Open access peer-reviewed chapter

Enzymes and Phytohormones from Micromonospora

Written By

Waleed M. Abdulkhair and Mousa A. Alghuthaymi

Submitted: 03 March 2015 Reviewed: 27 July 2015 Published: 11 February 2016

DOI: 10.5772/61261

From the Edited Volume

Actinobacteria - Basics and Biotechnological Applications

Edited by Dharumadurai Dhanasekaran and Yi Jiang

Chapter metrics overview

2,157 Chapter Downloads

View Full Metrics

Abstract

Actinobacteria produce diverse and huge amounts of enzymes that are widely used in different industrial purposes. Specific properties of enzymes allow to run the reactions under milder conditions with improved yield and reduced wastes. Further redesign for natural enzymes is very essential because they are often not suitable for biocatalytic processes. Recently, new microbial natural and creating enzymes are discovered synchronous with the late advanced technologies of genomics, metagenomics, proteomics, efficient expression systems and emerging recombinant DNA. The ongoing development of enzyme biotechnology aids in the improvement of the industrial biocatalysis field. Thermophilic actinobacteria produce thermostable enzymes that are widely used in industrial processes. In contrast, psychrophilic actinobacteria grow well at low temperatures and subsequently their enzymes are more effective at low temperatures. The advanced DNA sequencing technique allows determining and identifying the sequences and functions of all the genes that synthesize proteins that are widely use in the industry. Recombinant strains can be obtained by using certain biotechnological tools to potentially increase enzymes production on a large scale. The ongoing development in this field will lead to the improvement of different industrial purposes such as food, chemicals, textiles, leather, pharmaceuticals, and so on.

Keywords

  • Actinomycetes
  • biocatalysts
  • enzyme biotechnology
  • industrial microbiology

1. Introduction

Actinobacteria are recognized by their morphology thin, elongated cells with branching filaments, as its name denotes. Nevertheless, morphology alone is not enough to add an organism to the actinobacterial group, so the advanced identification depends on other techniques such as cultural, physiological, biochemical and molecular characteristics. Moreover, phylogenetic analysis is necessary to distinguish between true actinobacteria and those that have closely phylogenetic affinities, and at the same time to precisely determine the similarity among different species of the same genus. A lot of enzymes produced from actinobacteria are used in industries thus they are called "industrial enzymes". These enzymes employ in the industry of paper, leather, detergents, textiles, pharmaceuticals, chemicals, foods, beverages, biofuels, animal feed, personal care, and others [1]. The sustainable industry based on enzymes technology depends on the biotechnological development, which is represented in the modern molecular techniques such as metagenomics and genomics. The latter allows the discovery of new potential recombinant-biocatalytic enzymes used in different industries needed by society. These industrial enzymes are employed to yield over 500 products [2, 3]. Although chemical and organic methods are used in industries, they have several disadvantages non-economic, unfriendly to the environment, lacking of enantiomeric specificity for chiral synthesis, need for high temperature, low pH and high pressure and leads to organic waste and pollutants. On the other hand, enzymes are more useful in industrial applications because they act at mild reaction conditions, have a long half-life, and are friendly to the environment [4]. Moreover, gene-encoded enzymes can be modified to improve their properties stability, substrate specificity and specific activity. There are many competitive companies that participate in the enzyme industry Novozymes being the largest one followed by DSM and DuPont among others. The companies' competition relies on certain criteria such as product quality, performance, use of intellectual property rights, and the ability to innovate.

Advertisement

2. Enzymes detection

Recently, new enzymes can be detected by using certain biotechnological tools such as metagenome screening [5, 6], microbial genome mining [7, 8], and exploring the diversity of extremophiles [9, 10].

2.1. Metagenome screening

Metagenome is a new manner for microbial screening based on the function and/or sequence of the gene [6, 11, 12]. Function-based screening is a method by which gene isolation is carried out depending on the desired gene function through phenotype detection, heterologous complementation, and induced gene expression [13]. Whilst sequence-based screening is a method performed by either polymerase chain reaction (PCR) or hybridization procedures. Strains producing industrial enzymes are isolated from different environments such as volcanic vents, arctic tundra, cow rumen [14], marine environments [15], and termite guts [16] such as lipase [17, 18], oxidoreductase [19, 20], amidase [21], amylase and nitrilase [22], beta-glucosidase [23, 24], decarboxylase, and epoxide hydrolase [25]. Function-based screening method has main disadvantages due to its inefficiency and biased expression of foreign genes in Escherichia coli as an alternative host [12]. To overcome these problems, alternative bacterial host and expression systems are currently being examined including Streptomyces lividans, Pseudomonas putida and Rhizobium leguminosarum, among others [26, 27]. Target rate depends on other factors such as gene size and the assay method. Enzyme activity can be assayed by agar plate method, which has a sensitivity developed by using cell lysates [28], toxics resistant genes [29, 30], correlation of enzyme activity with a phenotypic characteristic such as green fluorescent protein (GFP) [31], or β-galactosidase [32]. Also, development of flow cytometry-based screens as SIGEX are leading the way as they enable more rapid screening of large metagenomic libraries [33].

2.2. Microbial genome mining

New enzymes particularly used in industries are explored from huge genome sequence databases [34]. The advanced genome sequencing programs, such as 454 from Roche, Solexa from Illumina, or SoLiD from ABI, reduce time and cost. Using these programs support the complete and accurate reading of multiple whole genomes in less than two weeks [35]. Two approaches are being followed to discover new enzymes [36].

2.3. Exploring the diversity of extremophiles

More than 30 papers were published on extremophiles in a special issue [37]. Extremophiles are promising microorganisms for enzyme production because they can grow under extreme physical conditions such as temperature (2-12°C, 60-110°C), pressure or radiation, and/or geochemical conditions such as salinity (2-5% NaCl) and pH (< 2, > 9). High biodiversity for extremophiles is present under all extreme conditions mentioned [10, 38]. For example, there are some thermostable enzymes such as proteases, lipases, cellulases and amylases that are successfully used in different industrial applications [10, 39]. Thermostable enzymes are isolated from different thermophilic bacteria such as Clostridium, Thermus, Thermotoga, and Bacillus, as well as Archaeabacteria such as Pyrococcus, Thermococcus or Methanopyrus. The most common thermostable enzyme is Taq DNA polymerase which isolated from Thermus aquaticus [40]. Moreover, thermostable enzymes are greatly used for detergent manufacturing. Polymer-degrading enzymes such as cellulases and xylanases are used for pulp and paper industries, and are also used with other applications such as extraction and clarification of fruit juices, improvement of bakery products, polishing and stone-washing of textiles, and bioremediation of waters contaminated with oils or hydrocarbons [10, 41 - 43]. Halophites are produced for specific enzymes that tolerate salt stress by containing a considerable number of negatively-charged amino acids that prevent precipitation [44]. The most common halophites that used in industrial applications are Halobacterium, Halobacillus and Halothermothrix [45, 46]. Thermoalkaliphilic enzymes, such as proteases and lipases, are produced by microorganisms that grow under extreme pH values and used as additives in laundry and dishwashing detergents [10, 47].

Advertisement

3. Improvement of microbial enzymes

Improvement of enzyme properties can be carried out by creating a growing demand [40]. Industrial enzymes usually need further fine tuning to achieve industrial scale production [48]. Using recombinant DNA technology in large industrial scales is very urgent due to the increase in production by 100-fold [49]. There are two main manners for enzymes modification to adapt their functions rational redesign and combinatorial methods.

3.1. Rational redesign

This strategy is based on using direct mutation to hit amino acid substitutions, which require complete information about the 3D structure and chemical mechanism of the enzymatic reaction. Databases include protein structures and sequences, so the sequence comparison of a new enzyme with the references can identify related enzymes whose functions or/and structures are already known [50 - 54].

