Novel Food Grade Enzymes: Abhishek Dutt Tripathi Kianoush Khosravi-Darani Suresh Kumar Srivastava
Novel Food Grade Enzymes: Abhishek Dutt Tripathi Kianoush Khosravi-Darani Suresh Kumar Srivastava
Kianoush Khosravi-Darani
Suresh Kumar Srivastava Editors
Novel Food
Grade
Enzymes
Applications in Food Processing and
Preservation Industries
Novel Food Grade Enzymes
Abhishek Dutt Tripathi •
Kianoush Khosravi-Darani •
Suresh Kumar Srivastava
Editors
# The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Singapore
Pte Ltd. 2022
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by
similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this
book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or
the editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Preface
Enzymes play a significant role in pharma, agriculture, and other allied industries. In
the modern era, there is significant application of enzymes in food processing
industries and our proposed book will give deeper insight into food applications of
enzymes. This book covers all the aspects of enzyme including its classification,
kinetics, microbial production, biosynthetic pathways, commodity-wise industrial
applications, and its downstream processing strategies. The broad focus of this book
will be on the application of various classes of enzymes in dairy, fruits and vegeta-
ble, cereals and oilseeds, meat and poultry, brewing and food packaging industries.
The first chapter of this book gives a brief introduction about enzymes classifications
and their kinetics. Chapters 2, 3, 5, 6, 7, and 8 provide information regarding enzyme
applications in different food commodities like fruits and vegetables, dairy, bever-
age, and meat processing industries. These chapters also focus on different food
grade enzyme biosynthesis mechanism in simpler and illustrative way. Besides that,
this book will also allow the readers to understand the biosynthesis, production,
optimization, and purification of certain less known enzymes such as CGTase and
naringinase. Chapters 9 and 12 of this book will be giving some novel information
related to application of enzymes specifically for improvement of flavor, color,
organoleptic attributes, shelf-life extension, and consumer acceptability of value-
added diversified food products. Chapters 10 and 11 also cover some of the very
interesting aspects of enzymes application in the food packaging and development of
biosensors which will be highly beneficial for those researchers and students who are
interested in learning the food safety issues and indulged in product quality improve-
ment. Enzymes play a significant role in the development of GM (Genetically
Modified) food production. Chapters 13 and 14 provide detailed information related
to enzymes application in GM foods. Chapter 15 highlights the various enzyme
infusion strategies adopted in the food processing industries that will enable the
students and entrepreneurs to understand the basic concept of enzyme activity and
stability after infusion in food. Future prospects of enzymes and their
nanotechnological applications have been discussed in Chapters 16 and 17 of this
book. The illustrated case studies related to certain novel enzymes will allow the
academicians and researchers to broaden their knowledge and will provide an
opportunity to carry out extensive research work pertaining to the applications of
such enzymes. The authors contributing to this book have already done extensive
v
vi Preface
research work in the proposed area and the contributed chapters will also be
highlighting some of the salient research work already published in the reputed
journals by internationally recognized authors in the proposed area. The flowchart
described in certain chapters will provide all the necessary information related to the
production, optimization, and characterization of food grade enzymes in simpler
manner. This book will not only cover the basic enzyme kinetics part but also
provide information related to the efficient enzyme recovery and purification pro-
cess. It also gives broad information on the factors affecting enzyme activity and
stability as well as involvement of enzyme in the development of functional food
combating chronic diseases and disorders.
As per our survey, only few books are available on the role of enzymes in food
processing although this is a prominent area of study and needs to be covered in
detail. The earlier books primarily focused on the general enzyme properties and its
application. However, no deep insight knowledge pertaining to the commodity-wise
application of food grade enzymes has been previously illustrated. We have tried to
cover all the aspects of food grade enzymes including its general properties, kinetics,
wide application in all the food processing and packaging industries. Although
numerous books on medical and therapy-based application of enzymes are available
in the market, no books on wider perspective of food grade enzymes applications are
available till now. The simpler language, clearly illustrated concepts, and broadly
discussed case studies related to various food grade enzymes will enable the readers
to strengthen their concept and knowledge. This book will be beneficial for the
faculty members, undergraduate, postgraduate, and PhD students of Microbiology,
Food Technology, Dairy Technology, Horticulture, Biotechnology, Biochemistry,
and other allied discipline of Life Science. This book will also be beneficial to those
authors who are indulged in the research activities related to the above-mentioned
subject areas such as food science, enzyme and fermentation technology, bioprocess
technology, and biochemical engineering. The involvement of foreign authors and
editors will provide deeper insight about the subject knowledge and will improve the
quality of book content making it readable and acceptable at global level. I hope the
readers will enjoy reading this book and will develop interest in the area of enzyme
technology.
vii
Contents
ix
x Contents
xi
Food Enzymes: General Properties
and Kinetics 1
S. M. Khade, S. K. Srivastava, L. H. Kamble, and J. Srivastava
Abstract
Enzymes are the biocatalysts having a catalytic power that speeds up a chemical
reaction without changing the equilibrium of the reaction. Almost all enzymes are
protein in nature and the biocatalytic power lies in the integrity of their structural
conformation. The enzymes are highly specific with their substrate molecules that
convert into the product by decreasing the activation energy, without getting
changed itself. However, the biocatalytic power of the enzymes also depends on
several physico-chemical parameters such as temperature, pH, salt
concentrations, etc. of the reaction. The enzyme catalysis can be quantitatively
revealed by the enzyme kinetics mechanism which measures the reaction rates
and the affinity of enzymes towards the substrates and inhibitors. The enzymes
can be isolated from the various biological factories such as plant, animal, or
microbial cells depending upon the type of enzyme. However, in order to produce
at large scale, the microbial sources are the best choice which can effectively
reduce the production and purification cost of enzymes. The enzymes are widely
used in various sectors such as agriculture, environmental, leather tanning, paper
and pulp, chemical and pharmaceutical, detergent, food and beverages, etc. In this
chapter, we are mainly focussing on the role of enzymes in the food industry.
Keywords
1.1 Introduction
General Properties:
1. Almost all enzymes are protein in nature except a small group of catalytic RNA
molecules. The catalytic activity of the enzyme relies on the structural integrity of
native protein conformation.
2. Enzymes are very specific to their substrates.
3. Enzymes possess huge catalytic power which increases the rate of a chemical
reaction by reducing the activation energy without altering the equilibrium
position of the reaction.
4. The biocatalytic power of the enzyme depends on the optimum physio-chemical
parameters of the reaction such as pH, temperature, and salt concentration.
5. Enzymes are highly regulated in nature.
(a) Simple enzymes: The enzymes which are entirely made up of the proteins
constituting the amino acids only, e.g. trypsin, pepsin, etc. (Bisswanger, 2017).
(b) Conjugated enzymes: The enzymes which contain the non-protein part called
cofactor along with the protein part, called apoenzyme. In these enzymes, the
apoenzyme part is generally biologically inactive, if the cofactor is removed.
The complete biologically active conjugated enzyme (apoenzyme and cofactor)
1 Food Enzymes: General Properties and Kinetics 3
Most of the enzymes are named with the suffix –ase; however, some old names are
exceptions, e.g. trypsin, ptyalin, pepsin, chymotrypsin, renin, etc. Also, some of the
old names indicate the source rather than the action, e.g. papain from Papaya and
bromelain from Pineapple (Bromeliaceae family). Many enzymes are named based
on substrate and the chemical reaction they catalyse with the suffix –ase,
e.g. pyruvate kinase that helps in the synthesis of pyruvate from the phosphoenol-
pyruvate, whereas pyruvate dehydrogenase enzyme which converts pyruvate to
acetyl-CoA. To avoid the confusion arose due to the common names, an Interna-
tional Commission on enzymes was established for the systematic nomenclature of
enzymes.
The enzyme commission (EC) has set a regulation for the nomenclature of
enzyme. According to EC, each enzyme is classified and named based on the type
of chemical reaction, and for each enzyme, number with four parts is given, e.g. EC
1.7.3.3 (Uricase); EC 3.5.1.1 (Asparaginase). The first three numbers denote major
class, subclass, and sub-subclass, respectively, and the last number is a serial number
in the sub-subclass, which indicates the order in which each enzyme is added to the
list (Boyce & Tipton, 2001) (Table 1.1).
All the enzymes are classified into six classes based on the type of chemical
reaction they catalyse. The first integer in the EC number denotes the class of
enzymes.
1.2.1 EC 1 Oxidoreductase
The enzymes which catalyse the oxidation-reduction reaction are classified under
this class. The enzymes are involved in the transfer of electrons from one molecule
(oxidant) to another molecule (reductant) (Table 1.2).
1.2.2 EC 2 Transferase
The enzymes which catalyse the transfer of groups from one molecule to another are
classified under transferase class. The groups involved in the transfer are phosphoryl
(–PO32), methyl (–CH3), carboxyl (–COOH), amino (–NH2), acyl (–RC¼O), and
carbonyl (–C¼O). The common names for these enzymes start with prefix trans with
exceptions of kinases, phosphorylases, etc. (Table 1.3).
XAþY!XþYA
1.2.3 EC 3 Hydrolases
These are the enzymes which catalyse the cleavage of a covalent bond by adding
water (Table 1.4).
X Y þ H2 O ! X H þ Y OH
1.2.4 EC 4 Lyases
These are the enzymes which cleave C–C, C–O, C–N, C–S, and other bonds without
hydrolysis or oxidation, but by means of elimination, which results into the forma-
tion of double bond or a new ring or contrarily adding groups to double bonds
(Table 1.5).
X ¼ Y þ SP ! X S þ Y P
1.2.5 EC 5 Isomerases
XY!YX
1.2.6 EC 6 Ligases
These are the enzymes which join the two molecules by forming a chemical bond
like C–C, C–N, C–O, and C–S. The energy required for these is gained by the
cleavage of ATP or NAD. Ligases are also called synthetases, because of synthesis
of new molecule (Table 1.7).
The collision of an atom, ion, or molecule of one substance with an atom, ion, or
molecule of other results into a chemical reaction between the two substances. After
this collision, the intermediate product is produced, having higher chemical energy
than the chemical energies of both the reactants together. The transition state in the
reaction is achieved with the help of some of the energy entering the reaction apart
from the chemical energy of the reactants. The transition state is the stage with
highest free energy. The difference in free energy between the transition state and the
reactants is known as the Gibbs free energy of activation or only the activation
energy.
The enzyme plays an important role in lowering the activation energy of a
reaction which results in increase in the rate of reaction in both the directions.
However, the enzyme cannot alter the relative energies of the initial and final states.
In addition, the catalyst does not alter the position of equilibrium of a chemical
1 Food Enzymes: General Properties and Kinetics 7
reaction, but only increases the rate of a reaction by lowering the activation energy
(Fig. 1.1).
The biocatalysis of a chemical reaction begins with the binding of enzyme and the
substrate, the substrate to be catalysed. The substrate specifically binds at the
specialized region in the enzyme called active site with multiple weak
non-covalent interactions such as hydrophobic, hydrogen bond, ionic interaction,
or reversible covalent bonds. Upon binding of the enzyme and substrate, a large
amount of free energy is released, called binding energy, which is used to lower the
activation energy. The general catalysed reaction between enzyme and substrate may
be represented as
EþSES!EþP
This model assumes that the binding of substrate at the active site of enzyme is
highly compact, which may be due to the high degree of similarity between the shape
of the substrate and the geometry of the binding site, i.e. active site on the enzyme.
This binding of substrate and enzyme is just like a lock and a specific key (Fig. 1.2).
8 S. M. Khade et al.
S
Acve site P
E-S E
E
Enzyme changes
S Slightly different shape slightly as P
acve site substrate binds
E E-S E
This model describes the behaviour of protein with some three-dimensional flexibil-
ity. Based on this model, the conformational change occurs in the enzyme upon
binding of the substrate which leads to complementary fit as soon as substrate is
bound. However, prior to substrate binding, the active site has slightly different
three-dimensional shape. After binding of the substrate, the enzyme-substrate com-
plex with transition state is formed which subsequently converts into product and
free enzyme (Fig. 1.3).
Leonor Michaelis and Maud Menten in 1913 proposed a model for the kinetics study
of enzyme-catalysed reaction of hydrolysis of disaccharide sugar and sucrose by
invertase enzyme into monosaccharide sugars, glucose, and fructose units.
Michaelis-Menten assessed the relationship between the reaction velocity and the
substrate concentration. The results of this disaccharide hydrolysis into the two
monosaccharides by the invertase are represented with the following reaction:
k2
E þ S kk11 E S ! E þ P
Vmax
0 Km [S]
1 K 1 1
¼ m þ ð1:1Þ
v V max ½S V max
v
v ¼ K m þ V max ð1:2Þ
½S
Vmax
- Km
[S]
½ S ½S K
¼ þ m ð1:3Þ
v V max V max
1
The straight line graph will yield V max as the slope, whereas VKmax
m
as the y-intercept
and Km as the x-intercept (Fig. 1.7).
12 S. M. Khade et al.
The enzymes play a vital role either in the food processing or additives in the food
industries. The major applications of food enzymes are in the baking industries,
dairy industries, starch processing, juice industries, brewing industries, etc.
The enzymes employed for the processing of dairy products become essential in the
improvement of the quality and physiologically health benefits. The enzymes,
chymosin (coagulation), lipases (low-down fat level), and lysozymes (to avoid late
blowing), are used in the production of cheese (Kilcawley et al., 1998; Marseglia
et al., 2013). In addition, β-galactosidases and lactases enzymes are predominantly
employed in the hydrolysis of lactose sugar in milk into glucose and galactose to
avoid lactose intolerance.
The α-amylase is the prominent enzyme used in the baking industry to act on the
starch present in the dough of the bread into the dextrins, which enhances the
fermentation process. The released extra sugar in the dough improves the taste and
textural properties of the bread (Souza, 2010).
The enzymes, amylases and glucoamylases, are mainly used in the clarification of
cloudy juices. Pectinases are used in the hydrolysis of structural
heteropolysaccharide of fruit cell wall pectin, leading to increase in the juice
production. Naringinases are mainly employed to reduce the bitterness of the citrus
juices (Tapre & Jain, 2014).
1 Food Enzymes: General Properties and Kinetics 13
The α-amylase hydrolyses the starch into maltose and glucose units with reduced
viscosity. β-Glucanases hydrolyse glucans to reduce the viscosity and smooth wort
separation. Proteases are used to process the malt which allows favourable content
for yeast growth. Xylanases are used to enhance the 5-C pentose utilization in malt,
barley, and wheat with extraction and beer filtration (Li et al., 2012).
References
Barnes-Svarney, P., & Svarney, T. E. (2014). The handy biology answer book. Visible Ink Press.
Bisswanger, H. (2017). Enzyme kinetics: Principles and methods. John Wiley & Sons.
Boyce, S., & Tipton, K. F. (2001). Enzyme classification and nomenclature. eLS. https://doi.org/10.
1038/npg.els.0000710
Dai, N., Schaffer, A. A., Petreikov, M., & Granot, D. (1995). Arabidopsis thaliana hexokinase
cDNA isolated by complementation of yeast cells. Plant Physiology, 108(2), 879.
El Enshasy, H. A., Elsayed, E. A., Suhaimi, N., Abd Malek, R., & Esawy, M. (2018). Bioprocess
optimization for pectinase production using Aspergillus niger in a submerged cultivation
system. BMC Biotechnology, 18(1), 71.
Faulds, C. B., Mandalari, G., LoCurto, R., Bisignano, G., & Waldron, K. W. (2004). Arabinoxylan
and mono-and dimeric ferulic acid release from brewer’s grain and wheat bran by feruloyl
esterases and glycosyl hydrolases from humicolainsolens. Applied Microbiology and Biotech-
nology, 64(5), 644–650.
Gazel, N., & Yildiz, H. B. (2016). Enzyme-based biosensors in food industry via surface
modifications. In Surface treatments for biological, chemical, and physical applications
(pp. 227–252). Wiley.
Gramss, G., & Rudeschko, O. (1998). Activities of oxidoreductase enzymes in tissue extracts and
sterile root exudates of three crop plants, and some properties of the peroxidase component. New
Phytologist, 138(3), 401–409.
Heinstra, P. W., Geer, B. W., Seykens, D., & Langevin, M. (1989). The metabolism of ethanol-
derived acetaldehyde by alcohol dehydrogenase (EC 1.1. 1.1) and aldehyde dehydrogenase
(EC 1.2. 1.3) in Drosophila melanogaster larvae. Biochemical Journal, 259(3), 791–797.
Huang, S. K., Chiu, A. W. H., Pu, Y. S., Huang, Y. K., Chung, C. J., Tsai, H. J., et al. (2008).
Arsenic methylation capability, heme oxygenase-1 and NADPH quinone oxidoreductase-1
genetic polymorphisms and the stage and grade of urothelial carcinomas. Urologia
Internationalis, 80(4), 405–412.
Huhtanen, P., & Khalili, H. (1992). The effect of sucrose supplements on particle-associated
carboxymethylcellulase (EC 3.2. 1.4) and xylanase (EC 3.2. 1.8) activities in cattle given
grass-silage-based diet. British Journal of Nutrition, 67(2), 245–255.
Janeček, S. (2009). Amylolytic enzymes-focus on the alpha-amylases from archaea and plants.
Nova Biotechnologica et Chimica, 9(1), 5–25.
Kiess, M., Hecht, H. J., & Kalisz, H. M. (1998). Glucose oxidase from Penicillium amagasakiense:
Primary structure and comparison with other glucose-methanol-choline (GMC)
oxidoreductases. European Journal of Biochemistry, 252(1), 90–99.
Kilcawley, K. N., Wilkinson, M. G., & Fox, P. F. (1998). Enzyme-modified cheese. International
Dairy Journal, 8(1), 1–10.
Kohler, R. (1971). The background to Eduard Buchner’s discovery of cell-free fermentation.
Journal of the History of Biology, 4, 35–61.
Kouker, G., & Jaeger, K. E. (1987). Specific and sensitive plate assay for bacterial lipases. Applied
and Environmental Microbiology, 53(1), 211–213.
14 S. M. Khade et al.
Mahmoud Aminlari
Abstract
Enzymes are biological catalysts which increase the rate of biochemical reactions
in living cells. It is important that enzymologist understands the specific action of
particular enzyme in a plant or animal tissue and applies these properties in vitro
and in a food product. Most enzymes can be used as processing aids and as
protection agents against microbial and deteriorative processes. Although the
advent of recombinant DNA technology and advances made in food applications
by microorganisms are more practical and economic, some of these enzymes are
sufficiently abundant in their natural sources to make them amenable to large-
scale production (for example, egg-white lysozyme and plant proteases). In this
chapter, several plant and animal enzymes, their occurrence, and potential
applications in food industry will be presented. Emphasis will be made on
enzyme working on carbohydrates, proteins, and lipids. A section is devoted to
miscellaneous enzymes used in food industry, such as phenylalanine ammonia
lyase of wheat seedling which metabolizes Phe, thereby rendering foods suitable
for PKU patients. In the final section of this chapter, examples of chemical
modification of enzymes to improve their properties will be discussed and
examples of the studies on modification of chicken egg-white lysozyme to
enhance its functional and antimicrobial activities, performed in the laboratory
of this author, will be presented.
M. Aminlari (*)
Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
Department of Biochemistry, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
e-mail: aminlari@shirazu.ac.ir
Keywords
2.1 Introduction
in the laboratory of this author will be presented. Table 2.1 summarizes the use and
suggested use of enzymes in food industry.
Most of the enzymes working on food carbohydrates are hydrolase, i.e., use water as
one of the substrates. These enzymes are commonly referred to as glycosyl
hydrolases or glycosidases.
They catalyze the hydrolysis of glycosidic bonds in complex sugars. They are
amazingly normal enzymes with roles in nature including degrading of complex
biomass like starch (amylase), cellulose (cellulase), hemicellulose, and
as antibacterial safeguard methodologies (e.g., lysozyme), in pathogenesis
components (e.g., viral neuraminidases), and in ordinary cell work (e.g., managing
mannosidases associated with N-connected glycoprotein biosynthesis). Along with
glycosyltransferases, glycosidases’ are the major synergist apparatus for the synthe-
sis and breakage of glycosidic bonds (Bourne & Henrissat, 2001). Glycosidases
hydrlyze α-1,4 or α-1,6 glycosidic bonds. Glycosidases can be classified either as
“retaing” or “invering” types, in light of the destiny of the anomeric configuration (α
or β) of the hydrolyzed glycosidic bond. One more broad differentiation among
glycosidases is whether they are “endo” or “exo” splitting. Exo-acting works for the
most part on the non-reducing terminal of the substrate, while endo-acting types
rondomly assault internal glycosidic bonds of the substrate. Naming glycosidases as
“α” and “β” (as in amylases and glucosidases) perceives the anomeric configuration
of the freed reducing group as being axial and equatorial, respectively (Whitaker,
1994). Specialty applications for various glycosidases continue to emerge.
Glycosidases account for about half of enzyme used as processing aids in the food
industry, primarily for the production of low molecular weight sweetener and
bulking or thickening agents (dextrins) from starch (such as amylases), and for
carbohydrate modification in baking and fruit products (such as cellulases and pectic
enzymes) (Parkin, 2017). Most of these enzymes are commercially available. A
summary of the types and classification of glycosidases of most importance in foods
is provided in Table 2.2.
2.2.1 Amylases
2.2.1.1 a-Amylases
The amylases are utilized to hydrolyze starch into lower molecular weight dextrins
and consequently “thin” starch suspensions. α-Amylase is an endo- splitting glycosi-
dase that hydrolyses α-1,4 glycoside bond. The initial product is dextrin and the final
main products are maltose or maltotriose. The α-1,4 bonds close to α-1,6 branches
are impervious to hydrolysis. Broad hydrolysis of amylopectins with α-amylases
produces “α-limit dextrins,” as cleavage stops at the α-1,6 bonds. Figure 2.1 shows
the impacts of various glycosidases on starch molecule (Wong & Robertson, 2007;
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 21
Table 2.1 Some uses and suggested uses of enzymes in food industries (from Whitaker, 1994)
Enzyme Food Purpose or action
Amylases Baked goods Increase in maltose content for food fermentation
Brewing Conversion of starch to maltose for fermentation, removal of
starch turbidity
Cereals Conversion of starch to dextrins and maltose, increase water
absorption
Confectionary Recovery of sugar from candy scraps
Fruit juices Removal of starch to increase sparkling properties
Jellies Removal of starch to increase sparkling properties
Pectin Aid in preparation of pectin from apple pomace
Syrup Conversion of starch to low molecular weight dextrins (corn
syrup)
Vegetables Hydrolysis of starch as in tenderization of peas
Cellulase Brewing Hydrolysis of complex carbohydrates cell walls
Coffee Hydrolysis of cellulose during drying of beans
Fruits Removal of graininess of pears, peeling of apricot, tomatoes
Invertase Artificial Conversion of sucrose to glucose and fructose
honey
Candy Manufacture of chocolate-coated soft-cream candies
Lactase Ice cream Prevention of crystallization which results in grainy, sandy
texture
Milk Stabilization of milk proteins in frozen milk by removal of
lactose. Hydrolysis of lactose, permitting use by adults deficient
in lactase in intestinal and infants with congenital lactase
deficiency
Tannase Brewing Removal of polyphenolic compounds
Pentosanase Milling Recovery of starch from wheat flour
Naranginase Citrus Debittering citrus juice by hydrolysis of the glucoside naringin
Pectic Chocolate, Hydrolytic activity during fermentation of cocoa
enzymes cocoa
Coffee Hydrolysis of gelatinous coating during fermentation of beans
Fruits Softening
Fruit juice Improving yield of press juice, prevention of cloudiness,
improving concentration process
Olive Extracting oil
Wines Clarification
Proteases Baked goods Softening action in dough, cut mixing time, increase
extensibility of dough, improvement in grain, texture, loaf
volume, liberate β-amylase
Brewing Body, flavor, and nutrient development during fermentation,
aid in liberation and clarification, chill-proofing
Cereals Modification of proteins to increase drying rate, improve
product handling characteristics, manufacture of misu and tofu
Cheese Casein coagulation, characteristic flavor during aging
Chocolate, Action on beans during fermentation
cocoa
(continued)
22 M. Aminlari
Table 2.2 Catalytic properties of some glycolytic enzymes important in food technology (from
Whitaker, 1994; Parkin, 2017)
Bond Product
Enzyme selectivity selectivity Products
α-Amylase (endo) α-1!4 glucose Retaining Initial major product dextins, final
α!α product maltose, maltotriose
β-Amylase (exo) α-1!4 glucose Inverting β-Maltose
α!β
Pullulanase (endo) α-1!6 glucose Retaining Acts on pullulan to give maltotriose and
α!α on starch to give linear dextrins
Glucoamylase α-1!4 Inverting β-Glucose
(α-1!6) α!β
glucose
Cyclomaltodextrin α-1!4 glucose Retaining Acts on cyclodextrins and linear dextrins
transferase α!α to give maltose and maltotriose
Cellulase β-1!4 glucose 1,4-β-dextrins, mixed 1,3–1,4-β-dextrins
Invertase β-1!2 fructose Retaining Glucose and fructose
β!β
β-Galactosidase β-1!4 Retaining Galactose and glucose
galactose β!β
β-Gluctosidase β-1!4, β-1 Retaining Aglycan and glucose
aglycan, β!β
glucose
Polygalactourinase α-1!4 Inverting Galactouronic acid
α!β
Xylanase α-1!4 xylose Retaining
β!β
Lysozyme α-1!4 Retaining
NAM-NAG α!α
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 23
Fig. 2.2 Effect of pH, calcium ion, and salts on α-amylases: (a) pancreatic, (b) Bacillus subtilis
α-amylases, (c) effect of different salts on α-amylase activity (A: No salt, B: 0.04 M NaCl, C:
0.04 M bromide, D: 0.04 M iodide, E: 1.0 M nitrate, F: 1. 0 M chlorate), effect of β-amylase (d) and
α-amylase (e) on viscosity and reducing sugar content during starch hydrolysis. (From Wong &
Robertson, 2007; Saini et al., 2017)
Starch Hydrolysis
Ordinary applications include preparation of starch syrup, dextrose, liquor, brewing,
and bakery. α-Amylase is utilized economically in the liquefaction of starch to create
dextrins which are further saccharified by glucoamylase to yield glucose feed stock
for corn syrup, fuel ethanol, or alcoholic beverages. Heat stable α-amylase in the
presence of calcium ion and a 105 degree centigrade is used to produce a 30-40%
starch slurry.
This yields a mixture of linear and branched dextrins (maltodextrins) with extent
of hydrolysis ranging from 8 to 15 DE (DE: dextrose equivalence). The product of
this reaction is used for the production of 15–40 DE maltodextrins (corn syrups, used
as thickening, bulking, and viscosity building), sweetener production, and a 95–98%
glucose syrup (95 DE) in which other enzymes such as glucoamylase (which is often
used as an immobilized enzyme column), with or without pullulanase, might also be
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 25
used. The glucose syrup can then be treated with an immobilized glucose isomerase
column to generate a high-fructose corn syrup of 42% fructose (52% glucose)
(Wong & Robertson, 2007; Van der Maarel et al., 2002).
In future, improving starch handling and change will zero in on broadening
optimum pH (to pH 4–5), heat stability and decreasing Ca2+ prerequisite of
α-amylase, thereby improving starch processing and transformation, and improving
the capacity to process crude starch by β-amylases. For all enzymes included,
improving heat stability is of significance as this will increase efficiency in handling
just as advancing a single-step process. Likewise, improvement of determinants of
selectivity is also important.
2.2.1.2 b-Amylases
β-Amylase is an exoglucanase that catalyzes the hydrolysis of 1,4-α-D-glucosidic
bonds in polysaccharides to separate progressively maltose units from the nonreduc-
ing end of α-1,4-glucans like starch and glycogen. Sweet potato contains a bountiful
β amylase which represents 5% of the total soluble proteins in the tuber; while only
little α-amylase is available. The activity of this endogenous catalyst brings about
production of maltose which renders a pleasant sweet flavor to potato (Thacker et al.,
1992).
2.2.2.1 Introduction
Pectins or pectic substances are collective names for a group of closely associated
polysaccharides present in plant cell walls, where they contribute to complex
physiological processes like cell growth and cell differentiation and so determine
the integrity and rigidity of plant tissue. They also play an important role in the
defense mechanisms against plant pathogens and wounding. Commercial pectins are
polymers of α-D-galactopyranosyluronic acids with various contents of methyl ester
groups and are obtained from citrus peel and apple pomace. Pectins have a unique
ability to form spreadable gels in the presence of sugar and acid or in the presence of
calcium ions and are used primarily in these types of applications. By definition,
preparations in which more than half of the carboxyl groups are in the methyl ester
form are classified as high-methoxyl (HM) pectins, the remainder of the carboxyl
groups will be present as a mixture of free carboxyl group and their sodium salt.
Preparations in which less than half of the carboxyl groups are esterified with
methanol are called low-methoxyl (LM) pectins. The percentage of carboxyl groups
is the degree of esterification (DE) or degree of methylation (DM) (Benen et al.,
2003; Schols & Voragen, 2003; Yapo, 2011).
The pectin degrading enzymes or pectic enzymes are classified into three gen-
eral groups, namely polygalacturonase, pectate and pecyin lyases, and pectin methyl
esterase. The specific responses brought about by these three pectinase activities are
displayed in Fig. 2.3. The depolymerizing enzymes are hydrolases and lyases. The
methylesteraseases hydrolyze the methylesterase at O6 of a
galacturonicacid residues, though acetylesterases hydrolyze the acetylester at O2
or potentially O3 of a galacturonic acids. These enzymes are commonly found in
plants and microorganisms (particularly in fungi like A. niger) and have been
purified and widely studied (Benen et al., 2003).
Pectic enzymes are normally present in many vegetables and fruits (endoge-
nous enzymes), yet they are also added as processing aids as exogenous enzymes.
In higher plants, especially pectin methyl esterase and polygalacturonase, both endo-
and exo-asplitting are present. Pectin acetylesterase and pectate lyase have been
found in citrus products of plants. The endogenous enzymes are expected to assume
significant parts in plant improvement and during maturing. The cutting edge
genomic approach will empower us to more readily comprehend their functions in
plants in various isoforms and their role in plant improvements. Taking into account
that pectic enzymes alone record for around one fourth of the world’s food enzymes
production, one can securely conclude that the technological innovations and
applications designed to reduce economical costs and increase the productivity of
these enzymes will be of great value (Alkorta et al., 1998; Whitaker, 1990).
2.2.2.2 Polygalacturonases
Polygalacturonases hydrolyze the α-1,4-D-galacturonosidic linkage (Fig. 2.3).
Whereas endo-polygalacturonases hydrolyze the polymer substrate randomly, the
exo-polygalacturonases are confined to cleave off galacturonic acid monomers or
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 29
Fig. 2.3 Site of action and reaction mechanism of pectin-degrading enzyme. (From Whitaker,
1994)
digalacturonides from the nonreducing end. The enzymes, as found for the pectate
lyases, use both polygalacturonic acid and low to moderate degree of esterification of
galactouronic acids as their substrate (Benen et al., 2003). Polygalacturonases are
widely distributed in higher plants and are believed to contribute to fruit softening of
pears, peaches, and avocado.
These enzymes assume significant parts in, food technology, for example, vege-
table and fruit juice extraction, such as, apples, grape wine, strawberries, grapes,
raspberries, and so on, degumming of plant fiber, waste water treatment, oil extrac-
tion, coffee and tea aging, paper and pulp industry, and animal feed. To clarify fruit
juices, a combination of amylases and pectinases is utilized. The time of filtration is
decreased up to half. Tomato is a rich source of polygalactouronase. Brilliant
practices are utilized in the tomato processing to heat inactivate the enzymes momen-
tarily (“hot break” process) to prepare thick juice liked by the purchaser and the high-
consistency concentrated juice (tomato pase) utilized for sauces, soups, catsup, and
comparable items. At the point when tomatoes are utilized for color and flavor, and
consistency is obtained by different agents like starch, thiner, “cold break”, juice is
the beginning material for paste production. A holding time is applied
30 M. Aminlari
2.2.3 Cellulases
Cellulose is the most plentiful biopolymer on the planet. It is mostly created in higher
plants in which it shapes the inflexible skeleton of the plant. Cellulose is a homopol-
ymer comprising of up to 1000 β-1,4-linked-anhydroglucopyranoside units. Single
glucose polymers are packed onto one another to shape an exceptionally crystalline
fibrillar material in which the singular cellulose chains are held together by hydrogen
bonds. Cellulose microfibrils likewise contain some amorphous areas, the level of
which depends on the source. The most crystalline cellulose is produced by algae,
and the least crystalline by plants (Klemm et al., 2005).
Cellulases belongs to the group of β-glucan hydrolases that can break down
cellulose. Several microorganisms, including filamentous organisms, yeast, and
microbes existing in the digestive tract and colon of monogastric, can hydrolyze
cellulose to oligosaccharides and in the long run to glucose. Ruminants can
completely degrade cellulose in their rumen by the wide range of microorganisms
present. Cellulases have generally been classified into two specific groups
endoglucanases and exoglucanases, which act in the middle or at either ends of the
cellulose chain, respectively (Tenkanen et al., 2003). Cellulases are produced by
fungi, bacteria, protozoans, and animals. There are additionally a few reports on the
presence of cellulases in plants. Up to this point, just the endoglucanases (1,4-β-D-
glucan-4-glucanohydrolase) have been found in plants. This enzyme catalyzes
random cuts in cellulose chains, subsequently creating more shorter cello-oligomers,
which can be additionally degraded by exoglucanases. Endogenous plant
endoglucanases have been recommended to assume a part in plant development,
for example, in fruit maturing and leaf abscission, or in the rearranging of
polysaccharides in developing cells. The producion of microbial cellulase in trans-
genic plants, for example, sugarcane and corn, has been reported. These advances
can offer one potential utilization of plant biomass as biofuel harvests or remaining
horticultural waste as a sustainable fuel source (Hefferon, 2017).
Cellulases are not among the central enzymes used in the food industries. They
are, nevertheless, applied in treatment of grain-based beverages and food
assortments like beer and bread. Cellulases are used in rather immense totals in
animal feed (Zhang & Zhang, 2013). Cellulase has a wide scope of utilizations in
industrial biotechnology and is the second most utilized modern enzyme after
protease. Cellulases are utilized in the textile industry, in cleansers, pulp and paper
industry, further developing edibility of animal feeds, and food industry. In numer-
ous food applications, cellulases are not utilized alone, but are added to help the
activity of other enzymes. In collaboration with enzymes, such as pectinases and
hemicellulases, cellulases disrupt the underlying rigidity of the plant cell walls and
upgrade the extraction efficiency of the molecules of interest. Thus, they are utilized
in any process of handling plant-based materials. Cellulases are utilized in wine
and production of fruit juice to facilitate the maceration, and, the extraction of color
and flavor compounds, fermentable sugars and the extraction of juices. Macerating
enzymes mix containing cellulases are likewise utilized in fruit juice production.
Furthermore, the utilization of these enzymes offers benefits in the treatment of
32 M. Aminlari
waste of the fruit juice industry by further developing the extraction yield and the
general process efficiency. In baking industry, cellulase is utilized to degrade gums
in the dough, improving bread structure. Added enzymes are utilized progressively
for improving the processes and final quality of products. Obviously, the biggest
modern utilization of cellulases is outside the food and feed area but in material
handling of fibers made out of cellulose (Zhang & Zhang, 2013; Autio et al., 1996).
Cellulases are likewise utilized for the extraction of nutraceuticals from plants.
Nutraceuticals are compounds from normal sources with extra medical advantages,
other than a nutritious agent (Fernandes, 2018). Cellulases and hemicellulases can be
utilized for the separation of pectin from citrus fruits to separate the cell wall,
expanding the pectin yield. Cellulases can be utilized alone or in blend with other
cell wall degrading enzymes in all processes in which significant mixtures like juice,
oil, polysaccharides, protein, and so on are removed from the plant material
(Laurikainen et al., 1998).
2.2.3.1 b-Glucosidases
β-Glucosidases are enzymes with great practical significance to natural systems.
These are divided in various glycoside hydrolase families in light of their catalytic
mechanism and amino acid sequences. Most investigations completed on
β-glucosidases are centered around their applications rather than their endogenous
capacity in the their natural environment (Singh, 2016). In a large number of leafy
foods tissues, significant flavor compounds are glycosylated. β-Glucosidases cata-
lyze the hydrolysis of alkyl and aryl β-glycosides disaccharides and short-chain
oligosaccharides. β-Glucosidases have an extraordinary potential to be utilized in
different biotechnological processes from freeing flavours, fragrances, and isofla-
vone aglycons to the biosynthesis of oligosaccharides and alkylglycosides. The
utilization of glycosidases to deliver flavor compounds from glycosidic compounds
was at first analyzed in wines. β-Glucosidases are omnipresent, generally dispersed
in nature, and can be found in microorganisms, plants, and animals (Romero-Segura
et al., 2009; Zhu et al., 2017).
One more attribute of glucosidases critical to food processing and quality is the
presence of cyanogenic glycoside in many plants that can potentially produce
hydrogen cyanide during preparation or by eating. The cyanogenic glucoside
linamarin in cassava roots and leaves (widely used as food staple in tropical areas
of Africa, Asia, and South America), lima beans, bitter almonds, and flax seeds are
examples of these glycosides. There are roughly 25 cyanogenic glycosides referred
to, for example, linamarin in cassava and lima beans; dhurrin in sorghum; amygdalin
in almonds, peaches, and apricot pits (Hughes, 1999). The enzyme rhodanese is a
ubiquitous enzyme active in all living organisms from bacteria to man.
It is a multifunctional enzyme, yet appears to have important role in cyanide
detoxification. This enzyme is likewise broadly present in plants (Chaudhary &
Gupta, 2012). The primary source of this enzyme was reported to be the liver of
animals. Nonetheless, broad examinations on the distribution of rhodanese in our
research have exhibited that different organs, specifically the gastrointestinal system,
is more prominent source of rhodanese, which show that cyanide delivered from
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 33
2.2.3.2 Xylanases
After cellulose, xylan is the most abundant polysaccharide in nature. Xylans are a
major hemicellulose component, and together with cellulose, they comprise the bulk
of the cell wall material in botanical products. Xylanases are β-retaining
glycosidases capable of hydrolyzing linear β-1,4-linked polymers of xylose (with
various substitutive groups such as arabinose). Multiple isoforms exist and these
enzymes can be endo- or exo-acting (endo-acting are more important in foods). The
xylanases are ubiquitous in nature and their presence is observed diversely in a wide
range of organisms (Biely, 2003). They are in plants (especially important in
cereals), bacteria, and fungi, and they typically range in molecular mass from
16 to 40 kDa. Xylanolytic enzymes also occurring in plants participate in the process
of cell wall extension, cell division, seed germination, and other morphological and
physiological events in plants.
During germination, endogeneous xylanases catalyze hydrolysis of these
polysaccharides to eliminate the actual boundary imposed by cell wall on the free
dispersion of starch and protein degrading enzymes through the germinating
grain. Enzymes responsible for breaking down hemicellulose polysaccharides are
likewise reported to be available in wheat grain and wheat flour (Kumar et al., 2017).
Xylanase enzymes are beneficial to food handling by depolymerizing water-
unextractable arabinoxylan pentosans, which have high water-holding capacity
This increases the viscosity and thickness of the dough, gluten strength, and final
bread volume. The mixed xylanases are especially significant in the preparation of
frozen dough. Endoxylanases are one of the hemicellulases that are utilized in fruit
and vegetable processing. Xylanases are likewise utilized in brewing to lower
viscosity of the wort in preparing, easeing partition/filtration steps, decreased haze
formation, and marginally further developed process yields (Kumar et al., 2017;
Bhardwaj et al., 2019).
2.2.3.3 Glucoamylase
Glucoamylase catalyzes the hydrolysis of α-(1,4), α,β-(1,1), α-(1,6), α-(1,3), and
α-(1,2) glycosidic connections between adjoining glucosyl residues, arranged by
their decreasing rates of hydrolydid. It starts hydrolysing from nonreducing end of
starch-based substrates to liberate glucose. Glucoamylas assume a significant func-
tion in food processing to saccharify starch in cereals and produce glucose syrups
from maltodextrins after the activity of α-amylase. Glucoamylas is produced by
certain eubacteria, some archaea, various yeasts, and numerous filamentous fungi.
Despite the fact that there are reports of animal and plant glucoamylas, these seem,
by all accounts, to be essentially various enzymes with analytic properties that
crossover those of genuine glucoamylas. Thus, and notwithstanding wide utilization
of this enzyme in food industry, this catalyst won’t be additionally discussed in this
section.
34 M. Aminlari
Proteases are found in all living life forms (creatures, plants, and
microorganisms). They are fundamental for separating of proteins to peptides and
amino acids. In the human digestive system, the proteolytic enzymes pepsin (stom-
ach) and several proteolytic enzymes in small intestine (trypsin, chymotrypsin,
carboxypeptidases, leucine aminopeptidases, tripeptidases, and dipeptidases) con-
vert the proteins of ingested food varieties to amino acids.
Proteases are by far the best-portrayed enzymes as far as their fundamental roles
in the human digestion and early commercialization (the originally normalized calf
rennet for cheddar making was promoted in 1874) is concerned (Parkin, 2017).
Peptidases breakdown food proteins in situ or are added exogenously to cause
protein change.
2.3.1 Specificity
In the protease active site, S1, S10 , . . . refer to the amino acids that provide the
chemical and spatial environment that complements the substrate amino acids
referred to as P1, P10 , . . . . Arrow shows the scissile peptide bond. These sites
have been mapped and characterized for many proteases (Parkin, 2017; Vizovišek
et al., 2018).
2.3.2 Classification
There are four distinct kinds of proteases, specifically, the serine proteases, the
cysteine proteases, the aspartic proteases, and the metalloproteases, in light of the
active site group of the enzyme engaged with the nucleophilic attack at the carbonyl
carbon of the scissile peptide bond of the substrate. These are portrayed below
(Whitaker, 2003).
tree, Carica papaya (papaya), actinidin from kiwi fruit, ficin from fig latex, bromelain
from pineapple, and lysosomal cathepsins from animal tissues. Among them, papain,
bromelain, and ficin address 5% of the worldwide sale of proteases (Illanes, 2008;
Verma et al., 2016).
The most studied cysteine protease is papain, which will be examined later in
light of its significant uses in the food industry for chill proofing of brew and meat
tenderization, and for synthesis of peptides and other organic compounds. An
interesting cysteine protease in muscle is calpain (numerous isoforms) that is
activated by Ca2+ and plays a part in after death tenderization of muscle. Commonly,
the enzymes of this group are 24–35 kDa in mass, are active at pH 6.0–7.5, and can
endure temperatures up to 60–80 C (due in some extent by three disulfide bonds)
(Whitaker, 2003; Otto & Schirmeister, 1997).
2.3.2.4 Metalloproteases
A large group of the metalloproteases are exopeptidases. These are the
carboxypeptidases A and B, glycyl-glycine dipeptidase, carnosinase (work on
β-alanyl-L-histidine and related compounds), amino-acyl-histidine hydrolase, and
cytosolic aminopeptidase (α-amino-acyl-peptide hydrolase). All require Zn2+ as
cofactor. Some of these enzymes, for instance, prolidase (where proline or hydroxy-
proline is the carboxyl terminal residue) and iminodipeptidase (in which proline
or hydroxyproline is the N-terminal residue) require Mn2+. Other metalloprotases are
endo-acting thermolysin (from Bacillus thermoproteolyticus) and the nreutal
endoprotease from Bacillus amyloliquefaciens (Tavano et al., 2018).
Commercial proteases are available at various levels of purity, and some contain
multiple proteolytic agents. Some of the important commercial applications of
proteolytic enzymes are shown in Table 2.3 and are described below (Whitaker,
2003).
(ordinarily meat, milk, fish, wheat, vegetable, vegetable, and yeast sources) might be
exposed to a pretreatment that renders them to some degree of denaturation as this
improves peptidase access and hydrolytic activity. Protein hydrolysates can improve
functional properties, for example, emulsifying capacity, foam capacity and stability,
antioxidative effect, water absorption capacity, and nutritional properties
(Chalamaiah et al., 2012; Thiansilakul et al., 2007; Sinha et al., 2007; Clemente,
2000). Protein hydrolysates constitute an alternative to intact proteins and elemental
formulas in the development of special formulations designed to provide nutritional
support to patients with different needs. The production of extensive protein
hydrolysates by sequential action of endopeptidases and exoproteases coupled
with the development of post-hydrolysis procedures is considered the most effective
way to obtain protein hydrolysates with defined characteristics (Rocha et al., 2009).
Protein levels in production of protein hydrolysates are often 8–10%, provided there
are no limitations on solubility, and the amount of enzyme added is generally 2%
on a protein basis, depending on purity. Protein:enzyme levels are sufficiently high
so that the enzyme reacts at nearly Vmax with limited autodigestion of the enzyme,
although product inhibition by accumulating peptides may attenuate reactivity. The
hydrolysates can be characterized by the degree of hydrolysis (DH), sequence of
produced peptides, functional properties such as solubility, emulsifying properties,
38 M. Aminlari
Cheese Manufacturing
The main gastric enzyme of neonatal ruminants is chymosin or rennet. Chymosin
has low broad proteolytic action, but high milk-coagulating action. Calf chymosin
(rennet) and chymosin substitutes (see beneath) are added to milk to cause the
underlying milk-clotting reaction prompting cheese formation. The rennet coagula-
tion of milk is a two-stage process. The first (essential) stage includes specific
hydrolysis of the PHE105-MET106 bond of κ-casein and creation of “para-casein”
and TCA (trichloroacetic acid)-soluble peptides (glycomacropeptides), while the
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 39
Aspartic protease are present in many plant species and have aspartic acid
residues in their active site. They participate in many activities in plants. Examples
include:
Serine proteases have in their active sites serine residues. Their main role in plants
is almost the same as the aspartic proteases, with some additional features. Serine
proteases are widespread in plants and belong to several taxonomic groups. They are
extracted, purified and characterized from several parts of plants, especially fruits.
Some of the enzymes in this group are (Di Cera, 2009):
3. Ficin from branches latex of figs tree Ficus carica sylvestris and an enzyme from
stem latex of Sideroxylon obtusifolium (Sideroxylon is a genus of trees in the
family Sapotaceae. They are collectively known as bully trees).
4. Actinidin from kiwi (Actinidia chinensis) fruits.
5. Calotropain from latex of crown flower Calotropis gigantean.
proteolysis and meat tenderization. Cathepsins were the main enzymes utilized in
meat tenderization, after which calpain was applied owing to its action in changing
the Z-line thickness found in post-mortem, despite the fact that it was not at first
connected with meat tenderization. Calpains are calcium-dependent proteases that
break down myofibrillar proteins. Calpastastin, on the other hand, inactivates
calpains, decreases the myofibrillar degradation, and hence lessens the tenderness.
Calpastatin effect is finished after being inactivated by cooking. The amount of the
enzymes changes among species, which affect the degree of meat tenderness,
because of expanded or diminished proteolysis of myofibrillar proteins (Lian et al.,
2013).
Meat tenderness relies upon the kind of muscle, pre- and post-mortem variables,
and after death pH and temperature. The research for significant proteases with
unique specificity for industrial applications is consistently a continued challange.
Proteolytic enzymes from plant sources have gotten extraordinary attention for being
active over a wide range of temperatures and pH’s. Treatment by proteolytic
enzymes is one of the most well-known techniques for meat tenderization. The
utilization of exogenous proteases for meat tenderization is a rather modern strategy
to further develop meat quality. There are few exogenous proteolytic enzymes, plant
proteases (papain, bromelain, ficin, and actinidin), and proteases from Aspergillus
oryzae and Bacillus subtilis, which have been considered as generally regarded as
safe (GRAS) for use in the meat. Papain and other sulfhydryl endopeptidases
(bromelain and ficin) are applied to muscle or meats that don’t turn out to be
sufficiently tendered during post-mortem aging. These enzymes are powerful in
this application since they can hydrolyze collagen and elastin, connective tissue
proteins that cause toughness in meat. These proteolytic enzymes are blended in the
meat to break down the proteins in muscle and hydrolyze collagen and elastin, which
helps in meat tenderization. Papain and bromelain are the most regularly involved
plant enzymes for meat tenderization. As meat tenderizers, proteolytic enzymes are
the most ideal for breakdown of collagen and elastin in connective tissue at some-
what low pH and low temperature. The structure of myosin and actin fibers is
impacted by the plant proteases. The tenderness of meat is deteremined by enzyme
activity assessment, myofibrillar fragmentation index, hydroxyproline estimation,
and examining by electron microscopy. The ideal meat tenderizer would be a
proteolytic enzyme with specificity for collagen and elastin in connective tissue, at
the generally low pH of meat, which would act either at the low temperature at which
meat is stored or at the high temperature accomplished during cooking (Arshad et al.,
2016; Lian et al., 2013).
Papain
Papain is a nonspecific thiol protease and the significant protein constituent of the
latex in the tropical plant Carica papaya. It is an important plant protease. The latex is
obtained by scoring, and afterward permitting it to dry on the fruit, and a crude
material is resulted. In the food industry, papain is regularly utilized for meat
tenderization, production of protein hydrolysate, and the clarifying of juice and
beer. Papain is also used in the baking and dairy industries (in cheese production),
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 43
and for the extraction of flavor and color compounds from plants. Papain is
prepared by decersing contaminants and further extraction. The enzyme has high
thermal and pressure resistances, requiring exceptional process conditions for suffi-
cient inactivation (to accomplish 95% inactivation of papain at 900 MPa and 80 C,
22 min of processing is required). The physiological job of papain in plants is to
protect them from insects. Papain tends to over tenderize the meat surface, making it
“soft”. Papain is utilized at a portion of 300 units/kg to release free amino acids in
dry fermented frankfurters. The three-dimentional structure for papain is reported.
Wide range enzymatic activity has been shown by papain in the pH range 5.9–7.5
and the temperatures of 70–90 C with optimum of 65–75 C. Under these
conditions, papain has superb activity in hydrolysis of myofibrillar proteins and
moderate impact on hydrolysis of collagen (Istrati, 2008; Gokoglu et al., 2017).
Papain can be utilized in the fruit juice and beer clarification. In beer, an
imperfection alluded to as chill-haze might be brought about by the combina-
tion (complexing) of tannins and proteins. Papain has for some time been utilized
to hydrolyze protein and limit cloudiness, despite the fact that bromelain and ficin
just as other bacterial and fungal proteases may now be utilized for this reason. The
endopeptidase is added after aging and preceding final filtering. Papain is finally
inactivated by pasturization, and unnecessary activity of papain might cause loss of
foam stability. Controlled or evaluated proteolysis in brew is important, since some
remaining protein is important to preserve specific quality attributes (Mosafa et al.,
2013).
Bromelain
Bromelain (or bromelin) enzymes are present in huge amounts in fruits, leaves, root,
and stems of the Bromeliacea family, of which pineapple (Ananas comosus) is the
best known. The juice of various pieces of the plant contains bromelain in soluble
form. Its high proteolytic action has made a wide interest in various applications,
principally in tenderization, food, detergents, and the textile industry. Bromelain
is commercially accessible in powdered form. It is assessed that 95% of the
utilized proteases in the United States are derived from plant proteases like papain
and bromelain, though microbial tenderizers are not utilized broadly. Reverse
micellar extraction is utilized for isolation and purification of bromelain from
pineapple. There is high bromelain recovery and purification in this strategy. This
enzyme, as different proteases, breakdown myofibrillar proteins and collagen, caus-
ing overtenderization of meat. Bromelain can be utilized in beef at 10 mg/100 g meat
and ideal tenderization happens after 24 h at 4 C. Following this time span, the
enzyme can be inactivated by heating at 70 . pH range for enzymatic activity for
bromelain is 4–7 and optimum pH is 5–6. The range of temperature for activity is at
50–80 C and optimum is 65–75 C. Under these conditions, bromelain has moder-
ate hydrolysis activity of myofibrillar proteins and good effect on hydrolysis of
collagen (Arshad et al., 2016; Gokoglu et al., 2017).
44 M. Aminlari
Ficin
Due to the problems raised by the use of animal or microbial recombinant proteases,
the use of ficin is becoming increasingly popular. Ficin is an outstanding example of
proteases from plants. This enzyme can be applied for protein hydrolysis, the
production of bioactive peptides, meat tenderization, milk coagulations in cheese
making, or peptide synthesis. Ficin does have huge potential and brilliant prospect in
the near future. Ficin is a sulfhydryl or cysteine protease commonly obtained from
Ficus carica (fig tree) that enhances the solubility of muscle proteins (Englund et al.,
1968; Morellon-Sterling et al., 2020). Ficin has a molecular weight of 44.5 kDa and
shows maximal activity at pH 5.0–9.0 and optimum pH is 7.0. The enzyme is fully
active at 45–75 C with optimum temperature of 60–70 C. Under these conditions,
ficin has moderate activity in hydrolysis of myofibrillar proteins and excellent effect
on hydrolysis of collagen. These properties make ficins a beneficial class of plant
proteases for use. We used ficin to tenderize beef used for manufacture of sausages.
Results indicated that solubility of meat proteins increased and SDS-PAGE results
showed the disappearance of several protein bands in ficin-treated meat. Ficin-
tenderized meat substantially improved water-holding capacity and emulsion stabil-
ity. The results of this study indicated that some quality attributes of meat products
can be improved by enzymatic modification of protein sources in the manufacture of
meat products (Ramezani et al., 2003).
Actinidin
Actinidin is a novel sulfhydryl protease extracted from gooseberry or the kiwi fruit.
Actinidin is the predominant enzyme in kiwifruit and can play a role in aiding the
digestive process. It has a molecular weight of 32 kDa. It is used commercially in
meat industry to tenderize meat. The ability of pre-rigor infusion of kiwifruit juice to
improve the tenderness of lamb was investigated. It was shown that actinidin in kiwi
fruit juice enhances proteolytic activity, resulting from the infused kiwifruit juice in
carcasses, and is associated with significant degradation of the myofibrillar proteins,
appearance of new peptides, and activation of m-calpain during post mortem aging.
Thus, kiwifruit juice is powerful and easily prepared meat tenderizer, which could
contribute efficiently and effectively to the meat tenderization process (Boland,
2013; Morton et al., 2009).
Actinidin has many applications in the food industry because of its advantages
over other plant proteases such as papain and ficin. Actinidin shows mild tenderizing
activity even at high concentrations, preventing surface mushiness. It has a relatively
low inactivation temperature (60 C), which makes it easier to control the tenderiza-
tion process without overcooking. A group in our lab studied the effect of actinidin
on beef for use in manufacture of sausage. Actinidin from kiwi fruit was partially
purified by precipitation with ammonium sulfate, followed by DEAE-Sephadex
column chromatography. The effect of purified actinidin on the protein solubility
(nitrogen solubility index [NSI]), water-holding capacity (WHC), texture, and
SDS-PAGE pattern of beef was studied and the quality attributes of a sausage
product were evaluated. Actinidin significantly increased NSI and WHC of beef;
the highest NSI and WHC (approximately 20% and 8% increase, respectively) was
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 45
observed when beef was incubated with 0.9 unit enzyme/g beef. Texture analysis
indicated increased tenderization (10% decrease in shear force) when slices of beef
were treated with actinidin at 37 C for 2 h. SDS-PAGE results indicated appearance
of several low molecular weight bands (<10 kDa) after treating beef with different
levels of actinidin for 30 or 60 min. Slight changes in protein band in the range of
100–120 kDa and 13–25 kDa were also observed. Use of actinidin-tenderized beef
significantly improved emulsion stability, texture, and organoleptic properties of the
sausage product (Lewis & Luh, 1988). We applied actinidin to tenderize camel meat
and beef and used the tenderized meats in the manufacture of emulsion type
sausages (Gheisari et al., 2008). The properties of sausages made from the meat of
both species were similar. Emulsion satiability, folding, texture, taste, and overall
quality of sausages produced from actinidin-tenderized meat were superior than
untreated samples (Aminlari et al., 2009). The actinidin has less tenderization
property as compared to other traditional plant proteases and still not approved as
GRAS by FDA (Lewis & Luh, 2007; Toohey et al., 2011).
Calpains
Endogenous proteolytic systems are responsible for modifying proteinases as well as
the meat tenderization. Abundant evidence has shown that calpains and calpastatin
(CAST) have the closest relationship with tenderness in livestock. They are involved
in a wide range of physiological processes including muscle growth and differentia-
tion, pathological conditions and postmortem meat aging (Lian et al., 2013). Meat
tenderness undergoes changes after slaughtering due to the activity of the endoge-
nous calpains and calpastatin. These calcium-dependent proteases degrade the
myofibrillar proteins. The calpain system consists of three members—m-calpain,
μ-calpain, and calpastatin, which is the calpain-specific endogenous inhibitor. Both
m-calpain and μ-calpain are cysteine proteases, and their proteolytic activity is
affected by oxidation, which can influence the quality of fresh meat. In the presence
of calcium, calpains autolyze, and this autolysis is indication of their proteolytic
activation during postmortem changes in muscles (Zhang et al., 2013). Calpain is an
important enzyme that is chiefly used for degradation of myofibrillar proteins. It also
aids in meat tenderizing and improves water holding capacity during postmortem
aging.
2.3.3.4 Conclusion
In meat, tenderness is the most important factor associated with meat palatability and
consumer satisfaction. Different plant proteases like papain, bromelain, actinidin,
and ficin have been used for tenderization of meat and meat products. These
enzymes are effective in this application because they can hydrolyze collagen and
elastin, connective tissue proteins that cause toughness in meat. Antemortem appli-
cation of enzyme is possible, as a fairly pure solution in saline can be injected
intravenously into animals 2–10 min before slaughter, sometimes after stunning; this
helps distribute the enzyme throughout the muscle tissues. Injection of inactivated
papain (disulfide form) obviates any discomfort among animals, since the enzyme
46 M. Aminlari
becomes activated by the reducing conditions that soon prevail postmortem (Arshad
et al., 2016).
Bioactive peptides have been defined as specific protein fragments that have a
positive impact on body functions or conditions and may influence health (Sánchez
& Vázquez, 2017). In recent years, peptides with known sequences have been
identified which have been shown to contain biological activities such as opiate-
like, mineral binding, immunomodulatory, antimicrobial, antioxidant,
antithrombotic, hypocholesterolemic, and antihypertensive actions. These bioactive
peptides benefit the human physiological system through the cardiovascular, ner-
vous, gastrointestinal, and immune systems (Choi et al., 2012). Most of the bioactive
peptides are inactive in the native protein sequences and become active only when
released when the parent proteins are hydrolyzed in vivo by the action of proteolytic
enzymes during digestion or in vitro by intentionally added proteases (Korhonen &
Pihlanto, 2006). The activity of these peptides is based on the inherent amino acid
composition and sequence. Bioactive peptides usually contain 3–20 amino acid
residues per molecule (Bhat et al., 2015), but in some cases may consist of more
than 20 amino acids. These peptides may be used as components of functional foods
or nutraceuticals because of their health-enhancing potential and safety profiles.
There is increasing commercial interest in the production of bioactive peptides
from various sources. Some of rich sources of bioactive peptides include milk and
egg, meat of various species of animals, fish, many plants including soy bean,
chickpeas, rice, and many other food-proteins and nonconventional protein sources
(Mazorra-Manzano et al., 2018; Sabbione et al., 2016).
Proteolytic enzymes have the ability to modify proteins through limited or
extensive cleavage, releasing free amino acids, peptides, or polypeptides with
physicochemical properties different from the original protein. Proteolytic enzymes
from proteolytic system of starters, proteases endogenous to food, or added enzymes
(e.g., rennet) differ in their specificity and therefore in their capacity to release
bioactive sequences (Korhonen & Pihlanto, 2006; Sabbione et al., 2016). Most
hydrolytic processes use one protease at a time; however, a combination of two
enzymes with different specificity has also been explored. Sometimes, proteins
partially hydrolyzed are further treated with pepsin and trypsin to simulate gastroin-
testinal digestion. Other proteases widely used for bioactive peptides’ production
include commercial preparations such as Alcalase, Neutrase, Flavourzyme,
Thermolysin derived from bacteria and fungi (Rui et al., 2012), and ficin (Shahidi
et al., 2018).
We have studied the production of peptide fragments produced from goat’s milk
whey proteins using trypsin and ficin and evaluated the bacterial growth inhibitory
activity of peptides. Goat’s milk whey proteins were subjected to enzymatic hydro-
lysis and peptides were purified by ultrafiltration followed by reverse-phase high-
performance liquid chromatography (RP-HPLC). Growth inhibitory activities of
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 47
hydrolysates ranged from 4.67% to 87.46% for E. coli and 3.03% to 98.63% for
B. cereus. Among all peptide fragments, permeate containing 3 kDa peptides
produced by trypsin showed maximum inhibition against Gram-positive and
Gram-negative bacteria. This fraction was further purified by HPLC. Fourteen
peptide fractions were collected and evaluated for their growth inhibitory activities.
Two fractions showed the highest growth inhibitory activities with MIC50’s of
383 8 and 492 10 μg/mL against E. coli and B. cereus, respectively. In a similar
study, we used trypsin and ficin to generate antibacterial peptides from goat milk
caseins. The peptide obtained by ficin with MW of <3 kDa showed the highest
antimicrobial activity and was selected for further purification by reversed-phase
high-performance liquid chromatography (RP-HPLC). Twenty-seven peptide
fractions were separated, and their antimicrobial activities were evaluated. The
results showed that one of the fractions (No. 14) possessed the highest activity
against Escherichia coli and Bacillus cereus (Esmaeilpour et al., 2016, 2017). The
authors of both papers suggested these novel antibacterial peptides can potentially
replace synthetic food preservatives in food industries.
In another research project, the yeasts Kluyveromyces marxianus and Saccharo-
myces cerevisiae protein hydrolysates were prepared by trypsin and chymotrypsin
hydrolysis and the peptides purified by reverse-phase high-performance liquid
chromatography (RP-HPLC). The antioxidant and ACE (angiotensin-converting
enzyme) inhibitory activities of the generated peptides were determined. From
K. marxianus two new peptides, LL-9, MW 1180 Da and VL-9, MW 1118 Da,
were identified. These peptides were sequenced and their functional properties
studied. Both peptides exhibited significant ACE inhibitory activity (IC50 of
22.88 mM for LL-9 and 15.20 mM, for VL-9). Molecular docking studies revealed
that the ACE inhibitory activities are due to interaction with the His513, His353,
Glu281 and Tyr520, Lys511, Gln281 pockets of ACE by LL-9 and VL-9, respec-
tively. In the case of S. cerevisiae, a fraction with molecular weight of <3 kDa
exhibited the highest activity. RP-HPLC resolved this fraction into five fractions,
one of which (fraction F3) with amino acid sequence of Tyr-Gly-Lys-Pro-Val-Ala-
Val-Pro-Ala-Arg (MW: 1057.45 Da) exhibited ACE inhibitory
(IC50 ¼ 0.42 0.02 mg/mL) and antioxidant activities (26.25 0.13 μM TE/μg
protein). Taken together, the results of these studies showed that K. marxianus and
S. cerevisiae proteins contain specific peptides in their sequences which can be
released by enzymatic hydrolysis.
These peptides have excellent bioactive properties that can potentially replace the
antioxidant and antihypertensive agents with chemical origin (Mirzaei et al., 2015,
2017).
The use of plant proteases in the production of bioactive peptides is still scarce.
Cysteine proteases such as papain, ficin, and bromelain are currently the most used
plant proteases. However, serine proteases such as zingibain, cucumisin, and
actinidin obtained from ginger rhizome, melon, and kiwifruit, respectively, are
three new emerging plant proteases which have been considered recently (Nafi
et al., 2013).
48 M. Aminlari
The search for novel specialized proteolytic enzymes with preference for specific
peptide bonds for the selective release of bioactive peptides requires further study in
order to improve process efficiency. The various plant proteases, the protein
substrates used, and the bioactive properties of peptides produced and their potential
to prevent or treat disorders such as hypertension, diabetes, obesity, and cancer have
been extensively reviewed (Mazorra-Manzano et al., 2018).
2.4.1 Lipases
Another major use of lipases is production of “structured lipids” in which the fatty
acyl groups are rearranged by lipases to yield a novel distribution along sn-glycerol
to create high-value lipids from low-value acylglycerol (Gunstone, 1999).
Lipases are used in baking industry and are added to bread doughs to supplement
endogenous cereal grain lipases and are added as dough improvers. This functional-
ity results in increased bread volume, more uniform crumb and air cell size, and
lesser tendency to stale, hydrolysis of lipids, thereby producing mono- and
diacylglycerolipids which function as emulsifying agents in the dough (Sonnet &
Gazzillo, 1991; Parkin, 2017).
2.4.2 Lipoxygenases
2.4.3 Phospholipases
2.5.1 Catalase
The enzyme catalase is known to catalyze the breakdown of hydrogen peroxide into
oxygen and water. It is an oxidoreductase enzyme as it plays a crucial role in
quenching the reactive oxygen species (ROS), i.e., hydrogen peroxide, often pro-
duced as a by-product of aerobic respiration. Hence, it acts as an antioxidant and
protects the cell against oxidative stress (Kaushal et al., 2018).
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 51
Biogenic amines and polyamines are present in variety of foods, such as fish, meat,
cheese, vegetables, and wines. These are organic bases with aliphatic, aromatic, and
heterocyclic structures. They are produced by decarboxylation of amino acid
substrates catalyzed by enzymes from contaminating bacteria. The most common
biogenic amines found in foods are histamine, tyramine, cadaverine,
2-phenylethylamine, spermine, spermidine, putrescine, tryptamine, and agmatine
(Ruiz-Capillas & Herrero, 2019). The formation of biogenic amines in food can
result in consumers suffering allergic reactions, characterized by difficulty in breath-
ing, itching, rash, vomiting, fever, and hypertension. Several methods for prevention
of the production or removal of biogenic amine in food have been practiced,
primarily by limiting microbial growth through chilling and freezing. However,
for many socioeconomic and technological reasons, such approaches are not always
practical. Therefore, alternative measures to prevent biogenic amine formation in
foods or to reduce their levels once formed are needed. One approach might be
application of enzymes which metabolite these amines (Naila et al., 2010).
Amine oxidase catalyzes the reaction with mono-, di-, or polyamine as substrate
(Ito & Ma, 2003):
The activity of these enzymes has been detected in Arthrobactor, yeast Candida
boidinii, Asp. niger, pea seedling, bovine plasma, and bovine lung. In human and
animals, the enzyme plays an important role for the metabolism of biogenic
monoamines in the central nervous system and peripheral tissues. Pea seedling
amine oxidase, a copper containing diamine oxidase, catalyzes the oxidative
52 M. Aminlari
2.5.4 Lysozyme
In recent years, consumer demand for ‘natural’ foods has driven development of
products without additives. In order to meet this demand, much attention and interest
have been directed towards identification and application of naturally made
compounds such as antimicrobial agents, in food and pharmaceuticals (Branen &
Davidson, 2004). Some naturally occurring proteins such as lactoperoxidase,
lactoferrin, and lysozyme have received much attention and are being considered
as potential antimicrobial agents to replace the currently used synthetic food
preservatives (Demain, 2009).
Lysozyme, also known as muramidase or N-acetylmuramide glycanhydrolase, is
an antimicrobial enzyme found in many different sources, from viruses to
vertebrates, and has been subjected to extensive scrutiny, both as a protein model
and a natural antimicrobial and pharmaceutical agent. It forms part of the innate
immune system. Lysozyme is abundant in secretions including tears, saliva, human
milk, and mucus. Chicken egg white has the highest content of lysozyme
(it constitutes 3.5% of the total egg white proteins), from which this enzyme is
purified and is commercially produced (Proctor & Cunningham, 1988). Lysozyme is
a glycoside hydrolase that catalyzes the hydrolysis of 1,4-beta-linkages between N-
acetylmuramic acid and N-acetyl-D-glucosamine residues in peptidoglycan, which is
the major component of gram-positive bacterial cell wall. In general, lysozyme
shows in vitro antimicrobial activity against some Gram-positive bacteria such as
Staphylococcus aureus, Micrococcus luteus, Bacillus stearothermophilus, and Clos-
tridium tyrobutyricum, but little action against Gram-negative bacteria (Cunningham
et al., 1991). Gram-negative bacteria, including foodborne pathogens, resist lyso-
zyme due to steric hindrance posed by the outer LPS layer, hence lysozyme cannot
access the peptidoglycan (Ibrahim et al., 1994). Thus, modification of lysozyme,
which can broaden its antibacterial properties against both Gram-negative and
Gram-positive bacteria, can also increase the usefulness of lysozyme (Seo et al.,
2013).
54 M. Aminlari
Table 2.4 Modified lysozymes and their properties (from Aminlari et al., 2014)
Modified lysozymes Effect on functionality
Palmitic acid Enhanced antimicrobial against E. coli (WT-3301)
Short and middle chain Enhanced antimicrobial against G-positive bacteria
saturated fatty acids
Glucose–stearic acid Enhanced activity against E. coli and E. tarda (G8104)
monoester
Perillaldehyde Enhanced activity against E. coli K12 and S. aureus
Cinnamaldehyde Enhanced activity against E. coli and S. aureus
Glucosamine Improved solubility at different pHs and temperatures, increased
heat stability, emulsion activity and stability, and foam capacity
Caffeic acid–cinnamic acid Antimicrobial activity against E. coli (ATCC 8739), decreased
activity against S. aureus (ATCC 6538)
Dextran Enhanced activity against E. coli and S. aureus in cheese
Enhanced activity against E. coli and S. aureus in milk
Treatment of bacterial isolates from cows with mastitis
Preparation of a lysozyme–dextran nanogel
Increased heat stability, higher emulsifying property
Galactomannan Antibacterial emulsifier
Emulsifier, antibacterial against G-negative pathogen E. tarda
Chitosan Enhanced bactericidal action against E. coli K12
Lysozyme-composite film with activity against E. coli,
L. monocytogenes, and S. faecalis
Cellulose Preparing a textile with potential barrier to microbial invasion
Gum Arabic Enhanced activity against E. coli and S. aureus in mayonnaise
Xanthan gum Enhanced activity against E. coli and S. aureus
Dextran sulfate Enhanced activity against E. coli and S. aureus
Barley beta-glucan Enhanced activity against E. coli and S. aureus
Tragacanth Enhanced activity against E. coli, S. typhimyrium, B. cereus, and
S. aureus
Inulin Improved functional properties
Trypsin and ficin digestion Enhanced activity of peptides E. coli and B. cereus dextran-
of conjugated lysozyme
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 55
2.6 Conclusions
References
Aehle, W. (Ed.). (2007). Enzymes in industry, production and applications (3rd ed., pp. 1–12).
Wiley-VCH Verlag GmbH and Co. KGaA.
Aiyer, P. V. (2005). Amylases and their applications. African Journal of Biotechnology, 4,
1525–1529.
Al Hafid, N., & Christodoulou, J. (2015). Phenylketonuria: A review of current and future
treatments. Translational Pediatrics, 4, 304–317.
Alirezaei, M., Aminlari, M., Gheisar, H. R., & Tavana, M. (2011). Actinidin: A promising milk
coagulating enzyme. European Journal of Food Research and Review, 1, 43–51.
Alkorta, I., Garbisu, L. M. J., & Serra, J. L. (1998). Industrial applications of pectic enzymes: A
review. Process Biochemistry, 33, 21–28.
56 M. Aminlari
Amal Ben Amira, A., Besbes, S., Attia, H., & Blecker, C. (2017). Milk-clotting properties of plant
rennets and their enzymatic, rheological and sensory role in cheese making: A review. Interna-
tional Journal of Food Properties, 20, S76–S93.
Aminlari, L., Mohammadi Hashemi, M., & Aminlari, M. (2014). Modified lysozymes as novel
broad spectrum natural antimicrobial agents in foods. Journal of Food Science., 79, R1077–
R1090.
Aminlari, L., Roshanzadeh, S., Jafarpoor, D., & Aminlari, M. (2010). Changes in phenylalanine
ammonia lyase activity of wheat seedling during storage at different temperatures. In First
International Congress on Food Technology, November 3–6, 2010, Antalya, Turkey.
Aminlari, M., Shekarforoush, S. S., Gheisari, H. R., & Golestan, L. (2009). Effect of actinidin on
the protein solubility, water holding capacity, texture, electrophoretic pattern of beef, and on the
quality attributes of a sausage product. Journal of Food Science, 74, C221–C226.
Aminlari, M., & Vaseghi, T. (2006). Biochemical properties and biological functions of the enzyme
rhodanese in domestic animals. Iranian Journal of Veterinary Research, 7, 1–13.
Arshad, M. S., Kwon, J. H., Imran, M., Sohaib, M., Aslam, A., Nawaz, I., Amjad, Z., Khan, U., &
Javed, M. (2016). Plant and bacterial proteases: A key towards improving meat tenderization, a
mini review. Cogent Food and Agriculture, 2, 1261780. https://doi.org/10.1080/23311932.
2016.1261780.
Autio, K., Harkonen, H., Parkkonen, T., Frigard, T., Poutanen, K., Siika-Aho, M., & Aman,
M. (1996). Effects of purified endo β-glucanase on the structural and baking characteristics of
rye doughs. Lebensmittel-Wissenschaft und -Technologie, 29, 18–27.
Bamforth, C. W. (2009). Current perspectives on the role of enzymes in brewing. Journal of Cereal
Science, 50, 353–357.
Bell, S. M., Wendt, D. J., Zhang, Y., Taylor, T. W., Long, S., TsurudaL, Z. B., Laipis, P., &
Fitzpatrick, P. A. (2017). Formulation and PEGylation optimization of the therapeutic
PEGylated phenylalanine ammonia lyase for the treatment of phenylketonuria. PLoS One, 12,
e0173269. https://doi.org/10.1371/journal.pone.0173269.
Benen, J. A. E., Voragen, A. G. I., & Visser, J. (2003). Pectic enzymes. In J. R. Whitaker,
A. G. J. Voragen, & D. W. S. Wong (Eds.), Handbook of food enzymology. Marcel Dekker.
Bhardwaj, N., Kumar, B., & Verma, P. A. (2019). Detailed overview of xylanases: An emerging
biomolecule for current and future prospective. Bioresources and Bioprocessing, 6, 40.
Bhat, Z. F., Kumar, S., & Bhat, H. F. (2015). Bioactive peptides of animal origin: A review. Journal
of Food Science and Technology, 9, 5377–5392. https://doi.org/10.1007/s13197-015-1731-5
Biely, P. (2003). Xylanolytic enzymes. In J. R. Whitaker, A. G. J. Voragen, & D. W. S. Wong
(Eds.), Handbook of food enzymology. Marcel Dekker.
Boland, M. (2013). Kiwifruit proteins and enzymes: Actinidin and other significant proteins.
Advances in Food and Nutrition Research, 68, 59–80.
Bourne, Y., & Henrissat, B. (2001). Glycoside hydrolases and glycosyltransferases: Families and
functional modules. Current Opinion in Structural Biology, 11, 593–600. https://doi.org/10.
1016/s0959-440x(00)00253-0
Branen, K. J., & Davidson, P. M. (2004). Enhancement of nisin, lysozyme and monolaurin
antimicrobial activities by ethylenediaminetetraacetic acid and lacto-ferrin. International Jour-
nal of Food Microbiology, 90, 63–73.
Breaker, R. R. (1997). DNA enzymes. Nature Biotechnology, 15, 427–431.
Camm, E. L., & Towers, G. (1973). Phenylalanine ammonia lyase. Phytochemistry, 12, 961–973.
https://doi.org/10.1016/0031-9422(73)85001-0.
Casado, V., Martín, D., Torres, C., & Reglero, G. (2012). Phospholipases in food industry: A
review. In G. Sandoval (Ed.), Lipases and phospholipases: Methods and protocols, methods in
molecular biology (Vol. 861, pp. 495–523). Springer Science and Business Media. https://doi.
org/10.1007/978-1-61779-600-5_29
Cegielska-Radziejewska, R., & Szablewski, T. (2013). Effect of modified lysozyme on the micro-
flora and sensory attributes of ground pork. Journal of Food Protection, 76, 338–342.
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 57
Chahardahcherik, M., Ashrafi, M., Ghasemi, Y., & Aminlari, M. (2020). Effect of chemical
modification with carboxymethyl dextran on kinetic, structural and immunological properties
of asparaginase. Analytical Biochemistry, 591, 113537. https://doi.org/10.1016/j.ab.2019.
113537
Chalamaiah, M., Dinesh Kumar, B., Hemalatha, R., & Jyothirmayi, T. (2012). Fish protein
hydrolysates: Proximate composition, amino acid composition, antioxidant activities and
applications: A review. Food Chemistry, 135, 3020–3038.
Chaudhary, M., & Gupta, R. (2012). Cyanide detoxifying enzyme: Rhodanese. Current Biotech-
nology, 1, 327–335.
