechanisms have evolved for increasing the level of carbon
dioxide in the stroma (see C4 carbon fixation). The use of oxygen as a substrate
appears to be a puzzling process, since it seems to throw away captured energy.
However, it may be a mechanism for preventing overload during periods of high light
flux. This weakness in the enzyme is the cause of photorespiration, such that healthy
leaves in bright light may have zero net carbon fixations when the ratio of O2 to CO2
reaches a threshold at which oxygen is fixed instead of carbon. This phenomenon is
primarily temperature-dependent. High temperature decreases the concentration of
CO2 dissolved in the moisture in the leaf tissues. This phenomenon is also related to
water stress. Since plant leaves are evaporatively cooled, limited water causes high
leaf temperatures. C4 plants use the enzyme PEP carboxylase initially, which has a
higher affinity for CO2. The process first makes a 4-carbon intermediate compound,
which is shuttled into a site of C3 photosynthesis then de-carboxylated, releasing
CO2 to boost the concentration of CO2, hence the name C4 plants.
Crassulacean acid metabolism (CAM) plants keep their stomata (on the under-
side of the leaf) closed during the day, which conserves water but prevents the
light-independent reactions (i.e. the Calvin Cycle) from taking place, since these
reactions require CO2 to pass by gas exchange through these openings. Evaporation
through the upper side of a leaf is prevented by a layer of wax.
Sialic Acid Monooxygenase
Sialic acids are a family of nine-carbon acidic monosaccharides that occur obvi-
ously at the cease of sugar chains attached to the surfaces of cells and soluble
proteins. Sialic acids are sugars usually discovered on the outer terminal role of
glycan chains that cover the surface of all vertebrate cells inside the human body,
the very best concentration of sialic acid (as N-acetylneuraminic acid) takes place
13 Monosaccharide Oxygenase 339

within the brain in which it participates as an necessary a part of ganglioside


structure in synaptogenesis and neural transmission.
The maximum generally expressed sialic acid is N-acetylneuraminic acid
(Neu5Ac) that is the precursor for N-glycolylneuraminic acid (Neu5Gc) synthesis—
a conversion mediated by means of motion of the CMP-N-acetylneuraminic acid
hydroxylase (CMAH) enzyme.
Various reactions involving the sialic acids has been given in figure 13.10.
Cells from higher animals and various microorganisms produce sialic acid in a
protracted pathway beginning from glucose.
The CMP-N-acetylneuraminate monooxygenase (CMAH) enzyme converts
Neu5Ac to the Neu5Gc “non-human” form of this sugar, (Fig. 13.10) (Irie et al.
1998). However, the Varki group has proposed that the functional loss of
CMP-N-acetylneuraminate monooxygenase (CMAH) during evolution of humans
from the great apes leads to several possible implications for its role in human
development, the most notable of which being less constrained brain growth (Varki
1992). This premise is supported by evidence that the mutation that abrogated the
activity of CMAH transcripts found in present-day humans occurred after the
Homo-Pan divergence (Chou et al. 1998) but prior to brain expansion during
human evolution (Chou et al. 2002). Unlike most primate brains that stop growing
relatively soon after birth, human brains continue to grow postnatally at a rate
similar to the whole body, presumably because they were freed from the constraints
imposed by Neu5Gc in other mammals that limit brain expansion after birth (Chou
et al. 2002).
In the human body, the highest concentration of sialic acid (as N-acetylneur-
aminic acid) occurs in the brain where it participates as an integral part of gan-
glioside structure in synaptogenesis and neural transmission. Human milk also
contains a high concentration of sialic acid attached to the terminal end of free
oligosaccharides, but its metabolic fate and biological role are currently unknown.
Sialic acids are components of the carbohydrate chains of glycoconjugates and
are involved in cell–cell recognition (Rutishauser et al. 1988; Kelm et al. 1996) and
cell-pathogen interactions (Smit et al. 1984; Suzuki et al. 1987). Sialic acid is a
generic designation used for N-acylneuraminic acids and their derivatives (Varki
1992). N-Acetylneuraminic acid (NeuAc)1 and N-glycolylneuraminic acid (NeuGc)
are two of the most abundant derivatives.
N-Glycolylneuraminic acid (NeuGc) is abundantly expressed in most mammals,
but it is not detectable in humans. The expression of NeuGc is controlled by
cytidine monophospho-N-acetylneuraminic acid (CMP-NeuAc) hydroxylase
activity (Irie et al. 1998).
Monosaccharide monooxygenases are noticeably specialized enzymes that have
evolved to catalyze the insertion of single oxygen atom from molecular oxygen
(O2) into a monosacchride. These enzymes activate molecular oxygen through
generation of a peroxy-intermediate that is capable to carry out the monooxy-
genation reaction. In most cases, monooxygenases utilize bound cofactor(s) which
include flavin, or metal ions to facilitate the activation of molecular oxygen.
340 A. Kumar et al.