3.2. Combinatorial methods

Combinatorial methods rely on certain factors relevant to an enzyme molecule such as chirality, biocatalytic effect, catalytic rate, solubility, specificity and stability. Combinatorial methods are faster and cheaper than methods for finding new enzymes it acts better than natural methods under specific conditions [3, 55 - 57]. Combinatorial methods, also called "directed evolution" have random mutagenesis in the protein-encoding gene using different techniques such as PCR [58], repeated oligonucleotide directed mutagenesis [59], and chemical agents [60]. The PCR technique introduces both random and point mutations in a large number of enzymes. These techniques perform random recombinant in vitro, typically between parent genes with homology higher than 70% [61]. The point mutation is carried out by the target active site residues (about 10–15 amino acids) and those closest to it (another 20–30 amino acids) [62]. There is another way, called CAS Ting, which is based on a combinatorial active site testing in which libraries are generated from groups of two or three residues made from the active site residues [63]. While running PCR, the amino acid alphabet is reduced and new proteins composed of only 12 amino acids are synthesized [64 - 66]. Further techniques can create shuffle exons or domains [67], loop regions [68], random truncations [69], or insertions and deletions of codons [70]. Moreover, random redesign technology is used to create new and improved enzymes that have the highest activity, are more stable at different pH values and temperatures [71], have increased chirality [72], altered substrate specificity [73], stability in organic solvents [74], novel substrate specificity and activity [75] and increased biological activity of protein pharmaceuticals and biological molecules [76, 77]. Directed evolution method could improve the activity of glyphosate-N-acetyltransferase by 10,000-fold and thermostability by 5-fold [78]. Directed evolution work has resulted in the presence of new and improved enzymatic proteins in the market since 2000 [79, 80].

Advertisement

4. Production of recombinant microbial proteins

Molecular biology techniques, particularly recombinant DNA, have a great effect in enzyme production from different microorganisms such as Bacillus spp, Ralstonia eutropha, Pseudomonas fluorescens, Saccharomyce cerevisiae, Pichia pastoris, Hansenula polymorpha, Aspergillus spp, Trichoderma and E. coli [81]. The latter is most common bacteria used in this purpose due to its accurate genome modification, rapid growth, and good growth on different media [82, 83]. Moreover, the Pseudomonas fluorescens bacteria of the Pfenex Company can produce 20 g/L of protein [84]. On the other hand, Saccharomyces cerevisiae is more useful than bacteria because it usually used as a cloning host, it has high cell density, produces heterologous proteins, and its genetics are more advanced than any other eukaryote. Despite all the benefits mentioned above, this yeast is not convenient for use in the mammalian proteins industry at the large-scale because drawbacks may occur such as hyperglycosylation, antigenic response in patients due to accumulation of α-1,3- mannose residues, and absence of strong and tightly-regulated promoters [85]. P. pastoris is regarded as one of the most common industrial microorganism and could produce over 700 proteins [81, 86, 87]. Recombinant protein production had 22 g/L for intracellular proteins [88] and 14.8 g/L for secreted proteins [89]. P. pastoris can produce up to 30 g/L of recombinant proteins [90 - 94]. Although DNA recombination technology is more easy and available, gene insertion and deletion remain difficult [95, 96]. The problem at industrial level is that non-fungal proteins production is low compared to homologous proteins. There are many strategies used to overcome this problem such as the establishment of weak protease strains [97], insertion of a considerable number of gene copies [98], use of strong fungal promoters, efficient secretion signals [98, 99], and gene fusions with a gene that encodes part of or an entire well-secreted protein [99]. Chrysosporium lucknowense was found to have a great ability for protein production (50–80 g/L), and from which low-viscosity and low-protease mutants have been obtained [100, 101].

Advertisement

5. Biocatalysts

Biocatalysts are widely used in different economic industries such as food industry [102]. Biocatalysis can be carried out by using intact cells, immobilized cells, cell free extracts, purified enzymes, or immobilized enzymes [103,104]. Microbial industry has been developed rapidly due to due to using the recent techniques such as genome sequencing, directed evolution, protein expression, metabolic engineering, and structural biology [105,106].

5.1. Enzymes applications

Enzymes have great importance in industrial processes [107, 108]. These enzymes are used in the detergent, textiles, pulp, paper, leather, and biofuels industries. Biofuels have the highest sales [109, 110]. In the textiles industry, enzymes are used as cleaners, reducing the use of raw materials and waste production [111]. Pectate lyase is used in the cotton industry [112]. Lipases, xylanases, and laccases are used in removing pitch in the pulp industry [113 - 115]. Cellulases are widely used in the textiles industry and are also used in the degradation of lignocellulosic feed stocks. There are several cellulases such as endoglucanases that degrade cellulose randomly, cellobiohydrolases that release glucose dimers from both ends of cellulose chains, and beta-glucosidases that hydrolyze oligomer chains to liberate glucose molecules [116 - 118]. The main microbial source of cellulases is Trichoderma reesei which depolymerizes plant blocks to free sugar molecules [119 - 124].

5.2. Enzymatic food industry

A certain group of enzymes plays an important role in the food industry and subsequently achieves great revenue [125]. The mechanism of feed enzyme action supports nutrient digestion and therefore leads to a much easier feed utilization. Moreover, they hydrolyze complex components that can be deleterious or have no value [126]. The most common commercial feed enzymes are phytases, proteases, α-galactosidases, glucanases, xylanases, α-amylases, and polygalacturonases, which are used in swine and poultry products industries [127, 128]. Lipase is a distinguishable group of enzymes that is used in the food industry. The maximum yield obtained from lipases requires optimization of enzyme concentration, pH, temperature, and emulsion content. The most common recombinant fungal lipases are produced from Rhizomucor miehi, Thermomyces lanuginosus and Fusarium oxysporum [129, 130]. Protease is also an important group of enzymes that is mainly used in the dairy industry, such as cheese manufacturing where it hydrolyzes a specific peptide bond that generates para-k-casein and macropeptides [131]. Recombinant protease of A. niger var awamori can produce 1 g/L of chymosin after nitrosoguanidine mutagenesis and the selection for 2-deoxyglucose resistance [132, 133]. There are four recombinant proteases that have been registered by the FDA for cheese production [128, 134]. Although all enzymes mentioned above are the most common in the food industry, there are others used in this application such as invertase used for candy and jam manufacture, β-galactosidase (lactase) used for hydrolysis of lactose from milk or whey, and galactosidase used for crystallization of beet sugar [135 - 137].

5.3. Enzymatic processing of chemicals and pharmaceuticals

The chemicals industry depends on enzyme technology that is low cost, has easy methods, and high quality [138]. Enzymatic processing requires lower energy, which has many benefits such as high yield, high catalytic activity, low releasing of wastes and byproducts, and lower volumes of wastewater streams [139]. Genomic and proteomic technology development improved enzyme properties which in turn improved the chemicals industry [140, 141]. L-tyrosine is a main compound for protease conversion [142, 143]. Beta-lactam antibiotics manufacturing is one of the common pharmaceuticals industries that depend on enzymes technology [144]. Esterases, lipases, proteases and ketoreductases are used in the industry of chiral alcohols, carboxylic acids, amines or epoxides [110, 145, 146]. Sometimes, recombinant microbial enzymes raise the yield percentage up to 100% [147]. A combination of random gene mutagenesis and ProSAR analysis increase the chirality of ketoreductase enzyme toward tetrahydrothiophene-3-one from 63 to 99% [148 - 152]. The improved enzymatic biotechnology supports the production of 2-methyl pentanol as an important intermediate for pharmaceuticals manufacture [153 - 155]. Also, an improved acyltransferase aids the conversion of cholesterol-lowering agent, lovastatin, to simvastatin [156, 157]. Moreover, lipase is used in stereoselectivity for acetylation of asymmetrical diol during an antifungal agent production [158]. Enzymatic biotechnology has many advantages such as higher substrate solubility, reversal of hydrolytic reactions, and modified enzyme specificity, which result in new enzyme activities [159].

Advertisement

6. Industrial enzymes

6.1. Pectinases

Actinobacterial pectinases are widely used in various industrial applications such as food and beverages industries, as well as fruit treatment including fruit maturation, viscosity rising, decreasing of must, preliminary treatment of must for wine industries, extraction of tomato pulp, and tea and chocolate fermentation [160, 161]. Pectinases are also used in the textile and paper industries in plant fiber degumming [162 - 165]. Moreover, the combination of pectinase and β-glucosidase supports the scent and volatile substances of fruits and vegetables, and raise the content of antioxidants [166, 167]. Pectinases are produced from several genera of microorganisms such as Bacillus, Aspergillus, Rhizopus, Trichoderma, Pseudomonas, Penicillium and Fusarium [168, 169].

