GET THE APP

An Overview on Fungal Cellulases with an Industrial Perspective
Journal of Nutrition & Food Sciences

Journal of Nutrition & Food Sciences
Open Access

ISSN: 2155-9600

Research Article - (2016) Volume 6, Issue 1

An Overview on Fungal Cellulases with an Industrial Perspective

Sajith S, Priji P, Sreedevi S and Benjamin S*
Enzyme Technology Laboratory, Biotechnology Division, Department of Botany, University of Calicut, Kerala - 673635, India, E-mail: benjamin@uoc.ac.in
*Corresponding Author: Benjamin S, Enzyme Technology Laboratory, Biotechnology Division, Department of Botany, University of Calicut, Kerala - 673635, India, Tel: +91-494-2407406, Fax: +91-494-2400269

Abstract

Lignocellulose is the most abundant biopolymer available on earth. Hydrolysis of lignocellulose into fermentable sugars, sugar acids and phenolics is the pre-requisite for its successful exploitation as substrates for the large scale production of industrially significant value added products. Though remarkable collection of lignocellulolytic microorganisms has been brought to limelight, only a few especially fungi have been studied extensively as they secrete copious lignocellulolytic enzymes extracellularly. Enzymatic hydrolysis of lignocelluloses is carried out by a group of enzymes viz., cellulases, hemicellulases, ligninases in unison or individually, and are collectively known as lignocellulolytic enzymes. In this light of this background, followed by a short introduction on lignocellulose, lignocellulolytic enzymes and mainly focused on fungal cellulase, this review discusses recent approaches on the production of cellulase by submerged and solid-state fermentation strategies; current knowledge on the purification, characterisation of cellulase; and finally concluded with detailed industrial applications of fungal cellulase.

Keywords: Fungal cellulase; Lignocellulose; Production strategies; Purification; Characterisation; Lignocellulolytic enzymes

Introduction

As a consequence of industrialisation and rapid growth in population hike globally, utilisation of natural resources has been increased exponentially during the past few decades. Increasing concern regarding the environmental pollutions and the exhaustion of fossil fuels compelled us to exploit alternative renewable energy resources so as to meet the ever increasing energy requirements [1]. Now-a-days, the concept of waste-to-energy has become the primary focus of many industries with an economic perspective, and sustainable processes signifying the utilization of biomass judiciouslythe most abundant organic renewable resource accounting for around 10-14% of the world’s energy supply. The term biomass is the collective term denoting all the organic materials found on earth including terrestrial and aquatic plants and animals as well as the organic wastes [2]. Generally, the biomass encompasses: plant-based woody biomass (mainly lignocelluloses), plant-based non-woody biomass (starch, sugar and oils), and animal/human based biomass (animal fats and proteins, slurry/slaughter wastes, house hold wastes, etc.,). Among the plant-based woody biomass, lignocellulosic biomass is considered as a potential resource for renewable energy, which is normally used for land filling or simply burned off. Lignocellulose constitutes 60% of the plant cell wall, and is made up of three biopolymers of sugars and their derivatives, viz., lignin, hemicelluloses and cellulose. Annually, about 100 billion tons of plant dry material is generated in the world by photosynthetic activity. The surplus of lignocellulosic biomass is mainly treated as waste (though nothing is literally waste in this universe); hence, intensive research has been accomplished for the effective utilization of the lignocellulosic materials for the production of enzymes, biofuels, antioxidants, feeds, etc.,

Degradation of lignocellulosic biomass is carried out primarily by microbial intervention – i.e., utilize it as carbon and nutrient/energy source for their growth. They include species of bacteria (Clostridium, Cellulomonas, Bacillus, Pseudomonas, Fibribacter, Ruminococcus, Butyrivibrio, etc.,), fungi (Aspergillus, Rhizopus, Trichoderma, Fusarium, Neurospora, Penicillium, etc.,), and actinomycetes (Thermomonospora, Thermoactinomyces, etc.). Of them, fungi are the principal agents involved in the degradation of lignocelluloses. The fungal degradation of lignocellulose is mainly accomplished by producing two types of extracellular enzyme systems: hydrolytic and oxidative catalytic systems. The hydrolytic system produces hydrolases to degrade polysaccharides and the lignolytic system produces ligninases to degrade lignin and opens phenyl rings [3]. The bioconversion of lignocellulosic materials to various value added products mainly includes two processes; hydrolysis of lignocelluloses in to fermentable reducing sugars is carried out by a group of enzymes collectively known as lignocellulolytic enzymes, and subsequent fermentation of these sugars to various value added bio-products. Thus, this review mainly focused on the general aspects of lignocellulose, lignocelluolytic enzymes with special emphasis on fungal cellulases; moreover, characteristics, structure, application and production strategies of cellulase are discussed with appropriate illustrations [4].

Lignocelluloses

Lignocelluloses are the structural polysaccharides of plants that composed of cellulose (~50%), hemicellulose (~30%), and lignin (~20%), and are widely distributed among the vascular plants [5,6]. Cellulose and hemicellulose are polysaccharides composed of simple sugars; whereas, lignin is a complex network of aromatic alcohols. In general, hemicelluloses and lignin provide an amorphous matrix to which crystalline cellulose microfibrils are dispersed (Figures 1 and 2) [7,8]. Cellulose microfibrils are stabilised by intra- and inter-molecular hydrogen bonds, and are surrounded by hemicellulosic polymers. The cellulose-hemicellulose matrices are protected by lignin, an amorphous insoluble polymer which impedes the microbial attack on internal cellulosic structures [9].

nutrition-food-sciences-Structure-lignocelluloses

Figure 1: Structure of lignocelluloses (courtesy: Rubin, [7]; Streffer, [167]).

nutrition-food-sciences-Schematic-representation-lignocelluloses

Figure 2: Schematic representation showing the contents present in the lignocelluloses.

Lignin

Lignin is the most complex polymer made up of phenylpropane units such as coumaryl alcohol, coniferyl alcohol and sinapyl alcohol (Figure 3) linked in a three-dimensional structure, which is difficult to degrade. Lignin component of soft woods contain more than 90% of coniferyl alcohol, and that of hardwood is composed of coniferyl and sinapyl alcohols in varying proportions [10]. They provide rigidity to the plant body, and play a critical role in liquid transport and prevention of microbial attack as well [11].

nutrition-food-sciences-phenylpropane-monomers-lignin

Figure 3: Three phenylpropane monomers in lignin.

Hemicellulose

Hemicellulose-the second most copious part (15-35%) of lignocellulosic biomass - are heterogeneous polymers of sugar acids, pentoses (including arabinose and xylose), and hexoses (glucose, galactose, mannose) [9]. They are highly branched polymers and lack crystalinity. In nature, the composition of hemicellulose is uneven, which depends on the nature of plant source (Figure 4). Generally, in softwoods (e.g., gymnosperm) like spruce and pine, hemicellulosic part is composed mainly of mannan, especially glucomannan and galactoglucomannan; whereas, secondary walls of hardwood (e.g., angiosperm) and herbaceous plants embody chiefly of xylans [12].

nutrition-food-sciences-Structure-hemicelluloses

Figure 4: Structure of hemicelluloses.

Cellulose

About 50% of the CO2 fixed photo-synthetically is stored in the form of cellulose in plants [13]. Cellulose is a high molecular weight (MW) homopolymer constituting (1,4)-D-glucopyranose units joined by β-1,4 linkages with the repeating units of the disaccharide (the cellobiose) (Figure 5). The cellulose chains interact with each other via numerous cross linkages such as hydrogen bonds and van der Waal’s interactions to form bundles which aggregate to form microfibrils with of 5-15 nm diameters [5,6].

nutrition-food-sciences-cellulose-consisting-cellobiose

Figure 5: Structure of cellulose consisting of cellobiose as repeating units.

Each chain of cellulose contributes to the microfibril, as if a small thread contributes in making a rope. Generally, the cellulose chains are arranged parallely in the microfibrils as a highly ordered form, resulting in crystalline structures; while, the less ordered structures contribute to the amorphous regions [14]. The rigidity of cellulose is a direct consequence of its structure. Thus, even though cellulose is a homo-polysaccharide, its structure is heterogeneous; for instance, filter paper is considered as highly crystalline cellulose, therefore, IUPAC has recommended the hydrolysis of filter paper as a standard measurement for total cellulolytic activity [15]. The most prevalent component in lignocelluloses is cellulose.

Lignocellulolytic enzymes

Even though a large chunk of lignocelluloses are formed annually, its bulkiness on earth does not accumulate due to the swift action of microorganisms on it. They efficiently degrade these organic materials so as to provide themselves with carbon and energy source for their growth, and allow the recycling of carbon back into the ecosystem [16]. Although a large number of microbes can thrive on lignocelluloses, a few of them produce the complete battery of enzymes necessary for the breakdown of lignocelluloses into simpler molecules for further aerobic or anaerobic catabolism. In nature, a wide variety of bacteria and fungi are evolved to produce lignocellulolytic enzymes as a part of ecological recycling.

The advent of biotechnology and bioprocess technology has boosted up the effective utilization of lignocelluloses for the production of these valuable lignocellulolytic enzymes. Fungi are mainly exploited for the production of lignocellulolytic enzymes on large scale for use in industry, as they grow attached to the solid substrates with limited moisture content, i.e., natural solid-state fermentation (SSF) [17].

Degradation of lignocelluloses is generally carried out by a complex array of enzymes including ligninases, hemicellulases and cellulases (Figure 6).

nutrition-food-sciences-lignocellulolytic-enzymes

Figure 6: Schematic representation of lignocellulolytic enzymes.