Neu5Ac
Neu5Ac
LYSOSOME
Neu5Ac
CMP
CYTOSOL
CMP-Neu5Ac
CMP GOLGI
ManNAc
+
Pyruvate

Glucose
ATP N-Acetylneuramic Acid
9
ADP (Neu5Ac)
Fructose -6- PO4 8 PO4
Glutamine H2 O CMP-N-Acetylneuramic Acid + PPi
1
NH3
Glucosamine -6- PO4 +Glutamic acid N-Acetylneuramic Acid -9- PO4 +Pi
Acetyl CoA
2 7 10
PEP NADPH
N-Acetylglucosamine -6- PO4 N-Acetylmannosamine -6- PO4

3
ADP
N-Acetylglucosamine -1- PO4
CMP-N-glycolylneuramic Acid
UTP ATP
4 O2 & H 2 O
6

UDP-N-Acetylglucosamine
N-glycolylneuraminic Acid
5*
(Neu5Gc)
ManNAc

GlcNAc

Fig. 13.10 Schema displaying the metabolism of sialic acids modified from Varki et al. (1999):
1 Glycosamine-6-phoshate synthase; 2 Glucosamine-6-phosphate N-acetyltransferase; 3
N-acetylglucosamine-!6-phosphate mutase; 4 UDP-N-acetylglucosaminepyrophos-phorylase; 5 *key
enzyme: UDP-N-acetylglucosamine-2-epimerase; activity is initially low in young rats; 6
N-acetylmannosamine kinase; 7 N-acetylneuraminate-9-phosphate synthase; 8 N-acetylneuraminate-9-
phosphatephatase; 9 CMP-N-acetylneuraminatesynthetase; 10 monooxygenase

Examples were described in which monooxygenases utilize the substrate as a


“cofactor” to generate the active peroxy-intermediate. By formation of a
peroxy-intermediate, these enzymes are capable of catalyze an extensive range of
oxidative reactions. The oxidative power of monooxygenases enables to catalyze
13 Monosaccharide Oxygenase 341

wide range of oxidation reactions; e.g., hydroxylations, epoxidations,


heteroatom-dealkylations, dehalogenations, dehydrogenations and oxidation of
heteroatoms. Monooxygenases containing other (or no) cofactors are more limited
of their reactivity scope. Besides the formation of the peroxy-intermediate,
cofactor-dependent monooxygenases have some common characteristic; the elec-
trons which can be required for the reduction of cofactor and thus for the activation
of molecular oxygen are (in)directly provided by nicotinamide coenzymes NADH
or NADPH. Therefore, cost-effective regeneration of these coenzymes is imperative
for successful industrial applications of monooxygenases. Of the various possible
approaches which are described in this chapter. The enzymatic regeneration of
nicotinamide coenzymes is the most advanced and successful so far. Unfortunately,
this confined substrate acceptance limits the applicability of these monooxygenases
as biocatalyst.

13.5 Future Recommendations

A few unidentified monosaccharide monooxygenases and dioxygenses can be


existed in nature. With the identification of new members of cofactor-independent
monooxygenase-family, various studies had been accomplished in order to
understand how these monooxygenases perform their oxidative reactions in the
absence/presence of cofactors. This is indicated by the proposed catalytic mecha-
nism of these enzymes; in contrast to other monooxygenases, molecular oxygen is
activated by the substrate, after which the oxygenation reaction occurs. This sug-
gests that the nature of the substrate is important for the catalytic activity of these
monooxygenases. Internal cofactor-independent monooxygenases are interesting
enzymes for biotechnological applications as they do not require expensive
cofactors/coenzymes.