6.2. Lipases

Lipases are used in various commercial applications, such as in the detergents industry. The most common lipase-producing microorganisms are Penicillium restrictum, Candida rugosa, Candida antarctica, Pseudomonas alcaligenes, Pseudomonas mendocina, Burkholderia cepacia [72], Geotrichum candidum DBM 4013, Pseudomonas cepacia, Bacillus stearothermophilus, Burkholderia cepacia, Candida lipolytica, Bacillus coagulans, Bacillus coagulans BTS-3, Pseudomonas aeruginosa PseA, Clostridium thermocellum 27405, Yarrowia lipolytica and Yarrowia lipolytica CL180 [170 - 179].

6.3. Lactases

Lactases or β-galactosidases are hydrolyzing enzymes that catalyze the hydrolysis of lactose at terminal residues and produce glucose and galactose. The microbial source of lactases are yeasts such as Kluyveromyces lactis, K. fragilis and Candida pseudotropicalis; bacteria such as Escherichia coli, Lactobacillus bulgaricus, Streptococcus lactis and Bacillus sp; and fungi, such as Aspergillus foetidus, A. niger, A. oryzae and A. Phoenecia [180,181]. Beta-galactosidase is widely used in the dairy industry, and also used in dairy products crystallization such as milk candy, condensed milk, frozen concentrated milk, yoghurt and ice cream mixtures [182 - 185].

6.4. Cellulases

Cellulases are hydrolyzing enzymes for cellulose substances to produce cellobiose and then glucose [186]. The combination of cellulases and pectinases provides well in applications in juice and wine industries [187]. There are a wide array of microorganisms that produce cellulases, such as Trichoderma, Penicillium, Aspergillus, Fusarium, Phoma, Acidothermus, Bacillus, Pseudomonas, Staphylococcus, Streptomyces, Xanthomonas, Acetovibrio, Clostridium, Pseudonocardia, and Thermoanaerobacter [188, 189].

6.5. Amylases

Amylases are used in the textiles, beer, liquor, bakery, infant feeding cereals, and animal feed industries. Aspergillus and Rhizopus are the most common amylases producers [190,191]. Amylases are used in the food industry in the conversion of starch into dextrin which in turn is converted to maltose, which used in the manufacture of soft drinks, beer, jellies, and ice cream. Maltose can be converted into glucose, which used in the soft-drinks and bakery industries [192,193]. Amylases are the most used enzymes in bread baking [194]. Amylases also play an important role in the pharmaceuticals industry [195 - 199].

6.6. Proteases

Proteases are used in the baking, feed and brewing industries. Proteases catalyze the splitting of peptide linkages in proteins. Proteases are classified into exopeptidases and endopeptidases according to the site of the concerned peptide bond to be cleaved. Recently proteases represent 60% of industrial enzymes on the market because they are easy to obtain and to recover. Proteases have high coagulation activity, with fewer risks [200]. Proteases are mainly used in the industry of soluble proteins, chemicals and pharmaceuticals [201]. Proteases are produced by Bacillus, Thermus caldophilus, Desulfurococcus mucosus, Streptomyces and Escherichia coli [202].

6.7. Glucose oxidase

Glucose oxidase can oxidize β-D-glucose with the formation of D-gluconolactone. The enzyme is used to remove harmful oxygen in the food industry to avoid toxicity [203].

6.8. Glucose isomerase

Glucose isomerase catalyzes the reversible isomerase from D-glucose and D-xylose into D-fructose and D-xylulose respectively. Glucose isomerase plays an important role in the food industry such as in the production of fructose-rich corn syrup [197]. The cloned gene xylA of Thermus themophilus is introduced to Saccharomyces cerevisiae to be expressed under the control of the yeast PGK1 promoter [205,206].

6.9. Invertase

Invertase is produced by Saccharomyces cerevisiae and other microorganisms. The enzyme catalyzes the hydrolysis from sucrose to fructose and glucose. The supplementation of an invertase to banana juice is to increase its sweetness and viscosity [207 - 210].

