Open access peer-reviewed chapter

Prospective Application of Aspergillus Species: Focus on Enzyme Production Strategies, Advances and Challenges

Written By

Mohammadhassan Gholami-Shabani, Masoomeh Shams-Ghahfarokhi, Fatemehsadat Jamzivar and Mehdi Razzaghi-Abyaneh

Submitted: 18 July 2021 Reviewed: 22 November 2021 Published: 03 January 2022

DOI: 10.5772/intechopen.101726

From the Edited Volume

Natural Food Additives

Edited by Miguel A. Prieto and Paz Otero

Chapter metrics overview

488 Chapter Downloads

View Full Metrics

Abstract

Fungal enzymes that catalyze different types of biochemical reactions play a significant role in modern industry by improving existing processes. Also, the use of enzymes to replace some traditional toxic chemical or mechanical approaches helps decrease energy demand and environmental pollution. However, enzymes must be able to compete commercially with relatively low-priced traditional approaches. Meeting economical and commercial feasibility criteria depends on a number of enzymatic properties including the specificity to the substrate, stability in industrial enzymatic reaction conditions and catalytic efficiency. Fungi used as an enzyme manufacture host should be appropriate for industrial scale fermentation. Aspergillus species are being developed as one of the best enzyme manufacture factories due to their capability to secrete high quantities of enzymes suitable for industrial applications. The industrial importance of Aspergillus species also includes the progress and commercialization of new products derived from genetically engineered modified strains. Hence, the main aim of this chapter investigation is to analyze the secreted and cellular proteins from Aspergillus species and their application in industries.

Keywords

  • filamentous fungi
  • Aspergillus species
  • fermentation
  • enzymes
  • intra- and extracellular secretion

1. Introduction

With the exponential increase in science and knowledge about biochemical processes; it would be fair to say that it is inconceivable to consider any biological process without an enzyme. They are biocatalysts that enhance the rate of reaction in numerous folds. Enzymes usually are reusable. In other words, they are not used up by the reaction and can be reused. Once an enzyme binds to a substrate and catalyzes the reaction, the enzyme is released, unchanged, and can be used for another reaction. This means that for each reaction, it is not necessary to have a ratio of 1:1 between enzymatic molecules and substrate molecules. Enzymes mostly are proteinaceous-based in nature (there are a few RNA-based enzymes) and necessary for all living organisms [1]. A significant number of them have been recognized as safe from a biotechnological perspective. Fungi, as one of the simplest organisms, are often used to produce enzymes. In addition, factors such as low energy, low cost, non-toxic and environmentally friendly nature make them popular in many industrial processes [2]. Also, the need for gentle temperature and pressure for enzymes to function enables them to become a viable alternative to hazardous chemical catalysts [3]. Enzymes are commonly used to make wine, beer, bread, cheese, vinegar, and leather and textiles. However, the pure and clean form of enzymes has found wide applications in industry only a few decades ago [4]. Enzymes produced by the fungal system are commonly used in various sectors including food, chemicals, medicine, agriculture and energy [5]. Today, due to multiple applications, the demand for different kind of enzymes in various food sectors has increased greatly [6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18], as shown in Table 1. Additionally, the manipulation of strains through recombinant DNA techniques and protein engineering technology has made it possible to meet the growing demand for enzymes [19]. Fungi are metabolically dynamic, simple to ferment and can work on an industrial scale, require simple nutrients and can be used throughout the year and are not subject to seasonal conditions [20]. The genus Aspergillus has more than 340 officially known species [21]. These fungi are characterized by an extraordinary capability to produce and secrete large amounts of proteins, metabolites, and organic acids into their growth medium [22]. Notwithstanding the existence of different pathogenic Aspergillus strains, a significant number of them have been recognized from a biotechnological perspective [23]. Characteristics such as the existence of a secretory pathway, the eventuality of genetic manipulation, and high productivity using diverse fermentative processes are beneficial and positive for the make use of Aspergillus species [23]. Enzymes produced by Aspergillus species have been broadly investigated for their potential in the formulation of commercial products [24]. Hence, the main goal of this chapter investigation is to analyze the secreted and cellular proteins from Aspergillus species and their utilization in food industries.

EnzymesFood sectorsApplications
AmylasesBrewing industryFermentation of alcohol by converting starch to sugars
Baking industryBreakdown of starch into simple sugars; thereby allowing the bread to rise and impart flavor
Dough conditioning
Generates additional sugar in the bread, which improves the taste, crust color and toasting quality
Anti-staling effect during bread making; improves the softness and shelf-life
CellulasesFruit industryFruit and vegetable juice clarification
Reducing the viscosity of nectars
Alteration of fruit sensory properties
Beverages industryConcentrating purees
Health food industryCarotenoids extraction
Edible oil extraction industryOlive oil extraction
Baking industryImprovement quality of bakery products
ChitosanasesSeafood industryThe degradation of crustacean chitinous waste
Agriculture industryBiological activities such as antifungal effect
GalactosidasesDairy industryProduction of low lactose/milk free lactose
Production of prebiotics
Prevents crystallization of lactose Improves the scoop ability and creaminess of the product
Production of ice creams, sweetened flavor and condensed milks
Improves the scoop ability and creaminess of the product
InvertasesFood sweetener marketInvert sugar production
Confectionery food industryProduction of high fructose syrup
Manufacturing of soft-centered candies
Manufacture of artificial honey
LaccaseWine industryRemoval of polyphenol, thereby providing stability to wines
Preparation of cork stoppers of wine bottles
Reduces cork taint generally imparted to aged wine bottles
Brewing industryRemoval of oxygen at the end of beer fermentation process
Prevent the formation of off-flavors (trans 2-nonenal)
Fruit industryJuice clarification
Baking industryIncrease strength, stability and reduce stickiness
Increase volume, improved crumb structure and softness of the product
LipasesFats and oils food industryProduction of mayonnaise and other emulsifiers, Triglycerides synthesis and trans-esterification of triglycerides in non-aqueous media; specially fat production
Dairy industryDevelopment of flavoring agent in milk, cheese, and butter
Hydrolysis of milk, fat, cheese ripening, and modification of butter fats
Meat industryDegumming during the refining of vegetable oil
Flavor development, meat and fish product fat removal
Baking industryFlavor development, shelf-life prolongation
NaringinasesFruit industryDebittering of citrus fruit juices
Wine industryEnhances the aroma in the wine
Production of pruning, a flavonoid
PectinasesFruit industryClarification of the fruit juices
Enhanced levels of fruit juice volume when fruit pulps treated with pectinase
Soften the peel of citrus fruits
Enhances the citrus oil extraction such as lemon oil
Beverages industryAccelerates tea fermentation
Reduces foam forming property in instant tea powders
Remove mucilaginous coat from coffee beans
Wine industryImparts stability of red wine
PhytasesBaking industryReduction of phytate content in dough & fresh breads
Shortening of formulation time without any change in pH
Increase in bread volume and an improvement in crumb texture
Softer bread crumbs were obtained
Other texture parameters like gumminess and chewiness were also decreased
ProteasesDairy industryPrevent coagulation of casein during cheese production
Flavor development
Meat industryMeat tenderization
Baking industryAssures dough uniformity
Improve dough consistency
Gluten development
Improve texture and flavor
Reduce mixing time
TannaseBrewing industryRemoval of polyphenolic compounds
Beverages industryManufacture of instant tea

Table 1.

The main applications of enzymes in different food sectors [6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18].

Advertisement

2. History and background

The use of various Aspergillus species dates back to almost a century ago, when it was discovered that the fungus Aspergillus niger was able to produce citric acid, which is a food and beverage additive and is normally extracted from citrus fruits [25]. Nowadays, A. niger is preferable to other microbes for the commercial production of citric acid because of its improved manufacturing yield. Aspergillus species are easily manipulated and can ferment different low-priced raw materials and offer high yields [26]. Thus, Aspergillus strains can be improved to generate industrial strains for application in the commercial manufacture, and mutagenesis and strain choice have been performed for such improvement. Through the application of different mutagenic agents, including various radiation for example ultraviolet, gamma and X-rays radiation, various chemicals for example, ethyl methane sulphonate and diethyl sulphonate have been commonly used to induce the mutation of Aspergillus species [26]. However, the industrial applications of Aspergillus species are not limited to the manufacture of citric acid. They have the capability to produce other organic acids such as gluconic, malic and itaconic acids, secondary metabolites and industrially important enzymes [26]. For example, A. niger has been used to manufacture different extracellular enzymes such as glucose oxidase, pectinase, α-amylase and glucoamylase, organic acids, and recombinant proteins [26]. A. oryzae is a fungus that widely plays an indispensable role in Asian food industrialized, such as soy sauce (sake and shoyu) and soybean paste (miso). Moreover, it has been applicable in the manufacture of industrial enzymes used in food processing [27]. A. terreus has attracted interest due to its ability to produce a group of secondary metabolites called statins that are used in the manufacture of cholesterol-lowering drugs [28]. A. nidulans is a capable fungal cell factory that can produce different industrial enzymes such as amylase, cellulases, cutinases, glucosidases, etc. [29]. There are many companies (such as Genencor, Novozymes, Pfizer, Amano Enzyme, Verenium, etc.) that used enzymes, proteins and secondary metabolites produced by different species of Aspergillus on a large scale to produce commercial products [30].