Ligninases/lignases

Ligninases break down lignin into low MW compounds that are assimilated by other microorganisms. Generally, ligninases are of two types: (a) phenol oxidases or laccases and (b) peroxidases (lignin peroxidase and manganese peroxidase). Laccases (EC 1.10.3.2) are copper containing glycoproteins, which may be monomeric, dimeric or tetrameric in nature. The MW of the monomer varies from 50 to 100 kDa. Laccases are involved in the degradation of lignin via oxidation of phenolic compounds to yield phenoxy radicals and quinines [18]. Aspergillus nidulans, Phanerochaete chrysosporium, Lentinula edodes, Phellinus ribis, Pleurotus pulmonarius are the known producers of laccase [19]. Peroxidases are included in the family of oxidoreductases that catalyse the depolymerisation of lignin utilising H2O2. Lignin peroxidase (EC 1.11.1.14) is a heme protein possessing high redox potential with low optimum pH, nearly 3 [19]. Lignin peroxidase is less specific towards its substrates and oxidises a wide range of phenolic, aromatic, non-phenolic and organic substrates. Usually, the MW of Lignin peroxidase isoenzymes ranges from 38 to 46 kDa [20]. In contrast, manganese peroxidases (EC 1.11.1.13) utilise Mn2+ as electron donor and oxidises phenolic structures to phenoxyl radicals; the MW of manganese peroxidise ranges from 38 to 62.5 kDa [21].

Hemicellulase

Hemicellulases are glycoside hydrolases or carbohydrate esterases represented by xylanases (EC 3.2.1.8), β-mannanases (EC 3.2.1.78), arabinofuranosidases (EC 3.2.1.55), and β-xylosidases (EC 3.2.1.37). Xylan constitutes around 70% of hemicelluloses, and is hydrolyzed by xylanases to oligomers, which are further degraded to xylose by β- xylosidases. Moreover, other hemicellulases like mannanase and arabinase are also required for the complete degradation of hemicelluloses, which depends on its chemical composition [1,9].

Cellulases

The third but the most significant group of lignocellulolytic enzyme is cellulases, the key enzymes for the conversion of cellulose into simple sugars [22]. Cellulase is a family of enzymes hyrolysing β-1,4- glycosidic bonds of intact cellulose and other related cellooligosaccharide derivatives. Synergistic action of three principal types of the enzymes, viz., endoglucanase (EC 3.2.1.4), exoglucanase (EC 3.2.1.91) and β-glucosidase (EC 3.2.1.21) is required to accomplish the degradation of intact hydrogen-bond-ordered cellulose. Endoglucanases preferentially hydrolyze the amorphous (internal) regions of the fibrils randomly by cleaving β-glucosidic bonds; cellobiohydrolases are exoglucanases releasing cellobiose, from the termini of the chains, while β-glucosidases complete the degradation process by hydrolyzing cellobiose and other cellodextrins with a low degree of polymerization to glucose units (Figure 7) [23]. Among the lignocellulytic enzymes, cellulases found significant potential for industrial applications, especially in sectors of foods, chemicals, detergents, cosmetics, pulp and paper, etc. [17].

nutrition-food-sciences-synergistic-action-cellulases

Figure 7: Diagramatic representation of the enzymatic hydrolysis of cellulose by the synergistic action of cellulases.

Classification of cellulases

Cellulase is a complex enzyme system comprising of endo-1,4-β-Dglucanase (endoglucanase, EC 3.2.1.4), exo-1,4-β-D-glucanase (exoglucanase, EC 3.2.1.91) and β-D-glucosidase (β-D-glucoside glucanhydrolase, EC 3.2.1.21) [13].

Endoglucanase

Endoglucanase (endo-β-1,4-D-glucanase, endo-β-1,4-D-glucan-4- glucano-hydrolase) - often called as CMCase - hydrolyses carboxymethyl cellulose (CMC) or swollen cellulose in a random fashion. Accordingly, the length of the polymer decreases, resulting in the increase of reducing sugar concentration [24,25]. Endoglucanase also acts on cellodextrins - the intermediate product of cellulose hydrolysis-and converts them to cellobiose (disaccharide) and glucose. These enzymes are inactive against crystalline celluloses such as cotton or avicel.

Exoglucanase

Exoglucanase (exo-β-1,4-D glucanase, cellobiohydrolase) degrades cellulose by splitting-off the cellobiose units from the non-reducing end of the chain. It is also active against swollen, partially degraded amorphous substrates and cellodextrins, but does not hydrolyze soluble derivatives of cellulose like carboxymethyl cellulose and hydroxyethyl cellulose. Some cellulase systems also contain glucohydrolase (exo-1,4-D-glucan-4-glucohydrolase) as a minor component [13].

β- glucosidase

β-glucosidase completes the process of hydrolysis of cellulose by cleaving cellobiose and removing glucose from the non-reducing end (i.e., with a free hydroxyl group at C-4) of oligosaccharides. The enzyme also hydrolyzes alkyl and aryl β-glucosides [26].

Fungal cellulases

The microbial hydrolysis of insoluble cellulose requires the action of multiple cellulases (endoglucanases, exoglucanases and β-glucosidases) in a synergistic manner, so that the complex polymer is converted to simple sugars. Among the microorganisms, fungi in particular are dynamic cellulose decomposers, and possibly responsible for 80% of the cellulose breakdown on earth [27]. This is particularly true in forest ecosystem, where fungi are the principal agents decomposing cellulose and lignin [28]. The cellulose decomposing fungi include members of the ascomycota, basidiomycota, and deuteromycota, as well as some chytrids that occur in the rumen of some animals. Efficient cellulolytic fungi are represented by the species of Aspergillus, Penicillium, Chaetomium, Trichoderma, Fusarium, Stachybotrys, Cladosporium, Alternaria, Acremonium, Ceratocystis, Myrothecium, Humicola, etc. [29,30].

Moreover, the aerobic fungal cellulases are usually preferred by the industry, because they are extracellular, adaptive in nature and usually secreted in large quantities during growth. This is in sharp contrast to many bacterial as well as anaerobic fungal cellulases which exist as tight multi-enzyme complexes; often membrane bound as cellosomes, from which it is difficult to recover individual active enzyme species; hence, economically less important [31]. Production of cellulolytic enzyme from aerobic fungi is wide spread; among them, species of Aspergillus, Trichoderma, Penicillium and Sclerotium are found as highly cellulolytic, and are mainly considered for commercial exploitation [32,33].

Structurally, fungal cellulases are simple and modular enzyme with functionally distinct modules or domains [34]. Some of them possess two domains, catalytic domain and carbohydrate binding domains connected by a serine- and threonine-rich polylinker with varying chain length and structure (Figure 8) [35]. The carbohydrate binding modules vary in size ranging from 4 to 20 kDa, and are rich in aromatic and often polar amino acid residues that immobilize the substrate during catalysis. The active site of the catalytic domain may be topologically tunnel, cleft or pocket in shape allowing the hydrolysis of the substrate efficiently [36].

nutrition-food-sciences-carbohydrate-binding-domain

Figure 8: General structure of cellulase consisting of catalytic domain (CD) and carbohydrate binding domain (CBD) joined via a linker peptide.

One of the most extensively studied aerobic fungi is T. reesei , which is capable of hydrolyzing native cellulose [37,38]. T. reesei possesses two genes encoding for exoglucanase, eight for endoglucanases and seven for glucosidases [39,40]. In the last few decades, thermophilic fungi have also been studied widely because of the fact that cellulose fibers bulge/swell up at higher temperatures, so that they become easily accessible for hydrolytic enzymes [41]. Talaromyces emersonii is a typical thermophilic fungus capable of producing cellulase, which was active even at 70°C and decomposes the intact cellulose [42]. Two strains of Penicillium were identified from subtropical soils with potentials for the production of cellulase [43]; Chaetomium thermophilum, Sporotrichum thermophile, Talaromyces emersonii and Thermoascus aurantiacus grew well and decomposed cellulose very rapidly, producing thermostable cellulases [41,44].

Cellulase production by fermentation

Submerged fermentation (SmF): The SmF - the most commonly used technology for the large scale production of enzymes - is generally carried out in the presence of free flowing liquid, in which soluble substrates are dispersed; however, compared to liquid medium, filamentous fungi like Aspergillus, Penicillium and Trichoderma normally produce large quantity of cellulase in solid medium. In fact, the easiness in controlling the process parameters, monitoring and downstream process make SmF more attractive [40]. In SmF, various factors such as pH, temperature, substrate concentration, inducer, medium composition, etc., influence the production of cellulase significantly. The main drawback in SmF is the requirement of long fermentation time (i.e., gestation period) with less production [45]. Among microorganisms, bacteria are most commonly used for the production of enzymes by SmF [46,47]; but, some species of fungi such as Aspergillus, Penicillium, Trichoderma, etc. are also being cultivated under SmF for the production of cellulase. Table 1 shows major cellulase producing species/strains of fungi by utilizing different carbon sources.

Fungi Substrate Cellulase (U/mL) Reference
A. heteromorphus wheat straw 83 Singh et al.[67]
A. flavus BS1 CMC 2793 Sajith et al. [61]
A. fumigatus  wheat straw 321  Saqib et al. [136]
A. niger CMC 1.6 Narasimha et al. [50]
A. niger maize straw 102 Milala et al. [33]
A. niger banana peel 12.4 Jadhav et al. [137]
A. niger coir waste 3.3  Mrudula and Murugammal, [138]
A. niger sawdust 3.9 Devi and Kumar [139]
A. niger MS82 grass 6.8 Sohail et al. [60]
P. fellutanum lactose 81 Kathiresan and Manivannan, [140]
R. oryzae water hyacinth 450 Karmakar and Ray [141]
T. harzianum CMC 150 Rubeena et al. [142]
T. harzianum MTCC 8230 rice straw 1.7 Kocher et al. [143]
T. reesei RUTC30 sugarcane baggase 121  da Silva Delabona et al., [144]
T. reesei ZU-02 corn cob residue 5.5 Liming and Xueliang, [145]
T. viride CMC 173 Neethu et al., [146]

Table 1: Production of cellulase by various fungi employing submerged fermentation.