References

  1. 1. Adrio JL, Demain AL. Microbial Cells and Enzymes—A Century of Progress. In Methods in Biotechnology. Microbial Enzymes and Biotransformations. 2nd ed. Barredo JL, Ed; Humana Press: Totowa, NJ, USA; 2005. Vol. 17, p. 1–27.
  2. 2. Johannes TW, Zhao H. Directed evolution of enzymes and biosynthetic pathways. Curr. Opin. Microbiol. 2006; 9: 261–267.
  3. 3. Kumar A, Singh S. Directed evolution: Tailoring biocatalysis for industrial application. Crit. Rev. Biotechnol. 2013; 33: 365–378.
  4. 4. Johnson EA. Biotechnology of non-Saccharomyces yeasts the ascomycetes. Appl. Microbiol. Biotechnol. 2013; 97: 503–517.
  5. 5. Rondon MR, Goodman RM, Handelsman J. The Earth’s bounty: Assessing and accessing soil microbial diversity. Trends Biotechnol. 1999; 17: 403–409.
  6. 6. Gilbert JA, Dupont CL. Microbial metagenomics: Beyond the genome. Annu. Rev. Mar. Sci. 2011; 3: 347–371.
  7. 7. The Institute for Genomic Research (TIGR)-JCVI. http://cmr.jcvi.org/tigr (accessed on 1 July 2013).
  8. 8. Kaul P, Asano Y. Strategies for discovery and improvement of enzyme function: State of the art and opportunities. Microbiol. Biotechnol. 2012; 5: 18–33.
  9. 9. Schiraldini C, de Rosa M. The production of biocatalysts and biomolecules from extremophiles. Trends Biotechnol. 2002; 20: 515–521.
  10. 10. Kumar L, Awasthi G, Singh B. Extremophiles: A novel source of industrially important enzymes. Biotechnol. 2011; 10: 1–15.
  11. 11. Steele HL, Jaeger KE, Daniel R, Streit WR. Advances in recovery of novel biocatalysts from metagenomes. J. Mol. Microbiol. Biotechnol. 2009; 16: 25–37.
  12. 12. Uchiyama T, Miyazaki K. Functional metagenomics for enzyme discovery: Challenges to efficient screening. Curr. Opin. Biotechnol. 2009; 20: 616–622.
  13. 13. Li S, Yang X, Yang S, Zhu M, Wang X. Technology prospecting on enzymes: Application, marketing and engineering. Comput. Struct. Biotechnol. J. 2012; 2: 1–11.
  14. 14. Hess M, Sczyrba A, Egan R, Kim TW, Chokhawala H, Schroth G, Luo S, Clark DS, Chen F, Zhang T, et al. Metagenomic discovery of biomass-degrading genes and genomes from cow rumen. Sci. 2011; 331: 463–467.
  15. 15. Kennedy J, Marchesi JR, Dobson AD Marine metagenomics: Strategies for the discovery of novel enzymes with biotechnological applications from marine environments. Microb. Cell Fact. 2008; 7: 27–37.
  16. 16. Warnecke F, Luginbühl P, Ivanova N, Ghassemian M, Richardson TH, Stege JT, Cayouette M, McHardy AC, Djordjevic G, Aboushadi N, et al. Metagenomic and functional analysis of hindgut microbiota of a wood-feeding higher termite. Nature 2007; 450: 560–565.
  17. 17. Jeon JH, Kim JT, Kim YJ, Kim HK, Lee HS, Kang SG, Kim SJ, Lee JH. Cloning and characterization of a new-cold active lipase from a deep-se sediment metagenome. Appl. Microbiol. Biotechnol. 2009; 81: 865–874.
  18. 18. Fernandez-lvaro E, Kourist R, Winter J, Böttcher D, Liebeton K, Naumer C. Enantioselective kinetic resolution of phenylalkyl carboxylic acids using metagenome-derived esterases. Microb. Biotechnol. 2010; 3: 59–64.
  19. 19. Knietsch A, Waschkowitz T, Bowien S, Henne A, Daniel R. Construction and screening of metagenomic libraries derived from enrichment cultures: Generation of a gene bank for genes conferring alcohol oxidoreductase activity on Escherichia coli. Appl. Environ. Microbiol. 2003; 69: 1408–1416.
  20. 20. Gabor EM, de Vries EJ, Janssen DB. Construction, characterization, and use of small-insert gene banks of DNA isolated from soil and enrichment cultures for the recovery of novel amidases. Environ. Microbiol. 2004; 6: 948–958.
  21. 21. Rondon MR, August PR, Betterman AD, Brady SF, Grossman TH, Liles MR. Cloning the soil metagenome: A strategy for accessing the genetic and functional diversity of uncultured microorganisms. Appl. Environ. Microbiol. 2000; 66: 2541–2547.
  22. 22. Bayer S, Birkemeyer C, Ballschmiter M. A nitrilase from a metagenomic library acts regioselectively on aliphatic dinitriles. Appl. Microbiol. Biotechnol. 2011; 89: 91–98.
  23. 23. Wang K, Li G, Yu SQ, Zhang CT, Liu YH. A novel metagenome-derived β-galactosidase: Gene cloning, overexpression, purification and characterization. Appl. Microbiol. Biotechnol. 2010; 88: 155–165.
  24. 24. Jiang C, Shen P, Yan B, Wua B. Biochemical characterization of a metagenome-derived decarboxylase. Enzyme Microb. Technol. 2009; 45: 58–63.
  25. 25. Kotik M, Stepanek V, Grulich M, Kyslik P, Archelas A. Access to enantiopure aromatic epoxides and diols using epoxide hydrolases derived from total biofilter DNA. J. Mol. Catal. B Enzym. 2010; 65: 41–48.
  26. 26. Martinez A, Kolvek SJ, Yip CLT, Hopke J, Brown KA, MacNeil IA, Osburne MS. Genetically modified bacterial strains and novel bacterial artificial chromosome shuttle vectors for constructing environmental libraries and detecting heterologous naturals products in multiple expression hosts. Appl. Environ. Microbiol. 2004; 70: 2452–2463.
  27. 27. Wexler M, Bond PL, Richardson DJ, Johnston AW. A wide host-range metagenomic library from a waste water treatment plants yields a novel alcohol/aldehyde dehydrogenase. Environ. Microbiol. 2005; 7: 1917–1926.
  28. 28. Suenaga H, Ohnuki T, Miyazaki K. Functional screening of a metagenomic library for genes involved in microbial degradation of aromatic compounds. Environ. Microbiol. 2007; 9: 2289–2297.
  29. 29. Mirete S, de Figueras CG, Gonzalez-Pastor JE. Novel nickel resistance genes from the rhizosphere metagenome of plants adapted to acid mine drainage. Appl. Environ. Microbiol. 2007; 73: 6001–6011.
  30. 30. Mori T, Mizuta S, Suenaga H, Miyazaki K. Metagenomic screening for bleomycin resistance genes. Appl. Environ. Microbiol. 2008; 74: 6803–6805.
  31. 31. Uchiyama T, Abe T, Ikemura T, Watanabe K. Substrate-induced gene-expression screening of environmental metagenome libraries for isolation of catabolic genes. Nat. Biotechnol. 2005; 23: 88–93.
  32. 32. Schipper C, Hornung C, Bijtenhoorn P, Quistchau M, Grond S, Streit WR. Metagenome-derived clones encoding two novel lactonase family proteins involved in biofilm inhibition in Pseudomonas aeruginosa. Appl. Environ. Microbiol. 2009; 75: 224–233.
  33. 33. Uchiyama T, Watanabe K. Substrate-induced gene expression (SIGEX) screening of metagenome libraries. Nat. Proto. 2008; 3: 1202–1212.
  34. 34. Van Lanen S, Shen B. Microbial genomics for the improvement of natural product discovery. Curr. Opin. Microbiol. 2006; 9: 252–260.
  35. 35. [35]NCBI Microbial Genomes. Available online: http://www.ncbi.nlm.nhi.gov/genomes/microbial (accessed on 10 July 2013).
  36. 36. Luo XJ, Yu HL, Xu JH. Genomic data mining: An efficient way to find new and better enzymes. Enzyme Eng. 2012; 1: 104–108.
  37. 37. Cavicchioli R, Amils R, Wagner D, McGenity T. Life and applications of extremophiles. Environ. Microbiol. 2011; 13: 1903–1907.
  38. 38. Pikuta EV, Hoover RB, Tang J. Microbial Extremophiles at the limit of life. Crit. Rev. Microbiol. 2007; 33: 183–209.
  39. 39. Atomi H, Sato T, Kanai T. Application of hyperthermophiles and their enzymes. Curr. Opin. Biotechnol. 2011; 22: 618–626.
  40. 40. De Carvalho CC. Enzymatic and whole cell catalysis: Finding new strategies for old processes. Biotechnol. Adv. 2011; 29: 75–83.
  41. 41. Hess M. Thermoacidophilic proteins for biofuels production. Trends Microbiol. 2008; 16: 414–419.
  42. 42. Chen ZW, Liu YY, Wu JF, She Q, Jiang CY, Liu SJ. Novel bacterial sulphur oxygenase reductases from bioreactors treating gold-bearing concentrates. Appl. Microbiol. Biotechnol. 2007; 74: 688–698.