Choi, J., Latha, S., Hasan, A., & Anad, S. (2012). Bioactive peptides in dairy products. Interna-
tional Journal of Dairy Technology, 65, 1–12.
Christophersen, C., Otzen, D. E., Norman, B. E., Christensen, S., & Schäfer, T. (1998). Enzymatic
characterization of Novamyl[R], a thermostable alpha-amylase. Starch-Stuttgart, 50, 39–45.
Clemente, A. (2000). Enzymatic protein hydrolysates in human nutrition. Trends in Food Science
and Technology, 11, 254–262.
Cunningham, F. E., Proctor, V. A., & Goetsch, S. J. (1991). Egg-white lysozyme as a food
preservative: An overview. World’s Poultry Science Journal, 47, 141–163.
Das, R., & Kayastha, A. M. (2019). β-Amylase: General properties, mechanism and panorama of
applications by immobilization on nano-structures. In Q. Husain & M. Ullah (Eds.), Biocataly-
sis, enzymatic basics and applications (pp. 17–38). Springer.
Demain, A. L. (2009). Antibiotics: Natural products essential to human health. Medicinal Research
Reviews, 29, 821–842.
Di Cera, E. (2009). Serine proteases. IUBMB Life, 61, 510–515.
Diamond, S. L. (2007). Methods for mapping protease specificity. Current Opinion in Chemical
Biology, 11, 46–51.
Ding, Z., Harding, C. O., & Thöny, B. (2004). State-of-the-art 2003 on PKU gene therapy.
Molecular Genetics and Metabolism, 81, 3–8.
Dixit, S., Upadhyay, S. K., Singh, H., Pandey, B., Chandrashekar, K., & Verma, P. C. (2013).
Pectin methylesterasease of Datura species, purification, and characterization from Datura
stramonium and its application. Plant Signaling & Behavior, 8, e25681. https://doi.org/10.
4161/psb.25681
Domingo, C., Roberts, K., Stacey, N. J., Connerton, I., Ruíz-Teran, F., & MC, M. C. (1998).
Apectatelyase from Zinnia elegans is auxin inducible. The Plant Journal, 13, 17–28.
Ebrahimnejad, H., Gheisari, H. R., & Khan Nazer, A. H. (2013). Pea seedling amine oxidase
application: An emerging antihistamine strategy in tuna fish. Journal of Food Processing and
Technology, 4, 242–248. https://doi.org/10.4172/2157-7110.1000242
El-Sohaimy, S. A., Hafez, E. E., & El-Saadani, M. A. (2010). Cloning and in vitro-transcription of
chymosin gene in E. coli. The Open Nutraceuticals Journal, 3, 63–68.
Englund, P. T., King, T. P., Craig, L. C., & Walti, A. (1968). Studies on ficin. I. Its isolation and
characterization. Biochemistry, 7, 163–175.
Esmaeilpour, M., Ehsani, M. R., Aminlari, M., Shekarforoush, S. S., & Hoseini, E. (2016).
Antimicrobial activity of peptides derived from enzymatic hydrolysis of goat milk caseins.
Comparative Clinical Pathology, 25, 599–605. https://doi.org/10.1007/s00580-016-2237-x
Esmaeilpour, M., Ehsani, M. R., Aminlari, M., Shekarforoush, S. S., & Hoseini, E. (2017).
Antimicrobial peptides derived from goat’s milk whey proteins obtained by enzymatic hydro-
lysis. Journal of Food Biosciences and Technology, 7, 65–72.
Fernandes, P. (2018). Enzymatic processing in the food industry. Elsevier.
Fox, P. F. (1993). Exogenous enzymes in dairy technology—A review. Journal of Food Biochem-
istry, 17, 173–199.
Frushicheva, M. P., Mills, M. J. L., Schopf, P., Singh, M. K., Prasad, R. B., & Warshel, A. (2014).
Computer aided enzyme design and catalytic concepts. Current Opinion in Chemical Biology,
21, 56–62.
58 M. Aminlari
Gheisari, H. R., Shekarforoush, S. S., & Aminlari, M. (2008). Application of fresh, defrosted and
actinidin-tenderized camel and cattle meat in the production of emulsion type sausages.
Advances in Food Sciences, 30, 1–7.
Gigot, C., Ongena, M., Fauconnier, M., Wathelet, J., Jardin, P. D., & Thonart, P. (2010). The
lipoxygenase metabolic pathway in plants: Potential for industrial production of natural green
leaf volatiles. Biotechnology, Agronomy, Society and Environment, 14, 451–460.
Gokoglu, N., Yerlikaya, P., Ucak, I., & Yatmaz, H. A. (2017). Effect of bromelain and papain
enzymes addition on physiochemical and textural properties of squid (Loligo vulgaris). Food
Measure, 11, 345–353. https://doi.org/10.1007/s11694-016-9403-3
Goldson, A., Lam, M., Scaman, C. H., Clemens, S., & Kermode, A. (2008). Screening of
phenylalanine ammonia lyase in plant tissues, and retention of activity during dehydration.
Journal of the Science of Food and Agriculture, 88, 619–625.
Gomaa, A. M. (2018). Application of enzymes in brewing. Journal of Nutrition and Food Science
Forecast, 1, 1–5.
Grahn, S., Kurth, T., Ullmann, D., & Jakubke, H. (1999). S0 subsite mapping of serine proteases
based on fluorescence resonance energy transfer. Biochimica et Biophysica Acta, 1431,
329–337.
Green, M. L. (1977). Review of the progress of dairy science: Milk coagulants. The Journal of
Dairy Research, 44, 159–188.
Gunstone, F. D. (Ed.). (1999). Lipid synthesis and manufacture (Vol. 472). CRC Press LLC.
Gupta, R., Kohli, P., & Kalia, M. (2015). Pectin methylesteraseases: A review. Journal of
Bioprocessing and Biotechniques, 5, 1–5. https://doi.org/10.4172/2155-9821.1000227
Hebeda, R. E., Bowles, L. K., & Teague, W. M. (1991). Use of intermediate stability enzymes for
retarding staling in baked goods. Cereal Foods World, 36, 619–624.
Hedstrom, L. (2002). Serine protease mechanism and specificity. Chemical Reviews, 102,
4501–4523.
Hefferon, K. (2017). Cellulase production in transgenic plants: Molecular pharming with a twist.
Advances in Biotechnology and Microbiology, 6, 555685. https://doi.org/10.19080/AIBM.
2017.06.555685
Hughes, H. A. (1999). Biosynthesis and degradation of cyanogenic glycosides. In D. Barton,
K. Nakanishi, & O. Meth-Cohn (Eds.), Comprehensive natural product chemistry
(pp. 881–895). Pergamon Press.
Ibrahim, H. R., Hl, H., Fujiki, M., Kim, M., & Yamamoto, T. (1994). Enhanced anti-microbial
action of lysozyme against gram-negative and gram-positive bacteria due to modification with
perillaldehyde. Journal of Agricultural and Food Chemistry, 42, 1813–1817.
Ikeda, K., Inada, Y., Schiltz, E., Fujii, T., Takahashi, M., Mitsui, K., Kodera, Y., Matsushima, A.,
Schulz, G. E., & Nishimura, H. (2005). Phenylalanine ammonia-lyase modified with polyethyl-
ene glycol: Potential therapeutic agent for phenylketonuria. Amino Acids, 29, 283–287. https://
doi.org/10.1007/s00726-005-0218-5
Illanes, A. (2008). Enzyme production. In A. Illanes (Ed.), Enzyme biocatalysis (pp. 57–106).
Springer.
Istrati, D. (2008). The influence of enzymatic tenderization with papain on functional properties of
adult beef. Journal of Agroalimentary Processes and Technologies, 14, 140–146.
Ito, A., & Ma, J. (2003). Amine oxidase. In J. R. Whitaker, A. G. J. Voragen, & D. W. S. Wong
(Eds.), Handbook of food enzymology. Marcel Dekker.
Javed, R., Nawaz, A., Munir, M., Hanif, M., Mukhtar, H., Ul Haq, I., & Abdullah, R. (2018).
Extraction, purification and industrial applications of pectinase: A review. Journal of Biotech-
nology and Bioresearch, 1, 1–6.
Kaushal, Y., Singh, S. G., Raina, A., & Arya, S. K. (2018). Catalase enzyme: Application in
bioremediation and food industry. Biocatalysis and Agricultural Biotechnology, 16, 192–199.
https://doi.org/10.1016/j.bcab.2018.07.035
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 59
Kawaguchi, Y., Kosugi, S., Sasaki, K., Uozumi, T., & Beppu, T. (1987). Production of chymosin in
Escherichia coli cells and its enzymatic properties. Agricultural and Biological Chemistry, 51,
1871–1877. https://doi.org/10.1080/00021369.1987.10868318
Kilcawley, K. N., Wilkinson, M. G., & Fox, P. F. (1998). Enzyme-modified cheese. International
Dairy Journal, 8, 1–10.
Kim, S. Y., Gunasekaran, S., & Olson, N. F. (2004a). Combined use of chymosin and protease from
cryphonectria parasitica for control of meltability and firmness of cheddar cheese. Journal of
Dairy Science, 87, 274–283.
Kim, W., Erlandsen, E., Surendran, S., Stevens, R., Gamez, A., Michols-Matalon, K., Tyring, S. K.,
& Matalon, R. (2004b). Trends in enzyme therapy for phenylketonuria. Molecular Therapy, 10,
220–224. https://doi.org/10.1016/j.ymthe.2004.05.001
Klemm, D., Heublein, B., Fink, H. P., & Bohn, A. (2005). Cellulose: Fascinating biopolymer and
sustainable raw material. Angewandte Chemie International Edition, 44, 3358–3393. https://
doi.org/10.1002/anie.200460587
Korhonen, H., & Pihlanto, A. (2006). Bioactive peptides: Production and functionality. Interna-
tional Journal of Dairy Technology, 16, 945–960.
Kumar, D., Kumar, S. S., Kumar, J., Kumar, O., Mishra, S. V., Kumar, R., & Malyan, S. K. (2017).
Xylanases and their industrial applications: A review. Biochemical and Cellular Archives, 17,
353–360.
Laurikainen, T., Harkonen, H., Autio, K., & Poutanen, K. (1998). Effects of enzymes in fibre-
enriched baking. Journal of the Science of Food and Agriculture, 76, 239–249.
Levy, H. L. (1999). Phenylketonuria: Old disease, new approach to treatment. Proceedings of the
National Academy of Sciences of the United States of America, 96, 1811–1813.
Lewis, D., & Luh, B. S. (2007). Development and distribution of actinidin in kiwifruit (Actinidia
chinensis) and its partial characterization. Journal of Food Biochemistry, 12, 109–116. https://
doi.org/10.1111/j.1745-4514.1988.tb00363.x
Lewis, D. A., & Luh, B. S. (1988). Application of actinidin from kiwifruit to meat tenderization and
characterization of beef muscle protein hydrolysis. Journal of Food Biochemistry, 12, 147–158.
Lian, T., Wang, L., & Liu, Y. (2013). A new insight into the role of calpains in post-mortem meat
tenderization in domestic animals: A review. Asian-Australasian Journal of Animal Sciences,
26, 443–454. https://doi.org/10.5713/ajas.2012.12365
Lo Piero, A. R., Puglisi, I., & Petrone, G. (2011). Characterization of the purified actinidin as a plant
coagulant of bovine milk. European Food Research and Technology, 233, 517–524. https://doi.
org/10.1007/s00217-011-1543-4
Losso, J. N., Nakai, S., & Charter, E. A. (2000). Lysozyme. In A. S. Naidu (Ed.), Natural food
antimicrobial systems (pp. 185–210). CRC Press LLC.
MacGregor, A. W., & Morgan, J. E. (1992). Determination of specific activities of malt α-amylases.
Journal of Cereal Science, 16, 267–277.
Mansfeld, J. (2009). Plant phospholipases A2: Perspectives on biotechnological applications.
Biotechnology Letters, 31, 1373–1380.
Marques, A. C., Marostica, M. R., Jr., & Maria Pastore, G. M. (2010). Some nutritional, techno-
logical and environmental advances in the use of enzymes in meat products. Enzyme Research,
480923. https://doi.org/10.4061/2010/480923
Mazorra-Manzano, M. A., Ramírez-Suarez, J. C., & Yada, R. (2018). Plant proteases for bioactive
peptides release: A review. Critical Reviews in Food Science and Nutrition, 58, 2147–2163.
https://doi.org/10.1080/10408398.2017.1308312
Mirzaei, M., Mirdamadi, S., Ehsani, M. R., & Aminlari, M. (2016). Antioxidant, ACE-inhibitory
and antimicrobial activities of Kluyveromyces marxianus protein hydrolysates and their peptide
fractions. Functional Foods in Health and Disease, 6, 425–439. https://doi.org/10.31989/ffhd.
v6i7.250
Mirzaei, M., Mirdamadi, S., Ehsani, M. R., & Aminlari, M. (2017). Production of antioxidant and
ACE-inhibitory peptides from Kluyveromyces marxianus protein hydrolysates: Purification and
60 M. Aminlari
Rui, X., Boye, J., Simpson, B. K., & Prasher, S. O. (2012). Angiotensin I-converting enzyme
inhibitory properties of Phaseolus vulgaris bean hydrolysates: Effects of different thermal and
enzymatic digestion treatments. Food Research International, 49, 739–746.
Ruiz-Capillas, C., & Herrero, A. M. (2019). Impact of biogenic amines on food quality and safety.
Food, 8, 62. https://doi.org/10.3390/foods8020062
Sabbione, A. C., Ibañez, S. M., Martínez, E. N., Añón, M. C., & Scilingo, A. A. (2016).
Antithrombotic and antioxidant activity of amaranth hydrolysate obtained by activation of an
endogenous protease. Plant Foods for Human Nutrition, 71, 174–182.
Saini, R., Saini, H. S., & Dahiya, A. (2017). Amylases: Characteristics and industrial applications.
Journal of Pharmacognosy and Phytochemistry, 6, 1865–1871.
Sánchez, A., & Vázquez, A. (2017). Bioactive peptides: A review. Food Quality Safety, 1, 29–46.
https://doi.org/10.1093/fqs/fyx006
Sarkissian, C. N., Shao, Z., Blain, F., Peevers, R., Su, H., Heft, R., Chang, T. M. S., & Scriver,
C. R. A. (1999). A different approach to treatment of phenylketonuria: Phenylalanine degrada-
tion with recombinant phenylalanine ammonia lyase. Proceedings of the National Academy of
Sciences of the United States of America, 96, 2339–2344.
Schmid, R. D., & Verger, R. (1998). Lipases. Interfacial enzymes with attractive applications.
Angewandte Chemie International Edition, 37, 1608–1633.
Schols, H. A., & Voragen, A. G. J. (2003). Pectic polysaccharides. In J. R. Whitaker,
A. G. J. Voragen, & D. W. S. Wong (Eds.), Handbook of food enzymology. Marcel Dekker.
Semenova, M. V., Sinitsyna, O. A., Morozova, V. V., Fedorova, E. A., Gusakov, A. V., Sokolova,
L. M., Koshelov, A. A., Bubonova, T. V., Vinetskii, Y. P., & Sinitsyn, A. P. (2006). Use of a
preparation from fungal pectin lyase in the food industry. Applied Biochemistry and Microbiol-
ogy, 42, 598–602.
Seo, S., Karboune, S., L’Hocine, L., & Yaylayan, V. A. (2013). Characterization of glycated
lysozyme with galactose, galactooligosaccharides and galactan: Effect of glycation on func-
tional properties of lysozyme. LWT—Food Science and Technology, 53, 44–53.
Shah, M. H., Mir, S. A., & Paray, M. A. (2014). Plant proteases as milk-clotting enzymes in
cheesemaking: A review. Dairy Science and Technology, 94, 5–16.
Shahidi, S., Jamili, S., Mostafavi, P. G., Rezaie, S., & Khorramizadeh, M. (2018). Assessment of
the inhibitory effects of ficin-hydrolyzed gelatin derived from squid (Uroteuthis duvauceli) on
breast cancer cell lines and animal model. Iranian Journal of Allergy, Asthma and Immunology,
17, 436–452. https://doi.org/10.18502/ijaai.v17i5.302
Shi, Y., Mandal, R., Singh, A., & Singh, A. P. (2020). Legume lipoxygenase: Strategies for
application in food industry. Legume Science, e44. https://doi.org/10.1002/leg3.44
Singh, G., Verma, A. K., & Kumar, V. (2016). Catalytic properties, functional attributes and
industrial applications of β-glucosidases. 3 Biotech, 6, 1–14. https://doi.org/10.1007/s13205-
015-0328-z
Sinha, R., Radha, C., Prakash, J., & Kaul, P. (2007). Whey protein hydrolysate: Functional
properties, nutritional quality and utilization in beverage formulation. Food Chemistry, 101,
1484–1491.
Song, H., Wang, P., Li, C., Han, S., Lopez-Baltazar, J., Zhang, X., & Wang, X. (2016). Identifica-
tion of lipoxygenase (LOX) genes from legumes and their responses in wild type and cultivated
peanut upon Aspergillus flavus infection. Scientific Reports, 6, 35245. https://doi.org/10.1038/
srep35245
Sonnet, P. E., & Gazzillo, J. A. (1991). Evaluation of lipase selectivity for hydrolysis. Journal of the
American Oil Chemists’ Society, 68, 11–15.
Stránská, J., Sebela, M., Tarkowski, P., Rehulka, P., Chmelík, J., Popa, I., & Peč, P. (2007).
Inhibition of plant amine oxidases by a novel series of diamine derivatives. Biochimie, 89,
135–144.
Tavano, O. S., Berenguer-Murcia, A., Secundo, F., & Fernandez-Lafuente, R. (2018). Biotechno-
logical applications of proteases in food technology. Comprehensive Reviews in Food Science
and Food Safety, 17, 412–436. https://doi.org/10.1111/1541-4337.12326
62 M. Aminlari
Tenkanen, M., Niku-Paavola, M. L., Linder, K., & Viikari, L. (2003). Cellulases in food processing.
In J. R. Whitakerm, A. G. J. Voragen, & D. W. S. Wong (Eds.), Handbook of food enzymology.
Marcel Dekker.
Thacker, S. P., Kothari, R. M., Ramamurthy, V., & Baroda, P. (1992). Characterization of barley
β-amylase for application in maltose production. Starch/Starke, 44, 339–341.
Thalmann, M., Coiro, M., Meier, T., Wicker, T., Zeeman, S. C., & Santelia, D. (2019). The
evolution of functional complexity within the β-amylase gene family in land plants. BMC
Evolutionary Biology, 19, 66–84. https://doi.org/10.1186/s12862-019-1395-2
Thiansilakul, Y., Benjakul, S., & Shahidi, F. (2007). Compositions, functional properties and
antioxidative activity of protein hydrolysates prepared from round scad (Decapterus maruadsi).
Food Chemistry, 103, 1385–1394.
Toohey, E. S., Kerr, M. J., van de Ven, R., & Hopkins, D. L. (2011). The effect of a kiwi fruit based
solution on meat traits in beef m. semimembranosus (topside). Meat Science, 88, 468–471.
https://doi.org/10.1016/j.meatsci.2011.01.028
Uluisik, S., & Seymour, G. B. (2020). Pectate lyases: Their role in plants and importance in fruit
ripening. Food Chemistry, 309, 125559. https://doi.org/10.1016/j.foodchem.2019.125559
Ulusu, Y., Şentürk, S. B., Kuduğ, H., & Gökçe, I. (2016). Expression, purification and characteri-
zation of bovine chymosin enzyme using an inducible pTOL system. Preparative Biochemistry
and Biotechnology, 46, 596–601.
Van der Maarel, M. J. E. C., ven der Veen, B., Uitdehaag, J. C. M., Leemhius, H., & Dijkhuizem,
L. (2002). Properties and applications of starch-converting enzymes of the α-amylase family.
Journal of Biotechnology, 94, 137–165.
Verma, S., Dixit, R., & Pandey, K. C. (2016). Cysteine proteases: Mode of activation and future
prospects as pharmaceutical targets. Frontiers in Pharmacology, 7, 107. https://doi.org/10.
3389/fphar.2016.00107
Vizovišek, M., Vidmar, R., Drag, M., Fonovic, M., Salvesen, G. S., & Turk, B. (2018). Protease
specificity: Towards in vivo imaging applications and biomarker discovery. Trends in Biochem-
ical Sciences, 43, 829–844. https://doi.org/10.1016/j.tibs.2018.07.003
Whitaker, J. R. (1990). New and future uses of enzymes in food processing. Food Biotechnology, 4,
669–697.
Whitaker, J. R. (1994). Principles of enzymology for food scientists (pp. 391–422). Marcel Dekker.
Whitaker, J. R. (2003). Proteolytic enzymes. In J. R. Whitaker, A. G. J. Voragen, & D. W. S. Wong
(Eds.), Handbook of food enzymology. Marcel Dekker.
Williams, D. C., Lim, M. H., Chen, A. O., Pangborn, R. M., & Whitaker, J. R. (1986). Blanching of
vegetables for freezing: Which indicator enzyme to choose. Food Technology, 40, 130–140.
Wong, D. W. S. (2003). Lipase. In J. R. Whitaker, A. G. J. Voragen, & D. W. S. Wong (Eds.),
Handbook of food enzymology. Marcel Dekker.
Wong, D. W. S., & Robertson, G. H. (2007). α-Amylases. In J. R. Whitaker, A. G. J. Varagen, &
D. W. S. Wong (Eds.), Handbook of food enzymology. Marcel Dekker.
Wong, D. W. S., & Whitaker, J. R. (2003). Catalase. In J. R. Whitaker, A. G. J. Voragen, &
D. W. S. Wong (Eds.), Handbook of food enzymology. Marcel Dekker.
Woolcock, K. (2016). Structure of a DNA enzyme. Nature Structural and Molecular Biology,
23, 97.
Yapo, B. M. (2011). Pectic substances: From simple pectic polysaccharides to complex pectins. A
new hypothetical mode. Carbohydrate Polymers, 86, 373–385. https://doi.org/10.1016/j.
carbpol.2011.05.065
2 Plants- and Animal-Derived Enzymes and Their Potential Application in. . . 63
Zhang, W., Xiao, S., & Ahn, D. U. (2013). Protein oxidation: Basic principles and implications for
meat quality. Critical Reviews in Food Science and Nutrition, 53, 1191–1201. https://doi.org/
10.1080/10408398.2011.577540
Zhang, X. Z., & Zhang, Y. H. P. (2013). Cellulases: Characteristics, sources, production, and
applications. In S. T. Yang, H. A. El-Enshasy, & N. Thongchul (Eds.), Bioprocessing
technologies in biorefinery for sustainable production of fuels, chemicals, and polymers (1st
ed.). John Wiley & Sons.
Zhao, X., Shi-Jian, D., Tao, G., et al. (2010). Influence of phospholipase A2 (PLA2)-treated dried
egg yolk on wheat dough rheological properties. LWT—Food Science and Technology, 43,
45–51.
Zhu, F., Du, B., Ma, Y., & Li, J. (2017). The glycosidic aroma precursors in wine: Occurrence,
characterization and potential biological applications. Phytochemistry Reviews, 16, 565–571.
Role of Enzymes in Fruit and Vegetable
Processing Industries: Effect on Quality, 3
Processing Method, and Application
Abstract
Keywords
Enzymes · Fruit and vegetable processing · HPP · PEF · Shelf life extension
Abbreviations
3.1 Introduction
Enzymes play a very critical role in the growth and maturation of fruits and
vegetables. They also make significant contributions during the post-harvest phase
in maintaining stability of raw food materials and improving their quality attributes
like aroma, colour, texture, flavour, and nutritional quality. Apart from these,
enzymes also have the ability to act as catalysts in transforming raw materials to
food products during processing. They are also found to have extensive applications
in food processing and production by enhancing the nutritional, safety, functional
qualities, and overall acceptability of ingredients and processed products. Moreover,
enzymes are also known for their substrate specificity, effectiveness in catalytic
reactions, and enhancement rate under controlled conditions of moisture, pH, and
temperature (Berg et al., 2010).
Due to rapid growth of population and fast changing lifestyle, the production and
demand of convenience and processed food products have also been gradually
increasing. As per the study of Markets and Markets (2020), the fruit and vegetable
processing enzyme global market is expected to touch around $41.39 billion by
2022 at 6.7% CAGR from 2016 to 2020. During this corresponding period, the Asia-
Pacific region enzyme market is projected to grow at the highest CAGR due to high
growth of the food and beverage industry in big emerging economies like India and
China. Therefore, productive usage of the existing enzymes and the development of
new enzymes have become very significant in meeting the ever-increasing demands
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 67
Knowing the composition of fruit and vegetable is the first step in determining the
benefits and drawbacks of them. Fruits and vegetables are mainly composed of
cellulose (7.20–43.60%), hemicelluloses (4.26–33.50%), pectin (1.50–13.40%),
lignin (15.30–69.40%), and starch (3–21%) (Dhillon et al., 2013). The primary
cell walls in the young plants comprise largely of cellulose and tend to thicken and
become higher in hemicelluloses and lignin as they grow (Albersheim et al., 1996).
Now, let’s discuss in detail the different types of composition in fruits and
vegetables.
3.2.1 Cellulose
Cellulose is one of the main components in fruits and vegetables ranging from 7.20%
to 43.60%. It is a linear polymer of D-glucose present in the primary cell wall. It
forms microfibrils, which give a stiff shape, structure, and tensile strength to the cell
wall. It also helps to form a resistance against the degradation of this polymer
(Szymańska-Chargot et al., 2017). These microfibrils are coated with hemicelluloses
and help in binding firmly to their surface (Carpita & Gibeaut, 1993). Cellulose is
known for its diverse applications in food processing industry. It is used as an
emulsifier, bulking agent, texturizer, and as a fat substitute (Grassino et al., 2016).
However, the application of celluloses during the extraction process needs extra
precaution in order to avoid breaking down the cellulose network in maceration
processes as it decreases the quantity of juice production.
68 M. D. Heirangkhongjam et al.
3.2.2 Hemicellulose
3.2.3 Pectin
Pectin is one of the least present components in fruits and vegetable ranging from
1.50% to 13.40%. It forms a class of complex polysaccharides commonly found in
the cell walls of higher plants. It provides structure and firmness to the plant tissue in
the primary cell wall and middle lamella component involved in intercellular
adhesion (Thakur et al., 1997). There are three pectic polysaccharides, namely,
homogalacturonan, rhamnogalacturonans, and substituted galacturonans isolated
from primary cell walls. Homogalacturonan (HG) is a homopolymer, a linear
chain of 1,4-linked α-D-galactosyluronic residues, known for its gel forming ability.
In HG, some of the carboxyl groups are methylesterified at the C-2 and C-3
positions. Similarly, the backbone of Rhamnogalacturonan-II (RG-II) is composed
of 1,4-linked D-galactosyluronic residues like HG, with a non-saccharide and an
octa-saccharide side chains attached to C-2 and two disaccharides attached to C-3 of
the backbone residues. RG-II is significantly used in winery and fruit juice industries
because of its exceptional quality of binding heavy metals, thereby reducing the
toxicity level in the final product. It also has immunomodulating activities (Grassin
& Coutel, 2009). Galarturonan fractions are generally separated from other high
molecular weight pectin fractions by degrading purified pectins specifically present
in the galacturonan backbone either enzymatically or chemically. In fact, as
explained above, the different types of pectins perform certain specific functions,
but pectins in general are largely used as gelling and thickening agent in dairy and
baking. Also, pectins are being widely used as a carrier of drug delivery system in
cosmetic and pharmaceutical industries (Kollarigowda, 2015).
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 69
3.2.4 Starch
Starch is another main component in fruits and vegetables which is known very
commonly. It is a polysaccharide composed of amylose and amylopectin. Amylose
has linear chains of about 840–22,000 units of α-D-glucopyranosyl residues linked
by 1!4-α-D-glucan, whereas amylopectin is usually comprised of highly branched
α-1!6 and α-1!4 glucosidic linkages. Starch is a form of carbohydrate or energy
reserve mostly found in storage organs, seeds, unripe fruits, and vegetables. The
most important quality of starch is its water holding capacity which depends on the
specific shape and size of each granule. For example, a potato granule can hold
approx. 5–100 μm water/glucose unit (Jobling, 2004). Moreover, each granule
contains ‘blockets’ of amylopectin which are crystalline and amorphous in nature.
They are responsible for absorption of water, swelling, lose crystallinity, and seep
amylose. However, the swelling capacity tends to be lower with higher amylose
content and thus results in reduction in gel strength (Li & Yeh, 2001). Therefore,
because of its inconsistent natural properties due to the vagaries of weather and
agricultural conditions, most of the suppliers constantly try to make uniform starches
as functional ingredients. In food processing industry, the usage of starch is mani-
fold. It acts as an innate natural ingredient with different added functionalities like an
emulsifier, stabilizer, water binder, thickener, and gelling agent.
3.2.5 Lignin
Lignin is one of the major components present in fruits and vegetables ranging from
15.30% to 69.40%. It represents a class of natural aromatic polymers of
4-hydroxyphenylpropanoids units connected by ether and carbon-carbon linkages.
Lignins are generally considered a part of dietary fiber. They are mainly present in
cereals, fruit, and vegetables, in which wheat bran has the richest source of lignin.
Lignins have efficient antioxidant properties, mainly scavenging superoxide and
hydroxyl radicals. It also inhibits the activity of xanthine oxidase, glucose-6-phos-
phate dehydrogenase, and non-enzymatic and enzymatic lipid peroxidation
(Lu et al., 1998). In addition to this, its concentration and composition influence
the adsorption capacity of cell walls. Therefore, due to these properties, lignins are
considered as efficient adsorbers of hydrophobic heterocyclic aromatic amines
(Funk et al., 2006). Although lignin has many applications in other different
industries, in food industry it is mainly used as an additive, provides roughage to
foods, and as sequestering agents.
70 M. D. Heirangkhongjam et al.
As discussed, the role of enzymes in the overall natural growth, maturation, and
ripening of fruits and vegetables is very critical. This very importance of enzymes
remains the same or even more in the industrial processing of fruit and vegetables.
Another important positive aspect for enzymes is their ability in keeping the quality
of fresh fruits and vegetables post-harvest and during storage conditions, which
ultimately plays a very pivotal role in the industrial food processing.
In industrial processing, the ripening process with the application of enzymes is a
major step, wherein several changes like alterations in the cell wall, middle lamellae,
and membrane occur resulting in softening of tissues. Naturally, most of the
enzymes in fruits and vegetable tissues are important for the maintenance of metab-
olism; however, they are also associated with undesirable effects on colour, flavour,
odour, texture, and nutritional value. For example, in some vegetables, flavour and
odour development is affected by lipoxygenase, lipase, and peroxidase (Fleuri et al.,
2016). Furthermore, phenol oxidases result in discolouration and unfavourable
effects on the taste and nutritional quality of fruits and vegetables. Also, fruits and
vegetables containing pectic substances and α-amylases have major effects on their
textural integrity (Berg et al., 2010; Fleuri et al., 2016).
With the advancement in enzymology, different types of new enzymes have been
developed and are being used in improving the quality of products, development of
new products, and in processing aids such as peeling, extraction of juice, and
clarification, thereby increasing the efficiency in processing operation. For example,
amylases, cellulases, and pectinases facilitate maceration, liquefaction, and
clarification in processing of fruit juice, and hence they are cost-effective and
increase the yield. Moreover, different enzyme extracts from plant tissues, bacteria,
yeast, and fungi are applied in fruit and vegetable processing industries for the same
purpose (Leadlay, 1993). In Table 3.1, some of the important microorganism origin
enzymes used in industrial processing and its application are summarized. In
Table 3.2, different types of enzymes, product type, and its application in fruit and
vegetable processing are highlighted.
Enzymes are classified broadly into four types: Pectinases, Cellulase, Xylanase,
and Amylase. In the proceeding paragraphs, we will be discussing in brief the
different types of enzymes in fruit and vegetable processing industry.
3.3.1 Pectinases
Table 3.1 Enzymes derived from microorganisms and their application in fruits and vegetable
processing
Enzyme Microorganism Action Application
Pectinase Aspergillus spp., Pectin hydrolysis Degradation of pectins,
Penicillium funiculosum increases the overall juice
production, fruit juice
clarification
Pectinesterase Aspergillus spp. Remove methyl Increase firmness of
groups from vegetables and also used
galactose units of with pectinase
pectin depectinisation
technology
Protopectinase Kluyveromyces fragilis, Catalyse pectin Clarification and
Galactomyces reesei, solubilization reduction of viscosity in
Trichosporon fragilis, fruit juices
Bacillus subtilis
Hemicellulase Aspergillus spp., Bacillus Hemicellulose Helps extraction of fruit
subtilis, Trichoderma hydrolysis juices, vegetable oils, and
reesei aromatic compounds,
acts on hydrolysis of
soluble pectin and cell
wall components with
pectinases, lowers
viscosity and texture
α-Amylase Aspergillus spp., Bacillus Random Hydrolysing starch to
spp., Microbacterium hydrolyses α-1,4 reduced viscosity,
imperiale bounds to rupture liquefying adjunct, helps
starch and produce in sugar production, for
maltose softness and increases
volume of fruit juices and
vegetables
Glucoamylase Aspergillus niger, Hydrolyse dextrin Fruit juice extraction and
Rhizopus spp. from starch in also used for corn syrup
glucose and glucose production
Glucose Streptomyces rubiginosus, Conversion of Helps in high-fructose
isomerise Streptomyces lividans, glucose to fructose corn syrup production
Actinplanes (beverage sweetener)
missouriensis, Bacillus
coagulans
Cellulase Trichoderma spp., Hydrolyses Liquefaction of fruit in
Aspergillus niger cellulose juice production
Note: Adapted from “Enzymes in food and beverage processing”, Fleuri, L. F., Delgado, C. H. O.,
Novelli, P. K., Pivetta, M. R., Do Prado, D. Z., & Simon, J. W., 2015, p. 257, London, New York:
CRC Press, Taylor & Francis Group
neutralization, which form gels with the addition of sugars and acids under
favourable conditions (Guo et al., 2014). Pectic substances are classified into
protopectins, pectic acids, and pectin which are partially soluble in water (Uneojo
& Pastore, 2007). These substances are generally degraded by the enzyme
pectinases. They are further classified into different sub-types, namely,
72 M. D. Heirangkhongjam et al.
pectatelyases (E.C. 4.2.2.9 and E.C. 4.2.2.2), or pectin lyases (E.C. 4.2.2.10)
(Kc et al., 2020).
As per a recent study, it is reported that pectinases from microbial origin account
for about 25% of global industrial enzymes market which is projected to reach USD
6.3 billion by 2021 (Oumer, 2017; Oumer & Abate, 2018). This clearly highlights
the significance of microorganism origin enzyme as a reliable source of industrial
enzyme production. Pectinases or pectinolytic enzymes are also naturally produced
by many other organisms like insects, bacteria, nematodes, and protozoans (Khairnar
et al., 2009). Some of the other commonly used microorganisms for extensive
production of pectinases are Aspergillus spp., Bacillus spp., Erwinia spp., and
Penicillium spp. (Oumer, 2017).
In food processing industries, especially in fruits and vegetables processing,
pectinases have multiple usage. They are mainly used during processing of citrus
juice. Moreover, pectinases are also generally recommended for use with the com-
bination of other enzymes such as cellulases and hemicellulases. Such combinations
of enzymes are generally used in facilitating the process of maceration, liquefaction,
and clarification. It ultimately helps in increasing the extraction yield and enhancing
the concentration of acids, colourants, and flavourings (Oumer & Abate, 2018).
Apart from these, pectinases are also widely used in wine clarification, concentra-
tion, and fermentation of tea, cocoa, and coffee. Further, these enzymes are also used
regularly in pickling, preparation of jam and jellies, syrups, starches, and vegetable
oil extraction (Kubra et al., 2018).
3.3.2 Cellulases
substances present in the grape skin. Besides releasing the flavouring substances and
improving the aroma and flavour of beverages, it also breaks the unpleasant-tasting
compounds present in the fruits and vegetables (Juturu & Wu, 2013).
Hemicellulase is an enzyme complex that breaks down the backbone of xylan and
arabinose side chains and releases pentoses (xylose and arabinose) (Yang et al.,
2017). Similarly, xylanases (endo-1,4-β-D-xylanohydrolase; E.C. 3.2.1.8) are hydro-
lytic enzymes involved in depolymerization of xylan. They are usually present in
superior plants, such as cereals, grasses, and trees that present noncellulosic
polysaccharides, such as D-glucose, L-arabinose, D-xylose, D-mannose, D-galactose,
D-glucuronic, and D-galacturonic acid (Cunha & Gandini, 2010). These enzymes
also degrade hemicellulose polymers, including acetyl xylan esterase
(E.C. 3.1.1.72), α-arabinofuranosidase (α-L-arabinofuranosidase, E.C. 3.2.1.55),
arabinase (endo-α-L-arabinase, E.C. 3.2.1.99), endo-xylanase (endo-1,4–βxylanase,
E.C. 3.2.1.8), feruloyl esterase (E.C. 3.1.1.73), α-glucuronidase (α-glucosiduronase,
E.C. 3.2.1.139), and β-xylosidase (xylan-β-1,4-xylosidase, E.C. 3.2.1.37) (Juturu &
Wu, 2013). Polymers such as xylan and arabinoxylan are completely hydrolysed by
the synergistic action of several xylanolytic enzymes: endo-1,4-β-D-xylanases. The
hydrolysis of these polymers degrades the β-D-xylan linkages; β-D-xylosidases
release a xylose monomer from the cleavage of the non-reducing end of xylo-
oligosaccharides and xylobiose (Terrasan et al., 2010).
Currently, commercial xylanases are produced on a large scale in many countries,
such as United States, Japan, Finland, Germany, Ireland, Canada, and Denmark, by
fermentation processes using bacteria, yeast, and fungi (Polizeli et al., 2005).
Substantial increase in the production of xylanases has been observed after the
development of improved microbial strains and efficient fermentation techniques
and recovery systems. It has several applications in food industries, agriculture as
well as in human health.
Furthermore, during processing of beer, the cellular wall is generally hydrolysed
releasing long chains of arabinoxylans which in turn increase the viscosity of beer
rendering it “muddy” in appearance. Then, xylanases are added to hydrolyse
arabinoxylans in order to lower oligosaccharides, thereby reducing the viscosity
and consequently improving its appearance (Dervilly et al., 2002). Xylanases in
combination with amylases, cellulases, and pectinases provide multiple advantages
such as increase yield of juice, stabilization of fruit pulp, and increased recovery of
flavours and aromas, essential oils, mineral salts, vitamins, etc.
3.3.4 Amylases
Amylases are enzymes that act as catalysts in the hydrolysis of starch into sugars
such as glucose and maltose (Sundarram & Murthy, 2014). These enzymes are
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 75
extensively found in plant, animal, and microbial kingdoms. Amylases are classified
into exo-amylases and endo-amylases based on their action. Exo-amylases are
involved in hydrolysis of α-glucan into maltose and glucose, whereas endo-amylase
hydrolyses α-glucan-forming oligosaccharides. Starch which is a polysaccharide that
is an essential factor of structure, consistency, and texture in foods is hydrolysed by
β-amylase. Moreover, the non-reducing terminals in starch polysaccharides and
malto-oligosaccharides are also hydrolysed by β-amylase (Fleuri et al., 2016).
Such enzymes are responsible for the degradation of starch and its related polymers
to yield products characteristic of individual amylolytic enzymes.
Amylase enzymes have some common types, namely, amylolytic, α-amylase,
β-amylase, and glucoamylase. These enzymes are largely present in a wide range of
organisms, including plants, animals, and microorganisms. Aspergillus spp. is the
most extensively used fungi for the production of amylolytic enzymes. The major
sources of α-amylase enzymes are A. niger, Aspergillus oryzae, B. circulans, Bacil-
lus amyloliquefaciens, B. subtilis, B. licheniformis, and B. stearothermophilus.
β-Amylase enzymes are obtained from species like Bacillus spp., Pseudomonas
spp., and Streptomyces spp. However, out of these enzymes, the production of
α-amylases accounts for about 30% of enzymes in world market for their extensive
use in food industrial processes, such as in baking, brewing, fruit juices, syrups,
starch, etc. It is also widely used in the production of drugs and other pharmaceutical
products (van der Maarel et al., 2002).
The colour of a product is the primary assessment factor of quality more than any
other factor. So, maintaining the natural colour of food is very critical. The colour of
food products is affected during pre-harvest and post-harvest factors which are
coupled by enzymatic reactions. The main enzymes responsible for changes of
colour in fruits and vegetables are polyphenol oxidase (PPO), peroxidase (POD),
and β-glucosidase (Zabetakis et al., 2000). Among the various processing changes,
enzymatic browning causes major problem in food processing (Terefe et al., 2014).
76 M. D. Heirangkhongjam et al.
Kojic acid is another natural compound; it chelates metal ions such as Fe3+ and Cu2+
in addition to impounding of free radicals (Kim & Uyama, 2005).
Proteases are another group of enzymes that catalyse the hydrolysis of proteins
which are degraded to peptides and amino acids (Omaña-Molina et al., 2013). It
inhibits PPO activity through proteolysis or binding to specific sites. According to a
study report, it was found that the browning reaction was decreased in apple when
papain was used (Labuza et al. 1992). Also, when pineapple juice and high-pressure
technique were applied, the browning reaction was reduced in apple slices. Such
inhibitory effect is due to the presence of bromelin, sulfhydryl groups, citric, or malic
acid (Perera et al., 2010). Similarly, the prevention of browning effect was also
observed in peeled banana when pineapple juice was used (Chaisakdanugull et al.,
2007). However, in some studies, bromelin was found to be ineffective in preventing
browning of apple juice (Tochi et al., 2009).
Peroxidases (POD) are generally isolated from plants, animals, and
microorganisms. These are oxidase enzymes which use hydrogen peroxide as a
catalyst in the oxidation reaction of polyphenols, aminophenols, monophenols,
and diphenols (Fatibello-Filho & Vieira, 2002). They are heat-stable and hence
used as a parameter to increase the efficiency in bleaching (Aguero et al., 2008).
POD lead to undesirable changes in colour, flavour, texture, and nutritional values in
foods (Gonçalves et al., 2007). The browning of sugar cane juice is due to the
presence of enzymes, POD and PPO, which oxidize phenolics compounds (Qudsieh
et al., 2002). The enzymatic activities of POD and PPD enzymes were studied in
different cultivars of grapes. In these studies, the POD extracts showed similar
activity in both the soluble and bound fractions, and highest PPD activity was
observed in cultivar Ruby. PPD and POD activities in cultivars Benitaka and
Ruby decrease when juice extracts were treated with higher temperature and longer
duration. It was observed that enzyme inactivation was achieved at 85 C with
10 min exposure time, but the thermal treatments were not sufficient to inactivate the
enzymes completely. For example, the thermal treatment in jam, jellies, and juices
causes reduction in PPO and POD activities, but not sufficient to inactivate the
enzymes (Freitas et al., 2008). Similar activities were also reported in processing of
guavas (Zanatta et al., 2006).
In other studies, it was shown that apple peel from Fuji and Gala cultivars when
compared to its pulp had elevated enzymatic activity both for PPO and POD. When
heat treatment was done in the concentrated extracts of pulp and peel, there was a
decline in PPO enzyme activity and total inactivation was also achieved after 10 min
of heat treatment at 75 C. However, such case of total inactivation was not observed
for POD enzyme activity (Valderrama et al., 2001). Studies have shown that the
enzymatic activity of PPO in fresh broccoli was higher compared to bleached
broccoli. In case of POD, the activity was found to be lower in bleached broccoli,
indicating that bleaching was partially effective in denaturation of these enzymes
(Lopes & Clemente, 2002).
78 M. D. Heirangkhongjam et al.
Taste and flavour are the two major attributes in determining the overall quality of
various fruits and vegetables. Taste is determined by the contents of sugar, tannins,
phenols, organic acids, and other compounds. Analysis of flavour compounds has
given us an inclusive knowledge on the chemical compounds responsible for flavour
sensations of fruits and vegetables. However, different enzymes have multiple
effects on these attributes of fruits and vegetables. Enzymes such as peroxidases
and lipoxygenases are responsible for off-flavour formation in fruits and vegetables
(Bhowmik & Dris, 2004). The enzyme peroxidases are responsible for deterioration
in colour, flavour, texture, and loss in nutritional qualities in raw or processed fruits
and vegetables. The off-flavour is often linked with the oxidation of phenolic
compounds and indigenous lipids. It was observed that enzyme inactivation was
achieved at 70 C with 15 min exposure time (Sessa & Anderson, 1981). Therefore,
inhibition of such enzymes during processing is a must in order to retain or improve
the aroma and flavour.
Another enzyme that has a major effect in flavour and aroma is lipogenase. This
enzyme produces free radicals and conjugated unsaturated hydroperoxy acids by
catalytic oxidation of polyunsaturated fatty acids. Then, these free radicals interact
with other constituents like proteins, vitamins, phenolics, etc. present in fruits and
vegetables which helps in enhancing the flavour and aroma of the product. However,
these aromatic compounds may produce off-flavours in Brassicaceae family.
According to the reports of Sheu and Chen (1991), increase in colour losses and
development of off-flavour were observed in broccoli and asparagus during storage
of non-blanched and under-blanched products.
Furthermore, the volatile compounds such as aldehydes, alcohols, ketones, esters,
lactones, etc. are related to flavour and aromatic characteristics in foods (Beaulieu &
Baldwin, 2002). For example, the compounds present in alcohols and aldehydes are
extensively used as food additives due to the aroma referred to as ‘green touch’, a
characteristic observed in freshly harvested fruits (Schwab et al., 2008). These
volatile compounds are synthesized using various substrates like amino acids, fatty
acids, and carotenoids (Goff & Klee, 2006). The main enzymes involved in the
synthesis of volatile compounds from fatty acids are lipoxygenase, alcohol dehydro-
genase, hydro peroxide lyase, and (3Z ): (2E)-enal isomerise (Schwab et al., 2008).
These enzymes are used in the extraction from fruits and vegetables such as banana,
soy, tomato, and olive where they subsequently react with fatty acids to produce
volatile compounds (Akacha & Gargouri, 2009).
In order to improve the taste and flavour in citrus fruits, enzymes limoninase and
naringinase can be enzymatically tailored by degrading the bitter taste compounds
such as limonin and naringin (Ribeiro et al., 2010). The formation of limonin can
also be prevented by using limonoate dehydrogenase, as it catalyses the oxidation of
its precursor lactone-A-ring to 17-dehydrolimonoate, a non-bitter derivative which
cannot be changed into limonin (Merino et al., 1997). Bitter compounds in citrus
fruits can be reduced by using adsorbing polymers, such as Amberlite XAD-16HP
and Dowex Optipore L285 resins. Besides acting as a debittering agent, Dowex
80 M. D. Heirangkhongjam et al.
Optipore L285 can also induce other modifications in juice processing like reduction
of total titratable acidity (TTA), increasing total soluble solids (TSS), the ratio of
TSS to TTA, pH, etc. (Kola et al., 2010).
Texture is one of the important quality attributes of fruits and vegetables (Pan et al.,
2014; Grassin & Coutel, 2009; Guillemin et al., 2008; Jensen et al., 2004; Degraeve
et al., 2003). However, the texture of processed fruits and vegetables, especially the
softer ones, such as strawberry, raspberry, and tomatoes, is adversely affected by
thermal processing treatments like blanching, sterilization, freezing or pasteuriza-
tion, and other mechanical method which may result in softening (Grassin & Coutel,
2009; Guillemin et al., 2008; Degraeve et al., 2003).
The texture and structural integrity of fruits and vegetables depend on the
composition of cell walls present in them. These cell walls are composed of an
interlinked fibrous structure of cellulose embedded in a matrix of pectin,
hemicelluloses, and celluloses (Guillemin et al., 2008; Sila et al., 2008). The enzyme
pectin methylesterase (PME) bound to the cell wall is a pectin degrading enzyme
which results in demethoxylation of pectin (de-esterification of the methylated
carboxy groups of polygalacturonic pectin), releasing methanol and forming
carboxylated pectin. This carboxylic acid is said to interact with calcium, resulting
in firmness of fruits by strengthening the cell wall (strong pectate network with
added calcium) (Pan et al., 2014; Sila et al., 2008; Guillemin et al., 2008; Degraeve
et al., 2003). This process is known as chelation, and it helps to overcome the
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 81
or suberin in vegetable surface (Suutarinen et al., 2003). As per a study by Toker and
Bayindirli, it was observed that the enzymatic peeling was easier at relatively
moderate temperatures such as 44–47 C for nectarines, 41–46 C for peaches,
and 45 C for apricots with a pH ranging from 3 to 4.1 (Toker & Bayιndιrlι,
2003). The steps involved during enzymatic peeling are illustrated in Fig. 3.1.
The first step is selection of fruits or vegetables and washing them with water. At
times, hot water treatment or scalding is done prior to enzymatic peeling to improve
the peeling process and have a high quality end product (Pretel et al., 2008, 2010). A
study on grapefruit as early as 1974 demonstrated dipping the fruit in a water bath of
60 C for 30–35 min resulted in a good quality product (Bruemmer et al., 1978). The
advantage of scalding is that it reduces the viscosity of pectin as well as enhanced the
ability of the peel to absorb the enzyme solution, thereby helping in enzymatic
peeling process (Pretel et al., 2008) (Fig. 3.1).
Therefore, enzymatic peeling has wide range of applications in fruits and
vegetables such as grapefruit, orange, mandarin, apricot, peaches, potato, carrot,
and Swedish turnips (Suutarinen et al., 2003; Toker & Bayιndιrlι, 2003). This
method is a significant alternative in food processing industry because of its
advantages in improving the quality of products, reducing wastage, minimal water
use, reduction in contamination, and being cost-effective (Toker & Bayιndιrlι,
2003).
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 83
Fig. 3.1 Steps in enzymatic peeling. (Adapted from Pretel MT, Sánchez-Bel P, Egea I & Romojaro
F. (2010). Enzymatic peeling of citrus fruits. In Bayindirli (Ed.), Enzymatic processing of fruits and
vegetables: Chemistry and engineering applications (pp. 145–174). CRC Press, Taylor and Francis
Group)
The natural liquid substance contained in fruits and vegetables is commonly known
as juice. It can be obtained or prepared directly by extraction, pressing, or diffusion
from fruits or vegetables and such juices are meant for direct consumption. They can
be broadly categorized as juices without pulp (clarified/cloudy) or juices with pulp
such as purees, nectars, and pulps (Ceci & Lozano, 2010; Cautela et al., 2010). In
juice processing, different enzymes are extensively used in fruit and vegetable for
higher yield, clarification, and improvement in filtration, resulting in higher quality
of juices (Fleuri et al., 2016). Some of the commonly used enzymes in fruit and
vegetable juice processing are pectinases, hemicellulases, and cellulases. In juice
processing of fruits and vegetables, multiple steps are involved and below are the
standard procedures. The steps involved in fruit juice processing are also illustrated
in Fig. 3.2.
Fig. 3.2 Steps in juice processing. (Adapted from Fleuri, L. F., Delgado., C. H. O., Novelli, P. K.,
Pivetta, M. R., do Prado, D. Z., & Simon, J. W. (2016). Enzymes in food and beverage processing
(pp. 255–280). CRC Press, Taylor and Francis Group)
extraction, clarification, and filtration (Claus & Mojsov, 2018). Moreover, the skin
and cell walls of grapes contain several essential compounds such as anthocyanins
and tannins. These compounds are responsible for colour and structure of wine
(Gómez-Plaza et al., 2010; Bruchmann & Fauveau, 2009).
The natural enzymes present in grapes and those obtained from yeast, fungi, and
bacteria collectively play an important role in wine making process (Gómez-Plaza
et al., 2010). Usually, the manufacturers extend the action of endogenous enzymes
by adding exogenous enzymes during production (Gómez-Plaza et al., 2010). The
commonly used commercial enzymes include pectinases, glucanases, glycosidases,
lysozymes, and ureases (Gómez-Plaza et al., 2010). The application of these exoge-
nous enzymes and yeast strains during processing increases the rate of production,
yield, and quality of the wine (Toushik et al., 2017).
The standard steps in wine processing are illustrated in Fig. 3.3.
These steps can be broadly categorized in three phases, namely pre-fermentation,
fermentation, and post-fermentation.
Pre-fermentation
This phase involves crushing the fruit and extraction of juice. In red wine prepara-
tion, the skin is not separated, while it is separated in white wine. During the
processing of red wine, pulp, skin, and seeds of grapes are kept together after
crushing and during fermentation in order to extract colour and flavour
(Byarugaba-Bazirake, 2008). The colour of red wine is due to the presence of
anthocyanins and tannins in grape skin and seed (Gómez-Plaza et al., 2010). In
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 87
this phase, exogenous enzymes derived from Aspergillus species such as pectinases,
cellulases, and hemicellulases (xylanases and galactanases) are employed, which
help in expediting the extraction of juice and colour control (Espejo, 2020; Toushik
et al., 2017; El Darra et al., 2016; Kelebek et al., 2007; Bautista-Ortίn et al., 2005).
These macerating enzymes also modify the stability, taste, structure, and mouth feel
of red wine (Gao et al., 2019).
Furthermore, maceration of the cell walls by pectinases results in the liberation of
liquid and other phenolic compounds (Espejo, 2020; Gao et al., 2019). It has been
observed in different studies that wines prepared by enzyme treatment tend to have
phenolic and tannin content, as well as colour intensity (Romero-Cascales et al.,
2008; Revilla & Gonzalez-San Jose, 2003). Sometimes, maceration is accompanied
by pressing (wine press extract must or wine from crushed grapes), resulting in better
yield (Gómez-Plaza et al., 2010). Another important attribute in wine making is its
aroma; reductases such as glycosidases and polyphenol reductase help in aroma
extraction and polyphenol reduction, respectively (Espejo, 2020). Glycosidases
improve the aroma of wines by releasing aromatic compounds from their
non-aromatic precursors or non-odouriferous sugar compounds (Zhu et al., 2014).
As per a study, treatment of Albarino wine with pectinolytic preparations and
glycosidase resulted in an improved aroma (Armada et al., 2010). However, in
industrial processing, usually exoglycosidases and β-glucosidase from Aspergillus
niger are used (Gómez-Plaza et al., 2010).
Fermentation
In this phase, yeast is added to the must in red wine to initiate fermentation
(Byarugaba-Bazirake, 2008). During the process, the sugars present in the juice
are converted into alcohol and release carbon dioxide. In this period, pectin plays an
important role by preventing the diffusion of intercellular components such as
phenolic and aroma compounds into the must (Claus & Mojsov, 2018; Bruchmann
& Fauveau, 2009).
Post-fermentation
This phase involves different steps such as clarification, filtration, and microbial
stabilization with the help of different enzymes. Proteases are used at the time of
clarification, while pectinase and β glucanase are used during filtration and lysozyme
for microbial stabilization (Espejo, 2020). After pressing, the grape must is quite
turbid and is rich in solid particles (Armada & Falqué, 2007). Thus, clarification is an
important step to improve the quality of wine, especially in white wine (Gómez-
Plaza et al., 2010). Moreover, activated carbon, bentonite, and pectinase may be
added to aid in clarification or physical removal of suspended matter in the must
(Gómez-Plaza et al., 2010). Commercial enzymes that help in clarification are:
(a) Pectin lyase which results in destabilization of the cloud, thereby reducing
viscosity.
(b) Pectin methylesterase (PME) results in demethylation of pectin, aiding
polygalacturonase in hydrolysis of pectin and leading to cloud flocculation
88 M. D. Heirangkhongjam et al.
Enzymes present in fruits and vegetables play a huge role in determining the texture,
colour, flavour, and taste attributes of the processed products (Oey, 2010). The
continued enzymatic activity in fruits and vegetables affects the storage quality,
shelf life, and palatability of the product. Therefore, several processing methods such
as grinding, crushing, slicing, juices, or preservation (pickling) are used to prolong
the shelf life and reduce wastage of fruits and vegetables (Oey, 2010). Hence, it is
very important to control the stability and activity of endogenous enzymes present in
fruits and vegetables during food processing (Oey, 2010).
Apart from the conventional techniques of processing such as blanching, heating,
or freezing, new and highly advanced processing techniques like high hydrostatic
pressure (HHP), pulse electric field (PEF), and ultrasound have been introduced
successfully. The major advantage of these new techniques is the use of non-thermal
technology, which helps in retaining the sensory attributes and nutritional content of
the product (Oey, 2010; Jaiswal & Sharma, 2016). In HHP technique, it employs
high pressure range of 100–600 MPa, resulting in enzyme and microbial inactivity
which may affect the shelf life of the products (Briones-Labarca et al., 2015). In PEF
technique, being a non-thermal processing, it uses a series of short and high voltage
pulses to inactivate microbes or enzymes (such as peroxidases and polyphenol
oxidases) in food (Segovia et al., 2015). However, in ultrasound processing tech-
nique, high frequency short pulses are used for inactivation (Jaiswal & Sharma,
2016). These non-thermal processing techniques are effective at ambient or
sub-lethal temperatures and minimize the adverse thermal effects on the nutritional
content and quality of fruits and vegetables (Tiwari et al., 2009).
However, these non-thermal processing techniques still have several drawbacks,
such as high equipment and processing cost, tedious to operate, hazardous, requires
stringent process control operations, etc. (Jaiswal & Sharma, 2016). Nonetheless,
multiple advantages are rendered by these techniques in food processing industry
over conventional thermal techniques, which affect not only the enzymes, but also
the texture, taste, and colour of the product compelled for further investigation and
improvement (Oey, 2010).
2018). This technique can be applied in solid, semi solid liquid, or particulate food
products (Augusto et al., 2018).
The high pressure (HP) may increase the shelf life of fruits and vegetables,
especially when combined with temperature by resulting in enzyme activity or
inactivity and pressure-induced gel formation (Guerrero-Beltrán et al., 2005). In
HHP technique, enzyme inactivation may be affected by the type of food, pH,
temperature, and duration of treatment (de Castro Leite Júnior et al., 2017;
Guerrero-Beltrán et al., 2005).
The HHP unit consists of a pressure vessel, pressure generator, temperature
control, and pressure handling system. The HHP processing can be of two types—
batch (closed vessel system) or semi-continuous (number of closed vessels). In this
process, the hydrostatic pressure at a given point is transformed rapidly and uni-
formly in all directions. The intensity of HHP is determined by the process
parameters like pressure, treatment duration, and temperature. The entire HHP
processing cycle takes a few minutes and it can be performed at pressures as high
as 1400 MPa and temperatures between less than 0–150 C (Oey, 2010) (Augusto
et al., 2018; Oey, 2010). The maintenance of a product inside a vessel at a high-
pressure results in molecular changes affecting the enzymes (de Castro Leite Júnior
et al., 2017).
Several studies have been conducted to determine the effect of HHP (at times
combined with thermal processing) on enzyme activity in various fruits and
vegetables as shown in Table 3.5. Enzymes like PME, POD, and PPO are considered
baroresistant with POD and PME being the most and least resistant, respectively
(Augusto et al., 2018). Hence, some studies have found that PPO and POD are not
affected substantially by HHP (Augusto et al., 2018).
the enzyme pectate lyase at pressures more than 150 MPa (Calligaris et al., 2012).
Therefore, it is not recommended to use HPH independently and preferably; it needs
to be coupled with other processing methods for microbial or enzyme inactivation
(Augusto et al., 2018; de Castro Leite Júnior et al., 2017; Calligaris et al., 2012;
Welti-Chanes et al., 2009).
when the technique, high power ultrasound with low frequency, is combined with
temperature/heat, the process is known as thermosonification or pressure
(manosonification) (Bourke et al., 2010). The combination of heat and ultrasound
helps to ensure product stability and inactivation of enzymes and microbes, thereby
resulting in retaining the quality and extending the shelf life of fruits and vegetables,
especially juices (Rojas et al., 2017; Saeeduddin et al., 2015). The inactivation of
microorganisms or enzymes in this technique may be due to physical factors such as
caviation or mechanical effects (Bourke et al., 2010; O’Donnell et al., 2010).
Ultrasonic processing in fruits and vegetables focuses on the inactivation of
endogenous enzymes which are more resistant to heat treatments (Feng et al.,
2008). Studies have It has been observed that ultrasonic processing either in combi-
nation with heat or pressure has minimal effect on quality of fruit juices such as
orange juice, guava, and strawberry juice (Bourke et al., 2010). Further, it has been
observed that high ultrasound and longer processing times may be required for
enzyme inactivation as the pulp complicates the inactivation process (Rojas et al.,
2017). Hence, ultrasound technique is usually combined with a thermal process to
increase the rate of inactivation of enzymes in juices (Chen et al., 2019; Rojas et al.,
2017; Anaya-Esparza et al., 2017; Abid et al., 2014). PME was also found to be
inactivated in lemon juice (Knorr et al., 2004) and mousambi (Siwach & Kumar,
2012) by using ultrasonic processing along with heat. Likewise, peroxidase enzyme
present in watercress was also found to be inactivated by the use of thermosonication
(Cruz et al., 2006).
In PEF processing technique, electrochemical effects and ohmic heating are respon-
sible for changing the structure and function of enzymes present in the food. At
times, large specific energy inputs are required to inactivate enzymes (Poojary et al.,
2017). Enzyme activity can be affected by a number of factors such as properties of
the enzyme, treatment parameters, processing conditions as well as the condition of
the medium (Poojary et al., 2017).
The PEF unit consists of a treatment chamber, pulse generator, a pulse monitoring
system, and a temperature monitor. It can be conducted in two ways—continuous or
batch processing. The unit also contains a fluid handling system for liquid foods in
continuous PEF processing (Min et al., 2007). The design of the treatment chamber
plays a key role in distribution of uniform temperature inside the PEF chamber (Oey,
2010). In PEF, the duration of processing takes substantially lesser time (micro to
milli seconds) as compared to other methods (Oey, 2010).
However, in PEF processing, the activity on enzyme may either limit its activa-
tion/inactivation or may not be affected at all (Poojary et al., 2017). In this process,
multiple factors such as food matrix, pH, enzyme dissolving medium, and certain
treatment conditions affect the enzyme treatment in fruits and vegetables (Poojary
et al., 2017; Oey, 2010). Two important parameters that affect the intensity of PEF
processing are electric field intensity and the total duration of treatment per energy
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 93
(Oey, 2010). A higher electric field and treatment duration may result in enzyme
inactivation, while a lower electric field and duration may result in enzyme activa-
tion (Poojary et al., 2017). Also, increasing pulse width lowers enzyme activity
(Poojary et al., 2017).
Furthermore, several studies have been conducted to assess the effect of PEF
processing on various food enzymes such as polyphenol oxidase, lipoxygenase,
peroxidase, and pectin methyl esterase (Poojary et al., 2017; Aguiló-Aguayo et al.,
2009; Noci et al., 2008; Espachs-Barroso et al., 2006) and are given in Table 3.6.
The findings from these studies indicate that a longer treatment duration, higher
electric field intensity, and pulse width result in enzyme inactivation.
that contain water and ionic salts in higher amount are found to be more effective
with this treatment (Miller & Silva, 2012). In other foods, this process has many
advantages like uniform and rapid heating which ultimately helps in nutritional and
sensory attributes of the processed products. As per the study by Somavat et al.
(2013), B. coagulans (ATCC 8038) in tomato juice when treated with OH at 60 Hz
and 10 kHz expedites the inactivation process as compared to conventional treatment
(Somavat et al., 2013). However, microwave heating is another common
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 95
The regulation of enzymes for its application in food industry is very complex and
differs from county to country. The European Union (EU) classifies food enzymes as
processing aids of food additives. Majority of the enzyme preparations used in food
processing are categorized as processing aids by European Union since they have a
role in the technological aspect of food processing stage and not in the final product
(Aehle, 2007). The processing aid may not be labelled (Bruchmann & Fauveau,
2009). The enzyme lysozyme used in wine processing is considered as a food
additive (Bruchmann & Fauveau, 2009) since it has a role to play in the final product
and is regulated under Food Additives Directive (95/2). A vertical legislation of EU
on fruit juices (Council Directive 93/77/EEC) allows pectinolytic, proteolytic, and
amylolytic enzymes, while another regulation (Council Regulations 82/87/EEC)
allows only pectinolytic enzymes on wines (Aehle, 2007). The Organization
Internationale de la Vigne et du Vin (OIV) regulates the enzymes used in wine
processing in EU (Gómez-Plaza et al., 2010; Bruchmann & Fauveau, 2009; Aehle,
2007) and has recognized the important role of pectinases, hemicellulases, cellulose,
β-glucanase, and glycosidase. The European Commission Regulation 1493/1999
authorises the use of pectinases from Aspergillus niger, β-glucanase from
Trichoderma harzanium and urease from Lactobacillus fermentum. Pectinases are
assigned a GRAS status by United States Food and Drug Administration
(Bruchmann & Fauveau, 2009). In India, Food Safety and Standards Authority of
India (FSSAI) regulates the use and safety of food enzymes. As per the regulation
Food Safety and Standards (Food Products Standards and Food Additives) (Amend-
ment) Regulations, 2015, the food regulator had permitted the use of processing aids
in bread. The enzymes permitted for usage are glucose oxidase, lipase, and xylanase.
They are obtained from various microbial sources.
In some EU countries, namely Denmark, France, and United Kingdom, approval
is required for the use of any food enzymes (Aehle, 2007). However, in countries
like Poland, China, Japan, Australia, New Zealand, Canada, Brazil, and Mexico,
approval is needed for the use of enzymes produced traditionally, especially if they
are new enzymes (Aehle, 2007). On the other hand, countries such as Thailand,
Korea, and Taiwan require registration prior to their enzyme use, while USA
requires a GRAS (generally regarded as safe) assessment, GRAS notice, or Food
additive (Aehle, 2007). Even though countries have different set regulations, enzyme
preparations must comply with specifications recommended by Joint FAO/WHO
Expert Committee on Food Additives (JECFA) and by Food Chemical Codex (FCC)
for food enzymes (Bruchmann & Fauveau, 2009; Grassin & Coutel, 2009).
96 M. D. Heirangkhongjam et al.
References
Abid, M., Jabbar, S., Hu, B., Hashim, M. M., Wu, T., Lei, S., Khan, M. A., & Zeng, X. (2014).
Thermosonication as a potential quality enhancement technique of apple juice. Ultrasonics
Sonochemistry, 21(3), 984–990. https://doi.org/10.1016/j.ultsonch.2013.12.003
Achir, N., Dhuique-Mayer, C., Hadjal, T., Madani, K., Pain, J. P., & Dornier, M. (2016). Pasteuri-
zation of citrus juices with ohmic heating to preserve the carotenoid profile. Innovative Food
Science and Emerging Technologies, 33, 397–404.
Aehle, W. (Ed.). (2007). Enzymes in industry: Production and applications (3rd completely rev.
ed.). Wiley-VCH.
Aguero, M. V., Ansorena, M. R., Roura, S. I., & Del Valle, C. E. (2008). Thermal inactivation of
peroxidase during blanching of butternut squash. Food Science and Technology, 41, 401–407.
Aguilar-Rosas, S., Ballinas-Casarrubias, M., Elias-Ogaz, L., Martin-Belloso, O., & Ortega-Rivas,
E. (2013). Enzyme activity and colour changes in apple juice pasteurised thermally and by
pulsed electric fields. Acta Alimentaria, 42, 45–54.
Aguiló-Aguayo, I., Oms-Oliu, G., Soliva-Fortuny, R., & Martín-Belloso, O. (2009). Changes in
quality attributes throughout storage of strawberry juice processed by high-intensity pulsed
electric fields or heat treatments. LWT—Food Science and Technology, 42(4), 813–818. https://
doi.org/10.1016/j.lwt.2008.11.008
Akacha, N. B., & Gargouri, M. (2009). Enzymatic synthesis of green notes with hydroperoxide-
lyase from olive leaves and alcohol-dehydrogenase from yeast in liquid/gas reactor. Process
Biochemistry, 44(10), 1122–1127.
Albersheim, P., Darvill, A. G., O’Neill, M. A., Schols, H. A., & Voragen, A. G. J. (1996). An
hypothesis: The same polysaccharides are components of the primary cell walls of all higher
plants. In J. Visser & A. G. J. Voragen (Eds.), Pectin and pectinases (pp. 47–55). Elsevier
Science B.V.
Alkorta, I., Garbisu, C., Llama, M. J., & Serra, J. L. (1998). Industrial applications of pectic
enzymes: A review. Process Biochemistry, 33(1), 21–28.
Anaya-Esparza, L. M., Velázquez-Estrada, R. M., Roig, A. X., García-Galindo, H. S., Sayago-
Ayerdi, S. G., & Montalvo-González, E. (2017). Thermosonication: An alternative processing
for fruit and vegetable juices. Trends in Food Science & Technology, 61, 26–37. https://doi.org/
10.1016/j.tifs.2016.11.020
Andreou, V., Dimopoulos, G., Katsaros, G., & Taoukis, P. (2016). Comparison of the application of
high pressure and pulsed electric fields technologies on the selective inactivation of endogenous
enzymes in tomato products. Innovative Food Science and Emerging Technologies, 38,
349–355. https://doi.org/10.1016/j.ifset.2016.07.026
Arjmandi, M., Otón, M., Artés, F., Artés-Hernández, F., Gómez, P. A., & Aguayo, E. (2016). Semi-
industrial microwave treatments positively affect the quality of orange-colored smoothies.
Journal of Food Science and Technology, 53, 3695–3703.
Armada, L., & Falqué, E. (2007). Repercussion of the clarification treatment agents before the
alcoholic fermentation on volatile composition of white wines. European Food Research and
Technology, 225(3–4), 553–558. https://doi.org/10.1007/s00217-006-0453-3
Armada, L., Fernández, E., & Falqué, E. (2010). Influence of several enzymatic treatments on
aromatic composition of white wines. LWT—Food Science and Technology, 43(10),
1517–1525.
Augusto, P. E. D., Tribst, A. A. L., & Cristianini, M. (2018). High hydrostatic pressure and high-
pressure homogenization processing of fruit juices. In Fruit juices (pp. 393–421). Elsevier.
https://doi.org/10.1016/B978-0-12-802230-6.00020-5
Baker, R. A., & Bruemmer, J. H. (1989). Quality and stability of enzymatically peeledand sectioned
citrus fruit. In J. J. Jen (Ed.), Quality factors of fruits and vegetables (pp. 140–148). American
Chemical Society.
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 97
Bansal, N., Tewari, R., Gupta, J. K., Soni, S. K., & Soni, R. (2011). A novel strain of Aspergillus
niger producing a cocktail of industrial depolymerising enzymes for the production of second
generation biofuels. BioResources, 6, 552–569.
Bartolome, L. G., & Hoff, J. E. (1972). Firming of potatoes. Biochemical effects of preheating.
Journal of Agricultural and Food Chemistry, 20(2), 266–270. https://doi.org/10.1021/
jf60180a028
Bautista-Ortίn, A. B., Martinez-Cutillas, A., Ros-Garcia, J. M., Lopez-Roca, J. M., & Gomez-Plaza,
E. (2005). Improving colour extraction and stability in red wines: The use of maceration
enzymes and enological tannins. International Journal of Food Science and Technology,
40(8), 867–878. https://doi.org/10.1111/j.1365-2621.2005.01014.x
Baysal, A. H., & Icier, F. (2010). Inactivation kinetics of Alicyclobacillus acidoterrestris spores in
orange juice by ohmic heating: Effects of voltage gradient and temperature on inactivation.
Journal of Food Protection, 73, 299–304.
Beaulieu, J. C., & Baldwin, E. A. (2002). Flavor and aroma of fresh-cut fruits and vegetables. In
O. Lamikanra (Ed.), Fresh-cut fruits and vegetables: Science, technology, and market
(pp. 391–425). CRC Press.
Belluzzo, A. S. F., Fleuri, L. F., Macedo, J. A., & Macedo, G. A. (2009). Characterization of Biuti
peach polyphenol oxidase. Food Science and Biotechnology, 18, 878–883.
Ben-Shalom, N., Levi, A., & Pinto, R. (1986). Pectolytic enzyme studies for peeling of grapefruit
segment membrane. Journal of Food Science, 51(2), 421–423.
Berg, M., Roubos, J., & A.; & Parenicova, L. (2010). Enzymes in fruit and vegetable processing:
Future trends in enzyme discovery, design, production and application. In A. Bayindirli (Ed.),
Enzymes in fruit and vegetable processing: Chemistry and engineering applications
(pp. 341–358). CRC Press.
Berry, R. E., Baker, R. A., & Bruemmer, J. H. (1988). Enzyme separated sections: A new lightly
processed citrus product. In R. Goren & K. Hendel (Eds.), Proceedings of the Sixth Interna-
tional Citrus Congress (pp. 1711–1716). Balaban.
Bhowmik, P. K., & Dris, R. (2004). Enzymes and quality factors of fruits and vegetables. In R. Dris
& S. M. Jain (Eds.), Production practices and quality assessment of food crops (pp. 1–25).
Springer.
Bourke, P., Tiwari, B., O’Donnell, C., & Cullen, P. J. (2010). Effect of ultrasonic processing on
food enzymes of industrial importance. Trends in Food Science and Technology, 21(7), 358.
Briones-Labarca, V., Plaza-Morales, M., Giovagnoli-Vicuña, C., & Jamett, F. (2015). High hydro-
static pressure and ultrasound extractions of antioxidant compounds, sulforaphane and fatty
acids from Chilean papaya (Vasconcellea pubescens) seeds: Effects of extraction conditions and
methods. LWT—Food Science and Technology, 60(1), 525–534. https://doi.org/10.1016/j.lwt.
2014.07.057
Bruchmann, A., & Fauveau, C. (2009). Enzymes in potable alcohol and wine production. In R. J.
Whitehurst & M. van Oort (Eds.), Enzymes in food technology (pp. 195–210). Wiley-Blackwell.
https://doi.org/10.1002/9781444309935.ch9
Bruemmer, J. H., Griffin, A. W., & Onayami, O. (1978). Sectionizing grapefruit by enzyme
digestion. Proceedings of the Florida State Horticultural Society, 91, 112–114.
Byarugaba-Bazirake, G. W. (2008). The effect of enzymatic processing on banana juice and wine.
Dissertation, Institute of Wine Technology at Stellenbosch University.
Calligaris, S., Foschia, M., Bartolomeoli, I., Maifreni, M., & Manzocco, L. (2012). Study on the
applicability of high-pressure homogenization for the production of banana juices. LWT—Food
Science and Technology, 45(1), 117–121. https://doi.org/10.1016/j.lwt.2011.07.026
Carpita, N., & Gibeaut, D. (1993). Structural models of primary cell walls in flowering plants. The
Plant Journal: For Cell and Molecular Biology, 3, 1–30.
Carrín, M. (2004). Characterization of starch in apple juice and its degradation by amylases. Food
Chemistry, 87(2), 173–178. https://doi.org/10.1016/j.foodchem.2003.10.032
Castro, S. M., Saraiva, J. A., Lopes-da-Silva, J. A., Delgadillo, I., Loey, A. V., Smout, C., &
Hendrickx, M. (2008). Effect of thermal blanching and of high pressure treatments on sweet
98 M. D. Heirangkhongjam et al.
green and red bell pepper fruits (Capsicum annuum L.). Food Chemistry, 107(4), 1436–1449.
https://doi.org/10.1016/j.foodchem.2007.09.074
Cautela, D., Castaldo, D., Servillo, L., & Giovan, A. (2010). Enzymes in citrus juice processing. In
A. Bayindirli (Ed.), Enzymatic processing in fruits and vegetables: Chemistry and engineering
applications (pp. 197–214). CRC Press, Taylor and Francis Group.
Ceci, L. N., & Lozano, J. E. (2010). Use of enzymes for non-citrus fruit juice production. In
A. Bayindirli (Ed.), Enzymatic processing in fruits and vegetables: Chemistry and engineering
applications (pp. 175–195). CRC Press, Taylor and Francis Group.
Chaisakdanugull, C., Theerakulkait, C., & Wrolstad, R. E. (2007). Pineapple juice and its fractions
in enzymatic browning inhibition of banana (Musa [AAA Group] Gros Michel). Journal of
Agriculture and Food Chemistry, 55, 4252–4257.
Chen, D., Xi, H., Guo, X., Qin, Z., Pang, X., Hu, X., Liao, X., & Wu, J. (2013). Comparative study
of quality of cloudy pomegranate juice treated by high hydrostatic pressure and high tempera-
ture short time. Innovative Food Science and Emerging Technologies, 19, 85–94.
Chen, L., Bi, X., Guo, D., Xing, Y., & Che, Z. (2019). The effect of high-power ultrasound on the
quality of carrot juice. Food Science and Technology International, 25(5), 394–403. https://doi.
org/10.1177/1082013219825736
Claus, H., & Mojsov, K. (2018). Enzymes for wine fermentation: Current and perspective
applications. Fermentation, 4(3), 52. https://doi.org/10.3390/fermentation4030052
Constabel, C. P., & Ryan, C. A. (1998). A survey of wound- and methyl jasmonate-induced leaf
polyphenol oxidase in crop plants. Phytochemistry, 47(4), 507–511.