Advertisement

3. Enzymes production process

A general overview of the enzyme production process has been shown in Figure 1. Fermentation has two parts, upstream processes (UsP) and downstream processes (DsP). The UsP for the enzyme manufacture include the selection of Aspergillus strain, inoculum and sterilization of media [31]. For primary culture, the frozen culture of the Aspergillus strain is inoculated into the medium. The culture media are sterilized before inoculations for primary and secondary culture. Once the strain is inoculated, primary cultivation is acceptable for primary fermentation [31, 32]. The primary culture is used as inoculum during the secondary fermentation process [32]. The DsP of the fermented product depends on the nature of the secretion of an enzyme (intracellular/extracellular) [31]. If the enzymes are intracellular, the cells are first collected with the help of centrifugation and other filtration methods [32]. Disruption of cells by diverse techniques such as sonication, french press, enzymatic lysis and freezing/thawing process is necessary to get the intracellular content [31, 32]. After the cell breaking, the cell residue is excluded by filtration. In the case of extracellular proteins, the enzymes are secreted in the culture medium, so they are easier to purify. The enzymes produce using both intracellular and extracellular fermentation are subsequently concentrated using concentrators. The fermentation is generally followed by cell lysis or cell extraction then elimination of cells-residue from the culture, either using centrifugation, filtration or sedimentation. The cell-free culture is then concentrated using ultra-filtration, ammonium sulphate precipitation or liquid extraction with organic solvents [33]. About 80% of the purification processes employed thus far have used a precipitation step, with 65% of these using ammonium sulphate and 35% using ethanol, acetone or an acid (usually hydrochloric) followed by a combination of ultra-filtration system and various chromatographic techniques (20%) for example gel filtration and affinity chromatography [33]. The purification of an enzyme leads to the procedure of formulation [34]. The formulation may include methods such as drying, vacuum drying, spray drying, and freezedrying [34]. These processes make a decision how an enzymatic product will be launched in the market. It can be a mixture of two enzymes or more of them. The formulation is exclusively based upon the research and development of an industry or a lab. Formulation development is a fundamental factor not only in the early stages of enzymes/protein development, but also in the future marketing success of a promising investigational product. This is the operation of discovering novel sciences which can be used to generate novel and valuable products. The development part comes after the study and analysis and is the act of turning the discovered science into a precious product that the company can commercialize and sell. All the UsP and DsP lead to the formation of the final product.

Figure 1.

Process steps for conversion of low-cost substrate to various enzymes by Aspergillus strains. The fermentation (submerged or solid-state) is generally followed by cell-lysis/extraction, then elimination of cells-residue from the culture, either using centrifugation, filtration, sedimentation. The cell-free culture is then concentrated using ultra-filtration, ammonium sulphate precipitation or liquid extraction with organic solvents. Then followed by a combination of various chromatographic techniques for example gel filtration and affinity chromatography.

3.1 Fermentation

Both submerged (SmF) and solid-state (SSF) fermentation are used for making different enzymes by fungi [35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99]. Due to easy measurement and of control fermentation parameters, reduction fermentation time and basic ways for harvesting and refining enzymatic products, more attention has been paid to SmF [62, 63, 64, 65]. In recent time, intensive investigate on SSF has been conducted and has gained reliability due to low water consumption, low energy necessities, less contamination and high manufacture yields [35, 52]. However, Aspergillus species have shown potential for producing enzymes under both SSF and SmF (Table 2).

EnzymesAspergillus speciesSubstrateType of fermentationFermentation conditionsYield of enzymesReference
pHT (°C)DM (%)
AmylaseA. niger JGI 24Wheat branSSF4.5–9.022–40443–8174 U/mgds[34]
SmF58.06 U/ml
A. terreus NCFT4269.10Pearl milletSSF7.03047019.19 U/gds[35]
A. fumigatusMaltoseSmF6.0308–1060–130 U/mgds[36]
A. tamariiStarch or maltoseSmF4.0–10.025–426ND[37]
A. oryzae LS1Wheat branSSF6.028–3075014,249 U/gds[38]
A. oryzaeGroundnut oil cake, coconut oil cake, sesame oil cakeSSF4.532.54–5649868.12 U/gds[39]
A. flavus AUMC 11685Mandarin (Citrus reticulata) peelSmF4.0–5.528–404–526.90 U/ml[40]
A. awamori ATCC 22342Rice flourSmF6.53020.18 U/ml[41]
A. niger0.08 U/ml
A. awamori nakazawa MTCC 6652Wheat branSSF5.5354854528.4 ± 121 U/gds[42]
A. carneus SA 1326Ground millet, starch or carboxymethylcelluloseSmF5.6284ND[43]
A. sydowii IMI 502692Cassava root fiberSSF3.62ND2ND1.327 U/ml[44]
CellulasesA. niger NS-2Agricultural and kitchen waste residuesSSF3.0–8.020–50457–6717–310 U/gds[45]
A. heteromorphusWheat strawSmF5.03053.2 U/ml[46]
A. fumigatusRice straw and wheat BranSSF5.0–6.0404750.68–42.7 U/gds[47]
A. terreusLantana leavesSmF5.0257213.3 U/ ml[48]
A. nigerWheat bran, rice bran, rice husk, coir waste and saw dustSSF6.03045029.11 U/gds[49]
SmF2.04 U/ml
A. niger USM AI 1Corn steep liquorSSF7.0305703. 4 U/gds[50]
ChitosanasesA. nigerWheat branSSF6.62856541.33 U/gds[51]
Aspergillus sp. QD-2Yeast glucoseSmF4.030185.816 U/ml[52]
A. fumigates ATCC13073Vogel’s mediumSmF6.03718.80 U/mg[53]
A. oryzae SU-B2Yeast-peptone glucoseSmF5.0304352 mg/l[54]
GalactosidasesA. niger ATCC 9142Rice straw and wheat strawSSF7.0306704681 U/mg[55]
A. oryzaeRed gram and waste-wheat branSSF5.535650ND[56]
A. oryzae ATCC 20423Lactose and wheat branSmF4.8307ND[57]
A. oryzaeWheat bran and rice huskSSF5.030790146.6–386.6 U/ml[58]
InvertasesA. caespitosusWheat branSSF4–630–40370117.4 U/gds[59]
SmF19.1 U/ml
A. niger MTCC 282Orange fruit peelSSF5.03048043 U/ml[60]
A. nidulansRye flourSmF6.030330–33.6 U/ml[61]
LaccaseA. flavusStarch and yeast extractSmF7.0351417.39 U/ml[62]
A. sydowii NYKA 510Banana peel and peptone excelledSmF5.231715.1 and 2.60 g/l[63]
A. nidulansGlucose and strawSmF5–72820.052 and 0.0677 U/ml[64]
LipasesA. niger NCIM 1207Wheat bran + synthetic oil basedSSF5.530640630 U/gds[65]
A. niger AS-02Sheanut cakeSSF7.03076049.37 U/gds[66]
A. oryzae RBM4Sorghum, wheat branSmF5.53035.66 U/ml[67]
A. flavus PW2961Bran-wood flour-olive oil, bran-soy beanSSF5.0283–45037.4 U/gds[68]
A. niger MTCC 872Rice husk, cottonseed cake and red gram huskSSF6.04017528.19 U/gds[69]
NaringinasesA. niger van Tieghem MTCC 2425Citrus wastesSmF3–526–306–8426.4–545.2 U/gds[70]
A. foetidusOrange and grapefruit rindSSF5.4358ND2.58 U/ml[71]
A. aculeatus JMUdb058Yeast extract, naringinSmF6.02871.16 U/ml[72]
A. oryzae 11,250Orange peelSmF5.04542194.62 U/mgds[73]
A. niger MTCC 1344Rice bran, wheat bran, sugar cane bagasse, citrus peel, and press mudSSF4.02745058.1 U/gds[74]
A. brasiliensis 1344Cassava wasteSSF5.0275ND889.91 U/mg[75]
A. tubingensis MN589840Mildew pomelo peelSSF4.0305ND808.85 U/mg[76]
A. sojaeSoybeansSmF4.52861.5 U/mgds[77]
PectinasesA. nigerWheat branSSF4.03036368 U/gds[78]
A. carneus NRC1Orange peels and pulpsSmF5.0–5.530–55540 U/ml[79]
A. flavipes FP-500Lemon peelSmF4.2376ND[80]
A. tamariiWheat bran, banana peel, sugarcane bagasse, lemon peel, coffee pulp and orange peelSSF6.030470101.05 U/ml[81]
A. flavus CECT-2687Agroindustrial residues and polysaccharidesSmF3.5–9.03751.35–7.89 U/ml[82]
A. japonicusPolygalacturonic acid, citrus pectinsSmF4.0–5.530ND805 and 839 U/mg[83]
PhytasesA. niger CFR 335Wheat bran, rice bran, and groundnut cakeSSF2.0–7.530810–8060.6 U/gds[84]
SmF109.6 U/mL
A. ficuum SGA 01SSF810–8038 U/gds
SmF108.2 U/mL
A. niger NCIM 563Chickpea flourSmF7.0354164 U/mL[85]
A. ficuumPotato wasteSSF6.12767912.93 U/gds[86]
A. terreusRice branSmF4.5304ND[87]
ProteasesA. nigerWheat branSSF8.04083.330.21 U/mg[88]
A. oryzae LBA 01Wheat branSSF5–5.5233503961.30 U/gds[89]
A. clavatusVogel medium with glucoseSmF9.5371038 U/ml[90]
A. flavus IMI 327634Wheat branSSF7.5–9.5322636.8 U/ml[91]
TannaseA. tamariiTannic acid, gallic acid and methyl gallateSmF5.030220.6 U/ml[92]
A. ruberJamun (Syzygium cumini) leavesSSF5.53045069 U/gds[93]
A. niger MTCC 5898Cashew testaSSF3.0–8.032–353–56097.32–301.7 U/gds[94]
A. heteromorphus MTCC 8818Rosewood (Dalbergia sissoo) sawdust—a timber industry wasteSSF5.5304701.84 U/gds[95]
A. ochraceusKhanna mediumSmF5.04030.92 U/mgds[96]
A. melleusAchachairu seed powderSSF5.540260452.55 U/ml[97]
A. aculeatus DBF9Wheat bran, rice bran, saw dust, rice straw dust, sugarcane pithSSF5.0303801.32–3.95 U/gds[98]

Table 2.