  43. 43. Margesin R, Feller G, Gerday C, Rusell N. Cold-Adapted Microorganisms: Adaptation Strategies and Biotechnological Potential. In The Encyclopedia of Environmental Microbiology; Bitton, G Ed.; John Wiley and Sons: New York, NY, USA, 2002; p. 871–885.
  44. 44. Klibanov AM. Improving enzymes by using them in organic solvents. Nature 2001; 409: 241–246.
  45. 45. Gomes I, Gomes J, Steiner W. Highly thermostable amylase and pullulanase of the extreme thermophilic eubacterium Rhodothermus marinus: Production and partial characterization. Bioresour. Technol. 2003; 90: 207–214.
  46. 46. Van den Burg B. Extremophiles as a source for novel enzymes. Curr. Opin. Microbiol. 2003, 6: 213–218.
  47. 47. Shukla A, Rana A, Kumar L, Singh B, Ghosh D. Assessment of detergent activity of Streptococcus sp. AS02 protease isolated from soil of Sahastradhara, Doon Valley, Uttarakhand. Asian J. Microbiol. Biotechnol. Environ. Sci. 2009; 11: 587–591.
  48. 48. Marrs B, Delagrave S, Murphy D. Novel approaches for discovering industrial enzymes. Curr. Opin. Microbiol. 1999; 2: 241–245.
  49. 49. Singhania RR, Patel AK, Pandy A. Industrial Biotechnology: Sustainable Growth and Economic Success; Soetaert W, Vandamme EJ, Eds.; Wiley VCH Verlag GmbH: Weinheim, Germany, 2010; p. 207–226.
  50. 50. Cedrone F, Menez A, Quemeneur E. Tailoring new enzyme functions by rational redesign. Curr. Opin. Struct. Biol. 2000; 10: 405–410.
  51. 51. Beppu T. Modification of milk-clotting aspartic proteinases by recombinant DNA techniques. Ann. N. Y. Acad. Sci. 1990; 613: 14–25.
  52. 52. Van den Burg B, de Kreij A, van der Veek P, Mansfeld J, Venema G. Engineering an enzyme to resist boiling. Proc. Natl. Acad. Sci. USA 1998; 95: 2056–2060.
  53. 53. Bolon DN, Voigt CA, Mayo SL. De novo design of biocatalysts. Curr. Opin. Struct. Biol. 2002; 6: 125–129.
  54. 54. Shimaoka M, Shiftman JM, Jing H, Tagaki J, Mayo SL, Springer TA. Computational design of an integrin I domain stabilized in the open high affinity conformation. Nat. Struct. Biol. 2000; 7: 674–678.
  55. 55. Turner NJ. Directed evolution drives the next generation of biocatalysts. Nat. Chem. Biol. 2009; 5: 567–573.
  56. 56. Schmidt M, Boettcher D, Bornscheuer UT. Industrial Biotechnology: Sustainable Growth and Economic Success; Soetaert W, Vandamme EJ, Eds; Wiley VCH Verlag GmbH: Weinheim, Germany, 2010; p. 155–187.
  57. 57. Dalby PA. Strategy and success for the directed evolution of enzymes. Curr. Opin. Struct. Biol. 2011; 21: 473–480.
  58. 58. Leung DW, Chen E, Goeddel DV. A method for random mutagenesis of a defined DNA segment using a modified polymerase chain reaction. Technique 1989; 1: 11–15.
  59. 59. Reidhaar-Olson J, Bowie J, Breyer RM, Hu JC, Knight KL, Lim WA, Mossing MC, Parsell DA, Shoemaker KR, Sauer RT. Random mutagenesis of protein sequences using oligonucleotide cassettes. Methods Enzymol. 1991; 208: 564–586.
  60. 60. Taguchi S, Ozaki A, Momose H. Engineering of a cold-adapted protease by sequential random mutagenesis and a screening system. Appl. Environ. Microbiol. 1998; 64: 492–495.
  61. 61. Ness JE, del Cardayre SB, Minshull J, Stemmer WP. Molecular breeding: The natural approach to protein design. Adv. Protein Chem. 2000; 55: 261–292.
  62. 62. Morley KL, Kazlauskas RJ. Improving enzyme properties: When are closer mutations better? Trends Biotechnol. 2005; 3: 231–237.
  63. 63. Reetz MT, Kahakeaw D, Lohmer R. Addressing the numbers problem in directed evolution. ChemBioChem 2008; 9: 1797–1804.
  64. 64. Munoz E, Deem MW. Amino acid alphabet size in protein evolution experiments: Better to search a small library thoroughly or a large library sparsely? Protein Eng. Des. Sel. 2008; 1: 311–317.
  65. 65. Reetz MT, Wu S. Greatly reduced amino acid alphabets in directed evolution: Making the right choice for saturation mutagenesis at homologous enzyme positions. Chem. Commun. 2008; 5499–5501. doi:10.1039/b813388c.
  66. 66. Crameri A, Raillard SA, Bermudez E, Stemmer WP. DNA shuffling of a family of genes from diverse species accelerates directed evolution. Nature 1998; 391: 288–291.
  67. 67. Kolkman JA, Stemmer WP. Directed evolution of proteins by exon shuffling. Nat. Biotechnol. 2001; 19: 423–428.
  68. 68. Hackel BJ, Kapila A, Wittrup KD. Pico molar affinity fibronectin domain engineering utilizing loop length diversity, recursive mutagenesis, and loop shuffling. J. Mol. Biol. 2008; 381: 1238–1252.
  69. 69. Ostermeier M, Nixon AE, Shim JH, Benkovic SJ. Combinatorial protein engineering by incremental truncation. Proc. Natl. Acad. Sci. USA 1999; 96: 3562–3567.
  70. 70. Fujii R, Kitakoa M, Hayashi K. A simple and novel method of generating random insertion and deletion mutations. Nucleic Acid Res. 2006; 34: e30.
  71. 71. Ness JE, Welch M, Giver L, Bueno M, Cherry JR, Borchert TV, Stemmer WP, Minshull J. DNA shuffling of subgenomic sequences of subtilisin. Nat. Biotechnol. 1999; 17: 893–896.
  72. 72. Jaeger KE, Reetz MT. Directed evolution of enantioselective enzymes for organic chemistry. Curr. Opin. Chem. Biol. 2000; 4: 68–73.
  73. 73. Suenaga H, Mitsokua M, Ura Y, Watanabe T, Furukawa K. Directed evolution of biphenyl dioxygenase: Emergence of enhanced degradation capacity for benzene, toluene, and alkylbenzenes. J. Bacteriol. 2001; 183: 5441–5444.
  74. 74. Song JK, Rhee JS. Enhancement of stability and activity of phospholipase A(1) in organic solvents by directed evolution. Biochim. Biophys. Acta 2001; 1547: 370–378.
  75. 75. Raillard S, Krebber A, Chen Y, Ness JE, Bermudez E, Trinidad R, Fullem R, Davis C, Welch M, Seffernick J, et al. Novel enzyme activities and functional plasticity revealed by recombining highly homologous enzymes. Chem. Biol. 2001; 8: 891–898.
  76. 76. Kurtzman AL, Govindarajan S, Vahle K, Jones JT, Heinrichs V, Patten PA. Advances in directed protein evolution by recursive genetic recombination: Applications to therapeutic proteins. Curr. Opin. Biotechnol. 2001; 12: 361–370.
  77. 77. Patten PA, Howard RJ, Stemmer WP. Applications of DNA shuffling to pharmaceuticals and vaccines. Curr. Opin. Biotechnol. 1997; 8: 724–733.
  78. 78. Siehl DL, Castle LA, Gorton R, Chen YH, Bertain S, Cho HJ, Keenan R, Liu D, Lassner MW. Evolution of a microbial acetyl transferase for modification of glyphosate: A novel tolerance strategy. Pest Manag. Sci. 2005; 61: 235–240.
  79. 79. Tobin MB, Gustafsson C, Huisman GW. Directed evolution: The rational basis for irrational design. Curr. Opin. Struct. Biol. 2000; 10: 421–427.
  80. 80. Cherry JR, Fidanstel AL. Directed evolution of industrial enzymes: An update. Curr. Opin. Biotechnol. 2003; 14: 438–443.
  81. 81. Demain AL, Vaishnav P. Production of recombinant proteins by microbes and higher organisms. Biotechnol. Adv. 2009; 27: 297–306.
  82. 82. Swartz JR. Escherichia coli Recombinant DNA Technology. In Escherichia coli and Salmonella: Cellular and Molecular Biology, 2nd ed.; Neidhardt, FC Ed.; American Society of Microbiology Press: Washington, DC, USA, 1996; p. 1693–1771.
  83. 83. Barnard GC, Henderson GE, Srinivasan S, Gerngross TU. High level recombinant protein expression in Ralstonia eutropha using T7 RNA polymerase based amplification. Protein Expr. Purif. 2004; 38: 264–271.
  84. 84. Squires CH, Lucy P. Vendor voice: A new paradigm for bacterial strain engineering. BioProcess Int. 2008; 6: 22–27.
  85. 85. Romanos MA, Scorer CA, Clare JJ. Foreign gene expression in yeast: A review. Yeast 1992; 8: 423–488.