The cellulolytic activities of fungi may vary depending upon the medium as well as the culture conditions. Hence, the formulation of suitable fermentation strategies is the key factor for deciding the efficiency of a fungus in terms of the production of cellulase. In order to obtain better productivity, various synthetic or natural carbon sources are used in SmF; for instance, Acharya et al. [48] studied the production of cellulase by A. niger on pretreated (by alkali) sawdust and the maximum cellulase activity was observed under optimized condition, i.e., pH between 4 and 4.5, 120 rpm, at 28°C and peptone as nitrogen source. Similarly, Karthikeyan et al. [49] investigated the production of cellulase by Penicillium strain K-P in liquid medium by supplementing various carbon and nitrogen sources at varying pH and temperature; the fungus showed the maximum cellulase activity in the presence of fructose, ammonium nitrate, pH 3.0 and 30°C on day 5. Production of cellulases was studied by A. niger in the presence of various carbon and nitrogen sources at varying pH; and found that the maximum production of cellulase was on Czapek-Dox medium supplemented with 1% CMC or sawdust at pH 5 [50]. From the aforesaid reports, it is clear that the production of cellulase mainly observed under acidic pH and 20-30°C; in addition to the ability of the fungi in utilizing the carbon and nitrogen sources present in the liquid medium.

Solid-state fermentation

The SSF is one of the important strategies employed in the industries for the enhanced production of various enzymes. SSF is carried out on the solid substrates in the presence of no free water (i.e., water is available in the bound form only), which acts as both solid support and source of nutrients. In the recent years, SSF is gaining more interest as a suitable strategy for the recycling of nutrient-rich wastes such as lignocelluloses. SSF facilitates not only the possibilities for the bioconversion of agro-residues to value-added products, but also it enables the efficient recycling of lignocellulosic materials with the expenditure of less energy [51]. Earlier days, SSF was considered as suitable only for the fermentation of food or food-associated products; but further studies showed several benefits of this technology; such as high enzyme yield at low cost, use of agricultural waste as substrate, and wider range of additional enzyme activities than found in SmF [52,53]. Thus, SSF is an attractive means to produce cellulase economically, because of its lower capital investment and operating cost [54]. Due to the discrete nature of SSF, the physico-chemical characteristics of substrate such as crystallinity, bed porosity and enormous surface area can influence the production of cellulolytic enzyme system in fungal cultures. In SSF, the operating conditions like temperature, pH and moisture content are vital factors influencing the microbial growth and production of cellulase. Availability of oxygen in the open space between substrate particles (i.e., porosity) and generation of heat are the major challenges in SSF, which have to be addressed properly [55,56].

Agricultural wastes such as brans of wheat and rice, corn stover, straws of wheat and rice, sugarcane baggase, sawdust, etc. are the most commonly used substrates for the production of cellulase. For instance, Liu et al. [57] investigated the production of cellulase on different lignocellulosic substrates such as straws of rice, wheat and cotton, corn stover and corncob using A. fumigatus Z5; of them, the corn comb supported the maximum production of endoglucanase. P. echinulatum 9A02S1 showed the maximum production of cellulolytic enzymes on the medium containing a mixture of pretreated sugarcane bagasse and wheat bran [58]. Dutta et al. [59] studied the production of cellulases from P. citrinum using brans of wheat and rice and rice straw as substrate; all these substrates supported the production of cellulases. A. niger MS82 efficiently utilized the lignocellulosic substrates like grass, bagasse and corncob with variable cellulase activities [60]. A. flavus BS1 competently utilized different lignocellulosic substrates and supported higher levels of cellulase activity [61]. A. flavus Linn NSPR 101 showed the production of cellulase on various natural substrates like bagasse, corncob and sawdust [62]. Thus, production of cellulase may vary between species of fungi; and from the above studies, it is evident that the wide range and higher enzyme activities are possible on waste lignocellulosic biomass as substrate. It is difficult to compare cellulolytic enzymes reported in literature; because different authors demonstrated the activity of these enzymes in different units. Nevertheless, Table 2 demonstrates the production of cellulase on various solid substrates by different species of fungi, and the activities are expressed in gram dry fermented substrates (gds).

Fungi Substrate Cellulase(U/gds) Reference
A. flavus BS1 tapioca flour and sawdust 5408.5 Sajith et al., [14]
A. fumigatus  rice straw and wheat bran 14.7 Sherief et al., [147]
A. niger wheat bran 3.2 Chandra et al., [148]
A. niger 38 wheat bran and wheat straw 14.8 Jecu, [149]
A. niger KK2 rice straw 130 Kang et al., [150]
A. niger MS82 grass 100 Sohail et al., [60]
A. terreus M11 corn stover 581 Gao et al., [151]
A. fumigatus Z5 corn stover 526.3 Liu et al., [57]
Aspergillus spp. MAM-F35 wheat straw 487 AboState et al., [152]
Fomitopsis sp. RCK2010 wheat bran 84 Deswal et al., [153]
niger USM AI sugarcane bagasse and palm kernel cake 3.2 Lee et al., [84]
P. citrinum rice bran 2 Dutta et al., [59]
P. decumbens  wheat straw 52.8 Mo et al., [154]
P. echinulatum 9A02S1  sugarcane bagasse and wheat bran 282.4 Camassola and Dillon, [58]
T. reesei sugarcane bagasse and palm kernel cake 2.2 Lee et al., [84]
T. reesei LW1 corn straw and wheat bran 452.5 Wang et al., [155]
T. reesei NRRL11460 sugarcane bagasse 154.6 Singhania et al., [38]
T. reesei LM-UC4 bagasse 38.6 Duenas et al., [156]
A. phoenicis QM 329
T. viride wheat straw 555 AboState et al., [152]
Thermoascus aurantiacus wheat straw 1572 Kalogeris et al., [157]

Table 2: Production of cellulase by fungi on various lignocellulosic substrates employing solid-state fermentation.

Statistical optimization

The major challenge in the development of economically feasible bioprocess for the production of industrially significant enzymes is to identify the potential microorganisms, composition of media and the optimization of various process parameters that influence the microbial growth and production of enzyme [63]. Statistical tool such as response surface methodology (RSM) coupled with artificial neural networks (ANN) is generally employed to optimize the parameters influencing a particular biological response, that critically evaluates the interactive effects normally neglected in conventional one-at-a-time cultivation strategy. Moreover, it significantly reduces the number of experiments required for the standardization of a biotechnological process with remarkable reproducibility [64]. Since microbial cellulases are inducible in nature and their production is influenced by several parameters such as pH, temperature, agitation, substrate concentration, type of nitrogen and carbon source etc. RSM is effectively employed to standardize these process parameters. The statistical tools such as Plackett-Burman, Box-Behnken, central composite designs, ANN alone or in their combination are the most commonly used methods for the screening of independent variables that significantly affect the enzyme production. For instance, Singhania et al. [65] employed a combination of Plackett-Burman and central composite designs to maximize the production of cellulase produced by T. reesei RUT C30, which resulted in 6.2 folds increase in cellulase production in the presence of 37.5% moisture content and 30°C. Mixed cultures of T. reesei and A. phoenicis showed β-glucosidase activity and filter paper activity of 0.64 IU/mL and 1.54 FPU/mL, respectively on dairy manure at 27°C and pH 5, which was optimized using Box- Behnken design [66]. Similarly, Singh et al. [67] utilized Box-Behnken design for effectively optimizing the temperature, pH and substrate concentration as 60°C, pH 4.8 and 148.9 mg/mL, respectively [68]. Thus, the statistical methods improve the effectiveness of fermentation processes prior to industrialization, thereby promoting the utilities of various microorganisms as potential sources of bioproducts.

Purification and characterization of cellulase

Purification and characterization are the vital steps required for developing the improved performance/functioning of an enzyme. Enzymes in the culture supernatant could be purified by the classical methods including precipitation, dialysis and column purification [69]. Different types of columns are used for the purification of cellulase, among which sephadex with different sieve sizes is the most popular matrix used for gel exclusion chromatography [70]. The efficiency of purification is generally analyzed in terms of purification folds and yield. Table 3 represents the columns used, purification fold and yield of cellulase produced by different species of fungi.

Fungi Column Yeild (%) Purification fold Reference
A. aculeatus DEAE-Sephadex 25 4 Naika et al., [90]
A. glaucus XC9 Sephadex G-100 22.3 21.5 Tao et al., [70]
A. kawachii G3000-SW 7.7 494 Iwashita et al., [158]
A. niger Sephacryl S-300 22 23 Yan et al., [159]
A. niger 322 Sephadex G-75 33 6 Peshin and Mathur, [160]
A. niger IF031125 TSK-gel G2000SW 21.8 46.1 Akiba et al., [71]
A. oryzae TSK DEAE-5PW 4.5 176.9 Riou et al., [74]
A. terreus AN1 DEAE-sepharose 1.3 40 Nazir et al., [78]
A. terreus DSM Sepharose-4B column F11 16.6 15.4 Elshafei et al., [161]
A. terreus M11 Sephadex G-100 14 18 Gao et al., [72]
Alternaria alternata Sephadex 13 4.2 Macris, [162]
Fusarium oxysporum Column PBE94 65.4 17.6 Christakopoulos et al., [163]
Mucor circinelloides Bio-Gel A-0.5 m 3 408 Saha, [164]
P. purpurogenum KJS506 Hydroxyapatite column 22.3 34 Lee et al., [84]
T. harzianum Sephadex G-50 10.3 21.9 Ahmed et al., [165]
T. viride Sephadex-G100 2.1 2.3 Nasir [166]

Table 3: Purification fold and yield of fungal cellulase based on various column packing materials.