Production of industrial enzymes from various Aspergillus species.

ND = not determined.

SSF: solid-state fermentation; Smf: submerged fermentation; T: incubation temperature; D: day incubation time of fermentation; M: moisture content in case of SSF.

U/ml = μmoles of fatty acids released per minute per ml of crude enzyme; U/gds = μmoles of fatty acids released per minute per gram of dry solid substrate from which enzyme has been extracted; U/mgds = μmoles of fatty acids released per minute per milligram of dry solid substrate from which enzyme has been extracted; mg/l = milligram per liter is a unit of measurement of mass concentration that shows how many milligram of a certain substance are present in one liter of a mixture; g/l = gram per liter is a unit of measurement of mass concentration that shows how many gram of a certain substance are present in one liter of a mixture.

3.2 Use of low-cost/economical substrates for enzyme cost-effective/commercial production

A large amount of agro-industrial waste like wheat straw, rice straw, rice husk, bagasse, potato waste, industrial effluents, citrus wastes, sludge, etc., are produced annually. They are rich sources of sugars, mineral elements, vitamins, fiber and different phenolic compounds, etc. Consequently, they can be used for the manufacture of commercially important products like enzyme, due to their nutritional potential (Table 2). Enzymes like amylases, celluloses, chitosanases, galactosidases, invertases, laccase, lipases, naringinases, pectinases, phytases, proteases, tannase, etc., have industrial significance and are broadly used in different industries such as pulp and paper, textile, wine and brewery, food processing, laundry and detergent, agricultural industries and bio-ethanol production [6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18]. The cost of these enzymes is a big subject faced by these industries, and efforts are going on to decrease the cost through strain improvement, better fermentation and recovery system and utilization of easily available low-cost substrates [60]. These agro-industrial wastes have exposed potential for the production of various kinds of enzymes using Aspergillus species [46, 60, 100].

3.3 Recovery purification and formulation

Enzymes are recovered from fermentation through chemical engineering operations that are broadly used to produce enzymes [101]. When the enzyme is intracellular, the cells must be broken down to release the enzyme. This can be done using mechanical methods (such as high-pressure press, grinding, or ultrasound) or non-mechanical methods (such as drying or lysis). In the case of extracellular enzymes, an early stage of isolation (centrifugation, filtration or both) is often used to eliminate residue of the cells [101]. Then the dissolved enzyme is concentrated by eliminating the water (cross-flow filtration or evaporation), resulting in an enzyme concentrate. Alternatively, a whole enzyme preparation containing inactivated cells or cell debris may be suitable where the resulting food undergoes further refinement, for example in potable alcohol production. In all cases, the prepared enzyme is free of viable fungi [101]. The concentrate is then formulated using the correct ingredients to stabilize and standardize the enzyme [101]. The raw materials used for recovery and formulation require to be of suitable purity for the future use and require to be used according to good manufacturing Practices, i.e., in the minimal quantities required to achieve the desired effect [101]. The utilize of potential allergens in the process of producing food enzymes must be addressed and, if necessary, included in the enzyme preparation. At the end of the manufacturing procedure, the last formulated enzyme generate is introduced in the market after testing to verify agreement with qualifications for contaminants (microbial and chemical) established for enzyme preparations by the Food Chemicals Codex and FAO/WHO’s JECFA. In other words, enzymes come in three diverse forms. Firstly, there is the enzymatic protein itself, which is a pure substance that is used in labs [102]. Secondly, enzymatic concentrates are products produced following fermentation or extraction [102]. They contain the enzyme produced in much smaller amounts from other substances obtained during the fermentation process, like other (secondary) enzymes or the remainder of the fermentation [102]. These enzymatic concentrates are evaluated for safety prior to being approved for marketing. Finally, there are enzymatic preparations, which are formulations containing one or more enzymatic concentrates with added stabilizers, preservatives, and diluents to stabilize enzymes and maintain activity. These formulas are sold commercially. In general, a proven quality feature of enzymes produced by microorganisms is the lack of viable cells. In addition, other specific features of microbial enzymes include their ability and significant activity under abnormal conditions, mostly temperature and pH. For example, some microbial enzymes are produced in thermophilous, acidophilic or alkalophilic forms.

Advertisement

4. Recombinant DNA (rDNA) technology

In industries, the rDNA technique will contribute to the manufacture of chemicals of commercial importance, to the advancement of existing fermentation processes and protein/enzyme production from waste materials. For this purpose, more effective strains of microorganisms can be developed. Thus, the technology of rDNA has many useful applications in crop betterment, medication and industry.

The rDNA in microorganisms occurs through three different parasexual processes namely conjugation, transduction, and transformation [103]. Internal genetic rearrangements can also occur via translocatable DNA segments (insertion sequences or transposons) [103]. Conjugation implies DNA transfer through cell-to-cell contact. Transduction occurs from the host cell to the recipient cell through bacteriophaging mediation. Transformation involves the absorption and expression of bare DNA by the appropriate cells. Competence occurs naturally but can also be induced by changes in the physical and chemical environment. In the laboratory, it can be induced by cold calcium chloride treatment, protoplasting, electroporation and heat shock [103]. After 1980, there was a heightened interest in the application of genetic recombination to the production of important microbial products such as antibiotics. The use of rDNA technology has made it possible to produce new enzymes appropriate for specific food-processing conditions [104]. Various substantial enzymes (lipases, pectinases, cellulases, amylases, etc.) are useable for the specific manufactures because of their exclusive roles and utilization in food and feed industries. Manufacture of microbial strains is another vast accomplishment that became feasible with the assist of rDNA technology. Various microbial strains have been expanded to manufacture superior enzymes by particular engineering. Specific strains of fungi have been modified in order that their capability of manufacturing toxic and hazardous materials could be decreased. Wide ranges of recombinant proteins/enzymes have been expressed in different species of fungi to be used as enzymes in industries [105]. Aspergillus species are regarded as promising candidates for developing large-scale heterologous protein production platforms. However, production yields of heterologous proteins are usually significantly lower than those detected for native proteins. Failure to achieve the favorable protein amounts in Aspergillus cultures can be ascribed to limitations associated to transcription, translation, and the post-translation processing and modifications during protein manufacture. Furthermore, bottlenecks in the fungal secretion system and the problem of extracellular breakdown further impede the effective production of foreign proteins in Aspergillus species [106].

Several Aspergillus species, in particular A. niger and A. oryzae, are widely used as protein-production hosts in a variety of biotechnology applications. To improve the expression and secretion of recombinant proteins from these filamentous fungi, several novel genetic engineering strategies have been developed over the past few years. Yu et al. reported construction and application of a novel genetically engineered A. oryzae for expressing proteases [107]. Their results showed different degrees of improvement in the protease activity when compared with wild-type A. oryzae. A major improvement in the polypeptide yield was achieved when these strains were used in soybean meal fermentation. The polypeptide conversion rate of transformants A. oryzae reached 35.9%, which was approximately twofold higher than that exhibited by wild-type A. oryzae [107]. Amino acid content analysis showed that the essential amino acid content and amino acid composition of the fermentation product significantly improved when engineered A. oryzae strains were used for soybean meal fermentation [107]. Prathumpai et al. reported lipase production by recombinant strains of A. niger expressing a lipase-encoding gene from Thermomyces lanuginosus [108]. Their heterologous lipase was expressed using the TaKa amylase (the A. oryzae amylase is known as Taka-amylase) promoter from A. oryzae. Their results showed the transformants strain was found to be the best producer. Record et al. reported expression of the Pycnoporus cinnabarinus laccase gene in A. niger. A. niger preprosequence allowed an 80-fold increase in laccase production [109]. Bohlin et al. reported heterologous expression of Trametes versicolor laccase in A. niger. Their results showed recombinant laccase from A. niger harboring the lcc2 cDNA was purified to homogeneity and it was found to be a 70 kDa homogeneous enzyme with biochemical and catalytic properties similar to those of native T. versicolor laccase [110]. Dragosits et al. reported recombination for the production and purification of two previously uncharacterized β-galactosidases from A. nidulans as well as one β-galactosidase from A. niger. Their results showed that A. niger and A. nidulans are suitable for various glycobiological and biotechnological applications [111].

Advertisement

5. Enzymes production by Aspergillus species and their application

5.1 Amylases

The Aspergillus species produce a wide range of intra- and extracellular enzymes, and amylases are one of the main enzymes used in industrial markets (Table 2). Amylases are broadly employed in food manufacturing such as brewing, baking, preparation of digestive aids, manufacture of cakes, fruit juices and starch syrups [112]. They have been extensively used in the baking industry. Amylases can be added to the dough of bread which degrade the starch in the flour into smaller dextrins, and these smaller dextrins are subsequently fermented by the yeast. The addition of these enzymes to the dough results in improving the rate of fermentation and the reduction of the viscosity of dough, resulting in progresses in the volume and texture of the product. Moreover, they produce additional sugar in the dough that improves the flavor, crust color and toasting qualities of the bread [112]. In addition to producing fermentable compounds, they also have an antistaling effect in baking bread which advances the retention of the softness of bakery products, increasing the shelf life of these products. These enzymes are also used for the clarification of fruit and veggie juices or beer or for the pretreatment of animal feed to improve the digestibility of fiber [6, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44].