  86. 86. Higgins DR, Cregg JM. Introduction to Pichia pastoris. In Pichia Protocols; Higgins DR, Cregg J.M Eds.; Humana Press: Totowa, NJ, USA, 1998; p. 1–15.
  87. 87. Zhang AL, Luo XJ, Zhang TY, Pan YN. Recent advances on the GAP promoter-derived expression system of Pichia pastoris. Mol. Biol. Rep. 2009; 36: 1611–1619.
  88. 88. Hasslacher M, Schall M, Hayn M, Bora R, Rumbold K, Luecki J, Grieng H, Kohlwein SD, Schwab H. High-level intracellular expression of hydroxynitrile lyase from the tropical rubber tree Hevea brassiliensis in microbial hosts. Protein Expr. Purif. 1997; 11: 61–71.
  89. 89. Werton MWT, Van den Bosch TJ, Wind RD, Mooibroek H, de Wolf FA. High-yield secretion of recombinant gelatins by Pichia pastoris. Yeast 1999; 15: 1087–1096.
  90. 90. Morrow KJ. Improving protein production processes. Gen. Eng. Biotechnol. News 2007; 27: 50–54.
  91. 91. Bretthauer RK, Castellino FJ. Glycosylation of Pichia pastoris-derived proteins. Biotechnol. Appl. Biochem. 1999; 30: 193–200.
  92. 92. Romanos MA. Advances in the use of Pichia pastoris for high-level expression. Curr. Opin. Biotechnol. 1995; 6: 527–533.
  93. 93. Giuseppin M, van Eijk HM, Bes BC. Molecular regulation of methanol oxidase activity in continuous cultures of Hansenula polymorpha. Biotechnol. Bioeng. 1988; 32: 577–583.
  94. 94. Egli T, van Dijken JP, Veenhuis M, Harder W, Feichter A. Methanol metabolism in yeasts: Regulation of the synthesis of catabolite enzymes. Arch. Microbiol. 1980, 124: 115–121.
  95. 95. Shuster JR, Connelley MB. Promoter-tagged restriction enzyme-mediated insertion mutagenesis in Aspergillus niger. Mol. Gen. Genet. 1999; 262: 27–34.
  96. 96. Gouka RJ, Gerk C, Hooykaas PJJ, Bundock P, Musters W, Verrips CT, de Groot MJA. Transformation of Aspergillus awamori by Agrobacterium tumefaciens-mediated homologous recombination. Nat. Biotechnol. 1999; 6: 598–601.
  97. 97. Van der Hombergh JP, van de Vondervoort PJ, van der Heijden NC, Visser J. New protease mutants in Aspergillus niger result in strongly reduced in vitro degradation of target proteins; genetical and biochemical characterization of seven complementation groups. Curr. Genet. 1997; 28: 299–308.
  98. 98. Gouka RJ, Punt PJ, van den Hondel CAMJJ. Efficient production of secreted proteins by Aspergillus: Progress, limitations and prospects. Appl. Microbiol. Biotechnol. 1997; 47: 1–11.
  99. 99. Moralejo FJ, Cardoza RE, Gutierrez S, Martin JF. Thaumatin production in Aspergillus awamori by use of expression cassettes with strong fungal promoters and high gene dosage. Appl. Environ. Microbiol. 1999; 65: 1168–1174.
  100. 100. Gusakov AV, Salanovich TN, Antonov AI, Ustinov BB, Okunev ON, Burlingame R, Emalfarb M, Baez M, Sinitsyn AP. Design of highly efficient cellulase mixtures for enzymatic hydrolysis of cellulose. Biotechnol. Bioeng. 2007; 97: 1028–1038.
  101. 101. Verdoes JC, Punt PJ, Burlingame R, Bartels J, van Dijk R, Slump E, Meens M, Joosten R, Emalfarb M. A dedicated vector for efficient library construction and high throughput screening in the hyphal fungus Chrysosporium lucknowense. Ind. Biotechnol. 2007; 3: 48–57.
  102. 102. Wohlgemuth R. Asymmetric biocatalysis with microbial enzymes and cells. Curr. Opin. Microbiol. 2010; 13: 283–292.
  103. 103. Gong JS, Lu ZM, Li H, Shi JS, Zhou ZM, Xu ZH. Nitrilases in nitrile biocatalysis: Recent progress and forthcoming research. Microb. Cell Fact. 2012; 11: 142–160.
  104. 104. Schmidt A, Dordick JS, Hauer B, Kiener A, Wubbolts M, Witholt B. Industrial biocatalysis today and tomorrow. Nature 2001; 409: 258–268.
  105. 105. Ran N, Zhao L, Chen Z, Tao J. Recent applications of biocatalysis in developing green chemistry for chemical synthesis at the industrial scale. Green Chem. 2008; 10: 361–372.
  106. 106. Bornscheuer UT, Huisman GW, Kazlauskas RJ, Lutz S, Moore JC, Robins K. Engineering the third wave of biocatalysis. Nature 2012; 485: 185–194.
  107. 107. Sanchez S, Demain AL. Enzymes and bioconversions of industrial, pharmaceutical, and biotechnological significance. Org. Process Res. Dev. 2011; 15: 224–230.
  108. 108. BBC Research. Report BIO030 F Enzymes in Industrial Applications: Global Markets; BBC Research: Wellesley, MA, USA, 2011.
  109. 109. World Enzymes. Freedonia Group: Cleveland, OH, USA, 2011.
  110. 110. Kirk O, Borchert TV, Fuglsang CC. Industrial enzyme applications. Curr. Opin. Biotechnol. 2002; 13: 345–351.
  111. 111. Araujo R, Casal M, Cavaco-Paulo A. Application of enzymes for textiles fibers processing. Biocatal. Biotechnol. 2008; 26: 332–349.
  112. 112. Tzanov T, Calafell M, Guebitz GM, Cavaco-Paulo A. Bio-preparation of cotton fabrics. Enzyme Microb. Technol. 2001; 29: 357–362.
  113. 113. Gutiérrez A, del Rio JC, Martinez AT. Microbial and enzymatic control of pitch in the pulp and paper industry. Appl. Microbiol. Biotechnol. 2009; 82: 1005–1018.
  114. 114. Farrell RL, Hata K, Wall MB. Solving pitch problems in pulp and paper processes by the use of enzymes or fungi. Adv. Biochem. Eng. Biotechnol. 1997; 57: 197–212.
  115. 115. Rodriguez-Couto S, Toca-Herrera JL. Industrial and biotechnological applications of laccases: A review. Biotechnol. Adv. 2006; 24: 500–513.
  116. 116. Rubin EM. Genomics of cellulosic biofuels. Nature 2008; 454: 841–845.
  117. 117. Wilson BD. Cellulases and biofuels. Curr. Opin. Biotechnol. 2009; 20: 295–299.
  118. 118. Zhang YHP. What is vital (and not vital) to advance economically-competitive biofuels production. Process Biochem. 2011; 46: 2091–2110.
  119. 119. Zhang YHP, Himmmel ME, Mielenz JR. Outlook for cellulose improvement: Screening and selection strategies. Biotechnol. Adv. 2006; 24: 452–481.
  120. 120. Kumar R, Singh S, Singh OV. Bioconversion of lignocellulosic biomass: Biochemical and molecular perspectives. J. Ind. Microbiol. Biotechnol. 2008; 35: 377–391.
  121. 121. Kubicek CP, Mikus M, Schuster A, Schmoll M, Seiboth B. Metabolic engineering strategies for the improvement of cellulase production by Hypocrea jecorina. Biotechnol. Biofuels 2009; 2: 19–31.
  122. 122. Dashtban M, Qin W. Overexpression of an exotic thermotolerant β-glucosidase in Trichoderma reesei and its significant increase in cellulolytic activity and saccharification of barley straw. Microb. Cell Fact. 2012; 11: 63–78.
  123. 123. Martinez D, Berka RM, Henrissat B, Saloheimo M, Arvas M, Baker SE, Chapman J, Chertkov O, Coutinho PM, Cullen D, et al. Genome sequence analysis of the cellulolytic fungus Trichoderma reesei (syn. Hypocrea jecorina) reveals a surprisingly limited inventory of carbohydrate active enzymes. Nat. Biotechnol. 2008; 26: 553–560.
  124. 124. Kanafusa-Shinkai S, Wakayama J, Tsukamoto K, Hayashi N, Miyazaki Y, Ohmori H, Tajima K, Yokoyama H. Degradation of microcrystalline cellulose and non-pretreated plant biomass by a cell-free extracellular cellulose/hemicellulase system from the extreme thermophilic bacterium Caldicellulosiruptor bescii. J. Biosci. Bioeng. 2013; 115: 64–70.
  125. 125. Feed Enzymes: The Global Scenario; Frost & Sullivan: London, UK, 2007.
  126. 126. Choct M. Enzymes for the feed industry: Past, present and future. World Poult. Sci. 2006; 62: 5–15.
  127. 127. Selle PH, Ravindran V. Microbial phytase in poultry nutrition. Anim. Feed Sci. Technol. 2007; 135: 1–41.
  128. 128. Pariza MW, Johnson EA. Evaluating the safety of microbial enzyme preparations used in food processing: Update for a new century. Regul. Toxicol. Pharmacol. 2001; 33: 173–186.