Cruz, R. M. S., Vieira, M. C., & Silva, C. L. M. (2006). Effect of heat and thermosonication
treatments on peroxidase inactivation kinetics in watercress (Nasturtium officinale). Journal of
Food Engineering, 72(1), 8–15. https://doi.org/10.1016/j.jfoodeng.2004.11.007
Cunha, A. G., & Gandini, A. (2010). Turning polysaccharides into hydrophobic materials: A critical
review. Part 2. Hemicelluloses, chitin/chitosan, starch, pectin and alginates. Cellulose, 17,
1045–1065.
de Carvalho, J. M., Maia, G. A., da Fonseca, A. V., de Sousa, P. H., & Rodrigues, S. (2015). Effect
of processing on physicochemical composition, bioactive compounds and enzymatic activity of
yellow mombin (Spondias mombin L.) tropical juice. Journal of Food Science and Technology,
52, 1182–1187.
de Castro Leite Júnior, B. R., Tribst, A. A. L., & Cristianini, M. (2017). Effect of high-pressure
technologies on enzymes applied in food processing. In M. Senturk (Ed.), Enzyme inhibitors
and activators. InTech. https://doi.org/10.5772/66629
Degraeve, P., Saurel, R., & Coutel, Y. (2003). Vacuum impregnation pretreatment with
pectinmethylesterase to improve firmness of pasteurized fruits. Journal of Food Science,
68(2), 716–721. https://doi.org/10.1111/j.1365-2621.2003.tb05738.x
Dervilly, G., Leclercq, C., Zimmerman, D., Roue, C., Thibault, J. F., & Sauliner, L. (2002).
Isolation and characterization of high molecular mass water-soluble arabinoxylans from barley
malt. Carbohydrate Polymers, 47, 143–149.
Dhillon, G. S., Kaur, S., & Brar, S. K. (2013). Perspective of apple processing wastes as low-cost
substrates for bioproduction of high value products: A review. Renewable and Sustainable
Energy Reviews, 27, 789–805.
Dinu, D., Nechifor, M. T., Stoian, G., Costache, M., & Dinischiotu, A. (2007). Enzymes with new
biochemical properties in the pectinolytic complex produced by Aspergillus niger MIUG 16.
Journal of Biotechnology, 131, 128–137.
Eisenmenger, M. J., & Reyes-De-Corcuera, J. I. (2009). High pressure enhancement of enzymes: A
review. Enzyme and Microbial Technology, 45, 331–347.
El Darra, N., Turk, M. F., Ducasse, M.-A., Grimi, N., Maroun, R. G., Louka, N., & Vorobiev,
E. (2016). Changes in polyphenol profiles and color composition of freshly fermented model
wine due to pulsed electric field, enzymes and thermovinification pretreatments. Food Chemis-
try, 194, 944–950. https://doi.org/10.1016/j.foodchem.2015.08.059
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 99
Espachs-Barroso, A., Van Loey, A., Hendrickx, M., & Martín-Belloso, O. (2006). Inactivation of
plant pectin methylesterase by thermal or high intensity pulsed electric field treatments. Innova-
tive Food Science and Emerging Technologies, 7(1–2), 40–48. https://doi.org/10.1016/j.ifset.
2005.07.002
Espejo, F. (2020). Role of commercial enzymes in wine production: A critical review of recent
research. Journal of Food Science and Technology. https://doi.org/10.1007/s13197-020-
04489-0
Fatibello-Filho, O., & Vieira, I. C. (2002). Uso analítico de tecidos e de extratos brutos vegetais
como fonte enzimática. Química Nova, 25(3), 455–464.
Feng, H., Yang, W., & Hielscher, T. (2008). Power ultrasound. Food Science and Technology
International, 14(5), 433–436. https://doi.org/10.1177/1082013208098814
Fleuri, L. F., Delgado, C. H. O., Novelli, P. K., Pivetta, M. R., do Prado, D. Z., & Simon, J. W.
(2016). Enzymes in food and beverage processing (pp. 255–280). CRC Press, Taylor and
Francis Group.
Freitas, A. A., Francelin, M. F., Hirata, G. F., Clemente, E., & Schmidt, F. L. (2008). Atividades das
enzimas peroxidase (POD) e polifenoloxidase (PPO) nas uvas das cultivares benitaka e rubi
e em seus sucos e geleias. Ciência e Tecnologia de Alimentos, 28, 172–177.
Funk, C., Weber, P., Thilker, J., Grabber, J. H., Steinhart, H., & Bunzel, M. (2006). Influence of
lignification and feruloyation of maize cell walls on the adsorption of heterocyclic aromatic
amines. Journal of Agricultural and Food Chemistry, 54, 1860–1867.
Gao, Y., Zietsman, A. J. J., Vivier, M. A., & Moore, J. P. (2019). Deconstructing wine grape cell
walls with enzymes during winemaking: New insights from glycan microarray technology.
Molecules, 24(1), 165. https://doi.org/10.3390/molecules24010165
Garg, G., Singh, A., Kaur, A., Singh, R., Kaur, J., & Mahajan, R. (2016). Microbial pectinases: An
ecofriendly tool of nature for industries. Biotech, 6(1), 47. https://doi.org/10.1007/s13205-016-
0371-4
Goff, S. A., & Klee, H. J. (2006). Plant volatile compounds: Sensory cues for health and nutritional
value? Science, 311(5762), 815–819.
Gómez-Plaza, E., Romero-Cascales, I., & Bautista-Ortίn, A. B. (2010). Use of enzymes in wine
production. In A. Bayindirli (Ed.), Enzymatic processing in fruits and vegetables: Chemistry
and engineering applications (pp. 215–243). CRC Press, Taylor and Francis Group.
Gonçalves, E. M., Pinheiro, J., Abreu, M., Brandão, T. R. S., & Silva, C. L. M. (2007). Modelling
the kinetics of peroxidase inactivation, colour and texture changes of pumpkin (Cucurbita
maxima L.) during blanching. Journal of Food Engineering, 81, 693–701.
Gonzalez, M. E., & Barrett, D. M. (2010). Thermal, high pressure, and electric field processing
effects on plant cell membrane integrity and relevance to fruit and vegetable quality. Journal of
Food Science, 75, R121–R130.
Granada, G. L., Vendruscolo, J. L., & Treptow, R. O. (2001). Caracterização química e sensorial de
sucos clarificados de amora-preta (Rubus spp. L.). Revista Brasileira de Agrociencia, 7,
143–147.
Grassin, C., & Coutel, Y. (2009). Enzymes in fruit and vegetable processing and juice extraction. In
R. J. Whitehurst & M. van Oort (Eds.), Enzymes in food technology (pp. 236–263). Wiley-
Blackwell. https://doi.org/10.1002/9781444309935.ch11
Grassino, A. N., Brnčić, M., Vikić-Topić, D., Roca, S., Dent, M., & Brnčić, S. R. (2016).
Ultrasound assisted extraction and characterization of pectin from tomato waste. Food Chemis-
try, 198, 93–100.
Guerrero-Beltrán, J. A., Barbosa-Cánovas, G. V., & Swanson, B. G. (2005). High hydrostatic
pressure processing of fruit and vegetable products. Food Reviews International, 21(4),
411–425. https://doi.org/10.1080/87559120500224827
Guillemin, A., Guillon, F., Degraeve, P., Rondeau, C., Devaux, M.-F., Huber, F., Badel, E., Saurel,
R., & Lahaye, M. (2008). Firming of fruit tissues by vacuum-infusion of pectin methylesterase:
Visualisation of enzyme action. Food Chemistry, 109(2), 368–378. https://doi.org/10.1016/j.
foodchem.2007.12.050
100 M. D. Heirangkhongjam et al.
Guo, X., Zhao, W., Pang, X., Liao, X., Hu, X., & Wu, J. (2014). Emulsion stabilizing properties of
pectins extracted by high hydrostatic pressure, high speed shearing homogenization and tradi-
tional thermal methods: A comparative study. Food Hydrocolloids, 35, 217–225.
Hsu, C. P., Deshpande, S. N., & Desrosier, N. W. (1965). Role of pectin methylesterase in firmness
of canned tomatoes. Journal of Food Science, 30(4), 583–588. https://doi.org/10.1111/j.
1365-2621.1965.tb01806.x
Huang, W., Bi, X., Zhang, X., Liao, X., Hu, X., & Wu, J. (2013). Comparative study of enzymes,
phenolics, carotenoids and colour of apricot nectars treated by high hydrostatic pressure and
high temperature short time. Innovative Food Science and Emerging Technologies, 18, 74–82.
İyidoǧan, N. F., & Bayındırlı, A. (2004). Effect of l-cysteine, kojic acid and 4-hexylresorcinol
combination on inhibition of enzymatic browning in Amasya apple juice. Journal of Food
Engineering, 62(3), 299–304. https://doi.org/10.1016/S0260-8774(03)00243-7
Jaiswal, A. K., & Sharma, S. (2016). Enzymes in synthesis of novel functional food ingredients. In
M. Chandrasekaran (Ed.), Enzymes in food and beverage processing (pp. 381–400). CRC Press,
Taylor and Francis Group.
Javeri, H., Toledo, R., & Wicker, L. (1991). Vacuum infusion of citrus pectinmethylesterase and
calcium effects on firmness of peaches. Journal of Food Science, 56(3), 739–742. https://doi.
org/10.1111/j.1365-2621.1991.tb05371.x
Jecu, L. (2000). Solid state fermentation of agricultural wastes for endoglucanase production.
Industrial Crops and Products, 11(1), 1–5. https://doi.org/10.1016/S0926-6690(99)00022-9
Jensen, M., Petersen, B. R., & Adler-Nissen, J. (2004). Enzymatic firming of processed red pepper
by means of exogenous pectinesterase. Food Biotechnology, 18(2), 217–227. https://doi.org/10.
1081/FBT-200025667
Jobling, S. (2004). Improved starch for food and industrial applications. Current Opinion of Plant
Biology, 7, 210–218.
Juturu, V., & Wu, J. C. (2013). Insight into microbial hemicellulases other than xylanases: A
review: Microbial hemicellulases other than xylanases. Journal of Chemical Technology and
Biotechnology, 88(3), 353–363. https://doi.org/10.1002/jctb.3969
Kant, S., Vohra, A., & Gupta, R. (2013). Purification and physicochemical properties of
polygalacturonase from Aspergillus niger MTCC 3323. Protein Expression and Purification,
87, 11–16.
Kavuthodi, B., & Sebastian, D. (2018). Review on bacterial production of alkaline pectinase with
special emphasis on Bacillus species. Bioscience Biotechnology Research Communications, 11,
18–30.
Kc, S., Upadhyaya, J., Joshi, D. R., Lekhak, B., Chaudhary, D. K., Pant, B. R., & Raghavan,
V. (2020). Production, characterisation and industrial applications pectinases enzyme isolated
from fungal strains. Fermentation, 6(2), 59.
Keenan, D. F., Rößle, C., Gormley, R., Butler, F., & Brunton, N. P. (2012). Effect of high
hydrostatic pressure and thermal processing on the nutritional quality and enzyme activity of
fruit smoothies. LWT—Food Science and Technology, 45(1), 50–57. https://doi.org/10.1016/j.
lwt.2011.07.006
Kelebek, H., Canbas, A., Cabaroglu, T., & Selli, S. (2007). Improvement of anthocyanin content in
the cv. Öküzgözü wines by using pectolytic enzymes. Food Chemistry, 105(1), 334–339.
https://doi.org/10.1016/j.foodchem.2006.11.068
Khairnar, Y., Krishna, V. K., Boraste, A., Gupta, N., Trivedi, S., Patil, P., Gupta, G., Gupta, M.,
Jhadhav, A., Mujapara, A., et al. (2009). Study of pectinase production in submerged fermenta-
tion using different strains of Aspergillus niger. International Journal of Microbiology
Research, 1, 13–17.
Kim, J. Y., Seo, Y. S., Kim, J. E., Sung, S.-K., Song, K. J., An, G., & Kim, W. T. (2001). Two
polyphenol oxidases are differentially expressed during vegetative and reproductive develop-
ment and in response to wounding in the Fuji apple. Plant Science, 161(6), 1145–1152. https://
doi.org/10.1016/S0168-9452(01)00522-2
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 101
Kim, Y. J., & Uyama, H. (2005). Tyrosinase inhibitors from natural and synthetic sources:
Structure, inhibition mechanism and perspective for the future. Cellular and Molecular Life
Sciences, 62, 1707–1723.
Knorr, D., Zenker, M., Heinz, V., & Lee, D.-U. (2004). Applications and potential of ultrasonics in
food processing. Trends in Food Science and Technology, 15(5), 261–266. https://doi.org/10.
1016/j.tifs.2003.12.001
Kola, O., Kaya, C., Duran, H., & Altan, A. (2010). Removal of limonin bitterness by treatment of
ion exchange and adsorbent resins. Food Science and Biotechnology, 19(2), 411–416.
Kollarigowda, R. H. (2015). Novel polysaccharide nanowires; synthesis from pectin-modified
methacrylate. RSC Advances, 5, 102143–102146.
Kubra, K. T., Ali, S., Walait, M., & Sundus, H. (2018). Potential applications of pectinases in food,
agricultural and environmental sectors. Journal of Pharmaceutical, Chemical and Biological
Sciences, 6, 23–34.
Labuza, T. P., Lillemo, J. H., & Taoukis, P. S. (1992). Inhibition of polyphenol oxidase by
proteolytic enzymes. Fruit Processing, 2(1), 9–13.
Leadlay, P. F. (1993). An introduction to enzyme chemistry (p. 82). The Royal Society of
Chemistry.
Lee, J. Y., Kim, S. S., & Kang, D. H. (2015). Effect of pH for inactivation of Escherichia coli O157:
H7, Salmonella Typhimurium and Listeria monocytogenes in orange juice by ohmic heating.
LWT—Food Science and Technology, 62, 83–88.
Li, J.-Y., & Yeh, A.-I. (2001). Relationships between thermal, rheological characteristics and
swelling power for various starches. Journal of Food Engineering, 50, 141–148.
Liu, F., Osman, A., Yusof, S., & Ghazali, H. M. (2005). Effects of enzyme-aided peeling on the
quality of local mandarin (Citrus reticulata b.) segments: Enzyme-aided peeling of mandarins.
Journal of Food Processing and Preservation, 28(5), 336–347. https://doi.org/10.1111/j.
1745-4549.2004.21145.xC
Loizzo, M. R., Tundis, R., & Menichini, F. (2012). Natural and synthetic tyrosinase inhibitors as
antibrowning agents: An update. Comprehensive Reviews in Food Science and Food Safety,
11(4), 378–398. https://doi.org/10.1111/j.1541-4337.2012.00191.x
Lopes, A. S., & Clemente, E. (2002). Minerais e enzimas oxidativas em brócolis (Brassica oleracea
L. cv. Itálica) minimamente processado. Acta Scientiarum, 24(6), 1615–1618.
Lu, F.-J., Chu, L. H., & Gau, R.-J. (1998). Free radical—Scavenging properties of lignin. Nutrition
and Cancer, 30, 31–38.
Markets & Markets. (2020). Retrieved June 20, 2020 from https://www.marketsandmarkets.com/
Market-Reports/fruit-vegetable-processing-enzymes-market
Marshall, M. R., Kim, J., & Wei, C. (2000). Enzymatic browning in fruits, vegetables and
seafoods. FAO.
Marszałek, K., Krzyżanowska, J., Woźniak, L., & Skąpska, S. (2017). Kinetic modelling of
polyphenol oxidase, peroxidase, pectin esterase, polygalacturonase, degradation of the main
pigments and polyphenols in beetroot juice during high pressure carbon dioxide treatment.
LWT—Food Science and Technology, 85, 412–417.
Marszałek, K., Mitek, M., & Skąpska, S. (2015). The effect of thermal pasteurization and high
pressure processing at cold and mild temperatures on the chemical composition, microbial and
enzyme activity in strawberry purée. Innovative Food Science and Emerging Technologies, 27,
48–56. https://doi.org/10.1016/j.ifset.2014.10.009
Merino, M. T., Humanes, L., Lopez-Ruiz, A., Diez, J., & Roldan, J. M. (1997). High-performance
liquid chromatography quantitation of limonin D-ring lactone hydrolase and limonoate dehy-
drogenase activities. Journal of Chromatography A, 760(2), 173–178.
Miller, F. A., & Silva, C. L. M. (2012). Thermal treatment effects in fruit juices. In S. Rodrigues &
F. Fan (Eds.), Advances in fruit processing technologies (e-book ed., pp. 363–383). CRC
Press. ISBN: 978-1-4398-5153-1.
102 M. D. Heirangkhongjam et al.
Min, S., Evrendilek, G. A., & Zhang, H. Q. (2007). Pulsed electric fields: Processing system,
microbial and enzyme inhibition, and shelf life extension of foods. IEEE Transactions on
Plasma Science, 35(1), 59–73. https://doi.org/10.1109/TPS.2006.889290
Nandakumar, R., & Wakayama, M. (2015). Enzymes in flavours and food additives. In
M. Chandrasekaran (Ed.), Enzymes in food and beverage processing (pp. 321–340). CRC Press.
Noci, F., Riener, J., Walkling-Ribeiro, M., Cronin, D. A., Morgan, D. J., & Lyng, J. G. (2008).
Ultraviolet irradiation and pulsed electric fields (PEF) in a hurdle strategy for the preservation of
fresh apple juice. Journal of Food Engineering, 85(1), 141–146. https://doi.org/10.1016/j.
jfoodeng.2007.07.011
O’Donnell, C. P., Tiwari, B. K., Bourke, P., & Cullen, P. J. (2010). Effect of ultrasonic processing
on food enzymes of industrial importance. Trends in Food Science and Technology, 21(7),
358–367. https://doi.org/10.1016/j.tifs.2010.04.007
Oey, I. (2010). Effect of novel food processing on fruit and vegetable enzymes. In A. Bayindirli
(Ed.), Enzymatic processing in fruits and vegetables: Chemistry and engineering applications
(pp. 245–312). CRC Press, Taylor and Francis Group.
Oliveira, C. M., Ferreira, A. C. S., De Freitas, V., & Silva, A. M. S. (2011). Oxidation mechanisms
occurring in wines. Food Research International, 44(5), 1115–1126. https://doi.org/10.1016/j.
foodres.2011.03.050
Omaña-Molina, M., González-Robles, A., Iliana Salazar-Villatoro, L., Lorenzo-Morales, J.,
Cristóbal-Ramos, A. R., Hernández-Ramírez, V. I., et al. (2013). Reevaluating the role of
Acanthamoeba proteases in tissue invasion: Observation of cytopathogenic mechanisms on
MDCK cell monolayers and hamster corneal cells. BioMed Research International, 2013,
461329.
Oumer, O. J. (2017). Pectinase: Substrate, production and their biotechnological applications.
International Journal of Environment, Agriculture and Biotechnology, 2, 1007–1014.
Oumer, O. J., & Abate, D. (2018). Screening and molecular identification of pectinase producing
microbes from coffee pulp. BioMed Research International. https://doi.org/10.1155/2018/
2961767
Pagán, A., Ibarz, A., & Pagán, J. (2005). Kinetics of the digestion products and effect of temperature
on the enzymatic peeling process of oranges. Journal of Food Engineering, 71(4), 361–365.
https://doi.org/10.1016/j.jfoodeng.2004.10.039
Palma-Fernandez, E. R. D., Gomes, E., & Silva, R. D. (2002). Purification and characterization of
two β-Glucosidases from the thermophilic Fungus Thermoascus aurantiacus. Folia
Microbiologica, 47(6), 685–690.
Pan, X., Tu, T., Wang, L., Luo, H., Ma, R., Shi, P., Meng, K., & Yao, B. (2014). A novel low-
temperature-active pectin methylesterase from Penicillium chrysogenum F46 with high effi-
ciency in fruit firming. Food Chemistry, 162, 229–234. https://doi.org/10.1016/j.foodchem.
2014.04.069
Perera, N., Gamage, T. V., Wakeling, L., Gamlath, G. G. S., & Versteeg, C. (2010). Colour and
texture of apples high pressure processed in pineapple juice. Innovative Food Science and
Emerging Technologies, 11, 39–46.
Placido, D., Fernandes, C. G., Isidro, A., Carrondo, M. A., Henriques, A. O., & Archer, M. (2008).
Auto-induction and purification of Bacillus subtilis transglutaminase (Tgl) and its preliminary
crystallographic characterization. Protein Expression and Purification, 59, 1–8.
Polizeli, M. L. T. M., Rizzatti, A. C. S., Monti, R., Terenzi, H. F., Jorge, J. A., & Amorim, D. S.
(2005). Xylanases from fungi: Properties and industrial applications. Applied Microbiology and
Biotechnology, 67(5), 577–591. https://doi.org/10.1007/s00253-005-1904-7
Poojary, M. M., Roohinejad, S., Koubaa, M., Barba, F. J., Passamonti, P., Režek Jambrak, A., Oey,
I., & Greiner, R. (2017). Impact of pulsed electric fields on enzymes. In D. Miklavčič (Ed.),
Handbook of electroporation (pp. 2369–2389). Springer International Publishing. https://doi.
org/10.1007/978-3-319-32886-7_173
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 103
Prakash, S., Singhal, R., & Kulkarni, P. (2001). Enzymic peeling of Indian grapefruit (Citrus
paradisi). Journal of the Science of Food and Agriculture, 81(15), 1440–1442. https://doi.org/
10.1002/jsfa.969
Pretel, M. T., Botella, M. Á., Amorós, A., Serrano, M., Egea, I., & Romojaro, F. (2007a). Obtaining
fruit segments from a traditional orange variety (Citrus sinensis (L.) Osbeck cv. Sangrina) by
enzymatic peeling. European Food Research and Technology, 225(5–6), 783–788. https://doi.
org/10.1007/s00217-006-0482-y
Pretel, M. T., Botella, M. A., Amorós, A., Zapata, P. J., & Serrano, M. (2007b). Optimization of
vacuum infusion and incubation time for enzymatic peeling of ‘Thomson’ and ‘Mollar’ oranges.
LWT—Food Science and Technology, 40(1), 12–20. https://doi.org/10.1016/j.lwt.2005.07.021
Pretel, M. T., Sánchez-Bel, P., Egea, I., & Romojaro, F. (2008). Enzymatic peeling of citrus fruits:
Factors affecting degradation of the albedo. In T. da Silva & J. A. Islework (Eds.), Tree and
forestry science and biotechnology (Vol. 2, pp. 52–59). Global Science Books.
Pretel, M. T., Sánchez-Bel, P., Egea, I., & Romojaro, F. (2010). Enzymatic peeling of citrus fruits.
In A. Bayindirli (Ed.), Enzymatic processing of fruits and vegetables: Chemistry and engineer-
ing applications (pp. 145–174). CRC Press, Taylor and Francis Group.
Qudsieh, H. Y. M., Yusof, S., Osman, A., & Rahman, R. A. (2002). Effect of maturity on
chlorophyll, tannin, color and polyphenol oxidase (PPO) activity of sugarcane juice (Saccharum
officinarum var. Yellow cane). Journal of Agricultural and Food Chemistry, 50, 1615–1618.
Rawson, A., Patras, A., Tiwari, B. K., Noci, F., Koutchma, T., & Brunton, N. (2011). Effect of
thermal and non thermal processing technologies on the bioactive content of exotic fruits and
their products: Review of recent advances. Food Research International, 44(7), 1875–1887.
https://doi.org/10.1016/j.foodres.2011.02.053
Revilla, I., & Gonzalez-San Jose, M. L. (2003). Addition of pectolytic enzymes: An enological
practice which improves the chromaticity and stability of red wines. International Journal of
Food Science and Technology, 38(1), 29–36. https://doi.org/10.1046/j.1365-2621.2003.
00628.x
Ribeiro, D. S., Henrique, S. M. B., Oliveira, L. S., Macedo, G. A., & Fleuri, L. F. (2010). Enzymes
in juice processing: A review. International Journal of Food Science and Technology, 45(4),
635–641. https://doi.org/10.1111/j.1365-2621.2010.02177.x
Rojas, M. L., Miano, A. C., & Augusto, P. E. D. (2017). Ultrasound processing of fruit and
vegetable juices. In Ultrasound: Advances for food processing and preservation
(pp. 181–199). Elsevier. https://doi.org/10.1016/B978-0-12-804581-7.00007-5
Rolff, M., Schottenheim, J., Decker, H., & Tuczek, F. (2011). Copper–O2 reactivity of tyrosinase
models towards external monophenolic substrates: Molecular mechanism and comparison with
the enzyme. Chemical Society Reviews, 40(7), 4077. https://doi.org/10.1039/c0cs00202j
Romero-Cascales, I., Fernández-Fernández, J. I., Ros-García, J. M., López-Roca, J. M., & Gómez-
Plaza, E. (2008). Characterisation of the main enzymatic activities present in six commercial
macerating enzymes and their effects on extracting colour during winemaking of Monastrell
grapes. International Journal of Food Science and Technology, 43(7), 1295–1305. https://doi.
org/10.1111/j.1365-2621.2007.01608.x
Rouhana, A., & Mannheim, C. H. (1994). Optimization of enzymatic peeling of grapefruit.
Lebensmittel Wissenschaft und Technology, 27, 103–107.
Saeeduddin, M., Abid, M., Jabbar, S., Wu, T., Hashim, M. M., Awad, F. N., Hu, B., Lei, S., &
Zeng, X. (2015). Quality assessment of pear juice under ultrasound and commercial pasteuriza-
tion processing conditions. LWT—Food Science and Technology, 64(1), 452–458. https://doi.
org/10.1016/j.lwt.2015.05.005
Schwab, W., Davidovich-Rikanati, R., & Lewinsohn, E. (2008). Biosynthesis of plant-derived
flavor compounds. The Plant Journal, 54, 712–732.
Segovia, F. J., Luengo, E., Corral-Pérez, J. J., Raso, J., & Almajano, M. P. (2015). Improvements in
the aqueous extraction of polyphenols from borage (Borago officinalis L.) leaves by pulsed
electric fields: Pulsed electric fields (PEF) applications. Industrial Crops and Products, 65,
390–396. https://doi.org/10.1016/j.indcrop.2014.11.010
104 M. D. Heirangkhongjam et al.
Sessa, D. J., & Anderson, R. L. (1981). Soybean peroxidases: Purification and some properties.
Journal of Agricultural and Food Chemistry, 29, 960.
Sharma, H. P., Patel, H., & Sugandha. (2017). Enzymatic added extraction and clarification of fruit
juices—A review. Critical Reviews in Food Science and Nutrition, 57(6), 1215–1227. https://
doi.org/10.1080/10408398.2014.977434
Sheu, S. C., & Chen, A. O. (1991). Lipoxygenase as blanching index for frozen vegetable soybeans.
Journal of Food Science, 56(2), 448–451.
Sila, D. N., Duvetter, T., De Roeck, A., Verlent, I., Smout, C., Moates, G. K., Hills, B. P., Waldron,
K. K., Hendrickx, M., & Van Loey, A. (2008). Texture changes of processed fruits and
vegetables: Potential use of high-pressure processing. Trends in Food Science and Technology,
19(6), 309–319. https://doi.org/10.1016/j.tifs.2007.12.007
Singh, N., Singh, J., Kaur, L., Sodhi, N. S., & Gill, B. S. (2003). Morphological, thermal and
rheological properties of starches from different botanical sources. Food Chemistry, 81(2),
291–231.
Singhania, R. R., Saini, J. K., Saini, R., Adsul, M., Mathur, A., Gupta, R., & Tuli, D. K. (2010).
Bioethanol production from wheat straw via enzymatic route employing Penicillium
janthinellum cellulases. Bioresource Technology, 169, 490–495.
Siwach, R., & Kumar, M. (2012). Comparative study of thermosonication and thermal treatments
on pectin methyl esterase inactivation in mosambi juice. Journal of Dairying, Foods and Home
Sciences, 31, 290–296.
Somavat, R., Mohamed, H. M. H., & Sastry, S. K. (2013). Inactivation kinetics of Bacillus
coagulans spores under ohmic and conventional heating. LWT—Food Science and Technology,
54, 194–198.
Sundarram, A., & Murthy, T. P. K. (2014). α-Amylase production and applications: A review.
Journal of Applied and Environmental Microbiology, 2(4), 166–175.
Suutarinen, M., Mustranta, A., Autio, K., Salmenkallio-Marttila, M., Ahvenainen, R., & Buchert,
J. (2003). The potential of enzymatic peeling of vegetables. Journal of the Science of Food and
Agriculture, 83(15), 1556–1564. https://doi.org/10.1002/jsfa.1579
Swami Hulle, N. R., & Rao, P. S. (2016). Effect of high pressure and thermal processing on quality
changes of Aloe vera-litchi mixed beverage (ALMB) during storage. Journal of Food Science
and Technology, 53, 359–369.
Szymańska-Chargot, M., Chylińska, M., Gdula, K., Koziol, A., & Zdunel, A. (2017). Isolation and
characterisation of cellulose from different fruit and vegetable pomaces. Polymers, 9(10), 495.
Terefe, N. S., Buckow, R., & Versteeg, C. (2014). Quality related enzymes in plant based products:
Effects of novel food processing technologies, Part 1: High-pressure processing. Critical
Reviews in Food Science and Nutrition, 54, 24–63.
Terefe, N. S., Tepper, P., Ullman, A., Knoerzer, K., & Juliano, P. (2016). High pressure thermal
processing of pears: Effect on endogenous enzyme activity and related quality attributes.
Innovative Food Science and Emerging Technologies, 33, 56–66. https://doi.org/10.1016/j.
ifset.2015.12.001
Terrasan, C. R. F., Temer, B., Duarte, M. C. T., & Carmona, E. C. (2010). Production of xylanolytic
enzymes by Penicillium janczewskii. Bioresource Technology, 101, 4139–4143.
Thakur, B. R., Singh, R. K., Handa, A. K., & Rao, M. A. (1997). Chemistry and uses of pectin—A
review. Critical Reviews in Food Science and Nutrition, 37(1), 47–73.
Tiwari, B. K., O’Donnell, C. P., & Cullen, P. J. (2009). Effect of non-thermal processing
technologies on the anthocyanin content of fruit juices. Trends in Food Science and Technology,
20(3–4), 137–145. https://doi.org/10.1016/j.tifs.2009.01.058
Tochi, B. N., Wang, Z., Xu, S. Y., & Zhang, W. (2009). Effect of stem bromelain on the browning
of apple juice. American Journal of Food Technology, 4(4), 146–153.
Toker, İ., & Bayιndιrlι, A. (2003). Enzymatic peeling of apricots, nectarines and peaches. LWT—
Food Science and Technology, 36(2), 215–221. https://doi.org/10.1016/S0023-6438(02)
00203-7
3 Role of Enzymes in Fruit and Vegetable Processing Industries: Effect. . . 105
Tourino, M. C. C., Chitarra, A. B., & Gavilanes, M. C. (1993). Injúria mecânica em tecidos de
frutos de pessegueiros (Prununs persica [L.] Batsch): Mecanismos de cura. Boletim da
Sociedade Brasileira de Ciência e Tecnologia de Alimentos, 27(2), 69–78.
Toushik, S. H., Lee, K.-T., Lee, J.-S., & Kim, K.-S. (2017). Functional applications of
lignocellulolytic enzymes in the fruit and vegetable processing industries: Applications of
lignocellulolytic enzymes. Journal of Food Science, 82(3), 585–593. https://doi.org/10.1111/
1750-3841.13636
Uneojo, M., & Pastore, G. M. (2007). Pectinases: Aplicações industriais e perspectivas. Química
Nova, 30(20), 1–14.
Valderrama, P., Marangoni, F., & Clemente, E. (2001). Efeito do tratamento térmico sobre a
atividade de peroxidase (POD) e polifenoloxidase (PPO) em maçã (Mallus comunis). Ciência
e Tecnologia de Alimentos, 21, 321–325.
van der Maarel, M. J. E., van der Veen, B., Uitdehaag, J. C. M., Leemhuis, H., & Dijkhuizen,
L. (2002). Properties and applications of starch-converting enzymes of the alpha-amylase
family. Journal of Biotechnology, 94, 137–155.
Villena, M. A., Iranzo, J. F. Ú., & Pérez, A. I. B. (2007). β-Glucosidase activity in wine yeasts:
Application in enology. Enzyme and Microbial Technology, 40(3), 420–425. https://doi.org/10.
1016/j.enzmictec.2006.07.013
Vincken, J. P., Beldman, G., & Voragen, A. G. J. (1994). The effect of xyloglucans on the
degradation of cell wall embdedded cellulose by the combined action of cellobiohydrolase
and endoglucanases from Trichoderma viride. Plant Physiology, 104(1), 99–107.
Wang, K., Jiang, X., Xu, F., Sun, R. C., & Baird, M. S. (2010). Influence of steam pressure on the
physiochemical properties of degraded hemicelluloses obtained from steam-exploded Lespe-
deza stalks. BioResources, 5, 1717–1732.
Welti-Chanes, J., Ochoa-Velasco, C. E., & Guerrero-Beltrán, J. Á. (2009). High-pressure homoge-
nization of orange juice to inactivate pectinmethylesterase. Innovative Food Science and
Emerging Technologies, 10(4), 457–462. https://doi.org/10.1016/j.ifset.2009.05.012
Yang, Y., Zhu, N., Yang, J., Lin, Y., Liu, J., Wang, R., Wang, F., & Yuan, H. (2017). A novel
bifunctional acetyl xylan esterase/arabinofuranosidase from Penicillium chrysogenum P33
enhances enzymatic hydrolysis of lignocellulose. Microbial Cell Factories, 16(1), 166.
https://doi.org/10.1186/s12934-017-0777-7
Yoruk, R., & Marshall, M. R. (2003). Physiochemical properties and function of plant polyphenol
oxidase: A review. Journal of Food Biochemistry, 27(5), 361–422. https://doi.org/10.1111/j.
1745-4514.2003.tb00289.x
Zabetakis, I., Leclerc, D., & Kajda, P. (2000). The effects of high hydrostatic pressure on the
strawberry anthocyanins. Journal of Agriculture and Food Chemistry, 48, 2749–2754.
Zanatta, C. L., Zotarelli, M. F., & Clemente, E. (2006). Peroxidase (POD) e Polifenoloxidase (PPO)
em polpa de goiaba (Psidium guajava R.). Ciência e Tecnologia de Alimentos, 26(3), 705–708.
Zhang, L., Zhang, L., Yi, H., et al. (2012). Enzymatic characterization of transglutaminase from
Streptomyces mobaraensis DSM 40587 in high salt and effect of enzymatic cross-linking yak
milk proteins on functional properties of stirred yogurt. Journal of Dairy Science, 95,
3559–3568.
Zhu, F.-M., Du, B., & Li, J. (2014). Aroma enhancement and enzymolysis regulation of grape wine
using β-glycosidase. Food Science and Nutrition, 2(2), 139–145. https://doi.org/10.1002/
fsn3.84
Production of a, b, and g-Cyclodextrin
Gluconotransferase (CGTase) and Their 4
Applications in Food Industry
Abstract
Keywords
α-Cyclodextrin · β-Cyclodextrin · γ-Cyclodextrin · Food industry · Applications
4.1 Introduction
Cyclodextrins (CDs) are a family of cyclic oligosaccharides that show the ability to
form inclusion complex (IC) through variety of molecules (like Allyl isothiocyanate
(AITC)) (Del Valle, 2004). These molecules are produced through the action of
# The Author(s), under exclusive license to Springer Nature Singapore Pte 107
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_4
108 R. Gautam and S. K. Arya
Fig. 4.1 Basic structure of alpha, beta, and gamma cyclodextrins (Radi & Eissa, 2010; Das et al.,
2013; Astray et al., 2009)
the most marketed utilized CD is β-CD and to lesser extent α-CD. The production of
gamma CD is less due to high cost and low production associated with it. The
CGTases producing cyclodextrin of single type are of interest as it is difficult to
isolate the CD of interest from a complex mixture of CDs. Cyclodextrins are
hydrophobic molecules and their solubility increases with ethanol concentration.
The spectroscopic studies imply that the confirmation of cyclodextrins in solution is
similar to that in crystalline state; however, β-CD has a perfect regularity, whereas
alpha and gamma are slightly distorted (Astray et al., 2009).
Cyclodextrins are useful because of following reasons: They are made from
renewable material (starch). They are relatively cheap. Due to their biodegradability,
they do not produce any harmful effect to the environment. They are nontoxic for
human body (Duca & Boldescu, 2008). The stability of the cyclodextrins follows
α > β > γ. The cyclodextrins are as stable as sucrose in solid state and are resistant to
degradation and can be stored for several years (Kurkovsergey &
Loftssonthorsteinn, 2013). These are homogenous, crystalline, and
non-hygroscopic substances (Stella & Rajewski, 1997). The alpha cyclodextrin
has six glucopyranose rings, beta has seven, and gamma has eight glucopyranose
units (Astray et al., 2009). The rate of hydrolysis accelerates in the order of
α-CD < β-CD < γ-CD. Gamma cyclodextrins have been proved to retain the
maximum retention properties than the three cyclodextrins. Cyclodextrins are also
used to retain colours of food substances (Duca & Boldescu, 2008; Lopez-Nicolas
et al., 2007a, b). The hydrophobic interior of the cavity possesses some lewis base
characteristics due to the presence of high electron density.
4.2 History
First time cyclodextrin was discovered by Villiers in 1891 from starch using a strain
of Bacillus amylobacter (Das et al., 2013; Szejtli, 1990). The following table
represents the discoveries of cyclodextrins according to time passage and work
done on cyclodextrins in different fields (Del Valle, 2004; Dardeer, 2014).
Year of
working Work done on cyclodextrin References
1891 First time CDs produced from starch by Singh et al. (2002)
digesting it with Bacillus amylobacter
strain by scientist Villiers. Villiers also
produces cyclodextrin from impure culture
by using strain Bacillus macerans.
1903 Schardinger produces two crystalline Eastburn and Tao (1994)
products and named it as dextrin A and
dextrin B, but the strain from which they
are produced are not maintained.
1904 Schardinger discovered a new organism Eastburn and Tao (1994)
that has capability of producing ethyl
(continued)
110 R. Gautam and S. K. Arya
Year of
working Work done on cyclodextrin References
alcohol and acetone from sugar and starch-
containing plant materials.
1911 Schardinger produced large amount of Eastburn and Tao (1994)
cyclodextrin from strain Bacillus macerans
about 25–30% and named it α-CD and
β-CD.
1935 Schardinger discovered γ-CD. After that, Loftsson and Brewester (1996),
Freudenberg and Cramer explained the Matsuda and Arima (1999), Mabuchi
existence of large cyclodextrin molecules. and Ngoa (2001)
1942 Schardinger demonstrated the structures of Buschmann and Schollmeyer (2002)
β-CD and α-CD using X-ray
crystallography.
1948 γ-CD structure is determined by using Buschmann and Schollmeyer (2002)
X-ray crystallography. At that time, it is
observed that cyclodextrins can form
inclusion complexes.
1961 Some evidences show the existence of Qi and Romberger (1988), Hirose and
natural cyclodextrins like ξ, δ, ζ, and even Yamamoto (2001)
η-cyclodextrin.
1965 A method is developed in this year for Kainuma (1984)
isolation of larger homologues structures
of ζ-cyclodextrin and η-cyclodextrin,
method named fractionation method.
1970 In 1970, several numbers of Kainuma (1984)
microorganisms are considered as a source
for cyclodextrin glucanotransferases and
glucanotransferase was purified and named
during the exploration period. Applications
of cyclodextrins in different industries
started in this period.
1980 Development in biotechnology in the mid Crini (2014)
of 1980s, improved the purification and
production of cyclodextrins and led to
increases in their value for
commercial uses.
1982 In 1982, first book on cyclodextrins was Crini (2014)
published, written by Szejtli.
1992 In this period, Hedges R.A. published a Hedges (1992)
book on cyclodextrin under a title
Cyclodextrins and Their Applications in
Biotechnology.
2003 In this period, Tomasik P. published a Szejtli (2004a)
book on cyclodextrins, under a title
Cyclodextrins Chemical and Functional
Properties of Food Saccharides.
2004 In this year, review articles were published Szejtli (2004b)
on data related to cyclodextrin and its
(continued)
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 111
Year of
working Work done on cyclodextrin References
future aspects and also on application of
cyclodextrins.
2006 Helena Dodziuk published a book on Kainuma (1984)
Cyclodextrins and Their complexes:
Chemistry, Analytical Methods and
Applications.
2007 Investigation of cyclodextrin Kunamneni et al. (2007)
glucanotransferase production using
alginate-immobilized cells of alkalophilic
Bacillus sp. in an airlift reactor.
2010 Biochemical characterization of Zhaofeng et al. (2010)
α-cyclodextrin glycosyltransferase and its
extracellular expression from
Paenibacillus macerans have been
published by Zhaofeng Li et al.
2011 Encapsulation of ethylene gas into Ho et al. (2011)
α-cyclodextrin and characterization of the
inclusion complexes.
2012 Effects of cyclodextrins on the Liang et al. (2012)
antimicrobial activity of plant-derived
essential oil compounds.
2013 Optimization of reaction of γ-cyclodextrin Wang et al. (2013)
glycosyltransferase and isoamylase for
enhanced production of c-cyclodextrin.
2014 Cyclodextrins in capillary electrophoresis: Escuder-Gilabert et al. (2014)
Recent developments and new trends.
2015 Experimental setup and kinetic study of Maneechakr et al. (2015)
ultrasonic-assisted transesterification of
waste cooking oil over sulfonated carbon
catalyst derived from cyclodextrin.
2016 Use of cyclodextrins to recover catechin Lopez-Miranda et al. (2016)
and epicatechin from red grape pomace.
CD have hydrophobic cavity and the ability to form reversible host–guest complexes
or termed as supramolecular complex via weak forces like, dipole–dipole
interactions, Van der Waals interactions, and hydrogen bonding with molecules
(Escuder-Gilabert et al., 2014). CDs have the ability to solubilize hydrophobic
materials and thus can entrap volatile components by forming inclusion complexes
with organic compounds, which results in enhancing their physical and chemical
properties. They have the property of selective recognition of a chiral compound and
enantio recognition of enantiomers of chiral compounds (Del Valle, 2004;
Steinbrunn & Wenz, 1996). Low toxicity and immunogenicity of CDs allow for
112 R. Gautam and S. K. Arya
their use as a transport system into the living systems. Also, CDs have the capacity to
form pseudorotaxanes and rotaxanes through number of linear species. Rotaxanes
are the compounds that consist linear and cyclic species and are linked together by
non-covalent interaction (Isnin & Kaifer, 1993; Buston et al., 2001). As inclusion
bodies, CDs help in increasing the stability, solubility, and bioavailability of
lipophilic organic molecules, improve the stability, increase the permeability of
water-insoluble compounds, and diminish the drug toxicity by manufacturing the
compound effective at lesser doses (Del Valle, 2004; Kamimura et al., 2014).
Fig. 4.2 Cyclodextrins structure and inclusion complex formation (Schmid, 1989; Gomes et al.,
2014; Garnero et al., 2010)
solution is then stirred for complete 5 min. Saturated alpha cyclodextrin obtained is
added to plastic container and ethylene gas was then pressurized at 0.2, 0.5, 1, and
1.5 Pa at 25 C. Afterwards, crystal precipitates formed are collected by vacuum
filtration and then analysed for ethylene gas concentrations and further characterized
for their physiochemical properties (Ho et al., 2011).
CDs have hydrophobic cavity. CDs have the ability to form host–guest
complexes via weak forces, like Van der waal interactions, dipole–dipole
interactions, and hydrogen bonding with molecules (Szejtli, 1988). CDs can solubi-
lize hydrophobic materials and thus entrap volatile components by forming inclusion
complex with organic compounds, resulting in enhancing their chemical and physi-
cal properties. CGTase production at large scale by using Bacillus sp. depends upon
the kinetic model of cyclodextrin-glucanotransferase (Burhan et al., 2005).
Advantages of CD to form IC are: higher thermal stability, high solubility, bioavail-
ability of hydrophobic guests, control volatility, masking off unpleasant odours, and
controlling release of drugs and flavours.
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 115
This table represents the various sources of CGTase from different spices, their mode
of production, and their optimum parameters like temperature and pH.
Optimum
Sources of CGTase Production temperature and pH References
Bacillus macerans Bacillus macerans Optimum Fujiwara et al.
IFO 3490 IFO 3490 mainly temperature is 55 C (1974), Kitahata
produces α-CGTase. and pH 5.2–5.7. et al. (1974),
Fermentation medium Rimphanitchayakit
is soluble starch, corn et al. (2005)
steep liquor.
Bacillus circulance E Bacillus circulance E Optimum Charoenlap et al.
192 192 produces mainly temperature 60 C (2004), Vassileva
β-CGTase. Agar- and pH 5.5–5.8. et al. (2005)
immobilized cells are
used for high
productivity of
CGTase. B. circulance
requires alkaline pH
for an efficient
CGTase formation.
Paenibacillus Main product is β-CD. Optimum Larsen et al. (1998)
temperature is 50 C
and pH is 6.0–8.0.
Klebsiella pneumonia The CGTase from It can be preferred at Gawande and
Klebsiella pneumonia. pH 6–8, temperatures Patkar (2001)
Main product is α-CD. of 40–50 C.
Bacillus CGTase from Bacillus Its optimum activity Rahman et al.
stearothermophillus stearothermophillus at 80 C temperature. (2004)
ET1 is a potential
antistaling enzyme
with cyclodextrin
(CD)-producing
activity. Main product
is α- and β-CD.
Bacillus lehensis Bacillus lehensis is Optimum Blanco et al. (2014)
isolated from cassava temperature is 55 C
starch wastewater. and pH 8.0.
Main product is
β-CD. Its CGTase
activity is 18.9 μm/L
Micrococcus Micrococcus variant Its optimum activity Kim et al. (1997)
M 849 produces α and temperature is
β-CD as main 55–65 C and pH is
products. 5.0–9.0.
Thermoanaerobacter Thermoanaerobacter Its optimum activity Starnes et al. (1992)
sp. sp. ATCC 53627 temperature is
mainly used for the 90–95 C.
(continued)
116 R. Gautam and S. K. Arya
Optimum
Sources of CGTase Production temperature and pH References
production of
β-CGTase.
Alkalophilic Bacillus Alkalophilic Bacillus Optimum Sian et al. (2005)
sp. sp. 38-2 mainly temperature is 45 C.
produces β-CD.
Bacillus CGTase is synthesized Growth medium is Andel-Naby et al.
amyloliquefaciens by maintained at pH 6.5 (2000)
B. amyloliquefaciens and microorganism is
by immobilized maintained at 4 C on
method in batch and potato dextrose agar
continuous cultures. slants.
In this table, methods for CDs complexes or inclusion complexes formation have
been explained and the manufacturing of CDs.
Methods for
CD-complex
formations Process References
Precipitation Precipitation method is most widely used for Das et al. (2013),
method inclusion complex production. According to Sapkal et al. (2007)
Sapkal et al., by using co-precipitation method, a
poor aqueous solubility guest molecule is
combined with β-cyclodextrin to form inclusion
complex. In this method, drug and CD are
dissolved in water and solvent (like acetone) is
heated at 75 C. Stirring is done for 1 h at 75 C to
obtain concentrated, viscous, and translucent liquid
and then cooled at room temperature. Precipitates
are filtered or separated than dried and stored to get
solid inclusion complex.
Kneading method Conventional kneaders like low and high shear Das et al. (2013),
mixers are used to prepare CD paste, paste is Miller et al. (2007)
prepared with small amount of water in which
guest molecule is added (without a solvent) in a
small amount of ethanol, and then after grinding,
solvent is evaporated and powdered form
cyclodextrin-inclusion formed.
Spray drying In this method, first cyclodextrin is dissolved in Das et al. (2013),
solution that is already alkalinized with 25% Arias et al. (2000)
aqueous ammonia, then guest molecule is
dissolved in 96% ethanol alcohol solution in
100 mL, both solutions are then mixed, and finally
sonicated. The l solution obtained is then spray-
dried to get inclusion complexes; this is done at
(continued)
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 117
Methods for
CD-complex
formations Process References
equilibrium state which is attained by removing the
solvent by spray drying.
Neutralization The guest molecule is dissolved in basic or acidic Das et al. (2013),
method aqueous cyclodextrin solution. Cyclodextrin and Choi et al. (2001)
drug are dissolved separately in 0.1 N NaOH
solution, mixed and stirred for half an hour; pH is
maintained by 0.1 N HCl at 7.5; at this pH,
complexes precipitate and then residues are
separated and washed until free from chlorine and
then dried at 250 C for 24 h
(a) Batch process: Generally CDs are produced through batch process which is
quite simple and easy to control process. But there are several limitations of this
method, like long operation hours, high labour cost, and high enzyme concen-
tration requirement that leads to switching to other production method (Sakinah
et al., 2014; Lia et al., 2014).
(b) Enzymatic membrane reactor (EMR): EMR is basically a stirred tank reactor
combined with a separation membrane that recirculates the reaction mixture
through a membrane module placed outside the enzymatic reactor. As compared
to batch process, this is a continuous process method and is more practical and
economical. Also, reusability of the enzyme results in high yield productions of
cyclodextrins as compared to batch process (Lia et al., 2014). In this process,
product (CD) is separated from the enzyme and substrate through a semi-
permeable membrane, which is composed of an ultra-thin separation layer
widely used in enzyme separation. During the separation process, CDs penetrate
through the membrane and the enzymes get retained by the membrane within the
reaction reactor (Bodalo et al., 2001).
The working principle is such that the pressure difference across the membrane,
also called the transmembrane pressure (TMP), impels the product through the
membrane, while the unreacted substrate and free enzyme are recirculated to the
enzymatic reactor (Sakinah et al., 2009). Advantages of enzymatic membrane
reactor over batch reactor are its high production flow rates; reductions in cost,
energy, and waste products by the recycling practice; easy reactor operation and
control; and the high yields of pure material (Katzir, 1993). But the main
limitation of this method is fouling of membrane as the substrate gets deposited
onto the membrane surface and enzyme precipited within the membrane pores
(Hughes & Field, 2006; Balakrishnan & Dua, 2000; Yuan & Zydney, 1999;
Bansal et al., 2006). However, this problem can be overcome by employing a
high cross-flow velocity (CFV), but this condition requires large amount of
118 R. Gautam and S. K. Arya
energy and sometimes reduces the enzyme activity (Sakinah et al., 2014;
Gugliemi et al., 2007; Priyananda & Chen, 2006).
4.4.1 Production
(Rakmai et al., 2015; Iyer et al., 2003). The result is a mixture of α-, β-, and
γ-cyclodextrins, containing trace amounts of cyclodextrins (Sakinah et al., 2014).
α-CD has the lowest hydrogen bond strengths between the 3-OH and 2-OH groups
around the wider rim. Diameter of internal cavity is 4.7–5.3 Å and the cavity volume
is about 66% or 41% of that of β- or γ-cyclodextrins, respectively (Li et al., 2007). It
typically forms inclusion complexes with benzene derivatives because of Van der
Waals interactions. α-Cyclodextrin has modest solubility in water which means it is
almost eight times greater than that of β-cyclodextrin, but 1.6 times lower than that of
γ-cyclodextrin at 25 C. Also, it is more resistant to hydrolysis in acid solutions. It
was observed that even after 3 h of incubation period in extremely acidic conditions
(pH ¼ 2.4) at 100 C, no breakdown of α-cyclodextrin is recognized (Jodai et al.,
1984; Saha & Zeikus, 1992). But α-cyclodextrin can’t be hydrolysed by human
salivary and pancreatic amylases for its commercial production (Kondo et al., 1990).
α-CGTases from Bacillus macerans or Paenibacillus macerans are most commonly
used. It is also effective at solubilizing free fatty acids (Qi & Zimmermann, 2005).
There are two types of processes that result in the production of cyclodextrins:
4.4.4 Applications
Cyclodextrins are used as browning inhibitors in different fruit juices. The oxidation
of volatile precursors present in freshly squeezed pear juices can be prevented by
120 R. Gautam and S. K. Arya
y
Filtration and drying of crystals
Fig. 4.3 Flow chart is showing the production of α-CD (Lia et al., 2014; Rendleman, 1997)
adding α-cyclodextrin (90 mM). It is one of the most suitable agents for
encapsulating flavours that are encapsulated in powder form by spray drying with
α-cyclodextrin (Shiga et al., 2004). α-Cyclodextrin is also a dietary fibre as it forms
complex with triglyceride at a ratio 1:1 that is typical for dietary fibres and form a
stable complex with dietary fat (Ferrari et al., 2013; Threapleton et al., 2013; Zhang
et al., 2013a, b). It reduces the absorption and bioavailability of dietary fat, which
makes it practical as a weight loss supplement (Suzuki & Sato, 1985).
α-Cyclodextrin is effective in reducing and/or maintaining body weight with
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 121
increasing energy intake for obese patients with type 2 diabetes (Gallaher et al.,
2007; Grunberger et al., 2007). It is effective in improving metabolic syndrome by
reducing serum triglyceride and leptin levels and increasing insulin sensitivity and
faecal fat excretion in rats. α-Cyclodextrin may also be useful as an ingredient for
reducing the glycaemic impact of foods (Penninga et al., 1995).
Alpha CGTase from Paenibacillus macerans is most commonly used for the
production of alpha cyclodextrin. However, the production of enzyme can be
increased by expressing it in E. coli which can bring a benefit in reducing cost of
alpha cyclodextrin. Alpha CGTases produced in E. coli are accumulated in the
cytosol in the form of inactive inclusion bodies. In this case, alpha CGTase from
P. macerans JFB05-01 is expressed extracellularly in E. coli. The recombinant
enzyme is then purified and characterized with respect to cyclization activity,
including optimum conditions effect of metal ions, etc.
The plasmid pET20b is used for the production of alpha cyclodextrin. At 25 C, very
little recombinant alpha CGTase was secreted into the medium. But after 90 h of
incubation period, the production rate increased rapidly and the cyclization activity
of alpha CGTase also increased which was 42 times higher than the parent strain. A
single colony of E. coli BL21cells harbouring plasmid CGT/pET20B was inoculated
into LB medium containing ampicillin. For inducing the expression of α-CGTase,
different amounts of isopropyl 1-thio-b-D-galactopyranoside (IPTG) were added.
The recombinant alpha CGTase contains a histidine sequence on a C-terminal
domain which can be separated by metal binding affinity. To overcome the hetero-
geneity, the recombinant enzyme is purified by chromatographic separation
techniques which include Q-sepharose and phenyl-superose chromatography. The
purified protein obtained is then separated by SDS page. From the SDS PAGE, it
was found that alpha CGTase migrated as a single band with molecular mass of
approximately 72 kDa. The optimum temperature of alpha CGTase recombinant
enzyme was 45 which was below that of the native enzyme which is 50 C. The
recombinant and native CGTase both showed the best cyclization activity at
pH 5.5 at a pH value; above or below this, the cyclization activity decreases. Enzyme
is stable in Na2HPO4/NaH2PO4 at the same pH. The cyclization activity of alpha
CGTase is inhibited completely by Hg2+ and slightly by Fe2+ or Co2+. The addition
of 2 mM Ca2+, 2 mM Ba2+, or 0.1 mM Zn2+ activated the cyclization activity
considerably. It is also observed that the growth medium has a great impact on the
secretion of proteins in E. coli. However, TB medium was considered the best for
extracellular production of alpha CGTase as TB medium has beneficial effect on cell
growth because of its relative high buffering capacity. Also, the low IPTG
122 R. Gautam and S. K. Arya
concentration (0.01 mM) favoured the extracellular enzyme production. The extra-
cellular productivity of the recombinant enzyme was 40 fold higher than the parent
enzyme P. macerans JFB05-01. The metal chelator like EDTA doesn’t show any
effect on the activity of α-CGTase enzyme even at relatively high concentration
(10 mM), which indicates that a metal cofactor was not required for the function of
the enzyme. Furthermore, α-CGTase could be activated in the presence of Ca2+,
which was probably because of the binding of Ca2+ with its binding sites which are
located at the active site of the protein molecule, resulting in stabilizing the substrate
binding cleft of CGTase (Eastburn & Tao, 1994). Following table represents number
of the sources of alpha cyclodextrin production, optimum temperature, optimum pH,
and main products (Stella & Rajewski, 1997; Buschmann & Schollmeyer, 2002;
Hirose & Yamamoto, 2001).
Optimum
Source of α-CD Optimum temperature
production pH ( C) Main product References
CGTase 6–8 40–50 In this method, alpha- Flaschel
produced from cyclodextrin is the main et al. (1984)
Klebsiella product, produced by
pneumoniae M5 enzymatic degradation of
starch, starch used as a
substrate. The yield of alpha-
cyclodextrin is lower by the
enzymatic method.
CGTase 5.0–8.0 55–65 Main product is α-CDs, Larsen et al.
produced from studied by purification and (1998), Yagi
Micrococcus characterization of et al. (1980)
variant M 849 cyclodextrin
glycosyltransferase from
Micrococcus variant M 849.
CGTase 5.0–7.5 – By using this sp., CGTase Larsen et al.
produced from produces α-CDs as a main (1998),
Klebsiella product. Purification and Bender
oxytoca M5A1 characterization of CGTase (1983)
are done in this article.
CGTase 5.2–5.7 55 In this paper, we study the Larsen et al.
produced from production of α-CDs by using (1998),
Bacillus sp. Bacillus macerans. Fujiwara
macerans IFO et al. (1974)
3490
4.5 Beta-Cyclodextrin
Beta-cyclodextrin (β-CD) has the ability to encapsulate molecules that are hydro-
phobic in nature which helps in improving their aqueous solubility and also reduces
their volatility (Lin et al., 2016).
Production of β-CD can be achieved in both batch and continuous process. But
continuous process is preferred over batch because of its advantages like greater
process control, high productivity, and improvement of quality and yield. Continu-
ous process includes the combination of both CSTR (continuous stirred tank reactor)
and PFR (plug flow reactor) and is considered to be more suitable for long-term use.
Also in case of industrial scale production, CSTR is preferred over batch process
because of its properties like high stability and low by-product formation, but the
disadvantage of using CSTR is the undesired thermal gradients in case of PBR, poor
radial mixing, and low mass transfer rate (Santos et al., 2015; Rakmai & Cheirsilp,
2016; Galan et al., 2011; Kittikun et al., 2008).
Gram-positive microorganisms Bacillus firmus strain and Bacillus sphaericus
strain 41 were immobilized on a loofa sponge and are used as the direct source for
the CGTase and CD production (Moriwak et al., 2014). The CGTase produced by
Bacillus sp. C26 has been reported to have high activity for β-cyclodextrin produc-
tion; immobilized CGTase is then applied for the production of β-cyclodextrin
(Rakmai et al., 2015) (Fig. 4.4).
CGTase was produced by bacillus firmus isolated from brazillian soil of oat culture.
The medium contained KH2PO4, MgSO4, and Na2CO3 and the pH was maintained
at 10.3. Then the medium is transferred to a 5 L medium and proceeded for
cultivation for 5 days at 37 C. The specific enzyme is isolated using affinity column
chromatography and the protein concentration is estimated using Bradford method
using bovine serum as standard, and using SDS-PAGE, molecular mass of protein is
estimated. The CGTase activity is measured by replacing maltodextrin by corn-
starch and potato starch. The maximum activity of enzyme is seen at a pH greater
than 5 and less than 10 and at a temperature of 50 C (Natalia et al., 2007).
Production of β-CD (10 whole till last): Immobilization of CGTases is more
commonly used as it reduces the production costs and makes it feasible to be used in
various food industries (Garcia-Galan et al., 2011). The enzyme immobilization has
a lot of advantages as the enzyme can be reused, makes downstream processing easy,
and increases thermo-stability of enzyme (Rodrigues et al., 2013; Natividade
Schöffer et al., 2013). Chitosan derivative of chitin has required properties for
enzyme immobilization (Muzzarelli, 1980). Chitosan has reactive amino and
hydroxyl groups in its glucosamine chain (Chiou & Wu, 2004). Chitosan also
exhibits mechanical stability, rigidity, and biocompatibility which permit its use in
food industries (Krajewska, 2004). The following table represents the sources of
β-cyclodextrins from various papers and its optimum pH, temperature, and fermen-
tation methods.
124 R. Gautam and S. K. Arya
Fig. 4.4 Flow chart showing production of β-cyclodextrin using entrapped CGTase (Rakmai et al.,
2015; Muria et al., 2011)
Optimum
Source of Optimum temperature Main
γ-CGTase pH ( C) product References
Bacillus subtilis 8.0 65 γ-CD Li et al. (2007), Kato and
strain 313 Horikoshi (1986)
Bacillus 7.5–10.5 55 γ-CD Li et al. (2007), Fugita et al.
sp. strain AL-6 (1990)
Bacillus firmus 6–8 60 γ-CD Li et al. (2007), Veen et al.
strain 290-3 (2000)
Bacillus cereus 8.0 37 γ-CD Li et al. (2007), Lin et al.
(2016), Ng et al. (2011)
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 127
4.6.1 Applications
1. Cyclodextrins are used to preserve the natural flavours, vitamins, and natural
colours of foods
Manufacturing, packaging, and other factors modify the flavours by reducing
aroma. There are wide techniques and processes used for retaining the properties
and flavour of the food like spray drying, freeze-drying, fluidized bed coating,
and crystallization. However, those involving the formation of flavour/CD
molecular-inclusion complexes offer great potential for the protection of volatile
substances throughout many rigorous food-processing methods such as freezing,
thawing, and microwaving. The addition of β-CD has proved to be most effective
in retaining the flavour against heat, evaporation, and also against thermal
degradation (Jouquand et al., 2004). The flavour enhancing complexes consists
of many components, thus it is important that there is proper incorporation of
these components in the complex without causing any change in its composition
(Astray et al., 2009). Natural and synthetic coffee flavours have also been retained
by the use of beta cyclodextrins. When these complex bound flavours come in
contact with water molecules, they get released immediately and thus responsible
for the taste or flavour (Astray et al., 2009; Szente & Szejtli, 1986). In addition to
coffee, β-CD is also used for tea aromatization (jasmine, peppermint, and cinna-
mon) and also results in stability as well as efficient storage.
2. For the removal of undesired taste, flavour, microbial contamination, and other
undesired products
For the rejection of various food items, bitter taste is responsible. It has been
found that there are two classes of chemical compounds like flavonoids and
limonoids that are responsible for bitterness. Fresh citrus juice turns bitter in
case of storage which is dependent on pH and temperature. It is highly desirable
to remove the bitter components without the addition of any other substance to
the fruit juice. Therefore, cyclodextrins are used for masking the undesired
components from food. When the cyclodextrins are added to the fruit juice
which has a peculiar odour and an undesirable taste, it forms complexes and
retains the flavour of the components. For example, this process is used for
deodourizing soybean milk and soy protein which removes the peculiar fish
odours. A mixture of cyclodextrins alpha, beta, and gamma is ordinarily used.
Rice which gives an unpleasant odour when stored for a long time can be made
odour-free by cooking the rice in the presence of 0.01–0.4% CD (Sakakibara
et al., 1985). The taste of cooked ice can be improved by addition of maltosyl beta
CD. When CDs are complexed with sweetening agents such as aspartame; it
helps in stabilizing and improving the taste of product. This complexation process
also helps in eliminating the bitter aftertaste of other sweeteners like stevioside,
glycyrrhizin, and rubusoside. CD itself is a promising new sweetener (Singh
128 R. Gautam and S. K. Arya
and also improve the barrier properties (diffusion rate and transmission rate), thus
maintaining the food quality (Suzuki & Sato, 1985). Cyclodextrins can release
antimicrobials and antioxidant compounds using CDs as carriers. As the humidity
levels in freshly cut vegetables or fruits increase, they can lead to spoilage; this
can be reduced by using cyclodextrins. At high relative humidity, high water–CD
interaction weakens the host guest interaction and consequently there is a release
of antimicrobial molecule which should protect the molecule against microbial
growth (Astray et al., 2009; Ayala-Zavala et al., 2008).
5. Cyclodextrins as stabilizers
Cyclodextrins are used for the preparation of stable water in oil emulsion such as
mayonnaise and salad dressing due to their hydrophobic cavity and hydrophilic
outer surface. In tomato ketchup, natural food colouring components can be
stabilized by the addition of beta cyclodextrins. This ketchup when prepared by
heating at 100 for 2 h did no discolouration, while the control did. Also, the
addition of cyclodextrins to emulsified cheese increased the water retention and
shelf life. However, the control discoloured when heated to this temperature
(Szente & Szejtli, 2004). Cyclodextrins also help in processed meat products in
improving water retention and texture (Ota & Takeda, 1981).
6. The browning or caking formation in the solid compositions containing sugars
and amino acids can be prevented by addition of oligosaccharides such as
cyclodextrin
By the addition of 20% of β-CD to powdered juice containing anhydrous glucose,
aspartic acid, d-alanine, citric acid, etc., the shelf life and also stability of the
product are enhanced. Even after 30–35 days at 40 C, no change in colour was
observed (Fujimoto, 1981). The control started to discolour even on the second
day and turned brown on fourth day (Szente & Szejtli, 2004). Starch gelatiniza-
tion is the process in which heat treatment is given to starch and water, resulting in
swelling of starch granules. However, addition of β-CD considerably improves
the gelatinization of wheat flour. Addition of 1.5% beta cyclodextrin improves the
solubility and swelling properties of starch granules. It has been observed that the
adding up of 1.5% of beta cyclodextrin augmented the viscosity of starch paste to
4%. Milk casein hydrolysate is a readily digestible protein and its bitter protein
can be removed by addition of 10% of beta cyclodextrin (Specht et al., 1981).
Similarly, the bitter taste of ginseng extract and propylene glycol can also be
eliminated by the use of beta cyclodextrin (Akiyama & Miyao, 1979).
7. Extraction efficiency is increased with the use of beta cyclodextrin
When the roasted coffee beans are extracted using beta cyclodextrin, the extrac-
tion becomes more effective and the complex containing beta cyclodextrin and
coffee can be used for effectual preservation of volatile components and also
provide better aroma of the product (Wagner et al., 1988). Low cholesterol butter
is also made by the addition of beta cyclodextrin. Molten butter is blended with
crystalline beta cyclodextrin, which forms stable complexes with triglycerides/
cholesterol and the cholesterol/β-CD is easily removed from molten butter.
130 R. Gautam and S. K. Arya
Ninety percent of cholesterol in fat milk can also be separated by the addition of
beta cyclodextrin; the butter obtained after this process does not retain any of the
cyclodextrin (Szente & Szejtli, 2004). Cyclodextrins are widely used in order to
modify the natural properties of materials, for example simply eggs have been
produced in USA which means a low cholesterol egg which is produced by the
addition of cyclodextrins to reduce cholesterol.
8. β-Cyclodextrin is more extensively used in food applications due to its availabil-
ity at low cost, chemically stable structure, and non-hygroscopic, non-toxic, and
edible nature. Ethyl vinyl alcohol copolymers are widely used in food industry
due to their properties like gas barrier to oxygen and organic compounds and high
transparency (Sanchez-Chaves et al., 2007; Lopez-de-Dicastilloa et al., 2010).
On comparing all the three forms of cyclodextrins, γ-cyclodextrins show the
highest flavour retention capacity among the other three.
4.6.2 Properties
References
Arya S. K., & Srivastava S. K. (2006, July 3). Enzyme and microbial technology, 39 (3), 507–510.
Akiyama, Y., & Miyao, K. (1979). Microencapsulation of royal jelly. Japanese Kokai JP 77 80 463.
Andel-Naby, M. A., Reyad, R. M., & Abdel-Fattah, A. F. (2000). Biosynthesis of cyclodextrin
glucosyltransferase by immobilized Bacillus amyloliquefaciens in batch and continuous
cultures. Biochemical Engineering Journal, 5, 1–9.
Arias, M. J., Moyano, J. R., Munoz, P., Gines, J. M., Justo, A., & Giordano, F. (2000). Study of
omeprazole-gamma-cyclodextrin complexation in the solid state. Drug Development and
Industrial Pharmacy, 26, 253–259.
Artiss, J. D., Brogan, K., Brucal, M., Moghaddam, M., & Jen, K. L. (2006). The effects of a new
soluble dietary fiber on weight gain and selected blood parameters in rats. Metabolism, 55,
195–202.
Astray, G., Barreiro, C. G., Mejuto, J. C., Otero, R. R., & Gándara, J. S. (2009). A review on the use
of cyclodextrins in foods. Food Hydrocolloids, 23(7), 1631–1640.
Ayala-Zavala, J. F., Del-Toro-Sánchez, L., Lvarez-Parrilla, A. E., & Gonzalez-Aguilar, G. A.
(2008). High relative humidity in-package of fresh-cut fruits and vegetables: Advantage or
disadvantage considering microbiological problems and antimicrobial delivering systems. Jour-
nal of Food Science, 73(4), R41–R47.
Balakrishnan, M., Dua, M., & Bhagat, J. J. (2000). Effect of operating parameters on sugarcane
juice ultrafiltration: Results of a field experience. Separation and Purification Technology, 19,
209–220.
Bansal, B., Ali, R. A., Prieto, R. M., & Chen, X. D. (2006). Rinsing and cleaning of alactalbumin
fouled MF membranes. Separation and Purification Technology, 48, 202–207.
Bender, H. (1983). An improved method for the preparation of cyclooctaamylose, using starches
and the cyclodextrin glycosyltransferase of Klebsiella pneumoniae M5 al. Carbohydrate
Research, 124, 225–233.
Blackwood, A. D., & Bucke, C. (2000). Addition of polar organic solvents can improve the product
selectivity of cyclodextrin glycosyltransferase solvent effects on CGTase. Enzyme and Micro-
bial Technology, 27, 704–708.
Blanco, K. C., Moraes, F. F. D., Bernardi, N. S., Vettori, M. H. P. B., Monti, R., & Contiero,
J. (2014). Cyclodextrin production by Bacillus lehensis isolated from cassava starch:
Characterisation of a novel enzyme. Czech Journal of Food Sciences, 32(2014), 48–53.
Bodalo, A., Gomez, J. L., Gomez, E., Bastida, J., Maximo, M. F., & Montiel, M. C. (2001).
Ultrafiltration membrane reactors for enzymatic resolution of amino acids: Design model and
optimization. Enzyme and Microbial Technology, 28, 355–361.
132 R. Gautam and S. K. Arya
Burhan, N., Sapundzhiev, T. S., & Beschkov, V. (2005). Mathematical modeling of cyclodextrin-
glucanotransferase production by batch cultivation. Biochemical Engineering Journal, 24,
73–77.
Buschmann, H. J., & Schollmeyer, E. (2002). Applications of cyclodextrins in cosmetic products: A
review. Journal of Cosmetic Science, 53, 575–592.
Buston, J. E. H., Marken, F., & Anderson, H. L. (2001). Enhanced chemical reversibility of redox
processes in cyanine dye rotaxanes. Chemical Communications, 11, 1046–1047.
Charoenlap, N., Dharmsthiti, S., Sirisansaneeyakul, S., & Lertsiri, S. (2004). Optimization of
cyclodextrin production from sago starch. Bioresource Technology, 92, 49–54.
Cheirsilp, B., Kitcha, S., & Maneerat, S. (2010). Kinetic characteristics of β-cyclodextrin produc-
tion by cyclodextrin glycosyltransferase from newly isolated Bacillus sp. C26. Electronic
Journal of Biotechnology, 13, 717–3458.
Chiou, S. H., & Wu, W. T. (2004). Immobilization of Candida rugosa lipase on chitosan with
activation of the hydroxyl groups. Biomaterials, 25(2), 197–204.
Choi, H. G., Lee, B. J., Han, J. H., Lee, M. K., Park, K. M., Yong, C. S., Rhee, J. D., Kim, Y. B., &
Kim, C. K. (2001). Terfenadine-β-cyclodextrin inclusion complex with antihistaminic activity
enhancement. Drug Development and Industrial Pharmacy, 27(8), 857–862.
Crini, G. (2014). Review: A history of cyclodextrins. Chemical Reviews, 114, 10940–10975.
Dardeer, H. M. (2014). Importance of cyclodextrins into inclusion complexes. International
Journal of Advances Research, 2(4), 414–428.
Das, S. K., Rajabalaya, R., David, S., Gani, N., Khanam, J., & Nanda, A. (2013). Cyclodextrins:
The molecular container. Research Journal of Pharmaceutical, Biological and Chemical
Sciences, 4(2), 1694.
Del Valle, E. M. M. (2004). Cyclodextrin and their uses: A review. Process Biochemistry, 39,
1033–1046.
Duca, G., & Boldescu, V. (2008). Cyclodextrin: Fields of application. Chemistry Journal of
Moldova: General, Industrial and Ecological Chemistry, 3, 30–37.
Eastburn, S. D., & Tao, B. Y. (1994). Applications of modified cyclodextrins. Journal of Biotech-
nology Advances, 12, 325–339.
Escuder-Gilabert, L., Martín-Biosca, Y., Medina-Hernández, M. J., & Sagrado, S. (2014).
Cyclodextrins in capillary electrophoresis: Recent developments and new trends. Journal of
Chromatography A, 1357, 2–23.
Ferrari, P., Rinaldi, S., Jenab, M., Lukanova, A., Olsen, A., Tjonneland, A., et al. (2013). Dietary
fiber intake and risk of hormonal receptor-defined breast cancer in the European Prospective
Investigation into Cancer and Nutrition study. American Journal of Clinical Nutrition, 97,
344–353.
Ferrarotti, S., Rosso, A., Marechal, M., Krymkiewicz, N., & Marechal, L. (1996). Isolation of two
strains (S–R type) of Bacillus circulans and purification of a cyclomaltodextrin-
glucanotransferase. Cellular and Molecular Biology, 42(5), 653–657.
Flaschel, E., Landert, J. P., Spiesser, D., & Renken, A. (1984). The production of alpha-
cyclodextrin by enzymatic degradation of starch. Annals of the New York Academy of Sciences,
434, 70–77.
Fugita, Y., Tsubouchi, H., Inagi, Y., Tomita, K., Ozaki, A., & Nakanishi, K. (1990). Purification
and properties of cyclodextrin glycosyltransferase from Bacillus sp. AL-6. Journal of Fermen-
tation and Bioengineering, 70, 150–154.
Fujimoto, S. (1981). Prevention of quality deterioration in rice. Japanese Kokai JP 8196670.
Fujiwara, S., Kakihara, H., Woo, K. B., Lejeune, A., Kanemoto, M., Sakaguchi, K., & Imanaka,
T. (1974). Cyclization characteristics of cyclodextrin glucanotransferase are conferred by the
NH2-terminal region of the enzyme. Applied and Environmental Microbiology, 58, 4016–4025.
Galan, C. G., Murcia, A. B., Lafuente, R. F., & Rodrigues, R. C. (2011). Potential of different
enzyme immobilization strategies to improve enzyme performance. Advanced Synthesis and
Catalysis, 353(16), 2885–2904.
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 133
Gallaher, D. D., Gallaher, C. M., & Plank, D. W. (2007). Alphacyclodextrin selectively increases
fecal excretion of saturated fats. FASEB Journal, 21, A730.
Garcia-Galan, C., Berenguer-Murcia, Á., Fernandez-Lafuente, R., & Rodrigues, R. C. (2011).
Potential of different enzyme immobilization strategies to improve enzyme performance.
Advanced Synthesis & Catalysis, 353(16), 2885–2904.
Garnero, C., Zoppi, A., Genovese, D., & Longhi, M. (2010). Studies on tri-methoprim:
Hydroxypropyl-β-cyclodextrin: Aggregate and complex formation. Carbohydrate Research,
345, 2550–2556.
Gawande, B., & Patkar, A. (2001). Alpha-cyclodextrin production using cyclodextrin
glycosyltransferase from Klebsiella pneumoniae AS-22. Starch-Starke, 53(2), 75–83.
Gawande, B. N., Singh, R. K., Chauhan, A. K., Goel, A., & Patkar, A. Y. (1998). Optimization of
cyclomaltodextrin glucanotransferase production from Bacillus firmus. Enzyme and Microbial
Technology, 22, 288–291.
Goel, A., & Nene, S. (1995). Modifications in the phenolphthalein method for spectro-photometric
estimation of beta cyclodextrin. Starch, 47, 399–400.
Gomes, L. M. M., Petito, N., Costa, V. G., & Deborah, Q. F. (2014). Inclusion complexes of red bell
pepper pigments with b-cyclodextrin preparation, characterisation and application as natural
colorant in yogurt. Food Chemistry, 148, 428–436.