5.2 Cellulases

Cellulases are groups of enzymes that are secreted using a wide range of Aspergillus species (Table 2). According to current enzyme market information, the main areas of the industry where cellulases enzymes are progressively being applied are textile, pulp and paper, laundry detergents, healthcare, food and beverages. Their extensive application in coffee and tea processing, fruit juice production and wine making is associated to food and beverage segment. In other industrial applications, it is widely used to generate laundry detergents and cleaning and washing agents. These enzymes are also being highly known as an effective alternative to available various antibiotics and antifungal. Lemnaru Popa et al. reported bacterial cellulose in skin wound treatment is very attractive due to its unique characteristics. Their results confirmed the drugs’ presence in the bacterial-cellulose dressing’s structure as well as the antimicrobial efficiency against Staphylococcus aureus and Escherichia coli [113]. In another study, Limón et al. reported antifungal and chitinase specific activities of Trichoderma harzianum CECT 2413 by addition of a cellulose binding domain was increased [114]. Consequently, the potential of these enzymes is a wonderful trend to fight against antibiotic-resistant bacteria which may overcome problems in the healthcare sector [7]. Furthermore, in the food market these enzymes have many applications. Fruit and veggie juice clarification, carotenoid extraction, reducing the viscosity of nectars, alteration of fruit sensory properties, concentrating purees, olive oil extraction, and the quality betterment of bakery products are among the different processes in food industry and biotechnology where cellulases are exploited worldwide. Consequently, these enzymes can cause a huge economic impact. However, there are some significant bottlenecks of employing this enzyme in the industry such as loss of enzyme activity, immobilization of enzyme in undesired conformation and subsequent loss of activity, cost of carrier and additional preparation materials and methods, as well as laborious training strategies, mass transfer limitations, laborious and time-consuming immobilization processes [7, 46, 47, 48, 49, 50].

5.3 Chitosanases

Chitosanases can degrade chitin and it can generate using various Aspergillus species (Table 2). These enzymes play a significant role in nutrition and defense. Due to its antimicrobial activity against a wide range of filamentous fungi, yeasts and bacteria, they have attracted much attention as a potential food preservative [8]. These enzymes potentially used in preparation of N-acetyl d-glucosamine and chitooligosaccharides. Chitooligosaccharides and N-acetyl glucosamine are currently of enormous relevance to pharmaceutical, nutraceutical, cosmetics, food, and agriculture industries due to their wide range of biological activities, which include antimicrobial, antitumor, antioxidant, anticoagulant, wound healing, immunoregulatory, and hypocholesterolemic effects. Furthermore, the control of pathogenic fungi in agriculture could be done using chitonases. The degradation of crustacean chitinous waste in seafood industry could be enhanced using them. It is also valuable for the preparation of single-cell protein and also for the isolation of protoplasts from fungi and yeast, etc. [8, 51, 52, 53, 54, 55].

5.4 Galactosidases

Galactosidases can be generated by various Aspergillus species and being used in food industry for hydrolysis of lactose in milk and milk by-products (Table 2). They have attracted much attention in view of lactose intolerance in human population and due to importance of milk in human diet. These enzymes can hydrolyze galactopyranosides, that is, lactose, and form a wide range of trans-galactosylation products or galactooligosaccharides capable of providing some health benefits as prebiotics. Furthermore, these enzymes also find applications in production of lactose based sweeteners from high lactose containing effluents of cheese manufacturing industries. At present, these enzymes are mainly obtained from various A. niger strains [9, 56, 57, 58, 59].

5.5 Invertase

Invertases are generated using plants, bees, and microorganisms [10], but the filamentous fungi belonging to the Aspergillus species are the most prominent organisms used for invertases manufacture (Table 2). They are used to hydrolyze sucrose and polysaccharides, which have the same type of β-d-fructofuranosyl bond, to obtain fructose and glucose as final products [10]. They are significant in the food biotechnology, especially in confectionery and candy manufacturers, as a catalytic factor in obtaining an artificial sweetener. Hence, they are used for the construction of formulations that prevent crystallization of certain sweet preparations, using in the chocolate industry markets. In some syrup, it is also used to growth its sweetening properties such as producing of soft caramel fillings. Honey is the most common form of this inverted sugar that is a supersaturated mixture of fructose and glucose. Furthermore, they are able to synthesize fructooligosaccharides through fructotransferase where sucrose is presented in high concentrations. The fructooligosaccharides are related to improve human health [9, 60, 61, 62].

5.6 Laccases

Fungi such as Aspergillus species have been used to generate Laccases (Table 2). Laccases are a broadly studied enzyme because of its potential use in some industries areas such as textile, paper and pulp [16]. They can be used in bioremediation, beverage processing (such as wine, fruit juice and beer), sugar beet pectin gelation, ascorbic acid determination, baking, and as biosensor and to progress food sensory parameters [17]. They can increase the productivity, efficiency and quality of food goods without costly investment, and this is an advantage. Wide areas of the food industry that benefit from processing with these enzymes include juice processing, wine stabilization, baking industry, and bioremediation of waste water [16, 17, 63, 64, 65].

5.7 Lipases

Lipases are one of the most important biocatalysts that perform different reactions in aqueous and non-aqueous media [11]. These enzymes usually catalyze the hydrolysis of long-chain triglycerides. They can operate on a diversity of substrates counting natural oils, artificial triglycerides, and esters of fatty acids. They are manufactured using animals, plants, and microorganisms. Presently, fungal lipases are achieving much consciousness with the rapid development of enzyme technology. Fungi-produced lipases have played an interesting role in industrial biotechnology because many of them are stable in a wide range of pH, high temperatures, and organic solvents. They are signifying one of the most important groups of biocatalysts for industrial applications. Aspergillus species is an important chief manufacturer of lipases (Table 2). They are capable to modify the characteristics of lipids by altering the location of fatty acid chains in the glyceride and exchange one or more fatty acid with new ones. Cocoa butter is a crucial ingredient in chocolate that has a high butterfat value because it contains palmitic and stearic acids and has a melting point of almost 37°C. Melting of cocoa butter in the mouth creates a positive favorable cooling sensation in a product such as chocolate [11]. Lipases are used ex-situ to create taste and to change the formation using inter- or transesterification that obtain products of improved nutritional value, or appropriate for feeding [11]. They are also been used in food to modify taste using manufacture of esters of short-chain fatty acids and alcohol, which are known flavor and fragrance compounds. Lipases facilitate the elimination of fat from meat and fish products. They are used for the manufacture of maltose and lactose like sugar fatty acid esters. They have many applications in food and flavor industry and in the production of ice cream [11, 66, 67, 68, 69, 70].

5.8 Naringinases

Various microbial sources of naringinases have been reported worldwide by various investigators [12]. Production of naringinases has been very well studied in fungal sources. Among fungi, Aspergillus species (especially A. niger) have been reported as major producers of naringinases (Table 2). Naringin is a flavonoid naturally which present in citrus fruits (such as oranges, lemon, and grapefruit). Flavonoids may cause interference during the citrus fruit juice processing and are responsible for the bitter taste. The processing of citrus fruit juice has faced formidable problems in terms of bitterness and delayed onset of bitterness. The bitterness affects its consumer acceptability. In citrus juices two compounds namely flavonoids and limonoids are established responsible for bitterness. The bitterness in grapefruit juice can be reduced by using enzymes such as naringinases which hydrolyzes naringin into relatively non-bitter compounds. They are an enzyme which catalyzes the hydrolysis of naringin into pruning, rhamnose, glucose and then into naringenin, which is non-bitter and tasteless. This enzyme has two different enzyme activities (due to two different subunits). One is α-rhamonosidase which acts on naringin to release prunin and α-rhamnose. Second is β-D-glucosidase which acts on prunin to release naringenin and β-D-glucose. There are only few reports on the commercial manufacture of this enzyme. These enzymes have been used for elimination of bitter flavor in citrus fruit juice (due to naringin) [12, 71, 72, 73, 74, 75, 76, 77, 78].

5.9 Pectinases

Pectinases have the most important role in fruit and veggie juice marketing by breaking the pectin (polysaccharide) structure present in the cell wall of plants. They are mainly manufactured using microorganisms and plants. Among microorganisms, fungi (especially Aspergillus species) have a high ability to secrete them (Table 2). Pectinases are a class of enzymes that catalyzes the degradation of pectic substances [13]. They have broad applications in food and agricultural industries. Pectinases can be used and commercially applied for the processing of fruit juices and wines. In food industry, these enzymes are used especially for clarification, maceration, extraction and stabilization of fruit juices. They also enhance fruit juice yield and involved in fermentation of coffee, cocoa and tea and preparation of jams and jellies. They are used in oil extraction from plant but olive oil extraction is the most common. These enzymes are added for easy oil extraction during grinding of olives. They have capability to reduce feed viscosities which directly increase the nutrients absorption capability of animals. These nutrients are released from fibers using hydrolysis process and it also reduces animal defecation [13, 79, 80, 81, 82, 83, 84].