Appendix 1: Some Reactions of Oxidoreductases

1. Higher plants incorporate large amounts of D-GluA residues into cell wall
glucuronoarabinoxylans. D-GluA forms irreversibly from UDP-D-Glc by UDP-
D-glucose dehydrogenase (EC 1.1.1.22)

+ +
2NAD 2NADH + H

UDP-D-glucose UDP-D-glucuronic acid


(EC1.1.1.22)
342 A. Kumar et al.

2. GDP-L-fucose formation requires subsequent activities of GDP-D-mannose


4,6-dehydratase (EC 4.2.1.47) and a bifunctional GDP-L-fucose synthase (EC
1.1.1.271).

NADPH + H+ NADP+

GDP-6-deoxy-D-lyxo-hexos-4-ulose GDP-L-fucose
(EC 1.1.1.271)

3. N-acetyl-D-glucosamine can be further converted to UDP-N-acetyl-D-glucuronic


acid (UDP-N-acetylglucosamine 6-dehydrogenase, EC 1.1.1.136).

2NAD+ +H2O 2NADH + 2H+


UDP-N-acetyl-2-
UDP-N-acetyl-D-
amino-2-deoxy-D-
glucosamine
glucuronic acid
(EC 1.1.1.136)

4. The peptidoglycan building stone, UDP-N-MurAc, forms from UDP-N-acetyl-


D-glucosamine and phosphoenolpyruvic acid via the intermediate UDP-N-
acetyl-3-O-(1-carboxyvinyl)-D-glucosamine (UDP-N-acetyl-D-glucosamine
1-carboxyvinyl transferase, EC 2.5.1.7), which is reduced to UDP-N-MurAc by
UDP-N-acetylmuramate dehydrogenase (EC 1.1.1.158)

NADH + H+ NAD+
UDP-N-acetyl-3-O- UDP-N-
(1-carboxyvinyl)-D- acetylmuramic
glucosamine (EC 1.1.1.158) acid

5. Biogenesis of UDP-D-GlcA can also occur via an alternative pathway from


myo-inositol, which involves the action of myo-inositol oxygenase (EC
1.13.99.1)

O2 H2O

myo-inositol D-glucuronic
acid
(EC 1.13.99.1)
13 Monosaccharide Oxygenase 343

Appendix 2

Table 13.2 List of oxedoreductases as per their EC number—the enzymes oxidizes a substrate by
transferring the oxygen from molecular oxygen O2 (as in air) to it are called as oxygenases (EC
1.13 or EC 1.14.)
EC 1.1 Acting on the CH–OH group of donors
EC 1.2 Acting on the aldehyde or oxo group of donors
EC 1.3 Acting on the CH–CH group of donors
EC 1.4 Acting on the CH–NH2 group of donors
EC 1.5 Acting on the CH–NH group of donors
EC 1.6 Acting on NADH or NADPH
EC 1.7 Acting on other nitrogenous compounds as donors
EC 1.8 Acting on a sulfur group of donors
EC 1.9 Acting on a heme group of donors
EC 1.10 Acting on diphenols and related substances as donors
EC 1.11 Acting on a peroxide as acceptor
EC 1.12 Acting on hydrogen as donor
EC 1.13 Acting on single donors with incorporation of molecular oxygen (oxygenases)
EC 1.14 Acting on paired donors, with incorporation or reduction of molecular oxygen
EC 1.15 Acting on superoxide radicals as acceptor
EC 1.16 Oxidizing metal ions
EC 1.17 Acting on CH or CH2 groups
EC 1.18 Acting on iron-sulfur proteins as donors
EC 1.19 Acting on reduced flavodoxin as donor
EC 1.20 Acting on phosphorus or arsenic in donors
EC 1.21 Acting on the reaction X–H + Y–H = X–Y
EC 1.22 Acting on halogen in donors
EC 1.23 Reducing C–O–C group as acceptor
EC 1.97 Other oxidoreductases