  129. 129. Mendez C, Salas JA. Altering the glycosylation pattern of bioactive compounds. Trends Biotechnol. 2001; 19: 449–456.
  130. 130. Okanishi M, Suzuki N, Furita T. Variety of hybrid characters among recombinants obtained by interspecific protoplast fusion in streptomycetes. Biosci. Biotechnol. Biochem. 1996; 6: 1233–1238.
  131. 131. Rao MB, Tanksale AM, Ghatge MS, Deshpande VV. Molecular and biotechnological aspects of microbial proteases. Microbiol. Mol. Biol. Rev. 1998; 62: 597–635.
  132. 132. Dunn-Coleman NS, Bloebaum P, Berka R, Bodie E, Robinson N, Armstrong G, Ward M, Przetak M, Carter GL, LaCost R, et al. Commercial levels of chymosin production by Aspergillus. Biotechnol. 1991; 9: 976–981.
  133. 133. Bodie EA, Armstrong GL, Dunn-Coleman NS. Strain improvement of chymosin-producing strains of Aspergillus niger var awamori using parasexual recombination. Enzyme Microb. Technol. 1994; 16: 376–382.
  134. 134. US FDA. Center for Food Safety and Applied Nutrition. http://vm.cfsan.fda.gov (accessed on 1 July 2013).
  135. 135. Vandamme EJ, Cerdobbel A, Soetaert W. Biocatalysis on the rise. Part 1. Principles. Chem. Today 2005; 23: 57–61.
  136. 136. Collection of Information on Enzymes; Austrian Federal Environment Agency: Vienna, Austria, 2002.
  137. 137. Olempska-Beer ZS, Merker RI, Ditto MD, DiNovi MJ. Food-processing enzymes from recombinant microorganisms—A review. Regul. Toxicol. Pharmcol. 2006; 45: 144–158.
  138. 138. Tufvesson P, Lima-Ramos J, Nordblad M, Woodley JM. Guidelines and cost analysis for catalyst production in biocatalytic processes. Org. Process Res. Dev. 2011; 15: 266–274.
  139. 139. Panke S, Wubbolts M. Advances in biocatalytic synthesis of pharmaceutical intermediates. Curr. Opin. Chem. Biol. 2005; 9: 188–194.
  140. 140. Jȃckel C, Hilvert D. Biocatalysts by evolution. Curr. Opin. Biotechnol. 2010; 21: 753–759.
  141. 141. Lutz S. Reengineering enzymes. Sci. 2010; 329: 285–287.
  142. 142. Kim DY, Rha E, Choi SL, Song JJ, Hong SP, Sung MH, Lee SG. Development of bioreactor system for L-tyrosine synthesis using thermostable tyrosine phenol-lyase J. Microb. Biotechnol. 2007; 17: 116–122.
  143. 143. Volpato G, Rodrigues RC, Fernandez-Lafuente R. Use of enzymes in the production of semi-synthetic penicillins and cephalosporins: Drawbacks and perspectives. Curr. Med. Chem. 2010; 17: 3855–3873.
  144. 144. Kirchner G, Scollar MP, Klibanov A. Resolution of racemic mixtures via lipase catalysis in organic solvents. J. Am. Chem. Soc. 1995; 107: 7072–7076.
  145. 145. Zhang GW, Xu JH. New opportunities for biocatalysis: Driving the synthesis of chiral chemicals. Curr. Opin. Biotechnol. 2011; 22: 784–792.
  146. 146. Wohlgemuth R. Biocatalysis—Key to sustainable industrial chemistry. Curr. Opin. Biotechnol. 2010; 21: 713–724.
  147. 147. Liang J, Lalonde J, Borup B, Mitchell V, Mundorff E, Trinh N, Kochreckar DA, Cherat RN, Ganesh PG. Development of a biocatalytic process as an alternative to the (-)-DIP-Cl-mediated asymmetric reduction of a key intermediate of Montelukast. Org. Process Res. Dev. 2010; 14: 193–198.
  148. 148. Ma SK, Gruber J, Davis C, Newman L, Gray D, Wang A, Grate J, Huisman GW, Sheldon RA. A green-by-design biocatalytic process for atorvastatin intermediate. Green Chem. 2010; 12: 81–86.
  149. 149. Sheldom RA. E factors, green chemistry and catalysis: An odyssey. Chem. Commun. 2008; 3352–3365. doi:10.1039/b803584a.
  150. 150. Höhne M, Schitzle S, Jochens H, Robins K, Bornscheuer UT. Rational assignment of key motifs for function guides in silico enzyme identification. Nat. Chem. Biol. 2010; 6: 807–813.
  151. 151. Koszelewski D, Göritzer M, Clay D, Seisser B, Kroutil W. Synthesis of optically active amines employing recombinant ω-transaminases in E. coli cells. ChemCatChem 2010; 2: 73–77.
  152. 152. Gooding O, Voladri R, Bautista A, Hopkins T, Huisman G, Jenne S, Ma S, Mundorff EC, Savile MM, Truesdell SJ. Development of a practical biocatalytic process for (R)-2-methylpentanol. Org. Process Res. Dev. 2010; 14: 119–126.
  153. 153. Savile CK, Janey JM, Mundorff EC, Moore JC, Tam S, Jarvis WR, Colbeck JC, Krebber A, Fleits FJ, Brands J, et al. Biocatalytic asymmetric synthesis of chiral amines from ketones applied to sitagliptin manufacture. Sci. 2010; 329: 305–309.
  154. 154. Liang J, Mundorff E, Voladri R, Jenne S, Gilson L, Conway A, Krebber A, Wong J, Huisman G, Truesdell S, et al. Highly Enantioselective Reduction of a small heterocyclic ketone: Biocatalytic reduction of tetrahydrothiophene-3-one to the corresponding (R)-alcohol. Org. Process Res. Dev. 2010; 14: 185–192.
  155. 155. Ritter S. Green chemistry awards. Honors: Presidential challenge highlights innovations that promote sustainability. Chem. Eng. News 2012; 90: 11.
  156. 156. Ema T, Sayaka I, Nobuyasu O, Sakai T. Highly efficient chemoenzymatic synthesis of methyl (R)-o-chloromandelate, a key intermediate for clopidogrel, via asymmetric reduction with recombinant Escherichia coli. Adv. Synth. Catal. 2008; 350: 2039–2044.
  157. 157. Zaks A, Dodds DR. Application of biocatalysis and biotransformations to the synthesis of pharmaceuticals. Drug Discov. Today 1997; 2: 513–531.
  158. 158. Lee MY, Dordick JS. Enzyme activation for non-aqueous media. Curr. Opin. Biotechnol. 2002; 13: 376–384.
  159. 159. Almeida C, Brinyik T, Moradas-Ferreira P, Teixeira J. Continuous production of pectinase by immobilized yeast cells on spent grains Process Biochem. 2005; 40: 19 - 37.
  160. 160. Da Silva EG, Borges MF, Medina C, Piccoli RH, Schwan RF. FEMS Yeast Res. 2005; 5: 859.
  161. 161. Sorensen JF, Krag KM, Sibbesen O, Delcur J, Goesaert H, Svensson B, Tahir TA, Brufau J, Perez-Vendrell AM, Bellincamp D, D’Ovidio R, Camardella L, Giovane A, Bonnin E, Juge N. Biochim. Biophys. Acta. 2004; 1696: 275.
  162. 162. Kaur G, Kumar S, Satyanarayama T. Production, characterization and application of a thermostable polygalacturonase of a thermophilic mould Sporotrichum thermophile Apinis. Bioresour. Technol. 2004; 94: 239.
  163. 163. Taragano VM, Pilosof AMR. Enzyme Microb. Technol. 1999; 25: 411.
  164. 164. Lima AS, Alegre RM, Meirelles AJA. Carbohydr. Polym. 2000; 50: 63.
  165. 165. Uenojo M, Pastore GM. Pectinases: Aplicaçȗes Industriais e Perspectivas. Quȃmica Nova, 2007; 30: 388 - 394.
  166. 166. Paula B, Moraes IVM, Castilho CC, Gomes FS, Matta VM, Cabral LMC. Melhoria na eficiência da clarificaçio de suco de maracuj pela combinaço dos processos de microfiltraçio e enzimjtico. Boletim CEPPA, 2004; 22: 311324.
  167. 167. De Gregorio A, Mandalani G, Arena N, Nucita F, Tripodo MM, Lo Curto RB. SCP and crude pectinase production by slurry-state fermentation of lemon pulps. Bioresour. Technol., 2002; 83: 8994.
  168. 168. Blandino A, Dravillas K, Cantero D, Pandiella SS, Webb C. Process Jaeger, KE & Reetz MT. 1998. Microbial lipases form versatile tools for biotechnology. Tibtech, 2001; 16: 396 - 403.
  169. 169. Zarevöcka M, Kejak Z, Saman D, Wimmer Z, Demnerovj K. Enantioselective properties of induced lipases from Geotrichum. Enzyme and Microbial Technology, 2005; 37: 481 - 486.
  170. 170. Bradoo S, Rathi P, Saxena RK, Gupta R. Microwave-assisted rapid characterization of lipase selectivities. J. Biochem. 2002; 51: 115 - 120.
  171. 171. Tan T, Zhang M, Wang B, Ying C, Deng Li. Screening of high lipase producing Candida sp. And production of lipase by fermentation. Process Biochem. 2003; 39: 459 - 465.