5.10 Phytases

Phytases have a role in food and feed industry. They are synthesized using fungi, mainly from Aspergillus species (Table 2). They can reduce the antinutritional effect of phytate and improve the digestibility of phosphorous, calcium, amino acids and energy, as well as minimize the negative impact of inorganic phosphorous excretion on the environment [14]. The benefits of using phytase in animal feed are well recognized [14]. Especially, phytate-degrading enzymes from Aspergillus species offer industrial and economical feasibility for their manufacture and application. Phytates have been considered as a threat in human diet due to its antinutrient behavior, which is known as strong chelator of divalent minerals such as Ca2+, Mg2+, Zn2+ and Fe2+. There is a high potential for the employ of phytases in processing and developed of food for human consumption. Investigation in this aspect focuses on the progress of the nutritional value of plant-based food and feed as well as on the technical improvement of food processing. A diet rich in phytate leads to a significantly decreased absorption of dietary minerals and the dephosphorylation of phytate during food processing results in the formation of only partially phosphrylated myo-inositol phosphate esters with a lower capability to impair with the intestinal uptake of dietary minerals. Individual myo-inositol phosphate esters have been shown to have some important physiological functions in man. Consequently, phytases may find application in food processing to generate functional foods. Also, phytases were showing to be an excellent bread making improver [14, 85, 86, 87, 88].

5.11 Proteases

Proteases are produce in all organisms, such as plants, animals, and microbes [15]. The peptide bond present in the polypeptide chain is hydrolyzed by proteases. They are degradative enzymes and demonstrate specificity and selectivity in protein modification. They are one of the most important industrial enzymes and their international market is significantly growing annually. Of the 60% of enzymes marketed worldwide, proteases account for 20%. Proteases have been successfully produced by researchers from various microbial sources [15]. Reports suggest that two-thirds of the world’s commercial proteases are produced by microorganisms because of their greater yield, reduction in time consumption, reduction in space requirement, lofty genetic manipulation, and cost-effectiveness, which have made them suitable for biotechnological application in the market. Among microbes, Aspergillus species have been extensively studied for protease manufacture in a large scale (Table 2). Proteases are used on a large commercial scale in the production of baked goods, bread, crackers and waffles. Proteases produced by Asperjillus oryzae have been used to modify wheat gluten using limited proteolysis. They are also used in the dairy industry. Their main application in the dairy industry is in cheese manufacture. In cheese manufacture, the primary function of proteases is to hydrolyze the certain peptide bond to generate para-k-casein and macro peptides [15, 89, 90, 91, 92].

5.12 Tannase

Tannases are a group of enzymes that are employed in multitudinous industries such as food, brewing, and pharmaceutical [18]. They have an expansive range of scattering and are generated form animals, plants, and microbial sources. However, manufactured tannins of microbial origin are favored over other sources for industrial utilization. Fungi such as Aspergillus species have been used to generate tannases (Table 2). They operate upon hydrolyzable tannins by cracking the ester and depside bonds so as to release glucose and gallic acid. One of the most significant commercial applications of tannases is gallic acid manufacture. Besides that, they are widely employed in the food industry, especially in the manufacture of instant tea, where it increases the extractability and cold water solubility of key compounds [18]. Another significant applying of tannase is the elimination of haze formation and unflavored phenolic compounds from beer and wine [18]. Moreover, the quality of fruit juices can be enhanced using tannases enzymes. The turbidity and bitterness of fruit and veggie juices are minimized by using these enzymes. While using agro-industrial residues as animal feed, tannins-rich biomass are considered as anti-nutritional factors. De-tannification of feed using tannases enzymes treatment can extensively progress the quality of animal feed [18, 93, 94, 95, 96, 97, 98, 99].

Advertisement

6. Future perspectives and conclusions

At present, enzymes have become an important part of various industries [1]. The total enzymes market size in the worldwide is anticipated to reach over $13–14 billion by 2027. However, the manufacture of different enzymes has always been a challenge. They are produced from plants, animals and microorganisms [4]. Microbial enzyme production is generally accepted and occupies approximately 85–90% of the global enzyme market. In microbial enzyme manufacture, the localization of enzyme is a major aspect to be considered. If an enzyme is extracellular, the cost of downstream processing is reduced [100]. However, when it comes to intracellular enzymes, it becomes an expensive process to purify such enzymes. The degree of purification also varies according to the use of enzymes. Among microorganisms, fungi are especially used for the manufacture of various enzymes in a wide range [5]. Out of about 260 commercial enzymes, 60% are sourced from about 25 fungal genera [115]. Fungi can produce a number of industrial enzymes that are used in variety different industrial processes. Owing to their ability to use low-value substrates, their ability to handle and their ability to produce high enzymatic titres, fungi are the subject of extensive studies for industrial enzymes. Enzymes of fungal origin (especially from Aspergillus species) are employed in a wide range of industrial applications. The advantages of using Aspergillus species in the industry are multiple and they are a rich source of enzymes with valuable properties in industrial processes. A good example is their outstanding high stability, as they naturally evolved to work in a relatively harsh extracellular environment. They can use common “types of waste” as a source of carbon and energy and release the valuable enzymatic product from their cells into the medium. The cost of enzyme manufacture by Aspergillus species is another attractive parameter and an essential prerequisite for the exploitation of these catalysts in large-scale industrial settings. In addition, Aspergillus species will be able to produce strong, multifunctional (chimeric) enzymes using recombinant DNA technology, high-efficiency screening of the latest, metagenomic screening, silicon enzyme engineering and site-to-site mutagenesis to meet future needs.

Advertisement

Acknowledgments

Research reported in this publication was supported by Elite Researcher Grant Committee under award numbers [958634 and 963646] from the National Institute for Medical Research Development (NIMAD), Tehran, Iran to MRA.

Advertisement

Conflict of interest

Authors declare no conflict of interest.