Goo, B. G., Hwang, Y. J., & Park, J. K. (2014). Bacillus thuringiensis: A specific gamma-
cyclodextrin producer strain. Carbohydrate Research, 386, 12–17.
Grunberger, G., Jen, K. L., & Artiss, J. D. (2007). The benefits of early intervention in obese
diabetic patients with FBC—A new dietary fibre. Diabetes/Metabolism Research and Reviews,
23, 56–62.
Gugliemi, G., Chiarani, D., Judd, S. J., & Andreottola, G. (2007). Flux criticality and sustainability
in a hollow fibre submerged membrane bioreactor for municipal wastewater treatment. Journal
of Membrane Science, 289, 241–248.
Hara, H., & Hashimoto, H. (2002). Antimicrobial and insect-repellent cyclodextrin films. Japanese
Kokai JP 2002029901.
Hedges, R. A. (1992). Cyclodextrins and their applications in biotechnology. In Minutes of the
Sixth International Symposium on Cyclodextrins (pp. 380–389). Editions de Sante.
Higuti, I. H., da Silva, P. A., Papp, J., de Eiro, M., Okiyama, V., Alves de Andrade, E., Abreu
Marcondes, A., & do Nascimento, A. J. (2004). Colorimetric determination of α &
β-cyclodextrins and studies on optimization of CGTase production from B. firmus using
factorial designs. Brazilian Archives of Biology and Technology, 47(6), 837–841.
Hirose, T., & Yamamoto, Y. (2001). Hinokitol containing cyclodextrin polymer compositions and
their molding with excellent antimicrobial and gas barrier properties. Journal of Japanese
Patent 55480.
Ho, B. T., Joyce, D. C., & Bhandari, B. R. (2011). Encapsulation of ethylene gas into
α-cyclodextrin and characterisation of the inclusion complexes. Food Chemistry, 127, 572–580.
Hughes, D., & Field, R. W. (2006). Cross-flow filtration of washed and unwashed yeast suspensions
at constant shear under nominally sub-critical conditions. Journal of Membrane Science, 280,
89–98.
Isnin, R., & Kaifer, A. E. (1993). A new approach to cyclodextrin-based rotaxanes. Pure and
Applied Chemistry, 65, 495–498.
Iyer, J. L., Shetty, P., & Pai, J. S. (2003). Immobilization of cyclodextrin glucanotransferase from
B. circulans ATCC21783 on purified seasand. Journal of Industrial Microbiology & Biotech-
nology, 30, 47–51.
Jodai, I., Kandra, L., Harangi, J., Nanasi, P., Debrecen, & Szejtli, J. (1984). Hydrolysis of
cyclodextrin by Aspergillus oryzae alphaamylase. Starch-Starke, 36, 140–143.
Jouquand, C., Ducruet, V., & Giampaoli, P. (2004). Partition coefficients of aroma compounds in
polysaccharide solutions by the phase ratio variation method. Food Chemistry, 85, 467–474.
134 R. Gautam and S. K. Arya
Qi, Z., & Romberger, M. L. (1988). Cyclodextrin. In R. H. Walter (Ed.), Polysaccharide associa-
tion structures in food (pp. 207–226). Marcel Dekker.
Radi, A. E., & Eissa, S. (2010). Electrochemistry of cyclodextrin inclusion complexes of pharma-
ceutical compounds. Journal of the Open Chemical and Biomedical Methods, 3, 74–85.
Rahman, R. A., Illias, R. M., Nawawi, M. G. M., Ismail, A. F., Hassan, Q., & Kamaruddin,
K. (2004). Optimisation of growth medium for the production of cyclodextrin
glucanotransferase from Bacillus stearothermophillus HR1 using response surface methodol-
ogy. Process Biochemistry, 39, 2053–2060.
Rakmai, J., & Cheirsilp, B. (2016). Continuous production of β-cyclodextrin by cyclodextrin
glycosyltransferase immobilized in mixed gel beads: Comparative study in continuous stirred
tank reactor and packed bed reactor. Biochemical Engineering Journal, 105, 107–113.
Rakmai, J., Cheirsilp, B., & Prasertsan, P. (2015). Enhanced thermal stability of cyclodextrin
glycosyl transferase in alginate–gelatine mixed gel beads and the application for
β-cyclodextrin production. Biocatalysis and Agricultural Biotechnology, 4, 717–726.
Rendleman, J. A. J. (1997). Enhancement of cyclodextrin production through use of debranching
enzymes. Applied Biochemistry and Biotechnology, 26, 51–61.
Rimphanitchayakit, V., Tonozuka, T., & Sakano, Y. (2005). Construction of chimeric cyclodextrin
glucanotransferases from Bacillus circulans A11 and Paenibacillus macerans IAM1243and
analysis of their product specificity. Carbohydrate Research, 340, 2279–2229.
Rios, G. M., Belleville, M. P., Paolucci, D., & Sanchez, J. (2004). Progress in enzymatic membrane
reactors—A review. Journal of Membrane Science, 242, 189–196.
Rodrigues, R. C., Ortiz, C., Berenguer-Murcia, A., Torres, R., & Fernandez-Lafuente, R. (2013).
Modifying enzyme activity and selectivity by immobilization. Chemical Society Reviews, 42,
6290–6307.
Rosso, A., Ferrarotti, S., Krymkiewicz, N., & Nudel, C. (2002). Optimisation of batch culture
conditions for cyclodextrin glucanotransferase production from Bacillus circulans DF 9R.
Microbial Cell Factories, 1(3), 1–10.
Saha, B. C., & Zeikus, J. G. (1992). Cyclodextrin degrading enzymes. Starch-Starke, 44, 312–315.
Sakakibara, S., Sugisawa, K., Matsui, F., & Sengoku, K. (1985). Cyclodextrins: New versatile food
additive. Indian. Japanese Patent JP 851 248 075.
Sakinah, A. M. M., Ismail, A. F., Hassan, O., Zularisam, A. W., & Illias, R. M. (2009). Influence of
starch pretreatment on yield of cyclodextrins and performance of ultrafiltration membranes.
Desalination, 239, 317–333.
Sakinah, A. M. M., Ismail, A. F., Illias, R. M., Zularisam, A. W., Hassan, O., Matsuura, T., &
T. (2014). Effect of substrate and enzyme concentration on cyclodextrin production in a hollow
fibre membrane reactor system. Separation and Purification Technology, 124, 61–67.
Sanchez-Chaves, M., Fernandez-García, M., & Cerrada, M. L. (2007). Ethylene-vinyl alcohol
copolymers partially modified with benzoate groups: Study of their polymorphic behaviour.
Journal of Polymer Science Part B: Polymer Physics, 45, 1026–1036.
Santos, E. H., Kamimura, J. A., Hill, L. E., & Gomes, C. L. (2015). Characterization of carvacrol
beta-cyclodextrin inclusion complexes as delivery systems for antibacterial and antioxidant
applications. LWT—Food Science and Technology, 60, 583–592.
Sapkal, N. P., Kilor, V. A., Bhusari, K. P., & Daud, A. S. (2007). Evaluation of some methods for
preparing gliclazide-β-cyclodextrin inclusion complexes. Tropical Journal of Pharmaceutical
Research, 6(4), 833–840.
Schmid, G. (1989). Cyclodextrin glucanotransferase production: Yield enhancement by over
expression of cloned genes. Trends in Biotechnology, 7, 244–248.
Shiga, H., Yoshii, H., Ohe, H., Yasuda, M., Furuta, T., Kuwahara, H., et al. (2004). Encapsulation
of shiitake (Lenthinus edodes) flavours by spray drying. Bioscience, Biotechnology and Bio-
chemistry, 68, 66–71.
Sian, H. K., Said, M., Hassan, O., Kamaruddin, K., Ismail, A. F., Rahman, R. A., Mahmood,
N. A. N., & Illias, R. M. (2005). Purification and characterization of cyclodextrin
glucanotransferase from alkalophilic Bacillus sp. G1. Process Biochemistry, 40, 1101–1111.
4 Production of a, b, and g-Cyclodextrin Gluconotransferase (CGTase). . . 137
Singh, M., Sharma, R., & Banerjee, U. C. (2002). Biotechnological applications of cyclodextrins.
Biotechnology Advances, 20, 341–359.
Specht, M. Rothe, M., Szente, L., & Szejtli, J. (1981). Removal of phenylalanine from protein
hydrolysates. Ger. Offen. 147 615.
Starnes, R. L., Amemiya, K., Jorgensen, P. L., & Jorgensen, S. (1992). Cloning and expression of
the Thermoanaerobacter sp. cyclodextrin glycosyl transferase gene in E. coli and B. subtilis. In
A. R. Hedges (Ed.), Minutes of the Sixth International Symposium on Cyclodextrins
(pp. 46–52). Editions de Sante.
Steinbrunn, M. B., & Wenz, G. (1996). Synthesis of water-soluble inclusion compounds from
polyamides and cyclodextrins by solid-state polycondensation. Angewandte Chemie Interna-
tional Edition in English, 35, 2139–2141.
Stella, V. J., & Rajewski, R. A. (1997). Cyclodextrins: Their future in drug formulation and
delivery. Pharmaceutical Research, 14, 556–5567.
Suzuki, M., & Sato, A. (1985). Nutritional significance of cyclodextrins: Indigestibility and
hypolipemic effect of alpha-cyclodextrin. Journal of Nutritional Science and Vitaminology
(Tokyo), 31, 209–223.
Szejtli, J. (1988). Cyclodextrin technology (pp. 1–78). Kluwer Academic Publishers.
Szejtli, J. (1990). The cyclodextrin and their applications in biotechnology. Journal of Carbohy-
drate Polymers, 12, 375–392.
Szejtli, J. (2004a). Chapter 17: Cyclodextrins. In P. Tomasik (Ed.), Chemical and functional
properties of food saccharides (pp. 271–290). CRC Press.
Szejtli, J. (2004b). Past, present and future of cyclodextrin research. Pure and Applied Chemistry,
76, 1825–1845.
Szente, L., & Szejtli, J. (1986). Molecular encapsulation of natural and synthetic coffee flavor with
β-cyclodextrin. Journal of Food Science, 51(4), 1024–1027.
Szente, L., & Szejtli, J. (2004). Cyclodextrins as food ingredients. Trends in Food Science and
Technology, 15, 137–142.
Threapleton, D. E., Greenwood, D. C., Evans, C. E., Cleghorn, C. L., Nykjaer, C., Woodhead, C.,
et al. (2013). Dietary fiber intake and risk of first stroke: A systematic review and meta-analysis.
Stroke, 44, 1360–1368.
Tonkova, A. (1998). Bacterial cyclodextrin glucanotransferase. Enzyme and Microbial Technology,
22, 678–686.
Vassileva, A., Atanasova, N., Ivanova, V., Dhulster, P., & Tonkova, A. (2007). Characterisation of
cyclodextrin glucanotransferase from Bacillus circulans ATCC 21783 in terms of cyclodextrin
production. Annals of Microbiology, 57(4), 609–615.
Vassileva, A., Beschko, V., Ivanova, V., & Tonkova, A. (2005). Continuous cyclodextrin
glucanotransferase production by free and immobilized cells of Bacillus circulans ATCC
21783 in bioreactors. Process Biochemistry, 40, 3290–3295.
Veen, B. A. V., Uitdehaag, J. C. M., Dijkstra, B. W., & Dijkhuizen, L. (2000). The role of arginine
47 in the cyclization and coupling reactions of cyclodextrin glycosyltransferase from Bacillus
circulans strain 251. The FEBS Journal, 267(12), 3432–3441.
Wagner, C. W., Wilson, C. W., & Shaw, P. E. (1988). Reduction of grape fruit bitter components by
cyclodextrin polymers. Journal of Food Science, 53, 516.
Wang, L., Wu, D., Chen, J., & Wu, J. (2013). Enhanced production of γ-cyclodextrin by optimiza-
tion of reaction of γ-cyclodextrin glycosyltransferase as well as synchronous use of isoamylase.
Food Chemistry, 141, 3072–3076.
Yagi, Y., Kouno, K., & Inui, T. (1980). Purification and characterization of cyclodextrin;
glycosyltransferase from Paenibacillus sp. F8. US Patent 4 317 881.
138 R. Gautam and S. K. Arya
Yuan, W., & Zydney, A. L. (1999). Humic acid fouling during microfiltration. Journal of Mem-
brane Science, 157, 1–12.
Zhang, Z., Xu, G., Liu, D., Zhu, W., Fan, X., & Liu, X. (2013a). Dietary fiber consumption and risk
of stroke. European Journal of Epidemiology, 28, 119–130.
Zhang, Z., Xu, G., Ma, M., Yang, J., & Liu, X. (2013b). Dietary fiber intake reduces risk for gastric
cancer: A meta-analysis. Gastroenterology, 145(1), 113–120.
Zhaofeng, L., Bin, L., Zhengbiao, G., Dua, G., Jing, W., & Chen, J. (2010). Extracellular expression
and biochemical characterization of alpha-cyclodextrin glycosyltransferase from Paenibacillus
macerans. Journal of Carbohydrate Research, 345, 886–892.
Enzyme in Milk and Milk Products: Role
and Application 5
Aparna Agarwal, Naman Kaur, Nidhi Jaiswal,
Memthoi Devi Heirangkhongjam, and Kanika Agarwal
Abstract
Enzymes are biocatalysts that catalyse a desired chemical reaction. Enzymes are
specific in their action and yield into products. The enzymes that are utilized in
the dairy industry for processing milk and milk products, like yoghurt, cheese,
and fermented milks, are commonly known as dairy enzymes. These enzymes
mostly aid in coagulation, cheese production, and enhancing shelf life of various
dairy products. The most used dairy enzymes include lactase, amylase, lipases,
transglutaminase, protease, catalase, and rennet. The functions of enzymes vary
with the kind of the product to be processed. Both endogeneous and exogeneous
enzymes are important for dairy application to provide functionality and safety to
the product along with shelf life extension.
Keywords
Enzymes · Dairy products · Cheese · Fermented products
5.1 Introduction
# The Author(s), under exclusive license to Springer Nature Singapore Pte 139
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_5
140 A. Agarwal et al.
and hence, of various reactions; they catalyse only some of the many possibilities.
Generally, the suffix ‘ase’ is added to the substrate name (e.g. glucose-oxidase, an
enzyme which oxidizes glucose) or the type of reaction (e.g. a polymerase for a
polymerization). However, pepsin, rennin, and trypsin are enzymes that were studied
originally, and hence, are the exceptions to this rule. Enzymes can be categorized
based upon the reactions they catalyse. These include, EC 1 oxidoreductases,
enzymes that immobilize oxidation or reduction reactions (e.g. oxidases or
dehydrogenases); EC 2 transferases enzymes (e.g. transphosphorylases and
phosphomutases, example of a phosphate group), transfer a functional group such
as a methyl or phosphate group; EC 3 hydrolases (e.g. hydrolases, including
carbohydrases, deaminases, and proteases; hydrases such as fumarase, enolase,
and carbonic anhydrase) are enzymes which catalyse the process of hydrolysis of
different bonds (involving mostly, addition or removal of water); EC 4 lyase
enzymes split bonds by methods other than hydrolysis and oxidation or form a
C¼C bond; EC 5 isomerases stimulate isomerization modifications within a mole-
cule (e.g. glucose-isomerase); and EC 6 ligases combine two molecules using
covalent bonds (Whitehurst & Van Oort, 2009).
All enzymes reduce the energy required by a reaction to take place. Different
enzymes have different mechanisms to catalyse reactions, which involve creating
conditions in which the transition state is stabilized in order to reduce the activation
energy. This can be accomplished by providing a substitute way which involves
bonding, and consequently, stabilizing the transition-state conformation of the
substrate. For example, temporary reaction with the substrate in order to form an
intermediate enzyme–substrate complex, where an enzyme plays an indispensable
role. Enzymes possess an interesting property which is their specificity. For exam-
ple, certain enzymes display absolute specificity which implies that they immobilize
only one specific reaction or are selective for a certain kind of chemical bond or
functional group. Generally, there exist four types of specificity which are: Absolute
specificity; here, highly specific enzymes stimulate only one reaction; Group speci-
ficity; here, enzymes that are group-specific act only on molecules with certain
functional groups, such as phosphate, amino, or methyl groups; Linkage specificity,
where enzymes act on chemical bonds of specific nature, regardless of the molecular
structure; and, Stereochemical specificity, where enzymes act only on a certain steric
or optical isomer and not on their isomeric counterparts (Whitehurst & Van Oort,
2009). These biocatalysts are highly potent, as even in a very small quantity, they are
capable of augmenting the rate of reactions to several folds. For example, orotidine-
50 -phosphate-decarboxylase is an enzyme that facilitates a reaction to occur in
milliseconds which would generally take millions of years (Callahan & Miller,
2007; Radzicka & Wolfenden, 1995). As a result, enzymes have advanced as a
vital component for the sustenance of life. Evidently, over 5000 types of biochemical
reactions have been found to be catalysed by enzymes (Schomburg et al., 2013).
Enzymes take part in several metabolic functions of living organisms. For
example, they (kinases and phosphatases) are crucial for processes like signal
transduction and cell regulation. They are also responsible for inducing movement,
with adenosine triphosphate (ATP) which hydrolyses myosin in order to produce
5 Enzyme in Milk and Milk Products: Role and Application 141
contraction in the muscles and to also mobilize cargo around the cell as part of the
cytoskeleton (Macrae, 1985). They also serve a vital role in the ‘digestive systems’
of various mammals and animals. For example, amylases cleave starch molecules
and proteases cleave protein molecules (Whitehurst & Van Oort, 2009). Besides,
enzymes have also been utilized at an industrial scale for antibiotic production.
Moreover, there are certain enzymes which are used in household products like
detergents that help in cleaving several stains on clothes. Additionally, as a result of
the ability of enzymes to alter and enhance the nutritional, functional, and organo-
leptic properties of various ingredients and products, they have also found a wide-
spread application in the food industry. They are employed in the food industries to
manufacture bread, alcoholic drinks, and milk-based products like cheese and
yoghurt, and to improve the flavour and texture of dairy-based products (Qureshi
et al., 2015).
The enzymes that are utilized in the dairy industry for processing milk and milk
products like yoghurt and cheese are commonly known as dairy enzymes. These
enzymes mostly aid in coagulation, cheese production, and enhancing shelf life of
various dairy products. The most used dairy enzymes include lactase, amylase,
lipases, transglutaminase, protease, catalase, and rennet. Lactase enzyme is mostly
utilized for hydrolysing lactose molecules to glucose and galactose sugars and
augment the solubility and sweet flavour in various dairy products. Amylases are
biocatalysts which break down starch by immobilizing the process of hydrolysis of
internal α-1,4-glycosidic bonds of polysaccharide sugars like maltose, glucose,
dextrin, and maltotriose, while preserving the α-anomeric configuration in the
products (Takata et al., 1992). Lipase is another commonly used enzyme which is
employed mostly to mature cheese and improve flavours. Another dairy enzyme is
transglutaminase, commonly known as TGases, which plays a pivotal role in the
enhancement of protein functionality in dairy products. Similar to lipases are
different types of protease enzymes which are also utilized in manufacturing of
cheese to increase the rate of cheese aging, as a functional property, and modifying
milk protein to lower the allergic effects in cow milk-based infant food products
(Fox, 2002). Another frequently used dairy enzyme in cheese production is catalase.
It is employed in producing some cheeses in lieu of pasteurization (e.g. Swiss
cheese) to save regular milk proteins that are responsible for providing certain
benefits to finished product and flavour enhancement of the cheese (Fox, 2002).
Rennet, which is also known as rennin, is a combination of pepsin and chymosin
extracted from animals as well as microbiological sources. It is also employed in the
cheese manufacturing industry to curdle milk (Merheb-Dini et al., 2010).
This chapter aims to emphasize on various endogenous and exogenous dairy
enzymes like lactase, amylase, lipases, transglutaminase, protease, catalase, and
rennet which are used widely, not only in the production of milk and milk-based
products, but also lactose-free products. It briefly delineates the structure, sources,
and applications of these enzymes in the dairy industry. Additionally, the chapter
also meticulously explains the use of these dairy enzymes in the production of
cheese, lactose-free milk, fermented milk as well as in packaging.
142 A. Agarwal et al.
All biochemical processes and metabolic pathways are propelled and modulated by a
group of enzymes mobilizing a series of reactions. These enzymes catalyse reactions
and improve the product yield up to several folds. However, in their absence, the rate
of reaction decelerates, and eventually reaches impasse. Enzymes, on the basis of
their source, can be broadly classified into two categories—exogenous enzymes and
endogenous enzymes (EHEs). Enzymes secreted by animals are known as exoge-
nous enzymes. These are used to enhance the process of digestion in animals.
Optimizing the conditions required for exogenous enzymes is a tiresome job,
which include optimum temperature, pH, and enzyme-substrate concentration.
Additionally, these enzymes are expensive, and hence, increase the ultimate cost
of the by-product. Contrariwise, EHEs are enzymes that are added to the animal
feed. The efficacy, ease of use, unique properties, and profitability of these enzymes
make them an appealing candidate for industries for the production of numerous
by-products (Ramawat & Mérillon, 2015).
5.2.1 Lactase
5.2.1.1 Structure
Lactase or β-Galactosidase is a tetramer, that is four identical polypeptide chains.
Each chain comprises 1023 amino acids that amalgamate to create five distinct
structural domains. Of the five domains, one is a jelly roll barrel, whereas the rest
comprise β-sandwich, fibronectin, and a central domain. The central domain with
TIM-type barrel is made of tetramer subunits and acts as the active site (Huber et al.,
1976). However, cleavage of tetramer to dimers causes the active site to inactivate.
The α-peptide present at the n-terminal of lactase is involved in α-complementation
and contributes in subunit interface (Corral et al., 2006).
5 Enzyme in Milk and Milk Products: Role and Application 143
5.2.1.2 Sources
Lactase can be found in fungi, bacteria as well as in yeasts. It can be mostly found in
plants, like apples, apricots, peaches, and almonds. However, for industrial and
commercial scale, lactase is usually acquired from fungi like Aspergillus and
Kluyveromyces (Zhou & Chen, 2001).
Bacterial Sources
Lactase derived from bacterial sources possesses high activity, stability, and ease of
fermentation, and hence, it is utilized to hydrolyse lactose. Studies have reported
Lactobacillus and Bifidobacterium species to be an efficacious probiotic. As a result,
they are commonly used to obtain lactase. It has been reported that lactase derived
from bacterial strains like Bifidobacterium longum strain CCRC 15708,
Bifidobacterium infantis strain CCRC 14633, and Bifidobacterium longum CCRC
15708 possess high enzyme activity (Hsu et al., 2007).
Yeast Sources
Lactase is extracted from yeast to produce lactose-free products. Kluyveromyces
lactis, a variety of yeast found in dairy environment, is one of the most commercially
used sources of lactase (Pivarnik & Senecal, 1995). The optimal pH of lactase
obtained from a yeast lies between 6.0 and 7.0. Studies have reported that
Kluyveromyces marxianus, another variety of yeast, can be utilized to produce
homologous enzymes of lactase and other heterologous proteins that have the ability
to thrive on a variety of substrates with lactose as their only source of energy. It has
been found that a cold-active acidic lactase extracted from Guehomyce spullulans, a
strain of psychrophilic yeast species, can be utilized for whey and milk hydrolysis in
food industries (Saqib et al., 2017).
Fungal Sources
Fungi have been known to yield enzymes that are highly stable. Lactases obtained
from fungal sources have an optimal acidic pH range of 2.5–5.4. This makes them
highly potent for hydrolysing lactose of an acidic material such as whey. The most
commonly used fungal sources to produce lactases that have been accepted as
“generally recognized as safe” (GRAS) by FDA are Kluyveromyces lactis,
Kluyveromyces fragilis (Saccharomyces fragilis), and certain Aspergillus species.
Lactase extracted from Aspergillus oryzae is used on commercial scale and has been
reported to be an effective source for whey utilization. Lactase generated from
another fungus, that is, Aspergillus niger, is used to remove galactose residues
from oligosaccharides and polysaccharides that are obtained from plant (Kazemi
et al., 2016).
Plants
Lactase can also be obtained from a variety of plant sources like apple, tomato,
mango muskmelon, avocado, coffee, kiwifruit, and Japanese pear plants (Seddigh &
Darabi, 2014). These enzymes contribute to plant growth, ripening of fruits, and
hydrolysis of lactose. It has been reported that this enzyme reduces galactosyl
144 A. Agarwal et al.
content of cell wall of persimmons, causes cell wall hydrolysis in papaya, and
facilitates the process of fruit ripening (Saqib et al., 2017).
Whey Utilization
Cheese industry has been known to yield a massive amount of whey as a derivative.
The chief constituents of whey include lactose, proteins, and minerals. The lactose in
whey is associated with chemical oxygen demand and biochemical oxygen demand.
Therefore, despite the fact that in developing countries a substantial amount of whey
is used to produce permeates and protein concentrates, whey, when disposed in
water streams, leads to severe water pollution. Nevertheless, whey, when converted
into beneficial products such as ethanol as well as lactase, can be utilized for the
production of lactose-free food products (Kokkiligadda et al., 2016). Additionally,
treating whey with lactase can help in converting it into a substrate that is readily
available for cell cultivation (Parashar et al., 2016). Another application of lactase
5 Enzyme in Milk and Milk Products: Role and Application 145
Production of Galacto-Oligosaccharides
Another application of lactase enzyme includes the production of galacto-
oligosaccharides (GOS). GOS are nondigestible prebiotics and hence play a crucial
role in improving human health by modifying intestinal microflora of human and
promoting the growth of useful bacteria in the intestine such as Lactobacillus and
Bifidobacterium species. GOS are produced from the transglycosylation activity of
lactase during lactose hydrolysis. It can be stored at room temperature for longer
period of time due to its acidic conditions. This characteristic of GOS makes it
capable of being used for the production of different products without any decom-
position. The oligosaccharide content in GOS ranges from 1% to 45% and depends
further on the source of enzyme and the total amount of saccharides (Pazur, 1953).
GOS is also present in human milk and is known to help in increasing the natural
microflora of small intestine of a breast-fed infant, that is bifidobacteria. This
bifidogenic activity of GOS plays a vital role in reducing the number of pathogenic
bacteria. In view of this characteristic, companies dealing with infant foods are now
producing milk- and cereal-based food products containing GOS (Matto & Husain,
2006). Additionally, GOS also functions as soluble fibres and gets fermented by the
natural microflora present in large intestine. This fermentation produces end
products that are short-chain fatty acids that help in reducing the pH of fecal matter.
The GOS is indigestible by the bacteria present in mouth; due to this reason they do
not contribute to the development of cariogenicity (Matioli et al., 2003).
5.2.2 Amylase
Amylases are enzymes that degrade starch by catalysing the process of hydrolysis of
internal α-1,4-glycosidic bonds of polysaccharides like maltose, glucose, dextrin,
and maltotriose, while retaining the α-anomeric configuration in the products
(Takata et al., 1992). They can be found in a variety of organisms. These enzymes
are known to have multiple applications, and hence, hold a major share in market for
the sales of enzymes (Vaidya et al., 2015). For centuries together, amylases derived
from microbial and plant sources have been utilized in the brewing industry.
Amylases extracted from fungal sources have been employed in the Asian cuisine
(Hernández et al., 2006). The growing research interest in amylases obtained from
bacterial, fungal, and viral sources in comparison to amylases that are derived from
animals and plants sources can be ascribed to the ease in its large-scale production
and industrial application (Mukherjee et al., 2009).
Amylases (EC 3.2.1.0) are classified into three subtypes that are α-amylase,
ß-amylase, and γ-amylase. The two types of amylases—endoamylases and
exoamylases, are categorized on the basis of the method by which the glycosidic
bond is attacked (Vaidya et al., 2015). α-Amylases (α-1,4-glucan-glucanohydrolase,
146 A. Agarwal et al.
EC 3.2.1.1) are extracellular enzyme (Cherry et al., 2004) that is responsible for
degrading α-1,4-glucosidic linkage of starch and associated products and producing
oligosaccharides (Baysal et al., 2008). β-Amylase (1,4-α-D-glucan maltohydrolase;
glycogenase; saccharogen amylase, EC 3.2.1.2), another form of amylase, works
from its non-reducing end and mobilizes the process of hydrolysis of the second
α-1,4-glycosidic bond by cleaving two maltose units at a time. At the time of fruit
ripening, this enzyme helps in cleaving starch present in the fruit and results in the
sweet flavour of ripe fruit. γ-Amylase (EC 3.2.1.3) (Alternative names: Glucan
1,4-α-glucosidase; amyloglucosidase; Exo-1,4-α-glucosidase; glucoamylase; lyso-
somal α-glucosidase; 1,4-α-D-glucan glucohydrolase), the third sub-type of
amylases, is known to cleave α-(1–6)-glycosidic linkage along with the cleavage
of the last α-(1–4)-glycosidic linkages at the non-reducing end of amylose and
amylopectin to yield glucose. This sub-type has an optimum pH of 3, and hence,
is efficacious in acidic conditions (Saini et al., 2017).
5.2.2.1 Structure
Amylase possesses a three-dimensional structure that enables it to bind to substrates.
The high activity of specific catalytic groups helps in the cleavage of the glycoside
links (Iulek et al., 2000). The human α-amylase is made of 512 amino acids in a
single oligosaccharide chain. It comprises calcium and has a molecular weight of
57.6 kDa (Whitcomb & Lowe, 2007). Amylase possesses three domains: A, B, and
C. The A domain which is the largest exhibits a classical barrel-shaped (β/α)8 super
structure. The B domain is cemented to A domain by disulphide bond and is
positioned between the A and C domains. The C domain possesses a sheet-like
organization and is attached to the A domain by a simple polypeptide chain. The
substrate binding site or the active site of the enzyme is positioned in a long cleft
which is situated between the carboxyl end of the A and B domains. The calcium ion
(Ca2+) is positioned amid the A and B domains. This ion may serve in the process of
stabilizing the three-dimensional structure of the enzyme and act as an allosteric
activator. The active site of the enzyme comprises 5 sub-sites with the catalytic site
located at sub-site 3. Substrate can attach to the first glucose residue present in
sub-site 1 or 2, thereby permitting the division to take place between the first and
second or second and third glucose residues (Whitcomb & Lowe, 2007).
5.2.2.2 Sources
Amylases are a group of enzymes produced in nature by plants, animals, and
microorganisms.
Alpha Amylase
Alpha amylase can be isolated from a variety of sources like plants, animals, and
microbes (Pandey et al., 2000). Although alpha amylase has been extracted from a
variety of bacteria, fungi, actinomycetes, and yeasts, the enzymes obtained from
fungi and bacteria have been found to have major industrial applications. This can be
ascribed to their consistency, cost-effectiveness, less time and space requirement for
their production, and ease of process optimization and modification (Ellaiah et al.,
5 Enzyme in Milk and Milk Products: Role and Application 147
2002). The microbial enzyme derived from the microbial sources is capable of
meeting the industrial demand and a huge amount of them are commercially
available. For centuries, amylases derived from microbes and plants have been
used as food additives (Hernández et al., 2006). The bacterial amylases are mostly
derived from Bacillus sp. like B. subtilis, B. stearothermophilus, B. licheniformis,
and B. amyloliquefaciens. Most of the fungal sources of amylases belong to terres-
trial isolates like Aspergillus species (Jamal et al., 2011). The amylases derived from
fungi are extensively utilized to prepare oriental foods (Popovic et al., 2009). The
alpha amylases have also been isolated from filamentous fungi, which are high-
yielding producers of extracellular proteins (Coradin et al., 2011).
5.2.3 Transglutaminase
For the first time, TGases were derived from tissues of mammals and body fluids
to investigate their applications in industry. The first commercially available TGase,
extracted from the liver of guinea pig, was not found to be appealing candidate for
industrial use ascribing to the limited supply and requirement of Ca2þ for activation
which resulted in the high production cost of the enzyme. Likewise, another TGase
known as Factor XIII, which was derived from blood plasma, exhibited a major
drawback of red pigmentation and the requirement of thrombin for activation. In
1980s, during the screening of microorganisms for TGase activity, microbial TGase,
which is independent of Ca2þ and possesses low substrate specificity, was identified
from Streptomyces mobaraensis. Production of mTGase results from conventional
fermentation of the wild-type S. mobaraensis strain (GRAS status) with consequent
extraction of the protein that is secreted. Thus far, mTGase is the only commercially
available TGase in the food industry (Jaros & Rohm, 2016).
5.2.3.1 Structure
mTGase, a monomeric protein with 331 amino acids, has an isoelectric point of 8.9.
Its secondary structure comprises 8 β-strands adjoined with 11 α-helices. The
enzyme acquires a disk-like shape that has a deep cleft at the edge. At this cleft is
situated a single cysteine residue (Cys64). Molar mass of mTGase as evaluated by
the technique of mass spectrometry and amino acid sequencing is 37.86 kDa.
Usually, mTGase exhibits activity at pH range of 4 and 9. However, the optimum
pH of the enzyme lies in the range 6–7. The optimum temperature for mTGase is
50 C. Although it is immobilized at 70 C within minutes, it maintains some
residual activity at near-freezing temperatures. Another characteristic of mTGase is
its stability under high hydrostatic pressure. This characteristic provides the
prospects of alteration of proteins, even in situations where they are not available
for mTGase under atmospheric pressure (Jaros & Rohm, 2016).
5.2.3.2 Sources
TGases are widely spread in nature (Kashiwagi et al., 2002). They can be derived
from various invertebrates and microbial cells (Yu et al., 2008; Griffin et al., 2002) as
well as mammalian tissues (Yasueda et al., 1994). TGases can also be found in
tissues of various plants such as orchard apple, soy, fodder beet, and topinambour
(Falcone et al., 1993). These enzymes have been extracted from microbes belonging
to Streptoverticillium sp. and Physarum polycephalum. TGases are biosynthesized
as an extracellular enzyme by Streptoverticillium sp., Streptoverticillium
cinnamoneum subsp. cinnamoneum (Duran et al., 1998), Streptomyces netropsis
(Yu et al., 2008), Streptoverticillium griseocarneum (Gerber et al., 1994),
Streptoverticillium ladakanum (Ho et al., 2000), and Streptomyces lydicus
(Færgemand & Qvist, 1997).
(Kieliszek & Misiewicz, 2014). A study reported that TGases at the level of 0.02%
improve melting, firmness, flexibility, and organoleptic properties of high fat Moz-
zarella cheese. However, in the case of Mozzarella with low fat content, 0.05% of
TGase provides adequate meltability, flexibility, free oil formation, and organolepti-
cally to the cheese. The cross-linking property of TGase helps in augmenting soft
cheese production, reducing syneresis, extending shelf life, and decreasing nutrient
bioavailability for deteriorative microorganisms (Mahmood, 2009). It has also been
reported that this enzyme can be utilized to partially replace fat in ice-creams, and it
is also responsible for increasing overrun, partial amalgamation of fat globules,
consistency, melting resistance, firmness, and viscosity in ice cream (Tafes, 2019).
A study reported that up to 0.5% of TGase was more efficacious to enhance the
functional properties of yogurt that was made from goat milk. The cross-linking
property of TGase was found to be accountable for enhancing the gel consistency
and reducing the whey separation substantially in yoghurt (Farnsworth et al., 2006).
Another study reported that when stirred yogurt was developed through covalent
cross-linking by the combined action of inactivated mTGase and glutathione, no
negative effects on the fermentation of yogurt were observed. However, a substantial
rise in apparent viscosity as well as protein polymerization was observed in yogurt
that was only developed by TGase (Bönisch et al., 2007). The use of TGase in high
concentration was found to reduce syneresis and increase the viscosity of yogurt.
However, a minor bottleneck was observed with regard to the growth of Lactic Acid
Bacteria. This led to reduced production of acid and acetaldehyde in comparison to
those reported for the control. Nevertheless, TGase concentration up to 0.3 g/L was
reported to be optimum and was recognized as an acceptable substitute to stabilizers
to be added in the production of fat-free yogurt (Ozer et al., 2007). Studies identified
TGase as an efficacious candidate to enhance the physical properties of yogurt when
its constitution was modified by the addition of whey (Şanli et al., 2011).
Low levels of lactase production a human beings has been known to be a leading
cause of “lactose intolerance”, which is an intestinal symptomatic condition. A
lactose-intolerant individual may experience clinical symptoms like nausea, abdom-
inal pain, flatulence, diarrhea, and bloating after consuming food that contains
lactose. It has been estimated that about 70% of the world population, including
all the age groups, is suffering from lactase deficiency (Xavier et al., 2018). Lactose-
intolerant individuals can consume dairy-fermented products that comprise very
little or no lactose in them (Saqib et al., 2017). Currently, there is an increase in
demand for lactose-free dairy among the lactose-intolerant populations. This can be
ascribed to the presence of little or no lactose in these products and their ability to
provide vital macro- and micro-nutrients present in milk to individuals suffering
from lactose intolerance (Dekker et al., 2019).
150 A. Agarwal et al.
The dairy industry employs different processing techniques to reduce the lactose
content in the dairy products to produce lactose-free products. Cheese production
involves certain techniques which help in reducing the lactose content. For example,
Gouda cheese production involves a curd washing step to reduce the lactose content.
Although the lactose content in most of the cheeses is already relatively low without
ripening, in several other types of cheeses, the lactose content is reduced during the
process of ripening with the help of lactic acid bacteria. Generally, aged and hard
cheeses like Parmesan, Swiss, or Cheddar cheeses have a very low concentration of
lactose. Contrariwise, young and fresh cheeses may consist enough lactose that may
be capable of inducing a reaction among individuals with lactose intolerance,
depending on the quantity consumed. Butter is another dairy product with low
lactose content. The majority of water-soluble constituents of milk inclusive of
lactose are eliminated, during the production of butter resulting in the reduction of
lactose concentration in butter (Dekker et al., 2019).
Besides reducing the lactose content in dairy products, another solution to
produce lactose-free dairy products includes hydrolysing lactose into glucose and
galactose using the lactase enzyme. The monosaccharides produced get easily
adsorbed in the small intestine and avert the incidence of symptoms associated
with lactose intolerance. Currently, neutral lactases and acid lactases are the two
kinds of commercially available lactases (Dekker & Daamen, 2011). The former is
chiefly employed on an industrial scale to produce lactose-free dairy products, while
the latter is provided as a dietary supplement to the consumers for its consumption
along with regular dairy products to induce cleavage of lactose in the stomach
(Dekker et al., 2019).
Researches have reported that lactose dosage of less than 12 g per meal may result in
mild symptoms among lactose-intolerant individuals; however, the dairy industry
aims to maintain the lactose content as low as possible (Suchy et al., 2010).
Currently, there does not exist global agreement on the regulatory requirements for
lactose-free claims. In the past, lactose reduction to 0.5% or 0.1% was considered
enough by the dairy producers, but, in some countries, lactose content less than
0.01% is considered to be the current requirement to consider milk to be lactose-free.
To achieve such low lactose concentration in milk production entails special atten-
tion to the milk processing, to the dosage and efficiency of the enzyme used in this
process, and to sensitive analytical methods to determine such low amounts of
lactose (van Scheppingen et al., 2017).
Presently, there exist two processes that are used in the production of lactose-free
milk (Troise et al., 2016; Harju, 2004). These include batch process and aseptic
process and both of them use soluble lactase enzyme. Although various studies have
suggested the processes that rely on an immobilized enzyme, these processes have
not been employed in industrial practice to produce lactose-free milk either in the
5 Enzyme in Milk and Milk Products: Role and Application 151
past or in present due to issues related to the microbial stability of the end product
(Finocchiaro et al., 1980).
Fundamentally, there exist two techniques to obtain sterile lactase enzyme. The
first technique involves pre-sterilization of lactase by the enzyme manufacturer and
usage of distinct sterile dosing equipment that is required for the sterile injection.
The second technique involves the filter-sterilization of the unsterile enzyme imme-
diately before incorporating it to the sterilized milk at the dairy industry (Dekker
et al., 2019).
Considering the high incubation time and temperature required in this process,
the enzyme dosage can be kept much lower. Process control in this process is not
present as the enzyme is vigorous only in the final milk package. The dairy
manufacturer should also consider the aspects such as deviation in the storage
temperature in un-thermostated warehouses, resulting from the seasonal transitions
such as summer to winter, when dosing the enzyme (Dekker et al., 2019).
During the aseptic process, special equipment and consumable costs are essential
requirements specifically for the in-factory filtration. Highly skilled operators are
employed during the process of lactose injection in order to prevent microbial
contamination in the milk. When organized appropriately, the process can function
as a completely continuous operation, thus making the aseptic process a desired
technique in industries requiring a high yield (Dekker et al., 2019). The aseptic
process for the production of lactose-free UHT milk can only be considered to be a
fully developed technique after the crucial refinement of the lactase enzyme quality.
Researches have reported that the arylsulfatase side activity in the preparation of
lactose may result in serious medicinal off-flavours during storage. This has been
ascribed to formation of p-cresol from sulphonated-cresol, which occurs naturally in
the milk (De Swaaf et al., 2006). Currently, arylsulfatase-free lactase enzymes are
available commercially. It is important for a lactose-free UHT milk producer to
consider utilizing only the supreme quality lactase enzymes for this process in order
to avert shelf life-related complication (Dekker et al., 2019).
Lactose hydrolysis in milk has been known to augment the monosaccharides’
content, resulting in an efficient Maillard reaction, which may be amplified due to the
inadequate proteolysis by proteases. These proteases may be present naturally in the
milk or they may originate from the lactase preparation. The amplified reactions lead
to the increase in the formation of off-flavours and greater browning of lactose-free
milk in comparison to regular milk. When stored at higher temperature, it also results
in a reduced nutritional value of the product (Troise et al., 2016; Jansson et al., 2014;
Evangelisti et al., 1999). The enhanced maillard reaction may be considered as the
critical determinant of the decreased shelf life of lactose-free UHT milk in compari-
son to regular UHT milk. In the past, various researches have suggested that the
batch process for the production of lactose-free UHT milk may result in compara-
tively higher browning than the milk produced using the aseptic process (Mendoza
et al., 2005). However, recent studies have reported the relevance of the storage
conditions (temperature) and lactase quality for determining shelf life (Troise et al.,
2016). Excellent shelf life was found for lactose-free UHT milk produced with the
batch process, and milk browning during storage is, therefore, largely independent of
the production process that is used (Dekker et al., 2019).
5 Enzyme in Milk and Milk Products: Role and Application 153
Besides lactose-free milk, various other lactose-free dairy products are manufactured
with the use of enzymatic treatment. Flavoured milk is one such product that is
produced using a similar process (McCain et al., 2018). The comparatively stronger
production of colour and flavour by the flavoured lactose-free milk has been reported
to reduce browning and production of off-flavours due to the maillard reaction.
Unlike regular lactose-free milk, the flavoured lactose-free milk requires less addi-
tion of extra sugar, due to the lactose treatment; however, in certain flavoured milks
like highly sugared chocolate milk, the process of lactose hydrolysis may not be
adequate for complete replacement of sugar addition (Li et al., 2015), and thus there
may be a requirement to add additional sweeteners. Another lactose-free dairy
product is dairy powder, which can be developed from either milk or whey, which
is made lactose-free with the batch process. A major setback reported during the
production of lactose-free dairy powder is the high monosaccharide content in the
treated milk, which causes a reduction in the glass-transition temperature. This
phenomenon further results in the contamination of the spray dryer when the drying
conditions are not adjusted (Torres et al., 2017). Additionally, packaging of the
lactose-free dairy powder should be done cautiously as the high hygroscopic nature
of the powder results in its caking during storage. It is because of these challenges
that the lactose-free dairy powders are not a part of big markets like regular milk
powders. Cheese is yet another dairy product that can be made lactose-free. This is
done using lactase enzyme to incubate the cheese milk before renneting. This
method is used majorly for cheeses that are young and fresh, as they comprise a
considerable quantity of lactose. In the case of ripened cheeses, lactase incubation is
not required as lactose gets completely consumed by the lactic acid bacteria. The
studies conducted in the past have reported that unlike yoghurt, lactase treatment of
cheese milk stimulates the process of acidification during cheese manufacturing.
Furthermore, the addition of lactase during the process of ripening enhances the
flavour of cheese. It is unclear if these effects are a result of the stimulation of the
cheese microbial flora caused either by lactose hydrolysis or residual proteolytic
activity occurring during lactase preparations that were available commercially in
the past (Marschke et al., 1980). Lactose-free ice cream can also be produced using
either lactose-free milk or powders in the ice cream mix (Abbasi & Saeedabadian,
2015). The production of lactose-free ice cream involves the addition of lactase
during the ageing period before freezing, once the process of pasteurization and
incubation is complete. The lactose hydrolysis increases the monosaccharide con-
centration which reduces the freezing point of the ice cream. This results in the
production of ice cream with a soft texture at the same temperature. Although this
may be a desirable attribute in several frozen desserts, it may also cause faster
melting. Additionally, the sweetness resulting due to the lactose hydrolysis may
help in reducing the addition of extra sugar in the ice creams, and consequently,
raising the melting temperature again. In order to avert crystallization of lactose in
the ice creams which causes a sensory defect known as “sandiness”, lactase treat-
ment is employed to convert lactose into two monosaccharides—glucose and
154 A. Agarwal et al.
galactose, which are highly soluble at high temperature. This treatment is essential
when ice cream is produced using whey powder or WPC as the high lactose content
may result in the formation of crystals during freezing (Abbasi & Saeedabadian,
2015).
5.4 Lipase
position of fatty acid in glycerol backbone, and degree of unsaturation. Also, its
activity depends upon the pH of the solution such as lipase is stable at neutral pH and
pH 4.0–8.0. For the catalytic activity of enzyme lipase, greater mild conditions and
large surface area are required (Aravindan et al., 2007). Among the different lipases
produced from plants, animals, and microorganisms, the most vital are microbial
lipases. Microbial lipases are gaining greater attention due to their non-toxic nature,
convenience, high catalytic activity, greater yield, rapid growth, and usage due to
absence of seasonal interference as well as availability of cheaper media for growth
(Ray, 2012). Microbial lipases are utilized in different areas such as in cosmetics,
detergents, pharmaceuticals, formation of aliphatic acids, leather industry, and in
treatment of waste excreted from different industries (Seitz, 1974).
Lipases obtained from microorganisms are broadly classified into two major
categories on the basis of their specificity, namely, non-specific and 1,3-specific
lipases. Non-specific lipases are those lipases which catalyse the reactions indepen-
dent of the position esterified in glyceride molecules such as lipases obtained from
Staphylococcus aureus, Pseudomonas spp., Candida cylindracea, and
Chromobacterium viscosum. On the other hand, 1,3-specific lipases are those
which react and catalyse only outer, i.e. sn-1 and sn-3, positions of the glycerol
backbone such as lipases obtained from Rhizopus oryzae, Rhizopus delemar, Rhizo-
pus niveus, Aspergillus niger, Mucor javanicus, Candida lipolytica, and Humicola
lanuginose (Balcao & Malcata, 1998).
Lipases are used as biocatalysts, especially microbial lipases, in various industries
such as food industry due to their various functions and properties such as
regioselectivity, stereospecificity, able to function in different pH and temperature,
substrate specificity, and potential to act upon heterogeneous reactions at the inter-
face between water-soluble and water-insoluble systems (Verma & Kanwar, 2008).
On the other hand, bacterial lipases are stable at a temperature range of 30–60 C and
under neutral or alkaline pH except the lipase produced from P. fluorescens SIK W1,
which is stable under acidic pH of 4.8. Bacterial lipases are also stable in organic
solvents (Verma et al., 2012).
Lipase has a wide range of applications in dairy industry such as in improving the
flavor and modifying the fatty acid chain of dairy products including cheese. The
major role of lipases in dairy industry involves the hydrolysis of milk fat. Tissues
from animals such as pancreatic glands, i.e. bovine and porcine, and pre-gastric
tissues of young ruminants, i.e. lamb and calf for lipase production, are widely used
in traditional cheese flavor enhancement process (Ray, 2012).
Recent applications of lipase in dairy industry involve the production of cheese,
its flavor enhancement, and lipolysis of cream and butterfat. Lipases are utilized in
the manufacturing of enzyme-modified cheese for providing concentrated flavor to
cheese. In this process, cheese is incubated with enzymes at high temperature and
thus leads to the formation of enzyme-modified cheese which can be used as an
156 A. Agarwal et al.
ingredient in various products such as sauces, dressings, dips, snacks, and soups.
Addition of lipases in the production of dairy products leads to the release of short
chain fatty acids, namely C4 and C6, which provide sharp and tangy flavor to the
products along with the release of medium chain fatty acids, namely C12 and C14,
which provide soapy taste to the finished products (Hasan et al., 2006). Exogenous
lipase enzyme helps in accelerating the cheese ripening process. On the other hand,
addition of free lipases results in greater lipolysis which deteriorates the texture as
well as flavor of cheese, but encapsulating the lipase helps in maintaining and
stabilizing the enzyme-substrate ratio, thus eliminating the problem of excessive
lipolysis (Houde et al., 2004). It is also used in the manufacturing of chocolates by
production of cocoa butter-equivalent from palm oil with the technique using
interesterification. There are many commercial lipase enzymes which are used for
the production of different milkfat flavor profiles for milk-based products, such as
Snow plum blossom and Palatase 20,000 L (Sarmah et al., 2017).
In addition to the dairy industry, lipase also plays a vital role and possesses various
applications in other industries such as cosmetics, pharmaceutical, leather, paper,
food industry, etc. In food industry, lipase is widely used in the preparation of infant
formulas which can provide an alternative of breast milk. The enzyme is also used
for the modification of lipid characteristics of oils rich in omega-6 fatty acids such as
coconut, sunflower, olive, rice bran, corn oil, and oils rich in omega-3 fatty acids
such as linseed and fish oil. In pharmaceutical industries, lipase is used as an
ingredient in the formation of products and also utilized in the interesterification of
vegetable oil. It helps in enhancement of colour and texture of the dough and used as
an emulsifier in many bakery industries. Lipase is also essential in the formation of
cosmetic component, i.e. isopropyl myristate, for manufacturing of cosmetic items
(Sarmah et al., 2017; Houde et al., 2004).
Lipases are also utilized in detergent industry as an additive in household and
laundry detergents in order to vanish the oil stains from the fabrics as these enzymes
can withstand harsh pH (10–11) and temperature (30–60 C) required for washing. It
also plays a vital role in paper manufacturing industry by removing the pitch, which
is a hydrophobic portion of wood, i.e. waxes and triglycerides, from the pulp
required for manufacturing paper (Sharma et al., 2001). In addition to this, microbial
lipases such as Candida rugosa, Mucor miehei, Pseudomonas fluorescens, Rhizopus
oryzae, and Aspergillus niger are used for the production of biodiesel. Presently,
lipase produced from the microbe Streptomyces sp. was considered as most effective
for biodiesel production (Chandra et al., 2020).
5 Enzyme in Milk and Milk Products: Role and Application 157
Different types of rennet are used in cheese manufacturing which are classified on
the basis of their source such as animal rennet, plant rennet, and fungal rennet. Under
the classification of animal rennet, calf rennet is widely utilized for cheese produc-
tion, the reason being its high amount of chymosin. Presently, adult bovine rennet is
used as an alternative towards calf rennet due to high pepsin content in adult bovine
rennet that results in a product with high pH sensitivity and greater proteolytic
activity (Harboe et al., 2010).
Many proteolytic enzymes are derived from plants such as bromelain from
pineapple, ficin from Ficus spp., and papain from papaya. Other plants include fig,
pumpkin, soybean, ash gourd, and milkweed. The enzymes are embedded in the
buds, leaves, flowers, seeds, latex, and roots of the plants. But the proteolytic
capability of enzymes from vegetable sources is low which makes them unsuitable
for cheese production. And if excessive proteolysis occurs, it will lead to bitterness
in the finished products. Although for cottage cheese and soft cheese production,
extract from berries of Withania coagulans was used, it is also not suitable for
cheddar cheese manufacturing (Garg & Johri, 1994).
Rennet clotting enzymes are also produced from fungi such as rennet from Mucor
miehei and Endothia parasitica. Enzyme obtained from Mucro miehei results in an
excellent quality of cheddar cheese, even with high ripening process due to greater
stability between 4.0 and 6.0 pH with no loss of its activity and also due to its high
heat stability. However, rennet obtained from Endothia parasitica is also useful in
158 A. Agarwal et al.
cheddar cheese production, but its high proteolytic activity leads to 1.2% yield loss
of the cheese (Brown & Ernstrom, 1988).
References
Abbasi, S., & Saeedabadian, A. (2015). Influences of lactose hydrolysis of milk and sugar reduction
on some physical properties of ice cream. Journal of Food Science and Technology, 52(1),
367–374. https://doi.org/10.1007/s13197-013-1011-1
Andreazzi, A. S. R., Pereira, M. N., Reis, R. B., Pereira, R. A. N., Morais Júnior, N. N., Acedo,
T. S., Hermes, R. G., & Cortinhas, C. S. (2018). Effect of exogenous amylase on lactation
performance of dairy cows fed a high-starch diet. Journal of Dairy Science, 101(8), 7199–7207.
https://doi.org/10.3168/jds.2017-14331
Aravindan, R., Anbumathi, P., & Viruthagiri, T. (2007). Lipase applications in food industry.
Indian Journal of Biotechnology, 6, 141–158.
Balcao, V. M., & Malcata, F. X. (1998). Lipase catalyzed modification of milkfat. Biotechnology
Advances, 16, 1–33.
Baysal, Z., Uyar, F., Doğru, M., & Alkan, H. (2008). Production of extracellular alkaline α-amylase
by solid state fermentation with a newly isolated Bacillus sp. Preparative Biochemistry and
Biotechnology, 38(2), 184–190. https://doi.org/10.1080/10826060701885167
Beauchemin, K., Colombatto, D., Morgavi, D., & Yang, W. (2003). Use of exogenous fibrolytic
enzymes to improve feed utilization by ruminants. Journal of Animal Science, 81(14_Suppl_2),
E37–E47. https://doi.org/10.2527/2003.8114_suppl_2E37x
Bjorkling, F., Godtfredsen, S. E., & Kirk, O. (1991). The future impact of industrial lipases. Trends
in Biotechnology, 9(1), 360–363. https://doi.org/10.1016/0167-7799(91)90119-3
Bönisch, M. P., Huss, M., Weitl, K., & Kulozik, U. (2007). Transglutaminase cross-linking of milk
proteins and impact on yoghurt gel properties. International Dairy Journal, 17(11), 1360–1371.
https://doi.org/10.1016/j.idairyj.2007.01.019
Brandão, R. L., Nicoli, J. R., & de Souza Figueiredo, A. F. (1987). Purification and characterization
of a β-galactosidase from Fusarium oxysporum var. lini. Journal of Dairy Science, 70(7),
1331–1337. https://doi.org/10.3168/jds.S0022-0302(87)80152-2
Brown, R. J., & Ernstrom, C. A. (1988). Chapter 12: Milk-clotting enzymes and cheese chemistry.
In Fundamentals of dairy chemistry (pp. 609–654). Van Nostrand Reinhold. https://doi.org/10.
1007/978-1-4615-7050-9_12
Callahan, B. P., & Miller, B. G. (2007). OMP decarboxylase—An enigma persists. Bioorganic
Chemistry, 35(6), 465–469. https://doi.org/10.1016/j.bioorg.2007.07.004
Carrara, C. R., & Rubiolo, A. C. (1994). Immobilization of β-galactosidase on chitosan. Biotech-
nology Progress, 10(2), 220–224. https://doi.org/10.1021/bp00026a012
Champluvier, B., Kamp, B., & Rouxhet, P. G. (1988). Immobilization of β-galactosidase retained in
yeast: Adhesion of the cells on a support. Applied Microbiology and Biotechnology, 27(5–6),
464–469. https://doi.org/10.1007/BF00451614
Chandra, P., Enespa, Singh, R., & Arora, P. K. (2020). Microbial lipases and their industrial
applications: A comprehensive review. Microbial Cell Factories, 19(1), 2–42. https://doi.org/
10.1186/s12934-020-01428-8
Cherry, H. M., Hossain, M. T., & Anwar, M. N. (2004). Extracellular glucoamylase from the isolate
Aspergillus fumigatus. Pakistan Journal of Biological Sciences, 7(11), 1988–1992. https://doi.
org/10.3923/pjbs.2004.1988.1992
Coradin, J. H., Braun, A., Viccini, G., de Lima Luz, L. F., Krieger, N., & Mitchell, D. A. (2011). A
three-dimensional discrete lattice-based system for modeling the growth of aerial hyphae of
filamentous fungi on solid surfaces: A tool for investigating micro-scale phenomena in solid-
state fermentation. Biochemical Engineering Journal, 54(3), 164–171. https://doi.org/10.1016/j.
bej.2011.02.012
5 Enzyme in Milk and Milk Products: Role and Application 159
Corral, J. M., Bañuelos, O., Adrio, J. L., & Velasco, J. (2006). Cloning and characterization of a
β-galactosidase encoding region in Lactobacillus coryniformis CECT 5711. Applied Microbiol-
ogy and Biotechnology, 73(3), 640–646. https://doi.org/10.1007/s00253-006-0510-7
Dahlqvist, A., Asp, N. G., Burvall, A., & Rausing, H. (1977). Hydrolysis of lactose in milk and
whey with minute amounts of lactase. Journal of Dairy Research, 44(3), 541–548. https://doi.
org/10.1017/S0022029900020495
De Swaaf, M. P. M., van Dijk, A. A., Edens, L., & Dekker, P. J. T. (2006). Enzyme preparation
yielding a clean taste. EP1954808 B1, November 28, 2006.
Dekker, P. J. T., & Daamen, C. B. G. (2011). Enzymes exogenous to milk in dairy technology: β-d-
galactosidase. In Encyclopedia of dairy sciences (2nd ed., pp. 276–283). Academic Press.
https://doi.org/10.1016/B978-0-12-374407-4.00152-7
Dekker, P. J. T., Koenders, D., & Bruins, M. J. (2019). Lactose-free dairy products: Market
developments, production, nutrition and health benefits. Nutrients, 11(3), 1–14. https://doi.
org/10.3390/nu11030551
Domingues, L., Lima, N., & Teixeira, J. A. (2005). Aspergillus niger β-galactosidase production by
yeast in a continuous high cell density reactor. Process Biochemistry, 40(3–4), 1151–1154.
https://doi.org/10.1016/j.procbio.2004.04.016
Duran, R., Junqua, M., Schmitter, J. M., Gancet, C., & Goulas, P. (1998). Purification,
characterisation, and gene cloning of transglutaminase from Streptoverticillium cinnamoneum
CBS 683.68. Biochimie, 80(4), 313–319. https://doi.org/10.1016/S0300-9084(98)80073-4
Ellaiah, P., Adinarayana, K., Bhavani, Y., Padmaja, P., & Srinivasulu, B. (2002). Optimization of
process parameters for glucoamylase production under solid state fermentation by a newly
isolated Aspergillus species. Process Biochemistry, 38(4), 615–620. https://doi.org/10.1016/
S0032-9592(02)00188-7
Evangelisti, F., Calcagno, C., Nardi, S., & Zunin, P. (1999). Deterioration of protein fraction by
Maillard reaction in dietetic milks. Journal of Dairy Research, 66(2), 237–243. https://doi.org/
10.1017/S0022029999003453
Færgemand, M., & Qvist, K. B. (1997). Transglutaminase: Effect on rheological properties,
microstructure and permeability of set style acid skim milk gel. Food Hydrocolloids, 11(3),
287–292. https://doi.org/10.1016/s0268-005x(97)80058-6
Falcone, P., Serafini-Fracassini, D., & Del Duca, S. (1993). Comparative studies of
transglutaminase activity and substrates in different organs of Helianthus tuberosus. Journal
of Plant Physiology, 142(3), 265–273. https://doi.org/10.1016/S0176-1617(11)80421-9
Farnsworth, J. P., Li, J., Hendricks, G. M., & Guo, M. R. (2006). Effects of transglutaminase
treatment on functional properties and probiotic culture survivability of goat milk yogurt. Small
Ruminant Research, 65(1–2), 113–121. https://doi.org/10.1016/j.smallrumres.2005.05.036
Fazouane-Naimi, F., Mechakra, A., Abdellaoui, R., Nouani, A., Daga, S. M., Alzouma, A. M.,
Gais, S., & Penninckx, M. J. (2010). Characterization and cheese-making properties of rennet-
like enzyme produced by a local Algerian isolate of Aspergillus niger. Food Biotechnology,
24(3), 258–269. https://doi.org/10.1080/08905436.2010.507149
Felicilda-Reynaldo, R. F. D., & Kenneally, M. (2016, March 2–3). Therapy: Pancreatic enzymes.
Finocchiaro, T., Olson, N. F., & Richardson, T. (1980). Use of immobilized lactase in milk systems.
Advances in Biochemical Engineering, 15, 71–88. https://doi.org/10.1007/3540096868_3
Fox, P. F. (2002). Significance of indigenous enzymes in milk and dairy products. In Handbook of
food enzymology (pp. 270–293). CRC Press.
Garg, S. K., & Johri, B. N. (1994). Rennet: Current trends and future research. Food Reviews
International, 10(3), 313–355. https://doi.org/10.1080/87559129409541005
Gerber, U., Jucknischke, U., Putzien, S., & Fuchsbauer, H. L. (1994). A rapid and simple method
for the purification of transglutaminase from Streptoverticillium mobaraense. Biochemical
Journal, 299(3), 825–829. https://doi.org/10.1042/bj2990825
Godfrey, T., & Reichelt, J. (1983). Industrial enzymology: The application of enzymes in industry
(1st ed.). Macmillan Publishers Ltd.
160 A. Agarwal et al.
Griffin, M., Casadio, R., & Bergamini, C. M. (2002). Transglutaminases: Nature’s biological glues.
Biochemical Journal, 368(2), 377–396. https://doi.org/10.1042/BJ20021234
Harboe, M., Broe, M. L., & Qvist, K. B. (2010). Chapter 3: The production, action and application
of rennet and coagulants. In Technology of cheesemaking (2nd ed., pp. 98–125). John Wiley &
Sons, Ltd.
Harju, M. (2004). Chromatographic and enzymatic removal of lactose from milk. Bulletin of the
International Dairy Federation, 389, 4–8.
Harju, M., Kallioinen, H., & Tossavainen, O. (2012). Lactose hydrolysis and other conversions in
dairy products: Technological aspects. International Dairy Journal, 22(2), 104–109. https://doi.
org/10.1016/j.idairyj.2011.09.011
Hasan, F., Shah, A. A., & Hameed, A. (2006). Industrial applications of microbial lipases. Enzyme
and Microbial Technology, 39, 235–251. https://doi.org/10.1016/j.enzmictec.2005.10.016
Hernández, M. S., Rodríguez, M. R., Guerra, N. P., & Rosés, R. P. (2006). Amylase production by
Aspergillus niger in submerged cultivation on two wastes from food industries. Journal of Food
Engineering, 73(1), 93–100. https://doi.org/10.1016/j.jfoodeng.2005.01.009
Ho, M. L., Leu, S. Z., Hsieh, J. F., & Jiang, S. T. (2000). Technical approach to simplify the
purification method and characterization of microbial transglutaminase produced from
Streptoverticillium ladakanum. Journal of Food Science, 65(1), 76–80. https://doi.org/10.
1111/j.1365-2621.2000.tb15959.x
Houde, A., Kademi, A., & Leblanc, D. (2004). Lipases and their industrial applications. Applied
Biochemistry and Biotechnology, 118, 155–170.
Hristov, A. N., McAllister, T. A., & Cheng, K. J. (1998). Stability of exogenous polysaccharide-
degrading enzymes in the rumen. Animal Feed Science and Technology, 76(1–2), 161–168.
https://doi.org/10.1016/S0377-8401(98)00217-X
Hsu, C. A., Lee, S. L., & Chou, C. C. (2007). Enzymatic production of galactooligosaccharides by
β-galactosidase from Bifidobacterium longum BCRC 15708. Journal of Agricultural and Food
Chemistry, 55, 4–9. Retrieved from http://pubs.acs.org/doi/full/10.1021/jf063126+%0Apapers3
://publication/doi/10.1021/jf063126
Huber, R. E., Kurz, G., & Wallenfels, K. (1976). A quantitation of the factors which affect the
hydrolase and transgalactosylase activities of β-galactosidase (E. coli) on lactose. Biochemistry,
15(9), 1994–2001. https://doi.org/10.1021/bi00654a029
Iulek, J., Franco, O. L., Silva, M., Slivinski, C. T., Bloch, C., Rigden, D. J., & Grossi De Sá, M. F.
(2000). Purification, biochemical characterisation and partial primary structure of a new
α-amylase inhibitor from Secale cereale (rye). International Journal of Biochemistry and Cell
Biology, 32(11–12), 1195–1204. https://doi.org/10.1016/S1357-2725(00)00053-4
Jamal, P., Idris, Z. M., & Alam, M. Z. (2011). Effects of physicochemical parameters on the
production of phenolic acids from palm oil mill effluent under liquid-state fermentation by
Aspergillus niger IBS-103ZA. Food Chemistry, 124(4), 1595–1602. https://doi.org/10.1016/j.
foodchem.2010.08.022
Jansson, T., Jensen, H. B., Sundekilde, U. K., Clausen, M. R., Eggers, N., Larsen, L. B., Ray, C.,
Andersen, H. J., & Bertram, H. C. (2014). Chemical and proteolysis-derived changes during
long-term storage of lactose-hydrolyzed ultrahigh-temperature (UHT) milk. Journal of Agricul-
tural and Food Chemistry, 62(46), 11270–11278. https://doi.org/10.1021/jf504104q
Jaros, D., & Rohm, H. (2016). Enzymes exogenous to milk in dairy technology: Transglutaminase.
In Reference module in food science (Issue July). Elsevier. https://doi.org/10.1016/b978-0-08-
100596-5.21158-x
Jelen, P., & Tossavainen, O. (2003). Low lactose and lactose-free dairy products—Prospects,
technologies and applications. Australian Journal of Dairy Technology, 58(2), 161–165.
Karasova, P., Spiwok, V., Mala, S., & Kralova, B. (2002). Beta-galactosidase activity in
psychrotrophic microorganisms and their potential use in food industry. Czech Journal of
Food, 20(2), 43–47. Retrieved from http://81.0.228.28/publicFiles/50865.pdf
Kashiwagi, T., Yokoyama, K.-I., Ishikawa, K., Ono, K., Ejima, D., Matsui, H., & Suzuki, E.-I.
(2002). Crystal structure of microbial transglutaminase from Streptoverticillium mobaraense.
5 Enzyme in Milk and Milk Products: Role and Application 161
Moschopoulou, E. (2011). Characteristics of rennet and other enzymes from small ruminants used
in cheese production. Small Ruminant Research, 101, 188–195. https://doi.org/10.1016/j.
smallrumres.2011.09.039
Mukherjee, A. K., Borah, M., & Rai, S. K. (2009). To study the influence of different components
of fermentable substrates on induction of extracellular α-amylase synthesis by Bacillus subtilis
DM-03 in solid-state fermentation and exploration of feasibility for inclusion of α-amylase in
laundry detergent formulations. Biochemical Engineering Journal, 43(2), 149–156. https://doi.
org/10.1016/j.bej.2008.09.011
Najera, A. I., Renobales, M., & Barron, L. J. R. (2003). Effects of pH, temperature, CaCl2 and
enzyme concentrations on the rennet-clotting properties of milk: A multifactorial study. Food
Chemistry, 80, 345–352.
Nozière, P., Steinberg, W., Silberberg, M., & Morgavi, D. P. (2014). Amylase addition increases
starch ruminal digestion in first-lactation cows fed high and low starch diets. Journal of Dairy
Science, 97(4), 2319–2328. https://doi.org/10.3168/jds.2013-7095
Ozer, B., Avni Kirmaci, H., Oztekin, S., Hayaloglu, A., & Atamer, M. (2007). Incorporation of
microbial transglutaminase into non-fat yogurt production. International Dairy Journal, 17(3),
199–207. https://doi.org/10.1016/j.idairyj.2006.02.007
Pandey, A., Soccol, C. R., & Mitchell, D. (2000). New developments in solid state fermentation:
I—Bioprocesses and products. Process Biochemistry, 35(10), 1153–1169. https://doi.org/10.
1016/S0032-9592(00)00152-7
Parashar, A., Jin, Y., Mason, B., Chae, M., & Bressler, D. C. (2016). Incorporation of whey
permeate, a dairy effluent, in ethanol fermentation to provide a zero waste solution for the
dairy industry. Journal of Dairy Science, 99(3), 1859–1867. https://doi.org/10.3168/jds.
2015-10059
Pazur, J. H. (1953). The enzymatic conversion of lactose into galactosyl oligosaccharides. Science,
117(3040), 355–356. https://doi.org/10.1126/science.117.3040.355
Pivarnik, L. F., & Senecal, A. G. (1995). Hydrolytic and transgalactosylic activities of commercial
beta-galactosidase (lactase) in food processing. Advances in Food and Nutrition Research, 38,
1–102. https://doi.org/10.1016/s1043-4526(08)60083-2
Popovic, M. K., Jamrath, T., & Linder, C. (2009). Production of thermostable alpha amylase using
food industry wastes. New Biotechnology, 5(1), 206–210. https://doi.org/10.1016/j.nbt.2009.
06.147
Qureshi, M. A., Khare, A. K., & Pervez, A. (2015). Enzymes used in dairy industries. International
Journal of Applied Research, 1(110), 523–527. www.allresearchjournal.com
Radzicka, A., & Wolfenden, R. (1995). A proficient enzyme. Science, 267, 90–931.
Ramana Rao, M. V., & Dutta, S. M. (1978). Lactase activity of microorganisms. Folia
Microbiologica, 23(3), 210–215. https://doi.org/10.1007/BF02876581
Ramawat, K. G., & Mérillon, J.-M. (2015). Polysaccharides: Bioactivity and biotechnology.
Springer. https://doi.org/10.1007/978-3-319-16298-0
Ray, A. (2012). Application of lipase in industry. Asian Journal of Pharmacy and Technology, 2(2),
33–37.
Saini, R., Singh Saini, H., Dahiya, A., & Harnek Singh Saini, C. (2017). Amylases: Characteristics
and industrial applications. Journal of Pharmacognosy and Phytochemistry, 6(4), 1865–1871.
Şanli, T., Sezgin, E., Deveci, O., Şenel, E., & Benli, M. (2011). Effect of using transglutaminase on
physical, chemical and sensory properties of set-type yoghurt. Food Hydrocolloids, 25(6),
1477–1481. https://doi.org/10.1016/j.foodhyd.2010.09.028
Saqib, S., Akram, A., Halim, S. A., & Tassaduq, R. (2017). Sources of β-galactosidase and its
applications in food industry. 3 Biotech, 7(1), 1–7. https://doi.org/10.1007/s13205-017-0645-5
Sarmah, N., Revathi, D., Sheelu, G., Rani, K. Y., Sridhar, S., Mehtab, V., & Sumana, C. (2017).
Recent advances on sources and industrial applications of lipases. Biotechnology Progress, 34,
5–28.
5 Enzyme in Milk and Milk Products: Role and Application 163
Schmid, R. D., & Verger, R. (1998). Lipases: Interfacial enzymes with attractive applications.
Angewandte Chemie International Edition, 37(12), 1608–1633. https://doi.org/10.1002/(SICI)
1521-3773(19980703)37:12<1608::AID-ANIE1608>3.0.CO;2-V
Schomburg, I., Chang, A., Placzek, S., Söhngen, C., Rother, M., Lang, M., Munaretto, C., Ulas, S.,
Stelzer, M., Grote, A., Scheer, M., & Schomburg, D. (2013). BRENDA in 2013: Integrated
reactions, kinetic data, enzyme function data, improved disease classification: New options and
contents in BRENDA. Nucleic Acids Research, 41(D1), 764–772. https://doi.org/10.1093/nar/
gks1049
Seddigh, S., & Darabi, M. (2014). Comprehensive analysis of beta-galactosidase protein in plants
based on Arabidopsis thaliana. Turkish Journal of Biology, 38(1), 140–150. https://doi.org/10.
3906/biy-1307-14
Seitz, E. W. (1974). Industrial application of microbial lipases: A review. Journal of the American
Oil Chemists Society, 51(2), 12–16. https://doi.org/10.1007/BF02545206
Sharma, R., Chisti, Y., & Banerjee, U. C. (2001). Production, purification, characterization, and
applications of lipases. Biotechnology Advances, 19, 627–662.
Sitanggang, A. B., Drews, A., & Kraume, M. (2016). Recent advances on prebiotic lactulose
production. World Journal of Microbiology and Biotechnology, 32(9). https://doi.org/10.
1007/s11274-016-2103-7
Suchy, F. J., Brannon, P. M., Carpenter, T. O., Fernandez, J. R., & Gilsanz, V. (2010). National
Institutes of Health Consensus development conference: Lactose intolerance and health. Annals
of Internal Medicine, 152(12), 792–796.
Tafes, A. G. (2019). The application of novel inputs and advanced technology in dairy product
processing a review. Journal of Dairy and Veterniary Sciences, 14(2). https://doi.org/10.19080/
JDVS.2019.14.555882
Takata, H., Kuriki, T., Okada, S., Takesada, Y., Iizuka, M., Minamiura, N., & Imanaka, T. (1992).
Action of neopullulanase. Neopullulanase catalyzes both hydrolysis and transglycosylation at
α-(1!4)- and α-(1!6)-glucosidic linkages. Journal of Biological Chemistry, 267(26),
18447–18452.
Torres, J. K. F., Stephani, R., Tavares, G. M., de Carvalho, A. F., Costa, R. G. B., de Almeida,
C. E. R., Almeida, M. R., de Oliveira, L. F. C., Schuck, P., & Perrone, Í. T. (2017). Technologi-
cal aspects of lactose-hydrolyzed milk powder. Food Research International, 101(February),
45–53. https://doi.org/10.1016/j.foodres.2017.08.043
Trespalacios, P., & Pla, R. (2007). Simultaneous application of transglutaminase and high pressure
to improve functional properties of chicken meat gels. Food Chemistry, 100(1), 264–272.
https://doi.org/10.1016/j.foodchem.2005.09.058
Troise, A. D., Bandini, E., De Donno, R., Meijer, G., Trezzi, M., & Fogliano, V. (2016). The quality
of low lactose milk is affected by the side proteolytic activity of the lactase used in the
production process. Food Research International, 89, 514–525. https://doi.org/10.1016/j.
foodres.2016.08.021
Vaidya, S., Srivastava, P., Rathore, P., & Pandey, A. (2015). Amylases: A prospective enzyme in
the field of biotechnology. Journal of Applied Biosciences, 41(1), 1–18.
van Scheppingen, W. B., van Hilten, P. H., Vijverberg, M. P., & Duchateau, A. L. L. (2017).
Selective and sensitive determination of lactose in low-lactose dairy products with
HPAEC-PAD. Journal of Chromatography B: Analytical Technologies in the Biomedical and
Life Sciences, 1060, 395–399. https://doi.org/10.1016/j.jchromb.2017.06.024
Vandenplas, Y. (2015). Lactose intolerance. Asia Pacific Journal of Clinical Nutrition, 24-
(December), S9–S13. https://doi.org/10.6133/apjcn.2015.24.s1.02
Verma, M. L., & Kanwar, S. S. (2008). Properties and application of poly(methacryclic acid-co-
dodecyl methacrylate-cl-N,N-methylene bisacrylamide) hydrogel immobilized Bacillus cereus
MTCC 8372 lipase for the synthesis of geranyl acetate. Journal of Applied Polymer Science,
110(2), 837–846. https://doi.org/10.1002/app.28539
Verma, N., Thakur, S., & Bhatt, A. K. (2012). Microbial lipases: Industrial applications and
properties (a review). International Research Journal of Biological Sciences, 1(8), 88–92.
164 A. Agarwal et al.
Whitcomb, D. C., & Lowe, M. E. (2007). Human pancreatic digestive enzymes. Digestive Diseases
and Sciences, 52(1), 1–17. https://doi.org/10.1007/s10620-006-9589-z
Whitehurst, R. J., & Van Oort, M. (2009). Enzymes in food technology (2nd ed.). Wiley. https://doi.
org/10.1002/9781444309935
Xavier, J. R., Ramana, K. V., & Sharma, R. K. (2018). β-galactosidase: Biotechnological
applications in food processing. Journal of Food Biochemistry, 42(5). https://doi.org/10.1111/
jfbc.12564
Yasueda, H., Kumazawa, Y., & Motoki, M. (1994). Purification and characterization of a tissue-
type transglutaminase from Red Sea Bream (Pagrus major). Bioscience, Biotechnology, and
Biochemistry, 58(11), 2041–2045. https://doi.org/10.1271/bbb.58.2041
Yu, Y. J., Wu, S. C., Chan, H. H., Chen, Y. C., Chen, Z. Y., & Yang, M. T. (2008). Overproduction
of soluble recombinant transglutaminase from Streptomyces netropsis in Escherichia coli.