Characterization of cellulase for analyzing its optimum temperature and pH is required to reveal its possible industrial applications. Most of the fungal cellulases are active at a temperature range from 40 to 70°C and are acidophilic in nature. For instance, the cellulase produced by A. niger IFO31125 showed an optimum temperature of around 70°C at pH 6 with stability for 2 h [71]. The pH and the temperature optima of the cellulase produced by A. terreus M11 was found as 2 and 60°C, respectively; and 60% of the initial activity was maintained for 1 h at 70°C [72]. Two endoglucanases, RCE1 and RCE2 produced by Rhizopus oryzae showed 55°C as the optimum temperature for both the enzymes with different pH optima, i.e., 5 and 6, respectively [73]. A. oryzae secretes a highly glucose tolerant cellulase with the optimum activity at 50°C and pH 5 [74], while the cellulase produced by A. oryzae S/92Gbr showed the optimum activity at pH 5 and 45°C [75].

Influence of metal ions, detergents and surfactants

Various metal ions and chemical compounds may influence the cellulase activity. It was reported that usually metal ions such as Hg2+, Cu2+, Zn2+, Mg2+, Fe3+, Mn2+, Ag+, Mn2+, K+ are slightly or completely inhibitory to cellulase, whereas metal ions such as Ca2+, Na+ and Co2+ stimulate or unaffect the activity of cellulase [59,76-78]. Various chemicals or reagents such ethylenediaminetetraacetic acid (EDTA), sodium dodecyl sulphate (SDS), dicyclohexyl carbodiimide, sodium azide, β-mercaptoethanol, dithiothreitol, triton X-100, urea, etc. may increase, decrease or abolish the activity, based on the nature of enzymes [79]. Riou et al. [74] investigated the effect of different metal ions and detergents on cellulase produced by A. oryzae, and found that Ag+, Hg2+, Fe3+, SDS, diethylpyrocarbonate, castanospermine, dithiothreitol were slightly or completely inhibited the cellulase activity; whereas Mn2+ enhanced the activity. Similarly, the activity of cellulase produced by A. terreus M11 was increased in the presence of Mn2+; but the presence of Hg2+, Cu2+, Pb2+ and detergents slightly decreased or completely abolished the activity of cellulase [72]. Yang et al. [80] demonstrated that the cellulase produced by Paecilomyces thermophila was strongly inhibited by Hg2+; while the activity was highly enhanced in the presence Zn2+.

Enzyme kinetics

The maximum velocity (Vmax) and Michaelis-Menton constant (Km) are the two constants describing the kinetic characteristics of the enzyme action. The Vmax describes the specific point in an enzymatic reaction at which the rate of the reaction is catalyzed to the maximum, if other factors are optimum; the Vmax is attained only when the substrate concentration is sufficiently available to fill the enzyme’s active site. Km is the substrate concentration required to fill half of the enzyme’s active site. High Km describes that the enzyme has less affinity towards substrate; whereas low Km indicates high affinity towards substrate, thereby higher activity. Liu et al. [81] demonstrated the Km and Vmax of two cellulases produced by A. fumigatus Z5 as 37.8 mg/mL and 437.3 μmol/min/mg; 51.8 mg/mL and 652.7 μmol/min/mg, respectively. The Km and Vmax of cellulase produced by A. niger BCRC31494 were found to be 134 mg/mL and 4.6 U/min/mg [82]; while Km and Vmax of P. pinophilum were: 4.8 mg/mL and 72.5 U/mg, respectively [83]. Lee et al. [84] confirmed the Km and Vmax of P. purpurogenum KJS506 as 1.15 mg/mL and 220 U/mg, respectively.

Molecular weight of fungal cellulases

The MW of cellulase produced by different fungal species may vary from 12 kDa to 126 kDa [85,86]. SDS-polyacrylamide gel electrophoresis (SDS-PAGE) is the most commonly used method for judging the apparent MW of enzymes [87-89]. Fungal cellulase may be of monomeric [90] or dimeric [91] in nature. Cellulase produced by T. viride was purified to homogeneity using DEAE-sepharose column and the MW was estimated as 87 kDa by SDS-PAGE [92]. P. pinophilum MS 20 produced a monomeric cellulase with MW of 42 kDa, which appeared as a single band on SDS-PAGE gel [83]. It was reported that A. awamori VTCC-F099 and Fomitopsis pinicola produced monomeric thermo active cellulase with a MW of 32 kDa [93,94]. The cellulase produced by A. niger revealed a MW of 60 kDa on SDS-PAGE gel [95]. In all the aforesaid studies, the purified celluase appeared as single band upon SDS-PAGE, indicating that the cellulase produced by these fungi are active in solution as monomers or homodimers, consequently migrates through the SDS-PAGE gel according to their MW so as to segregate as single band. In contrast, some studies reported the identification of hetero-dimeric cellulases or isoforms which appeared as separate bands upon SDS-PAGE. For instance, A. niger Z10 produced two cellulase bands on SDS-PAGE gel with MWs 50 and 83 kDa [5,6]. Similarly, another strain of A. niger also reported as producing dimeric cellulase with MWs of 23 and 36 kDa; whereas A. fumigatus produced dimeric cellulase with MWs of 21 and 32 kDa [96]. Kaur et al. [97] reported 40 and 50 kDa isoforms of cellulase produced by Melanocarpus sp. MTCC 3922 with MW as judged by SDS-PAGE.

Applications of cellulases

Cellulases occupy the third most significant industrial enzymes on the global market (i.e, ≈15%) after amylase (≈25%) and protease (≈18%). Enzymatic hydrolysis of cellulosic biomass offers an attractive alternative for the generation of sugars, which can serve as the raw materials for the production of various value added products of commercial interest such as bioethanol [98]; organic acids [99,100]; free sugars, antibiotics and animal feeds [101]. Enzymatic hydrolysis of cellulose is favorably superior to acid and alkali hydrolyses - because enzymes are recoverable, specific, low in energy requirements, and nonpolluting. The production cost and the low yield of cellulase are the major constraints in the economics of the process, which hinder its application in industries. Therefore, the discovery of novel microbial species or mutant strains secreting higher levels of cellulases is still an emerging area of research to develop economically competitive bioprocess strategies applicable on large scale [102]. Cellulase has wide applications in various industries including detergents, textiles, in the production of paper and pulp, bioethanol and organics; some of them are discussed below.

Paper and pulp industries

Cellulases are mainly used for the pulping and deinking of waste papers. Bio-mechanical methods are widely used now-a-days to obtain the suspension of fibres from wood, i.e., the pulp. Application of cellulase for pulping enhances the energy efficiency of the process, and also improves the physical properties such as inter-fibre bonding and mechanical strength of the final paper product [103]. Moreover, it provides environment-friendly processes, limiting the use of harmful chemicals. For instance, cellulase was effectively used for refining the bleached Eucalyptus globules kraft pulp, which enhanced the drainability of the pulp by about 80% without any change in the energy consumption [104]. Similarly, cellulase was used for the modification of cellulose fibres of kraft/sulphate pulp, resulting in improved physical properties of the sheets [105]. Cellulase was also used for deinking of waste papers. During deinking, the ink attached to the surface of recycled cellulose fibres was released by the enzymatic hydrolysis of carbohydrates, leading to the peeling of individual fibres or bundles [106]. The waste papers from various fields offer as important raw material for the pulp and paper industries, as the recycling of used papers reduces the solid wastes and also lessens the burden of deforestation for wood fibres [107,108]. Enzymatic deinking of waste papers, especially employing the mixtures of cellulase and hemicellulase, enhance the quality and brightness of the recycled paper [109-111]. Cellulases are also used to improve the drainage of several paper mills by dissolving clogged fibre residues [112]. Moreover, the cellulase preparations are also used to make easily biodegradable cardboards, tissue papers and sanitary papers [113-115].

Textile industry

Cellulases are widely used to improve the softness and appearance of cellulose-based textiles. In textile industry, cellulases are mainly used for bio-polishing of cotton cloths and biostoning of denim jeans to impart stonewashed look for denims. During the biostoning process, cellulases hydrolyse the small fibre protrusions from the surface and release the indigo dye attached to it, resulting in the dull look of the jeans. It replaces the conventional pumice stones used for the purpose, thereby reducing the fibre damage and human labour [116]. Cellulase produced by Humicola insolens and Trichoderma is generally used for biostoning of jeans [40,117]. During the process of biopolishing, cellulase hydrolyzes the small protrusions of the fibres from the surface of cotton clothes, thus removes the fussiness of the surface so as to create a smooth and glossy appearance [107,118]. Cellulases are also used to improve the dye absorbance of the fibres and to remove excess dye, giving a colour gradient to the fabrics [112]. Cellulases found potential applications as additive in household laundry detergents for improving fabric softness and brightness. Mild alkaline and thermotolerent cellulases produced by fungi such as T. viride, T. hurzianum, T. reesei, A. niger and Humicola insolens are generally used for the purpose [119,120].