References

  1. 1. Patel AK, Singhania RR, Pandey A. Production, purification, and application of microbial enzymes. In: Brahmachari G, editor. Biotechnology of Microbial Enzymes. San Diego, CA: Elsevier; 2017. pp. 13-41. DOI: 10.1016/B978-0-12-803725-6.00002-9
  2. 2. Meyer V, Basenko EY, Benz JP, Braus GH, Caddick MX, Csukai M, et al. Growing a circular economy with fungal biotechnology: A white paper. Fungal Biology and Biotechnology. 2020;7:1-23. DOI: 10.1186/s40694-020-00095-z
  3. 3. Jun H, Kieselbach T, Jönsson LJ. Enzyme production by filamentous fungi: Analysis of the secretome of Trichoderma reesei grown on unconventional carbon source. Microbial Cell Factories. 2011;10:68. DOI: 10.1186/1475-2859-10-68
  4. 4. Singh R, Kumar M, Mittal A, Mehta PK. Microbial enzymes: Industrial progress in 21st century. 3 Biotech. 2016;6:174. DOI: 10.1007/s13205-016-0485-8
  5. 5. Robinson PK. Enzymes: Principles and biotechnological applications. Essays in Biochemistry. 2015;59:1-41. DOI: 10.1042/bse0590001
  6. 6. de Souza PM, de Oliveira Magalhães P. Application of microbial α-amylase in industry—A review. Brazilian Journal of Microbiology. 2010;41(4):850-861. DOI: 10.1590/S1517-83822010000400004
  7. 7. Jayasekara S, Ratnayake R. Microbial cellulases: An overview and applications. In: Pascual AR, Martín MEE, editors. Cellulose. London, England: IntechOpen; 2019. DOI: 10.5772/intechopen.84531
  8. 8. Muzaddadi AU, Devatkal S, Oberoi HS. Seafood enzymes and their application in food processing. In: Dhillon GS, Kaur S, editors. Agro-Industrial Wastes as Feedstock for Enzyme Production. San Diego, CA: Elsevier; 2016. pp. 201-232. DOI: 10.1016/B978-0-12-802392-1.00009-5
  9. 9. Xavier JR, Ramana KV, Sharma RK. β-Galactosidase: Biotechnological applications in food processing. Journal of Food Biochemistry. 2018;42:e12564. DOI: 10.1111/jfbc.12564
  10. 10. Veana F, Flores-Gallegos AC, Gonzalez-Montemayor AM, Michel-Michel M, Lopez-Lopez L, Aguilar-Zarate P, et al. Invertase: An enzyme with importance in confectionery food industry. In: Kuddus M, editor. Enzymes in Food Technology. Singapore: Springer Singapore; 2018. pp. 187-212. DOI: 10.1007/978-981-13-1933-4_10
  11. 11. Mehta A, Guleria S, Sharma R, Gupta R. The lipases and their applications with emphasis on food industry. In: Ray RC, editor. Microbial Biotechnology in Food and Health. San Diego, CA: Elsevier; 2021. pp. 143-164. DOI: 10.1016/B978-0-12-819813-1.00006-2
  12. 12. Yadav M. Naringinase: Microbial sources, production and applications in food processing industry. Journal of Microbiology, Biotechnology and Food Sciences. 2018;8:717-720. DOI: 10.15414/jmbfs.2018.8.1.717-720
  13. 13. Kubra KT, Ali S, Walait M, Sundus H. Potential applications of pectinases in food, agricultural and environmental sectors. Journal of Pharmaceutical, Chemical and Biological Sciences. 2018;6:23-34
  14. 14. Dahiya S. Role of phytate and phytases in human nutrition. International Journal of Food Sciences and Nutrition. 2016;1:39-42
  15. 15. Razzaq A, Shamsi S, Ali A, Ali Q, Sajjad M, Malik A, et al. Microbial proteases applications. Frontiers in Bioengineering and Biotechnology. 2019;7:110. DOI: 10.3389/fbioe.2019.00110
  16. 16. Minussi RC, Pastore GM, Durán N. Potential applications of laccase in the food industry. Trends in Food Science and Technology. 2002;13:205-216. DOI: 10.1016/S0924-2244(02)00155-3
  17. 17. Brijwani K, Rigdon A, Vadlani PV. Fungal laccases: Production, function, and applications in food processing. Enzyme Research. 2010;2010:149748. DOI: 10.4061/2010/149748
  18. 18. Kumar SS, Sreekumar R, Sabu A. Tannase and its applications in food processing. In: Parameswaran B, Varjani S, Raveendran S, editors. Green Bio-Processes: Energy, Environment, and Sustainability. Singapore: Springer Singapore; 2019. pp. 357-381. DOI: 10.1007/978-981-13-3263-0_19
  19. 19. Wang Q, Zhong C, Xiao H. Genetic engineering of filamentous fungi for efficient protein expression and secretion. Frontiers in Bioengineering and Biotechnology. 2020;8:293. DOI: 10.3389/fbioe.2020.00293
  20. 20. Hyde KD, Xu J, Rapior S, Jeewon R, Lumyong S, Niego AGT, et al. The amazing potential of fungi: 50 ways we can exploit fungi industrially. Fungal Diversity. 2019;97:1-136. DOI: 10.1007/s13225-019-00430-9
  21. 21. Park HS, Jun SC, Han KH, Hong SB, Yu JH. Diversity, application, and synthetic biology of industrially important Aspergillus fungi. Advances in Applied Microbiology. 2017;100:161-202. DOI: 10.1016/bs.aambs.2017.03.001
  22. 22. El-Enshasy HA. Filamentous fungal cultures—Process characteristics, products, and applications. In: Yang ST, editor. Bioprocessing for Value-Added Products from Renewable Resources. B.V., The Netherlands: Elsevier; 2007. p. 225–261. Doi:10.1016/B978-044452114-9/50010-4
  23. 23. Luis HSG, Patricia N d CS. Aspergillus biotechnology: An overview on the production of hydrolases and secondary metabolites. Current Biotechnology. 2017;6:283-294. DOI: 10.2174/2211550105666160607071134
  24. 24. Ward OP, Qin WM, Dhanjoon J, Ye J, Singh A. Physiology and biotechnology of Aspergillus. Advances in Applied Microbiology. 2005;58C:1-75. DOI: 10.1016/S0065-2164(05)58001-8
  25. 25. Max B, Salgado JM, Rodríguez N, Cortés S, Converti A, Domínguez JM. Biotechnological production of citric acid. Brazilian Journal of Microbiology. 2010;41:862-875. DOI: 10.1590/S1517-83822010000400005
  26. 26. Show PL, Oladele KO, Siew QY, Aziz Zakry FA, Lan JC-W, Ling TC. Overview of citric acid production from Aspergillus niger. Frontiers in Life Science. 2015;8:271-283. DOI: 10.1080/21553769.2015.1033653
  27. 27. Gomi K. Aspergillus: Aspergillus oryzae. In: Encyclopedia of Food Microbiology. 2nd ed. USA: Elsevier Academic Press; 2014. pp. 92-96. DOI: 10.1016/B978-0-12-384730-0.00011-2
  28. 28. Manzoni M, Rollini M. Biosynthesis and biotechnological production of statins by filamentous fungi and application of these cholesterol-lowering drugs. Applied Microbiology and Biotechnology. 2002;58:555-564. DOI: 10.1007/s00253-002-0932-9
  29. 29. Kumar A. Aspergillus nidulans: A potential resource of the production of the native and heterologous enzymes for industrial applications. International Journal of Microbiology. 2020;2020:8894215. DOI: 10.1155/2020/8894215
  30. 30. Cairns TC, Nai C, Meyer V. How a fungus shapes biotechnology: 100 years of Aspergillus niger research. Fungal Biology and Biotechnology. 2018;5:13. DOI: 10.1186/s40694-018-0054-5
  31. 31. Arnau J, Yaver D, Hjort CM. Strategies and challenges for the development of industrial enzymes using fungal cell factories. In: Grand Challenges in Fungal Biotechnology. Cham: Springer International Publishing; 2020. pp. 179-210. DOI: 10.1007/978-3-030-29541-7_7
  32. 32. Berenjian A, editor. Essentials in Fermentation Technology. 1st ed. Cham, Switzerland: Springer Nature; 2019. DOI: 10.1007/978-3-030-16230-6
  33. 33. Tan CH, Show PL, Ooi CW, Ng EP, Lan JC, Ling TC. Novel lipase purification methods–a review of the latest developments. Biotechnology Journal. 2015;10:31-44
  34. 34. Bhatia S. Introduction to Pharmaceutical Biotechnology. Volume 2: Enzymes, Proteins and Bioinformatics. London, England: Institute of Physics Publishing; 2018. pp. 2-42. DOI: 10.1088/978-0-7503-1302-5
  35. 35. Varalakshmi KN, Kumudini BS, Nandini BN, Solomon J, Suhas R, Mahesh B, et al. Production and characterization of alpha-amylase from Aspergillus niger JGI 24 isolated in Bangalore. Polish Journal of Microbiology. 2009;58:29-36
  36. 36. Sethi BK, Jana A, Nanda PK, DasMohapatra PK, Sahoo SL, Patra JK. Production of α-amylase by Aspergillus terreus NCFT 4269.10 using pearl millet and its structural characterization. Frontiers in Plant Science. 2016;7:639. DOI: 10.3389/fpls.2016.00639
  37. 37. Goto CE, Barbosa EP, Kistner LCL, Moreira FG, Lenartovicz V, Peralta RM. Production of amylase by Aspergillus fumigatus utilizing α-methyl-D-glycoside, a synthetic analogue of maltose, as substrate. FEMS Microbiology Letters. 1998;167(2):139-143. DOI: 10.1111/j.1574-6968.1998.tb13219.x
  38. 38. Moreira FG, Lima FA de, Pedrinho SRF, Lenartovicz V, Souza CGM de, Peralta RM. Production of amylases by Aspergillus tamarii. Revista de Microbiologia 1999;30:157–162. DOI:10.1590/S0001-37141999000200014
  39. 39. Fattah Farid MA, Halim Shata HMA. Amylase production from Aspergillus oryzae LS1 by solid-state fermentation and its use for the hydrolysis of wheat flour. Iranian Journal of Biotechnology. 2011;9:267-274
  40. 40. Balakrishnan M, Jeevarathinam G, Kumar SKS, Muniraj I, Uthandi S. Optimization and scale-up of α-amylase production by Aspergillus oryzae using solid-state fermentation of edible oil cakes. BMC Biotechnology. 2021;21:33. DOI: 10.1186/s12896-021-00686-7
  41. 41. Ali EH, El-Nagdy MA, Al-Garni SM, Ahmed MS, Rawaa AM. Enhancement of alpha amylase production by Aspergillus flavus AUMC 11685 on mandarin (Citrus reticulata) peel using submerged fermentation. European Journal of Biological Research. 2017;7:154-164. DOI: 10.5281/zenodo.818271