Applied Microbiology and Biotechnology, 81(3), 523–532. https://doi.org/10.1007/s00253-
008-1688-7
Zhou, Q. Z. K., & Chen, X. D. (2001). Effects of temperature and pH on the catalytic activity of the
immobilized β-galactosidase from Kluyveromyces lactis. Biochemical Engineering Journal,
9(1), 33–40. https://doi.org/10.1016/S1369-703X(01)00118-8
Enzymes in Brewing and Wine Industries
6
S. Pati and D. P. Samantaray
Abstract
Enzymes are the bio-catalysts used in breweries and wineries for the biochemical
transformation of a substrate to produce finished products. Several enzymes such
as amylase, glucanase, protease, glycosidase, pectinase, phytase and many more
exogenous enzymes are used in different stages of brewing to improve the
production level as well as the quality of beer. In contrast, lipase, lysozyme,
pectinase, glucanase, glycosidase, urease, protease, phenoloxidase and ester
hydrolase and synthetase are employed in oenology practices for the production
of wine with enormous organoleptic properties. This chapter postulates a bird’s
eye view of different key enzymes used in breweries and wineries, their mecha-
nism of action and their pros and cons.
Keywords
Bio-catalysts · Brewery · Winery · Exogenous · Oenology · Organoleptic
6.1 Background
Beer and wine have concealed a fascinating part of history and our social life.
Although the discovery and development of brewing and wine processing signify
a cornerstone achievement of humankind, its origin is still mysterious. In ancient
civilization, people used to preserve fruits and grains in wooden containers for an
extended period to produce beer and wine. The entire process of production is
termed as fermentation, which came from theLatin word ‘fervere’, meaning ‘to
# The Author(s), under exclusive license to Springer Nature Singapore Pte 165
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_6
166 S. Pati and D. P. Samantaray
boil’. The crushed fruits and grains in the wooden container produced bubbles due to
microbial action or activities as if they were boiling. During that period, people
didn’t fully understand that a tiny, eukaryotic fungus was making the entire recipe
work (Alba-Lois & Segal-Kischinevzky, 2010). Researchers took ten decades to find
out how a diminutive microbe makes the entire fermentation successful and resulted
in a potation that became a status symbol in the modern era. Nevertheless, the
concept of brewing and wine processing was established prior to thousands of
years, which was found in the graves and settlements of early civilizations. Ancient
Egyptians made wine 8000 years ago from fruits and labelled it as such by pouring it
in fruit-shaped flasks, while beer came to the limelight 7000 years ago from China.
Initially, Germany, the United Kingdom, the United States, Belgium, Spain and Italy
were the leading countries for beer and wine production. However, in the last five
decades, beer and wine production has confronted significant changes as per the
demand. Global beer production reached 1940 mhl in 2018, with China being the
largest producer as well as a consumer (https://www.statista.com/statistics/270275/
worldwide-beer-production). In contrast, as per the data of the International Organi-
zation of Vine and Wine 2018, the global wine production has reached 292.3 mhl,
with 246 mhl consumption, where, Italy, France and Spain are the highest producers.
Enzymes are the bio-macromolecules that act as catalysts in specific reactions and
play a vital role in food science and fermentation technology. Since 6000 BC,
enzymes have been recognized as a biochemical substance to be utilized in the
processing of various foods, such as cheese preparation, brewing, wine preparation
and meat tenderizing to enhance the nutritional, sensorial and functional values of
the finished products (Gomaa, 2018; Gurung et al., 2013). Amongst all, enzymes
offer a heroic performance in brewing and winery by accelerating the release of
digestive sugars and other nutrients used by the yeasts during fermentation (Spier
et al., 2016). Brewing refers to the oldest fermentation process where complex starch
is bio-transformed to ethanol by the action of yeasts. This traditional process
involves a series of complex endogenous and exogenous enzymes that regulate the
malting of grains, the mashing of grist and rate of fermentation to produce
low-calorie beer with admirable flavour, aroma and texture (Oliver, 2011; Bamforth,
2009). While grapes bio-transformed to wine by yeasts under fermentation. This
process is catalysed by a wide array of enzymes that not only convert complex sugar
to ethanol but also release several volatile and non-volatile substances to enhance the
quality and stability of the wine (Ottone et al., 2020). Traditionally, these enzymes of
brewing and winery were produced naturally by yeast or present in the grains or
grapes. However, recent trends in fermentation technology pave the way for com-
mercial production of enzymes to enhance the quality and productivity of wine and
beer (Claus & Mojsov, 2018). Hence, this chapter offers a comprehensive summary
on the sequential processing of brewing and winery using different enzymes as well
as their mechanism of action. Moreover, the effect of enzymes on the quality and
quantity of the final product and pros and cons have been enlightened.
6 Enzymes in Brewing and Wine Industries 167
The process of brewing evolved centuries ago as a result of the resourcefulness and
artistry of the brewers. Traditionally, lager type is the most common method of
brewing, comprising low-temperature fermentation of barley and pure water with an
extended maturation period. However, variations have developed in the young beer
style, where brewing is not possible without the application of enzymes that
hydrolyse complex starch to simple fermentable sugar and then convert it to ethanol
and CO2 by yeast or bacteria. In brewing, the keystones, malted grains and barley are
the sources of enzymes, including α- and β-amylase, exo-peptidase, carboxy-pepti-
dase, proteases glucanases and cellulases (Sammartino, 2015). The detailed key role
or mode of action of these enzymes is discussed below.
6.2.1.1 Malting
Malting is the initial and prime step of brewing, which stimulates complex carbohy-
drate and protein-reducing enzymes present in the barley grains. The most prominent
endogenous enzymes of malt include α-glucosidase, α-amylase, β-amylase, car-
boxypeptidase, dextrinase, lipoxygenase, xylanase and glucanase. Besides these,
peroxidase, acid or alkali phosphatases, catalase, polyphenol-oxidases and phytase
enzymes are also responsible for malting reactions (Spier et al., 2016; Van Oort,
2010). The malting process comprises steeping, germination and drying. In steeping,
barley grains are immersed in water for 48–60 h under oxic conditions that initiate
germination and biosynthesis of amylase, glucanase, proteases and carboxypepti-
dase. Amylase acts on the modification of complex starch, whereas β-glucanase
hydrolyses β-glucans for malting clarification and proteases act on the complex
protein content of the grain (Van Oort, 2010). Then malt is subjected to drying
with a temperature between 90 and 140 C to enhance the flavour and colour of the
finished product (Curtis, 2013). The challenge during this step is to maintain the
quality of beer as the application of heat can adversely affect the functionality of
enzymes.
168 S. Pati and D. P. Samantaray
6.2.1.2 Mashing
Mashing begins with boiling of malt with water, malt adjuncts and hops at different
temperatures such as 50, 62, 72 and 78 C for proteolysis, gelatinization, saccharifi-
cation and inactivation of malting enzymes, respectively. Mashing embraces α- and
β-amylases and protease, which degrade complex starch to maltose and dextrins, and
the undesirable barley endosperm complex proteins to simple proteins, respectively
(glutelin and hordein) (Dhillon et al., 2016; Spier et al., 2016). Additionally, exoge-
nous enzymes, glucoamylases and pullulanases are added during mashing that break
down the α-1,4 and α-1,6 linkages of starch to produce maltose, dextrin, maltotriose
and glucose (Briess, 2013).
6 Enzymes in Brewing and Wine Industries 169
6.2.1.3 Pitching
Pitching refers to the addition of different strains of Saccharomyces such as
S. cerevisiae, S. uvarum and S. carlsbergensis to the fermenter as inoculum. These
strains own the MEL gene that synthesize α-galactosidases to cleave oligosaccharide
melibiose to glucose and galactose. However, research is still on its way for geneti-
cally modified yeasts that can synthesize glucosidase, amylase and glucanase for
metabolization of wort and soluble proteins (Stewart et al., 2013; Van Oort, 2010).
6.2.1.4 Aging
The beer obtained after completion of fermentation is called green beer due to its
harsh taste and which can be removed by a process called aging. In aging, green beer
is collected from a fermentation vat and stored in refrigerated conditions from a few
weeks to several months. This process enhances the flavour of beer and precipitates
yeasts, proteins and resins. During this period, diacetyl reductase is secreted by
yeast, which reduces diacetyl to acetoin to develop an undesirable butterscotch
flavour in beer. Thus, exogenous enzyme acetolactate decarboxylases is supplied
that directly converts acetolactate to acetoin thereby reducing the undesirable flavour
of beer during aging (Spier et al., 2016). In addition, several other enzymes actively
involved in brewing are presented in Table 6.1.
6.2.2.1 Amylase
During brewing, both α- and β-amylases are involved in the biotransformation of
complex starch to simple sugars such as dextrin, maltose, glucose and oligosaccha-
ride (Bamforth, 2017). Endogenous α- and β-amylases are released from the barley,
when the outer membrane of the granules is hydrolysed by xylanases and
glucanases. During malting and mashing, amylase degrades complex starch, thereby
increasing the availability of fermentable sugar. Amylase (α) strikes the internal
α-(1–4) glycosidic linkage of α-glucose amylose and amylopectin to produce dex-
trin, whereas β-amylase cleaves the external α-(1–4) glycosidic bonds of amylose
170 S. Pati and D. P. Samantaray
6.2.2.2 Protease
Proteases have many reimbursements in brewing like digestion of peptide bonds of
proteins to enhance clarification of malting, augmentation of protein solubility,
lowering of beer viscosity and favour rapid yeast growth. Temperature labile
protease has a key role in the malthouse, functions optimally at 52 C and can be
deactivated at 70–75 C, however, it can function at a wide range of pH. Though
6 Enzymes in Brewing and Wine Industries 171
6.2.2.3 b-Glucanases
It is the key enzyme of malting and mashing involved in the digestion of the outer
membrane of starch that allows other hydrolysing enzymes to act on starch granules.
This enzyme not only lowers the viscosity of beer but also helps protease activity in
the hydrolysis of a matrix of starch granules to make the kernel soften during
germination. Though, β-glucanase are present naturally in barley, in the case of
light beer it is supplied externally (0.3–1 kg per ton of wort) for improvement of light
quality and texture of the beer (Bamforth, 2017; Lyven, 2016). β-Glucanase
hydrolyses the cell wall at an optimum pH of 6 and temperature of 45–50 C,
however, this extremely heat-sensitive enzyme denatures at 60 C (Bamforth,
2017). Thus, heat-stable microbial β-glucanases of Bacillus subtilis, Aspergillus,
Penicillium and Trichoderma are used exogenously in the mashing stage of brewing.
One of the most momentous advancements in brewing is to study the use and effect
of different enzymes for beer production in order to fulfil the demand of consumers
172 S. Pati and D. P. Samantaray
for a beer with an appealing taste and flavour. Therefore, it is the need of the hour to
know the effect of enzymes on the quality of beer. Malting is the sole source of
endogenous enzymes like α- and β-amylases that are actively involved in the
conversion of complex starch to simple fermentable sugar. These enzymes are also
responsible for increasing the fermentability by boosting the saccharification, higher
product yield and enhancing of thermal stability of mash during brewing, which
ultimately removes the bitter taste from beer. The poor activity of β-glucanases can
lead to deprived runoff, recovery, spent-grain drainage, filter performance and
sedimentation in beer. However, surpassing β-glucanases activity makes the mash
thermostable and boosts the fine ability and filterability of beer (Aehle, 2007).
Malthouse enzyme protease provides foam stability in brewing. Moreover, the
shelf life improvement, maturation and flavour of beer are also greatly affected by
enzymatic activity. These enzymes ensure protein solubilization during mashing,
together with sugar availability to yeast during fermentation. Enzymes like amylase
also decrease the fermentation period during pilot-scale brewing. Furthermore,
enzymes in combination with substrate and temperature affect the carbonation,
aroma and flavour of the beer. Many commercial enzymes are also cast-off to
boost the quality attributes (clarification, texture, colour and flavour) of beer. The
enzymes utilized in brewing are assorted in their action and properties and are key
factors for the improvement of each step of brewing. Hence, intensive research is
highly essential regarding the mode of action and properties of these brewing
enzymes.
Biotechnological advances facilitate the combined use of fruit juice, yeasts and
enzymes to make a surface for wine preparation. Wine is the partial or complete
alcoholic fermentation of grape juice by yeasts where sugars are bio-transformed to
ethanol and other metabolites. These metabolites, including volatile and non-volatile
composites add significant flavour, colour, odour, taste and aroma to wine. Enzymes
are either naturally present in the grapes or supplied exogenously to catalyse
fermentation process as well as enhance the sensory properties of the wine. In this
high-tech era of biotech, endogenous and exogenous enzymes are involved at
different stages of wine preparation for smooth management of pre and post-
fermentation conditions. Several enzymes such as pectinases, glucanases,
glycosidases, xylanases, glucose oxidases, proteases, ureases, peroxidases and
proteases are actively involved in the catalyzation of different reactions occurring
during wine preparation. The role of these enzymes at various stages are momentar-
ily discussed below.
6 Enzymes in Brewing and Wine Industries 173
Wine production begins with the collection of matured grapes followed by crushing
for extraction of juice. Then, the juice is subjected to maceration for must formation,
where enzymes start functioning as a part of pre-treatment. The must undergo
fermentation by yeasts to obtain raw wine with a precise aroma due to the involve-
ment of specific enzymes. In clarification, pectic enzymes are implied to reduce the
viscosity and turbidity of the wine. Finally, suitable enzymes are used in the ageing
process to get good quality wine, where pertinent physicochemical properties are
incorporated into the wine. The implementation of various enzymes in different steps
of wine preparation is presented in Fig. 6.2.
6.3.1.1 Maceration
Desired enzymes of maceration not only expedite extraction of colour, aroma and
antioxidant activity but also regulate ethanol concentration of the wine (Ottone et al.,
2020). The endogenous cellulases and hemicellulases catalyse the lysis of grape
cells, whereas pectinases lead to the degradation of polysaccharide to accelerate the
extraction of juice. These enzymes cause activation, release and solubilization of
derivatives of antioxidant activity and colour and fragrance precursors (Garg et al.,
2016). Besides this, the addition of glucose oxidase to the must regulate the ethanol
concentration of the wine, as it maintains the concentration of glucose in the must by
using it as an electron acceptor. However, the by-product H2O2 released during
maceration oxidized the phenolic components of the wine, resulting in reduced
antioxidant activity. Thus, exogenous catalase enzyme is added to the must for
conversion of toxic H2O2 to non-toxic H2O.
6.3.1.2 Fermentation
The overall flavour and aroma of the wine are determined by the concentration of
volatile components synthesized during bioprocess technology. Wine can be
prepared with four types of aromas such as varietal, pre-fermentative, fermentative
and post-fermentative. Production of varietal aroma is due to the partial metabolism
of grapes depending on the variety, ripening percentage, soil and climate, while pre
and post-fermentative aromas are developed during maceration and ageing
(Samoticha et al., 2017). Moreover, the fermentative aroma is generated by acids,
esters, alcohols and sulfur and carbonylated substrates produced by yeasts during
fermentation. Oenology is incomplete without the involvement of yeasts like Sac-
charomyces cerevisiae and Saccharomyces carlsbergensis. These yeasts release
certain enzymes that enhance the rate of fermentation and product yield and add
flavour and aroma to the wine. Invertase is one of the most vital enzymes that causes
hydrolysis of saccharose to fructose and glucose (Ribéreau-Gayon et al., 2006),
whereas β-1,3-glucanases catalyses the synthesis of mannoprotein in fermentation
medium and cell wall hydrolysis. Pectinases and β-glucosidase also found in yeasts
catalyse the degradation of pectic components and improve the aroma of the wine
respectively (Merín et al., 2014; Villena et al., 2007). Except for these endogenous
enzymes, exogenous β-glucanases are also implemented during fermentation for
better stability and structure of the wine (Spagna et al., 2002).
highly indispensable for clarification, they are not synthesized naturally during wine
preparation. Thus, commercially synthesized pectinase from microbes and plants is
employed during bioprocess technology. On the other hand, protease is added to the
medium to circumvent haze formation. Sometimes the reaction medium gets
contaminated by Lactobacillus sp. and Pediococcus sp. causing acidity, mousy
taint and buttery flavour in the wine (Bartowsky, 2009; Liburdi et al., 2014). Thus,
lysozyme is supplemented, which not only inhibits the growth of these contaminants
but also enhances malolactic fermentation (Ottone et al., 2020).
6.3.1.4 Ageing
Ageing potentially improves the quality of the wine, where it is aged for a significant
time period till the availability of oxygen for the development of premium flavour,
aroma and taste. However, throughout ageing the naturally occurring urea and
ethanol react to form ethyl carbamate, which has carcinogenic activity. Therefore,
acid urease is added in this period for removal of urea that effectively minimizes
ethyl carbamate formation to reduce the toxicity in the wine.
Over the past decades enzymes have been established as a processing aid in
oenology. The enzymes offer several advantageous bids during pre-fermentation,
fermentation, clarification, stabilization and ageing steps that ultimately enhance the
juice yield and add colour, odour and flavour to the wine, leading to the generation of
extremely fine-quality wine. Pectinase, lipase, lysozyme, glucanases, glycosidases,
ester hydrolases and synthetases, phenol oxidases and urease are the potential
enzymes widely used in wine preparation (Table 6.2). The above-said enzymes are
naturally synthesized during oenological practice; however, in some cases, they are
also supplied externally.
6.3.2.1 Pectinase
The grape cell comprises cellulose, hemicellulose, mannan, pectin and xyloglucan
linked with proteins. During wine processing, the highly viscous pectin impedes
juice extraction, filtration, clarification and diffusion of aromatic components into
the must (Claus & Mojsov, 2018). The enzyme pectinase hydrolyses pectin to
enhance the juice yield and ease clarification and filtration. Pectinase treatment
boosts the absorbance of phenols and anthocyanins by the must, as well as intensifies
the colour and clarity of the wine (Mojsov et al., 2011). Polygalacturonase, pectin
lyase, pectinesterase and acetylesterase are the pectinase enzymes actively involved
in wine preparation. However, pectinase is neither present naturally in grapes nor
secreted by the yeasts. Therefore, commercial pectinase solutions composed of
2–5% of active enzymes and additives (preservatives, sugar and salt) are used in
wine preparation (Mojsov, 2013). The pectinase available in the market is generally
of fungal origin or produced by Rhodotorula mucilaginosa and Cystofilobasidium
capitatum. Nevertheless, high amounts of tannin, alcohol (above 17%) and SO2
(above 500 mg/l) render the pectinase activity (Van Rensburg & Pretorius, 2000).
6.3.2.2 Lysozyme
Traditionally, SO2 was added to the medium to prevent microbial contamination in
the wine, but an allergic response caused by sulphites pushes the oenologist for an
enzymatic solution (König & Fröhlich, 2017; Campos et al., 2016). Thus, lysozyme
is used to prevent microbial contamination, which kills the bacteria by cell lysis.
Lysozyme stabilizes the wine by preventing malolactic fermentation. As per the
International Organization of Vine and Wine, hen’s egg lysozyme at a concentration
of 250–500 mg/l can be used in wine preparation. Moreover, after completion of
bioprocess technology, this enzyme can be removed from the wine by the imple-
mentation of fining agents.
6.3.2.3 Glycosidase
Almost 90% of aroma precursors such as phenolic compounds, nerol, terpenes
linalool and geraniol are present in grapes’ skin in conjugated form as odourless
compounds. Glycosidase hydrolyses these precursors to liberate volatile, aromatic
terpenes that ultimately activate the organoleptic properties of the wine. These
enzymes are naturally found in grapes and promote the liberation of aromatic
compounds under optimized condition. However, these grape glycosidases are
inactivated at high alcohol and glucose concentrations and at pH 5. Therefore,
commercially available glycosidases extracted from the species of Saccharomyces,
Pichia, Candida and Rhodotorula are also employed in wine preparation (Claus &
Mojsov, 2018; Ugliano, 2009).
6 Enzymes in Brewing and Wine Industries 177
6.3.2.4 Glucanase
Lactic acid bacteria and some fungus associated with the skin of grapes release
viscous polysaccharides, which hamper the wine filtration. Neither flocculants nor
filtration can remove these polysaccharides; thus, glucanase is used for reduction of
wine viscosity. Both endo andexo-glucanase release mannoproteins to enhance the
varietal flavour in the wine. Endo-glucanase is naturally synthesized by Saccharo-
myces species during fermentation, while exo-glucanase is supplemented to the
fermentation medium. Moreover, the commercially available exo-glucanase is
extracted from fungus like Taleromyces versatilis and Trichoderma sp., different
species of yeast like Kloeckera, Zygosaccharomyces and Pichia and lactic acid
bacteria (Claus & Mojsov, 2018).
6.3.2.5 Protease
Proteins present in must or synthesized by starter culture cause allergic reactions in
the consumer (Van Sluyter et al., 2015; Rizzi et al., 2016). Though proteins are
precipitated after fermentation, the presence of acid, proteolytic and heat-resistant
pathogenesis-related (PR) proteins causes undesirable turbidity in the wine. Gener-
ally, bentonite is used for the removal of PR proteins, but it can adversely affect the
quality and quantity of the wine (Jaeckels et al., 2015). Thus, researchers are now
focusing on protease as an attractive enzymatic solution for the removal of these
undesirable proteins. Protease can not only enhance the quality and quantity of the
wine but also reduce the haziness in the wine. Saccharomyces species doesn’t show
any protease activity; however, some fungus and non-Saccharomyces species depict
the same. So, nowadays researchers are shifting their attention to selecting or
developing protease positive starter culture for wine production.
6.3.2.6 Urease
In wine, fermentation yeast generates urea that is chemically converted to carcino-
genic ethyl carbamate (Lonvaud-Funel, 2016). In 1997, urease was established as an
enzymatic solution that cleaves urea into CO2 and NH3 and prevents ethyl carbamate
synthesis. Generally, urease of Lactobacillus origin is used at a concentration of
25–50 mg/l in the fermentation medium (Pozo-Bayón et al., 2012).
6.3.2.7 Lipase
Lipids are released during wine fermentation as a result of the autolysis of yeast or
grape cells, which cause significant changes in fermentation as well as in the finished
product. Hence, lipase is employed in wine preparation to decompose the lipids
present in the cell membrane for the improvement of the colour and texture of the
wine. It is found in a few wild strains of Lactobacillus and yeasts.
6.3.2.9 Phenoloxidases
Phenoloxidases affect not only the sensory attribute of the wine but also the final
phenol concentration of the wine. However, these oxygen-sensitive enzymes on
exposure to O2 cause enzymatic browning of wine. Phenoloxidases include tyrosi-
nase, responsible for implementation of colour to the wine, and laccase, which
causes phenol oxidation to enhance the organoleptic properties of the wine (Claus
& Mojsov, 2018).
Enzymes have a very controversial effect on wine preparation as they are highly
selective. Several studies have been directed to know the effect of different enzymes
on the organoleptic as well as quantitative properties of the wine. The colour
extraction of the wine is greatly influenced by xylanase and glycosidase activities.
During maceration, the enzymatic treatment enhances the extraction of phenolic
compounds while depicting undesirable anthocyanin content and colour parameters.
Similarly, glycosidase helps in aroma extraction in the wine; however, an abundance
of glucose inhibits its action. Lysozyme causes total inhibition of lactic acid bacteria
from wine, but the presence of the residual amount of lysozyme can cause an allergic
reaction in the consumer (Liburdi et al., 2014). Furthermore, protease is essential to
remove PR proteins from wine, but it is only functional under restricted alcohol
concentration, pH and temperature (Espejo, 2020). It can’t be ignored that enzymes
are highly essential for wine preparation, but the sensitivity of these enzymes
influences industry and academia to analyse the pros and cons before further
application.
Before taking any step forward towards the enzymology of brewing and wine
preparation, it is highly recommended to study their pros and cons. The application
of enzymes in brewing enhances the maturation of beer, catalyses low-calorie beer
production, stabilizes the beer by improving mashing and clarification and reduces
the viscosity, leading to fine-quality beer production (Gomaa, 2018). Nevertheless,
the cost is the prime barrier in the enzymatic treatment of beer. Besides, the
sensitivity of enzymes towards temperature, pH, alcohol and glucose concentration
also significantly affects the use of enzymes in brewing. Like in brewing, enzymes
also have a significant effect towards the enhancement of the flavour, aroma and
taste of the wine. Some enzymes are used to prevent contamination of the wine by
toxic microbes and compounds. Enzymes also ease the wine processing for higher
yield and lower production cost, leading to a booming profit. On the other hand, it is
always advisable to consider the drawbacks of these enzymes for a better future.
Sometimes specific activity and side effects of a few enzymes have a detrimental
effect on wine quality. Also, the use of commercial enzymes requires a lot of
6 Enzymes in Brewing and Wine Industries 179
6.5 Conclusion
References
Aehle, W. (2007). Industrial enzymes. In Enzymes in industry (pp. 99–262). Wiley-VCH Verlag
GmbH & Co. KGaA.
Alba-Lois, L., & Segal-Kischinevzky, C. (2010). Yeast fermentation and the making of beer and
wine. Nature Education, 3(9), 17.
Bamforth, C. W. (2009). Current perspectives on the role of enzymes in brewing. Journal of Cereal
Science, 50, 353–357.
Bamforth, C. W. (2017). Progress in brewing science and beer production. Annual Review of
Chemical and Biomolecular Engineering, 8, 161–176.
Bartowsky, E. J. (2009). Bacterial spoilage of wine and approaches to minimize it. Letters in
Applied Microbiology, 48(2), 149–156.
Boulton, C. (2013). Encyclopedia of brewing (Vol. 716). Wiley-Blackwell.
Briess. (2013). In E. Richard (Ed.), Mashing for optimal yield. Division Manager Brew East, Briess
Malt and Ingredients Company.
180 S. Pati and D. P. Samantaray
Campos, F. M., Couto, J. A., & Hogg, T. (2016). Utilization of natural and by-products to improve
wine safety. In M. V. Morena-Arribas & B. Bartolomé Sualdea (Eds.), Wine safety, consumer
preference and human health (pp. 27–49). Springer International Publishing.
Claus, H., & Mojsov, K. (2018). Enzymes for wine fermentation: Current and perspective
applications. Fermentation, 4(52), 1–19.
Curtis, T. (2013). A London encyclopedia or universal dictionary of science, art, literature and
practical mechanics (Vol. 13, pp. 484–485). Forgotten Books.
Dhillon, G. S., Kaur, S., Oberoi, H. S., Spier, M. R., & Brar, S. K. (2016). Agricultural-based
protein byproducts: Characterization and applications. In Protein by-products: Transformation
from environmental burden into value-added products. Elsevier.
Dulieu, C., Moll, M., Boudrant, J., & Poncelet, D. (2000). Improved performances and control of
beer fermentation using encapsulated alpha-acetolactate decarboxylase and modeling. Biotech-
nology Progress, 16, 958–965.
Espejo, F. (2020). Role of commercial enzymes in wine production: A critical review of recent
research. Journal of Food Science and Technology. https://doi.org/10.1007/s13197-020-
04489-0
Garg, G., Singh, A., Kaur, A., Singh, R., Kaur, J., & Mahajan, R. (2016). Microbial pectinases: An
ecofriendly tool of nature for industries. 3 Biotech, 6(1), 47.
Gomaa, A. M. (2018). Application of enzymes in brewing. Journal of Nutrition and Food Science
Forecast, 1(1), 1–5.
Gurung, N., Ray, S., Bose, S., & Rai, V. (2013). A broader view: Microbial enzymes and their
relevance in industries, medicine and beyond. BioMed Research International, 2013, 1–18.
Hans, S. O. (2008). Enzymes in brewing. Biokemisk Forening.
Jaeckels, N., Tenzer, S., Rosch, A., Scholten, G., Decker, H., & Fronk, P. (2015). β-Glucosidase
removal due to bentonite fining during wine making. European Food Research and Technology,
241, 253–262.
König, H., & Fröhlich, J. (2017). Lactic acid bacteria. In Biology of microorganisms on grapes, in
must and in wine. Springer International Publishing.
Liburdi, K., Benucci, I., & Esti, M. (2014). Lysozyme in wine: An overview of current and future
applications. Comprehensive Reviews in Food Science and Food Safety, 13(5), 1062–1073.
Lonvaud-Funel, A. (2016). Undesirable compounds and spoilage microorganisms in wine. In M. V.
Morena-Arribas & B. Bartolomé Sualdea (Eds.), Wine safety, consumer preference, and human
health (pp. 3–26). Springer International Publishing.
Lyven. (2016). Brewing enzymes. Retrieved from lyven.com
Martín, M. C., & Morata de Ambrosini, V. I. (2014). Effect of a cold-active pectinolytic system on
color development of Malbec red wines elaborated at low temperature. International Journal of
Food Science and Technology, 49(8), 1893–1901.
Merín, M. G., Mendoza, L. M., & Ambrosini, V. I. M. (2014). Pectinolytic yeasts from viticultural
and enological environments: Novel finding of Filobasidium capsuligenum producing
pectinases. International Journal of Basic Microbiology, 54, 835–842.
Mojsov, K. (2013). Use of enzymes in wine making: A review. International Journal of Technology
Marketing, 3, 112–127.
Mojsov, K., Ziberoski, J., & Bozinovic, Z. (2011). The effect of pectolytic enzyme treatments on
red grapes mash of Vranec on grape juice yields. Perspectives of Innovations, Economics and
Business, 7, 84–86.
Oliver, G. (2011). The oxford companion to beer (1st ed.). Oxford University Press.
Ottone, C., Romero, O., Aburto, C., Illanes, A., & Wilson, L. (2020). Biocatalysis in the
winemaking industry: Challenges and opportunities for immobilized enzymes. Comprehensive
Reviews in Food Science and Food Safety, 19, 595–621.
Pozo-Bayón, M. A., Monagas, M., Bartolomé, B., & Moreno-Arribas, M. V. (2012). Wine features
related to safety and consumer health: An integrated perspective. Critical Reviews in Food
Science and Nutrition, 52, 31–54.
6 Enzymes in Brewing and Wine Industries 181
Ribéreau-Gayon, P., Dubourdieu, D., Donèche, B., & Lonvaud, A. (2006). Handbook of enology—
The microbiology of wine and vinification (Vol. 1, 2nd ed.). John Wiley & Sons, Ltd.
Rizzi, C., Mainente, F., Pasini, G., & Simonato, B. (2016). Hidden exogenous proteins in wine:
Problems, methods of detection and related legislation—A review. Czech Journal of Food
Sciences, 34, 93–104.
Sammartino, M. (2015). Enzymes in brewing. Master Brewers Association of the Americas
Technical Quarterly, 52(3), 156–164.
Samoticha, J., Wojdyło, A., Chmielewska, J., Politowicz, J., & Szumny, A. (2017). The effects of
enzymatic pre-treatment and type of yeast on chemical properties of white wine. LWT—Food
Science and Technology, 79(Supplement C), 445–453.
Spagna, G., Barbagallo, R. N., Palmeri, R., Restuccia, C., & Giudici, P. (2002). Properties of
endogenous β-glucosidase of a Saccharomyces cerevisiae strain isolated from Sicilian musts
and wines. Enzyme and Microbial Technology, 31, 1030–1035.
Spier, M. R., Nogueira, A., Alberti, A., Gomes, T. A., & Dhillon, G. S. (2016). Potential
applications of enzymes in brewery and winery. In Agro-industrial wastes as feedstock for
enzyme production (pp. 261–278). Elsevier.
Stewart, G. G., Hill, A. E., & Russell, I. (2013). 125th Anniversary review: Developments in
brewing and distilling yeast strains. Journal of the Institute of Brewing, 119, 202–220.
Ugliano, M. (2009). Enzymes in winemaking. In M. V. Moreno-Arribas & M. C. Polo (Eds.), Wine
chemistry and biochemistry (pp. 103–126). Springer Science Business Media LLC.
Van Oort, M. (2010). Enzymes in food technology—Introduction. In R. J. Whitehurst & M. Van
Oort (Eds.), Enzymes in food technology (pp. 1–17). Wiley-Blackwell.
Van Rensburg, P., & Pretorius, I. S. (2000). Enzymes in winemaking: Harnessing natural catalysts
for efficient bio-transformations: A review. South African Journal for Enology and Viticulture,
21, 52–73.
Van Sluyter, S. C., McRae, J. M., Falconer, R. J., Smith, P. A., Bacic, A., Waters, E. J., &
Marangon, M. (2015). Wine protein haze: Mechanisms of formation and advances in preven-
tion. Journal of Agricultural and Food Chemistry, 63, 4020–4030.
Villena, M. A., Iranzo, J. F. Ú., & Pérez, A. I. B. (2007). β-Glucosidase activity in wine yeasts:
Application in enology. Enzyme and Microbial Technology, 40, 420–427.
Enzymes in Meat, Fish, and Poultry
Products Processing and Preservation-I 7
Khadijeh Abhari and Hedayat Hosseini
Abstract
Enzymes are used in the food industry widespread to modify and enhance the
nutritional value, and functional and sensorial characteristics such as color, smell,
and flavor of different types of food products.
The technological applications of enzymes in the meat industry are a hot topic
nowadays. The most common application of enzymes in red meat products is to
increase the meat tenderness. The toughness of beef meat produced from dairy
cattle is related to the volume of connective tissue, mainly collagen. Proteolytic
enzymes have the potential to tenderize lower-grade meats and increase their
market value. Consumers’ demand for fresh meat without undesirable excessive
fat and salt content leads the manufacturer to consider new possibilities of
enzyme application to increase the carcass efficiency by using more effective
slaughtering techniques.
The utilization of enzymes to maximize the by-products’ efficiency is an
undeniable possibility.
The common by-products of the poultry industry are feathers, which can be
digested by enzymes and applied for animal feed and also nonfood industries
such as films, coating, and packaging due to their high amount of keratins and
hydrophobic amino acids. Fat, bone, and mechanical flesh are common
by-products of meat processing, which can be digested by enzymatic reactions
and used for their special meat properties. Enzymes also can be applied in seafood
processing for deskinning and descaling, production of caviar, and recovery of
# The Author(s), under exclusive license to Springer Nature Singapore Pte 183
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_7
184 K. Abhari and H. Hosseini
Keywords
Calpains
The calpain system, which is activated by calcium and thiol, contains three enzymes
three protease enzymes: calpain I (μ), calpain II (m), calpain 3, and calpastatin,
which is recognized as calpain inhibitor. Calpain I is linked to myofibrils of muscles,
whereas calpain II and calpain 3 are situated in cytosol and sarcomere (near the Z and
M line in myofibrillar structure), respectively.
It has been suggested that μ-calpain has an actual role on postmortem proteolysis.
Many factors effect on calpain system activity, particularly the calcium ion
7 Enzymes in Meat, Fish, and Poultry Products Processing and Preservation-I 185
concentration; thus, any operation which increases the calcium ion can improve meat
tenderness (Moudilou et al., 2010).
Studies revealed that injection of calcium or application of electrical stimulation
causes an increase in meat tenderness due to increase in calcium level and decrease
in calpastatin activity. The changes in Z line and degradation of myofibrillar proteins
such as I troponins, T troponins, desmins, titins, and nebulins are the most common
phenomenon during caplain system activity.
Plant Enzymes
Papain
Papain is a protease enzyme which contains cysteine and extracted protease from the
latex of the papaya plant (Carica papaya). This enzyme is a part of defense system of
plant and protects the plant against insect. The optimum temperature and pH for
enzyme activity is 65 C and 5–8, respectively.
Commercial forms of papain contain various ratio of papain, papaya peptidase,
and chymopapain and lead to different biochemical properties. Papain tenderizes the
toughness of meat by acting on amino acids which have aromatic side chain like
tyrosine and phenylalanine. Papain can effectively hydrolyze collagen and myofi-
brillar proteins. Investigations show that injection of papain approximately 30 min
before slaughter can cause meat tenderness. As injection of papain to animal may
186 K. Abhari and H. Hosseini
cause stress symptoms and severe shock, using inactive papain can be an alternative
approach.
Ficin
Ficin is extracted from latex of fig fruits (Ficus glabrata, Ficus anthelmintica). Figs
contain ten different proteases and ficin is most popular for its tenderizing effect.
Studies revealed that application of ficin in temperatures of 60 and 70 C has the
most influence in tenderization process. It is suggested that ficin has the ability to
effect on collagen, elastin, and myofibrillar proteins in addition to increase in water-
holding capacity.
Cucumin
Cucumin is another proteolytic enzyme which is achieved from kachri (Cucumis
pubescens). The researches show that cucumin can be effective on tenderizing
buffalo meat, sheep meat, and chicken.
Bromelain
Bromelain is cysteine proteases which originated from pineapple. It has two sources:
fruit bromelain and stem bromelain; it has been shown that fruit bromelain is more
specific and more effective in comparison to stem bromelain. The optimum temper-
ature for enzyme activity is 50–60 C; however, the studies showed that the enzyme
activity remains at 0 C and it becomes inactive in temperatures more than 80 C.
The bromelain activity starts by degradation of collagen, and then attacks myosin.
Zingibain
Zingibain is a potent proteolytic enzyme which is obtained from ginger. The studies
reported that Zingibain is more effective in meat tenderization when heated. Ginger
is a great and economical source of proteases that begin degradation from the I band
of each sarcomere and go through M line.
Actinidin
Actinidin is a natural enzyme extracted from kiwi fruit which has been recognized as
an agent which can hydrolyze both connective tissue proteins specially collagen and
myofibrillar proteins. Studies show that applying actinidin in brine solution can
tenderize porcine muscles.
Flavor has a complex definition which includes several phenomenons such as taste,
odor, trigeminal senses, and texture. As well as the mentioned factors, tenderness
significantly affects the flavor and the relationship of all these together creates
consumer acceptability. Generally, the raw meat is tasteless due to absence of
nonvolatile components; however, the flavor precursors are present in the meat
7 Enzymes in Meat, Fish, and Poultry Products Processing and Preservation-I 187
tissue and it is necessary to undergo enzymatic reaction to tastes appears in the meat
(Fernandez et al., 2000; Toldra, 1998; Calkins & Hodgen, 2007).
As the results of several enzymes activity such as lipase, protease, glutaminase,
and peptidase, biochemical reactions occur and lead to enhance the flavor. These
enzymes may be endogenous natural lipases and protease, microbial originated
enzymes or those which added to the product throughout the manufacturing proce-
dure. Due to proteases activity mainly cathepsins, trypsin-like peptidases, and
Micrococcaceae originated enzymes (Fernandez et al., 2000), nitrogenous
complexes such as volatile compounds, small peptides, and free amino acids appear.
As well as micrococci, other species of lactic acid bacteria, such as Pediococcus and
Lactobacillus, are able to produce intracellular peptidases which contribute to
increase the levels of free amino acids. These substances have principal effect on
the taste of the product.
Another group of enzymes which takes part in flavor enhancement are lipases.
Lipases produced by some microorganisms like lactobacilli or micrococci could
effect on the lipids by their lipolytic activity and oxidation, leading to increase in the
level of free fatty acids and short chain fatty acids. The substances produced by
lipases are responsible to develop the flavor in final product. Although the optimum
temperature for maximum activity of microbial lipases is 30–40 C, some thermo-
philic microorganisms such as Pseudomonas, Bacillus sp., Thermomyces
lanuginosus, and Aspergillus niger release lipases which can stand in the higher
temperatures (50–65 C).
Glutaminase also play a role in creating the flavor of sausages by deamidation of
glutamine, leading to production of L-glutamic acid and ammonia through hydrolysis
of the glutamine amide group. Glutamic acid is an amino acid which is responsible
for the flavor enhancement and producing by starter cultures has an important role in
flavor in sausage, seasonings such as soya sauce and miso, and pickles. Glutamic
acid is the substance which increases “umami” taste in food products. The ammonia,
produced by glutaminase, performs as an acid neutralizer. Glutaminase can be
obtained from Bacillus amyloliquefaciens, Aspergillus oryzae, Debaryomyces, and
Rhizobium etli.
7.2.1 Deskinning
Fish protein hydrolyzates are recognized as one of the important by-products of the
seafood industry which can be obtained by enzymatic or chemical reactions. The
wasted of fish industry such as gills, head, skin, bones, viscera and liver are the main
sources for producing fish hydrolyzates which are in the forms of peptides with
2–20 amino acids depends on the type of enzyme, kind of fish and the time of
operation.
Fish hydrolyzates also can be obtained by alkali or acid reactions in high
temperature and pressure in the traditional method. The major acids utilized in
acid hydrolysis are sulfuric acid and hydrochloric acid. One of the disadvantages
of acid and alkali hydrolysis is production of great amount of sodium chloride and
sodium hydroxide, respectively, which decreases its functionality. Moreover,
destruction of tryptophan, a critical amino acid, and production of several toxic
compounds are other concerns regarding using acid hydrolysis approach.
On the other hand, enzymatic hydrolysis has several advantages, such as being
economical, and required slight conditions of pressure, temperature, and pH. Fish
hydrolyzates demonstrate functional properties in food formulation and can be
applied as emulsifier or a substance which increase water-holding capacity, protein
solubility, gelling activity, and oil-binding capability.
Fish hydrolyzates which are mostly produced by action of alcalase are more
constructive compared to poultry by-products.
Poultry industry with various products including chicken, egg, and turkey has a large
portion in food sector and human diet. Due to side effects of red meat consumption,
nutritional benefits of white meat and ease of preparation of such products for
customers and of the poultry industry has been growing fast, as the USA, China,
and Brazil have the maximum products, worldwide.
7 Enzymes in Meat, Fish, and Poultry Products Processing and Preservation-I 189
Feathers are a great source of inexpensive protein; however, due to its low nutritional
value and low digestibility, its application as a feedstuff has various concerns.
Conventional hydrothermal method of degradation for feathers causes losing some
essential amino acids, as well as production of non-nutritious amino acids such as
lysinoalanine and lanthionine. To overcome the mentioned problems, biotechnolog-
ical methods via microorganisms have been offered. The keratinase produced by
microorganisms such as Bacillus spp. has the capacity to hydrolyze keratin and
convert it to peptides which can be used as nitrogen fertilizers or animal feed.
Controlled enzymatic hydrolysis is an appropriate method for production of bioac-
tive peptides that have the potential to play role as ant antimicrobial, antioxidant and
antihypertensive as Fontoura et al. (2014) reported DPP-IV and ACE inhibitor
activities of hydrolyzed keratin obtained by feathers of raw chicken.
Another by-product of poultry industry is fat which is significantly located in the
skin. Chicken fat contains both ω-3 and ω-6 polyunsaturated fatty acids which can be
beneficial to reduce cholesterol. Lee and Foglia (2000) have indicated that Candida
rugosa and Geotrichum candidum are able to produce lipases which act on triglyc-
eride fractions that have been evoked from chicken fat by supercritical CO2.
190 K. Abhari and H. Hosseini
Due to consumer demand for fresh tender meat with minimum fat and salt, and also
the manufactures to produce the carcasses with sufficient profits, enzymatic
techniques have been utilized to achieve these aims. One of these methods which
recently become popular in meat industry is using TGase which is a cold binding is
enzymatic technology.
In this method, the cut and timed pieces of meat from one or several animals can
be attached together with TGase to form meat products with new shape and size,
which can be stored in every temperature.
References
Ashie, I., Sorensen, T., & Nielsen, P. (2002). Effects of papain and a microbial enzyme on meat
proteins and beef tenderness. Journal of Food Science, 67(6), 2138–2142.
Calkins, C. R., & Hodgen, J. M. (2007). A fresh look at meat flavor. Meat Science, 77(1), 6380.
Fernandez, M., et al. (2000). Accelerated ripening of dry fermented sausages. Trends in Food
Science & Technology, 11(6), 201–209.
Fontoura, R., Daroit, D. J., Correa, A. P. F., Meira, S. M. M., Mosquera, M., & Brandelli, A. (2014).
Production of feather hydrolysates with antioxidant, angiotensin-I converting enzyme- and
dipeptidyl peptidase-IV-inhibitory activities. New Biotechnology, 31(5), 506–513.
Herrera-Mendez, C. H., et al. (2006). Meat ageing: Reconsideration of the current concept. Trends
in Food Science & Technology, 17(8), 394–405.
Hwang, I., Devine, C., & Hopkins, D. (2003). The biochemical and physical effects of electrical
stimulation on beef and sheep meat tenderness. Meat Science, 65(2), 677–691.
Kemp, C. M., et al. (2010). Tenderness—An enzymatic view. Meat Science, 84(2), 248–256.
Koohmaraie, M., & Geesink, G. (2006). Contribution of postmortem muscle biochemistry to the
delivery of consistent meat quality with particular focus on the calpain system. Meat Science,
74(1), 34–43.
Koohmaraie, M., et al. (1988). Factors associated with the tenderness of three bovine muscles.
Journal of Food Science, 53(2), 407–410.
Lawrie, R. (1998). Meat science (6th ed.). Woodhead Publ. Ltd.
Lee, K., & Foglia, T. A. (2000). Synthesis, purification, and characterization of structured lipids
produced from chicken fat. Journal of the American Oil Chemists’ Society, 77, 1027–1034.
Lepetit, J. (2008). Collagen contribution to meat toughness: Theoretical aspects. Meat Science,
80(4), 960–967.
Moudilou, E., et al. (2010). Calpains expression during Xenopus laevis development. Tissue and
Cell, 42(5), 275–281.
7 Enzymes in Meat, Fish, and Poultry Products Processing and Preservation-I 191
Olson, D., & Parrish, F., Jr. (1977). Relationship of myofibril fragmentation index to measures of
beefsteak tenderness. Journal of Food Science, 42(2), 506–509.
Rhodes, D. N., & Dransfield, E. (1973). Effect of pre-slaughter injections of papain on toughness in
lamb muscles induced by rapid chilling. Journal of the Science of Food and Agriculture, 24(12),
1583–1588.
Simpson, B. K., et al. (2012). Food biochemistry and food processing. John Wiley & Sons.
Toldra, F. (1998). Proteolysis and lipolysis in flavour development of dry-cured meat products.
Meat Science, 49, S101–S110.
Enzymes in Meat, Fish, and Poultry Product
Processing and Preservation-II 8
Sandesh Suresh Karkal, Anushma Venmarath,
Suresh Puthenveetil Velappan, and Tanaji G. Kudre
Abstract
The current chapter provides insights into the applications of enzymes in meat,
fish, and poultry products’ processing industries. The mechanical and chemical
techniques mainly employed to process meat, fish and poultry products are not
eco-friendly, consume more energy, and generate by-products. Hence, the substi-
tution of these techniques with enzymes (biocatalysts) could be the best alterna-
tive. Besides, the application of enzymes in these industries is gaining momentum
since they are eco-friendly, consume less energy, and also aid in obtaining good-
quality products with better yield. The enzymes can catalyze the reaction process
at mild temperature and pH and are highly specific toward substrate molecules.
Enzymes such as proteases, lipases, transglutaminases from plant, animal, and
microbial origin are suitable and have been extensively studied to process meat,
fish, and poultry products. Currently, novel enzymes are also being developed as
a result of technological advancements. The recombinant enzymes being devel-
oped through technical advancements in biotechnology for their application in
meat, fish, and poultry products processing could be an excellent prospect. This
chapter describes the potential enzymes used in the processing and preservation
of meat, fish, and poultry products.
Keywords
# The Author(s), under exclusive license to Springer Nature Singapore Pte 193
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_8
194 S. S. Karkal et al.
8.1 Introduction
Enzymes are termed as “biocatalysts” owing to their natural catalytic nature and are
considered to be prime tools in biotechnology and related areas (Fernandes, 2016).
Enzymes are mainly protein molecules known to speed up the biochemical reaction
rate occurring in the cell (Singh, 2018). Enzymes have been utilized in the
processing of food since ancient times, long before their catalytic function was
discovered. Homer’s epic poem, Iliad, written around the eighth century BC,
describes the use of enzyme ficin for milk clotting (Copeland, 2000). Apart from
this, the original inhabitants of the Pacific Islands exploited papaya juice for meat
tenderization. The colonization of these islands by the British led to the growth of
enzyme technology for food processing, especially meat tenderization and also
wound healing in European nations (Gomes et al., 2018). At present, enzymes
have found their potential application in food, leather, animal feed, biofuel, deter-
gent, etc., industries. In these industries, enzymes have been employed as efficient
catalysts (Singh, 2018). The increased demand for enzymes in industries, especially
food industries, may be attributed to its high specificity towards the substrate
molecule. Besides, enzymes can catalyze the reactions under mild pH and tempera-
ture with minimal by-product formation (Shahidi & Kamil, 2001).
Food is one of the necessities of life required for human survival. Despite being
an important necessity, the global report on the food crisis (2019 Edition) by the
European Commission illustrates that across 53 countries, a population of more than
113 million is experiencing a food shortage, nutritional deficiency, and also
improper livelihood assistance (European Commission-GRFC, 2019). Due to the
rapid population explosion, extensive utilization of natural resources, disputes across
the globe, and variation in climate, there has been an increased demand for food
worldwide (Singh, 2018). According to the United Nation Department of Economic
and Social Affairs (UNDESA), New York, the population of the world is projected
to be increased from 6.9 to 9.1 billion (i.e., 32% rise) by 2050 and this would trigger
the hike in food demand up to 70% (UNDESA, 2017; Singh, 2018). Moreover, to
mitigate food shortage, in the year 2015, the United Nations (UN) Member States
adopted the 2030 Agenda, which mainly aims for the contribution of fisheries and
aquaculture in the use of natural resources toward food security and nutrition, and
thereby ensure sustainable development in economic, social, and environmental
terms (FAO, 2016). A considerable increment in the production of meat and poultry
products was also observed to meet the global food demands. According to the Food
and Agriculture Organization of the United Nations (FAO), by 2050, the global
production of meat is expected to increase to 455 million tons (Chandrasekaran et al.,
2015).
At present, combating the current global scenario of the food crisis, elimination of
malnutrition and hunger, and ensuring food security have been the most challenging
research and development fields. Food processing could play a pivotal role in
eradicating the persisting global food shortage (Singh, 2018). Food processing refers
to the process where the agricultural raw material is processed into a consumer-ready
product. The main objectives of modern food processing include (1) to prepare safe
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 195
Animals such as sheep, goat, pig, cattle, and other livestock are slaughtered,
processed, and distributed in the meat industry (Chandrasekaran et al., 2015).
According to FAO, in the year 2018, global meat production increased up to 1.2%
compared to 2017, which could be easily interpreted in Fig. 8.1 (FAO, 2019).
Figure 8.1 depicts the production of meat worldwide from the year 2016 to 2019.
Increased production of pork and beef meat was observed in the year 2018, but there
was a slight decrease in their production in 2019. However, sheep meat production
increased from 2016 until 2019.
196 S. S. Karkal et al.
Fig. 8.1 Global meat production from the year 2016 to 2019. (Source: Statista, Production of meat
worldwide, 2020)
In the past decades, global consumption of meat has increased due to population
explosion and the economic boost in developing nations. Currently, the consumers
consider the quality attributes of meat products rather than their price, which
signifies that the greater the quality traits of meat products, the greater their impor-
tance to meet the increasing consumer demands (Gomes et al., 2018). The most
appreciable quality trait of meat judged by consumers is “tenderness”. The tender-
ness is a textural property of food associated with chewiness or cohesiveness. This
quality trait of food during mastication manifests little resistance to fragmentation
(Ashie et al., 2002; Gomes et al., 2018). The tenderness of the meat is primarily
related to the structural integrity of connective tissue proteins and myofibrillar
proteins. The key constituent of connective tissue is collagen, and the tenderness
of the meat is determined by the amount, type, as well as the degree of intermolecular
cross-linking in collagen. Studies have depicted an increased cross-linking in aged
animals rather than young animals (Ashie et al., 2002). To improve the tenderness of
the meat, the application of enzymes has been found to be desirable in terms of both
producing quality products and the cost (Cazarin et al., 2015). There are two
underlying mechanisms to obtain the desired tenderness in meat products using
enzymes. The first mechanism involves the generation of amino acids or peptide
fragments by breaking the covalent bonds of protein molecules. This would result in
meat structure alteration. The second mechanism includes the formation of new
covalent bonds, which would ultimately change the meat tenderness. The enzyme
mainly involved in the new bond formation is transglutaminases (Cazarin et al.,
2015). Both the endogenous enzymes (calpains, cathepsins, capsases, and
proteosomes) and exogenous enzymes from the plant, animal, and microbial sources
(ficin, bromelain, papain, actinidin, cucumin, zinzibain, collagenase, elastase, animal
protease, and fungal protease) have been found to play a pivotal role in the tenderi-
zation of meat (Ashie et al., 2002; Cazarin et al., 2015; Gomes et al., 2018). Apart
from the application of enzymes in meat tenderization, other techniques involve
shock wave pressure, calcium chloride injection, mechanical tenderization, electrical
stimulation, and elevated temperature storage (Ashie et al., 2002).
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 197
Over the decades, a tremendous leap has been observed in fish production across the
globe. The global fish production in the year 2018 was 178.8 million metric tons, and
in the year 2019, it is projected to be 177.9 million metric tons (Statista, 2019;
FAO-GLOBEFISH, 2020). Figure 8.2 displays the total global fish production from
the year 2016 to 2019. The total global fish production includes aquaculture produc-
tion, which includes the production of even crustaceans and mollusks (seafood). In
the majority of the countries, fresh fish and seafood harvested are mainly sold for
local consumption (Suresh et al., 2015). From the total fish and seafood processed,
about 20–50% is available for edible purposes, and the rest of it is generated as
non-edible by-products or discarded as waste. These by-products are rich in various
biomolecules such as oil and lipids, proteins, bioactive peptides, pigments,
polymers, vitamins, and minerals (Suresh et al., 2015).
The utilization of enzymes in the processing of fish and seafood has gained
momentum due to the advancements in enzyme technology. Enzymes have been
employed in fish-processing industries to obtain a diversified range and improved
quality products. In fish-processing industries, enzymes have been used for
descaling, deskinning, salted fish ripening, production of products such as surimi,
fish sauce, and fish protein hydrolyzates, extraction of oil and biomolecules such as
chitin, carotenoproteins, and flavor compounds, and removal of exoskeleton from
shellfish (Suresh et al., 2015). The enzymes used for fish and seafood processing are
mainly derived from microorganisms, plants, and animals (Fernandes, 2016; Suresh
et al., 2015). Moreover, research has also been carried out on the enzymes isolated
from fish discards and its application in the processing of fish and seafood (Suresh
et al., 2015). This approach would help in waste management of fish and seafood
by-products, the majority of which are mainly dumped into the environment. The
enzymes—both natural and recombinants—derived from microorganisms have been
used in fish and seafood processing. Further, the demand for microbial enzymes is
more than that for enzymes isolated from plants and animals since the microbial
enzymes exhibit better catalytic activity and high yield, and can be genetically
176
174
172
170
168
166
2016 2017 2018 2019 (P)
Year
198 S. S. Karkal et al.
manipulated with ease, regular supply, and rapid microbial growth in low-priced
culture media (Suresh et al., 2015). Besides fish and seafood processing, enzymes
have also been used in the analysis of processed aquaculture and fish products
(Fernandes, 2016).
Similar to meat and fish production worldwide, poultry production has also
increased over the decades. Figure 8.3 displays the global poultry production from
the year 2016 to 2019. From the figure, it is evident that the global poultry
production is increasing every year and is higher than global sheep, pork and beef,
and veal production. Global poultry production has been soaring to meet the global
consumption demands due to overpopulation.
Besides the tenderization of pork, sheep, and beef meat, enzymes have also been
used for the tenderization of poultry products. Studies have been carried out on
improving the tenderness of poultry meat using exogenous enzymes such as papain,
bromelain, proteases, etc., and also endogenous enzymes such as calpains (Bawa
et al., 1981; Kang & Warner, 1974; Whipple et al. 1990).
As described in the previous sections, the enzymes used in meat, fish, and seafood as
well as poultry products processing may be from plants, animals, or microorganisms.
Apart from exogenous enzymes, the roles of endogenous enzymes are also crucial
for the processing of food products. The enzymes from the microbial source are most
widely preferred since they can be easily manipulated and microorganisms can be
easily cultured. Besides, the trend towards the use of enzymes from marine
microorganisms has been increasing from the last few years (Fernandes, 2016).
Proteases such as ficin, bromelain, and papain, which are primarily plant-based,
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 199
have been widely used in meat tenderization. These proteases possess a sulfhydryl
group in their active sites to carry out their catalytic activity. Apart from meat
tenderization, these proteases have also been used in the preparation of the fish
sauce. Moreover, papain has also been used in the preparation of fish protein
hydrolyzates (Fernandes, 2016). Research has also been carried out on the extraction
of enzymes from fish-processing discards and their application in the processing of
fish products. Enzymes including pepsin, trypsin, gastricin, alkaline proteinase,
acidic protease, calpain, lysosomal cathepsins type B, H, L, cathepsin and other
lysosomal cathepsins, chymotrypsin, collagenases, chymosin, elastase, lipase,
carbohydrases, and alkaline peptidase from fish-processing by-products have been
tested for their application in the processing of fish and seafood products
(Venugopal, 2016). Most of the enzymes used in meat, fish, and poultry product
processing are proteases. Besides proteases, other enzymes (lipases and
transglutaminases) play a major role in flavor modification, hydrolysis of fatty
acids, and other processing methods. The different enzymes used in the processing
of meat, fish, and poultry products, and also their sources are exhibited in Tables 8.1,
8.2 and 8.3, Fig. 8.4.
8.4.1 Calpains
Table 8.1 Proteases used in the processing of meat, fish, and poultry products
Source Enzyme Source Applications
Plants Ficin Fig (Ficus glabrata, Meat tenderization
Ficus laurifolia, Ficus
anthelmintica)
Papain Papaya (Carica papaya) Meat tenderization, preparation
of fish sauce, and fish protein
hydrolyzates
Bromelain Pineapple (Ananas Meat tenderization and
comosus) preparation of fish sauce
Actinidin Kiwi fruit (Actinida Meat tenderization
deliciosa)
Cucumin Kachri fruit (Cucumis Meat tenderization
pubescens, Cucumis
trigonas)
Zinzibain Ginger (Zingiber Meat tenderization
officinale)
Animal Porcine Pork Meat tenderization
pancreatin
Microorganisms Fungal Aspergillus oryzae, Meat tenderization, preparation
protease Penicilium of fish protein hydrolyzates
chrysogenum Pg222
Alkaline Bacillus subtilis Preparation of fish protein
protease hydrolyzates
Thermophilic Bacillus strain E.A.1 Meat tenderization
enzyme E
A1 protease Thermus strain Meat tenderization
4-1 A Rt4-1.A Meat tenderization
protease
Caldolysin Thermus strain T-35 Meat tenderization
Elastase Bacillus species Meat tenderization
EL31410
Collagenase Vibrio B-30 Meat tenderization
Source: Naveena et al. (2004), Venugopal et al. (2000), Cazarin et al. (2015)
8.4.2 Cathepsins
Cathepsins are lysosomal acid proteases found in almost all organisms. There are
13 types of lysosomal cathepsins, 7 of which exist in skeletal muscle (Goll et al.,
1983). Cathepsins comprise both exoproteases and endoproteases, and are
differentiated from the active site: aspartate (Cathepsin D and E), serine (Cathepsin
G), and cysteine (Cathepsin B, H, L, and X) (Sentandreu et al., 2002). They exist as a
proenzyme in living tissue. Due to pH fall after cell death, they are released into the
cytoplasm or intercellular space due to lysosomal disruption (Duston, 1983).
Cathepsins B and L are the major cathepsins involved in muscle proteolysis (Jamdar
& Harikumar, 2002). Protease inhibitor cystatin regulates cathepsin B, H, and L
in vivo (Turk & Bode, 1991).
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 201
Table 8.2 Lipases used in the processing of meat, fish, and poultry products
Source Source Applications
Animal Porcine pancreas Preparation of ω-3 PUFA concentrates
Atlantic cod Production of ω-3-enriched triglycerides, flavor
improvement
Sardine Production of ω-3-enriched triglycerides, flavor
improvement
Indian mackerel Production of ω-3-enriched triglycerides, flavor
improvement
Red sea bream Production of ω-3-enriched triglycerides, flavor
improvement
Salmon Production of ω-3-enriched triglycerides, flavor
improvement
Microorganisms Pseudomonas Enrichment of EPA and DHA in sardine oil
species
Candida Increasing DHA content, hydrolysis of menhaden oil
cylindracea
Lactobacillus Synthesis of short-chain fatty acid esters
plantarum
Source: Suresh et al. (2015), Venugopal et al. (2000), Cazarin et al. (2015), Uppada et al. (2017)
Table 8.3 Transglutaminase application in the processing of meat, fish, and poultry products
Source Source Applications
Microorganisms Streptoverticillum sp., Fish meat sheet formation, fish meat film and
Bacillus subtilis, mince formulations, surimi and restructured
Streptomyces sp. fishery products, texture modification of
finfish, collagen and gelatin bond formation,
improve the gel strength of chicken and beef
sausages, improve the gel strength of chicken
and beef meat
Source: Suresh et al. (2015), Cazarin et al. (2015)
Enzymes (Proteases,
Lipases,
Transglutaminases)
8.4.3 Papain
Papain is the most common enzyme used in meat tenderization. Papain, which
belongs to the C1 family of cysteine protease, also called the papain family
(Rawlings et al., 2010), is obtained from the latex of the papaya plant (Carica
papaya), and it plays a major physiological role in protecting the plant from insects
(Konno et al., 2004). Papain’s commercial importance is primarily due to its good
proteolytic activity against a broad variety of protein substrates, and because it is
active in a wide range of operating conditions. Smith and Hong-Shum (2003)
reported that it has a high optimal temperature (65 C) and broad pH range (5–8)
for its activity. Papain predominantly acts on the muscle structural component,
which enhances the meat tenderization (Gracey & Thronton, 1985). It is a highly
effective enzyme that is capable of inducing major degradation of collagen and
myofibrillar proteins (Ashie et al., 2002), and the maximum tenderizing activity
occurs during the cooking process (Tappel et al., 1956). Kang and Rice (1970)
reported that papain showed strong activity for a myofibrillar fraction with greater
solubilizing activity on the connective tissue. Use of papain leads to over tenderiza-
tion of meat which resulted in mushy meat, hence the commercial use of papain is
limited for meat tenderization (Han et al., 2009).
8.4.4 Ficin
Ficin, the proteolytic enzyme from fig trees (Ficus sp.), is a sulfhydryl protease.
About ten proteases are present in crude ficin latex (Kramer & Whitaker, 1964). The
mostly studied ficins are those from Ficus glabrata and Ficus carica. It is reported to
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 203
hydrolyze and increase the solubility of muscle proteins (El-Gharbawi & Whitaker,
1963). The solubility of meat protein might be increased by ficin by degrading the
proteins into units of smaller molecular weight, which, when aggregated, form a
three-dimensional network (Ramezani et al., 2006). El-Gharbawi and Whitaker
(1963) reported that the optimum pH was around 7 for enzyme activity for collagen
and myofibrillar proteins, and about 5.0–5.5 for elastin.
8.4.5 Bromelain
The proteolytic enzymes called bromelain are present in fruits, leaves, and stems of
the Bromeliaceae family of which pineapple (Ananas comosus) is the most com-
monly used enzyme in the processing of meat (Doko et al., 1991). The enzyme
extracted from the stem is called stem bromelain, and that extracted from the fruit is
called fruit bromelain (Vanhoof & Cooreman, 1997). Fruit bromelain is said to have
a higher proteolytic activity and specificity compared to stem bromelain (Barrett
et al., 2004; Grzonka et al., 2007). This enzyme, like other proteases, results in over-
tenderization by degrading myofibrillar proteins and collagen (Melendo et al., 1996).
At first, it degrades about 40% of the collagen in the sarcolemma, and then it
degrades myosin in the myofibrillar region (Kang & Rice, 1970; Wang et al.,
1958). The application of this enzyme is easy and cheap and can be exploited on a
household and industrial scale for tenderizing tough meat; it is also a better alterna-
tive to chemical tenderizers (Sunantha & Saroat, 2011). Ionescu et al. (2008)
investigated the use of bromelain in adult beef with best results at 10 mg/100 g
meat, with a tendering time of 24 h at 4 C, followed by thermal treatment with an
increase of 1 C/ min to 70 C (when enzyme inactivation occurs). These conditions
improved the tenderness of the beef.
8.4.6 Cucumin
Cucumin obtained from kachri (Cucumis pubescens) is a protease. Initially, the dried
coarsely ground fruits of C. trigonus Roxb (locally known as kachri) were tradition-
ally used as a meat tenderizer in some parts of India (Naveena et al., 2004). Cucumin
tenderizes meat to a significant degree, and may serve as a basis for commercial
meat-tenderizing mixtures (Hujjatullah & Baloch, 1970).
8.4.7 Zingibain
Zingibain protease was isolated from the plant Zingiber officinale roscoe (ginger
rhizome) by Thompson et al. (1973). This thiol protease shows optimum activity at
60 C (Naveena et al., 2004). The ginger extracts contain two cysteine proteases with
a molecular mass of 29 and 31 kDa, as reported by Su et al. (2009). This enzyme has
a greater proteolytic activity when heated (Naveena & Mendiratta, 2001). Its
204 S. S. Karkal et al.
proteolytic activity produces more tender meat by degrading collagen and actino-
mycin (Naveena & Mendiratta, 2001; Thompson et al., 1973). Lee et al. (1986)
reported that an extensive degradation of myofibrillar proteins is possible with a high
concentration of ginger extract: the degradation appears to begin at the I band of each
sarcomere and progresses to the M line.
8.4.8 Actinidin
Actinidin or actinidain is a cysteine protease obtained from the kiwi fruit (Actinidai
deliciosa). Actinidin is stable at a pH range of 7–10 and has optimal activity at
58–62 C (Yamaguchi et al., 1982). A pH range of 5–7 is also reported by Boyes
et al. (1997), which reflects variations in the cultivar and assay used to estimate the
proteolytic activity. Both myofibrillar proteins and connective tissue proteins are
hydrolyzed by this enzyme (Christensen et al., 2009; Han et al., 2009). But a higher
hydrolysis of collagen was reported by Wada et al. (2002).
8.4.9 Aminopeptidases
Aminopeptidases are the major enzymes involved in the characteristic flavor devel-
opment in meat products. They hydrolyze amino acids from the N terminus of
peptides and proteins during meat processing. This generates a large number of
free amino acids that produce flavor. In porcine skeletal muscle, aminopeptidase B
(BAP), leucyl (LAP), alanyl (AAP), and pyroglutamyl (PGAP) have been localized
in the cytosol, and are named according to the preference of specific N terminal
amino acid (Flores et al., 1993, 1996, 1997). Except for leucyl aminopeptidase, all
are active at acidic pH. In the cytosol of skeletal muscle, around 86% of the total
aminopeptidase consists of alanyl aminopeptidase (Lauffart & Mantle, 1998). As
reported by Toldra (1992), aminopeptidases show good stability during the
processing of dry-cured ham, with activity still recovered toward the end of the
process.
8.4.10 Lipases
Lipolysis is the process of degradation of lipids in meat during processing (Toldra &
Flores, 1998). Enzymes of fat cells and muscle fibers, as well as bacterial enzymes
are involved in lipolysis. However, it is established that in dry fermented sausages,
the contribution of bacterial lipase is weak because of many differences in the
medium and optimum conditions (Molly et al., 1997). Intense lipolysis occurs in
muscle and adipose tissues during the dry-curing of meat; most of the free fatty acid
generation occurs during the initial 5 months (Motilva et al., 1993). Important
lipolytic enzymes of adipose tissue are lipoprotein lipase, hormone-sensitive lipase,
and monoacylglycerol lipase (Belfrage et al., 1984). Lipoprotein lipase, specific for
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 205
8.4.12 Elastase
Elastase is the enzyme that breaks down elastin in connective tissue. A new elastase,
a protease with high proteolytic activity, was isolated from Bacillus sp. EL31410 by
Chen and He (2002). It had a marked preference for elastin and collagen, which can
contribute to meat hardness, over other myofibrillar proteins at the pH of meat,
usually ranging from 5.5 to 6.0, and it has the same tenderization effect as papain on
beef meat based on the results of texture, sensory, and structural analyses (Qihe et al.,
2006).
Many strains of bacteria play a major role in protein degradation in fermented meat
and fish products (Bekhit, 2010). Because of relatively specific activity and low
inactivation temperatures, proteases from bacterial sources make them useful for
meat tenderization. Alkaline elastase from alkalophilic Bacillus sp. strain Ya-B
206 S. S. Karkal et al.
Animal proteases are less used for meat tenderization. Injection of pancreatin on beef
semitendinosus tended to improve overall tenderness without affecting the taste
(Pietrasik et al., 2010).
Various enzymes from different sources have been exploited for the processing of
fish products and their preservation. The main enzymes, which play a key role in the
processing of fish and seafood are described below.
8.5.1 Proteases
Proteases find their wide application in the seafood processing industry, especially in
deskinning, descaling, salted fish ripening, fish sauce production, squid tenderiza-
tion, etc. Besides, proteases have also been examined for the extraction of
compounds (mainly pigments, oils and fats, and flavors) and the preparation of
hydrolyzates from seafood processing wastes (Suresh et al., 2015). Proteases from
plant, animal, microbial, and even aquatic sources have been used for the processing
of fish.
viscera) with proteases at ideal pH and temperature for a few hours, followed by
drying. The enzymes preferred for the production of fish protein hydrolyzates
include pepsin, trypsin, chymotrypsin, alcalase, etc. The prepared fish protein
hydrolyzates are amorphous powder and hygroscopic in nature. The fish protein
hydrolyzates contain proteins (80–90%), fats (<5%), minerals (5–8%) and mois-
ture (1–8%) (Venugopal et al., 2000; Venugopal, 2016). Traditionally, the prepa-
ration of fish protein hydrolyzates was carried out using either acids or bases.
Hydrochloric acid or sulfuric acid (occasionally) was preferred for acid hydroly-
sis, whereas alkali hydrolysis was carried out using sodium hydroxide. Both acid
and alkali hydrolysis are carried out at elevated temperature. Moreover, alkali
hydrolysis produces toxic compounds during the reaction process, and acid
hydrolysis destroys tryptophan. Besides, fish protein hydrolyzates preparation
using low-cost proteolytic enzymes can be carried out at a mild temperature,
pressure, and pH, and doesn’t produce toxic compounds. Thus, the preparation of
fish protein hydrolyzates using proteolytic enzymes is economical (Fernandes,
2016).
Gajanan et al. (2016) prepared protein hydrolyzates from threadfin breams
(Nemipterus japonicas) a fish frame waste using two different plant proteases,
papain and bromelain. The prepared protein hydrolyzates exhibited high
Angiotensin-Converting Enzyme (ACE) inhibitory and anti-oxidative activity.
The hydrolyzates also showed better functional properties. Similar results were
also exhibited by protein hydrolyzates prepared from the muscle of small-spotted
catshark (Scyliorhinus anicula) using esperase and alcalase (Vázquez et al.,
2017).
3. Fish sauce: Fish sauce is a product produced by the fermentation process using
endogenous or exogenous enzymes. The underlying process mainly involves the
solubilization and digestion of fish proteins by enzymes. The pre-dominant
enzymes involved in protein hydrolysis include chymotrypsin and trypsin, along-
side cathepsins. When the pH of fish sauce drops from 7 to 5, the action of trypsin
and chymotrypsin becomes complicated since they are active at near-neutral pH,
whereas cathepsins are active at acidic pH. Fish sauce production using endoge-
nous enzymes is a traditional process, and it’s a very time-consuming process.
Therefore, to hasten the process, exogenous enzymes such as papain, ficin, and
bromelain, and commercial enzymes such as protamex (Protex 51FP) and
neutrase have been used (Fernandes, 2016). About 80–90% of the Southeast
Asian people consume fish sauce. It is being used as a prime condiment in
Southeast Asian cuisines to improve the taste of food. Currently, Thailand is
the leading producer of fish sauce in the world (Gowda et al., 2020).
4. Recovery of proteins from fish processing waste: Fish waste is a rich source of
proteins such as collagen, myofibrillar proteins, and sarcoplasmic proteins.
Proteases derived from the fish wastes (collagenases and pepsin) have been
employed for the extraction of collagen from various fish offals, including
skins, fins, bones, scales, swim bladders, and heads (Venugopal, 2016).
5. Ripening: Proteases have been known to be involved in the ripening of fermented
fishery products as well as salted fish (Venugopal, 2016; Fernandes, 2016). The
208 S. S. Karkal et al.
8.5.2 Lipases
8.5.3 Transglutaminase
Besides proteases, lipases, and transglutaminases, other enzymes have also been
exploited for the fish products processing industries. Enzymes such as diamine
oxidase, xanthine oxidase, nucleotide phosphorylase, putrescine oxidase, and gluta-
mine dehydrogenase have been used for the evaluation of the quality of seafood and
freshness. Restriction endonucleases and Taq polymerase have been used for the
identification of fish species and adulteration detection. Furthermore, research works
have also been carried out in increasing the shelf-life of seafood products using
glucose oxidase enzymes.
Various exogenous and endogenous enzymes have been exploited for the processing
of poultry products. The enzymes used in the processing of pork, sheep, and beef
meat are also being used in the processing of poultry meat. Mainly the enzymes are
used to improve the tenderness of poultry meat. Papain, ficin, and bromelain
enzymes derived from plants have been extensively utilized for the tenderization
of poultry meat. The proteolytic enzymes derived from plants are considered to be
superior to enzymes derived from bacteria (Ketnawa & Rawdkuen, 2011). The
applications of commercial enzymes such as alcalase, neutrase, flavorzyme,
protamex, collupulin, bromelain, and alphalase in softening the chicken meat
(breast) texture have been studied. Of all the enzymes bromelain and collupulin
have shown the marked softening of chicken breast. The exploitation of proteolytic
enzymes from Calotropis procera latex has also shown to be effective in the
210 S. S. Karkal et al.
tenderization of poultry meat. Apart from the application of enzymes in poultry meat
processing, the addition of enzymes in the improvement of barley and wheat-based
feed has also been studied by Perić et al. (2011). They stated that incorporating the
enzyme Allzyme SSF into poultry feed had a positive effect on the growth of
chicken, and observed increased egg production in layers. Thus, enzymes find
extensive application in the poultry industry to prepare poultry feed and poultry
meat processing.
8.8 Conclusion
Currently, the meat, fish, and poultry industries play a significant role in meeting the
global food demand. The enzymatic processing of meat, fish, and poultry products
processing is still in the early stage, and the introduction of new techniques is
resulting in progress. The enzymatic processing technique has the potential to
overthrow the chemical and mechanical techniques employed in the processing of
meat, fish, and poultry products processing. This would help to overcome the
environmental problems caused by chemical and mechanical processing techniques.
The most suitable enzymes that could be employed in the processing of meat, fish,
and poultry products are discussed in this chapter. The meat and poultry industries
mainly utilize enzymes to improve the tenderness of the meat, whereas in fish
products processing enzymes have a wide array of applications. Technological
advancements have enabled the development of recombinant enzymes from
microorganisms for their application in these industries. However, recombinant
enzymes are yet to be used in the industries. Thus, the enzymes have a promising
future for their application in the processing of meat, fish, and poultry products.
Acknowledgments The authors are grateful to the CSIR-Central Food Technological Research
Institute (CSIR-CFTRI), Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, and
the Department of Biotechnology (Govt. of India, New Delhi) for the support.
References
Ashie, I. N. A., Sorensen, T. L., & Nielsen, P. M. (2002). Effects of papain and a microbial enzyme
on meat proteins and beef tenderness. Journal of Food Science, 67, 2138–2142.
Barrett, A. J., Rawlings, N. D., & Woessner, J. F. (2004). Handbook of proteolytic enzymes.
Elsevier Academic Press.
Bawa, A. S., Orr, H. L., & Usborne, W. R. (1981). Enzymatic tenderization of spent white leghorn
hens. Poultry Science, 60(4), 744–749.
Bekhit, A. E. D. (2010). Fermentation of fish roe. In D. R. Heldman, D. G. Hoover, & M. B.
Wheeler (Eds.), The encyclopedia of biotechnology in agriculture and food (Vol. 1, pp.
251–256). Taylor & Francis Group.
Belfrage, P., Fredrikson, G., Stralfors, P., & Tornqvist, H. (1984). Adipose tissue lipases. In
B. Borgström & H. L. Brockman (Eds.), Lipases (p. 365). Elsevier.
Benito, M. J., Rodrıguez, M., Acosta, R., & Córdoba, J. J. (2003). Effect of the fungal extracellular
protease EPg222 on texture of whole pieces of pork loin. Meat Science, 65(2), 877–884.
Benito, M. J., Rodrıguez, M., Martın, A., Aranda, E., & Córdoba, J. J. (2004). Effect of the fungal
protease EPg222 on the sensory characteristics of dry fermented sausage “salchichón” ripened
with commercial starter cultures. Meat Science, 67(3), 497–505.