Food and feeds

Cellulases found potential applications in food and feed processing industries as well. They are the integral part of the macerating enzyme complex (cellulase, xylanase and pectinase), that are used for the extraction and clarification of fruits and vegetable juices, nectars, oils and purees [121,123]. Cellulase-assisted extraction of flavanoids from flowers and seeds enhanced the yield and reduced the extraction time and heat damage, as compared to the conventional acid/alkali/organic solvent/heat extraction methods [101]. Cellulase in combination with other cell wall degrading enzymes can be used to increase the taste and aroma of citrus fruits by reducing the bitterness [112]. As cellulases favour the enhanced release of simple sugars, they found potential applications in alcoholic beverages including beer and wine. It was found that application of cell wall degrading enzymes during malting and fermentation improved liquor yield, aroma and stability [124]. Besides, cellulases are also used in fermented foods and feeds for improving the nutritional quality and digestibility.

Bioethanol

The massive exploitation and utilization of fossil fuels have insistently reduced its natural reserves and caused severe environmental pollution via the release of green house and toxic gases. Hence, the world economy is now focused on biofuels, especially bioethanol from renewable resources, which is expected to replace 20% of the fossil fuel consumption by 2020 [125,126]. The most actively investigated application of cellulase is the production of biofuels, especially bioethanol. Cellulases actively convert the cellulosic renewable resources in to glucose and other simple fermentable sugars that can be used as substrates for the production of bioethanol. Production of bioethanol from lignocelluloses is a multistep process. Initially, the lignocellulosic biomass is subjected to pre-treatment - either mechanically, chemically or enzymatically to remove lignin and hemicelluloses fractions, followed by the treatment with cellulase to release fermentable sugars (pentoses and hexoses). Then the hydrolyzed cellulosic residue is used for the microbial fermentation to produce bioethanol [127]. Agriculture residues such as sugarcane bagasse, straw of wheat, rice and corn; wheat bran, corn stover, etc. were successfully used as raw materials for the production of bioethanol, employing cellulases produced by various filamentous fungi including Aspergillus, Trichoderma, and Penicillium [41,128-130].

Other applications

Cellulases are also used to generate plant protoplast for genetic manipulation [131], to control industrial slime [132] and to produce cellulase-based chitosan with antibacterial, immunomodulatory and antitumour activities [133,134]. Han and He [135] reported that the commercially available cellulase from T. reesei successfully decomposed the straw thereby increasing the soil fertility and plant growth.

Conclusion

Overwhelming demand for natural products has elevated the significance of industrial enzymes; among which, cellulases occupy a pivotal position. The major bottleneck in the commercialization of cellulase is the lacuna in the economically feasible process and to improve the functioning/catalysis of cellulase in tune with the demand. However, the utilization of lignocellulosic wastes has proven as a main contender to overcome the problem to a great extent. Still, the exploration of sustainable substrates, microorganisms and fermentation strategies are to be evolved so as to achieve higher productivity, quality and economic feasibility. Moreover, further studies should be accelerated for the expansion of cellulase research by manipulating the ability of microorganism via gene/protein engineering for the effective utilization of biomass, facilitating better methods for bioconversion as well as solid waste management. In fact, the upper hand of SSF in alleviating the environmental burden due to the heaping up of lignocellulosic biomass has to be exploited with an economic and industrial perspective.

Acknowledgement

The authors gratefully acknowledge the financial support from the Kerala State Council for Science Technology and Environment (Grant No (T) 422/SRS/2009/CSTE). The authors also declare that there exists no conflict of interest.