  42. 42. Pestana F, Castillo FJ. Glucoamylase production by Aspergillus awamori on rice flour medium and partial characterization of the enzyme. World Journal of Microbiology and Biotechnology. 1985;1:225-237. DOI: 10.1007/BF01742317
  43. 43. Negi S, Banerjee R. Optimization of culture parameters to enhance production of amylase and protease from Aspergillus awamori in a single fermentation. African Journal of Biochemistry Research. 2010;4:73-80. DOI: 10.5897/AJBR.9000178
  44. 44. Shambe T, Ejembi O. Production of amylase and cellulase: Degradation of starch and carboxymethylcellulose by extracellular enzymes from four fungal species. Enzyme and Microbial Technology. 1987;9:308-312. DOI: 10.1016/0141-0229(87)90010-X
  45. 45. Adegoke SA, Odibo FJC. Production, purification and characterization of α-amylase of Aspergillus sydowii IMI 502692. Plant Cell Biotechnology and Molecular Biology. 2019;20:1050-1058
  46. 46. Bansal N, Tewari R, Soni R, Soni SK. Production of cellulases from Aspergillus niger NS-2 in solid state fermentation on agricultural and kitchen waste residues. Waste Management. 2012;32:1341-1346. DOI: 10.1016/j.wasman.2012.03.006
  47. 47. Singh A, Singh N, Bishnoi NR. Production of cellulases by Aspergillus heteromorphus from wheat straw under submerged fermentation. International Journal of Civil Engineering. 2009;1:23-26
  48. 48. Sherief AA, El-Tanash AB, Atia N. Cellulase production by Aspergillus fumigatus grown on mixed substrate of rice straw and wheat bran. Research Journal of Microbiology. 2010;5:199-211. DOI: 10.3923/jm.2010.199.211
  49. 49. Padmavathi T, Nandy V, Agarwal P. Optimization of the medium for the production of cellulases by Aspergillus terreus and Mucor plumbeus. European Journal of Experimental Biology. 2012;2:1161-1170
  50. 50. Mrudula S, Murugammal R. Production of cellulase by Aspergillus niger under submerged and solid state fermentation using coir waste as a substrate. Brazilian Journal of Microbiology. 2011;42:1119-1127. DOI: 10.1590/S1517-838220110003000033
  51. 51. Lee CK, Darah I, Ibrahim CO. 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. 2011;2011:658493. DOI: 10.4061/2011/658493
  52. 52. Xu X, Song Z, Yin Y, Zhong F, Song J, Huang J, et al. Solid-state fermentation production of chitosanase by Streptomyces with waste mycelia of Aspergillus niger. Advances in Enzyme Research. 2021;9:10-18. DOI: 10.4236/aer.2021.91002
  53. 53. Zhang H, Sang Q, Zhang W. Statistical optimization of chitosanase production by Aspergillus sp. QD-2 in submerged fermentation. Annales de Microbiologie. 2012;62:193-201. DOI: 10.1007/s13213-011-0246-1
  54. 54. Hirano K, Arayaveerasid S, Seki K, Adams DJ, Mitsutomi M. Characterization of a chitosanase from Aspergillus fumigatus ATCC13073. Bioscience, Biotechnology, and Biochemistry. 2012;76:1523-1528. DOI: 10.1271/bbb.120248
  55. 55. Jebur HA, Abdulateef AA, Thbit ZA. Chitosan production from Aspergillus oryzae SU-B2 by submerged fermentation and studying some of its physiochemical and antibacterial characteristics. Jordan Journal of Pharmaceutical Sciences. 2019;11:609-613
  56. 56. Kazemi S, Khayati G, Faezi-Ghasemi M. Β-galactosidase production by Aspergillus niger ATCC 9142 using inexpensive substrates in solid-state fermentation: Optimization by orthogonal arrays design. Iranian Biomedical Journal. 2016;20:287-294. DOI: 10.22045/ibj.2016.06
  57. 57. Kapnoor S, Mulimani VH. Production of α-galactosidase by Aspergillus oryzae through solid-state fermentation and its application in soymilk Galactooligosaccharide hydrolysis. Brazilian Archives of Biology and Technology. 2010;53:211-218. DOI: 10.1590/S1516-89132010000100026
  58. 58. Bailey MJ, Linko M. Production of β-galactosidase by Aspergillus oryzae in submerged bioreactor cultivation. Journal of Biotechnology. 1990;16:57-66. DOI: 10.1016/0168-1656(90)90065-J
  59. 59. Nizamuddin S, Sridevi A, Narasimha G. Production of β-galactosidase by Aspergillus oryzae in solid-state fermentation. African Journal of Biotechnology. 2008;7:1096-10100. DOI: 10.5897/AJB07.325
  60. 60. Alegre ACP, de Lourdes Teixeira de Moraes Polizeli M, Terenzi HF, Jorge JA, Guimarães LHS. Production of thermostable invertases by Aspergillus caespitosus under submerged or solid state fermentation using agroindustrial residues as carbon source. Brazilian Journal of Microbiology. 2009;40:612-622. DOI: 10.1590/S1517-838220090003000025
  61. 61. Raju CHAI, Pulipati K, Jetti A. Production of invertase by Aspergillus niger under solid state fermentation using orange fruit peel as substrate. Advances in Crop Science and Technology. 2016;04:1-4. DOI: 10.4172/2329-8863.1000247
  62. 62. Alves JNO, Jorge JA, Guimarães LHS. Production of invertases by anamorphic (Aspergillus nidulans) and teleomorphic (emericela nidulans) fungi under submerged fermentation using rye flour as carbon source. Advances in Microbiology. 2013;03:421-429. DOI: 10.4236/aim.2013.35057
  63. 63. Kumar R, Kaur J, Jain S, Kumar A. Optimization of laccase production from Aspergillus flavus by design of experiment technique: Partial purification and characterization. Journal, Genetic Engineering & Biotechnology. 2016;14:125-131. DOI: 10.1016/j.jgeb.2016.05.006
  64. 64. Abdallah YK, Estevez AT, Tantawy DEDM, Ibraheem AM, Khalil NM. Employing laccase-producing Aspergillus sydowii NYKA 510 as a cathodic biocatalyst in self-sufficient lighting microbial fuel cell. Journal of Microbiology and Biotechnology. 2019;29:1861-1872. DOI: 10.4014/jmb.1907.07031
  65. 65. Fokina O, Fenchel A, Winandy L, Fischer R. Immobilization of LccC laccase from Aspergillus nidulans on hard surfaces via Fungal hydrophobins. Applied and Environmental Microbiology. 2016;82(21):6395-3402. DOI: 10.1128/AEM.01413-16
  66. 66. Mahadik ND, Puntambekar US, Bastawde KB, Khire JM, Gokhale DV. Production of acidic lipase by Aspergillus niger in solid state fermentation. Process Biochemistry. 2002;38:715-721. DOI: 10.1016/S0032-9592(02)00194-2
  67. 67. Salihu A, Bala M, Alam MZ. Lipase production by Aspergillus niger using sheanut cake: An optimization study. Journal of Taibah University for Science. 2016;10:850-859. DOI: 10.1016/j.jtusci.2015.02.011
  68. 68. Ahmed A, Badar R, Khalique N. Screening and optimization of submerged fermentation of lipolytic Aspergillus oryzae. BioResources. 2019;14:7664-7674. DOI: 10.15376/biores.14.4.7664-7674
  69. 69. Kirsh D. Lipase Production by Penicillium oxalicum and Aspergillus flavus. Botanical Gazette. 1935;97:321-333. DOI: 10.1086/334555
  70. 70. Nema A, Patnala SH, Mandari V, Kota S, Devarai SK. Production and optimization of lipase using Aspergillus niger MTCC 872 by solid-state fermentation. Bulletin of the National Research Centre. 2019;43:1-8. DOI: 10.1186/s42269-019-0125-7
  71. 71. Borkar V, Chakraborty S, Gokhale JS. Fermentative production of naringinase from Aspergillus niger van Tieghem MTCC 2425 using citrus wastes: Process optimization, partial purification, and characterization. Applied Biochemistry and Biotechnology. 2021;193:1321-1337. DOI: 10.1007/s12010-020-03385-9
  72. 72. Mendoza-Cal A, Cuevas-Glory L, Lizama-Uc G, Ortiz-Vázquez E. Naringinase production from filamentous fungi using grapefruit rind in solid state fermentation. African Journal of Microbiology Research. 2010;4:1964-1969
  73. 73. Chen Y, Ni H, Chen F, Cai H, Li L, Su W. Purification and characterization of a naringinase from Aspergillus aculeatus JMUdb058. Journal of Agricultural and Food Chemistry. 2013;61:931-938. DOI: 10.1021/jf303512q
  74. 74. Zhu Y, Jia H, Xi M, Li J, Yang L, Li X. Characterization of a naringinase from Aspergillus oryzae 11250 and its application in the debitterization of orange juice. Process Biochemistry. 2017;62:114-121. DOI: 10.1016/j.procbio.2017.07.012
  75. 75. Shanmugaprakash M, Vinoth Kumar V, Hemalatha M, Melbia V, Karthik P. Solid-state fermentation for the production of debittering enzyme naringinase using Aspergillus niger MTCC 1344. Engineering in Life Sciences. 2011;11:322-325. DOI: 10.1002/elsc.201000128
  76. 76. Shanmugaprakash M, Vinothkumar V, Ragupathy J, Reddy DA. Biochemical characterization of three phase partitioned naringinase from Aspergillus brasiliensis MTCC 1344. International Journal of Biological Macromolecules. 2015;80:418-423. DOI: 10.1016/j.ijbiomac.2015.06.057
  77. 77. Xia X-K, Zhang Y-E, Lei S-J, Hu B, Fu C-X. Identification and iterative combinatorial mutagenesis of a new naringinase-producing strain, Aspergillus tubingensis MN589840. Letters in Applied Microbiology. 2021;72:141-148. DOI: 10.1111/lam.13379
  78. 78. Chang H-Y, Lee Y-B, Bae H-A, Huh J-Y, Nam S-H, Sohn H-S, et al. Purification and characterisation of Aspergillus sojae naringinase: The production of prunin exhibiting markedly enhanced solubility with in vitro inhibition of HMG-CoA reductase. Food Chemistry. 2011;124:234-241. DOI: 10.1016/j.foodchem.2010.06.024
  79. 79. Sandri I, Silveira M. Production and application of pectinases from Aspergillus niger obtained in solid state cultivation. Beverages. 2018;4:48. DOI: 10.3390/beverages4030048