References

Aboelella NW, Kryatov SV, Gherman BF, Bernnessel WW, Young VG Jr, Sarangi R,
Rybak-Akimova EV, Hogdson KO, Hedman B, Solomon EI, Cramer CJ, Tolman WB (2004)
Dioxygen activation at a single copper site: structure, bonding and mechanism of formation of
1:1 Cu-O2 adducts. J Am Chem Soc 126:16896–16911
Adams MA, Jia Z (2005) Structural and biochemical evidence for an enzymatic quinone redox
cycle in Escherichia coli—identification of a novel quinol monooxygenase. J Biol Chem
280:8358–8363
Andralojc PJ, Dawson GW, Parry MA, Keys AJ (1994). Incorporation of carbon from
photosynthetic products into 2-carboxyarabinitol-1-phosphate and 2-carboxyarabinitol.
Biochem J 304(3):781–786. PMC 1137402. PMID 7818481
344 A. Kumar et al.

Axcell BC, Geary PJ (1975) Purification and some properties of a soluble benzene-oxidizing
system from a strain of Pseudomonas. Biochem J 146:173–183
Bauwe H, Hagemann M, Fernie AR (2010) Photorespiration: players, partners and origin. Trends
Plant Sci 15(6):330–336
Behrenfeld MJ et al (2001) Biospheric primary production during an ENSO transition. Science 291
(5513):2594–2597. Abstract/FREE Full Text
Brown R (1967) Pyrenoid: its structure distribution and function. J Phycol 3(Suppl 1):5–7
Burton SG (2003) Oxidising enzymes as biocatalysts. Trends Biotechnol 21:543–549
Chambers KJ, Tonkin LA, Chang E, Shelton DN, Linskens MH, Funk WD (1998) Identification
and cloning of a sequence homologue of dopamine betahydroxylase. Gene 218:111–120
Chollet R, Anderson LL, Hovsepian LC (1975) The absence of tightly bound copper, iron, and
flavin nucleotide in crystalline ribulose 1,5-bisphosphate carboxylase–oxygenase from tobacco.
Biochem Biophy Res Commun 64:97–107
Chou HH, Takematsu H, Diaz S, Iber J, Nickerson E, Wright KL, Muchmore EA, Nelson DL,
Warren ST, Varki A (1998) A mutation in human CMP-sialic acid hydroxylase occurred after
the Homo-Pan divergence. Proc Natl Acad Sci USA 95:11751–11756
Chou HH, Hayakawa T, Diaz S, Krings M, Indriati E, Leakey M, Paabo S, Satta Y, Takahata N,
Varki A (2002) Inactivation of CMP-N-acetylneuraminic acid hydroxylase occurred prior to
brain expansion during human evolution. Proc Natl Acad Sci USA 99:11736–11741
Crafts-Brandner SJ, Salvucci ME (2000) Rubisco activase constrains the photosynthetic potential
of leaves at high temperature and CO2. Proc Natl Acad Sci USA 97(24):13430–13435.
Bibcode:2000PNAS…9713430C. doi:10.1073/pnas.230451497. PMC 27241. PMID
11069297
Dixon M, Webb EC (1979) Enzymes, 3rd edn. pp 218–220, Academic Press: New York
Ellis RJ (1979) The most abundant protein in the world. Trends Biochem Sci 4(11):241–244
Engel BD et al (2015) Native architecture of the Chlamydomonas chloroplast revealed by in situ
cryo-electron tomography. eLife 4:e04889
Ensley BD, Gibson DT (1983) Naphthalene dioxygenase: purification and properties of a terminal
oxygenase component. J Bacteriol 155:505–511
Ensley BD, Gibson DT, Laborde AL (1982) Oxidation of naphthalene by a multicomponent
enzyme system from Pseudomonas sp. strain NCIB 9816. J Bacteriol 149:948–954
Entsch B, van Berkel WJH (1995) Flavoprotein structure and mechanism. 1. Structure and
mechanism of p-hydroxybenzoate hydroxylase. FASEB J 9:476–483
Field CB, Behrenfeld MJ, Randerson JT, Falkowski P (1998) Primary production of the biosphere:
integrating terrestrial and oceanic components. Science 281(5374):237–240
Fitzpatrick PF (1999) Tetrahydropterin-dependent amino acid hydroxylases. Ann Rev Biochem
68:355–381
Fitzpatrick PF (2003) Mechanism of aromatic amino acid hydroxylation. Biochemistry 42:14084–
14091
Gallagher SC, Cammark R, Dalton H (1997) Alkene monooxygenase from Nocardia corallina
B-276 is a member of the class of dinuclear iron proteins capable of stereospecific
epoxygenation reactions. Eur J Biochem 247:635–641
Ghisla S, Massey V (1989) Mechanisms of flavoprotein catalyzed reactions. Eur J Biochem 181:1–
17
Gibson DT, Koch JR, Kallio RE (1968) Oxidative degradation of aromatic hydrocarbons by
microorganisms. I. Enzymatic formation of catechol from benzene. Biochemistry 7:2653–2662
Giordano M, Beardall J, Raven JA (2005) CO2 concentrating mechanisms in algae: mechanisms,
environmental modulation, and evolution. Annu Rev Plant Biol 56:99–131
Gutteridge S, Gatenby AA (1995) RubisCO synthesis, meeting, mechanism, and regulation. Plant
Mobile 7:809–819
Hartman FC, Harpel MR (1994) Shape, function, law and assembly of D-ribulose-1,5-bisphosphate
carboxylase/oxygenase. Annu Rev Biochem 63:197–234
Irie A, Koyama S, Kozutsumi Y, Kawasaki T, Suzuki A (1998) The molecular basis for the
absence of N-glycolylneuraminic acid in humans. J Biol Chem 273:15866–15871
13 Monosaccharide Oxygenase 345