References

  1. Dashtban M, Schraft H, Qin W (2009) Fungal bioconversion of lignocellulosic residues; opportunities & perspectives. IJBS, 5: 578.
  2. McKendry P (2002) Energy production from biomass (Part 1): Overview of biomass. Bioresour Technol 83: 37-46.
  3. Sánchez C (2009) Lignocellulosic residues: biodegradation and bioconversion by fungi. Biotechnol Adv 27: 185-194.
  4. Sajith S (2015) Investigations on the lignocellulolytic activities of certain fungi with special reference to cellulase production. PhD Thesis, University of Calicut.
  5. Coral G, Arikan B, Ünaldi MN, Guvenmez H (2002a) Some properties of crude carboxymethyl cellulase of Aspergillus niger Z10 wild-type strain. TJB 26: 209-213.
  6. Coral G, Arikan B, Ünlandi MN, Guvenmez H (2002b) Some properties of crude carboxymethyl cellulase of Aspergillus niger Z10 wild-type strain. TJB 26: 209-213.
  7. Krogh KB, Mørkeberg A, Jørgensen H, Frisvad JC, Olsson L (2004) Screening genus Penicillium for producers of cellulolytic and xylanolytic enzymes. Paper presented at the Proceedings of the Twenty-Fifth Symposium on Biotechnology for Fuels and Chemicals Held in Breckenridge, CO.
  8. Howard R, Abotsi E, Van Rensburg EJ, Howard S (2004) Lignocellulose biotechnology: issues of bioconversion and enzyme production. AJB 2: 602-619.
  9. Harmsen P, Huijgen W, Bermudez L, Bakker R (2010) Literature review of physical and chemical pretreatment processes for lignocellulosic biomass.
  10. McCrady E (1991) The nature of lignin. Alkaline Paper Advocate 4: 33-34.
  11. Gírio FM, Fonseca C, Carvalheiro F, Duarte LC, Marques S, et al. (2010) Hemicelluloses for fuel ethanol: A review. Bioresour Technol 101: 4775-4800.
  12. Joshi V, Pandey A (1999) Biotechnology: Food Fermentation: Microbiology, Biochemistry, and Technology. Educational Publishers & Distributors.
  13. Gardner K, Blackwell J (1974) The structure of native cellulose. Biopolymers, 13: 1975-2001.
  14. Jun H, Bing Y, Keying Z, Xuemei D, Daiwen C (2009) Strain improvement of Trichoderma reesei Rut C-30 for increased cellulase production. Indian J Microbiol 49: 188-195.
  15. Sette LD, de Oliveira VM, Rodrigues MFA (2008) Microbial lignocellulolytic enzymes: industrial applications and future perspectives. Microbiology Australia 29: 18-20.
  16. Kunamneni A, Ballesteros A, Plou FJ, Alcalde M (2007) Fungal laccase-a versatile enzyme for biotechnological applications. Communicating current research and educational topics and trends in applied microbiology 1: 233-245.
  17. Arora DS, Sharma RK (2010) Ligninolytic fungal laccases and their biotechnological applications. Applied biochemistry and biotechnology 160: 1760-1788.
  18. Wong DW (2009) Structure and action mechanism of ligninolytic enzymes. Appl Biochem Biotechnol 157: 174-209.
  19. Hofrichter M (2002) Review: lignin conversion by manganese peroxidase (MnP). Enzyme and Microbial Technology, 30: 454-466.
  20. Chinedu SN, Okochi V, Smith H, Omidiji O (2005) Isolation of cellulolytic microfungi involved in wood-waste decomposition: Prospects for enzymatic hydrolysis of cellulosic wastes. International Journal of Biomedical and HealthSciences.
  21. Szijártó N, Szengyel Z, Lidén G, Réczey K (2004) Dynamics of cellulase production by glucose grown cultures of Trichoderma reesei Rut-C30 as a response to addition of cellulose. Paper presented at the Proceedings of the Twenty-Fifth Symposium on Biotechnology for Fuels and Chemicals Held in Breckenridge, CO.
  22. Robson LM, Chambliss GH (1989) Cellulases of bacterial origin. EMT 11: 626-644.
  23. Begum F, Absar N, Alam MS (2009) Purification and characterization of extracellular cellulase from A. oryzae ITCC-4857.01. JASR 5: 1645-1651.
  24. Kubicek CP, Messner R, Gruber F, Mach RL, Kubicek-Pranz EM (1993) The Trichoderma cellulase regulatory puzzle: from the interior life of a secretory fungus. Enzyme Microb Technol 15: 90-99.
  25. Moore Landecker E (1996) Fundamenttals of the Fungi. (4thedn) New Jersey: Prentice Hall, inc.
  26. Alexopoulos C, Mims C, Blackwell M (1996) Introductory mycology: New York: John Wiley pp. 869.
  27. Wood TM (1985) Properties of cellulolytic enzyme systems. Biochem Soc Trans 13: 407-410.
  28. Mehrotra R, Aneja K (1990) An introduction to mycology: New Age International.
  29. Mathew GM, Sukumaran RK, Singhania RR, Pandey A (2008) Progress in research on fungal cellulases for lignocellulose degradation. JSIR 67: 898.
  30. Pointing SB (1999) Qualitative methods for the determination of lignocellulolytic enzyme production by tropical fungi.
  31. Milala M, Shugaba A, Gidado A, Ene A, Wafar J (2005) Studies on the use of agricultural wastes for cellulase enzyme production by Aspergillus niger. RJABS 1: 325-328.
  32. Bayer EA, Chanzy H, Lamed R, Shoham Y (1998) Cellulose, cellulases and cellulosomes. Curr Opin Struct Biol 8: 548-557.
  33. Payne CM, Knott BC, Mayes HB, Hansson H, Himmel ME, et al. (2015) Fungal cellulases. Chem Rev 115: 1308-1448.
  34. Hildén L, Johansson G (2004) Recent developments on cellulases and carbohydrate-binding modules with cellulose affinity. Biotechnol Lett 26: 1683-1693.
  35. Reczey K, Szengyel Z, Eklund R, Zacchi G (1996) Cellulase production by Trichoderma reesei. Bio resource Technology, 57: 25-30.
  36. Singhania RR, Sukumaran RK, Pillai A, Prema P, Szakacs G, et al. (2006) Solid-state fermentation of lignocellulosic substrates for cellulase production by Trichoderma reesei NRRL 11460. Indian journal of Biotechnology 5: 332-336.
  37. Aro N, Pakula T, Penttilä M (2005) Transcriptional regulation of plant cell wall degradation by filamentous fungi. FEMS Microbiol Rev 29: 719-739.
  38. Sukumaran RK, Singhania RR, Pandey A (2005) Microbial cellulases-production, applications and challenges. JSIR 64: 832.
  39. Li DC, Li AN, Papageorgiou AC (2011) Cellulases from thermophilic fungi: recent insights and biotechnological potential. Enzyme Res 2011: 308730.
  40. Murray P, Aro N, Collins C, Grassick A, Penttilä M, et al. (2004) Expression in Trichoderma reesei and characterisation of a thermostable family 3 β-glucosidase from the moderately thermophilic fungus Talaromyces emersonii. PEP 38: 248-257.
  41. Picart P, Diaz P, Pastor FI (2007) Cellulases from two Penicillium sp. strains isolated from subtropical forest soil: production and characterization. Lett Appl Microbiol 45: 108-113.
  42. Mandels M (1975) Microbial sources of cellulase. Biotechnol Bioeng Symp : 81-105.
  43. Singhania RR, Sukumaran RK, Patel AK, Larroche C, Pandey A (2010) Advancement and comparative profiles in the production technologies using solid-state and submerged fermentation for microbial cellulases. EMT 46: 541-549.
  44. Smitha R, Jisha V, Sajith S, Benjamin S (2013) Dual production of amylase and d-endotoxin by Bacillus thuringiensis subsp. kurstaki during biphasic fermentation. Microbiology 82: 794-800.
  45. Sreedevi S, Sajith S, Benjamin S (2013) Cellulase producing bacteria from the wood-yards on Kallai river bank. Advances in Microbiology 3: 326.
  46. Acharya P, Acharya D, Modi H (2008) Optimization for cellulase production by Aspergillus niger using saw dust as substrate. AJB.
  47. Karthikeyan N, Sakthivel M, Palani P (2010) Screening, Identifying of Penicillium KP Strain and Its Cellulase Producing Conditions. JE 2: 4-7.
  48. Narasimha G, Sridevi A, Buddolla V, Subhosh CM, Rajasekhar RB (2006) Nutrient effects on production of cellulolytic enzymes by Aspergillus niger.
  49. Robinson T, Singh D, Nigam P (2001) Solid-state fermentation: a promising microbial technology for secondary metabolite production. AMB 55: 284-289.
  50. Couto SR, Sanromán MA (2006) Application of solid-state fermentation to food industry-a review. JFE 76: 291-302.
  51. Xia L, Cen P (1999) Cellulase production by solid state fermentation on lignocellulosic waste from the xylose industry. Process Biochemistry 34: 909-912.
  52. Thibault J, Pouliot K, Agosin E, Pérez-Correa R (2000) Reassessment of the estimation of dissolved oxygen concentration profile and KL in a solid-state fermentation. Process Biochemistry 36: 9-18.
  53. Oostra J, le Comte EP, van den Heuvel JC, Tramper J, Rinzema A (2001) Intra-particle oxygen diffusion limitation in solid-state fermentation. Biotechnol Bioeng 75: 13-24.
  54. Liu D, Zhang R, Yang X, Wu H, Xu D, Tang Z, et al. (2011a) Thermostable cellulase production of Aspergillus fumigatus Z5 under solid-state fermentation and its application in degradation of agricultural wastes. International Biodeterioration & Biodegradation 65: 717-725.
  55. Camassola M, Dillon A (2007) Production of cellulases and hemicellulases by Penicillium echinulatum grown on pretreated sugar cane bagasse and wheat bran in solid-state fermentation. JAM 103: 2196-2204.
  56. Dutta T, Sahoo R, Sengupta R, Ray SS, Bhattacharjee A, Ghosh S (2008) Novel cellulases from an extremophilic filamentous fungi Penicillium citrinum: production and characterization. JIMB 35: 275-282.
  57. Sohail M, Siddiqi R, Ahmad A, Khan SA (2009) Cellulase production from Aspergillus niger MS82: effect of temperature and pH. N Biotechnol 25: 437-441.
  58. Sajith S, Sreedevi S, Priji P, Unni KN, Benjamin S (2014) Production and partial purification of cellulase from a novel fungus, Aspergillus flavus BS1. Ann Microbiol 64: 763-771.
  59. Ojumu TV, Solomon BO, Betiku E, Layokun SK, Amigun B (2003) Cellulase Production by Aspergillus flavus Linn Isolate NSPR 101 fermented in sawdust, bagasse and corncob. African Journal of Biotechnology 2: 150-152.
  60. Brijwani K, Oberoi HS, Vadlani PV (2010) Production of a cellulolytic enzyme system in mixed-culture solid-state fermentation of soybean hulls supplemented with wheat bran. Process Biochemistry 45: 120-128.
  61. Myers RH, Montgomery DC, Anderson-Cook CM (2009) Response surface methodology: process and product optimization using designed experiments. John Wiley & Sons.
  62. Singhania RR, Sukumaran RK, Pandey A (2007) Improved cellulase production by Trichoderma reesei RUT C30 under SSF through process optimization. Appl Biochem Biotechnol 142: 60-70.
  63. Wen Z, Liao W, Chen S (2005) Production of cellulase/beta-glucosidase by the mixed fungi culture of Trichoderma reesei and Aspergillus phoenicis on dairy manure. Appl Biochem Biotechnol 121-124: 93-104.