Benito, M. J., Rodríguez, M., Núñez, F., Asensio, M. A., Bermúdez, M. E., & Córdoba, J. J. (2002).
Purification and characterization of an extracellular protease from Penicillium chrysogenum
(Pg222) active against meat proteins. Applied and Environmental Microbiology, 68,
3532–3536.
Boyes, S., Strubi, P., & Marsh, H. (1997). Actinidin levels in fruit of Actinidia species and some
Actinidia arguta rootstock scion combinations. LWT—Food Science and Technology, 30,
379–389.
212 S. S. Karkal et al.
Cazarin, C. B. B., Lima, G. C., da Silva, J. K., & Maroostica, M. R., Jr. (2015). Enzymes in meat
processing. In M. Chandrasekaran (Ed.), Enzymes in food and beverage processing
(pp. 354–377). CRC Press.
Chandrasekaran, M. (2015). Emerging trends and future prospects. In M. Chandrasekaran (Ed.),
Enzymes in food and beverage processing (pp. 354–377). CRC Press.
Chandrasekaran, M., Basheer, S. M., Chellappan, S., Krishna, J. G., & Beena, P. S. (2015).
Enzymes in seafood processing. In M. Chandrasekaran (Ed.), Enzymes in food and beverage
processing (pp. 354–377). CRC Press.
Chen, Q. H., & He, G. Q. (2002). Optimization of medium composition for the production of
elastase by Bacillus sp. EL31410 with response surface methodology. Enzyme and Microbial
Technology, 5, 667–672.
Christensen, M., Torngren, M. A., Gunvig, A., Rozlosnik, N., Lametsch, R., Karlsson, A. H., &
Ertbjerg, P. (2009). Injection of marinade with actinidin increases tenderness of porcine
M. biceps femoris and affects myofibrils and connective tissue. Journal of the Science of
Food and Agriculture, 89, 1607–1614.
Copeland, R. A. (Ed.). (2000). Enzymes: A practical introduction to structure, mechanism, and data
analysis (pp. 1–10). Wiley-VCH.
Cronlund, A. L., & Woychik, J. H. (1986). Effect of microbial rennets on meat protein fractions.
Journal of Agricultural and Food Chemistry, 34, 502–505.
Doko, M. B., Bassani, V., Casadebaig, J., Cavailles, L., & Jacob, M. (1991). International Journal
of Pharmaceutics, 76, 199–206.
Duston, T. R. (1983). Relationship of pH and temperature to disruption of specific muscle proteins
and activity of lysosomal proteases. Journal of Food Biochemistry, 7, 223–245.
El-Gharbawi, M., & Whitaker, J. R. (1963). Factors affecting enzymatic solubilization of beef
proteins. Journal of Food Science, 28, 168–172.
European Commission-GRFC. (2019). Food shortage. Retrieved July 7, 2020 from https://ec.
europa.eu/knowledge4policy/publication/global-report-food-crises-2019_en#:~:
text¼According%20to%20the%202019%20edition,3%20or%20above)%20in%202018
FAO. (2016). The state of world fisheries and aquaculture. Contributing to food security and
nutrition to all, Fish production to combat food shortage, Rome, 200 pp. Retrieved July 7, 2020.
FAO. (2019). Food basic necessity of life. Retrieved July 7, 2020 from https://
nutritionmeetsfoodscience.com/2019/01/03/food-more-than-an-essential-need/
FAO-GLOBEFISH. (2020). Information and analysis on world fish trade. FAO. Retrieved July
7, 2020 from http://www.fao.org/in-action/globefish/fishery-information/en/#:~:text¼Global%
20fish%20production%20growth%20is,have%20increased%203.9%20percent%20...
Fernandes, P. (2016). Enzymes in fish and seafood processing. Frontiers in Bioengineering and
Biotechnology, 4, 59.
Flores, M., Aristoy, M. C., & Toldrá, F. (1993). HPLC purification and characterization of porcine
muscle aminopeptidase B. Biochimie, 75, 861.
Flores, M., Aristoy, M. C., & Toldra, F. (1997). Curing agents affect aminopeptidase activity from
porcine skeletal muscle. Zeitschrift für Lebensmittel-Untersuchung und -Forschung A, 205,
343–346.
Flores, M., Aristoy, M.-C., & Toldrá, F. (1996). HPLC purification and characterization of soluble
alanyl aminopeptidase from porcine skeletal muscle. Journal of Agricultural and Food Chem-
istry, 44, 2578.
Gajanan, P. G., Elavarasan, K., & Shamasundar, B. A. (2016). Bioactive and functional properties
of protein hydrolysates from fish frame processing waste using plant proteases. Environmental
Science and Pollution Research, 23(24), 24901–24911.
Goll, D. E., Otusuak, Y., Nagainis, P. A., Shannon, J. D., Sathe, A. K., & Mururuma, M. (1983).
Role of muscle proteinases in maintenance of muscle integrity and mass. Journal of Food
Biochemistry, 7, 137–177.
Goll, D. E., Thompson, V. F., Li, H. Q., Wei, W., & Cong, J. Y. (2003). The calpain system.
Physiological Reviews, 83, 7.
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 213
Gomes, H. A., Moreira, L. R., & Edivaldo Filho, X. F. (2018). Enzymes and food industry: A
consolidated marriage. In A. M. Holban & A. M. Grumezescu (Eds.), Advances in biotechnol-
ogy for food industry (pp. 55–89). Academic Press.
Gowda, S. G. S., Narayan, B., & Gopal, S. (2020). Antioxidant properties and dominant bacterial
community of fermented Rohu (Labeo rohita) sauce produced by enzymatic and fermentation
method. Turkish Journal of Fisheries and Aquatic Science, 20(8), 583–592.
Gracey, J. F., & Thronton, S. (1985). Meat hygiene (7th ed.). Bailliere Tindall, English Language
Book Society.
Grzonka, Z., Kasprzykowski, F., & Wiczk, W. (2007). Cysteine proteases. In J. Polaina & A. P.
MacCabe (Eds.), Industrial enzymes (pp. 181–195). Springer.
Han, J., Morton, J. D., Bekhit, A. E. D., & Sedcole, J. R. (2009). Pre-rigor infusion with kiwifruit
juice improves lamb tenderness. Meat Science, 82(3), 324–330.
Hope-Jones, M., Strydom, P. E., Frylinck, L., & Webb, E. C. (2010). The efficiency of electrical
stimulation to counteract the negative effects of β-agonists on meat tenderness of feedlot cattle.
Meat Science, 86, 699–705.
Hopkins, D. L., & Taylor, R. G. (2002). Post-mortem muscle proteolysis and meat tenderization. In
M. te Pas, M. Everts, & H. Haagsman (Eds.), Muscle development of livestock animals
(pp. 363–389). CAB International.
Hujjatullah, S., & Baloch, A. K. (1970). Proteolytic activity of Cucumis trigonus Roxb. Extraction,
activity, characteristics. Journal of Food Science, 35(3), 276–278.
Imanaka, T., Amanuma-Muto, K., Ohkuma, S., & Takano, T. (1984). Characterization of lysosomal
acid lipase purified from rabbit liver. Journal of Biochemistry, 96, 1089.
Ionescu, A., Aprodu, I., & Pascaru, G. (2008). Effect of papain and bromelin on muscle and
collagen proteins in beef meat. The Annals of the University Dunarea de Jos of Galati. Fascicle
VI—Food Technology, New Series, pp. 9–16.
Jamdar, S. N., & Harikumar, P. (2002). Sensitivity of catheptic enzymes in radurized chicken meat.
Journal of Food Science and Technology, 39(1), 72–73.
Kang, C. K., & Rice, E. E. (1970). Degradation of various meat fractions by tenderizing enzymes.
Journal of Food Science, 35, 563–565.
Kang, C. K., & Warner, W. D. (1974). Tenderization of meat with papaya latex proteases. Journal
of Food Science, 39(4), 812–818.
Ketnawa, S., & Rawdkuen, S. (2011). Application of bromelain extract for muscle food tenderiza-
tion. Food and Nutrition Sciences, 2, 393–401.
Kim, T., Silva, J. L., Parakulsuksatid, P., & Wang, D. S. (2014). Optimization of enzymatic
treatments for deskinning of catfish nuggets. Journal of Aquatic Food Product Technology,
23(4), 385–393.
Konno, K., Hirayama, C., Nakamura, M., Tateishi, K., Tamura, Y., Hattori, M., & Kohno,
K. (2004). Papain protects papaya trees from herbivorous insects: Role of cysteine proteases
in latex. The Plant Journal, 37, 370–378.
Koohmaraie, M., & Geesink, G. H. (2006). Contribution of post mortem muscle biochemistry to the
delivery of consistent meat quality with particular focus on the calpain system. Meat Science,
74, 34–43.
Koohmaraie, M., Whipple, G., Kretchmar, D. H., Crouse, J. D., & Mersmann, H. J. (1991).
Postmortem proteolysis in longissimus muscle from beef, lamb and pork carcasses. Journal of
Animal Science, 69, 617–624.
Kramer, D. E., & Whitaker, J. R. (1964). Ficus enzymes: II. Properties of the proteolytic enzymes
from the latex of Ficus carica variety Kadota. Journal of Biological Chemistry, 239,
2178–2183.
Lauffart, B., & Mantle, D. (1998). Rationalisation of aminopeptidase activities in human skeletal
muscle soluble extract. Biochimica et Biophysica Acta, 956, 300.
Lee, Y. B., Kim, Y. S., & Ashmove, C. R. (1986). Antioxidant property of ginger rhizome and its
application in meat products. Journal of Food Science, 51, 20–23.
214 S. S. Karkal et al.
Melendo, J. A., Beltrán, J. A., Jaime, I., Sancho, R., Roncalés, P., & P. (1996). Limited proteolysis
of myofibrillar proteins by bromelain decreases toughness of coarse dry sausage. Food Chemis-
try, 57(3), 429–433.
Mikami, M., Whiting, A. H., Taylor, M. A. J., Maciewicz, R. A., & Etherington, D. J. (1987).
Degradation of myofibrils from rabbit, chicken and beef by cathepsin l and lysosomal lysates.
Meat Science, 21, 81–97.
Molly, K., Demeyer, D., Johansson, G., Raemaekers, M., Ghistelinck, M., & Geenen, I. (1997). The
importance of meat enzymes in ripening and flavour generation in dry fermented sausages. First
results of a European project. Food Chemistry, 54(4), 539–545.
Motilva, M.-J., Toldrá, F., Nieto, P., & Flores, J. (1993). Muscle lipolysis phenomena in the
processing of dry-cured ham. Food Chemistry, 48, 121.
Naveena, B. M., & Mendiratta, S. K. (2001). Tenderization of spent hen meat using ginger extract.
British Poultry Science, 42, 344–349.
Naveena, B. M., Mendiratta, S. K., & Anjaneyulu, A. S. R. (2004). Tenderization of buffalo meat
using plant proteases from Cucumis trigonus Roxb (Kachri) and Zingiber officinale roscoe
(Ginger rhizome). Meat Science, 68(3), 363–369.
Núñez, F., Rodríguez, M. M., Bermúdez, M. E., Córdoba, J. J., & Asensio, M. A. (1996).
Composition and toxigenic potential of the mould population on dry-cured Iberian ham.
International Journal of Food Microbiology, 32, 185–197.
O’Halloran, G. R., Troy, D. J., Buckley, D. J., & Reville, W. J. (1997). The role of endogenous
proteases in the tenderisation of fast glycolysing muscle. Meat Science, 47, 187–210.
Padmapriya, B., Rajeswari, T., Noushida, E., Sethupalan, D. G., & Venil, C. K. (2011). World
Applied Science Journal, 12, 1798–1802.
Perić, L., Sartowska, K., Milošević, N., Đukić-Stojčić, M., Bjedov, S., & Nikolova, N. (2011). The
effect of enzymes on the economics of poultry meat and egg production. Macedonian Journal of
Animal Science, 1, 113–117.
Pietrasik, Z., Aalhus, J. L., Gibson, L. L., & Shand, P. J. (2010). Influence of blade tenderization,
moisture enhancement and pancreatin enzyme treatment on the processing characteristics and
tenderness of beef semitendinosus muscle. Meat Science, 84(3), 512–517.
Qihe, C., Guoqing, H., Yingchun, J., & Hui, N. (2006). Effects of elastase from a Bacillus strain on
the tenderization of beef meat. Food Chemistry, 98(4), 624–629.
Ramezani, R., Aminlari, M., & Fallahi, H. (2006). Effect of chemically modified soy proteins and
ficin-tenderized meat on the quality attributes of sausage. Journal of Food Science: Food
Chemistry and Toxicology, 68, 85–88.
Rawlings, N. D., Barrett, A. J., & Bateman, A. (2010). MEROPS: The peptidase database. Nucleic
Acids Research, 38, D227–D233.
Rees, M. P., Trout, G. R., & Warner, R. D. (2002). Effect of calcium infusion on tenderness and
ageing rate of pork m. longissimus thoracis et lumborum after accelerated boning. Meat Science,
61, 169–179.
Sentandreu, M. A., Coulis, G., & Ouali, A. (2002). Role of muscle endopeptidases and their
inhibitors in meat tenderness. Trends in Food Science and Technology, 13, 400–421.
Shahidi, F., & Kamil, Y. J. (2001). Enzymes from fish and aquatic invertebrates and their applica-
tion in the food industry. Trends in Food Science & Technology, 12(12), 435–464.
Singh, S. B. (2018). Enzyme catalysis and its role in food processing industries. In M. Kuddus
(Ed.), Enzymes in food technology (pp. 143–165). Springer.
Smith, J., & Hong-Shum, L. (2003). Enzymes. In Food additives data book (pp. 389–462).
Blackwell Science.
Statista. (2019). Retrieved July 8, 2020 from https://www.statista.com/statistics/264577/total-
world-fish-production-since-2002/#:~:text¼The%20volume%20of%20global%20fish,million
%20metric%20tons%20in%202010
Statista. (2020). Retrieved July 8, 2020 from https://www.statista.com/statistics/237632/
production-of-meat-worldwide-since-1990/
8 Enzymes in Meat, Fish, and Poultry Product Processing and Preservation-II 215
Su, H. P., Huang, M. J., & Wang, H. T. (2009). Characterization of ginger proteases and their
potential as a rennin replacement. Journal of the Science of Food and Agriculture, 89(7), 1178–
1185.
Sunantha, K., & Saroat, R. (2011). Application of bromelain extract for muscle foods tenderization.
Food and Nutrition Sciences. https://doi.org/10.4236/fns.2011.25055
Suresh, P. V., Nidheesh, T., & Pal, G. K. (2015). Enzymes in seafood processing. In
M. Chandrasekaran (Ed.), Enzymes in food and beverage processing (pp. 354–377). CRC Press.
Suzuki, K., Saido, T. C., & Hirai, S. (1992). Modulation of the cellular signal by calpain. Annals of
the New York Academy of Sciences, 674, 218–227.
Takagi, H., Kondou, M., Hisatsuka, T., Nakamori, S., Tsai, Y. C., & Yamasaki, M. (1992). Effects
of an alkaline elastase from an alkalophilic Bacillus sytrain on the tenderization of beef meat.
Meat Science, 40, 577–583.
Tappel, A. L., Miyada, D. S., Sterling, C., & Maier, V. P. (1956). Meat tenderization. II. Factors
affecting the tenderization of beef by papain. Food Research, 21, 375–383.
Thompson, E. H., Wolf, I. D., & Allen, C. E. (1973). Ginger rhizome: A new source of proteolytic
enzyme. Journal of Food Science, 38, 652–655.
Toldra, F. (1992). The enzymology of dry curing of meat products. In F. J. M. Smulders, F. Toldra,
J. Flores, & M. Prieto (Eds.), New technologies for meat and meat products (p. 209).
Toldra, F., & Flores, M. (1998). The role of muscle proteases and lipases in flavor development
during the processing of dry-cure Ham. Critical Reviews in Food Science and Nutrition, 38(4),
331–352.
Turk, V., & Bode, W. (1991). The cystatins: Protein inhibitors of cysteine proteinases. FEBS
Letters, 285(2), 213–219.
United Nations-Department of Economic and Social Affairs (UN DESA). (2017). World population
projected to reach 9.8 billion in 2050, and 11.2 billion in 2100. Retrieved July 7, 2020, from
https://www.un.org/development/desa/en/news/population/worldpopulation-prospects-2017.
html
Uppada, S. R., Akula, M., Bhattacharya, A., & Dutta, J. R. (2017). Immobilized lipase from
Lactobacillus plantarum in meat degradation and synthesis of flavor esters. Journal of Genetic
Engineering and Biotechnology, 15(2), 331–334.
Vanhoof, G., & Cooreman, W. (1997). Bromelain. In A. Lauwers & S. Scharpe (Eds.), Pharma-
ceutical enzymes (pp. 131–154). Marcel Dekker.
Vázquez, J. A., Blanco, M., Massa, A. E., Amado, I. R., & Pérez-Martín, R. I. (2017). Production of
fish protein hydrolysates from Scyliorhinus canicula discards with antihypertensive and antiox-
idant activities by enzymatic hydrolysis and mathematical optimization using response surface
methodology. Marine Drugs, 15(10), 306.
Veeramuthu, G. I., & Sams, A. R. (1999). Post mortem pH, myofibrillar fragmentation, and calpain
activity in Pectoralis from electrically stimulated and muscle tensioned broiler carcasses.
Poultry Science, 78, 272–276.
Venugopal, V. (2016). Enzymes from seafood processing waste and their applications in seafood
processing. In S. Kim & F. Toldra (Eds.), Advances in food and nutrition research (Vol. 78, pp.
47–94). Academic Press.
Venugopal, V., Lakshmanan, R., Doke, S. N., & Bongirwar, D. R. (2000). Enzymes in fish
processing, biosensors and quality control: A review. Food Biotechnology, 14(1–2), 21–77.
Wada, M., Suzuki, T., Yaguti, Y., & Hasegawa, T. (2002). The effects of pressure treatments with
kiwi fruit protease on adult cattle semitendinosus muscle. Food Chemistry, 78, 167–171.
216 S. S. Karkal et al.
Wang, H., Weir, C. E., Birkner, M. L., & Ginger, B. (1958). Studies on enzymatic tenderization of
meat. III. Histological and panel analyses of enzyme preparations from three distinct sources.
Food Research, 23, 423–438.
Whipple, G., Koohmaraie, M., Dikeman, M. E., Crouse, J. D., Hunt, M. C., & Klemm, R. D.
(1990). Effect of fasting on energy metabolism and tenderizing enzymes in chicken breast
muscle early postmortem. Evaluation of attributes that affect longissimus muscle tenderness in
Bos taurus and Bos indicus cattle. Journal of Animal Science, 68(9), 2716–2728.
Yamaguchi, Y., Yamashita, Y., Takeda, I., & Kiso, H. (1982). Proteolytic enzymes in green
asparagus, kiwifruit and mint: Occurrence and partial characterisation. Agricultural and
Biological Chemistry, 46, 1983–1986.
Yeh, C. M., Yang, M.-C., & Tsai, Y.-C. (2002). Application potency of engineered G159 mutants
on P1 substrate pocket of subtilisin YaB as improved meat tenderizers. Journal of Agricultural
and Food Chemistry, 50, 6199–6204.
Enzymes in Functional Food Development
9
Iran Alemzadeh, Asma Sadat Vaziri, Kianoush Khosravi-Darani,
and Pierre Monsan
Abstract
# The Author(s), under exclusive license to Springer Nature Singapore Pte 217
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_9
218 I. Alemzadeh et al.
Keywords
Enzymes · Functional food · L-Asparaginase · β-Glucanase · Chlorophyllase ·
Lipase · L-Glutaminase · Phytase · Protease · Tannase
9 Enzymes in Functional Food Development 219
9.1 Introduction
Fig. 9.1 Enzymes accessible in functional food development and the selected bioactive
components
be further utilized as protein substrates for food applications (Pokora et al., 2013).
Bioactive peptides (BP) are one of the most important bioactive compounds
generated from protease hydrolysis. The protein fragments of BPs act as peptide
inhibitors of the angiotensin-converting enzyme (ACE), and antimicrobial,
anti-aggregating, antithrombotic, anticancer and antihypertensive, and immune-
modulatory peptides. The proteolysis of BPs is required for their release and activa-
tion since they are hidden and encrypted within the parent proteins, such as casein,
lactalbumin, beta-lactoglobulin, and lactoferrin (De Leo et al., 2009). Gamma-
aminobutyric acid (GABA) is another potent bioactive protein catalyzed by gluta-
mate decarboxylase. GABA functions as a major inhibitory neurotransmitter in the
mammalian brain and central nervous system that strongly effects the behavior and
stress management. It is widely distributed in nature and demand for microbial
GABA supply has raised to satisfy the needs of GABA-enriched foods (Cui et al.,
2020).
Tannase is another group of enzymes that are suitable candidates as biocatalysts
in the treatment of phenolic-rich foods. It catalysis the biotransformation of phenolic
compounds in beverages to generate a product with enhanced clarity and color
appearance, as well as promoting its functional activity. As an example, the process
of browning in tea beverages occurs due to the presence of caffeine and catechins in
the tea, which results in the formation of insoluble compounds in cold water.
Biotransformation by tannase reduces the gallic acid-binding and inhibits the
browning effect (Srivastava & Kar, 2009; Cao et al., 2019; Gligor et al., 2019).
The enzyme accessible in functional food development and bioactive components is
categorized in Fig. 9.1. In this chapter, we describe the functional food components,
functional food in health, the role of selected enzymes in functional foods
manufacturing, or food improving quality circumstances. Enzymes, such as
asparaginase, carbohydrase, chlorophyllase, lipase, L-glutaminase, phytase, prote-
ase, and tannase are introduced, and the beneficial health effects are discussed.
222 I. Alemzadeh et al.
Functional foods are enriched or dietary materials that may offer several health
benefits beyond basic nutrition. Functional foods for health are an essential factor of
an overall healthful lifestyle that consists of a balanced diet from various sources.
People should strive to consume a wide variety of foods and food components
including the examples listed in Table 9.1. These food components could naturally
exist in natural food, such as whole grains, fruits, vegetables, beverages, enriched
compounds, and dietary supplements, extracted by chemical approaches or produced
by enzymatic processes.
Obesity and malnutrition are major drawbacks associated with diet habits, which are
strongly influenced by urbanization, and lifestyle alterations (Tanna & Mishra,
2018). Functional food could inherently include bioactive ingredients or
nutraceuticals and could be added from various sources to produce fortified food.
The bioactive ingredients are in the group of carbohydrates, enzymes, protein,
phytochemicals, vitamins, and lipids (Suleria et al., 2015; Maqsood et al., 2020).
These bioactive compounds could be derived from different sources like cereals,
seeds, marine-based, microorganisms, plants, animals, and fishery. Since bioactive
compounds in natural food products are in low amounts, the bioactive ingredients
could be produced by a microbial or enzymatic process in high quantity. The
bioactive ingredients have antibiotic, antiparasitic, antiviral, anti-inflammatory,
anti-fibrotic, and anticancer activities. Health benefit of the bioactive compounds
affects metabolic syndrome (Mets), such as CVD, elevated triglycerides, cholesterol,
and blood pressure, besides insulin resistance (Sirtori et al., 2017). Nutraceutical
ingredients have specificity such as antimicrobial, antiviral, antioxidant, anti-
obesity, anticancer, antidiabetic, anti-inflammatory, nephrotoxic, and arthritis.
Enzymes are catalytic biomolecules responsible for the catalyzing diverse biochem-
ical reactions in several food industries. Enzymatic processes can be a promising
alternative to conventional organic synthesis, as they provide eco-friendly, more
viable and sustainable conditions, safer products, and use of renewable substrates.
Enzymes are therefore possessing a significant role in many biotechnological
applications including food quality improvement, as well as food manufacturing
operations. Selected enzymes representing these concerns are defined and evaluated
respectively.
9 Enzymes in Functional Food Development 223
Table 9.1 Examples of functional food sources selected nutraceuticals, and their biological
activity
Functional food Selected
sources nutraceutical Biological activity Reference
Coriander fruits Fatty oil Antimicrobial, Sahib et al. (2013)
(seed and antioxidant,
pericarp) antidiabetic
Non-starch Dietary fiber Cholesterol reduction Sirtori et al. (2017)
polysaccharides
(NPS)
Pumpkin seeds Traditional Antioxidative, Patel and Rauf (2017)
medicine hypoglycemic,
anticancer
Yeasts Various preventing oxidative Rai et al. (2019)
bioactive stress
components
Probiotics Traditional Intervention of IBD Al Mijan and Lim (2018),
medicine Grom et al. (2020)
Seaweed Metabolite Chronic diseases such Tanna and Mishra (2018)
as cancer, arthritis,
diabetes
Mediterranean Various Antidiabetes Alkhatib et al. (2017)
diet bioactive
components
Plant sterols and Phytosterols Plasma cholesterol Poli et al. (2018)
stanols control
Phytochemicals Various Cancer, CVD, anti- Gul et al. (2016)
bioactive obesity
components
Higher Dietary fibers Anti-cardiovascular Venkatakrishnan et al.
polysaccharide and cerebrovascular (2020)
disease
Food bioactive Polyphenol, Anticancer Adefegha (2018)
compounds, plant flavonoids
foods
CLA PUFAs Anti-obesity, Kouchak Yazdi et al.
antidiabetic (2017), Yazdi and
Alemzadeh (2017)
Cereals Polyphenols Anti-chronic, diabetes, Arab et al. (2011)
anticancer, anti-obesity
Marin Bioactive Anticoagulant, anti- Dewapriya and Kim (2014),
microorganisms polysaccharides inflammatory, antiviral Suleria et al. (2015)
Date fruit and Phytochemical Antioxidant, antifungal Maqsood et al. (2020)
seed and antiviral,
anticancer
224 I. Alemzadeh et al.
9.3.1 L-Asparaginase
been explored (Patro et al., 2014). Fungal sources are also considered safe
asparaginase producers (generally recognized as safe, GRAS). It has been reported
that fungic L-asparaginase utilization during food manufacturing leads to acrylamide
reduction without any negative impact on the final appearance and nutritional value
of the product. This approach mainly occurs from two aspects, interference with
Millard reactions and removal of L-asparagine as the main precursor of acrylamide
formation (Batool et al., 2016). However, the efficacy of asparaginase formation is
not sufficient for the industrial demands (FAO/WHO, 2002; Jha et al., 2012). Two
commercial samples of fungal asparaginase in the food industry, PreventAse (DSM)
and Acrylaway (Novozymes) are obtained from Aspergillus niger and Aspergillus
oryzae, respectively (Pedreschi et al., 2011; Xu et al., 2016; DSM, 2018).
PreventAse presents an optimum pH in the range of 4–5 and temperature at 50 C,
while these values for Acrylaway are 6–7 and 60 C. The activity of L-asparaginase
during food preparation and hydrolysis should be high, thus producing a stable
enzyme is of great importance.
The cultivation conditions including media composition, pH, temperature, oxy-
gen levels, and the fermentation technique, namely solid-state fermentation or
submerged fermentation, have considerable influence on the production of an ideal
enzyme (Brumano et al., 2019). Carbon and nitrogen are the most important
substrates in the culture medium used for L-asparaginase production (Cachumba
et al., 2016). Glucose is considered the most appropriate carbon source to achieve L-
asparaginase (Doriya & Kumar, 2016). L-Asparagine, L-proline, urea, yeast extract,
and peptone are some of the important sources of nitrogen for high-yield
asparaginase (de Moura Sarquis et al., 2004).
Determination of the optimum pH and temperature of the fermentation medium is
also an important step for producing high-yield enzymes. The emergence of
engineered enzymes according to the structural and mechanistic methods and direct
evolution is needed to enhance the efficiency of enzymes for the synthesis of various
products (Bornscheuer et al., 2012). Bakery products, French fries, and roasted
coffee are the most widely consumed products that have high acrylamide content.
Glucans are non-starch glucose polymers and one of the most abundant
polysaccharides found in the cell wall of fungi, plants, and bran cereal grains
(Wu et al., 2018). The glucose moieties link through either or both alpha (α) or
beta (β) linkages, so their structures can be either linear or branched, and microfi-
brillar or amorphous. α-1,3-Glucans sensu strictu (pseudonigerans) is known as the
most abundant α-glucans in the cell wall of fungi. Their structure is in the form of
microfibrils which make resistance to the cell wall. On the other hand, the structure
of β-glucans is a complex of linear or branched. In most cases, the structure is in the
form of microfibrils with mostly β-1,3-linear, and β-1,6-branched linkages. One such
example is a water-soluble low molecular weight polymers (LMW), which contains
β-1,2 besides β-1,6 joined branches and has been isolated from black yeast
226 I. Alemzadeh et al.
Table 9.2 The classification of β-glucans and β-glucanases enzymes from various sources
EC
β-Glucans type Structure Description number References
Bacterial Linear β-1,3-glucan Exo: Wu et al.
(Euglena EC (2018)
gracilis) 3.2.1.58
Endo:
EC
3.2.1.39
Fungal Branched β-1,6-branched, and Endo: Sutivisedsak
(Schizophyllum β-1,3-glucan EC et al. (2013)
commune) 3.2.1.6
Yeast Long branched β-1,6-branched and EC Sato et al.
β-1,3-glucan 3.2.1.39 (2012)
endo-β-1,3-glucanases EC
3.2.1.6
Cereal (barley) Linear β-1,3/1,4-glucan Endo Rodríguez-
3.2.1.6 Mendoza
et al. (2019)
Lichen Linear 1,3–1,4-β-glucan Endo You et al.
(lichenase, 3.2.1.73 (2016)
licheniase)
9.3.3 Chlorophyllase
Fig. 9.2 The initial steps of chlorophyll catabolism (CLH and MCS are refer to chlorophyllase and
magnesium-dechelating substance, respectively) (Hu et al., 2015)
9.3.4 L-Glutaminase
such as higher yields within a limited period of fermentation time. The enzymatic
biosynthesis of L-thiamine takes place in the presence of ethylamine and L-glutamine
as substrate and catalyst (glutaminase). L-Thiamine production was carried out under
solid-state fermentation by the fungal strain Trichoderma koningii. The enzymatic
synthetic reaction for theanine production is presented in Fig. 9.3.
9.3.5 Lipase
Lipases, also known as triacylglycerol acyl ester hydrolases (EC 3.1.1.3), are
capable to catalyze cleavage of carboxyl ester bonds in tri-, di-, and
monoacylglycerols. Most lipases are generally recognized as safe (GRAS) and can
be employed in the food industry, mainly for diary applications with the aim of
ripening acceleration in Italian-type cheeses, and hydrolysis of milk fat to further be
used as a flavor enhancer in cheese products (Afarin et al., 2018).
Further applications of lipases in pharmaceutical and fine chemical preparation
processes include (but are not limited to) lipase-mediated synthesis of optically pure
and optically active polymers, lipase-catalyzed resolution of racemic aqueous
mixtures, monoglycerides production (as an emulsifier in the food and cosmetic
products), fatty alcohols (as lubricants in hydrophobic waste treatment), and sugar
esters as biosurfactants (Tarahomjoo & Alemzadeh, 2003).
Another advantage of lipase incorporation in dairy products is fat and oil-tailoring
for enhancing functional and nutritional properties. One such example is employing
lipase in conjugated linoleic acid (CLA) large-scale production for the modification
of triglycerides and upgrading of low-value oils and fats like human milk fat
replacers, cocoa butter substitutes, and enrichment of nutraceutical fats with poly-
unsaturated fatty acids (Paiva et al., 2000; Yazdi & Alemzadeh, 2017). CLA is one
of the most physiological active fatty acids a natural derivative of linoleic acid, an
essential fatty acid, that possesses a conjugated double-bond system for describing
octadecadienoic acids mixtures (18:2) (Jiang et al., 1998; Lin, 2006; Gofferjé et al.,
2014).
Due to the global rising trend concerning diseases, such as obesity, hyperlipid-
emia, atherosclerosis, diabetes, cancer, and high blood pressure and the established
association of lifestyle on health management, the quality of dietary fat is an
important factor that should be considered for prevention and treatment of early
stage diseases. In this regard, conjugated fatty acids have gained much attention for
their potential health effects on lowering the risk of these diseases, such as
9 Enzymes in Functional Food Development 231
9.3.6 Phytase
(EC 3.1.3.72), and 6-phytases (EC 3.1.3.26) (Greiner & Konietzny, 2006). Enzy-
matic dephosphorylation of phytates was originally a strategy to improve the
nutritional value of feed by liberating phosphate, especially for mono-stomached
animals like fish, broilers, shrimp, pigs, and poultry diets (Konietzny & Greiner,
2004).
Recently, there have been numerous studies showing the potential use of these
enzymes for functional food development. One of the major concerns in phytate
hydrolysis content during food manufacturing is the selection of an ideal phytase.
The activity of added phytase has to be high enough during food preparation.
Therefore, a high capability of phytate degradation over a wide range of
temperatures and pH is essential (Greiner & Konietzny, 2006). According to previ-
ous studies the optimum pH range for acid phytase is 3.5–6.0, and for alkaline
phytase is 7.0–8.0. The optimum temperature for high phytate-degrading activity is
in the range of 35–80 C. The optimum temperature and pH of the added enzyme are
strongly affected by the source of phytase.
Phytase has been found in plants, microorganisms, and animal tissues. There are
already available commercial phytases from different microbial sources, such as
Escherichia coli, Peniophora lycii, Aspergillus niger, Schizosaccharomyces pombe.
Commercial production of phytase from Aspergillus, a soil fungus, has several
prominent drawbacks, such as fungal enzymes are catalytic efficiency, substrate
specificity, and resistance to proteolysis. Bacterial phytases have been distributed in
various aerobic and anaerobic species, including Bacillus strains, Pseudomonas
strains, Raoultella strains, Escherichia coli, Citrobacter braakii, Prevotella strains,
Selenomonas ruminantium, Mitsuokella jalaludinii, Megasphaera elsdenii, Lacto-
bacillus sanfranciscensis. The expression of phytase from these bacteria sources is
different and it can be regulated by the nature of the culture composition used for
growth and the nutrient or energy limitations. The level of inorganic phosphate on
the phytase synthesis induction was reported to be effective for all microbial sources,
except Raoultella terrigena and the rumen bacteria. In some phytase producers, the
nature of the carbon source is a significant inducible factor by involving in the
catabolism pathway (Konietzny & Greiner, 2004).
Generally, microbial-derived phytases are more favorable economically com-
pared to plant-derived ones, normally due to their thermostability and pH tolerance
as well as a broad specific activity. The specific activity of characterized phytasea, is
approximately in the range of 10–1000 U/mg at 37 C and various pH optimums
(Greiner & Konietzny, 2006). The highest specific activities are reported for
microbial-derived enzymes. In food applications, it is important that the enzymatic
properties of phytase to hydrolyze phytate in the gastric conditions be stable under
acidic pH and pepsin enzyme. Likewise, for food supplements, an enzyme with
acidic pH optimum and highly resistant to digestive enzymes is indisputably
attractive.
However, when exposed to pH values above pH ¼ 7.5 or below pH ¼ 4.0, most
of the plant enzymes dramatically lose their stability (Greiner & Konietzny, 2006).
Besides, the activation of these enzymes is irreversibly affected when exposed to
temperatures above 70 C within a few minutes. Despite the fact that most of the
234 I. Alemzadeh et al.
plant enzymes are inactivated at pH values lower or higher than the pH optima and
high temperatures, they are more acceptable among consumers as they are rarely
reported as an allergen.
The use of agro-industrial byproducts in the culture media has been suggested as
low-cost substrates for the production of phytase (Pires et al., 2019). Corn meal and
rice bran were used for the production of phytase from the fungus A. zeae B, and the
yeast K. marxianus. The activity of phytases from agro-industrial products strongly
depends on the species of the microorganism. For example, the determination of
total phytase activity in the mentioned study revealed tenfold higher values in the
yeast extraction compared to those of the fungus extraction (Pires et al., 2019).
Engineered phytases via genetic engineering are a promising type of enzyme that
show optimized catalytic features, thermal stability, and broad specific activity for
food application. Enhancement of thermal stability by a shift from 80 to 90 and
optimum temperature from 55 to 65 for Escherichia coli was achieved by using
mutagenesis technology. The basis of the phytase synthesis is to mutation of gene
site saturation by comparing the amino acid sequences derived from both the
available standard program and the homologous proteins (Rodriguez et al., 2000;
Garrett et al., 2004).
The incorporation of phytase in the food industry is a growing trend, mainly for
two purposes; increasing the mineral bioavailability of a given food by reducing its
phytate content and exerting nutritional health benefits in a supplemental product
(Konietzny & Greiner, 2004).
Natural phytate-degrading activity, observed in most plants, results in phytate
dephosphorylation during food processing such as soaking, germination, and fer-
mentation. This capability of intrinsic phytate-degrading activity in plant seeds
exhibits over a wide range of pH values from 3 to 10, and the maximum activity
peak occurs at pH ¼ 5–5.5 (Greiner & Konietzny, 2006). Therefore, phytate
hydrolysis by a certain phytase at optimum pH and temperature might differ greatly
among various plants and microorganisms. Some examples of optimum conditions
for maximum phytase activity are represented in Table 9.4.
9.3.7 Protease
Table 9.4 The effect of phytase sources on the specific activity and pH and temperature optimum for various applications
Specific pH Temperature optimum
Phytase source activity optimum ( C) Application References
Yersinia intermedia 3960 U/mg 4.5 55 Feed industry Huang et al. (2006)
Streptomyces sp. (NCIM – 2.5 70 Plant growth Puppala et al. (2019)
5533)
Sporotrichum thermophile 1190 U/mg 5.0 60 Dietary supplement Maurya et al. (2017)
Aspergillus aculeatus APF1 13.82 U/mg 3.0 50 Biofortified cereals Saxena et al. (2020)
Enterobacter sp. ACSS 805.98 U/mg 2.5 60 Dephytinizing animal feeds, baking Chanderman et al. (2016)
Enzymes in Functional Food Development
industry
Pichia anomala – 4.0 60 Fractionation of allergenic soya Joshi and Satyanarayana
proteins (2017)
Rhizopus oligosporus 31.3 U/gds 5.5 50 Food and feed industry Suresh and Radha (2016)
MTCC 556
Aspergillus niger NCIM 160 IU/mL 2.5 50 Organophosphorus pesticides Shah et al. (2017)
563 degradation
Penicillium oxalicum 12.8 U/g 7.0 40 Food industry Kaur (2017)
EUFR-3
Aspergillus ficuum 5.17 U/gds 6.1 37 Food industry Tian and Yuan (2016)
235
236 I. Alemzadeh et al.
A great variety of peptides are formed during cheese ripening which has been
shown to exert biological activities. Furthermore, secondary proteolysis during
cheese ripening may lead to the formation of bioactive peptides (Korhonen &
Pihlanto, 2006). The occurrence of the bioactive peptides which are naturally formed
in cheese depends on the equilibrium between their formation and the degradation of
the peptide during cheese ripening. Ripening is a time-consuming process during
cheese making. To overcome these problems, some economic and technological
methods for acceleration of cheese ripening have been reported (Fox, 1989). Accel-
eration of ripening may help to increase bioactive peptides. Encapsulation of
enzymes in liposome can accelerate cheese ripening (Jahadi et al., 2016;
Mohammadi et al., 2015; Zoghi et al., 2018). Vmax and Km and stability of the
enzyme were increased by encapsulation of the Flavourzyme in liposome
(Vafabakhsh et al., 2013; Jahadi et al., 2012, 2015, 2020; Jahadi & Khosravi-
Darani, 2017).
9 Enzymes in Functional Food Development 237
Gamma-amino butyric acid (GABA) is another amino acid that consists of four
carbons widely found in microorganisms, animals, and plants. GABA has been used
in pharmaceuticals and functional foods due to its blood pressure-lowering activity
and its role as a neurotransmitter signal in the central nervous system including the
mammalian brain (Takahashi et al., 2012). It can also be used for the synthesis of
nylon 4 as a monomer (Kim et al., 2007; Park et al., 2013). Its production has been
reported to occur by decarboxylation of glutamate, a reaction catalyzed by glutamate
decarboxylase (Pham et al., 2016).
The ever-increasing demand for GABA has generated the need for efficient
microbial production. Lactic acid bacteria (LAB) are one of the main microbial
sources for GABA production, mainly due to their food-grade property and high
ability in the direct production of GABA-rich components. The progress toward
culture conditions optimization and genetic engineering to achieve physiology-
oriented engineering approaches and co-culture methods for the GABA-producing
LAB species, the biosynthesis pathway of GABA by LAB, and the glutamate
decarboxylase (GAD), as the key enzyme in GABA biosynthesis, is growing rapidly
(Cui et al., 2020). The biosynthesis of GABA is known to exhibit superior perfor-
mance due to high product efficiency, mild reaction conditions, simple process, and
low environmental burden (Gavrilescu & Chisti, 2005; Alcalde et al., 2006;
Carvalho, 2017). Irreversible α-decarboxylation of L-glutamate is an important
pathway in GABA biosynthesis, which is catalyzed by the pyridoxal 50 -phosphate
(PLP)-dependent glutamate decarboxylase (GAD) (Xu et al., 2017).
It has been indicated that the enzymatic hydrolysis of collagen provides structural
cleavage and better functionality. Enzymatic hydrolysis of bovine collagen and beef
bone extract, which were undertaken by Flavourzyme® and Protamex®, respec-
tively, have shown better functional (antioxidant and antimicrobial activity) and
physicochemical properties of the final hydrolysates (Vidal et al., 2018).
Peptides have been proved to offer health-promoting benefits for hypertension, a
primary risk factor in cardiovascular disease. Short-chain peptides are frequently
favored compared to other protein resources like free amino acids combinations, due
to their enhanced absorption kinetics (Jahan-Mihan et al., 2011). Thus, the proteoly-
sis process to achieve short-chain peptides is of paramount importance. Angiotensin-
converting enzyme inhibitory (ACEI) peptides are among the most recognized
peptides. The proteolysis of ACEI peptides is necessary for their bioactivity and
bioaccessibility, as they are encrypted within the parent proteins (De Leo et al.,
2009).
Protein hydrolysis in peptides occurs both in vitro and in vivo. In vitro degrada-
tion takes place during food preparation processes by the addition of isolated or
microbial enzymes. The in vivo process occurs within the gut, in the presence of
digestive and microbial enzymes of the gut flora. Employing a blend of diverse
proteases and a step-by-step procedure should be carried out for consecutive prote-
olysis (Boutrou et al., 2013).
Recently, dietary supplements based on the protein hydrolysates have already
gained much attention in case of being unable to assimilate native proteins due to
specific illnesses, such as Crohn’s disease or pancreatitis (Posovszky, 2016). The
238 I. Alemzadeh et al.
9.3.8 Tannase
Table 9.5 Some examples of tannase production from various microorganisms and their related properties (tannase activity, pH, and temperature)
pH
optimal
Source Activity stability Substrate A half-life (t1/2) temperature Application Reference
Aspergillus 390.4 U/mL 6.0 Catechingallate 5.4 h at 60 C Green tea Shao et al.
niger rAntan1 3.0–8.0 0.5 h at 70 C extraction (2020)
Bacillus 2868.75 U/mg 5.0 Tamarind seed 4.5 h at 60 C Bacterial tannase Jana et al.
subtilis PAB2 3.0–8.0 production (2013)
Penicillium 34.7 U/mL 7.5 Keekar leaves (3% w/v) – Selwal and
atramentosum 32.8 U/mL 5.5 Amla leaves (2% w/v) Selwal (2012)
Pestalotiopsis 98.6 U/mL 6.9 Pomegranate seeds – Reges de
guepinii – Sena et al.
(2014)
Aspergillus 111.5 U/mL 7.0 Propyl gallate – – Liu et al.
niger Bde14 – (2018)
Enzymes in Functional Food Development
Aspergillus 28.5 U/mL 5–6 2% tannic acid 40–60 C (in the presence Sorghum and Cavalcanti
fumigates (B-cyclodextrin) – of B-cyclodextrin as an leather effluent et al. (2020)
CAS21 adjuvant) treatment
Propyl gallate
production
Talaromyces 32.18 U/gds 8.0 Acacia nilotica bark 60 C Fruit juice Aharwar and
verruculosus 4–8 detannification Parihar
(2019)
Bacillus 0.265 U/gds – Triphala (11.532 g) – – Selvaraj et al.
gottheilii – (2019)
M2S2
Aspergillus 21.94 4.0 1% tannic acid 30 C Industrial Cavalcanti
niger 31.89 4.0 30 C applications et al. (2017)
Aspergillus
fumigatus
241
(continued)
Table 9.5 (continued)
242
pH
optimal
Source Activity stability Substrate A half-life (t1/2) temperature Application Reference
Klebsiella 39.72 5.52 Indian gooseberry leaves 39.72 C – Kumar et al.
pneumoniae – (2016)
Serratia 42 U/mL – 2% tannic acid – Antibacterial Nsayef
marcescens – activity Muslim et al.
(2017)
Bacillus 49.32 U/L 4.74 4% tannic acid – Clarification of
gottheilii – acidic beverages
M2S2
Aspergillus 148.7 U/g – Rice bran and spent coffee ground – – Mansor et al.
niger PN1 116.52 – Coconut residue (2019)
88.64
Enterobacter 4 U/mL 6.0 Glucose 50 C – Govindarajan
cloacae – et al. (2019)
Aspergillus 12.26 U/g 5.5 Cashew bagasse 30 C Tannase Tatiana et al.
UCP1284 – production (2016)
Aspergillus 139.22 IU/mL 4.89 Pomegranate rind, Cassia 34.91 C – Varadharajan
oryzae – auriculata flower, black gram et al. (2017)
husk, and tea dust
I. Alemzadeh et al.
9 Enzymes in Functional Food Development 243
employed to enhance tannase activity and stability. The properties of the additive
compounds have a great influence on the properties of the final enzyme powder, like
glass transition temperature, density, particle size, and solubility. Therefore, the
adjuvants should be chosen considerably (Cavalcanti et al., 2020). The immobiliza-
tion method in multi-walled carbon nanotubes for tannase has increased tannase
stability (Ong & Annuar, 2018). Apart from the processing method, statistical tools,
mainly response surface methodology, have been considered for the optimization of
tannase production to reduce the cost and the time (Bhoite & Murthy, 2015).
Five main tannase applications are tea processing, fruit juices clarification and
de-bittering, animal feed treatment for nutritional improvement, gallic acid, and
propyl gallate production used in food, leather, dye, and chemical industries, and
wastewater treatment in agro-industries for decolorization and polyphenolic com-
pound reduction (Aharwar & Parihar, 2018; Cao et al., 2019).
9.4 Conclusion
In this chapter, we have reviewed some of the main enzymes incorporated in food
development, influencing the bioavailability and bioactivity of nutraceuticals.
Worldwide companies have been established to do high-tech research and develop-
ment of functional foods that have potentially positive effects on human health
beyond basic nutrition. However, scientists should bear in mind that in vitro and
in vivo experiments and clinical trials are essential steps that should be considered to
guarantee any health claims based on the major factors limiting their
bioaccessibility. According to traditional knowledge as well as scientific evidence,
enzymes have a dominant role in food biotechnology for the development and
commercialization of functional foods. Enzymes utilization in the production of
bioactive metabolites and development of bioprocesses has shown an increasing
trend. Several bioprocesses which involve enzymes have been established for the
development of commercially important nutraceuticals. Yet, many traditional
fermented foods have remained unexploited from technological aspects of functional
foods and nutraceuticals development. Enzymes, such as L-asparaginase,
chlorophyllase, beta-glucanase, L-glutaminae, lipase, and proteases are key enzymes
in nutraceutical and functional food production.
Exploring for high-efficient and cost-effective technologies should be concerned
in parallel with optimizing the experimental conditions to enhance the yield and
physiological stability of these bioactive compounds. Recently, food and agricultural
wastes have intensely attended as potential substrates to produce high-value
products, namely free and immobilized enzymes. They represent ideal sources of
proteins, lipids, and carbohydrates, and therefore, offer remarkable advantages from
economic and eco-sustainability aspects. Nevertheless, the large-scale applicability,
potential repeatability, and operation costs in real-industrial implementation are
some of the challenges that need to be considered more systematically. Another
emerging aspect of food enzyme application is the alteration of DNA encoding
native enzymes for the development of genetically modified enzymes. Although the
244 I. Alemzadeh et al.
References
Adebiyi, A. P., et al. (2009). Purification and characterization of antioxidative peptides derived from
rice bran protein hydrolysates. European Food Research and Technology, 228(4), 553–563.
Adefegha, S. A. (2018). Functional foods and nutraceuticals as dietary intervention in chronic
diseases; Novel perspectives for health promotion and disease prevention. Journal of Dietary
Supplements, 15(6), 977–1009.
Afarin, M., Alemzadeh, I., & Yazdi, Z. K. (2018). Conjugated linoleic acid production and
optimization via catalytic reaction method using safflower oil. International Journal of Engi-
neering, Transactions B: Applications, 31(8), 1166–1171.
Aharwar, A., & Parihar, D. K. (2018). Tannases: Production, properties, applications. Biocatalysis
and Agricultural Biotechnology, 15, 322–334.
Aharwar, A., & Parihar, D. K. (2019). Talaromyces verruculosus tannase production, characteriza-
tion and application in fruit juices detannification. Biocatalysis and Agricultural Biotechnology,
18, 101014.
Al Mijan, M., & Lim, B. O. (2018). Diets, functional foods, and nutraceuticals as alternative
therapies for inflammatory bowel disease: Present status and future trends. World Journal of
Gastroenterology, 24(25), 2673–2685.
Alam, S., et al. (2019) Recent development in the uses of asparaginase as food enzyme (pp. 55–81).
Alcalde, M., et al. (2006). Environmental biocatalysis: From remediation with enzymes to novel
green processes. Trends in Biotechnology, 24(6), 281–287.
Alemzadeh, I., et al. (2020). Encapsulation of food components and bioactive ingredients and
targeted release. International Journal of Engineering, Transactions A: Basics, 33(1), 1–11.
Alkhatib, A., et al. (2017). Functional foods and lifestyle approaches for diabetes prevention and
management. Nutrients, 9(12), 1310.
Ando, A. (2004). Conjugated linoleic acid production from castor oil by Lactobacillus plantarum
JCM 1551. Enzyme and Microbial Technology, 35, 40–45.
Arab, F., Alemzadeh, I., & Maghsoudi, V. (2011). Determination of antioxidant component and
activity of rice bran extract. Scientia Iranica, 18(6), 1402–1406.
Arshad, R., et al. (2019). Optimized production of tannase and gallic acid from fruit seeds by solid
state fermentation. Tropical Journal of Pharmaceutical Research, 18(5), 911–918.
Asselin, B., & Rizzari, C. (2015). Asparaginase pharmacokinetics and implications of therapeutic
drug monitoring. Leukemia and Lymphoma, 56(8), 2273–2280.
Baskar, G., Aiswarya, R., & Renganathan, S. (2019). Applications of asparaginase in food
processing. In Green bio-processes (pp. 83–98). Springer.
9 Enzymes in Functional Food Development 245
Batool, T., et al. (2016). A comprehensive review on L-asparaginase and its applications. Applied
Biochemistry and Biotechnology, 178(5), 900–923.
Berin, M. C. (2015). Immunopathophysiology of food protein-induced enterocolitis syndrome.
Journal of Allergy and Clinical Immunology, 135(5), 1108–1113.
Bhoite, R. N., & Murthy, P. S. (2015). Biodegradation of coffee pulp tannin by Penicillium
verrucosum for production of tannase, statistical optimization and its application. Food and
Bioproducts Processing, 94, 727–735.
Bornscheuer, U. T., et al. (2012). Engineering the third wave of biocatalysis. Nature, 485(7397),
185–194.
Boutrou, R., et al. (2013). Sequential release of milk protein-derived bioactive peptides in the
jejunum in healthy humans. American Journal of Clinical Nutrition, 97(6), 1314–1323.
Brumano, L. P., et al. (2019). Development of L-asparaginase biobetters: Current research status
and review of the desirable quality profiles. Frontiers in Bioengineering and Biotechnology,
6(January), 212.
Burton, S. G., Cowan, D. A., & Woodley, J. M. (2002). The search for the ideal biocatalyst. Nature
Biotechnology, 20(1), 37–45. https://doi.org/10.1038/nbt0102-37
Cachumba, J. J. M., et al. (2016). Current applications and different approaches for microbial
L-asparaginase production. Brazilian Journal of Microbiology, 47, 77–85.
Cao, Q. Q., et al. (2019). Improving the taste of autumn green tea with tannase. Food Chemistry,
277, 432–437.
Carle, R., & Schweiggert, R. (Eds.). (2016). Handbook on natural pigments in food and beverages:
Industrial applications for improving food color. Woodhead Publishing.
Carvalho, C. C. C. R. (2017). Whole cell biocatalysts: Essential workers from nature to the industry.
Microbial Biotechnology, 10(2), 250–263.
Cavalcanti, R. M. F., et al. (2017). Screening, selection and optimization of the culture conditions
for tannase production by endophytic fungi isolated from Caatinga. Journal of Applied Biology
& Biotechnology, 5(1), 1–9.
Cavalcanti, R. M. F., et al. (2020). Stabilization and application of spray-dried tannase from
Aspergillus fumigatus CAS21 in the presence of different carriers. 3 Biotech, 10(4), 1–14.
Chanderman, A., et al. (2016). Production, characteristics and applications of phytase from a
rhizosphere isolated Enterobacter sp. ACSS. Bioprocess and Biosystems Engineering, 39(10),
1577–1587.
Chu, H. M., et al. (2004). Structures of Selenomonas ruminantium phytase in complex with
persulfated phytate: DSP phytase fold and mechanism for sequential substrate hydrolysis.
Structure, 12(11), 2015–2024.
Chung, K. T., Wei, C. I., & Johnson, M. G. (1998). Are tannins a double-edged sword in biology
and health? Trends in Food Science and Technology, 9(4), 168–175.
Cui, Y., et al. (2020). Production of gamma-aminobutyric acid from lactic acid bacteria: A
systematic review. International Journal of Molecular Sciences, 21(3), 995.
De Leo, F., et al. (2009). Angiotensin converting enzyme (ACE) inhibitory peptides: Production
and implementation of functional food. Current Pharmaceutical Design, 15(31), 3622–3643.
de Moura Sarquis, M. I., et al. (2004). Production of L-asparaginase by filamentous fungi.
Memórias do Instituto Oswaldo Cruz, 99(5), 489–492.
de Paulo Farias, D., et al. (2019). Prebiotics: Trends in food, health and technological applications.
Trends in Food Science and Technology, 93, 23–35.
Dersjant-Li, Y., et al. (2015). Phytase in non-ruminant animal nutrition: A critical review on phytase
activities in the gastrointestinal tract and influencing factors. Journal of the Science of Food and
Agriculture, 95(5), 878–896.
Dewapriya, P., & Kim, S. K. (2014). Marine microorganisms: An emerging avenue in modern
nutraceuticals and functional foods. Food Research International, 56, 115–125.
Dewi, R. T. K., Mubarik, N. R., & Suhartono, M. T. (2016). Medium optimization of β-glucanase
production by Bacillus subtilis SAHA 32.6 used as biological control of oil palm pathogen.
Emirates Journal of Food and Agriculture, 28(2), 116–125.
246 I. Alemzadeh et al.
Doriya, K., & Kumar, D. S. (2016). Isolation and screening of l-asparaginase free of glutaminase
and urease from fungal sp. 3 Biotech, 6(2), 239.
DSM. (2018). DSM introduces PreventASe® XR to extend use of enzymatic acrylamide-reduction
solution to more snacks and baked goods. Retrieved from https://www.dsm.com/corporate/
media/informationcenter-news/2018/07/2018-07-10-dsm-introduces-preventase-xr-to-extend-
use-of-enzymatic-acrylamide-reduction-solution-to-more-snacks-and-baked-goods1.html
Ferruzzi, M. G., & Blakeslee, J. (2007). Digestion absorption and cancer preventative activity of
dietary chlorophyll derivatives. Nutrition Research, 27(1), 1–12. https://doi.org/10.1016/j.
nutres.2006.12.003
Filella, I., Serrano, L., Serra, J., & Peñuelas, J. (1995). Evaluating wheat nitrogen status with canopy
reflectance indices and discriminant analysis. Crop Science, 35(5), 1400–1405. https://doi.org/
10.2135/cropsci1995.0011183X003500050023x
Fox, P. F. (1989). Proteolysis during cheese manufacture and ripening. Journal of Dairy Science,
72(6), 1379–1400.
Gandul-Rojas, B., & Mínguez-Mosquera, M. I. (1996). Chlorophyllase activity in olive fruits and
its relationship with the loss of chlorophyll pigments in the fruits and oils. Journal of the Science
of Food and Agriculture, 72(3), 291–294.
García-Conesa, M. T., et al. (2001). Hydrolysis of diethyl diferulates by a tannase from Aspergillus
oryzae. Carbohydrate Polymers, 44(4), 319–324.
Garrett, J. B., et al. (2004). Enhancing the thermal tolerance and gastric performance of a microbial
phytase for use as a phosphate-mobilizing monogastric-feed supplement. Applied and Environ-
mental Microbiology, 70(5), 3041–3046.
Gavrilescu, M., & Chisti, Y. (2005). Biotechnology—A sustainable alternative for chemical
industry. Biotechnology Advances, 23(7–8), 471–499.
Georgetti, S. R., et al. (2009). Enhanced in vitro and in vivo antioxidant activity and mobilization of
free phenolic compounds of soybean flour fermented with different β-glucosidase-producing
fungi. Journal of Applied Microbiology, 106(2), 459–466.
Giese, E. C., Dekker, R. F. H., Barbosa, A., & da Silva, M. (2012). Production of β-(1,3)-glucanases
by Trichoderma harzianum Rifai: Optimization and application to produce gluco-
oligosaccharides from paramylon and pustulan. Fermentation Technology, 1(1), 1–5.
Gligor, O., et al. (2019). Enzyme-assisted extractions of polyphenols—A comprehensive review.
Trends in Food Science and Technology, 88, 302–315.
Global Medical Asparaginase Sales Market Report 2018. (2018). Retrieved from https://www.
themarketreports.com/report/global-medical-asparaginase-sales-market-report-2018
Gofferjé, G., et al. (2014). Kinetics of enzymatic esterification of glycerol and free fatty acids in
crude Jatropha oil by immobilized lipase from Rhizomucor miehei. Journal of Molecular
Catalysis B: Enzymatic, 107, 1–7.
Govindarajan, R. K., et al. (2019). Purification, structural characterization and biotechnological
potential of tannase enzyme produced by Enterobacter cloacae strain 41. Process Biochemistry,
77, 37–47.
Granato, D., et al. (2020). Functional foods: Product development, technological trends, efficacy
testing, and safety. Annual Review of Food Science and Technology, 11(1), 93–118.
Greiner, R., & Konietzny, U. (2006). Phytase for food application. Food Technology and Biotech-
nology, 44(2), 125–140.
Grom, L. C., et al. (2020). Probiotic dairy foods and postprandial glycemia: A mini-review. Trends
in Food Science and Technology, 101, 165–171.
Guan, H., et al. (2018). The enzymatic hydrolysis of soy protein isolate by Corolase PP under high
hydrostatic pressure and its effect on bioactivity and characteristics of hydrolysates. Food
Chemistry, 245, 89–96.
Gul, K., Singh, A. K., & Jabeen, R. (2016). Nutraceuticals and functional foods: The foods for the
future world. Critical Reviews in Food Science and Nutrition, 56(16), 2617–2627.
Handa, V., et al. (2020). Biotechnological applications of microbial phytase and phytic acid in food
and feed industries. Biocatalysis and Agricultural Biotechnology, 25, 101600.
9 Enzymes in Functional Food Development 247
Haskell, C. F., Kennedy, D. O., Milne, A. L., Wesnes, K. A., & Scholey, A. B. (2008). The effects
of L-theanine caffeine and their combination on cognition and mood. Biological Psychology, 77
(2), 113–122. https://doi.org/10.1016/j.biopsycho.2007.09.008
Hendriksen, H. V., Puder, K., & Goldbech Olsen, A. (2016). Method for reducing the level of
asparagine in a food material. U.S. Patent Application 14/781,760.
Hennessy, A. A., et al. (2012). The production of conjugated α-linolenic, γ-linolenic and stearidonic
acids by strains of bifidobacteria and propionibacteria. Lipids, 47(3), 313–327.
Holm, L., et al. (2017). Contraction mode and whey protein intake affect the synthesis rate of
intramuscular connective tissue. Muscle and Nerve, 55(1), 128–130.
Hosikian, A., Lim, S., Halim, R., & Danquah, M. K. (2010). Chlorophyll extraction from
microalgae: A review on the process engineering aspects. International Journal of Chemical
Engineering, 1–11. https://doi.org/10.1155/2010/391632
Hörtensteiner, S., & Kräutler, B. (2011). Chlorophyll breakdown in higher plants. Biochimica et
Biophysica Acta (BBA)-Bioenergetics, 1807(8), 977–988. https://doi.org/10.1016/j.bbabio.
2010.12.007
Hu, X., et al. (2015). Reexamination of chlorophyllase function implies its involvement in defense
against chewing herbivores. Plant Physiology, 167(3), 660–670.
Huang, H., et al. (2006). A novel phytase with preferable characteristics from Yersinia intermedia.
Biochemical and Biophysical Research Communications, 350(4), 884–889.
Ire, F. S., & Nwanguma, A. C. (2020). Comparative evaluation on tannase production by
Lasiodiplodia plurivora ACN-10 under submerged fermentation (SmF) and solid state fermen-
tation (SSF). Asian Journal of Biotechnology and Bioresource Technology, 39–49. https://doi.
org/10.9734/ajb2t/2020/v6i130074
Jahadi, M., & Khosravi-Darani, K. (2017). Liposomal encapsulation enzymes: From medical
applications to kinetic characteristics. Mini-Reviews in Medicinal Chemistry, 17, 366–370.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., Mozafar, M. R., Saboury, A. A., & Pourhosseini,
P. S. (2015). The encapsulation of flavourzyme in nanoliposome by heating method. Journal of
Food Science and Technology, 52(4), 2063–2072.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., Mozafari, M. R., Saboury, A. A., Zoghi, A., &
Mohammadi, M. (2016). Modeling of proteolysis in Iranian brined cheese using proteinase-
loaded nanoliposome. International Journal of Dairy Technology, 69(1), 57–62.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., et al. (2012). Evaluating the effects of process
variables on protease-loaded nano-liposome production by Plackett-Burman design for utilizing
in cheese ripening acceleration. Asian Journal of Chemistry, 24(9), 3891–3894.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., et al. (2020). Accelerating ripening of Iranian white
brined cheesesusing liposome-encapsulated and free proteinases. Biointerface Research in
Applied Chemistry, 10(1), 4966–4971.
Jahan-Mihan, A., et al. (2011). Dietary proteins as determinants of metabolic and physiologic
functions of the gastrointestinal tract. Nutrients, 3(5), 574–603.
Jana, A., et al. (2013). Structural characterization of thermostable, solvent tolerant, cytosafe tannase
from Bacillus subtilis PAB2. Biochemical Engineering Journal, 77, 161–170.
JECFA. (2002). Evaluation of certain food additives. Fifty-ninth report of the joint FAO/WHO
expert committee on food additives. World Health Organization technical report series, 913.
Jha, S. K., et al. (2012). Microbial L-asparaginase: A review on current scenario and future
prospects. International Journal of Pharmaceutical Sciences and Research, 3(9), 3076–3090.
Jiang, J., Björck, L., & Fondén, R. (1998). Production of conjugated linoleic acid by dairy starter
cultures. Journal of Applied Microbiology, 85(1), 95–102.
Joshi, S., & Satyanarayana, T. (2017). Characteristics and multifarious potential applications of
HAP phytase of the unconventional yeast Pichia anomala. In Developments in fungal biology
and applied mycology (pp. 265–278). Springer.
Kaur, R. (2017). Production and characterization of a neutral phytase of Penicillium oxalicum
EUFR-3 isolated from Himalayan region. Nusantara Bioscience, 9(1), 68–76.
248 I. Alemzadeh et al.
Khanbabaee, K., & van Ree, T. (2001). Tannins: Classification and definition. Natural Product
Reports, 18(6), 641–649.
Kim, S. H., et al. (2007). Cloning and expression of a full-length glutamate decarboxylase gene
from Lactobacillus brevis BH2. Biotechnology and Bioprocess Engineering, 12(6), 707–712.
Konietzny, U., & Greiner, R. (2004). Bacterial phytase: Potential application, in vivo function and
regulation of its synthesis. Brazilian Journal of Microbiology, 35(1–2), 11–18.
Korhonen, H., & Pihlanto, A. (2006). Bioactive peptides: Production and functionality. Interna-
tional Dairy Journal, 16, 945–960.
Koseki, T., et al. (2018). Characterization of a novel Aspergillus oryzae tannase expressed in Pichia
pastoris. Journal of Bioscience and Bioengineering, 126(5), 553–558.
Kouchak Yazdi, Z., & Alemzadeh, I. (2016a). Lipase catalyzed incorporation of conjugated linoleic
acid by transesterification into sunflower oil applying immobilized lipase (lipozyme
thermomyces lanuginosus and rhizomucor mehei). International Journal of Engineering,
Transactions A: Basics, 29(4), 436–443.
Kouchak Yazdi, Z., & Alemzadeh, I. (2016b). Lipase catalyzed incorporation of conjugated linoleic
acid by transesterification into sunflower oil applying immobilized lipase (Lipozyme TL im and
im RM). International Journal of Engineering, Transactions A: Basics, 29(4), 549–556.
Kouchak Yazdi, Z., Alemzadeh, I., & Vossoughi, M. (2017). Comparison and optimization of
conjugated linoleic acid production by Lactobacillus plantarum and Lactobacillus plantarum
subsp. plantarum. Scientia Iranica, 24(3), 1272–1280.
Kuddus, M. (2018). Enzymes in Food Biotechnology: Production, Applications, and Future
Prospects. Academic Press.
Kumar, D., et al. (2020). Barley grain beta glucan enrichment: Status and opportunities. In Wheat
and barley grain biofortification (pp. 295–308). Woodhead Publishing.
Kumar, M., et al. (2016). Improved production of tannase by Klebsiella pneumoniae using Indian
gooseberry leaves under submerged fermentation using Taguchi approach. AMB Express,
6(1), 46.
Lee, S. O., et al. (2003). Bioconversion of linoleic acid into conjugated linoleic acid during
fermentation and by washed cells of Lactobacillus reuteri. Biotechnology Letters, 25(12),
935–938.
Lin, T. Y. (2006). Conjugated linoleic acid production by cells and enzyme extract of Lactobacillus
delbrueckii ssp. bulgaricus with additions of different fatty acids. Food Chemistry, 94(3),
437–441.
Liu, F., et al. (2018). High level expression and characterization of tannase tan7 using Aspergillus
niger SH-2 with low-background endogenous secretory proteins as the host. Protein Expression
and Purification, 144, 71–75.
Lopez-Casado, G., et al. (2008). Plant glycosyl hydrolases and biofuels: A natural marriage.
Current Opinion in Plant Biology, 11(3), 329–337.
Mansor, A., et al. (2019). Evaluation of selected agri-industrial residues as potential substrates for
enhanced tannase production via solid-state fermentation. Biocatalysis and Agricultural Bio-
technology, 20, 101216.
Maqsood, S., et al. (2020). Bioactive compounds from date fruit and seed as potential nutraceutical
and functional food ingredients. Food Chemistry, 308, 125522.
Maurya, A. K., Parashar, D., & Satyanarayana, T. (2017). Bioprocess for the production of
recombinant HAP phytase of the thermophilic mold Sporotrichum thermophile and its structural
and biochemical characteristics. International Journal of Biological Macromolecules, 94,
36–44.
Meinlschmidt, P., Ueberham, E., Lehmann, J., Reineke, K., Schlüter, O., Schweiggert-Weisz, U., &
Eisner, P. (2016). The effects of pulsed ultraviolet light cold atmospheric pressure plasma and
gamma-irradiation on the immunoreactivity of soy protein isolate. Innovative Food Science &
Emerging Technologies, 38, 374–383. https://doi.org/10.1016/j.ifset.2016.06.007
Melini, F., et al. (2019). Health-promoting components in fermented foods: An up-to-date system-
atic review. Nutrients, 11(5), 1189.
9 Enzymes in Functional Food Development 249
Mitchell, L., et al. (1994). Increased endogenous thrombin generation in children with acute
lymphoblastic leukemia: Risk of thrombotic complications in L-sparaginase-induced antithrom-
bin III deficiency. Blood, 83(2), 386–391.
Mohammadi, R., Mahmoudzade, M., Atefi, M., Khosravi-Darani, K., & Mozafari, M. R. (2015).
Applications of nanoliposomes in cheese technology. International Journal of Dairy Technol-
ogy, 68(1), 11–23.
Mohanty, D. P., Mohapatra, S., Misra, S., & Sahu, P. S. (2016). Milk derived bioactive peptides and
their impact on human health—A review. Saudi Journal of Biological Sciences, 23, 577–583.
Momeni, V., Alemzadeh, I., & Vosoughi, M. (2015). Enhancement of L-asparaginase production
by candida utilis in a 13L fermenter and its purification. International Journal of Engineering,
Transactions B: Applications, 28(8), 1133–1141.
Niezgoda, N., & Gliszczyńska, A. (2019). Lipase catalyzed acidolysis for efficient synthesis of
phospholipids enriched with isomerically pure cis-9, trans-11 and trans-10, cis-12 conjugated
linoleic acid. Catalysts, 9(12), 1012.
Nsayef Muslim, D. S., Abbas Dham, Z., & Mohammed, D. N. J. (2017). Synthesis and characteri-
zation of nanoparticles conjugated tannase and using it for enhancement of antibacterial activity
of tannase produced by Serratia marcescens. Microbial Pathogenesis, 110, 484–493.
Ogawa, J., et al. (2005). Production of conjugated fatty acids by lactic acid bacteria. Journal of
Bioscience and Bioengineering, 100(4), 355–364.
Ong, C. B., & Annuar, M. S. M. (2018). Immobilization of cross-linked tannase enzyme on
multiwalled carbon nanotubes and its catalytic behavior. Preparative Biochemistry and Bio-
technology, 48(2), 181–187.
Paiva, A. L., Balcão, V. M., & Malcata, F. X. (2000). Kinetics and mechanisms of reactions
catalyzed by immobilized lipases. Enzyme and Microbial Technology, 27(3–5), 187–204.
Panda, R., et al. (2015). Enzymatic hydrolysis does not reduce the biological reactivity of soybean
proteins for all allergic subjects. Journal of Agricultural and Food Chemistry, 63(43),
9629–9639.
Pandey, A., et al. (1999). Fermentation for the production of industrial enzymes. Fermentation
Science and Technology, 77(1), 1–14.
Panesar, P. S., Kaur, R., Singla, G., & Sangwan, R. S. (2016). Bioprocessing of agro-industrial
wastes for production of foodgrade enzymes: Progress and prospects. Applied Food Biotech-
nology, 3(4), 208–227.
Park, S. J., et al. (2013). Synthesis of nylon 4 from gamma-aminobutyrate (GABA) produced by
recombinant Escherichia coli. Bioprocess and Biosystems Engineering, 36(7), 885–892.
Patel, S., & Rauf, A. (2017). Edible seeds from Cucurbitaceae family as potential functional foods:
Immense promises, few concerns. Biomedicine and Pharmacotherapy, 91, 330–337.
Patro, K. R., et al. (2014). Development of new medium composition for enhanced production of
L-asparaginase by Aspergillus flavus. Journal of Environmental Biology, 35(1), 295–300.
Pedreschi, F., et al. (2011). Acrylamide reduction in potato chips by using commercial asparaginase
in combination with conventional blanching. LWT—Food Science and Technology, 44(6),
1473–1476.
Pham, V. D., et al. (2016). Engineering the intracellular metabolism of Escherichia coli to produce
gamma-aminobutyric acid by co-localization of GABA shunt enzymes. Biotechnology Letters,
38(2), 321–327.
Phelan, M., Aherne, A., FitzGerald, R. J., & O’Brien, N. M. (2009). Casein-derived bioactive
peptides: Biological effects, industrial uses, safety aspects and regulatory status. International
Dairy Journal, 19, 643–654.
Philippaerts, A., Van Aelst, J., & Sels, B. (2013). Conjugated linoleic acids and conjugated
vegetable oils: From nutraceutical to bio-polymer. European Journal of Lipid Science and
Technology, 115(7), 717–720.
Pires, E. B. E., et al. (2019). Production of fungal phytases from agroindustrial byproducts for pig
diets. Scientific Reports, 9(1), 1–9.
250 I. Alemzadeh et al.
Pokora, M., et al. (2013). Biological and functional properties of proteolytic enzyme-modified egg
protein by-products. Food Science & Nutrition, 1(2), 184–195.
Poli, A., et al. (2018). Nutraceuticals and functional foods for the control of plasma cholesterol
levels. An intersociety position paper. Pharmacological Research, 134, 51–60.
Posovszky, C. (2016). Congenital intestinal diarrhoeal diseases: A diagnostic and therapeutic
challenge. Best Practice and Research: Clinical Gastroenterology, 30(2), 187–211.
Puppala, K. R., et al. (2019). Characterization of novel acidic and thermostable phytase secreting
Streptomyces sp. (NCIM 5533) for plant growth promoting characteristics. Biocatalysis and
Agricultural Biotechnology, 18, 101020.
Rai, A. K., Pandey, A., & Sahoo, D. (2019). Biotechnological potential of yeasts in functional food
industry. Trends in Food Science and Technology, 83, 129–137.
Reges de Sena, A., et al. (2014). Production, characterization and application of a thermostable
tannase from Pestalotiopsis guepinii URM 7114. Food Technology and Biotechnology, 52(4),
459–467.
Rittig, N., et al. (2017). Anabolic effects of leucine-rich whey protein, carbohydrate, and soy protein
with and without β-hydroxy-β-methylbutyrate (HMB) during fasting-induced catabolism: A
human randomized crossover trial. Clinical Nutrition, 36(3), 697–705.
Rizzari, C., et al. (2000). L-asparagine depletion and L-asparaginase activity in children with acute
lymphoblastic leukemia receiving i.m. or i.v. Erwinia C. or E. coli L-asparaginase as first
exposure. Annals of Oncology, 11(2), 189–193.
Rizzello, C. G., De Angelis, M., Di Cagno, R., Camarca, A., Silano, M., Losito, I., De Vincenzi, M.,
De Bari, M. D., Palmisano, F., Maurano, F., Gianfrani, C., & Gobbetti, M. (2007). Highly
efficient gluten degradation by lactobacilli and fungal proteases during food processing: New
perspectives for celiac disease. Applied and Environmental Microbiology, 73(14), 4499–4507.
https://doi.org/10.1128/AEM.00260-07
Rodriguez, E., et al. (2000). Site-directed mutagenesis improves catalytic efficiency and thermosta-
bility of Escherichia coli pH 2.5 acid phosphatase/phytase expressed in Pichia pastoris.
Archives of Biochemistry and Biophysics, 382(1), 105–112.
Rodríguez-Mendoza, J., et al. (2019). Purification and biochemical characterization of a novel
thermophilic exo-β-1,3-glucanase from the thermophile biomass-degrading fungus Thielavia
terrestris Co3Bag1. Electronic Journal of Biotechnology, 41, 60–71.
Ruiz-Herrera, J., & Ortiz-Castellanos, L. (2019). Cell wall glucans of fungi. A review. Cell Surface,
5, 100022.
Sahib, N. G., et al. (2013). Coriander (Coriandrum sativum L.): A potential source of high-value
components for functional foods and nutraceuticals—A review. Phytotherapy Research, 27(10),
1439–1456.
Sakhaei, M., & Alemzadeh, I. (2017). Enzymatic synthesis of theanine in the presence of
L-glutaminase produced by Trichoderma koningii. Applied Food Biotechnology, 4(2), 113–121.
Sato, H., et al. (2012). Inhibitory effects of water-soluble low-molecular-weight β-(1,3-1,6)
D-glucan isolated from aureobasidium pullulans 1A1 strain black yeast on mast cell degranula-
tion and passive cutaneous anaphylaxis. Bioscience, Biotechnology and Biochemistry, 76(1),
84–88.
Saxena, A., et al. (2020). Characteristics of an acidic phytase from Aspergillus aculeatus APF1 for
dephytinization of biofortified wheat genotypes. Applied Biochemistry and Biotechnology,
191(2), 679–694.
Schelbert, S., Aubry, S., Burla, B., Agne, B., Kessler, F., Krupinska, K., & Hörtensteiner, S. (2009).
The Plant Cell, 21(3), 767–785. https://doi.org/10.1105/tpc.108.064089
Selvaraj, S., et al. (2019). Modeling and optimization of tannase production with Triphala in packed
bed reactor by response surface methodology, genetic algorithm, and artificial neural network. 3
Biotech, 9(7), 259.