Jansson A, Niemi J, Lindqvist Y, Mantsala P, Schneider G (2003) Crystal structure of


aclacinomycin-10-hydroxylase, a S-adenosyl-L-methionine-dependent methyltransferase
homolog involved in anthracycline biosynthesis in Streptomyces purpurascens. J Mol Biol
334:269–280
Jansson A, Koskiniemi H, Erola A, Wang J, Mäntsälä P, Schneider G, Niemi J (2005)
Aclacinomycin 10-hydroxylase is a novel substrate-assisted hydroxylase requiring S-adenosyl-
L-methionine as cofactor. J Biol Chem 280:3636–3644
Jerina DM, Daly JW, Jeffrey AM, Gibson DT (1971) cis-1,2-dihydroxy-1,2-dihydronaphthalene: a
bacterial metabolite from naphthalene. Arch Biochem Biophys 142:394–396
Jordan DB, Ogren WL (1981) Species variant inside the specificity of ribulosebisphosphate
carboxylase/oxygenase. Nature 291:513–515
Kelm S, Schauer R, Crocker PR (1996) Glycoconj J 13:913–926
Khan S, Andralojc PJ, Lea PJ, Parry MA (1999). 2′-carboxy-D-arabitinol 1-phosphate protects
ribulose 1,5-bisphosphate carboxylase/oxygenase against proteolytic breakdown. Eur J
Biochem 266(3):840–847. doi:10.1046/j.1432-1327.1999.00913.x. PMID 10583377
Leahy JG, Batchelor PJ, Morcomb SM (2003) Evolution of the soluble diiron monooxygenases.
FEMS Microbiol Rev 27:449–479
Lemieux MJ, Ference C, Cherney MM, Wang M, Garen C, James MNG (2005) The crystal
structure of Rv0793, a hypothetical monooxygenase from M. tuberculosis. J Struct Funct
Genom 6:245–257
Levin EY, Levenberg B, Kaufman S (1960) Enzymatic conversion of
3,4-dihydroxypheylethylamine to norepinephrine. J Biol Chem 235:2080–2086
Long BM, Badger MR, Whitney SM, Price GD (2007) Analysis of carboxysomes from
Synechococcus PCC7942 reveals multiple Rubisco complexes with carboxysomal proteins
CcmM and CcaA. J Biol Chem 282(40):29323–29335
Loughlin WA (2000) Biotransformations in organic synthesis. Bioresour Technol 74:49–62
Mann GD (1996) Chloroplast morphology, movements and inheritance in diatoms. In:
Chaudhary BR, Agrawal SB (Eds) Cytology, genetics and molecular biology of algae. SPB
Academic Publishing, Amsterdam, pp 249–274. Google Scholar
Marcus Y, Gurevitz M (2000) Activation of cyanobacterial RuBP-carboxylase/oxygenase is
facilitated by inorganic phosphate via two independent mechanisms. Eur J Biochem 267
(19):5995–6003. doi:10.1046/j.1432-1327.2000.01674.x. PMID 10998060
Massey V (1994) Activation of molecular oxygen by flavins and flavoproteins. J Biol Chem
269:22459–22462
McKay RML, Gibbs SP (1991) Composition and function of pyrenoids: cytochemical and
immunocytochemical approaches. Can J Bot 69(5):1040–1052
Meyer M, Griffiths H (2013) Origins and diversity of eukaryotic CO2-concentrating mechanisms:
lessons for the future. J Exp Bot 64(3):769–786
Meyer MT et al (2012) Rubisco small-subunit a-helices control pyrenoid formation in
Chlamydomonas. Proc Natl Acad Sci USA 109(47):19474–19479
Monastirioti M, Linn CEJ, White K (1996) Characterization of Drosophila tyramine
beta-hydroxylase gene and isolation of mutant flies lacking octopamine. J Neurosci
16:3900–3911
Montersino S, Tischler D, Gassner GT, van Berkel WJH (2011) Catalytic and structural features of
flavoprotein hydroxylases and epoxidases. Adv Synth Catal 353:2301–2319
Murrell JC, Gilbert B, McDonald IR (2000) Molecular biology and regulation of methane
monooxygenase. Arch Microbiol 173:325–332
Nelson DR, Koymans L, Kamataki T, Stegeman JJ, Feyereisen R, Waxman DJ, Waterman MR,
Gotoh O, Coon MJ, Estabrook RW, Gunsalus IC, Nebert DW (1996) P450 superfamily: update
on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics
6:1–42
Nishimura M, Akazawa T (1974) Research on spinach leaf ribulosebisphosphate carboxylase.
Carboxylase and oxygenase response examined through immunochemical techniques.
Biochemistry 13(11):2277–2281
346 A. Kumar et al.