  64. Singh R, Kumar R, Bishnoi K, Bishnoi NR (2009) Optimization of synergistic parameters for thermostable cellulase activity of Aspergillus heteromorphus using response surface methodology. Biochemical Engineering Journal 48: 28-35.
  65. Singh A, Singh N, Bishnoi NR (2009) Production of cellulases by Aspergillus heteromorphus from wheat straw under submerged fermentation. International Journal of Civil and Environmental Engineering 1: 23-26.
  66. Benjamin S, Smitha R, Jisha V, Pradeep S, Sajith S, et al. (2013) A monograph on amylases from Bacillus spp. Advances in Bioscience and Biotechnology 4: 227-241.
  67. Tao YM, Zhu XZ, Huang JZ, Ma SJ, Wu XB, et al. (2010) Purification and properties of endoglucanase from a sugar cane bagasse hydrolyzing strain, Aspergillus glaucus XC9. J Agric Food Chem 58: 6126-6130.
  68. Akiba S, Kimura Y, Yamamoto K, Kumagai H (1995) Purification and characterization of a protease-resistant cellulase from Aspergillus niger. Journal of fermentation and bioengineering 79: 125-130.
  69. Gao J, Weng H, Xi Y, Zhu D, Han S (2008) Purification and characterization of a novel endo-beta-,4-glucanase from the thermoacidophilic Aspergillus terreus. Biotechnol Lett 30: 323-327.
  70. Murashima K, Nishimura T, Nakamura Y, Koga J, Moriya T, et al. (2002) Purification and characterization of new endo-1, 4-ß-D-glucanases from Rhizopus oryzae. Enzyme and Microbial Technology 30: 319-326.
  71. Riou C, Salmon J-M, Vallier M-J, Günata Z, Barre P (1998) Purification, characterization, and substrate specificity of a novel highly glucose-tolerant ß-glucosidase from Aspergillus oryzae. Applied and Environmental Microbiology 64: 3607-3614.
  72. Begum MF, Alimon AR (2011) Assessment of some wild Aspergillus species for cellulase production and characterization. African Journal of Microbiology Research 5: 4739-4747.
  73. Bagga PS, Sandhu DK, Sharma S (1990) Purification and characterization of cellulolytic enzymes produced by Aspergillus nidulans. J Appl Bacteriol 68: 61-68.
  74. Yan TR, Lin CL (1997) Purification and characterization of a glucose-tolerant beta-glucosidase from Aspergillus niger CCRC 31494. Biosci Biotechnol Biochem 61: 965-970.
  75. Nazir A, Soni R, Saini H, Manhas R, Chadha B (2009) Purification and characterization of an endoglucanase from Aspergillus terreus highly active against barley ß-glucan and xyloglucan. World Journal of Microbiology and Biotechnology 25: 1189-1197.
  76. Pal S, Banik SP, Ghorai S, Chowdhury S, Khowala S (2010) Purification and characterization of a thermostable intra-cellular ß-glucosidase with transglycosylation properties from filamentous fungus Termitomyces clypeatus. Bioresource Technology 101: 2412-2420.
  77. Yang S, Jiang Z, Yan Q, Zhu H (2008) Characterization of a thermostable extracellular beta-glucosidase with activities of exoglucanase and transglycosylation from Paecilomyces thermophila. J Agric Food Chem 56: 602-608.
  78. Liu D, Zhang R, Yang X, Xu Y, Tang Z, et al. (2011). Expression, purification and characterization of two thermostable endoglucanases cloned from a lignocellulosic decomposing fungi Aspergillus fumigatus Z5 isolated from compost. Protein Expression and Purification 79: 176-186.
  79. Li CH, Wang HR, Yan TR (2012) Cloning, purification, and characterization of a heat- and alkaline-stable endoglucanase B from Aspergillus niger BCRC31494. Molecules 17: 9774-9789.
  80. Pol D, Laxman RS, Rao M (2012) Purification and biochemical characterization of endoglucanase from Penicillium pinophilum MS 20. Indian J Biochem Biophys 49: 189-194.
  81. Lee CK, Darah I, Ibrahim CO (2011) Production and optimization of cellulase enzyme using Aspergillus niger USM AI 1 and comparison with Trichoderma reesei via solid state fermentation system. Biotechnology Research International 658493.
  82. Parry JB, Stewart JC, Heptinstall J (1983) Purification of the major endoglucanase from Aspergillus fumigatus Fresenius. Biochem J 213: 437-444.
  83. Bai H, Wang H2, Sun J, Irfan M2, Han M, et al. (2013) Production, purification and characterization of novel beta glucosidase from newly isolated Penicillium simplicissimum H-11 in submerged fermentation. EXCLI J 12: 528-540.
  84. Joo A-R, Jeya M, Lee K-M, Lee K-M, Moon H-J, et al. (2010) Production and characterization of ß-1, 4-glucosidase from a strain of Penicillium pinophilum. Process Biochemistry 45: 851-858.
  85. Lee K-M, Jeya M, Joo A-R, Singh R, Kim I-W, et al. (2010). Purification and characterization of a thermostable endo-ß-1, 4-glucanase from a novel strain of Penicillium purpurogenum. Enzyme and Microbial Technology 46: 206-211.
  86. Ramani G, Meera B, Vanitha C, Rao M, Gunasekaran P (2012) Production, purification, and characterization of a β-glucosidase of Penicillium funiculosum NCL1. Appl Biochem Biotechnol 167: 959-972.
  87. Naika GS, Kaul P, Prakash V (2007) Purification and characterization of a new endoglucanase from Aspergillus aculeatus. J Agric Food Chem 55: 7566-7572.
  88. Chaabouni SE, Mechichi T, Limam F, Marzouki N (2005) Purification and characterization of two low molecular weight endoglucanases produced by Penicillium occitanis mutant Pol 6. Appl Biochem Biotechnol 125: 99-112.
  89. Yasmin S, Mattoo R, Nehvi F (2013) Isolation, characterization and molecular weight determination of cellulase from Trichoderma viride. African Journal of Biotechnology 12: 4503-4511.
  90. Yoon JJ, Cha CJ, Kim YS, Kim W (2008) Degradation of cellulose by the major endoglucanase produced from the brown-rot fungus Fomitopsis pinicola. Biotechnol Lett 30: 1373-1378.
  91. Van Tuan Nguyen DTQ (2010) Purification and properties of a novel thermoactive endoglucanase from Aspergillus awamori VTCC-F099. Australian Journal of Basic and Applied Sciences 4: 6211-6216.
  92. Baraldo Jr A, Borges DG, Tardioli PW, Farinas CS (2014) Characterization of ß-Glucosidase Produced by Aspergillus niger under Solid-State Fermentation and Partially Purified Using MANAE-Agarose. Biotechnology research international 317092.
  93. Immanuel G, Bhagavath C, Raj PI, Esakkiraj P, Palavesam A (2007) Production and partial purification of cellulase by Aspergillus niger and A. fumigatus fermented in coir waste and sawdust. The Internet Journal of Microbiology 3: 1-20.
  94. Kaur J, Chadha BS, Kumar BA, Saini HS (2007) Purification and characterization of two endoglucanases from Melanocarpus sp. MTCC 3922. Bioresour Technol 98: 74-81.
  95. Yinbo Q, Zhu M, Liu K, Bao X, Lin J (2006) Studies on cellulosic ethanol production for sustainable supply of liquid fuel in China. Biotechnol J 1: 1235-1240.
  96. Shen X, Xia L (2006) Lactic acid production from cellulosic material by synergetic hydrolysis and fermentation. Appl Biochem Biotechnol 133: 251-262.
  97. Zhang ZY, Jin B, Kelly JM (2007) Production of lactic acid from renewable materials by Rhizopus fungi. Biochemical Engineering Journal 35: 251-263.
  98. Cao Y, Zhang H, Xiong P (2014) Application of Cellulase-assisted Method to Extract Flavonoids from Plants. Agricultural Science & Technology 15: 729.
  99. Kubicek CP, Mikus M, Schuster A, Schmoll M, Seiboth B (2009) Metabolic engineering strategies for the improvement of cellulase production by Hypocrea jecorina. Biotechnol Biofuels 2: 19.
  100. Chen Y, Wan J, Zhang X, Ma Y, Wang Y (2012) Effect of beating on recycled properties of unbleached eucalyptus cellulose fiber. Carbohydrate Polymers 87: 730-736.
  101. Gil N, Gil C, Amaral ME, Costa AP, Duarte AP (2009) Use of enzymes to improve the refining of a bleached Eucalyptus globulus kraft pulp. Biochemical Engineering Journal 46: 89-95.
  102. Cui L, Meddeb-Mouelhi F, Laframboise F, Beauregard M (2015) Effect of commercial cellulases and refining on kraft pulp properties: Correlations between treatment impacts and enzymatic activity components. Carbohydrate Polymers 115: 193-199.
  103. Kuhad RC, Singh A, Eriksson KE (1997) Microorganisms and enzymes involved in the degradation of plant fiber cell walls. Adv Biochem Eng Biotechnol 57: 45-125.
  104. Lee C, Ibrahim D, Ibrahim CO, Daud WRW (2011) Enzymatic and chemical deinking of mixed office wastepaper and old newspaper: Paper quality and effluent characteristics. BioResources 6: 3859-3875.
  105. Singh A, Yadav RD, Kaur A, Mahajan R (2012) An ecofriendly cost effective enzymatic methodology for deinking of school waste paper. Bioresour Technol 120: 322-327.
  106. Jeffries TW, Klungness JH, Sykes MS, Rutledge-Cropsey KR (1994) Comparison of enzyme-enhanced with conventional deinking of xerographic and laser-printed paper. Tappi Journal 77: 173-179.
  107. Lee C, Darah I, Ibrahim C (2007) Enzymatic deinking of laser printed office waste papers: some governing parameters on deinking efficiency. Bioresource Technology 98: 1684-1689.
  108. Ibarra D, Monte MC, Blanco A, Martínez AT, Martínez MJ (2012) Enzymatic deinking of secondary fibers: cellulases/hemicellulases versus laccase-mediator system. Journal of Industrial Microbiology & Biotechnology 39: 1-9.
  109. Kuhad RC, Gupta R, Singh A (2011) Microbial cellulases and their industrial applications. Enzyme Res 2011: 280696.
  110. Buchert J, Oksanen T, Pere J, Siika-Aho M, Suurnäkki A, et al. (1998) Applications of Trichoderma reesei enzymes in the pulp and paper industry. Trichoderma and gliocladium 2: 343-363.
  111. Bajpai P (1999) Application of enzymes in the pulp and paper industry Biotechnol Prog 15: 147-157.
  112. Hsu J, Lakhani N (2002) Softer and higher strength paper products and methods of making such products. US 20020162635 A1.
  113. Arja M-O (2007) Cellulases in the textile industry Industrial enzymes. Industrial Enzymes, Springer pp. 51-63.
  114. Cortez JM, Ellis J, Bishop DP (2001) Cellulase finishing of woven, cotton fabrics in jet and winch machines. J Biotechnol 89: 239-245.
  115. Saravanan D, Vasanthi N, Ramachandran T (2009) A review on influential behaviour of biopolishing on dyeability and certain physico-mechanical properties of cotton fabrics. Carbohydrate Polymers 76: 1-7.
  116. Mitchinson C, Wendt DJ (2001) Variant EGIII-like cellulase compositions. US 6268328 B1.
  117. Kottwitz B, Schambil F (2004) Cellulase and cellulose containing detergent. US 20050020472 A1.
  118. Rai P, Majumdar G, Gupta SD, De S (2007) Effect of various pretreatment methods on permeate flux and quality during ultrafiltration of mosambi juice. Journal of Food Engineering 78: 561-568.