Selwal, M. K., & Selwal, K. K. (2012). High-level tannase production by Penicillium
atramentosum KM using agro residues under submerged fermentation. Annals of Microbiology,
62(1), 139–148.
9 Enzymes in Functional Food Development 251
Shah, P. C., et al. (2017). Phytase production by Aspergillus niger NCIM 563 for a novel
application to degrade organophosphorus pesticides. AMB Express, 7(1), 1–11.
Shao, Y., et al. (2020). Thermostable tannase from aspergillus Niger and its application in the
enzymatic extraction of green tea. Molecules, 25(4), 952.
Sharafi, E., Dehestani, A., & Farmani, J. (2017). Bioinformatics evaluation of plant chlorophyllase,
the key enzyme in chlorophyll degradation, 4(3), 167–678.
Shi, R., et al. (2017). Biochemical characterization of a novel L-asparaginase from Paenibacillus
barengoltzii being suitable for acrylamide reduction in potato chips and mooncakes. Interna-
tional Journal of Biological Macromolecules, 96, 93–99.
Shoji, T., et al. (2006). Apple procyanidin oligomers absorption in rats after oral administration:
Analysis of procyanidins in plasma using the porter method and high-performance liquid
chromatography/tandem mass spectrometry. Journal of Agricultural and Food Chemistry,
54(3), 884–892.
Sicherer, S. H., & Sampson, H. A. (2014). Food allergy: Epidemiology pathogenesis diagnosis and
treatment. Journal of Allergy and Clinical Immunology, 133(2), 291–307.e5. https://doi.org/10.
1016/j.jaci.2013.11.020
Sirtori, C. R., et al. (2017). Nutraceutical approaches to metabolic syndrome. Annals of Medicine,
49(8), 678–697.
Song, H. Y., El Sheikha, A. F., & Hu, D. M. (2019). The positive impacts of microbial phytase on
its nutritional applications. Trends in Food Science and Technology, 86, 553–562.
Srivastava, A., & Kar, R. (2009). Characterization and application of tannase produced by Asper-
gillus niger ITCC 6514.07 on pomegranate rind. Brazilian Journal of Microbiology, 40(4),
782–789.
Suleria, H. A. R., et al. (2015). Marine-based nutraceuticals: An innovative trend in the food and
supplement industries. Marine Drugs, 13(10), 6336–6351.
Suresh, S., & Radha, K. V. (2016). Statistical optimization and mutagenesis for high level of
phytase production by Rhizopus oligosporus MTCC 556 under solid state fermentation. Journal
of Environmental Biology, 37(2), 253–259.
Sutivisedsak, N., et al. (2013). Novel sources of β-glucanase for the enzymatic degradation of
schizophyllan. Enzyme and Microbial Technology, 52(3), 203–210.
Takahashi, C., et al. (2012). Robust production of gamma-amino butyric acid using recombinant
Corynebacterium glutamicum expressing glutamate decarboxylase from Escherichia coli.
Enzyme and Microbial Technology, 51(3), 171–176.
Tanna, B., & Mishra, A. (2018). Metabolites unravel nutraceutical potential of edible seaweeds: An
emerging source of functional food. Comprehensive Reviews in Food Science and Food Safety,
17(6), 1613–1624.
Tarahomjoo, S., & Alemzadeh, I. (2003). Surfactant production by an enzymatic method. Enzyme
and Microbial Technology, 33(1), 33–37.
Tatiana, P. S. L. L., et al. (2016). Tannase production by Aspergillus spp. UCP1284 using cashew
bagasse under solid state fermentation. African Journal of Microbiology Research, 10(16),
565–571.
Tavano, O. L., et al. (2018). Biotechnological applications of proteases in food technology.
Comprehensive Reviews in Food Science and Food Safety, 17(2), 412–436.
Tian, M., & Yuan, Q. (2016). Optimization of phytase production from potato waste using
Aspergillus ficuum. 3 Biotech, 6(2), 256.
Torang, A., & Alemzadeh, I. (2016). Acrylamide reduction in potato crisps using: Asparaginase
from Candida utilis, commercial asparaginase, salt immersion, and pH treatment. International
Journal of Engineering, 29(7), 879–886.
Vafabakhsh, Z., Khosravi-Darani, K., Khajeh, K., Jahadi, M., Komeili, R., & Mortazavian, A. M.
(2013). Stability and catalytic kinetics of protease loaded liposomes. Biochemical Engineering
Journal, 72, 11–17.
252 I. Alemzadeh et al.
Varadharajan, V., et al. (2017). Modeling and verification of process parameters for the production
of tannase by Aspergillus oryzae under submerged fermentation using agro-wastes. Biotechnol-
ogy and Applied Biochemistry, 64(1), 100–109.
Vaziri, A. S., Alemzadeh, I., & Vossoughi, M. (2019). Survivability and oxidative stability of
co-microencapsulated L. plantarum PTCC 1058 and DHA as a juice carrier. Food Bioscience,
32, 100460.
Venkatakrishnan, K., Chiu, H. F., & Wang, C. K. (2020). Impact of functional foods and
nutraceuticals on high blood pressure with a special focus on meta-analysis: Review from a
public health perspective. Food and Function, 11(4), 2792–2804.
Vidal, A. R., et al. (2018). Effects of enzymatic hydrolysis (Flavourzyme®) assisted by ultrasound
in the structural and functional properties of hydrolyzates from different bovine collagens. Food
Science and Technology, 38, 103–108.
Wu, Q., et al. (2018). Isolation of β-1,3-glucanase-producing microorganisms from poria cocos
cultivation soil via molecular biology. Molecules, 23(7), 1555.
Xu, F., Oruna-Concha, M. J., & Elmore, J. S. (2016). The use of asparaginase to reduce acrylamide
levels in cooked food. Food Chemistry, 210, 163–171.
Xu, N., Wei, L., & Liu, J. (2017). Biotechnological advances and perspectives of gamma-
aminobutyric acid production. World Journal of Microbiology and Biotechnology, 33(3), 64.
Yang, L., et al. (2002). Production of conjugated linoleic acids through KOH-catalyzed dehydration
of ricinoleic acid. Chemistry and Physics of Lipids, 119(1–2), 23–31.
Yazdi, Z. K., & Alemzadeh, I. (2017). Kinetic mechanism of conjugated linoleic acid esterification
and production of enriched glycerides as functional oil. Canadian Journal of Chemical Engi-
neering, 95(11), 2078–2086.
You, S., et al. (2016). Improvement of the thermostability and catalytic efficiency of a highly active
β-glucanase from Talaromyces leycettanus JCM12802 by optimizing residual charge-charge
interactions. Biotechnology for Biofuels, 9(1), 1–12.
Yuan, J., Jiang, B., Li, K., Shen, W., & Tang, J. L. (2017). Beneficial effects of protein hydrolysates
in exercise and sports nutrition. Journal of Biological Regulators and Homeostatic Agents,
31(1), 183–188.
Zhao, H. W., Lv, J. P., & Li, S. R. (2011). Production of conjugated linoleic acid by whole-cell of
Lactobacillus plantarum A6-1F. Biotechnology and Biotechnological Equipment, 25(1),
2266–2272.
Zoghi, A., Khosravi-Darani, K., & Omri, H. (2018). Process variables and design of experiments in
liposome and nanoliposome research. Mini-Reviews in Medicinal Chemistry, 18(4), 324–344.
Zuo, S., et al. (2015). Recent research progress on microbial l-asparaginases. Applied Microbiology
and Biotechnology, 99(3), 1069–1079.
Enzymes as Active Packaging System
10
Amir Heydari and Negin Mahmoodi-Babolan
Abstract
Standard food packaging is the first step that has a vital role in protecting the
product from moisture, water vapor, odors, gases, dust, microorganisms,
vibrations, shocks, and pressure force, which preserved the freshness, quantity,
and quality of the packaging to reach the customer. Active and smart packaging
approaches enhance the communication and protective functions of food packag-
ing to fulfill the demands of consumers for high-quality, safe, and convenient
food products. Recently with the technology expansion, novel enzymes with
extended applications and specificity have been advanced, and new solicitation
areas are still being investigated. This chapter has focused on oxygen-reducing
enzymes in food-packaging material, and intelligent packaging technologies are
reviewed. The main lines are active packaging materials, enzymes as catalysts in
active scavenging systems, active releasing systems, antimicrobial packaging,
time–temperature indicators, removal of undesirable food components, oxygen-
sensing applications, and the effect of the coating process and composition on the
enzyme activity. Also, enzymatic reactions with a brief discussion investigated
and explained how enzymes are used in advanced smart and food-packaging
systems. Eventually, a comprehensive list of enzymes used in food smart pack-
aging and their application are presented.
Keywords
Enzyme · Intelligent and active packaging · Antimicrobial packaging · Shelf life
# The Author(s), under exclusive license to Springer Nature Singapore Pte 253
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_10
254 A. Heydari and N. Mahmoodi-Babolan
Food packaging is necessary to keep the safety and quality of the foods. Excellent
and well-designed packaging protects the product from external and natural agencies
that may cause damage to the quantity and quality of the product, tampering
resistance, contamination and spoilage, physical and chemical damage. It can main-
tain the freshness of the food. For making products more applicable and favorable
used to reclosable openings, dispensing caps, sprays, and other features. Packaging
in a suitable method assists transportation of the packaged goods with easiness and
reduces marketing costs (Robertson, 2016). Good packaging has less handling losses
and is easy to be handled by one person during loading; therefore, characteristics of
packaging cusses that marketing costs to be less (Han, 2003).
According to the degree of protection, food can influence maximum moisture gain or
oxygen uptake. Different packaging materials such as metal cans and glass
containers can be regarded as nearly impermeable to the passage of odors, gases,
and water vapor, although in comparison packaging materials based on the paper can
be considered penetrable. Also, packaging materials based on the plastics supply
varying degrees of protection, and be contingent mainly on the nature of the
polymers utilized in their manufacture.
For many foods and drinks in which quality declines with time, it pursues that there
will be a limited length of time. Shelf life is when the product becomes undesirable,
suitable, and safe to use in specified storage conditions (Labuza, 1982). Shelf life is a
usual scuffle for food companies. Manufacturers and producers want foodstuffs to
stay fresher to lengthen the time to sell or custom. Therefore, researchers are
believed to improve shelf life to give foods and drinks a better fortuity at high
quality when consumers are prepared to eat them (Labuza & Schmidl, 1985).
Provisions to enhance food product safety and quality, increase, or stabilize food
quickly become market demands (Labuza, 1982).
the shelf life of foods by Realini and Marcos (2014). In active packaging, different
additives may be used based on the packaging film and packaged food (Lorenzo
et al., 2014). Active packaging contains ingredients that can absorb carbon dioxide,
releasing oxygen, ethanol emitters, ethylene scavengers antioxidants, and other
preservatives (Conte et al., 2007).
Active packaging systems supply various solutions to be controlled by quality
attributes that are to be protected. This theme came up, for example, in discussions of
decelerating oxidation food products be considered, so the packaging must utilize an
active system that includes an oxygen scavenger or antioxidants. Also, if the decay
of the product is created by condensation or moisture, the packaging can include a
moisture absorber. Eventually, the motivation for the organization of any novel
technology is cost.
It should point out that the unique decay mechanism of different foods should be
considered previously designing the packaging. The reality, when we have active
packaging that it carries out some role other than supplying an inert obstacle to the
exterior environment. On the other hand, active packaging is identified as a system in
which the product, package, and environment interact in a positive method to expand
the shelf life or to obtain some properties (Conte et al., 2007).
The shelf life of packaged food is reliant on abundant features such as the food
properties or product-intrinsic (e.g., pH, product color, nutrient content, aroma,
texture, water activity, viscosity, biological structure and breathing rate, the exis-
tence of antimicrobial compounds) and product-extrinsic or related to the packaging,
receptacle, or external environment factors (e.g., visual, and tactile properties of food
packaging or dishware, smell, surroundings lighting, temperature and aroma, storage
temperature, relative humidity, and the ambient gaseous composition) (Lorenzo
et al., 2014).
These features will straight affect the physical, chemical, biochemical, and
microbiological deterioration of products and their shelf lives.
Active packaging aims to improve the maintenance of food in the package, and
delaying shelf life involves the application of various plans like oxygen removal,
temperature control, moisture control, besides chemicals, for example, sugar, salt,
carbon dioxide, or natural acids or a blend of these with active packaging.
These developments in active packaging have caused developments in numerous
fields, including controlled respiration rate in agricultural products, postponed
oxidation in muscle foods, microbial growth, and moisture movement to dried
products. Furthermore, active packaging similarly employs choosiness to modify
the atmospheric concentration of gaseous combinations interior of the package by
coating, lamination, micro-penetration, or polymer blending (Realini & Marcos,
2014).
The direct incorporation of active combinations into the polymer matrix of the
packaging system is the most regular kind of active packaging system. In this
form of the active packaging system, polymer and the active agent form a composite
matrix. In recent years, this type of active packaging system is the most frequently
considered for food active packaging systems. There are three types of release
mechanisms suggested for directly incorporated food active packaging systems.
1. Diffusion due to release: In this type of release, the active agent diffuses through
the macro-porous or micro-porous polymer matrix structure and is moved from
the surface of the film into the food. This mechanism is usual for releasing active
composites for water-resistant and synthetic polymers. The rate of this kind of
release is depended on polymer chemistry, permeability, and porosity.
2. Swelling due to release: low diffusion coefficient causes the incorporated active
agent unable to penetrate the polymer matrix. When polymer swell, the diffusion
coefficient of the active agent increases; therefore, it penetrates out. Since most
foods have moisture and the water is the most popular penetrant, this type of
release commonly happens in packing materials moisture-sensitive such as
polysaccharides or protein-based films.
3. Disintegration due to release: Deformation of polymer, cleavage, and degradation
are the main reasons for this kind of release. Absorption of liquids commonly
from aqueous fluids through the polymer matrix influences degradation and
deformation rate.
10 Enzymes as Active Packaging System 257
Fig. 10.2 Schematic illustration of different approaches used for release controlling from food
active packaging (Almasi et al., 2020)
Antimicrobial packing is a type of active packaging that use some agents which kill
or inhibit the growth of microorganisms that may be existing in the packaged
product or packaging material itself. For control of unfavorable microorganisms
on foodstuffs, antimicrobial materials can be combined and coated inside food-
packaging materials (Motta et al., 2004).
For example, antimicrobial polymers provide a good antimicrobial plan for
combating pathogens and have already drawn the attention of industrial and aca-
demic research (Prazak et al., 2002). Antimicrobial mechanisms demonstrate either
active or passive action and polymer material categories, including bound or
leaching antimicrobials used. Antimicrobial polymers are used in some industries,
such as medical, food, and textile (Motta et al., 2004). Compounding polymers and
258 A. Heydari and N. Mahmoodi-Babolan
10.6 Enzyme
Absolute specificity that the enzyme will catalyze only one reaction.
Group specificity in which the enzyme will affect performance only on molecules
with particular functional groups, such as phosphate, amino, and methyl groups
(Budny, 1989).
Linkage specificity that the enzyme, irrespective of the rest of the molecular struc-
ture, will act on a specific type of chemical bond and the best technique to
immobilize enzymes or other bioactive composites on polymers. The chemical
steps included in covalent bonding are complex and timewasting. Therefore, are
not presently practical for joint commercialization for food contact packaging.
Enzymes are effective biological catalysts frequently utilized in the food industry
to get better performance, quality, and shape of food. Using enzymes as active
agents in the food packaging system is not novel technology (Adrian, 1959).
Stereochemical specificity is the enzyme performance with an optical isomer or
specific esters. Enzymatic specificity requires two different forms: the first one is
the type of chemical reaction and the second one is no chemical reaction,
specificity for the reactant, or substrate. Therefore, for each chemical reaction
in a biological system, there is a particular enzyme demand for the optimal
production of reaction products. It is essential to consider many different
biological reactions to pursue numerous different enzymes (Brody & Budny,
1995).
Fig. 10.4 Schematic drawing of the enzyme immobilization process. (a) Immobilization in the
dispersion phase and (b) immobilization on the surface of polymer film formed from a latex
dispersion (Nestorson et al., 2008)
10 Enzymes as Active Packaging System 261
Time–temperature integrators (TTIs) are indicators that can display food safety and
quality changes during distribution, handling, and storage (Taoukis & Labuza,
Fig. 10.5 The enzymatic reactions in the oxygen scavenging system using the combination of
glucose oxidase and catalase. The mechanism complicated here is adapted from Shi et al. (2010)
and Tian et al. (2013)
262 A. Heydari and N. Mahmoodi-Babolan
1989). TTIs are necessary for temperature-sensitive foods to evaluate and control the
distribution chain (Giannakourou et al., 2005). TTIs must be low-priced, readable,
reliable, and without any poisonous materials. Most of the TTIs are very similar to
labels and can be used for food products. Several TTIs have been expanded based on
different operating principles. Enzymatic TTIs utilize α-amylase (Guiavarc’h et al.,
2004), lipase (Agerhem & Nilsson, 1981), or b-glucosidase (Adams & Langley,
1998). Presently, lipase is an available enzymatic TTI that can be utilized to control
quality assurance. Enzymatic TTIs that have been investigated to date have utilized
synthetic and then rather expensive chromogenic substrates hydrolyzed to generate a
color. Consequently, if an enzyme that uses natural dyes as its substrate were
available, a TTI produced using that enzyme would supply various benefits,
counting developed visibility, less expensive, and extensive use of dye coding.
Laccases are conventionally used for numerous industrial processes counting
paper processing, detoxification of environmental pollutants, oxidation of pigments,
prevention of wine discoloration, and production of chemicals from lignin (Kim
et al., 2012).
10.7 Conclusion
Active packaging technologies suggest novel chances for the industries for
protecting their products. Different active packaging systems are currently available
based on the chemical and physical structure of the foods. Oxygen, moisture, carbon
dioxide, ethylene, and ethanol are considered for the active packaging. The structure
of the active packaging and the potentials of the enzymes are mentioned. The
importance of this subject has produced many investigations to make more useful
systems. In the following, oxygen scavenger in enzyme packaging and time–tem-
perature integrator-indicators were investigated. Finally, investigation shows the
improvement of novel indicators and enzymes that are much more precisely
designed for the active packaging system.
References
Adams, J., & Langley, F. (1998). Nitrophenyl glucoside hydrolysis as a potential time-temperature
integrator reaction. Food Chemistry, 62(1), 65–68.
Adrian, J. (1959). Le dosage microbiologique des vitamines du groupe B. Annales de la nutrition et
de l’alimentation. JSTOR, 13, 1–175.
Agerhem, H., & Nilsson, H. J. (1981). Substrate composition and use thereof. Google Patents.
Ahvenainen, R. (2003). Novel food packaging techniques. Elsevier.
Almasi, H., Jahanbakhsh Oskouie, M., & Saleh, A. (2020). A review on techniques utilized for
design of controlled release food active packaging. Critical Reviews in Food Science and
Nutrition, 61, 1–21.
Blixt, K., & Tiru, M. (1976). An enzymatic time/temperature device for monitoring the handling of
perishable commodities. Developments in Biological Standardization, 36, 237–241.
Brody, A., & Budny, J. (1995). Enzymes as active packaging agents. In Active food packaging
(pp. 174–192). Springer.
10 Enzymes as Active Packaging System 263
Brody, A. L., Strupinsky, E., & Kline, L. R. (2001). Active packaging for food applications. CRC
Press.
Budny, J. (1989). A transporting storage or dispensing container with enzymatic reactor. Interna-
tional Patent Application W, 89.
Conte, A., et al. (2007). Innovative active packaging systems to prolong the shelf life of mozzarella
cheese. Journal of Dairy Science, 90(5), 2126–2131.
Effront, J. (1917). Biochemical catalysts in life and industry: Proteolytic enzymes. John Wiley &
Sons Inc.
Giannakourou, M., et al. (2005). Field evaluation of the application of time temperature integrators
for monitoring fish quality in the chill chain. International Journal of Food Microbiology,
102(3), 323–336.
Guiavarc’h, Y., et al. (2004). Bacillus licheniformis α-amylase immobilized on glass beads and
equilibrated at low moisture content: Potentials as a time–temperature integrator for sterilisation
processes. Innovative Food Science & Emerging Technologies, 5(3), 317–325.
Han, J. H. (2003). Antimicrobial food packaging. Novel Food Packaging Techniques, 8, 50–70.
Kim, K., Kim, E., & Lee, S. J. (2012). New enzymatic time–temperature integrator (TTI) that uses
laccase. Journal of Food Engineering, 113(1), 118–123.
Klibanov, A. M., & Dordick, J. S. (1989). Enzymatic temperature change indicator. Google
Patents.
Kothapalli, A., Morgan, M., & Sadler, G. (2008). UV polymerization-based surface modification
technique for the production of bioactive packaging. Journal of Applied Polymer Science,
107(3), 1647–1654.
Labuza, T. P. (1982). Shelf-life dating of foods. Food & Nutrition Press Inc.
Labuza, T. P., & Breene, W. (1989). Applications of “active packaging” for improvement of shelf-
life and nutritional quality of fresh and extended shelf-life foods. Journal of Food Processing
and Preservation, 13(1), 1–69.
Labuza, T. P., & Schmidl, M. K. (1985). Accelerated shelf-life testing of foods. Food Technology.
Liu, L., et al. (2014). Capillary electrophoresis-based immobilized enzyme reactor using particle-
packing technique. Journal of Chromatography A, 1352, 80–86.
Lopez-Rubio, A., et al. (2004). Overview of active polymer-based packaging technologies for food
applications. Food Reviews International, 20(4), 357–387.
Lorenzo, J. M., Batlle, R., & Gómez, M. (2014). Extension of the shelf-life of foal meat with two
antioxidant active packaging systems. LWT—Food Science and Technology, 59(1), 181–188.
Motta, G. J., Milne, C. T., & Corbett, L. Q. (2004). Impact of antimicrobial gauze on bacterial
colonies in wounds that require packing. Ostomy/Wound Management, 50(8), 48.
Narayanan, R., Stottrup, B. L., & Wang, P. (2009). Surface packing characterization of Langmuir
monolayer-anchored enzyme. Langmuir, 25(18), 10660–10665.
Nestorson, A., et al. (2008). Enzyme immobilization in latex dispersion coatings for active food
packaging. Packaging Technology and Science: An International Journal, 21(4), 193–205.
Ozdemir, M., & Floros, J. D. (2004). Active food packaging technologies. Critical Reviews in Food
Science and Nutrition, 44(3), 185–193.
Prazak, A. M., et al. (2002). Antimicrobial resistance of Listeria monocytogenes isolated from
various cabbage farms and packing sheds in Texas. Journal of Food Protection, 65(11),
1796–1799.
Realini, C. E., & Marcos, B. (2014). Active and intelligent packaging systems for a modern society.
Meat Science, 98(3), 404–419.
Robertson, G. L. (2016). Food packaging: Principles and practice. CRC Press.
Shi, X., Lim, J., & Ha, T. (2010). Acidification of the oxygen scavenging system in single-molecule
fluorescence studies: In situ sensing with a ratiometric dual-emission probe. Analytical Chemis-
try, 82(14), 6132–6138.
Soares, N., & Hotchkiss, J. (1998). Naringinase immobilization in packaging films for reducing
naringin concentration in grapefruit juice. Journal of Food Science, 63(1), 61–65.
264 A. Heydari and N. Mahmoodi-Babolan
Taoukis, P., & Labuza, T. P. (1989). Applicability of time-temperature indicators as shelf life
monitors of food products. Journal of Food Science, 54(4), 783–788.
Tian, F., Decker, E. A., & Goddard, J. M. (2013). Controlling lipid oxidation of food by active
packaging technologies. Food & Function, 4(5), 669–680.
Wierschem, M., et al. (2017). Enzyme kinetics for the transesterification of ethyl butyrate with
enzyme beads, coated packing and ultrasound assistance. Chemical Engineering and
Processing: Process Intensification, 111, 25–34.
Wu, X., et al. (2019). Packaging and delivering enzymes by amorphous metal-organic frameworks.
Nature Communications, 10(1), 1–8.
Enzymes as a Tool in Food Analysis
and Foodborne Pathogen Detection 11
Preethi Sudhakara, Jerrine Joseph, S Priyadharshini, Jemmy Chirsty,
Alex Anand, Davamani Christober,
and Aruni Wilson Santhosh Kumar
Abstract
P. Sudhakara
School of Medicine, University of Florida, Gainesville, FL, USA
J. Joseph
Center for Drug Discovery and Development, Sathyabama Institute of Science and Technology,
Chennai, India
S. Priyadharshini
Department of Food Science and Nutrition, The American College, Madurai, India
J. Chirsty · A. Anand
Center for Molecular Data Sciences and Systems Biology, Sathyabama Institute of Science and
Technology, Chennai, India
D. Christober
Department of Food Science and Nutrition, The American College, Madurai, India
The American College, Madurai, India
A. Wilson Santhosh Kumar (*)
Sathyabama Institute of Science and Technology, Chennai, India
Musculoskeletal Research Center, US Department of Veteran Affairs, Loma Linda VA, Loma
Linda, CA, USA
School of Medicine, Loma Linda University, Loma Linda, CA, USA
e-mail: vcaum@mum.amity.edu
# The Author(s), under exclusive license to Springer Nature Singapore Pte 265
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_11
266 P. Sudhakara et al.
Keywords
11.1 Introduction
Enzymes are defined as the biological materials responsible for fastening the reaction
rate. Enzymes are the so-called multifaceted biomolecules that are indispensable for
biochemical interconversions. They act as a catalyst for all the metabolic processes
in the body. The advancement in rDNA technology and protein engineering has
enabled us to capitalize on the various functions of enzymes from industrial to
biomedical applications. Enzymes are exceptionally effective biocatalysts
investigated for wide-reaching catalysis due to a few particular preferences that
run from their activity in milder response conditions to their excellent component
selectivity and lower ecological and physiological harmfulness (Chapman et al.,
2018). Enzymes are initially involved in the dissolvable or immobilized structure for
isolation, purification, and production to benefit humanity (Chaplin & Bucke, 1990).
The demand for the enzyme increases by 6% yearly, especially in food and feed
production (Nampoothiri et al., 2002). Enzymes are widely and progressively
utilized in exploration and food processing as significant handling biocatalysts
(Ermis, 2017). Enzymes are intricated in a living organism. Enzymes formed in
live cells can enhance reaction rates in vitro. The acclimatization of enzymes in the
food sector is known widely, and intense investigations consistently unravel the
global food crisis. This chapter highlights the fundamentals of enzymes, sources of
diverse enzymes, and their applications in the food sector.
Microbial enzymes have been catapulted to fame due to their inherent properties
of the ease of their production and yield manipulation in laboratory settings. This
highlights their significance and the opportunities that pave the way for
microbiologists by harnessing the biomolecules’ potential. The thoughtful consider-
ation of the enzymes in the food sector has amended the primary methods to afford
improved marketplaces with high quality and safety. Previously, this information
about enzymes has been used in meat tenderization, baking, brewing, etc. Mainly the
enzymes are produced by microbial synthesis. The microbial method for enzyme
production has been favored over other methods for quick and inexpensive
production.
Furthermore, microbially produced enzymes’ harmful content is lesser than other
sources. The first commercial application of enzymes was in the production of
cheese. Enzymes in the food sector alter traditional and toxic methods, substitute
them with sustainable methods, and reduce energy consumption levels. It may be
worthwhile to unclose an unexplored trail for enzyme applications, which could be
advantageous in the food industry.
Foodborne pathogens influence human health negatively and are comprehended
to induce financial losses. Therefore, it is essential to rapidly detect foodborne
pathogens and execute steps to assure their inactivation. Immunological, molecular,
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 267
and cultural approaches are often exploited to detect foodborne pathogens. High
expense, lengthy analysis duration, and the need for technical personnel are some of
the drawbacks of these approaches. Biosensors are known as analytical instruments.
Biosensors can be used as novel techniques for detecting toxins and pathogens.
These biosensors restore various signals (like biochemical signals) to electrical
signals that can be measured with the help of a transducer. Various kinds of
biosensors are being utilized for the detection of pathogenic bacteria. Biosensors
are prompt and cost-effective devices and are employed in multiple areas such as
food safety, medicine, pharmacy, measurement of environmental pollution, and
military defense. Electrochemical and optical biosensors and piezoelectric
immune-sensors are among the most repeatedly used biosensors in detecting
foodborne pathogens.
There are many viruses and bacteria, some that cause animal disease, and others
that contribute to foodborne human illness and disease that can be tested for the
occurrence of foodborne pathogens. Some of the significant subjects of testing
include several swine viruses, including swine influenza virus and porcine repro-
ductive and respiratory syndrome virus. Bovine viruses include bovine viral diar-
rhea virus, bovine syncytial virus, bovine rotavirus, coronavirus, and rhinotracheitis
virus. In addition, the Hepatitis A virus, flaviviruses, West Nile, and the Usutu virus
(USUV) that have recently been on the rise in Europe and some parts of South East
Asia are of much importance. The diagnosis and surveillance of bacterial pathogens
encompass the full spectrum of animal and foodborne pathogens. These include
Escherichia coli and bacteria in the Salmonella, Klebsiella, Clostridium, Listeria,
Staphylococcus, Streptococcus, Pseudomonas, Pasteurella, Actinobacillus, and
Mycoplasma genera.
Rapid, efficient pathogen detection and fingerprinting are crucial and usually
lifesaving to control the foodborne ailment. Presently, detecting and subtyping the
pathogens are distinct operations, but merging these two phases via a technique
known as “metagenomics analysis,” the detection is much prompter and more
sensitive as many microorganisms are now employed to produce food enzymes.
Enzyme technologies have been thoroughly revolutionized with the recent novel
trends in biotechnology applications to fulfill the new and changing requirements of
the food industry. There is an extensive tendency toward novelty in enzyme discov-
ery and creating tailored enzymes to make them inclined for industrial food
applications.
In the recent past, developments in enzyme informatics tools can simplify
designing the enzymatic structure. Also, its computational dynamic simulations
can produce the kinetic and energetic course of its mechanism. Enzyme informatics
can also recreate an exceptional part in mutant library design and other screening
platforms, but its execution of molecular approaches is still time consuming. Recent
improvements in the subject will hover around novel frontier in enzyme technologies
that could attribute various functional aspects to these enzyme biocatalysts. This
study will consolidate the extensive characteristics of food enzymes, their diversified
functions in the food industry, the destructive outcomes of foodborne pathogens, and
their current detection techniques.
268 P. Sudhakara et al.
Human health and healthcare predominately revolve around the quality of food
consumed. Appealing enzymes in the food industry can manage the quality food
supply problem. Hence, evaluating food quality and ensuring food safety standards
are highly important. This crucial factor plays a vital role in evaluating food quality,
safety, and the turnaround time to ascertain this. Enzymes are biomolecules that
inherently possess specificity and sensitivity traits (Fig. 11.1).
Hence enzymes are a better bet than the conventional diagnostic methods that are
dreary and overwhelming and require correlative modes to detect food quality and
safety. Enzyme-based analytical tools to measure biological functionality are more
reliable and consistent. The analytical tools should be high throughput and compe-
tent to authenticate and authorize the study of particular aspects of biology, either
qualitatively or quantitatively, which is possible by enzymes. Also, a crucial part of
developing such tools is to source or amalgamate the desired enzymes with the
required activity. Thus, the utilization of enzymes as analytical informers is an
inexpensive, simpler, and quicker manner of diagnostics. Microbes have been used
from bygone days; yeast is the first reported organism to produce alcoholic
beverages using barley in 6000 BC. The microbial enzymes have earned acknowl-
edgment globally for their extensive uses in several trades. Nevertheless, recombi-
nant DNA technology and protein engineering are assembled to confound the
demand. It improves consumer goods, expense deduction, natural resources short-
age, and environmental safety (Liu et al., 2013; Choi et al., 2015b).
Microbial enzymes play a crucial role (Raveendran et al., 2018). In terms of
analytical procedures in the Food and Pharmaceutical industries, enzymes play a
vital role. As indicated by food and regulatory agencies, food safety and quality are
the censorious criteria recurrent to all food constituents, and these criteria are of
substantial lucrative significance. Food safety depends on toxins, dangerous
microorganisms, toxins divulged by microbes, insecticides, and pesticides (Terry
et al., 2005). Processing of foods with biological enzymes is chronicled inveterate
approach. Earlier in 6000 BC, bread, brewing, wine, and cheese making were done
with biological agents, but the initial microbial fermentation was from 2000 BC for
vinegar production (Vasic-Racki, 2006). In 1930 for juice clarification, pectinases
were used. Invertase was used during World War II for the invert sugar syrup
production in the sugar industry that developed the use of an immobilized enzyme
(Vasic-Racki, 2006). In 1960, the extensive use of enzymes established during the
traditional acid hydrolysis was restored by slant based on utilizing amylase and
glucoamylases (Panesar et al., 2010).
The pattern for the plan and execution of procedures and creation of products
moored in the utilization of enzymes has consistently expanded (Norus, 2006).
Enzymes from microbes play a significant part in food ventures since microbial
enzymes’ stability is higher than plant and animal enzymes. They can be fabricated
through fermentation strategies in a lucrative way with less reality prerequisite and
high consistency and process adjustment (Gurung et al., 2013). In food analytical
strategies, food safety and quality control are essential, and they can be analyzed by
numerous analytical procedures like HPLC, GC-MS, and LC-MS (Castillo et al.,
2004). In any case, sample preparation strategies are the significant downside of this
technique. Progressing diagnostic strategies in the food industry are tedious and rely
upon skilled laborers, and these procedures depend on extended division techniques,
costly instruments, and exceptionally unadulterated chemicals. In the field of quick
screening, there is a requirement for complementary strategies to recognize food
safety and quality issues. A more specific and sensitive method for screening food
safety and quality issues is enzymatic analysis (Prodromidis & Karayannis, 2002).
Enzymes play a crucial role in the analytical process in the food, pharmaceutical, and
environmental industry. For governing the compound concentration that performs as
an activator, substrate, or inhibition of specific enzymes, enzymes are used as typical
analytical devices. For the analysis of modification induced physically and nutrition-
ally and quality criterion of food, the concentration and activity of specific enzymes
are used. The list of enzymes and their application, along with the microbial
enzymes, are listed in Table 11.1.
Enzymes are extensively used as an important marker, especially in the food and
pharmaceutical industry. The broad scope of enzymes has been boon to the food
analyst who investigates the different constituents of the food. Food, being more
complex with increased macronutrients and very few micronutrients analyzing these
food constituents, requires extremely simple and affordable, less time-consuming
feasible, and specific techniques. Enzymes are used as markers for food analysis and
their lustiness, adaptability, and cost-effective nature (Ashie, 2012).
270 P. Sudhakara et al.
Enzymes play a vital role in detecting milk pasteurization. For increasing the shelf
life of milk and milk products, heat treatment is essential. In many countries, an
alkaline phosphatase test is used as a standard technique for milk ratification.
Non-appearance of alkaline phosphatase in the heat-treated milk shows efficient
pasteurization. It is based on the inactivation of the alkaline phosphatase enzyme
using heat.
On the other hand, it reduces the pathogenic bacterial population in the milk. Not
only for milk, but this rapid detection method can also be used for other dairy
products like cheese production. Other primordial enzyme markers for milk include
lactoperoxidase and γ-glutamyl transpeptidase (Rankin et al., 2010).
The main aim of blanching is to remove the enzyme polyphenol oxidase. Apart from
this, another equitable goal is to decrease microbial growth and increase shelf life.
Time taken for blanching is enough to destroy the indicator enzyme. Blanching of
frozen foods, are done at different time intervals in order to recognize the relation-
ship between enzyme activity and sensory scores (Williams et al., 1986).
Enzymes are used in a variety of ways in the food industry. Enzyme activity aids in
the safe digesting of food in various situations. The thermal stability of enzymes has
been used extensively to quantify heat treatment; peroxidase activity, for example, is
used to determine the sufficiency of blanched vegetables. Lactoperoxidase has also
been used to test the effectiveness of the pasteurization process in milk. In some
cases, residual enzyme activity affects the flavors or color of products while they are
being stored. Lipoxygenase, for example, is responsible for off-flavors in frozen
veggies that have not been adequately blanched. The staining of wheat flour and
noodles and fruit liquids and pastes is catalyzed by polyphenol oxidase (Nielsen,
2017).
The enzyme is used to test the components of enzyme-substrate foods that are
commercially accessible. The glucose content of a complex dietary matrix
containing additional monosaccharides can be determined using commonly avail-
able enzymes. Enzyme activity measurement concerning food quality is also note-
worthy. Milk from mastitic udders, for example, has considerably increased catalase
activity. The quantity of bacteria in milk is also connected to catalase activity.
Another technique to use enzyme testing to determine food quality is to estimate
protein nutritional value by measuring the activity of added proteases on dietary
protein samples. Enzymes can track the appearance of breakdown products such as
trimethylamine in fish during storage. Enzymes are also used as preparatory tools in
food analysis. In fiber analysis, amylases and proteases are used, while enzymatic
hydrolysis of thiamine phosphate esters is used in vitamin analysis (Lee et al., 1991).
Table 11.2 lists some of the most often used enzymes in the food business.
Food scientists must understand that the environment significantly impacts
enzyme activity. At high temperatures or under other situations that are not too far
off from ideal, that enzyme can deteriorate and lose its activity. As a result,
therapeutic enzymes must be stored and handled with care, usually in the refrigerator
or freezer. To effectively carry out enzyme analyses in foods, certain basic enzymol-
ogy principles must be understood. Following a brief overview of these concepts,
instances of enzymatic analysis in food systems are discussed. Under physiological
conditions, enzymes are protein facilitators with high specificity and reactivity.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 273
Enzymes regulate the brewing process, ensure consistent, high-quality beer; enhance
animal and vegetable proteins’ functional and nutritive properties through enzymatic
hydrolysis; and increase juice yield while improving color and aroma. Enzyme use
274 P. Sudhakara et al.
in the food business is divided into several categories, including baking, dairy, juice
production, and brewing (Mojsov, 2011). Microbial enzymes are widely used in the
baking business, the most prominent application market in the food industry, to
increase dough stability, crumb softness and structure, and product shelf life. The
usage of enzymes in the dairy industry (Qureshi et al., 2015), which is the next
largest application area, is primarily due to the increased use of microbial enzymes in
cheese manufacturing (Qureshi et al., 2015). The laboratory must identify appropri-
ate enzymes with the required activity and understand the target analyte and the
complexity of the matrix in which the analyte must be measured (Kieliszek &
Misiewicz, 2014). Foods are typically very complex matrices, posing a significant
technical barrier to the analyst tasked with determining and establishing the levels of
various components. As a result, numerous analytical methods, including enzymes,
have been developed to meet this issue over time. The usage of enzymes is based on
specificity, which allows for the selective, sensitive, and precise measurement of
macro- and micronutrients in meals. The development of biosensors with greater
robustness and versatility has boosted the use of enzymes as an analytical tool in
recent years. Starch is a critical ingredient in baked goods because it aids in the
browning and flavoring of the crust. It is found primarily in plants in bread
production.
Many enzymes are used for milk processing and cheese production in the dairy
industry. Rennet is a coagulant enzyme mixture of chymosin and pepsin used in milk
clotting and cheese preparation. They have been extracted from animal sources,
mainly from the stomach of young calves (Theron & Divol, 2014). Plant extracts
also contain many enzymes, such as leaves of Sodom apple, berries of Solanum
dubium, Calotropis procera, and cardoon extracts, from microbial source
Rhizomucor miehei and Rhizomucor pusillus, contain protease enzyme that helps
in clotting. Protease enzyme obtained from Cryphonectria parasitica aggregate and
transferred milk into curd and liquid part.
Enzymes are ubiquitous, and their application has been used from the olden days in
human edification. Food processing methods modify the food materials and make
them appropriate for consumption (Monteiro et al., 2011). In processing, it has been
used in wine, beer brewing, cheese, and bread making from 6000 BC. Based on the
enzyme properties, the International Union of Biochemistry approved a class of
enzymes used in the food industry, shown in the figure below (Fig. 11.2).
Food enzymes are categorized into two types: food processing and food additives.
Most of them are used for food processing purposes, whereas lysozyme and inver-
tase are used as food additives. The main aim of these food processing agents is to
increase the edible foods’ shelf life and preserve the health and nutritional contents
of the food without affecting their taste and aroma. Uses of enzymes in the food
industry process carbohydrates, proteins, and fats.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 275
Lyases
Oxido
Hydrolases
reductases
Isomerases Pecnase
Types of
Enzyme
Ligases Invertase
Amylase
Protease
alpha, beta
and Gluco
Oxido
Transferase
reductases
Fig. 11.2 Types of major microbial enzymes used in the food industry
11.9.1 Alpha-Amylase
The most important industrial enzymes account for around 25% of the global
enzyme market, which comes from various sources, including plants, animals, and
microbes (Kandra, 2003; Reddy et al., 2004). Enzymes catalyze the hydrolysis of
internal 1,4-glycosidic bonds in starch to produce low molecular weight compounds
like glucose, maltose, and maltotriose (Gupta et al., 2003; Rajagopalan & Krishnan,
2008). The baking sector currently employs a thermostable maltogenic amylase from
Bacillus stearothermophilus. It is also used to clarify beer and fruit liquids and
improve fiber digestion in animal feed (van der Maarel et al., 2002; Gavrilescu &
Chisti, 2005; Ghorai et al., 2009).
sp. can produce these enzymes used to manufacture organic acids (Djekrif-
Dakhmouche et al., 2006; Hernández et al., 2006).
11.9.3 Glucoamylase
11.9.4 Cellulase
11.9.5 Pectinase
found in microbial strains such as Moniliella SB9, Penicillium spp., and Aspergillus
spp. (Priya & Sashi, 2014).
11.9.6 Invertase
11.9.7 Protease
Proteases represent one of the major groups of industrial enzymes, with a molecular
weight of 126 kDa. It breaks the peptide bonds present in the protein and separates
them as amino acids. Bacillus and Aspergillus spp. is the primary microorganism
that produces protease enzymes. It is used for several purposes such as brewing,
meat tenderization, baking, hydrolyzed animal proteins, functional meat
proteins, etc.
11.9.8 Lipase
Lipase enzymes deconstruct lipids into fatty acids and glycerol. This is something
that can be used in the baking sector. It separates milk fat and gives terrific flavors to
the cheese. The flavor of the cheeses improves the creamy texture and taste of the
products. Excessive lipid lysis leads to unavoidable odors. Most commercial lipases
develop flavor in dairy products, meat, vegetables, fruit, baked goods, milk, beer,
and others (Nagodawithana & Reed, 1993). The major lipase enzyme producers are
Pseudomonas sp., Bacillus sp., and Penicillium sp.
microorganisms demonstrated the need for improved and faster technology to ensure
the safety and quality of the food products. Conventional techniques for food quality
assessments were replaced with fast and easy techniques like biosensors. Biosensors
are more extraordinary than other demonstrative instruments due to different food
quality and security innovations. Enzymatic biosensors are commonly utilized in the
food business (Table 11.3). Different properties of biosensors, especially optical,
mechanical, and electrochemical, can be adjusted by utilizing nanomaterials.
Biosensors are an appropriate detecting tool for quality control and revealing the
contamination in food. It can be a systematic and feasible detecting method in the
food chain.
The most important part of the food industry is maintaining food safety and
quality control. Biosensors have numerous food safety and quality control
applications. Enzyme biosensors are an essential technique as it has a notable
usage compared to the conventional method. It is effortless and less time consuming,
and it does not involve skilled labor and costly equipment (Santos et al., 2006;
Hooda et al., 2018). There are two main biosensors’ main constituents: the
bioreceptor and the transducer (Viswanathan et al., 2009). Biosensors work on
three principles based on the device used to create biosensors, segments of molecular
remembrance, and transducer. There are few enzymes for specific foods to act as a
biosensor. There are numerous applications in the food industry in which biosensors
are involved in quantifying different food constituents to determine their shelf life.
Biosensor technologies have the insightful potential to screen adverse substances
in foods on-site and in-situ phases. Biological agents like enzymes usage in
biosensors improved the specificity due to their binding affinity to a specific sub-
strate and related catalytic effect. In addition, they lack interference with the
compounds present in a complex sample. Due to sensitivity and specificity, enzy-
matic biosensors are considered an inevitable technology for food quality screening.
Table 11.3 Enzymes utilized in biosensors (Modified from Akyilmaz et al., 2010)
Enzymatic biosensors Food constituents
Glucose oxidase, glucose dehydrogenase Glucose
Oxalate oxidase Oxalate
Tyrosinase Tyrosine
Malate dehydrogenase Malate
Galactose oxidase Galactose
Cholesterol oxidase Cholesterol
Alcohol oxidase Alcohol
Peptidase, glutamate oxidase Aspartame
Tyrosinase Phenols
Lipoxygenase Essential fatty acids
Fructose-5-dehydrogenase Fructose
Glucose oxidase, mutarotase, invertase Sucrose
Galactose oxidase, peroxidase Lactose
Glutaminase Glutamine
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 279
for food safety (Vasilescu et al., 2016). They are cheaper and can be obtained from
various materials with a minimal level of sample volumes (Power et al., 2018). The
enzyme-based sensor detects food contamination in various sectors of the food
industry as shown in Fig. 11.3.
In 1956, Leland Charles Clark Jr published research on the electrode used to measure
oxygen levels in the blood (Clark et al., 1958). Electrochemical sensors and enzyme
transducers were described as membranes at a 1962 Academy of Sciences sympo-
sium. Clark described the functioning enzyme electrode with glucose oxidase
attached to the oxygen sensor later in 1967. Guilbault and Montalvo first described
the potentiometric enzyme electrode in 1970. It works by immobilizing the enzyme
urease on an ammonium-selective liquid membrane electrode. In 1973, they
described a hydrogen peroxide-based glucose and lactate enzyme sensor using a
platinum electrode. Klaus Mosbach invented a thermistor (heat-sensitive sensor) in
1974. In 1975, Clark introduced the glucose analyzer from Yellow Springs Instru-
ment Company, which used an amperometric sensor to detect hydrogen peroxide.
Lubbers and Opitz described a fiber-optic sensor with an immobilized indication to
monitor carbon dioxide or oxygen levels. Later, by immobilizing alcohol oxidase,
the optical biosensor concept was expanded to detect alcohol levels.
With the help of an artificial pancreas, Clemens and his colleagues built an electro-
chemical glucose biosensor. Miles later marketed it in 1975 as the Bio-stator (Gouda
et al., 2002). Bio-stator was not accessible on the market. VIA Medical introduced a
new semi-continuous catheter-based blood glucose analyzer. The lactate analyzer
LA 640 was introduced by La Roche in 1976 to transport electrons from lactate
dehydrogenase to an electrode.
Advanced biosensors have both immobilized enzymes and mediators in the same
electrode. Liedberg used the surface plasmon resonance (SPR) method to track
affinity interactions in 1983. In 1984, ferrocene and its derivatives were employed
as immobilized mediators with oxidoreductases in the manufacture of low-cost
enzyme electrodes based on screen-printed enzyme electrodes, according to Turner
and his coworkers.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 281
processes and distribution lines or offline as quick, simple, and disposable sensors
for food contamination. Food contaminants may be challenging to detect in this
environment of chemicals. Nanosensors are an emerging technology that could be
used to detect a variety of food pollutants, including mycotoxins and food allergies,
due to their unique applications. Nanosensors are thus more cost-effective, quick,
and sensitive than instrumental and traditional methods. Recent advancements in
nanosensor technology may increase demand for its use in food contamination
detection. As the food laboratory faces increased pressure to minimize cost, time,
and complexity, the role of nanosensors will become increasingly more crucial. This
chapter seeks to provide a broad overview of the potential applications of
nanosensors in the detection and analysis of food contaminants. In recent years,
the traditional PCR technique has been superseded by a simplified application in
POC devices using multienzyme bioreceptors to determine DNA fragments’
properties from allergens or adulterants. Surface platform resonance (SPR)
characteristics and high conductivity are attributes of nanomaterials used in these
sensor systems, such as Au NPs and AgNPs. This will make it easier to incorporate
optical and electrochemical sensors. Magnetic nanoparticles, such as Fe2O3 NPs,
will aid in the separation of analytes and will boost their performance.
However, carbon nanotubes and graphene-based nanomaterials enhance electro-
chemical signals due to their outstanding electrical conductivity. In 2017, electro-
chemical affinity biosensing was utilized to test food allergies, adulterants, and
gluten. Andrei et al. (2016) used Nanoceria, a nanosensor device with oxidase-like
activity, to covert phenolic antioxidants caffeic acid, gallic acid, and quercetin to
quinones to measure wine’s antioxidant content. He et al. (2018) developed manga-
nese dioxide nanosheets to detect ascorbic acid levels in orange fruit and juice. The
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 285
hazardous chemical biphenol A (BPA) is released from the polycarbonate and epoxy
resin during the food packaging process. Tyrosinase converts BPA to quinine to
detect BPA NPs such as Au, Ni, or Fe2O3 (Alkasir et al., 2010).
Biosensors play a vital role in detecting foodborne pathogens, and it plays a crucial
role in food packaging as a part of smart packaging, which can be used in packaging
materials to check their freshness and shelf life. As a part of smart packaging, the
freshness indicator, time temperature indicators, integrity indicator, and radio fre-
quency identification are used (Park et al., 2015).
Food is required for the survival of life. We all eat food to obtain energy for various
metabolic functions. Food is required for all living species to grow, function, heal,
and maintain life processes. There are many different types of food on the market
today, and we all rely on diverse food sources daily, such as vegetables, fruits,
grains, pulses, legumes, etc. Small stones in cereals and grains, darkly stained
vegetables like cabbage and broccoli, fruits, dark red meat, and other items may
have been encountered while shopping for fresh vegetables and other foods. Food
adulteration is described as polluting food or food materials by adding a few
substances, collectively known as adulterants. Adulterants are substances or
products of lower quality that are added to food for economic or technical reasons.
The inclusion of these adulterants lowers the nutritional value of food and
contaminates it, rendering it unfit for ingestion. These adulterants can be found in
various foods that we regularly consume, including dairy, cereals, legumes, grains,
meat, vegetables, fruits, oils, and beverages. In underdeveloped countries, contami-
nation or adding to food components is typical. Milk, for example, can be diluted
with water to increase its volume, and starch powder is frequently added to increase
its solid content.
The urease microbial enzyme from Arthrobacter creatinolyticus was immobilized
on a poly (acrylonitrile-methyl methacrylate-sodium vinyl sulfonate) (PAN) mem-
brane to create a potentiometric urea biosensor. Glutaraldehyde was used as a cross-
linking agent in the biosensor. The following reaction is catalyzed by urease. The
production of ammonium ions produced by the breakdown of urea by urease was
used to detect urea in samples. A commercially accessible electrochemical worksta-
tion was used to perform potentiometric experiments. Milk samples were spiked
with known urea concentrations and evaluated using the biosensor for real-world
analysis. The measured potentiometric measurements were in good agreement with
the spiking concentrations. The biosensor had a detection limit of 0.3 mM and
linearity over the range of 1–100 mM. It was able to maintain appropriate sensitivity
for 13 cycles in a row. It could be kept for 70 days at 4 C.
286 P. Sudhakara et al.
11.14.1 Melamine
The demand for ready-to-eat halal foods such as burgers, pizzas, hot dogs,
sandwiches, soups, cookies, sweets, and creams has risen recently (Jeddah, 2011;
Mascini, 2015). Pork is being used more frequently in Halal meals, including beef
meatballs, to maximize profits (Mascini, 2015). It was identified utilizing NPs as a
colorimetric nanobiosensor, which changed the color of the meat from pinkish-red to
purple with a 525 nm absorption peak. Ali et al. (2011) used hybrid biomaterials
with functionalized nanoparticles that are noncovalently or covalently coupled to
biomolecules such as peptides, proteins, and DNA to detect pork adulteration in
processed meat.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 287
Food adulteration can arise from numerous sources, such as natural toxic
compounds, bacterial contamination, pesticides, veterinary drugs, and chemicals
used in food dispensation. Identifying this substance is essential for people affected
by diseases (Viswanathan et al., 2009). It has been mostly separated into two classes:
biological and chemical contaminants. Some critical assays analyze the
phytocompound such as sugars, alcohols, amino acids, flavors, and sweeteners.
Chromatography and spectroscopy have been used to detect contaminants from
the olden days. Currently, many sensors have been developed to detect the presence
of these contaminants in food without affecting their taste and flavor.
Toxic and pathogenic biological substances created during food preparation, pack-
ing, and storage are biological pollutants. Toxins such as aflatoxins, ochratoxins, and
other foodborne microorganisms are the most common pollutants that cause
foodborne illnesses. Pathogenic bacteria include Campylobacter, Salmonella,
Listeria monocytogenes, E. coli, Staphylococcus aureus, and Bacillus cereus. Toxi-
cology can be detected using a variety of approaches. The most prevalent
mycotoxins generated by Aspergillus spp. are aflatoxin and ochratoxins. It is
cancer-causing, teratogenic, mutagenic, and immunosuppressive. A nanosensor
has been created to detect this mycotoxin using the immunosensor approach.
(Sharma et al., 2010). Using antibody-conjugated MNPs and a magnetic field,
Salmonella in milk was detected. Sack et al. (2004) defined Vibrio cholera as an
acute intestine infection induced by eating tainted food or water. Cholera toxin, a
colorimetric nanobiosensor detected with glycol nanoparticles, is secreted by this
bacteria (Schofield et al., 2007).
Unwanted dangerous chemicals are introduced into foods directly or indirectly from
natural sources, pollution, or formation during food processing, distribution, and
storage. Pesticides, heavy metals, and melamine are just a few chemical pollutants
288 P. Sudhakara et al.
Table 11.5 Nanobiosensors used in the detection of chemical and biological contaminants
Nanomaterial Contaminants Biosensor
Biological
Gold nanoparticle Aflatoxin Immunoelectrode
Silver core and a gold shell AF B1 Immuno-dipstick assay
Antigen-modified and antibody-functionalized AF B1 and OTA Immunosensing probes
DCNPs
Nanostructured zinc oxide Mycotoxin ITO glass plate
Magnetic nanoparticles and TiOz nanocrystals Salmonella Optical nanocrystal
probes
Oligonucleotide-functionalized Au nanoparticles Escherichia coli Piezoelectric biosensor
Glyconanoparticles Cholera toxin Colorimetric bioassay
Chemical contaminants
Gold nanoparticle Carbofuran Electroimmunosensor
Antibody competitive nanoparticle Chloramphenicol Immunosensor IPR
MTT-stabilized gold nanoparticle Melamine Colorimetric sensor
Quantum dots Melamine Colorimetric sensor
CNTs Penicillin Enzyme-based
biosensor
that harm human and animal health. Nanosensors have the potential to be a helpful
instrument for detecting chemical pollutants. Table 11.5 shows a variety of
nanosensors and nanobiosensors for detecting chemical and biological pollutants.
designed as an effective strategy for facilitating quick and exact estimation in the
search for a steady approach to accurate discoveries. Many amine oxidase enzyme
designs have been used to construct biosensors, to increase precise biorecognition
and signal transduction.
Substances like histamine were biogenic amines. Their presence varies with degrees
in several food products, and their increased concentration is considered an indicator
of the depleted level of freshness. Histamine intoxication is the most prevalent
foodborne infection among humans who consume fish products. If histamine con-
centration exceeds 500 ppm in fish products induces histamine poisoning (Gonzaga
et al., 2009). Bacterial contamination in fish gut and skin induces histidine decar-
boxylase activity and results in histamine production. Histamine levels were
screened by immobilized histidine decarboxylase and horseradish peroxide loaded
on the electrodes using the bovine serum albumin and glutaraldehyde complex.
Another study used the histamine oxidase enzyme from the bacterial
sp. Arthrobacter crystallopoietes (HOD) to build a biosensor. HOD biosensor was
effectively employed to measure histamine levels in fish and meat products, com-
paratively yielding a good agreement with the results obtained from HPLC analyses
(Rosini et al., 2014).
Amino acids have been extensively researched due to their nutritional importance
and speed up metabolic processes. D-amino acids (D-AAs) are, on the other hand,
primarily found in microbiological, aqueous, soil, and other environmental
pollutants. In general, the presence of D-AAs reduces protein digestion in the
body, resulting in decreased bioavailability of essential amino acids and nutritional
value. D-AAs are also utilized to evaluate the adverse effects of heat and alkaline
treatments on dairy products. D-AA biosensors based on the D-amino acid oxidase
(DAAO) enzyme were frequently employed to evaluate milk samples (Sarkar et al.,
1999; Rosini et al., 2008). The FAD domain is a cofactor expressed in humans, not
bacteria, in this peroxisomal enzyme.
The antioxidant activity of phenolic chemicals found in berries has been shown to
help prevent cardiovascular disease and aging and have anti-cancer properties. Since
the TYN enzyme converts hydroquinone in foods like red wine to p-quinone, it then
reduces to hydroquinone at the electrode and generates a current. The TYR enzyme
from Agaricus bisporus is an oxidase enzyme that catalyzes the conversion of
290 P. Sudhakara et al.
In recent days, heavy metal-induced toxicity among the food has been observed and
studied extensively. Heavy metals like Fe, Zn, Cd, Ni, Mn, Cr, Co, Pb, Cu, and Al
were leached from the eating utensils as well as cookware made up of iron, old
stainless steel, new stainless steel, old and new aluminum, and clay pots lead to
metallic contamination of food and water (Ghorai et al., 2009). In practice, the usage
of enzyme-based biosensors is used to detect these toxic substances like heavy
metals. Since these heavy metals react with the thiol groups in the enzymes like
glucose oxidase, urease, glutathione S-transferase, alkaline phosphatase, lactate
dehydrogenase, acid phosphatase, and invertase and biosensors, the function is
based on enzymatic inhibition. Nevertheless, limited enzymes have an affinity to
heavy metals.
Assessing the ethanol concentration and control is vital in brewing and winemaking
and also needs the exact data about the ethanol content in the beverages.
Immobilized alcohol oxidase catalyzes the reaction on the ethanol biosensor. Earlier
studies on ethanol biosensors used the combination of enzymes of alcohol oxidase
(AOx) along with horseradish peroxidase (HRP) and a mediator ferrocene
immobilized on an electrode. Later, Shkotova et al. established an amperometric
transducer and AOx-based biosensor immobilized on resydrol polymer and used to
detect ethanol concentration in alcoholic beverages. Another enzyme,
pyrroloquinoline quinone alcohol dehydrogenase (PQQ-ADH), is used in ampero-
metric biosensors to detect ethanol. PQQ-ADH has advantages like oxygen inde-
pendence and does not require a soluble cofactor. Certain studies employed the
bi-enzymatic biosensor to detect the ethanol concentration in alcoholic beverages.
substitute for natural sugars. As a result, fructose has been used to manufacture
beverages and foods (Antiochia et al., 2013).
On the other hand, overuse of fructose has been associated with high blood
pressure, decreased glucose tolerance, insulin resistance, and hepatic steatosis. For
many years, biosensors based on FDH have been used to detect fructose content in
food and beverages. To immobilize the NDFDH-modified nanoporous gold
electrodes, FDH was coupled with a thiol and diazonium-bound carboxylic acid
functional group. The fructose level in beverages may be accurately measured using
this manufactured electrode.
Lactate concentrations in red wine, white wine, beer, yogurt, curd, and milk samples
were used as biomarkers to determine the food quality. The food industry utilized
lactate biosensors to detect the desired level of lactate content in fermentation. Lactic
acid fermentation is induced by Lactobacillus spp. in fermented food products, and
the lactate content suggests an enhanced Lactobacillus population. Batra et al.
created an electrochemical lactate sensor using a PGE modified with graphene
oxide (GO) nanoparticles and LDH in 2016. This upgraded LDH–GO–PGE lactate
biosensor is currently being utilized to assess lactate levels in fermentation industries
(Batra et al., 2016). Lactose-related biosensors were also frequently employed, as
lactose intolerance affects roughly 15% of Northern Europeans, 80% of black and
Latino persons, and almost 100% of American Indians and Asians. These
individuals lack the digestive enzyme galactosidase, which converts lactose to
292 P. Sudhakara et al.
Foodborne infections that are infectious and toxigenic have been known for more
than a century. Salmonella, Staphylococcus aureus, and Clostridium perfringens
were the most common pathogens of concern worldwide by the mid-1900s. Botu-
lism was a fatal disease rarely linked to commercially canned foods, home canning
of vegetables, or traditional Arctic marine mammal products.
There was little interest in or research on acute foodborne illness agents during
those days. According to what was learned during outbreaks, most outbreaks might
have been averted if adequate time and temperature handling and storage protocols
had been followed, especially with poultry and beef. Clostridium, Salmonella, and
Staphylococcus issues were mainly resolved after the population was educated.
However, additional agents like Campylobacter, E. coli O157:H7, Listeria, and
Yersinia species emerged in the 1980s. It took many more years to realize that
these disorders might lead to life-threatening complications or even death and that
several goods could transmit them. Only a few of these diseases are detectable by
existing monitoring systems (Kothary & Babu, 2001; Wang et al., 2010; Fukushima
et al., 2011; Lindström et al., 2011). However, more and more international
outbreaks are being recognized thanks to DNA typing and next-generation sequenc-
ing technology.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 293
Because of contamination from humans, cattle, and the environment, several Staph-
ylococcus genus species and subspecies are potentially harmful to foods. Both
coagulase-negative and coagulase-positive strains can produce enterotoxins that
cause human gastroenteritis. S. aureus is most commonly associated with food
poisoning. Enterotoxins are the most dangerous of the several toxins produced by
staphylococci. Enterotoxins are proteins generated by other Staphylococci strains
that, if allowed to flourish in foods, form enterotoxin. S. aureus is the primary
producer of structurally related, toxicologically comparable proteins (Lancette &
Bennett, 2001).
294 P. Sudhakara et al.
Meat and beef products, poultry and egg products, salads such as bacon, fish, rice,
potato, and macaroni, baking products such as cream-filled pastries, and cream pies
are the most common foods associated with staphylococcal food poisoning. Some of
these products are polluted and mismanaged as a result. Chemical, animal, or
environmental contaminants can contaminate pre-processed foods. At the same
time, foods exposed to temperatures favorable to S. aureus growth have a signifi-
cantly greater capability to form enterotoxin. This is also true of meat and milk
products that have been fermented. Secreted enterotoxins are a structurally and
functionally related family of virulence factors with critical super-antigenic qualities
that disrupt adaptive immunity. Standard enterotoxins (SEA-SEE) and modern
enterotoxins are two types of enterotoxins (SEG-SElY).
The pathophysiology of several human diseases, such as toxic shock syndrome
(TSS), sepsis-related infections, and pneumonia, is determined mainly by members
of these classes. These have emetic properties and are frequently linked to food
poisoning outbreaks. The enterotoxins are primarily responsible for the disease’s
consequences when the patient lacks denaturation resistance. The enterotoxin genes
are frequently found on many mobile genetic elements. The prevalence of
enterotoxins varies greatly among S. aureus isolates, and it is regulated by numerous
overlapping regulatory mechanisms (Lancette & Bennett, 2001).
of food poisoning; and (3) the ability to produce an intestinally active enterotoxin,
which is responsible for C. perfringens’ characteristic gastrointestinal symptoms.
C. perfringens-related food poisoning is one of the most common causes of food
contamination. The association between C. perfringens and foodborne sickness was
originally postulated around 100 years ago, it was not until the 1960s and 1970s that
conclusive evidence emerged demonstrating that an enterotoxin is linked to the
organism’s sporulation in the intestines. The bacteria have several characteristics
that contribute to its capacity to cause foodborne illness:
children who died of sudden infant death syndrome (SIDS) (Health Protection
Agency, 2011).
Shigella is spread through fecal–oral, feces, direct touch with people (fingers), food,
flies, and inanimate items (fomites). More than two-thirds of all shigellosis cases
occur in youngsters aged one to five who appear to use their lips to explore their
surroundings. Intake of contaminated water is frequently cited as a method of
shigella transmission. When people are crammed together, disease transmission
and bacillary dysentery epidemics are intensified. Inadequate sanitation creates an
environment for direct fecal–oral contamination, such as in daycare centers, custo-
dial institutions, psychiatric hospitals, nursing homes, and mass displacement and
gathering, such as in wartime refugee camps or political unrest.
In addition, those infected with the human immunodeficiency virus (HIV) might
develop more severe and chronic types of shigellosis. Secondary attack rates among
household contacts could be as high as 40% following primary case exposure.
Shigellosis outbreaks appear to follow seasonal patterns in some areas, with
298 P. Sudhakara et al.
transmission peaking in arid regions like Egypt’s hot, dry season. During times of
water scarcity, it is primarily ascribed to dirty water use and poor personal
cleanliness.
On the other hand, peak events occur in China and Thailand during the rainy
season because of water-washed-based transmission after heavy rains. These repre-
sent the link between shigellosis and the unclean settings that encourage fecal
transmission. Shigellosis is most common in developing countries during the sum-
mer months, when raw foods, fresh fruits and vegetables, and recreational facilities
are most plentiful. Years later, the proportion of S. flexneri, S. sunny, S. boydi, and
S. dysenteriae cases in developing countries was estimated to be 60% (mainly
serotype 2a), 15%, 6%, and 6% (30% of S. dysenteriae cases were type 1), respec-
tively; and 16% (predominantly serotype 3a), 77%, 2%, and 1% in developed
countries, with nearly half of travelers registering incidents and returning travelers
from (Kothary & Babu, 2001).
Vibrio vulnificus is the opportunistic bacteria that cause foodborne illness and
devastating infections in humans. Ingestion of raw or undercooked seafood, con-
tamination of an open wound, or a sore contaminated with seawater or seafood
drippings are all possible entrance points. V. vulnificus produces wound infections
by entering existing or parallel wounds, ulcers, and insect bites, in addition to being
foodborne. Exposure to seawater or shellfish leaking from a cut is nearly always the
cause of these wounds. Puncture wounds from a bite from a marine animal (e.g.,
crabs, stingrays), stabbing or laceration wounds from the use of shellfish/fish
cleaning equipment, lacerations from commercial or recreational use of fishing
gear, rocks, or debris in or near seawater, or contamination of an existing wound
or insect bite with seawater or shellfish drippings are all reported wound scenarios.
Puncture wounds from a bite from a marine animal (e.g., crabs, stingrays),
stabbing or laceration wounds from the use of shellfish/fish cleaning equipment,
lacerations from commercial or recreational use of fishing gear, rocks, or debris in or
near seawater, or contamination of an existing wound or insect bite with seawater or
shellfish drippings are all reported wound scenarios. Because V. vulnificus is
associated with various seafood species, the health status is unknown. The human
sickness produced by this organism and other marine vibrios could become a
growing food safety concern. This problem is exacerbated by the fact that demand
for seafood is increasing, and it can now only be fulfilled by aquaculture and marine
fisheries sources harvesting seafood. These issues highlight the importance of
monitoring systems to track the safety of our seafood (Kothary & Babu, 2001).
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 299
Although cholera is a watery illness, new evidence suggests that food, particularly
seafood, plays an essential role in cholera transmission. Foods are likely to be fecally
contaminated during preparation, primarily if the diseased food handlers work in an
unsanitary setting. High moisture content, neutral or alkaline pH, low temperature,
high organic content, and lack of other competing bacteria are physicochemical
features of foods that promote V. cholerae O1 survival and development. Seafood,
such as fish, shrimp, crabs, oysters, and clams, have been blamed for cholera
epidemics in various nations, including the United States and Australia.
Dirty rice, millet gruel, and vegetables have been linked to epidemics. Cholera
can be transmitted through various foods, including fruit (excluding sour fruit),
chicken, meat, and dairy products. Food must be made and consumed in a sanitary
environment free of feces to prevent foodborne cholera transmission hazards. To
reduce foodborne cholera transmission, proper food preparation, storage, and
reheating before eating and hand washing with clean water before and after defeca-
tion are beneficial health interventions (Sack et al., 2004; Parveen et al., 2008).
11.18.5 Campylobacter
C. jejuni, C. colli, and C. lari have also polluted natural water surfaces. Consuming
unchlorinated, contaminated water or eating food prepared with untreated or
inadequately treated water might lead to infection.
In every corner of the globe, Campylobacter is one of the most common human
bacterial causes of diarrhea. Approximately 80% of Campylobacter infections are
thought to be caused by diet. Infection among young children is hyperendemic in
developed countries. Domesticated animals (chickens, pigs, goats, horses, dogs,
cats, and birds) are likely to get infected. Ingesting undercooked poultry, pork, and
beef, unpasteurized milk, contaminated drinking water, and infected animal excre-
ment can spread this virus to humans.
Campylobacter jejuni and C. coli invade domestic fowl with incredible ease. At
least 60% of chickens sold in supermarkets are contaminated with Campylobacter,
and broiler chickens are responsible for 50–70% of human infections in underdevel-
oped nations. The most common way to contract Campylobacter jejuni is by eating
undercooked chicken. Red meat, on the other hand, can be contaminated. Infection
can also be spread by eating barbecued pork or sausages. Infection can also be spread
by eating raw or undercooked fish, shellfish, or mushrooms.
In the recent past, raw and unpasteurized milk was a common source of Cam-
pylobacter and was responsible for large foodborne outbreaks. However, outbreaks
caused by raw milk have dropped significantly in developing nations due to the
regular pasteurization of milk and improved public awareness (Acheson & Allos,
2001).
11.19.1 Aflatoxins
Fusarium is a fungus that causes plant disease and is commonly found in soil.
Because they infect plants in the field rather than in grain storage, Fusarium species
infect cereal grains, known as “field fungus.” As of 1996, more than 60 species of
Fusarium have been identified in raw and cooked foods worldwide, with more than
100 secondary toxigenic metabolites characterized. Variations in mycotoxin-
producing potential have been identified between strains within the same species.
Detailed assessments of the various presence of the numerous mycotoxins produced
by the Fusarium species have recently appeared in the literature. The most potent
poisons to Fusarium include deoxynivalenol (vomitoxin), fumonisins, and
zearalenone from human exposure and regulatory concern.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 303
Most fungal species found in soil are susceptible to invasion by field crops. Many
fungal species can infect growing plants and produce secondary poisonous
compounds termed mycotoxins under ideal temperature and humidity conditions.
Some fungal species can multiply as plants and grain kernels age, producing
poisonous compounds that are not eliminated or destroyed during harvesting.
Assume that harvested grains (crops) are not stored properly. In that instance,
some fungus on the crops brought in from the field may continue to thrive and
create mycotoxins, or they may eventually be replaced by fungi that may multiply
under different storage circumstances and produce new mycotoxins. As a result,
mycotoxins can infiltrate the food supply throughout preharvest and postharvest
periods. Small amounts of these compounds in foods and feeds can be permissible,
as long as the amounts are not considered detrimental to human and animal health.
Part of the goal of this chapter is to provide an overview of the significant
mycotoxins that are causing significant public health problems at both the national
and worldwide levels. Aflatoxin A, deoxynivalenol, fumonisins, patulin, and
zearalenone are the most commonly regulated mycotoxins (Zinedine et al., 2007).
11.21.1 Caliciviruses
Caliciviruses are an enteric virus family that infects a variety of marine and terrestrial
organisms. While there are three genogroups of human caliciviruses, there are two
distinct physical and biological forms of human viruses: classic human caliciviruses
and Norwalk-like viruses. These viruses are known as small round standardized
viruses (SRSVs) in the literature. While classic calicivirus strains have been shown
to induce gastroenteritis in adults, classical caliciviruses are primarily associated
with self-limiting gastroenteritis in newborns and babies. Traditional caliciviruses
account for just a small percentage of instances of infantile gastroenteritis. As a
result, traditional caliciviruses are generally considered less medical and foodborne
concern than the NLV.
Multiple genotypes are occasionally discovered during outbreak investigations.
This common group infection goes undiscovered until an infected person
contaminates a typical food or water source or fosters rapid human-to-human
transmission through intimate contact with people in a closed or institutional setting.
Contact with dirty linens, vomitus, excrement, aerosols, or fomites can direct person-
to-person transmission in these conditions. NL disease outbreaks have been reported
at banquets, geriatric institutions, mental units, emergency rooms, cafeterias, recrea-
tional lakes, swimming pools, dormitories, campgrounds, hotels, schools,
restaurants, and cruise and navy ships (Scharff, 2012).
Although mussels are well known for spreading NL illness, other foods have also
played a role in the disease’s transmission. Viruses are commonly transmitted
304 P. Sudhakara et al.
through raw fruits and salads. Surface contamination of fruits and vegetables can
occur due to irrigation or fertilization, harvesters or transporters, or food preparation
contamination. Sick food handlers have contributed significantly to the spread of NL
illness by contaminating foods with unwashed hands or exposing objects to unsani-
tary surfaces. One illness outbreak in Newfoundland was caused by mixing potato
salad in a sink where a restaurant worker had vomited the day before. According to
epidemiological studies, many NL disease outbreaks are caused by a general lack of
awareness among some food handlers due to inadequate sanitation and hygiene
training.
11.21.2 Hepatitis
Hepatitis A and E are the only hepatitis viruses spread through the feces–oral route;
the others are primarily transmitted through parenteral routes. Hepatitis A virus
(HAV) is transmitted chiefly through fecal–oral, direct person-to-person contact,
and contaminated food and drink. For example, HAV is sexually transmitted,
particularly among homosexuals, and has been documented on multiple occasions
via blood products, particularly factor VIII. The hepatitis E virus is spread through
contaminated food and infected water. Except for vertical transmission, there is no
known parenteral transmission of the hepatitis E virus.
Virus-related disease burden varies substantially around the globe and is directly
influenced by socioeconomic growth and sanitation rates. The disease is more
prevalent in small and high endemic countries; in the former, infection and disease
are uncommon due to low virus circulation in the population due to adequate
cleanliness and sanitation. In countries with high endemicity, on the other hand,
the virus circulates widely. As a result, infection is common in young children, often
asymptomatic and unnoticed, contributing to long-term safety. This is true in areas
of intermediate endemicity, or countries with the highest disease rates, where
developed and developing people coexist (Mesquita et al., 2011).
The development of hepatitis A and B viral serological tests revealed that neither
virus was responsible for most hepatitis cases. This resulted in the coining of the
term non-A, non-B hepatitis, which had two epidemiologically distinct variants: a
parenteral type with transmission routes similar to parenterally transmitted hepatitis
B virus infection and an enterically distributed non-A, non-B hepatitis with trans-
mission routes similar to hepatitis A virus infection.
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 305
Some of the new techniques for microbial detection are ELISA, Lateral Flow
Immunoassay (LFIA), colloidal gold label-paper-based dipstick, and quantum dots
labels. ELISA has better sensitivity and lesser time consuming compared to culture-
based technique. The LFIA technique is a simple, rapid, feasible, and potent
foodborne pathogen detector. Other novel techniques to detect the antibodies are
306 P. Sudhakara et al.
Foodborne illnesses depend on the biofilm formation on the surface of the food
industry. The occurrence of biofilm changes the type of the food industry, such as
pipes, water, animal carcasses, dispensing tubes, and packing materials (Camargo
et al., 2017). The five critical foodborne pathogens that can form biofilm are Bacillus
cereus, Escherichia coli, Listeria monocytogenes, Salmonella enterica, and Staphy-
lococcus aureus.
As the distance between the producer and the consumer has grown due to globaliza-
tion, one of the significant issues is supplying quality food without contamination.
Food traceability is an essential criterion in ensuring food safety and quality.
Globalization has posed a challenge not only to food safety but also to food quality.
Consumers want to know everything there is to know about retailing history.
Traceability is crucial in this case. Traceability is based on unit identification,
product tracking, and data storage principles. The importance of food in the country
cannot be overstated. Food production is a global phenomenon that influences social,
economic, and environmental factors. Assume that any food-related difficulties will
impact the country’s socioeconomic condition. It has an impact on the environment
because of increased transportation carbon labeling. Food providers have recently
relied on two techniques to assure global food safety: managing the food chain by
adhering to regulations and standards and regulating logistics through traceability.
FAO and WHO have a significant role in food safety, FDA legislation, and Interna-
tional Standardization Organizations (ISO) participating in the international food
trade. Analyzing the risk Hazard Analysis and Critical Control Points (HACCP) is
essential for maintaining food safety standards. After HACCP, good manufacturing
procedures are implemented. Many rules and regulations were in place to assure
food quality and safety across the food supply chain (Aung & Chang, 2014).
The essential aspects of food are maintaining food safety and food quality. There
were numerous techniques to detect food safety primarily. We rely upon the
traditional method, which is time consuming and high cost. Then after several
findings in the 1960s, an electronic nose was introduced, which will mimic the
human olfactory system with its sensors. There were many factors for accepting and
rejecting the food and one among the important factor is the aroma, the smell of the
food. Aroma plays a crucial role in food. Aroma acts as a biomarker to test food
contamination. Few biosensors are listed in Table 11.6.