  80. 80. El-Sheekh MM, Ismail A-MS, El-Abd MA, Hegazy EM, El-Diwany AI. Effective technological pectinases by Aspergillus carneus NRC1 utilizing the Egyptian orange juice industry scraps. International Biodeterioration & Biodegradation. 2009;63:12-18. DOI: 10.1016/j.ibiod.2008.06.002
  81. 81. Wolf-Márquez VE, Martínez-Trujillo MA, Aguilar Osorio G, Patiño F, Álvarez MS, Rodríguez A, et al. Scaling-up and ionic liquid-based extraction of pectinases from Aspergillus flavipes cultures. Bioresource Technology. 2017;225:326-335. DOI: 10.1016/j.biortech.2016.11.067
  82. 82. Shanmugavel M, Vasantharaj S, Yazhmozhi A, Bhavsar P, Aswin P, Felshia C, et al. A study on pectinases from Aspergillus tamarii: Toward greener approach for cotton bioscouring and phytopigments processing. Biocatalysis and Agricultural Biotechnology. 2018;15:295-303. DOI: 10.1016/j.bcab.2018.06.013
  83. 83. Pradal-Velázquez M, Martínez-Trujillo MA, Aguilar-Osorio G. Xylanases and pectinases of Aspergillus flavus CECT-2687 on different carbon sources and initial ph values. Revista Mexicana de Ingenieria Quimica. 2018;17:421-431. DOI: 10.24275/10.24275/uam/izt/dcbi/revmexingquim/2018v17n2/Pradal
  84. 84. Semenova MV. Isolation and properties of pectinases from the fungus Aspergillus japonicus. Biochemistry (Moscow). 2003;68:559-569. DOI: 10.1023/A:1023959727067
  85. 85. Shivanna GB, Venkateswaran G. Phytase production by Aspergillus niger CFR 335 and Aspergillus ficuum SGA 01 through submerged and solid-state fermentation. Scientific World Journal. 2014;2014:392615. DOI: 10.1155/2014/392615
  86. 86. Shah PC, Kumar VR, Dastager SG, Khire JM. Phytase production by Aspergillus niger NCIM 563 for a novel application to degrade organophosphorus pesticides. AMB Express. 2017;7:66. DOI: 10.1186/s13568-017-0370-9
  87. 87. Tian M, Yuan Q. Optimization of phytase production from potato waste using Aspergillus ficuum. 3 Biotech. 2016;6:256. DOI: 10.1007/s13205-016-0573-9
  88. 88. Yamada K, Minoda Y, Yamamoto S. Phytase from Aspergillus terreus: Part I. Production, purification and some general properties of the enzyme. Agricultural and Biological Chemistry. 1968;32:1275-1282. DOI: 10.1080/00021369.1968.10859223
  89. 89. Ahmed ME. Extraction and purification of protease from Aspergillus niger isolation. Pharmacy & Pharmacology International Journal. 2018;6:62-65. DOI: 10.15406/ppij.2018.06.00162
  90. 90. de Castro RJS, Sato HH. Protease from Aspergillus oryzae: Biochemical characterization and application as a potential biocatalyst for production of protein hydrolysates with antioxidant activities. Journal of Food Processing. 2014;2014:1-11. DOI: 10.1155/2014/372352
  91. 91. Tremacoldi CR, Carmona EC. Production of extracellular alkaline proteases by Aspergillus clavatus. World Journal of Microbiology and Biotechnology. 2005;21:169-172. DOI: 10.1007/s11274-004-2724-0
  92. 92. Malathi S, Chakraborty R. Production of alkaline protease by a new Aspergillus flavus isolate under solid-substrate fermentation conditions for use as a depilation agent. Applied and Environmental Microbiology. 1991;57:712-716. DOI: 10.1128/AEM.57.3.712-716.1991
  93. 93. Costa AM, Ribeiro WX, Kato E, Monteiro ARG, Peralta RM. Production of tannase by Aspergillus tamarii in submerged cultures. Brazilian Archives of Biology and Technology. 2008;51:399-404. DOI: 10.1590/S1516-89132008000200021
  94. 94. Kumar R, Sharma J, Singh R. Production of tannase from Aspergillus ruber under solid-state fermentation using jamun (Syzygium cumini) leaves. Microbiological Research. 2007;162:384-390. DOI: 10.1016/j.micres.2006.06.012
  95. 95. Viswanath V, Leo VV, Prabha SS, Potty VP, Jisha MS. Optimized production of tannase from cashew testa using Aspergillus niger MTCC 5898. Food Biotechnology. 2016;30:249-262. DOI: 10.1080/08905436.2016.1234392
  96. 96. Beniwal V, Rajesh, Goel G, Kumar A, Chhokar V. Production of tannase through solid state fermentation using Indian rosewood (Dalbergia sissoo) sawdust—A timber industry waste. Annales de Microbiologie. 2013;63:583-590. DOI: 10.1007/s13213-012-0508-6
  97. 97. Gonçalves HB, Riul AJ, Quiapim AC, Jorge JA, Guimarães LHS. Characterization of a thermostable extracellular tannase produced under submerged fermentation by Aspergillus ochraceus. Electronic Journal of Biotechnology. 2012;15:5. DOI: 10.2225/vol15-issue5-fulltext-5
  98. 98. Liu TPSL, Brandão Costa RMP, de Vasconcelos Freitas DJ, Oliveira Nacimento C, de Souza Motta CM, Bezerra RP, et al. Tannase from Aspergillus melleus improves the antioxidant activity of green tea: Purification and biochemical characterisation. International Journal of Food Science and Technology. 2017;52:652-661. DOI: 10.1111/ijfs.13318
  99. 99. Banerjee D, Mondal K, Pati B. Tannase production by Aspergillus aculeatus DBF9 through solid-state fermentation. Acta Microbiologica et Immunologica Hungarica. 2007;54:159-166. DOI: 10.1556/AMicr.54.2007.2.6
  100. 100. Bajar S, Singh A, Bishnoi NR. Exploration of low-cost agro-industrial waste substrate for cellulase and xylanase production using Aspergillus heteromorphus. Applied Water Science. 2020;10:153. DOI: 10.1007/s13201-020-01236-w
  101. 101. Sewalt V, Shanahan D, Gregg L, La Marta J, Carrillo R. The generally recognized as safe (GRAS) process for industrial microbial enzymes. Industrial Biotechnology. 2016;12:295-302. DOI: 10.1089/ind.2016.0011
  102. 102. Deckers M, Deforce D, Fraiture MA, Roosens NH. Genetically modified micro-organisms for industrial food enzyme production: An overview. Food. 2020;9:326. DOI: 10.3390/foods9030326
  103. 103. Adrio JL, Demain AL. Recombinant organisms for production of industrial products. Bioengineered Bugs. 2010;1:116-131
  104. 104. Khan S, Ullah MW, Siddique R, Nabi G, Manan S, Yousaf M, et al. Role of recombinant DNA technology to improve life. International Journal of Genomics. 2016;2016:2405954. DOI: 10.1155/2016/2405954
  105. 105. Archer DB. Enzyme production by recombinant Aspergillus. In: Yoshikatsu M, Tadayuki I, editors. Recombinant Microbes for Industrial and Agricultural Applications. Boca Raton, FL: CRC Press; 2020. pp. 373-393. DOI: 10.1201/9781003067191
  106. 106. Meyer V, Punt PJ, Ram AFJ. Genetics, genetic manipulation, and approaches to strain improvement of filamentous fungi. In: Bull AT, Junker B, Katz L, Lynd LR, Masurekar P, Reeves CD, Zhao H, editors. Manual of Industrial Microbiology and Biotechnology. 3rd ed. USA: American Society of Microbiology; 2010. pp. 318-329. DOI: 10.1128/9781555816827.ch22
  107. 107. Yu X-C, Ma S-L, Xu Y, Fu C-H, Jiang C-Y, Zhou C-Y. Construction and application of a novel genetically engineered Aspergillus oryzae for expressing proteases. Electronic Journal of Biotechnology. 2017;29:32-38. DOI: 10.1016/j.ejbt.2017.07.004
  108. 108. Prathumpai W, Flitter SJ, McIntyre M, Nielsen J. Lipase production by recombinant strains of Aspergillus niger expressing a lipase-encoding gene from Thermomyces lanuginosus. Applied Microbiology and Biotechnology. 2004;65:714-719. DOI: 10.1007/s00253-004-1699-y
  109. 109. Record E, Punt PJ, Chamkha M, Labat M, van Den Hondel CAMJJ, Asther M. Expression of the Pycnoporus cinnabarinus laccase gene in Aspergillus niger and characterization of the recombinant enzyme: P. cinnabarinuslaccase gene expression in A. niger. European Journal of Biochemistry. 2002;269:602-609. DOI: 10.1046/j.0014-2956.2001.02690.x
  110. 110. Bohlin C, Jönsson LJ, Roth R, van Zyl WH. Heterologous expression of Trametes versicolor laccase in Pichia pastoris and Aspergillus niger. Applied Biochemistry and Biotechnology. 2006;129-132:195-214
  111. 111. Dragosits M, Pflügl S, Kurz S, Razzazi-Fazeli E, Wilson IBH, Rendic D. Recombinant Aspergillus β-galactosidases as a robust glycomic and biotechnological tool. Applied Microbiology and Biotechnology. 2014;98(8):3553-3567. DOI: 10.1007/s00253-013-5192-3
  112. 112. Mojsov K. Microbial α-amylases and their industrial applications: A review. International Journal of Management in Education. 2012;2:583-609
  113. 113. Lemnaru Popa G-M, Truşcă RD, Ilie C-I, Țiplea RE, Ficai D, Oprea O, et al. Antibacterial activity of bacterial cellulose loaded with bacitracin and amoxicillin: In vitro studies. Molecules. 2020;25:4069. DOI: 10.3390/molecules25184069
  114. 114. Limón MC, Chacón MR, Mejías R, Delgado-Jarana J, Rincón AM, Codón AC, et al. Increased antifungal and chitinase specific activities of Trichoderma harzianum CECT 2413 by addition of a cellulose binding domain. Applied Microbiology and Biotechnology. 2004;64:675-685. DOI: 10.1007/s00253-003-1538-6
  115. 115. Kango N, Jana UK, Choukade R. Fungal enzymes: Sources and biotechnological applications. In: Satyanarayana T, Deshmukh S, Deshpande M, editors. Advancing Frontiers in Mycology & Mycotechnology. Singapore: Springer Singapore; 2019. pp. 515-538. DOI: 10.1007/978-981-13-9349-5_21

Written By

Mohammadhassan Gholami-Shabani, Masoomeh Shams-Ghahfarokhi, Fatemehsadat Jamzivar and Mehdi Razzaghi-Abyaneh

Submitted: 18 July 2021 Reviewed: 22 November 2021 Published: 03 January 2022