  119. De Carvalho LMJ, De Castro IM, Da Silva CAB (2008) A studies of retention of sugars in the process of clarification of pineapple juice (Ananas comosus, L. Merril) by micro-and ultra-filtration. JFE 87: 447-454.
  120. Ajayi A, Peter-Albert C, Adedeji O (2015) Modification of Cell Wall Degrading Enzymes from Soursop (Annona muricata) Fruit Deterioration for Improved Commercial Development of Clarified Soursop Juice (A Review). Med Aromat Plants 4: 2167-2412.
  121. Karmakar M, Ray R (2010) Current trends in research and application of microbial cellulases. RJM 6: 41-53.
  122. Rosegrant MW, Msangi S, Sulser T, Valmonte-Santos R (2006) Biofuels and the global food balance. Bioenergy and Agriculture: Promises and Challenges. Washington, DC: International Food Policy Research Institute (IFPRI).
  123. Msangi S, Sulser T, Rosegrant M, Valmonte-Santos R (2007) Global scenarios for biofuels: Impacts and implications for food security and water use. Paper presented at the 10th Annual Conference on Global Economic Analysis, Purdue University, Indiana.
  124. Sun Y, Cheng J (2002) Hydrolysis of lignocellulosic materials for ethanol production: a review. Bioresour Technol 83: 1-11.
  125. Zhou J, Wang YH, Chu J, Zhuang YP, Zhang SL, et al. (2008) Identification and purification of the main components of cellulases from a mutant strain of Trichoderma viride T 100-14. Bioresour Technol 99: 6826-6833.
  126. Binod P, Sindhu R, Singhania RR, Vikram S, Devi L, et al. (2010) Bioethanol production from rice straw: an overview. Bioresource technology 101: 4767-4774.
  127. Chen H, Qiu W (2010) Key technologies for bioethanol production from lignocellulose. Biotechnol Adv 28: 556-562.
  128. Liu W, Zhu WM (2000) Production and regeneration of Trichosporon cutaneum protoplasts. Process Biochemistry 35: 659-664.
  129. Wiatr CL (1990) Application of cellulase to control industrial slime: Google Patents.
  130. Qin C, Zhou B, Zeng L, Zhang Z, Liu Y, et al. (2004) The physicochemical properties and antitumor activity of cellulase-treated chitosan. Food Chemistry 1: 107-115.
  131. Wu GJ, Tsai GJ (2004) Cellulase degradation of shrimp chitosan for the preparation of a water-soluble hydrolysate with immunoactivity. Fisheries Science 70: 1113-1120.
  132. Han W, He M (2010) The application of exogenous cellulase to improve soil fertility and plant growth due to acceleration of straw decomposition. Bioresour Technol 101: 3724-3731.
  133. Saqib AA, Hassan M, Khan NF, Baig S (2010) Thermostability of crude endoglucanase from Aspergillus fumigatus grown under solid state fermentation (SSF) and submerged fermentation (SmF). Process Biochemistry 45: 641-646.
  134. Jadhav A, Girde A, More S, More S, Khan S (2013) Cellulase production by utilizing agricultural wastes. RJAFS 1: 6-9.
  135. Mrudula S, Murugammal R (2011) Production of cellulase by Aspergillus niger under submerged and solid state fermentation using coir waste as a substrate. BJM 42: 1119-1127.
  136. Devi MC, Kumar MS (2012) Production, Optimization and Partial purification of Cellulase by Aspergillus niger fermented with paper and timber sawmill industrial wastes. JMBR 2: 120-128.
  137. Kathiresan K, Manivannan S (2010) Cellulase production by Penicillium fellutanum isolated from coastal mangrove rhizosphere soil. RJM 5: 1160-1164.
  138. Karmakar M, Ray RR (2011) A statistical approach for optimization of simultaneous production of ß-glucosidase and endoglucanase by Rhizopus oryzae from solid-state fermentation of water hyacinth using central composite design. BRI.
  139. Rubeena M, Neethu K, Sajith S, Sreedevi S, Priji P, et al. (2013) Lignocellulolytic activities of a novel strain of Trichoderma harzianum. ABB, 4: 214-221.
  140. Kocher G, Kalra K, Banta G (2008) Optimization of cellulase production by submerged fermentation of rice straw by Trichoderma harzianum Rut-C 8230. TIJM.
  141. da Silva Delabona P, Farinas CS, da Silva MR, Azzoni SF, da Cruz Pradella JG (2012) Use of a new Trichoderma harzianum strain isolated from the Amazon rainforest with pretreated sugar cane bagasse for on-site cellulase production. Bioresource Technology 107: 517-521.
  142. Liming X, Xueliang S (2004) High-yield cellulase production by Trichoderma reesei ZU-02 on corn cob residue. Bioresour Technol 91: 259-262.
  143. Neethu K, Rubeena M, Sajith S, Sreedevi S, Priji P, et al. (2012) A novel strain of Trichoderma viride shows complete lignocellulolytic activities. ABB 3: 1160-1166.
  144. Sherief A, El-Tanash A, Atia N (2010) Cellulase production by Aspergillus fumigatus grown on mixed substrate of rice straw and wheat bran. RJM 5: 199-211.
  145. Chandra MS, Viswanath B, Reddy BR (2007) Cellulolytic enzymes on lignocellulosic substrates in solid state fermentation by Aspergillus niger. Indian J Microbiol 47: 323-328.
  146. Jecu L (2000) Solid state fermentation of agricultural wastes for endoglucanase production. ICP 11: 1-5.
  147. Kang SW, Park YS, Lee JS, Hong SI, Kim SW (2004) Production of cellulases and hemicellulases by Aspergillus niger KK2 from lignocellulosic biomass. Bioresour Technol 91: 153-156.
  148. Gao J, Weng H, Zhu D, Yuan M, Guan F, et al. (2008b) Production and characterization of cellulolytic enzymes from the thermoacidophilic fungal Aspergillus terreus M under solid-state cultivation of corn stover. Bioresour Technol 99: 7623-7629.
  149. AboState M, Hammad A, Swelin M, Gannam R (2010) Enhanced production of cellulases by Aspergillus spp. isolated from agriculture wastes by solid state fermentation. American Eurasium J Agric Environ Science 8: 402-410.
  150. Deswal D, Khasa YP, Kuhad RC (2011) Optimization of cellulase production by a brown rot fungus Fomitopsis sp. RCK2010 under solid state fermentation. Bioresour Technol 102: 6065-6072.
  151. Mo H, Zhang X, Li Z (2004) Control of gas phase for enhanced cellulase production by Penicillium decumbens in solid-state culture. PB 39: 1293-1297.
  152. Wang JS, Wang J, Gulfraz M (2005) Efficient cellulase production from corn straw by Trichoderma reesei LW1 through solid state fermentation process. Ethnobotanical Leaflets.
  153. Dueñas R, Tengerdy RP, Gutierrez-Correa M (1995) Cellulase production by mixed fungi in solid-substrate fermentation of bagasse. World J Microbiol Biotechnol 11: 333-337.
  154. Kalogeris E, Christakopoulos P, Katapodis P, Alexiou A, Vlachou S, et al. (2003) Production and characterization of cellulolytic enzymes from the thermophilic fungus Thermoascus aurantiacus under solid state cultivation of agricultural wastes. PB 38: 1099-1104.
  155. Iwashita K, Todoroki K, Kimura H, Shimoi H, Ito K (1998) Purification and characterization of extracellular and cell wall bound beta-glucosidases from Aspergillus kawachii. Biosci Biotechnol Biochem 62: 1938-1946.
  156. Yan TR, Lin YH, Lin CL (1998) Purification and Characterization of an Extracellular beta-Glucosidase II with High Hydrolysis and Transglucosylation Activities from Aspergillus niger. J Agric Food Chem 46: 431-437.
  157. Peshin A, Mathur J (1999) Purification and characterization of ß-glucosidase from Aspergillus niger strain 322. Letters in applied microbiology 28: 401-404.
  158. Elshafei AM, Hassan MM, Haroun BM, Abdel-Fatah OM, Atta HM, et al. (2009) Purification and properties of an endoglucanase of Aspergillus terreus DSM 826. J Basic Microbiol 49: 426-432.
  159. Macris BJ (1984) Production and Characterization of Cellulase and beta-Glucosidase from a Mutant of Alternaria alternata. Appl Environ Microbiol 47: 560-565.
  160. Christakopoulos P, Goodenough PW, Kekos D, Macris BJ, Claeyssens M, et al. (1994) Purification and characterisation of an extracellular beta-glucosidase with transglycosylation and exo-glucosidase activities from Fusarium oxysporum. Eur J Biochem 224: 379-385.
  161. Saha BC (2004) Production, purification and properties of endoglucanase from a newly isolated strain of Mucor circinelloides. Process Biochemistry 39: 1871-1876.
  162. Ahmed S, Bashir A, Saleem H, Saadia M, Jamil A (2009) Production and purification of cellulose-degrading enzymes from a filamentous fungus Trichoderma harzianum. PJB 41: 1411-1419.
  163. Hafiz Muhammad Nasir I, Ishtiaq A, Muhammad Anjum Z, Muhammad I (2011) Purification and characterization of the kinetic parameters of cellulase produced from wheat straw by Trichoderma viride under SSF and its detergent compatibility. ABB 2: 149-156.
  164. Streffer F (2014) Lignocellulose to biogas and other products. JSM Biotechnol Bioeng 2: 1023.
  165. Benjamin S, Pandey A (1998) Candida rugosa lipases: molecular biology and versatility in biotechnology. Yeast 14: 1069-1087.
  166. Berry DR, Paterson A (1990) Enzymes in the food industry Enzyme Chemistry: Springer 306-351.
  167. Gamarra NN, Villena GK, Gutiérrez-Correa M (2010) Cellulase production by Aspergillus niger in biofilm, solid-state, and submerged fermentations. Appl Microbiol Biotechnol 87: 545-551.
  168. Gokhan C, Arikan B, Ünaldi MN, Güvenmez H (2002) Some properties of crude carboxymethyl cellulase of Aspergillus niger Z10 wild-type strain. TJB 26: 209-213.
  169. Ikram-ul-Haq KS, Hameed U, Javed MM, Qadeer M (2006) Solid-State Fermentation of Cellulases by Locally Isolated Trichoderma harzianum for the Exploitation of Agricultural Byproducts. PJBS 9: 1779-1782.
  170. Ljungdahl LG, Eriksson KE (1985) Ecology of microbial cellulose degradation Advances in Microbial Ecology: Springer 237-299.
  171. Shankar T, Isaiarasu L (2012) Statistical optimization for cellulase production by Bacillus pumilus Ewbcm1 using response surface methodology. GJBB 7: 01-06.
  172. Sjostrom E (1993) Wood chemistry: fundamentals and applications: Elsevier.
  173. Stockton B, Mitchell D, Grohmann K, Himmel M (1991) Optimum β-D-glucosidase supplementation of cellulase for efficient conversion of cellulose to glucose. Biotechnology Letters 13: 57-62.
  174. Szengyel Z, Zacchi G, Varga A, Réczey K (2000) Cellulase production of Trichoderma reesei Rut C 30 using steam-pretreated spruce. Hydrolytic potential of cellulases on different substrates. App l Biochem Biotechnol 84-86: 679-91.
  175. Thiry M, Cingolani D (2002) Optimizing scale-up fermentation processes. Trends Biotechnol 20: 103-105.
Citation: Sajith S, Priji P, Sreedevi S, Benjamin S (2016) An Overview on Fungal Cellulases with an Industrial Perspective. J Nutr Food Sci 6:461.

Copyright: © 2016 Sajith S, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top