  172. 172. Alkan H, Baisal Z, Uyar F, Dogru M. Production of Lipase by a Newly Isolated Bacillus coagulans Under Solid-State Fermentation Using Melon Wastes. Appl. Biochem. Biotechnol. 2007; 136: 183 - 192.
  173. 173. Kumar S, Kikon K, Upadhyay A, Kanwar SS, Gupta R. Production, purification, and characterization of lipase from thermophilic and alkaliphilic Bacillus coagulans BTS-3. Protein Expr. Pur. 2005; 41: 38 - 44.
  174. 174. Chinn Ms, Nokes SE, Strobel HJ. Influence of moisture content and cultivation duration on Clostridium thermocellum 27405 endproduct formation in solid substrate cultivation on Avicel. Biores. Technol. 2008; 99: 2664 - 2671.
  175. 175. Dominguez A, Costas M, Longo MA, Sanromjn A. A novel application of solid culture: Production of lipases by Yarrowia lipolytica. Biotechnol. Lett. 2003; 25: 1225 - 1229.
  176. 176. Kim JT, Kang SG, Woo JH, Lee JH, Jeong BC, Kim SJ. Screening and its potential application of lipolytic activity from a marine environment: Characterization of a novel esterase from Yarrowia lipolytica CL180. Applied Microbiol. Biotechnol. 2007; 74: 820 - 828.
  177. 177. Colen G, Junqueira RG, Moraes-Santos T. Isolation and screening of alkaline lipase-producing fungi from Brazilian savanna soil. World J. Microbiol. Biotechnol. 2006; 22: 881 - 885.
  178. 178. Asther M, Haon M, Roussos S, Record E, Delattre M, Meessen-Lesage L, Labat M, Asther M. Feruloyl esterase from Aspergillus niger a comparison of the production in solid state and submerged fermentation. Process Biochem. 2002; 38: 685 - 691.
  179. 179. Holsinger VH, kilgerman KH. Application of lactose in dairy foods and other foods containing lactose. Food Technol. 1997; 45: 94-95.
  180. 180. Almeida MM de, PASTORE GM. Galactooligossacarideos-Produçao e efeitos benéficos, Ciência e tecnologia de Alimentos, Campinas, SBCTA, 2001; 35: 12 - 19.
  181. 181. Mahoney RR. Lactose: Enzymatic Modification. In: Lactose, water, salts and vitamins, London, Advanced Dairy Chemistry. 1997; 77 - 125.
  182. 182. Kardel G, Furtado MM, Neto JPML. Lactase na Indŏstria de Laticnios (Parte 1). Revista do Instituto de Laticnios “Cândido Tostes”. Juiz de Fora, 1995; 50: 15 - 17.
  183. 183. Pivarnik LF, Senegal AG, Rand AG. Hydrolytic and transgalactosil activities of commercial β-galactosidase (lactase) in food processing. Advances in Food and Nutrition Research, New York, 1995; Vol. 38: p. 33.
  184. 184. Milichovâ Z, Rosenberg M. Current trends of -galactosidase application in food technology. J. Food Nut. Res. 2006; 45: 47 - 54.
  185. 185. Dillon Aldo. Celulases. In: SAID, S.; PIETRO, R. C. L. Enzimas como agents biotecnologicos. Ribeirio Preto: Legis Summa. 2004; p. 243 - 270.
  186. 186. Pretel MT. Pectic enzymes in fresh fruit processing: optimization of enzymic peeling of oranges. Process Biochem. 1997; 32: 43 - 49.
  187. 187. Chandra MS, Viswanath B, Rajaseklar Reddy B. Cellulolytic enzymes on lignocellulosic substrates in solid state fermentation by Aspergillus niger. Indian J. Microbiol. 2007; 47: 323 - 328.
  188. 188. Castro AM, Pereira JrN. Produçio, propriedades e aplicaçio de celulases na hidrölise de resaduos agroindustriais. Quimica. Nova. 2010; 33: 181 - 188.
  189. 189. Pandey A, Benjamin S, Soccol CR, Nigam P, Kriger N, Soccol VT. The realm of microbial lipases in biotechnology. Biotechnol. Appl. Biochem. 1999; 29: 119 - 131.
  190. 190. Soccol CR, Vandenberghe LPS. Overview of applied solidstate fermentation in Brazil. Biochem. Engin. J. 2003; 13: 205 - 218.
  191. 191. Aquino ACMM, Jorge JA, Terenzi HF, Polizeli MLTM. Studies on athermostable α-amylase from thermophilic fungus Scytalidium thermophilum. Appl. Microbiol. Biotechnol. 2003; 61: 323 - 328.
  192. 192. Nguyen QD, Rezessy-SZABO JM, Claeyssens M, STALS I, Hoschke A. Purification and characterization of α-mylolytic enzymes from thermophilic fungus Thermomyces lanuginosus strain ATCC 34626. Enzimes. Microbial Technol. 2002; 31: 345 - 352.
  193. 193. Giménez A, Varela P, Salvador A, Ares G, Fiszman S, Garitta L. Shelf life estimation of brown pan bread: A consumer approach. Food Quality and Preference, Barking. 2007; 18: 196 - 204.
  194. 194. Peixoto SC, jorge JA, Terenzi HF, Polizeli MLTM. Rhizopus microsporus var. rhizopodiformis: A thermotolerant fungus with potential for production of thermostable amylases. Int. Microbiol. 2003; 6: 269 - 273.
  195. 195. Asghar M, Asad MJ, Rehman S, Legge RLA. Thermostable α-amylase from a Moderately Thermophilic Bacillus subtilis Strain for Starch Processing. J. Food Engin. 2006; 38: 1599 - 1616.
  196. 196. Mitidieri S, Martinelli AHS, SCHRANK A, VAINSTEIN MH. Enzymatic detergent formulation containing amylase from Aspergillus niger: A comparative study with commercial detergent formulations. Bioresource Technology. 2006; 97: 1217–1224.
  197. 197. Smith CA, Rangarajan M, Hartley BS. D-Xylose (D-glucose) isomerase from Arthrobacter strain N.R.R.L. B3728. Purification and properties. Biochemestry Journal. 1991; 1: 255–261.
  198. 198. Tunga R, Tunga BS. Extra-cellular Amylase Production by Aspergillus oryzae Under Solid State Fermentation. Japan: International Center for Biotechnology, Osaka University. 2003; p. 12
  199. 199. Melo IS, Valadares-Inglis MC, Nass LL, Valois ACC. Recursos Genéticos & Melhoramento- Microrganismos. (1 ed), Embrapa, ISBN. 2002; 85-85771-21-6- Jaguariauna-Sio Paulo- SP.
  200. 200. Rawling ND, Barret A. Families of serine peptidases. Meth. Enzymol. 1994; 244: 18 - 61.
  201. 201. Silva CR, Delatorre AB, Martins MLL. Effect of the culture conditions on the production of an extracellular protease by thermophilic Bacillus sp. and some properties of the enzymatic activity. Braz. J. Microbiol. 2007; 38: 253 - 258.
  202. 202. Mattila-Sandholm T, Crittenden R, Mogensen G, Fondén R, Saarela M. Technological challenges for future probiotic foods. Int. Dairy J. 2002; 12: 173 - 182.
  203. 203. Bhosale SH, Rao MB, Deshpande VV. Molecular and industrial aspects of glucose isomerase. Microbiol. Rev. 1996; 60: 280 - 300.
  204. 204. Walfridsson M, Bao X, Anderlund M, Lilius G, Bulow L, Hahn-Hagerdal B. Ethanolic fermentation of xylose with Saccharomyces cerevisiae harboring the Thermus thermophilus xylA gene, which expresses an active xylose (glucose) isomerase. Appl. Environ. Microbiol. 1996; 62: 4648 - 4651.
  205. 205. Hartley BS, Hanlon N, Robin J, Rangarajan J, Ragaranjan M. Glucose isomerase: insights into protein engineering for increased thermostability. Biochimica et Biophysica acta (BBA) - Protein Structure and Molecular Enzymol. 2000; 1543: 294 - 335.
  206. 206. Cardoso MH, Jackix MNH, Menezes HC, Gonçalves EB, Marques SVB. Efeito da associaçio de pectinase, invertase e glicose isomerase na qualidade do suco de banana. Ciênc. Tecnol. Aliment. 1998; 18: 2001 - 2061.
  207. 207. Goulart AJ, Adalberto PR, Monti R. Purificaçio parcial de invertase a partir de Rhizopus sp em fermentaçio semi-sölida. Alim. Nutr. 2003; 14: 199 - 203.
  208. 208. Novaki L, Hasan SDM, Kadowaku MK, Andrade D. Produçio de invertase por fermentaçȃo em estado sölido a partir de farelo de soja. Engevista. 2010; 12: 131 - 140.
  209. 209. Jiang C, Li SX, Luo FF, Jin K, Wang Q, Hao ZY. Biochemical characterization of two novel β-glucosidase genes by metagenome expression cloning. Bioresour. Technol. 2011; 102: 3272–3278.
  210. 210. Pariza MW, Cook M. Determining the safety of enzymes used in animal feed. Regul. Toxicol. Pharmcol. 2010; 56: 332–342.

Written By

Waleed M. Abdulkhair and Mousa A. Alghuthaymi

Submitted: 03 March 2015 Reviewed: 27 July 2015 Published: 11 February 2016