Omura T, Sato R (1964) Carbon monoxide-binding pigment of liver microsomes. 1. Evidence for
its hemoprotein nature. J Biol Chem 239:2370–2378
Panke S, Witholt B, Schmid A, Wubbolts MG (1998) Towards a biocatalyst for (S)-styrene oxide
production: characterization of the styrene degradation pathway of Pseudomonas sp. strain.
Appl Environ Microbiol 64:2032–2043
Prigge ST, Eipper BA, Mains RE, Amzel LM (2004) Dioxygen binds end-on to mononuclear
copper in a precatalytic enzyme complex. Science 304:864–867
Read BA, Tabita FR (1994) High substrate specificity element ribulosebisphosphate
carboxylase/oxygenase from eukaryotic marine algae and properties of recombinant cyanobac-
terial Rubisco containing “algal” residue adjustments. Arch Biochem Biophys 312:210–218
Rousseaux CS, Gregg WW (2013) Interannual variation in phytoplankton primary production at a
global scale. Remote Sens 6(1):1–19. CrossRefGoogle Scholar
Rutishauser U, Acheson A, Hall AK, Mann DM, Sunshine J (1988) Science 240:53–57
Sage RF, Sage TL, Kocacinar F (2012) Photorespiration and the evolution of C4 photosynthesis.
Annu Rev Plant Biol 63:19–47
Salvucci ME, Osteryoung KW, Crafts-Brandner SJ, Vierling E (2001) Exceptional sensitivity of
Rubisco activase to thermal denaturation in vitro and in vivo. Plant Physiol 127(3):1053–1064.
doi:10.1104/pp.010357
Schlaich NL (2007) Flavin-containing monooxygenases in plants: looking beyond detox. Trends
Plant Sci 12:412–418
Schmitz F (1882) Die Chromatophoren der Algen: Vergleichende Untersuchungen über Bau und
Entwicklung der Chlorophyllkörper und der analogen Farbstoffkörper der Algen. M. Cohen &
Sohn, Bonn
Shen B, Hutchinson CR (1993) Tetracenomycin F1-monooxygenase—oxidation of a naphtha-
cenone to a naphthacenequinone in the biosynthesis of tetracenomycin-C in Streptomyces
glaucescens. Biochemistry 32:6656–6663
Smit H, Gaastra W, Kamerling JP, Vliegenthart JF, de Graaf FK (1984) Infect Immun 46:578–584
Somerville CR, Ogren WL (1979) A phosphoglycolate phosphatase-deficient mutant of
Arabidopsis. Nature 280(5725):833–836
Suzuki Y, Nagao Y, Kato H, Suzuki T, Matsumoto M, Murayama J (1987) Biochim Biophys Acta
903:417–424
Tabita FR (1995) The biochemistry and metabolic regulation of carbon metabolism and CO2
fixation in red micro organism. In: Blankenship RE, Madigan MT, Bauer CE (eds) Anoxygenic
photosynthetic micro organism. Kluwer, Dordrecht, pp 885–914
Takabe T, Akazawa T (1975) Biochemistry 14:46–50
Thierstein HR, Young JR (eds) (2004) Coccolithophores: from molecular processes to global
impact. Springer, Heidelberg
Torres PDE, Winkler M, Glieder A, Fraaije MW (2010) Monooxygenases as biocatalysts:
classification, mechanistic aspects and biotechnological applications. J Biotechnol 146:9–24
Valentine AM, Stahl SS, Lippard SJ (1999) Mechanistic studies of the reaction of reduced
methane monooxygenase hydroxylase with dioxygen and substrates. J Am Chem Soc
121:3876–3887
van Beilen JB, Funhoff EG (2007) Alkane hydroxylases involved in microbial alkane degradation.
Appl Microbiol Biotechnol 74:13–21
van Berkel WJH, Kamerbeek NM, Fraaije MW (2006) Flavoprotein monooxygenases, a diverse
class of oxidative biocatalysts. J Biotechnol 124:670–689
van Hellemond EW, Janssen DB, Fraaije MW (2007) Discovery of a novel styrene monooxy-
genase originating from the metagenome. Appl Environ Microbiol 73:5832–5839
Varki A (1992) Diversity in the sialic acids. Glycobiology 2:25–40
Varki A, Cummings R, Kesko J, Freeze H, Hart G, Marth J (1999) Sialic acids. In: Varki A,
Cummings R, Kesko J et al. (eds) The Essentials of Glycobiology. Cold Spring Harbor, NY:
Cold Spring Harbor Laboratory Press, pp 195–209
13 Monosaccharide Oxygenase 347

Vaucher J-P (1803) Histoire des Conferves D’eau Douce: Contenant Leurs Différents Modes De
Reproduction, Et La Description De Leurs Principales Espèces, Suivie De L’histoire Des
Trémelles Et Des Ulves D’eau Douce. JJ Paschoud, Geneva
Wackett LP, Hershberger CD (2001) Biocatalysis and biodegradation: microbial transformation of
organic compounds, st edn. ASM Press, Washington
Wallar BJ, Lipscomb JD (1996) Dioxygen activation by enzymes containing binuclear non-heme
iron clusters. Chem Rev 96:2625–2657
Whited GM, Gibson DT (1991) Toluene-4-monooxygenase, a three-component enzyme system
that catalyzes the oxidation of toluene to p-cresol in Pseudomonas mendocina KR1. J Bacteriol
173:3010–3016
Xin X, Mains RE, Eipper BA (2004) Monooxygenase X, a member of the copper-dependent
monooxygenase family localized to the endoplasmic reticulum. J Biol Chem 279:48159–48167
Xu F (2005) Applications of oxidoreductases: recent progress. Ind Biotechnol 1:38–50
Yeh W-K, Gibson DT, Liu T-N (1977) Toluene dioxygenase: a multicomponent enzyme system.
Biochem Biophys Res Commun 78:401–410
Zhang N, Kallis RP, Ewy RG, Portis AR (2002) Light modulation of Rubisco in Arabidopsis
requires a capacity for redox regulation of the larger Rubisco activase isoform. Proc Natl Acad
Sci USA 99(5):3330–3334. Bibcode:2002PNAS…99.3330Z. doi:10.1073/pnas.042529999.
PMC 122518. PMID 11854454

You might also like