The food industry is benefiting from significant progress in developing enzymatic
biosensors with various transduction systems that can be used in food safety, quality,
and process control; studies are primarily focused on determining the composition
and contamination of raw materials and processed foods. The food industry is
benefiting from significant progress in developing enzymatic biosensors with vari-
ous transduction systems that can be used in food safety, quality, and process
control; studies are primarily focused on determining the composition and contami-
nation of raw materials and processed foods.
11.29 Conclusion
Intense research is ongoing to find a solution to the global food crisis. Enzyme
assimilation is well known in the food processing industry. This chapter addresses
the fundamentals of enzymes and the sources of various enzymes and their
applications in the food industry. The intrinsic features of microbial enzymes have
rocketed them to renown due to their simplicity of synthesis and yield manipulation
in laboratory settings. Enzymes in food processing convert physiochemical pro-
cesses to environmentally friendly processes, improve product biodegradability, and
reduce energy usage. The food processing industry could benefit from a new
approach for enzyme uses. Foodborne infections also harm human health and create
financial losses. As a result, rapid diagnosis of foodborne pathogens and the appli-
cation of countermeasures are critical. Preventing foodborne illness requires quick
detection of the pathogen. Enzymes can be used as biosensors to detect food
adulteration and contamination for faster food product screening. The dietary sub-
strate determines the enzyme biosensor’s characteristics. Molecular diagnostics and
serological methods are two new technologies for food sector monitoring.
While there are various methods for detecting food safety, the most common is
the time consuming and expensive method. The electronic nose stimulates the
olfactory system of humans using sensors, which is a breakthrough. Many aspects
influence whether or not a person accepts or rejects food. One of the most crucial
factors is the aroma of the dish. In food, the aroma is significant. The aroma as a
biomarker can detect the contamination in food. With the expanding dimensions of
the food industry, several critical procedures such as food preservation, monitoring,
adulteration, and foodborne diseases are critical to the industry’s overall perfor-
mance. Furthermore, enzymes perform multiple roles in all processes, one of which
is the constant monitoring of food and food products.
References
Abdollahzadeh, R., et al. (2020). Screening of pectinase-producing bacteria from farmlands and
optimization of enzyme production from selected strain by RSM. Folia Microbiologica, 65(4),
705–719. https://doi.org/10.1007/s12223-020-00776-7
Acheson, D., & Allos, B. M. (2001). Campylobacter jejuni infections: Update on emerging issues
and trends. Clinical Infectious Diseases, 32(8), 1201–1206. https://doi.org/10.1086/319760
Akyilmaz, E., Yorganci, E., & Asav, E. (2010). Do copper ions activate tyrosinase enzyme? A
biosensor model for the solution. Bioelectrochemistry, 78(2), 155–160. https://doi.org/10.1016/
j.bioelechem.2009.09.007
Ali, M., et al. (2011). Nanobiosensor for detection and quantification of DNA sequences in
degraded mixed meats. Journal of Nanomaterials, 2011. https://doi.org/10.1155/2011/781098
Alkasir, R. S. J., et al. (2010). Enzyme functionalized nanoparticles for electrochemical biosensors:
A comparative study with applications for the detection of bisphenol A. Biosensors and
Bioelectronics, 26(1), 43–49. https://doi.org/10.1016/j.bios.2010.05.001
Alvarez-Ordóñez, A., et al. (2013). Heterogeneity in resistance to food-related stresses and biofilm
formation ability among verocytotoxigenic Escherichia coli strains. International Journal of
Food Microbiology, 161(3), 220–230. https://doi.org/10.1016/j.ijfoodmicro.2012.12.008
Amine, A., et al. (2006). Enzyme inhibition-based biosensors for food safety and environmental
monitoring. Biosensors and Bioelectronics, 21(8), 1405–1423. https://doi.org/10.1016/j.bios.
2005.07.012
Andrei, V., et al. (2016). A single use electrochemical sensor based on biomimetic nanoceria for the
detection of wine antioxidants. Talanta, 156–157, 112–118. https://doi.org/10.1016/j.talanta.
2016.04.067
Antiochia, R., Vinci, G., & Gorton, L. (2013). Rapid and direct determination of fructose in food: A
new osmium-polymer mediated biosensor. Food Chemistry, 140(4), 742–747. https://doi.org/
10.1016/j.foodchem.2012.11.023
Aravindan, R., Anbumathi, P., & Viruthagiri, T. (2007). Lipase applications in food
industry. CSIR.
Aruna, K., Shah, J., & Birmole, R. (2014). Production and partial characterization of alkaline
protease from Bacillus tequilensis strains Csgab0139 isolated from spoilt cottage cheese.
International journal of applied biology an pharmaceutical technology, 5(3), 201–221.
310 P. Sudhakara et al.
Ashie, I. N. A. (2012). Enzymes in food analysis. In Food biochemistry and food processing
(pp. 39–55). Wiley Online Books. https://doi.org/10.1002/9781118308035.ch3
Aung, M. M., & Chang, Y. S. (2014). Traceability in a food supply chain: Safety and quality
perspectives. Food Control. https://doi.org/10.1016/j.foodcont.2013.11.007
Batra, B., Narwal, V., & Pundir, C. S. (2016). An amperometric lactate biosensor based on lactate
dehydrogenase immobilized onto graphene oxide nanoparticles-modified pencil graphite elec-
trode. Engineering in Life Sciences, 16(8), 786–794. https://doi.org/10.1002/elsc.201600082
Becker-Algeri, T. A., et al. (2016). Mycotoxins in bovine milk and dairy products: A review.
Journal of Food Science, 81(3), R544–R552. https://doi.org/10.1111/1750-3841.13204
Bhat, M. K. (2000). Cellulases and related enzymes in biotechnology. Biotechnology Advances,
18(5), 355–383. https://doi.org/10.1016/S0734-9750(00)00041-0
Bilal, M., & Iqbal, H. M. N. (2020). State-of-the-art strategies and applied perspectives of enzyme
biocatalysis in food sector—Current status and future trends. Critical Reviews in Food Science
and Nutrition, 60(12), 2052–2066. https://doi.org/10.1080/10408398.2019.1627284
Blanco, C. A., et al. (2014). Innovations in the brewing industry: Light beer. International Journal
of Food Sciences and Nutrition, 65(6), 655–660. https://doi.org/10.3109/09637486.2014.
893285
Borgmann, S., et al. (2012). Amperometric biosensors. In Advances in electrochemical science and
engineering (pp. 1–83). https://doi.org/10.1002/9783527644117.ch1
Health Protection Agency. (2011). Botulism, Epidemiological data.
Brown, C. A., et al. (2007). Outbreaks of renal failure associated with melamine and cyanuric acid
in dogs and cats in 2004 and 2007. Journal of Veterinary Diagnostic Investigation, 19(5),
525–531. https://doi.org/10.1177/104063870701900510
Calvo-Pérez, A., et al. (2013). Disposable amperometric biosensor for the determination of tyra-
mine using plasma amino oxidase. Microchimica Acta, 180(3), 253–259. https://doi.org/10.
1007/s00604-012-0926-y
Camacho, N. A., Aguilar, O., & G. (2003). Production, purification, and characterization of a low-
molecular-mass xylanase from Aspergillus sp. and its application in baking. Applied Biochem-
istry and Biotechnology, 104(3), 159–171. https://doi.org/10.1385/ABAB:104:3:159
Camargo, A. C., et al. (2017). Listeria monocytogenes in food-processing facilities, food contami-
nation, and human listeriosis: The Brazilian scenario. Foodborne Pathogens and Disease,
14(11), 623–636. https://doi.org/10.1089/fpd.2016.2274
Castillo, J., et al. (2004). Biosensors for life quality: Design, development and applications. Sensors
and Actuators B: Chemical, 102(2), 179–194. https://doi.org/10.1016/j.snb.2004.04.084
Chakdar, H., et al. (2016). Bacterial xylanases: Biology to biotechnology. 3 Biotech, 6(2), 150.
https://doi.org/10.1007/s13205-016-0457-z
Chandrasekaran, M. (2013). Valorization of food processing by-products. Retrieved from http://
www.crcnetbase.com/isbn/9781439848876.
Chaplin, M. F., & Bucke, C. (1990). Enzyme technology.
Chapman, J., Ismail, A. E., & Dinu, C. Z. (2018). Industrial applications of enzymes: Recent
advances, techniques, and outlooks. Catalysts, 8(6), 20–29. https://doi.org/10.3390/
catal8060238
Choi, E. J., Ahn, H. W., & Kim, W. J. (2015a). Effect of α-acetolactate decarboxylase on diacetyl
content of beer. Food Science and Biotechnology, 24(4), 1373–1380.
Choi, J.-M., Han, S.-S., & Kim, H.-S. (2015b). Industrial applications of enzyme biocatalysis:
Current status and future aspects. Biotechnology Advances, 33(7), 1443–1454. https://doi.org/
10.1016/j.biotechadv.2015.02.014
Christopher, N., & Kumbalwar, M. (2015). Enzymes used in food industry a systematic review.
International Journal of Inovative Research in Science Engineering and Technology, 4(10),
9830–9836. https://doi.org/10.15680/IJIRSET.2015.0410073
Clark, L. C., et al. (1958). Monitor and control of blood oxygen tension and pH during total body
perfusion. Journal of Thoracic Surgery, 36(4), 488–496. https://doi.org/10.1016/S0096-5588
(20)30126-4
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 311
Clark, L. C., & Lyons, C. (1962). Electrode systems for continuous monitoring in cardiovascular
surgery. Annals of the New York Academy of Sciences, 102, 29–45. https://doi.org/10.1111/j.
1749-6632.1962.tb13623.x
Colagiorgi, A., et al. (2017). Listeria monocytogenes biofilms in the wonderland of food industry.
Pathogens. Multidisciplinary Digital Publishing Institute, 6(3), 41.
Djekrif-Dakhmouche, S., et al. (2006). Application of a statistical design to the optimization of
culture medium for α-amylase production by Aspergillus niger ATCC 16404 grown on orange
waste powder. Journal of Food Engineering, 73(2), 190–197. https://doi.org/10.1016/j.
jfoodeng.2005.01.021
Ermis, E. (2017). Halal status of enzymes used in food industry. Trends in Food Science &
Technology, 64, 69–73. https://doi.org/10.1016/j.tifs.2017.04.008
Faulds, C. B. (2010). What can feruloyl esterases do for us? Phytochemistry Reviews, 9(1),
121–132. https://doi.org/10.1007/s11101-009-9156-2
Feng, W., et al. (2011). Isolation and characterization of lipase-producing bacteria in the intestine of
the silkworm, Bombyx mori, reared on different forage. Journal of Insect Science, 11, 135.
https://doi.org/10.1673/031.011.13501
Feuerstein, I., et al. (2006). Material-Enhanced Laser Desorption/Ionization (MELDI)—A new
protein profiling tool utilizing specific carrier materials for time of flight mass spectrometric
analysis. Journal of the American Society for Mass Spectrometry, 17(9), 1203–1208. https://doi.
org/10.1016/j.jasms.2006.04.032
Fukushima, H., Shimizu, S., & Inatsu, Y. (2011). Yersinia enterocolitica and Yersinia pseudotu-
berculosis detection in foods. Journal of Pathogens, 2011, 735308. https://doi.org/10.4061/
2011/735308
Galante, Y. M., De Conti, A., & Monteverdi, R. (1998). Application of trichoderma enzymes in
textile industry. In G. F. Harman & C. P. Kubicek (Eds.), Trichoderma and gliocladium
(Enzymes, biological control and commercial applications) (Vol. 2, pp. 311–326). Taylor and
Francis.
Gaur, R., & Tiwari, S. (2015). Isolation, production, purification and characterization of an organic-
solvent-thermostable alkalophilic cellulase from Bacillus vallismortis RG-07. BMC Biotechnol-
ogy, 15(1), 19. https://doi.org/10.1186/s12896-015-0129-9
Gavrilescu, M., & Chisti, Y. (2005). Biotechnology-a sustainable alternative for chemical industry.
Biotechnology Advances, 23(7–8), 471–499. https://doi.org/10.1016/j.biotechadv.2005.03.004
Ghorai, S., et al. (2009). Fungal biotechnology in food and feed processing. Food Research
International (Ottawa, Ont.), 42(5–6), 577–587. https://doi.org/10.1016/j.foodres.2009.02.019
Giaouris, E., et al. (2015). Intra-and inter-species interactions within biofilms of important
foodborne bacterial pathogens. Frontiers in Microbiology, 6, 841.
Gibson, G. R., & Wang, X. (1994). Regulatory effects of bifidobacteria on the growth of other
colonic bacteria. Journal of Applied bacteriology., 77(4), 412–420.
Gonzaga, V. E., et al. (2009). Histamine levels in fish from markets in Lima, Perú. Journal of Food
Protection, 72(5), 1112–1115. https://doi.org/10.4315/0362-028x-72.5.1112
Gouda, M. D., et al. (2002). Enhancement of operational stability of an enzyme biosensor for
glucose and sucrose using protein based stabilizing agents. Biosensors and Bioelectronics,
17(6), 503–507. https://doi.org/10.1016/S0956-5663(02)00021-0
Gupta, R., Gigras, P., Mohapatra, H., Goswami, V. K., & Chauhan, B. (2003). Microbial amylase:
A biotechnological perspective. Process Biochemistry, 38, 1599–1616.
Gurung, N., et al. (2013). A broader view: Microbial enzymes and their relevance in industries,
medicine, and beyond. BioMed Research International, 2013. https://doi.org/10.1155/2013/
329121
Hanft, F. and Koehler, P. (2006) ‘Studies on the effect of glucose oxidase in bread making’, Journal
of the Science of Food and Agriculture. John Wiley & Sons, Ltd, 86(11), pp. 1699–1704. doi:
https://doi.org/10.1002/jsfa.2455.
Hau, A. K., Kwan, T. H., & Li, P. K. (2009). Melamine Toxicity and the Kidney. Journal of the
American Society of Nephrology, 20(2), 245–250. https://doi.org/10.1681/ASN.2008101065
312 P. Sudhakara et al.
He, L., et al. (2018). Rapid and sensitive colorimetric detection of ascorbic acid in food based on the
intrinsic oxidase-like activity of MnO2 nanosheets. Luminescence, 33(1), 145–152. https://doi.
org/10.1002/bio.3384
Hernández, M. S., et al. (2006). Amylase production by Aspergillus niger in submerged cultivation
on two wastes from food industries. Journal of Food Engineering, 73(1), 93–100. https://doi.
org/10.1016/j.jfoodeng.2005.01.009
Hooda, V., et al. (2018). Alcohol quantification: Recent insights into amperometric enzyme
biosensors. Artificial Cells, Nanomedicine, and Biotechnology, 46(2), 398–410. https://doi.
org/10.1080/21691401.2017.1315426
Huck, C. W., et al. (2000). Development and evaluation of a new method for the determination of
the carotenoid content in selected vegetables by HPLC and HPLC—MS—MS. Journal of
Chromatographic Science, 38(10), 441–449.
Iqbal, S., & Selamat, J. (2016). Food safety; Basic concepts, recent issues, and future challenges.
https://doi.org/10.1007/978-3-319-39253-0.
Islam, F., & Roy, N. (2018). Screening, purification and characterization of cellulase from cellulase
producing bacteria in molasses. BMC Research Notes, 11(1), 445. https://doi.org/10.1186/
s13104-018-3558-4
Jeddah. (2011). Opinion: Halal food products: Lucrative business amid strong demand. The Saudi
Gazette - Business Section.
Jyoti, A., et al. (2011). Identification of environmental reservoirs of nontyphoidal salmonellosis:
Aptamer-assisted bioconcentration and subsequent detection of Salmonella typhimurium by
quantitative polymerase chain reaction. Environmental Science & Technology, 45(20),
8996–9002. https://doi.org/10.1021/es2018994
Kandra, L. (2003). α-Amylases of medical and industrial importance. Journal of Molecular
Structure: THEOCHEM, 666–667, 487–498. https://doi.org/10.1016/j.theochem.2003.08.073
Kaper, J. B., Nataro, J. P., & Mobley, H. L. T. (2004). Pathogenic Escherichia coli. Nature Reviews
Microbiology, 2(2), 123–140. https://doi.org/10.1038/nrmicro818
Karim, K. M. R., Husaini, A., & Tasnim, T. (2017). Production and characterization of crude
glucoamylase from newly isolated Aspergillus flavus NSH9 in liquid culture. American Journal
of Biochemistry and Molecular Biology, 7(3), 118–126.
Kieliszek, M., & Misiewicz, A. (2014). Microbial transglutaminase and its application in the food
industry. A review. Folia Microbiologica, 59(3), 241–250. https://doi.org/10.1007/s12223-013-
0287-x
Kim, C.-W., et al. (2010). Determination of spatial distribution of melamine cyanuric acid crystals
in rat kidney tissue by histology and imaging matrix-assisted laser desorption/ionization quad-
rupole time-of-flight mass spectrometry. Chemical Research in Toxicology, 23(1), 220–227.
https://doi.org/10.1021/tx900354z
Kothary, M. H., & Babu, U. M. A. S. (2001). Infective dose of foodborne pathogens in volunteers:
A Review. Journal of Food Safety, 21(1), 49–68. https://doi.org/10.1111/j.1745-4565.2001.
tb00307.x
Lancette, G., & Bennett, R. W. (2001). Staphylococcus aureus and Staphylococcal enterotoxins. In
K. Ito (Ed.), Compendium of methods for the microbiological examination of foods
(pp. 387–403). American Public Health Association.
Law, B. A. (2010). Enzymes in dairy product manufacture. In Enzymes in food technology
(pp. 92–93). Wiley Online Library.
Lee, B. L., Ong, H. Y., & Ong, C. N. (1991). Determination of thiamine and its phosphate esters by
gradient-elution high-performance liquid chromatography. Journal of Chromatography B: Bio-
medical Sciences and Applications, 567(1), 71–80. https://doi.org/10.1016/0378-4347(91)
80311-Y
Lépine, A. F. P., et al. (2019). Higher chain length distribution in debranched type-3 resistant
starches (RS3) increases TLR signaling and supports dendritic cell cytokine production. Molec-
ular Nutrition & Food Research, 63(2), e1801007. https://doi.org/10.1002/mnfr.201801007
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 313
Li, L., et al. (2010). Visual detection of melamine in raw milk using gold nanoparticles as
colorimetric probe. Food Chemistry, 122(3), 895–900. https://doi.org/10.1016/j.foodchem.
2010.03.032
Lindström, M., et al. (2011). Novel insights into the epidemiology of Clostridium perfringens type
A food poisoning. Food Microbiology, 28(2), 192–198. https://doi.org/10.1016/j.fm.2010.
03.020
Liu, L., et al. (2013). How to achieve high-level expression of microbial enzymes: Strategies and
perspectives. Bioengineered, 4(4), 212–223. https://doi.org/10.4161/bioe.24761
Luang-In, V., et al. (2019). Isolation and identification of amylase-producing bacteria from soil in
Nasinuan community forest, Maha Sarakham, Thailand. Biomedical and Pharmacology Jour-
nal, 12(3), 1061–1068. https://doi.org/10.13005/bpj/1735
Luong, J. H. T., Groom, C. A., & Male, K. B. (1991). The potential role of biosensors in the food
and drink industries. Biosensors and Bioelectronics, 6(7), 547–554. https://doi.org/10.1016/
0956-5663(91)80018-S
van der Maarel, M. J. E. C., et al. (2002). Properties and applications of starch-converting enzymes
of the α-amylase family. Journal of Biotechnology, 94(2), 137–155. https://doi.org/10.1016/
S0168-1656(01)00407-2
Marazuela, M. D., & Moreno-Bondi, M. C. (2002). Fiber-optic biosensors—An overview. Analyti-
cal and Bioanalytical Chemistry, 372(5–6), 664–682. https://doi.org/10.1007/s00216-002-
1235-9
Mascini, B. K. (2015) ‘Enzyme inhibition based biosensors for environmental monitoring’, in
Current enzyme inhibition. Bentham Science Publisher, p. 1–207. doi:10.2174/
157340805774580484.
Mecker, L. C., et al. (2012). Selective melamine detection in multiple sample matrices with a
portable Raman instrument using surface enhanced Raman spectroscopy-active gold
nanoparticles. Analytica Chimica Acta, 733, 48–55. https://doi.org/10.1016/j.aca.2012.05.001
Mello, L. D., & Kubota, L. T. (2002). Review of the use of biosensors as analytical tools in the food
and drink industries. Food Chemistry, 77(2), 237–256. https://doi.org/10.1016/S0308-8146(02)
00104-8
Mesquita, J. R., et al. (2011). Norovirus, hepatitis A virus and enterovirus presence in shellfish from
high quality harvesting areas in Portugal. Food Microbiology, 28(5), 936–941. https://doi.org/
10.1016/j.fm.2011.01.005
Mohan Kumar, N. S., et al. (2014). Reduction of acrylamide formation in sweet bread with
l-asparaginase treatment. Food and Bioprocess Technology, 7(3), 741–748. https://doi.org/10.
1007/s11947-013-1108-6
Mojsov. (2011) Applications of enzymes in the textile industry: A review. In: 2nd international
congress: Engineering, Ecology and Materials in the Processing Industry: Jahorina, Bosnia
and Herzegovina; Tehnoloski Fakultet Zvornik, pp. 230–239.
Monosik, R., et al. (2012). Application of enzyme biosensors in analysis of food and beverages.
Food Analytical Methods, 5(1), 40–53. https://doi.org/10.1007/s12161-011-9222-4
Monteiro, C. A., et al. (2011). Increasing consumption of ultra-processed foods and likely impact on
human health: Evidence from Brazil. Public Health Nutrition, 14(1), 5–13. https://doi.org/10.
1017/S1368980010003241
Moore, J. C., Spink, J., & Lipp, M. (2012). Development and application of a database of food
ingredient fraud and economically motivated adulteration from 1980 to 2010. Journal of Food
Science, 77(4), R118–R126. https://doi.org/10.1111/j.1750-3841.2012.02657.x
Nagodawithana, T. W., & Reed, G. (1993). Enzymes in food processing. Academic Press.
Nakamatsu, T., et al. (1975). Microbial production of glucose oxidase. Agricultural and Biological
Chemistry, 39(9), 1803–1811. https://doi.org/10.1080/00021369.1975.10861864
Nampoothiri, K. M., et al. (2002). Advances in fermentation technology. International Sugar
Journal, 104, 493–499.
Nielsen, S. S. (2017). Food analysis. Retrieved from http://rave.ohiolink.edu/ebooks/ebc/97833194
57765.
314 P. Sudhakara et al.
Norus, J. (2006). Building sustainable competitive advantage from knowledge in the region: The
industrial enzymes industry. European Planning Studies, 14(5), 681–696. https://doi.org/10.
1080/09654310500500239
Oumer, O. J., & Abate, D. (2018). Screening and molecular identification of pectinase producing
microbes from coffee pulp. BioMed Research International, 2018, 2961767. https://doi.org/10.
1155/2018/2961767
Panesar, P., Marwaha, S., & Chopra, H. (2010). Enzymes in food processing: Fundamentals and
potential applications.
Park, Y. W., et al. (2015). Application of biosensors in smart packaging. Molecular & Cellular
Toxicology, 11(3), 277–285. https://doi.org/10.1007/s13273-015-0027-1
Parveen, S., et al. (2008). Seasonal distribution of total and pathogenic Vibrio parahaemolyticus in
Chesapeake Bay oysters and waters. International Journal of Food Microbiology, 128(2),
354–361. https://doi.org/10.1016/j.ijfoodmicro.2008.09.019
Pasha, K. M., Anuradha, P., & Subbarao, D. (2013). Applications of pectinases in industrial sector.
International Journal of Pure and Applied Sciences and Technology, 16(1), 89.
Power, A. C., et al. (2018). Carbon nanomaterials and their application to electrochemical sensors:
A review. Nanotechnology Reviews, 7(1), 19–41. https://doi.org/10.1515/ntrev-2017-0160
Priya, V., & Sashi, V. (2014). Pectinase enzyme producing Microorganisms. International Journal
of Scientific and Research Publications, 4(1), 2250–3153. Available at: www.ijsrp.org
Prodromidis, M. I., & Karayannis, M. I. (2002). Enzyme based amperometric biosensors for food
analysis. Electroanalysis, 14(4), 241–261.
Qureshi, M. A., Khare, A. K., & Pervez, A. (2015). Enzymes used in dairy industries. International
Journal of Applied Research, 1(110), 523–527. Available at: www.allresearchjournal.com
Rajagopalan, G., & Krishnan, C. (2008). Alpha-amylase production from catabolite derepressed
Bacillus subtilis KCC103 utilizing sugarcane bagasse hydrolysate. Bioresource Technology,
99(8), 3044–3050. https://doi.org/10.1016/j.biortech.2007.06.001
Rankin, S. A., et al. (2010). Invited review: The application of alkaline phosphatase assays for the
validation of milk product pasteurization. Journal of Dairy Science, 93(12), 5538–5551. https://
doi.org/10.3168/jds.2010-3400
Raveendran, S., et al. (2018). Applications of microbial enzymes in food industry. Food Technol-
ogy and Biotechnology, 56(1), 16–30. https://doi.org/10.17113/ftb.56.01.18.5491
Rayappan, J. B. B., et al. (2017). Developments in electronic noses for quality and safety control.
Advances in Food Diagnostics, 63–96. https://doi.org/10.1002/9781119105916.ch3
Reddy, N., Nimmagadda, A., & Sambasiva Rao, K. R. S. (2004). An overview of the microbial
-amylase family. African Journal of Biotechnology, 2. https://doi.org/10.5897/AJB2003.
000-1119
Regalado, C., García-Almendárez, B. E., & Duarte-Vázquez, M. A. (2004). Biotechnological
applications of peroxidases. Phytochemistry Reviews, 3(1), 243–256. https://doi.org/10.1023/
B:PHYT.0000047797.81958.69
Richard, J., et al. (2003). Mycotoxins: Risks in plant, animal and human systems. CAST Task Force
Report, 139, 101–103.
Romero Gomez, S., Augur, C., & Viniegra-González, G. (2000). Invertase production by Aspergil-
lus niger in submerged solid-state fermentation. Biotechnology Letters, 22, 1255–1258. https://
doi.org/10.1023/A:1005659217932
Rosini, E., et al. (2008). A biosensor for all d-amino acids using evolved d-amino acid oxidase.
Journal of Biotechnology, 135(4), 377–384. https://doi.org/10.1016/j.jbiotec.2008.06.001
Rosini, E., et al. (2014). Evolution of histamine oxidase activity for biotechnological applications.
Applied Microbiology and Biotechnology, 98(2), 739–748. https://doi.org/10.1007/s00253-013-
5159-4
Sack, D. A., et al. (2004). Cholera. Lancet (London, England), 363(9404), 223–233. https://doi.org/
10.1016/s0140-6736(03)15328-7
11 Enzymes as a Tool in Food Analysis and Foodborne Pathogen Detection 315
Santos, A. S., et al. (2006). Amperometric biosensor for ethanol based on co-immobilization of
alcohol dehydrogenase and Meldola’s Blue on multi-wall carbon nanotube. Electrochimica
Acta, 52(1), 215–220. https://doi.org/10.1016/j.electacta.2006.04.060
Saqib, S., et al. (2017). Sources of β-galactosidase and its applications in food industry. 3 Biotech,
7(1), 79.
Sarkar, P., et al. (1999). Screen-printed amperometric biosensors for the rapid measurement of L-
and D-amino acids. The Analyst, 124(6), 865–870. https://doi.org/10.1039/a901404g
Sayali, P., & Satpute, S. (2013). Microbial esterases: An overview. International Journal of Current
Microbiology and Applied Sciences, 2, 135–146.
Scharff, R. L. (2012). Economic burden from health losses due to foodborne illness in the United
States. Journal of Food Protection, 75(1), 123–131. https://doi.org/10.4315/0362-028X.JFP-
11-058
Schofield, C. L., Field, R. A., & Russell, D. A. (2007). Glyconanoparticles for the colorimetric
detection of cholera toxin. Analytical Chemistry, 79(4), 1356–1361. https://doi.org/10.1021/
ac061462j
Sharma, A., et al. (2010). Antibody immobilized cysteamine functionalized-gold nanoparticles for
aflatoxin detection. Thin Solid Films, 519(3), 1213–1218. https://doi.org/10.1016/j.tsf.2010.
08.071
Sharma, S., et al. (2015). Bioprocessing of crop residues using fibrolytic enzymes and
Flavobacterium bolustinum for enriching animal feed. International Journal of Biotechnology
for Wellness Industries, 4(1), 12–17. https://doi.org/10.6000/1927-3037.2015.04.01.2
Siepenkoetter, T., Salaj-Kosla, U., & Magner, E. (2017). The immobilization of fructose dehydro-
genase on nanoporous gold electrodes for the detection of fructose. ChemElectroChem, 4(4),
905–912. https://doi.org/10.1002/celc.201600842
Sîrbu, T. (2011). The searching of active catalase producers among the microscopic fungi. Analele
Universitatii Din Oradea, Fascicula Biologie, 18(2), 164–167.
Sukumaran, R., Singhania, R., & Pandey, A. (2005). Microbial cellulases-production, applications
and challenges. Journal of Scientific & Industrial Research, 64, 832–844.
Tabssum, F., et al. (2018). RSM based optimization of nutritional conditions for cellulase mediated
Saccharification by Bacillus cereus. Journal of Biological Engineering, 12, 7. https://doi.org/10.
1186/s13036-018-0097-4
Terry, L. A., White, S. F., & Tigwell, L. J. (2005). The application of biosensors to fresh produce
and the wider food industry. Journal of Agricultural and Food Chemistry, 53(5), 1309–1316.
https://doi.org/10.1021/jf040319t
Theron, L. W., & Divol, B. (2014). Microbial aspartic proteases: Current and potential applications
in industry. Applied Microbiology and Biotechnology, 98(21), 8853–8868. https://doi.org/10.
1007/s00253-014-6035-6
Titball, R. W. (1993). Bacterial phospholipases C. Microbiology and Molecular Biology Reviews,
57(2), 347–366.
Tomme, P., Warren, R. A. J., & Gilkes, N. R. (1995). Cellulose hydrolysis by bacteria and fungi
(pp. 1–81). Academic Press. https://doi.org/10.1016/S0065-2911(08)60143-5
Tudorache, M., & Bala, C. (2007). Biosensors based on screen-printing technology, and their
applications in environmental and food analysis. Analytical and Bioanalytical Chemistry,
388(3), 565–578. https://doi.org/10.1007/s00216-007-1293-0
Turner, N. W., Subrahmanyam, S., & Piletsky, S. A. (2009). Analytical methods for determination
of mycotoxins: A review. Analytica Chimica Acta, 632(2), 168–180. https://doi.org/10.1016/j.
aca.2008.11.010
Vargas-Bernal, R., Rodríguez-Miranda, E., & Herrera-Pérez, G. (2012). Evolution and expectations
of enzymatic biosensors for pesticides (pp. 329–356). https://doi.org/10.5772/46227
Vasic-Racki, D. (2006). History of industrial biotransformations—Dreams and realities. Industrial
biotransformations, 40, 1–36.
316 P. Sudhakara et al.
Vasilescu, A., et al. (2016). Electrochemical affinity biosensors based on disposable screen-printed
electrodes for detection of food allergens. Sensors (Basel, Switzerland), 16(11). https://doi.org/
10.3390/s16111863
Venugopal, V. (2002). Biosensors in fish production and quality control. Biosensors and
Bioelectronics, 17(3), 147–157. https://doi.org/10.1016/S0956-5663(01)00180-4
Viswanathan, S., Radecka, H., & Radecki, J. (2009). Electrochemical biosensors for food analysis.
Monatshefte für Chemie - Chemical Monthly, 140(8), 891. https://doi.org/10.1007/s00706-009-
0143-5
Wachinger, G., et al. (1989). Identification of mycelium-associated cellulase from Streptomyces
reticuli. Applied and Environmental Microbiology, 55(10), 2653–2657. https://doi.org/10.1128/
AEM.55.10.2653-2657.1989
Wang, Q., Chang, B. J., & Riley, T. V. (2010). Erysipelothrix rhusiopathiae. Veterinary Microbiol-
ogy, 140(3–4), 405–417. https://doi.org/10.1016/j.vetmic.2009.08.012
Williams, D. C., Lim, M. H., Chen, A. O., Pangborn, R. M., & Whitaker, J. R. (1986). Blanching of
vegetables for freezing: Which indicator enzyme to choose. Food Technology, 40, 130–140.
Wingender, J., et al. (2016). Biofilms: An emergent form of bacterial life. Nature Reviews.
Microbiology, 14(9), 563–575.
Zhang, X.-Y., et al. (2015). Diversity of cultivable protease-producing bacteria in sediments of
Jiaozhou Bay, China. Frontiers in Microbiology, 6, 1021. https://doi.org/10.3389/fmicb.2015.
01021
Zhou, Q., et al. (2011). Development of gold nanoparticle-based rapid detection kit for melamine in
milk products. Journal of agricultural and food chemistry., 59(22), 12006–12011. https://doi.
org/10.1021/jf202919a
Zinedine, A., et al. (2007). Review on the toxicity, occurrence, metabolism, detoxification,
regulations and intake of zearalenone: An oestrogenic mycotoxin. Food and Chemical Toxicol-
ogy, 45(1), 1–18. https://doi.org/10.1016/j.fct.2006.07.030
Enzymes in Flavor Development and Food
Preservation 12
Fataneh Hashempour-Baltork and Parastou Farshi
Abstract
F. Hashempour-Baltork
Halal Research Center of IRI, Iran Food and Drug Administration, Ministry of Health and Medical
Education, Tehran, Iran
P. Farshi (*)
Food Science Institute, Kansas State University, Manhattan, KS, USA
e-mail: pfarshi@ksu.edu
# The Author(s), under exclusive license to Springer Nature Singapore Pte 317
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_12
318 F. Hashempour-Baltork and P. Farshi
Keywords
12.1.1 Introduction
In the modern food industry, different types of enzymes have been applied for the
synthesis of important food additives, food properties, and development of food
production processes. Flavor and fragrances have found wide application in the
food, feed, cosmetic, chemical and pharmaceutical sectors. Most of the flavor
compounds in the market are formed from animal and plant sources, which takes
place through chemical synthesis or extraction; however, there is a quick alteration
for the bio-production and use of flavor compounds with (micro) biological origin,
namely, bioflavors. Chemical synthesis usually results in an unfavorable and
environmentally unfriendly production process of racemic mixture compounds.
Moreover, the consumer has established a “chemophobia”-approach for synthetic
or chemical compounds, specifically for food and products being used in the home.
The definition of natural flavors by EC Flavor Directive (88/388/EEC), is “flavoring
ingredients or formulations which are achieved by suitable enzymatic,
microbiological, or physical processes from plant or animal origin materials”
(Vandamme & Soetaert, 2002). Until now, animal and plant sources were considered
to be essential sources of bioflavors; however, the amount of these compounds is
usually very low in these sources or they are only present in exotic (plant) species,
which results in their expensive formulation and isolation (Carocho et al., 2015).
However, nowadays biotechnology is considered as a preferred method with the
advantage of “natural” labeling that can draw consumers’ attention. De novo micro-
bial production (fermentation) and enzymatic synthesis are two biotechnological
ways to obtain bioflavors. Enzymatic flavor production is commonly preferred over
fermentation due to their high yields of production (Asunción Longo & Sanromán,
2005; Vandamme & Soetaert, 2002).
Enzyme immobilization and coenzyme regeneration techniques can lead to an
effective and exclusive biocatalytic processes for flavor synthesis (Kragl et al.,
1996). The benzaldehyde with almond and cherry taste can be made from the
cyanogenic glycoside amygdalin, which is present in almond meal and cherry
kernels, by mandelonitrile lyase and b-glucosidase enzymes (Fig. 12.1). An indus-
trial example for this is L-menthol production which is the major ingredient present
in peppermint oil (Fig. 12.2) (Menzel & Schreier, 2007).
Recently, synthesis of flavor compounds by biotechnological processes has
gained growing importance in the food industry. By considering the growing market,
and the growing public concern for the total wholesomeness and chemical safety of
food ingredients, researches on bioprocesses and inventing new products are also
12 Enzymes in Flavor Development and Food Preservation 319
increasing (Basset, 1990). In 2017, the total global flavors and fragrances market got
to US$28.2 billion, which means an increase of 4.6% rather than the previous year,
and it is estimated to increase at 4.9% average annual rate to get to approximately
US$36 billion in 2022 (Consultants, 2018). A wide range of options are provided by
enzymes, for food flavor production. The specificity of the enzymes and whether
they are being used through whole-cell or cell-free systems support the production of
chemicals that are hard to synthesize; also their stereoselectivity is considered as a
principal advantage for the food industry where a particular optical conformation can
affect flavor properties. Application of enzymes as food additives can produce or
liberate flavor from precursors, as well as correcting off-flavors produced by specific
naturally occurring compounds, or those that are produced during processing
(Bigelis, 1992). Besides intentional addition of enzymes to the food, they can also
be inherent to the food or may originate from microbial sources, or may come from
contamination.
In this chapter, several ways that enzymes can have effect on flavor are reviewed,
showing examples of actual research, their potential applications, as well as
examples of the fundamentals of biosynthesis and the market potential of some
products. In some instances, processes and reactions known for two decades are
stated and they are updated with recent developments, such as reaction in organic
media or whole-cell biocatalysts.
320 F. Hashempour-Baltork and P. Farshi
12.1.2.1 Lyases
Alliinases
Alliinases [S-alk(en)yl-L-cysteine sulfoxide lyase; alliin lyase; EC 4.4.1.4] remove
S-alk(en)yl-L-cysteine sulfoxides, and in this way they produce pyruvate, ammonia,
and thiosulfinates, (Fig. 12.3). Pyridoxal-50 -phosphate (PALP) is needed by this
enzyme as a cofactor.
The strong aroma and flavor of the most of the species of the Amaryllidaceae
family are because of the activity of this enzyme. Among other lyases, this enzyme is
detached from the substrate in the intact tissue, and the reaction occurs upon
breakdown of cells. The first alliinase to be explained was from garlic bulbs (Tishel
& Mazelis, 1966). Until now alliinases have been purified from onion, Welsh onion,
Chinese chive, garlic, and leek (Whitaker et al., 2002). Despite that they are from the
same genus, their specific alliinases are different in physicochemical properties. It is
many years that garlic and onion are being used by several cultures for different
purposes. In spite of having therapeutic and nutritional importance in diets, they are
being consumed around the world due to their unique flavor and their properties,
such as enhancing the flavors of other foods. Diallylthiosulfinate (allicin) is the
major sulfur compound in garlic that provides its distinctive aroma. This
thiosulfinate results from the elimination reaction by alliinase which is done on
S-2-propenyl-L-cysteine sulfoxide (alliin), via the intermediate 2-propenesulfenic
acid. There are three flavor precursors in onions, such as S-methyl-L-cysteine
sulfoxide, S-propyl-L-cysteine sulfoxide, and S-1-propenyl-L-cysteine sulfoxide.
Different thiosulfinates made by the activity of alliinase on the mentioned substrates
impart distinctive onion flavors, based on their concentrations (Masuda et al., 2012).
Other Allium spp. have diverse combinations of these substrates in addition to S-2-
propenyl-L-cysteine sulfoxide; this compound is not present in onion (Masuda et al.,
2012). It is stated that the lachrymatory factor in onion (syn-propanethial-S-oxide) is
originated from S-1-propenyl-L-cysteine sulfoxide (Jayathilaka et al., 2014).
Alliinases do the primary reaction which is responsible of producing distinctive
aromas and flavors in vegetables of the Allium genus. The maintenance of full
enzymatic flavor is the principal challenge of food processors who deals with
onion and garlic. Some preharvest and postharvest practices, such as environmental
factors influencing the flavor strength, including growing temperature, water supply,
and sulfur content of soil, are monitored in alliaceous foods to enhance their flavor
potential (Kamenetsky, 2007). Flavor potential of onions goes up with storage of its
bulb for a few months (Petropoulos et al., 2017). Nearly, half of the S-1-propenyl-L-
cysteine sulfoxide is bound as a peptide, L-glutamyl-S-allyl-L-cysteine sulfoxide, in
nature. It has been stated that the substrate is liberated by the activity of glutamyl
transferase. The most part of the flavor potential of the onion is lost during dehydra-
tion, which is due to the loss of the substrate more than the enzyme. In the other word
30% of the alliinase substrate is changed to cyclo alliin during the primary phases of
dehydration.
Cyclo alliin and the S-alkyl-L-cysteines are not substrates for alliinase. The
altered onion powder includes only one-third of the onion aroma of fresh onion
juice (Ramírez & Whitaker, 1999). Meticulous consideration of the speed of drying
is critical throughout the design of the dehydration process to diminish the substrate
loss; process with longer time normally leads to better yields. The activity of
alliinase can also lead to the development of unnecessary pigments in alliaceous
vegetables, such as pink pigments in onions and leeks, and green pigment in garlics
(Whitaker et al., 2002). Despite the ability of alliinase to catalyze the preliminary
reaction and thiopropanal-S-oxide formation (in onion), the formation of pigment is
dependent on the concentration of carbonyl and may differ from pink to red based on
the composition.
results from the activity of the mentioned enzymes and subsequently tissue softening
happens in these vegetables. Cystine lyase was identified to be the key enzyme in
broccoli, and its activity results in off-aroma decay (Barrett et al., 2000).
In a study, addition of the purified cystine to blanched broccoli was observed and
the enzyme-added broccoli could produce the aroma characteristic of the unblanched
broccoli. Chin and Lindsay (1993) determined the sulfur compounds produced in
damaged tissues of cabbage as dimethyl disulfide (DMDS), dimethyl trisulfide
(DMTS), and methanethiol (MT). Although these compounds are specific to brassica
vegetables, they are unfavorable for the consumer. These unpleasant sulfurous odors
make people to avoid consuming them, especially in the use of modified
atmospheres. The mechanisms of the formation of these compounds in cabbage,
which results from the activity of cystine or cystine sulfoxide lyase, were clarified
(Chin & Lindsay, 1994a). Researchers could not find significant differences between
the DMDS formation in anaerobic and aerobic circumstances; however, DMTS was
only identified in anaerobic conditions. Different treatments, such as soaking the
vegetable in caraway (Carum carvi) seed extract, ascorbic acid (500 ppm), phospho-
ric acid (0.1 M), sodium hydroxide (0.01 M), and tertiary butylhydroquinone
(TBHQ), have been done to decrease the levels of these undesirable volatiles in
broccoli stored in modified atmosphere condition (Chin & Lindsay, 1994b). All of
the mentioned treatments could decrease the intensity of sulfurous aromas, in which
alkaline treatment had the highest effect that could suppress the formation of DMDS.
Obenland and Aung (1996) discovered that the formation of MT and DMDS in an
intact broccoli flowerets can be nearly removed by infiltration of hydroxylamine
which is an inhibitor of cystine lyase, and can be increased 2.8 times by infiltration of
S-methyl cysteine sulfoxide before the anaerobiosis.
12.1.2.2 Isomerase
bonds in amylose and the main chain of amylopectin. However, the branches are
linked by 1,6-glycosidic bonds.
The starch which is produced from corn is exposed to liquefaction, in the first step
of enzymatic process, by application of a thermostable bacterial amylase
(EC 3.2.1.1) from Bacillus spp, such as B. stearothermophilus and
B. licheniformis. In this step, enzyme is able to produce a dextrin solution with a
dextrose equivalent (DE) of 10 by cleaving the 1,4 linkages in an endo fashion.
In the second step, other enzyme named as glucoamylase (amyloglucosidase; EC
3.2.1.3) releases single glucose moieties from the nonreducing end by hydrolyzing
the dextrins in an exo fashion.
Glucoamylase shows a higher activity for the 1,4-glycosidic bonds rather than the
1,6-branch bonds. Moreover, in some situations a debranching enzyme, such as
pullulanase (EC 3.2.1.41), is also used and the produced glucose solution has a DE
of 95.
In the next step of enzymatic process, other enzyme named as glucose isomerase
catalyzes the conversion of glucose into fructose until a concentration of 42% (molar
fraction based on total sugars). This reaction is exclusive because it is taking place in
a column reactor using immobilized form of the enzyme. Glucose isomerase was one
of the earliest enzymes used for this purpose on an industrial scale. This process is
done by encapsulating the enzyme in a packed-bed column reactor with 1- to 2-mm
particles (Ruiz-Matute et al., 2010). The diameter and height of the columns may be
1.5 m in and 4–5 m, respectively. This system has a high yield which produces
500,000 kg fructose (dry weight) per day by 4000 kg enzyme. High-glucose syrup
with DE ¼ 95, resulted from previous glucoamylase reaction with 45% solids and
pH adjusted to 7.5, is continuously pumped through the column. Several factors
have effect on the temperature and flow rate of the system. The flow rate should be
suitable to provide enough residence time for production of 42% fructose. Low flow
rates give rise to the risk of microbial contamination, also extremely high rates do not
provide enough conversion and result in channeling through the support material.
Temperatures below 55–60 C increase the syrup’s viscosity and increase the risk of
microbial contamination. However, higher temperatures decrease the stability of the
enzyme and results in the production of unfavorable by-products via chemical
reactions such as Maillard reactions.
Practically, temperatures of 55–60 C are being used. Regulation of the glucose
stream’s flow rate can compensate for the enzyme’s inactivation to maintain the
required conversion. These processes can continue for several weeks, under the
mentioned circumstances until the activity of the enzyme has become too low. After
getting to that point, the columns are disassembled and packed again with a batch of
fresh biocatalyst. The final HFCS is consisted of sugars, such as oligosaccharides
(up to 4%), fructose (42%), and glucose (54%) (Parker et al., 2010; Ruiz-Matute
et al., 2010).
12.1.2.3 Oxidoreductases
Oxidative and reductive processes play a significant role in foods. They affect many
aspects of food stuffs, such as taste, shelf life, texture, nutritional value, and
324 F. Hashempour-Baltork and P. Farshi
appearance. Both nonenzymatic and enzymatic redox processes are included. Some-
times processes lead to detrimental effects, such as textural problems, off-flavor, and
decreased shelf life. In some cases, also, they provide positive effects to the final
aroma, a better texture, a more desired appearance, or an improved shelf life. It is
very important to control redox behavior throughout all stages of processing and
storage, in food systems. Redox reactions in foods were controlled primarily by
addition of antioxidants or chemicals, designing air-tight packaging materials, cau-
tiously choosing raw materials, or by adjusting process situations, until now. How-
ever, there is not much paid attention to redox reactions in foods by the addition of
oxidoreductases or by modifying the profile or changing the content of
oxidoreductases in raw materials of foods by genetic implements. Apparently, the
main reason for this little attention for using oxidoreductases in foods is that it is still
not possible to produce most of the enzymes, cost-effectively. Moreover, general
concerns about the application of recombinant enzymes in foodstuffs are indicating
their commercial introduction. Various oxidoreductases are included in the in situ or
in vivo biogenesis of appropriate aroma compounds, in vitro production of flavors
and top notes, and in the endogenous development of off-flavors. The present
applications of oxidoreductases for controlling the taste of food products are
provided below:
Lipoxygenases
Lipoxygenase (LOX), which was previously known as carotene oxidase or
lipoxidase, is a dioxygenase containing iron, which catalyzes the oxidation of
polyunsaturated fatty acids comprising of cis,cis-1,4-pentadiene groups
(arachidonic, linolenic, and linoleic acids) to the equivalent conjugated cis-, trans-
dienoic monohydroperoxides. Moreover, LOX has a broad range of substrates which
are phenolic compounds (Markus et al., 1991); also it is able to oxidize other
substrates rather than the actual substrate. This process is identified as
co-oxidation and involves compounds, such as polyphenols and carotenoids. LOX
is one of the endogenous enzymes that plays a significant role in off-flavor formation
and flavor generation and in almost all food products it is originated from raw
materials of plant. Based on the type of food product and final concentration, a
flavor compound can be a desirable aroma component or an off-flavor at higher
concentrations. For instance, alcohols and C6-aldehydes derived from oxidation of
(poly)unsaturated fatty acids, which are catalyzed by the LOX, mostly have a
positive impact on the aroma profile (such as in wines and juices), but in other
products, such as beverages (e.g., beer), does not have desirable effect on flavor.
Similarly, endogenous LOX is known to have impact on the bread flavor due to
generating carbonyl compounds in dough systems (Pico et al., 2015).
The C6 compounds’ formation involves the consecutive action of four enzymes,
including two redox enzymes, such as a LOX, and an acylhydrolase, a yeast-derived
alcohol dehydrogenase, and a hydroperoxide lyase (Whitaker et al., 2002).
Normally, the formation of off-flavor is inhibited by the means of LOX
isoenzyme-deficient crop variants, by genetic tools or by screening or controlling
the oxygen levels during processing, in addition to eliminating the putrid oxidation
12 Enzymes in Flavor Development and Food Preservation 325
alcohol oxidases that are used for the formation of natural acetaldehyde from ethanol
(Lopez-Gallego et al., 2007). This enzyme is produced throughout growth on
methanol. Phase cells are collected and incubated with ethanol, at the final stage of
the logarithmic growth. Therefore, 1.5% natural acetaldehyde concentrations can be
reached, that can be further concentrated to the preferred application level. The
substrate specificity of the alcohol oxidase is different, in various yeasts. Hence, this
method can also be used to transform other alcohols, to their equivalent aldehyde.
Therefore, dehydrogenases can be used as substitutes to alcohol oxidases. However,
there is a limitation to use these dehydrogenases, because they need the costly
cofactor NAD(P)+instead of oxygen which is cheap, as an electron acceptor.
Although there are several designed complex NAD(P) cofactor stimulating systems
and significant cost reductions, it is obvious that oxidases are preferred in industrial
applications to their dehydrogenase equivalents. The application of dehydrogenases
for food purposes is limited to whole-cell transformations. There is a special kind of
alcohol oxidase, namely, vanillyl alcohol oxidase (VAO) which is derived from
Penicillium simplicissimum. Recently, it has been indicated that this enzyme, which
contains flavin and is relatively stable, has an extremely wide substrate specificity
and can convert para-substituted phenols into flavoring compounds or flavor
precursors (Ewing et al., 2018). In addition to the natural coniferyl alcohol and
vanillin, allylphenols and various vinylphenols, such as para-vinylguaiacol, can be
generated from cheap raw materials and oxygen (as an electron acceptor). VAO can
also be used for the production of building blocks of flavor. VAO can also be applied
to a natural mixture of phenolic compounds to enrich foods or flavor preparations
with a variety of aldehydic compounds and vinylic/allylic.
Sulfhydryl Oxidases
Formation of disulfide bonds from (protein) thiols is catalyzed by sulfhydryl oxi-
dase. This enzyme is a glycoprotein which contains Cu/Fe and has been detected in
goat, pig, human rabbit, bovine, and rat milks (Thorpe et al., 2002). The enzyme is
able to oxidize the cysteine and sulfhydryl groups of glutathione, and milk proteins
using molecular oxygen as electron acceptor, and transform them to their equivalent
disulfides (Faccio et al., 2011). SOX of bovine milk might be added to UHT milk for
reducing the cooked flavors. The enzyme has been immobilized on porous glass, and
its efficacy in improving the cooked flavor has been showed on a pilot scale through
the immobilized enzyme columns.
Peroxidases
Peroxidase (POX) happens extensively in nature and is the name which is generally
applied to a group of both nonspecific and specific enzymes which use hydrogen
peroxide as an electron acceptor. POXs, particularly the enzymes containing heme,
are able to catalyze a majority of various reactions, including N-demethylation,
oxidation, hydroxylation, and sulfoxidation; therefore, there is a special interest for
these enzymes in the production of specific flavoring top notes. Demethylation of
methyl N-methylanthranilate (ex Citrus) to mono-methylanthranilate is an example
of the application of these enzymes (Van Haandel et al., 2000). The mentioned
12 Enzymes in Flavor Development and Food Preservation 327
produced compound is the main top note flavor in Concord grapes. Horseradish,
microperoxidase, and soybean were found to be appropriate catalysts for this
reaction. Moreover, POX increases the fresh flavor profile when it is used in tomato
paste (Wilding & Woolner, 1997).
Polyphenol Oxidases
Polyphenol oxidase (PPO) enzymes are a group, including various enzymes with
different activities. Tyrosinase, a monophenol monooxygenase, is able to transform
a phenol into a catechol group. The other enzymes of this group, catechol oxidase or
1,2-diphenol oxidase (EC 1.10.3.1), converts catechol into an O-quinone, and
laccase is able to produce p-quinone from 1,4-diphenol. Generally, the first two
activities are related, since catechol is much more oxidized rather than a phenol. Also
most of the enzymes, that are able to catalyze the oxidation of 1,4-diphenol, can act
on 1,2-diphenols. In addition, guaiacols which are polyphenolic compounds impart
the bitter taste in several food products. PPOs are able to oxidize these compounds;
therefore, they can be used to decrease bitterness. There are several studies about the
application of these enzymes for this purpose, such as debittering of cacao beans
(Takemori et al., 1992), Adzuki beans (Okazawa et al., 1993), and coffee beans
(Small & Asquith, 1989). The other example of their application is the use of laccase
for debittering of olives (Charoenprasert & Mitchell, 2012).
12.1.2.4 Hydrolayses
rate of these systems is their main drawback (Aravindan et al., 2007). Commercial
lipases are mostly used in dairy products, meat industry, baked foods, fruit,
vegetables, beer, and wine (Nagodawithana & Reed, 1993). Immobilized lipase on
silica and microemulsion-based organogels are extensively being applied for syn-
thesis of ester (Ghosh et al., 1996; Sharma et al., 2001). Lipases are typically highly
specific. For instance, in a study Larios et al. (2004) in which the synthesis of esters
in n-hexane was considered, it was found that lipase (fraction B) from Candida
antarctica indicates substrate specificity. This specificity involves both acids and
alcohols. It was shown that it has activity on unsaturated fatty acids and short-chain
fatty acids with linear and branched chain structures, and 2-phenylethyl, n-butyl,
isopentyl, and geraniol as alcohols. Generally enantiomeric selectivity of enzymes is
among their important properties for food aroma synthesizing. As an example, only
(S)-form of 2-methylbutanoic acid methyl ester has the fruity flavor which is the
main flavor compound of apple and strawberry. It is found that lipases from
Aspergillus niger, Rhizomucor miehei, and Aspergillus javanicus have the selectivity
for this flavor compound (Kwon et al., 2000). Moreover, in a medium without
solvent, lipase from Candida rugosa was selected for L-menthol esterification
with long-chain unsaturated fatty acids to regulate the strong flavor related to it,
whereas it was acted weakly on D-menthol (Shimada et al., 1999). However, some
lipases indicate only restrained enantioselectivity, like Candida antarctica lipase
(fraction B) selectivity for chiral short-chain carboxylic acids (Larios et al., 2004).
Generally, lower water activity medium is preferred for lipase activity, so solvents
with high hydrophobic activity are more favorable due to their insolubility in water
layer around the enzyme surface which is important for protection of their active
stereoconfiguration (Huang et al., 1998; Langrand et al., 1990). Additionally, enzy-
matic synthesis of esters can take place in systems without solvent. In these systems,
reactant itself (i.e., an alcohol) acts as a solvent. This method is desirable in food
industry, but because of the heterogeneity of the reaction, there are limitations, such
as mass transfer and consequently low reaction rate in these systems. Researchers
could find a new thermostable esterase derived from a marine mud metagenomic
library, with potential use in industry, which is named as EST4. It is reported that this
enzyme can efficiently produce several short-chain flavor esters under high
concentrations of substrate and in media which is nonaqueous (Gao et al., 2016).
Researchers found that thermo-tolerant strain, such as Bacillus licheniformis
(cloned in E. coli), has good ability to produce thermophilic lipolytic enzymes.
These enzymes can tolerate high temperatures and organic solvents, which indicates
their promising feature to use them for ester synthesis (Alvarez-Macarie & Baratti,
2000; Dominguez et al., 2004; Fuciños et al., 2005). In a study by Chang et al.
(2001) the capability of mutant and wild-type lipases from Staphylococcus
epidermidis to catalyze the formation of flavor esters was investigated in aqueous
phase. This lipase was shown to have strong specificity for geranyl esters, unsatu-
rated esters, and medium-chain esters. Seitz has stated that Debaryomyces klockeri
and Candida mycoderma yeasts can produce lipases that have the best activity at
pH 4.5 (Seitz, 1974). In another study it is shown that lipases from Penicillium,
Aspergillus, and Paecilomyces, which are responsible for the fatty acid profile of the
12 Enzymes in Flavor Development and Food Preservation 329
cocoa bean, show different activities and specificities which are useful when they are
applied to milk fat (Hansen et al., 1973).
Applications of lipolytic enzymes require good interdependence between the total
substrate composition and the enzyme activity. For instance, the activity of milk
lipase was as a result of the effect of the both stimulatory and inhibitory activity of
several milk components on the enzyme (Hansen et al., 1973). In a study by Baianu
et al. (2003), the free lipase derived from M. miehei could conduct the direct
esterification of geraniol and citronellol with short-chain fatty acids, which had
high yields in n-hexane. According to Sánchez et al. (2002), flavor esters, such as
hexyl esters, produced by lipases are green note flavor compounds that are widely
used as flavor and fragrance in different industries, including food industry.
Recently, there is an increasing requirement for natural flavors, including “green
note” characterized by hexanol derivatives. For instance, hexyl butyrate is of
particular significance as it characterizes a model of flavor ester. Shieh and Chang
(2001) have investigated the capability of immobilized lipase from R. miehei to
catalyze the transesterification of tributyrin and hexanol. They determined that
decreasing the amounts of tributyrin as a co-substrate would decrease the cost for
the production of the hexyl butyrate. However, for transesterification of hexanol with
triacetin with the same lipase, results indicated that substrate molar ratio and
temperature of the reaction were the factors with the paramount importance and
the added water content had less impact on the reaction.
In dairy industry lipases and esterases are regularly used to enhance the buttery
flavor of the final product. Lipases are mostly used to hydrolyze milk fat. By
modifying fatty acid chain lengths, lipases can enhance flavor of different kinds of
cheeses. Short-chain fatty acids, specifically butanoic acid, play an essential role in
dairy products’ flavor and different commercial enzymes which have different
specificities towards short-chain fatty acids are considered as flavoring tools
(Saerens et al., 2008). There are several studies about the cheese ripening accelera-
tion and fat, cream, and butter lipolysis by lipases (Ghosh et al., 1996; Sharma et al.,
2001). Lipase from pregastric glands, such as bovine and porcine, and pregastric
tissues of ruminants were initially used for enhancing cheese flavor.
Various kinds of cheeses can be produced by using a specific kind of microbial
source or mixture of them. To produce enzyme-modified cheese (EMC) usually
cheese is incubated with enzymes at an increased temperature and in this way a
concentrated flavor is produced which can be used in sauces, snacks, soups, and
dips. In these cheeses the concentration of fat is tenfold higher rather than normal
cheeses (Ghosh et al., 1996; Sharma et al., 2001). Indigenous or exogenous milk
enzymes can contribute to lipolysis. Exogenous enzymes can be either microbial
lipases and/or mammalian pregastric esterase lipases. This process is an essential
factor in flavor development of cheese. From different reviews on lipolysis in cheese,
it can be derived that lipolysis results in the FFAs formation which are considered as
major flavor compounds of cheeses and can be transformed to other flavor
compounds, such as acetoacetate, aldehydes, β-keto acids, lactones, methyl ketones,
esters, and secondary alcohols which are more effective and can directly affect flavor
in different kinds of cheeses (Collins et al., 2003). Lactic acid bacteria (LAB) are
330 F. Hashempour-Baltork and P. Farshi
esters synthesizing in aqueous systems can be done through alcoholysis from other
alcohols and glycerides. For instance, it is found that St. thermophilus is able to
synthesize aromatic esters (2-phenyl ethyl hexanoate and 2-phenyl ethyl octanoate)
from an aromatic alcohol (2-phenyl ethanol) and two glycerides [monooctanoin
(mono-caprylin, C8) and dihexanoin (dicaproin, C6)](Liu et al., 2003a). Generally,
di- and monoglyceride substrates are preferred for alcoholysis by LAB esterases
which have the same specificity in hydrolysis (Liu et al., 2003a).
In cheese medium ester synthesizing can also done by selecting specific starters
that have high esterase or transferase activity, by changing the mono-, di-, and
triglyceride composition of the cheese fat glycerides, or by controlling ethanol
availability. For instance, in cheese made with mesophilic starters, by increasing
the ethanol level, ethyl hexanoate and ethyl butanoate concentrations are signifi-
cantly increased (Crow et al., 2002). In other study by spiking 1 M ethanol in
Cheddar-type cheese curd, during an incubation of 2 months, ethyl butanoate and
ethyl hexanoate are synthesized by the recombinant esterase from Lb. rhamnosus.
Availability of ethanol is possibly an essential factor in controlling ester formation in
cheese. Also primed cheeses with prehydrolyzed milk fat contain more esterases and
this is because of the presence of more di- and monoglycerides and it is found that
without addition of ethanol there is no ester formation in these cheeses. Moreover,
higher ester levels in cheese curd result in fruity aroma. In addition, some commer-
cial fungal lipases also found to synthesize esters from glycerides and ethanol in
cheese medium; also it is shown that they also have similar mechanism to LAB (Liu
et al., 2003b). Moreover, yeasts and molds have effect on ester formation in cheese
by producing the alcohols needed for ester synthesizing in cheese.
Salvadori (1961) could double the flavor of Pecorino cheese prepared with ewe’s
milk by addition of lipases taken from bacteria which were isolated from the
abomasum of lambs. In addition, it is found that esterase from Mucor miehei also
shows lipolytic activity in cheese similar to that achieved by pregastric oral esterases
(Richardson & Nelson, 1968).
In a study by comparing flavor characteristics of Romano cheese and Fontina
cheese containing Mucor miehei esterase to that of cheeses prepared with pregastric
esterase, it was found that by using fungal esterase and pregastric calf esterase at the
same lipolytic activity in the Fontina cheese, the desired flavor develops in both
cheeses. However, based on lipolytic activity, in Romano cheese it was necessary to
add a five time greater of fungal esterase, rather than pregastric kid esterase.
According to Baillargeon (1990), lipase from Geotrichum candidum which is a
dairy yeast has selectivity towards unsaturated fatty acids with cis-9 double bonds,
such as oleic, linoleic, and linolenic acids. Also, Yarrowia lipolytica (a dairy yeast)
could majorly release oleic and palmitic, from milk fat (Suzzi et al., 2001). It was
also previously found that G. candidum and Y. lipolytica can selectively release
unsaturated long-chain fatty acids from milk fat, in the cheese environment (Das
et al., 2005). However, these FAs have not favorable flavor and they impart a soapy
flavor to cheese, but they can be converted to conjugated linoleic acid (CLA) by
Propionibacteria which are usually used in Swiss cheese production (Jiang et al.,
1998). However, in another study researchers used three strains of
332 F. Hashempour-Baltork and P. Farshi
specificity to butanoic acid and production of buttery flavor in cream. Moreover, this
enzyme led to the highest concentration of free fatty acids (FFAs) (Saerens et al.,
2008).
Based on literatures milk lipases have insignificant importance in cheeses
prepared with pasteurized milk, and milk bacterial lipases, which are resistant to
thermal condition, can act better in cheese production from pasteurized milk.
Moreover, both bacterial and milk lipases have importance in milk fat hydrolysis,
whereas bacterial lipases and esterases (from Streptococcus cremoris) have signifi-
cant role in cheeses made from pasteurized milk (Chambers et al., 2010).
Using pregastric esterase in pasteurized milk for Cheddar cheese production
results in more FFA and flavor improvement; however, enzyme’s activity is lower
than its activity in raw milk. Also it was shown that the flavor of Cheddar cheese is
associated to a balance between acetate and FFAs and the ratio of 1.0 to 0.55 of
acetate: FFA results in the most favorable flavor. Microbial lipases increase FFAs
which affect the flavor of cheese (Collins et al., 2003). Kosikowski (1975) also
studied the prospective application of microbial lipases in continuous cheese
processing, and suggested good flavor development in Cheddar cheese from differ-
ent fungal lipases and in Blue cheese from Aspergillus sp lipase. However, organo-
leptic properties of Cheddar cheeses are increased in the presence of gastric lipase in
their production (Wolf et al., 2009). Moreover, there are studies about the effect of
lipase application in Feta (Karami et al., 2009) and Samsoe cheese (Jensen, 1970) in
their flavor development.
Lane and Hammer (1936) were the first people suggesting that homogenization of
cows’ raw milk or cream which is used in Blue cheese production can activate the
milk lipase in it; therefore, they concluded that homogenization has a great impact in
cheese flavor development and concentrations of volatile acids are two to fourfold
greater in cheese produced by homogenized milk. Studies showed that P. roqueforti
lipase acts specifically on triglycerides with smaller molecular weight. Secondary
alcohols and methyl ketones are fundamental compounds for the Blue cheese flavor
characteristics and it is known that they are derived from FFAs which are abundant
in Blue cheese (Tunick, 2007). Other researchers found out that methyl-n-amyl
ketone (2-heptanone) has a significant role in Blue cheese flavor characteristics,
which is suggested to be derived through the P. roqueforti activity on caprylic acid.
The methyl ketones’ formation and their metabolism by fungi in mold-ripened
cheeses act through different enzymatic mechanisms, such as release of FFAs
from triglycerides of milk fat by lipases (Walker & Mills, 2014). Dwivedi and
KINSELLA (1974) studied the continuous addition of lipolyzed milk fat and gradual
release of FFAs, in Blue cheese through the action of milk lipase. As a result, a
product was formed which was suitable for application in salad appetizers, dressings.
Its amount was 7- to 12-fold more than the ketone amount in a decent Blue cheese,
and its flavor efficacy was 4 times higher. Also these researchers reported the
production of methyl ketone by P. roqueforti from lipolyzed milk fat and suggested
that higher concentrations of FA had an impermanent inhibitory effect on the FAs
metabolization by enzyme.
334 F. Hashempour-Baltork and P. Farshi
Researchers, who studied the effect of pregastric esterases from lamb, kid, and
calf on bovine milk fat substrate, showed that these lipases have preference for
releasing short-chain FFAs. Each esterase had specificity to release butyric and other
short-chain FFAs and they were different in the release ratio of short-chain to long-
chain FFAs producing specific flavor characteristic while using cheese. It was found
that kid esterase has the highest specificity for butyric and other short-chain FFAs.
Studies on Provolone cheeses showed a direct relationship between FFAs (such
as butyric acid) and flavor quality and acceptance in Provolone and Romano cheeses
(Christensen, 1964; Collins et al., 2003). According to application of both lamb
pregastric esterase with lamb gastric extracts, results in more suitable “Provolone-
like” flavors, showing that addition of gastric lipase makes Provolone cheeses
organoleptically more acceptable. Overall, the extensive use of lipolytic enzymes
and commercial rennet paste in Italian cheese manufacture indicates the importance
of controlled lipolysis in this industry (Richardson & Nelson, 1968). It is found that
gastric lipase also has application for accelerating the cheese ripening and its flavor
development in different cheeses, such as Provolone, Ras, and Cheddar cheeses.
Addition of calf lipase increases the release of FAs (C4–C6) rate and accelerates the
development of flavor. The drawback of this method is that the lipase remains active
after ripening and can lead to development of an intense rancid flavor. By addition of
the mixture of fungal protease and lipase, relatively high-soluble proteins and FFAs
will develop in various cheeses which show better development of flavor in 3 months
of ripening. Also the amount of enzyme addition is of high importance and high level
of it results in unfavorable characteristics and decrease in the yield (Jahadi et al.,
2016).
Literatures indicated that using liposome technology for cheese ripening
decreases the losses in yield and bitterness (Jahadi et al., 2015; Vafabakhsh et al.,
2013). In cheese made from unpasteurized milk, the lipase which is inherent in the
cheese has significant effect in lipolytic action (Jahadi et al., 2020). P. roqueforti and
P. camemberti in Camembert and Blue-veined cheeses are considered as lipolytic
cultures and are responsible for lipases production, that accounts for lipolysis.
Moreover, lipases are often added to Italian cheese, such as Romano, Provolone,
and Paramesan to increase their flavor (Ghosh et al., 1996). Through ripening, there
is a constant rise in the concentration of released FAs and total soluble nitrogen
(Jahadi et al., 2012). Lipases are able to liberate the FAs from triglycerides, and in
this way they can trigger cheese flavor development (Maia et al., 1999; Mohammadi
et al., 2015).
In Gouda and Cheddar cheeses, esters, such as ethyl butyrate, have high contri-
bution to their flavor. Moreover, extra esters in Cheddar can impart fruity defect
(Caspia et al., 2006). Furthermore, there are compounds in Cheddar and Camembert
cheese, resulting from degradation of phenylalanine (such as phenylacetaldehyde,
2-phenylethanol, and the ester phenylethyl acetate), playing role in its floral rose-like
aroma and desirable floral note (Carunchia Whetstine et al., 2005).
There is an increasing interest for lipolytic enzymes in baking industry. Findings
suggested (phospho) lipases application as alternatives for producing emulsifying
lipids due to the enzymes ability to degrade polar wheat lipids to produce
12 Enzymes in Flavor Development and Food Preservation 335
emulsifying lipids in situ (Collar et al., 2000; Kirk et al., 2002). Lipase was mainly
used to increase the flavor in bakery products through esterification (releasing
short-chain fatty acids). In addition to flavor development, it also extended the
shelf life of most of the bakery products. Also softness and texture can be developed
by lipase catalyzation (Laboret & Perraud, 1999). Lipase from A. oryzae was also
used in baking industry, as processing aid (Greenough et al., 1996). Hydrolytic
enzymes, such as lipase, were discovered to be effective in decreasing the initial
firmness and enhancing the specific volume of breads (Laboret & Perraud, 1999).
According to Sánchez et al. (2002), yeasts with LIP A (bacterial lipase gene) result in
enzyme with high productivity which can be used in bread as an additive. Butter
flavor increases, by hydrolyzing the butterfat with an appropriate lipase in baked
foods.
Recent studies have investigated the possibility of application of various enzyme
systems for milk fat modification to incorporate them into bakery products. Gener-
ally, lipases of animal origin were found to be most suitable for milk fat modification
and including milk fat that is modified by these enzymes in bakery products led to
the formation of better flavors in them rather than milk fats modified by other
enzymes. Penicillium roqueforti, Geotrichum candidum, and Acromobacter
lipolyticurn lipases were mostly inappropriate due to the liberation of a large
concentrations of medium- or long-chain fatty acids and production of musty or
soapy flavors. Also lamb and kid esterases were not suitable due to the production of
excessive quantities of short-chain FAs imparting rancid flavors in bread
formulations (Arnold et al., 1975).
Vanillin (4-hydroxy-3-methoxybenzaldehyde), which is a generally desirable
flavor, mostly is present in Vanilla Planifolia beans. This flavor is extensively used
in foods (Priefert et al., 2001). Ferulic acid is precursor of vanillin and feruloyl
esterase has been recognized as the critical enzyme in the biosynthesis of it. This
enzyme can be produced in microbial cultures of several fungi grown on various
pretreated cereal brans (Mathew & Abraham, 2005). The metabolism of ferulic acid
in some microorganisms has also been investigated (Falconnier et al., 1994; Narbad
& Gasson, 1998).
Carboxyl esterases (carboxyl ester hydrolases) are enzymes extensively used in
different industries (Ewis et al., 2004). These enzymes prefer to catalyze the
hydrolysis of esters of short-chain FAs, but they also are able to catalyze the
synthesis of ester and transesterification reactions (Bornscheuer, 2002). Amid
these esters, flavor acetates from primary alcohols have a great application due to
their characteristic flavor and fragrance (Romero et al., 2005). Isoamyl acetate is also
one of the most significant flavor compounds used in the food industries and is
produced using lipases. In a research, the ability of type II esterase enzyme to
catalyze the esterification of isoamyl alcohol to isoamyl acetate which is less toxic
compound was studied. This compound is one of the most substantial flavor
compounds used in the food industries due to its distinctive banana flavor (Krishna
et al., 2001). This ester is widely used as a flavoring compound in several foods and
drinks, such as artificial coffee, honey, butterscotch, and alcoholic beverages. Pro-
duction of isoamyl acetate is typically doable through Fischer esterification
336 F. Hashempour-Baltork and P. Farshi
mechanism (Welsh et al., 1989) and all of the enzymatic synthesis reactions of this
compound were taken place using lipases (Krishna et al., 2001). Also there are
applications of lipases in ice cream and single cell protein in which C. rugosa lipase
has significant role in their flavor characteristics (Sánchez et al., 2002).
Tomato flavor, which is composed of a complex mixture of volatiles containing
multiple acetate esters, is resulted from esterases activity. It is believed that
red-fruited species of the tomato clade collects a quite low content of acetate esters
rather than the green-fruited species and the difference is related to the insertion of a
retrotransposon adjacent to the most enzymatically active member of family of
esterases, which results in higher expression of the esterase and decrease in the
levels of multiple esters that have negative impact on human preferences for tomato
(Goulet et al., 2012).
Studies on the flavor and odor characteristics of Suanyu (a traditional Chinese
fermented freshwater fish), in the presence of the esterase activity of Saccharomyces
cerevisiae 31, Lactobacillus plantarum 120, and Staphylococcus xylosus
135, indicated that all strains displayed esterase activities in both intracellular and
extracellular fractions against p-NP-butyrate, p-NP-octanoate, and p-nitrophenyl
(p-NP)-acetate. Moreover, it was found out that a low Aw (water activity), a low
pH, and also a high temperature would be more suitable for Suanyu processing,
which results in more flavor compounds which are favorable for consumers (Gao
et al., 2017).
Also in wine industry high levels of 4-vinylguaiacol and 4-vinylphenol were
identified from grape juice primarily treated with two enzymes. Firstly, cinnamoyl
esterase activity releases cinnamic acids from the related tartaric acid esters. Sec-
ondly, decarboxylase activity by the yeasts transforms cinnamic acids into 4-vinyl
phenol and 4-vinyl guaiacol (Dugelay et al., 1993).
In oil industry Cooney and Emerson strains of Mucor miehei esterase has some
functions. This enzyme have impact on natural fats such as lard oil, beef tallow, and
vegetable oils and some synthetic substrates, such as sorbitol esters of FAs. Also
fatty acid profiles which are resulted from the hydrolysis of beef tallow and soy oil at
pH 8.0 with both pancreatic lipase and M. miehei esterase, are comparable
(Moskowitz et al., 1977).
Protease
Proteases are enzymes able to break down the peptide bonds of proteins; they are
categorized as alkaline, neutral, and acid proteases. Animals, plants, and
microorganisms can be potential sources for these enzymes, in different conditions,
such as high salt concentrations. Wouters et al. (2016) explained that the flavor and
odor of the protein constituents have significant impact in food industry. In fact,
peptides and amino acids, besides other molecules, such as salts or sugars, define the
taste of foods. The human gustatory system is able to detect 5 basic flavors, such as
sweetness, bitterness, saltiness, sourness, and savoriness or umami. Sweetness,
bitterness, and savoriness are the major tastes associated with peptides (Iwaniak
et al., 2016). Bitterness needs a particular care, due to its cause for product rejection.
The peptides’ flavor can change based on their amino acid sequence. Studies have
12 Enzymes in Flavor Development and Food Preservation 337
indicated a correlation between the chain length of the peptides and their bitterness.
Their bitterness increases when they are made by up to 8 amino acid groups.
Furthermore, this is affected by its general hydrophobicity and the particular
amino acid positioned at the N- and C-terminus. For instance, the terminus position
of tyrosine or phenylalanine can determine the bitter taste (Iwaniak et al., 2016;
Maehashi & Huang, 2009). Therefore, selecting the right protease to accomplish the
hydrolysis of protein extract can reduce the unpleasant flavor in the final product and
also produce peptides with suitable tastes. For instance, during cheese production
and ripening, a continuous proteolysis is normally considered to be a required
condition for producing right flavor of cheeses (particularly Camembert or Brie)
(Singh et al., 2016). Moreover, proteolysis occurs during the main biochemical
modifications in cheese making, considering that in most circumstances, the first
step is an enzymatic coagulation. In the next steps, during the cheese ripening, a
progressive transformation of flavor and texture occurs. In fact, proteolysis is not
taking place only by the activity of external added enzymes but also it can be done by
enzymes of microorganisms. In Blue-veined cheeses, Blue molds generate a typical
odor and flavor through the conversions catalyzed by their enzymes. The same
happens at the moldy surfaces of cheeses, such as Camembert and Brie (Seratlić
et al., 2011).
Glutaminase
L-Glutamic acid is a verified flavor-enhancing amino acid which is extensively used
as a fundamental seasoning in food service, home cooking, and processed food
industries around the world. On the other hand, L-glutamic acid is originated from
proteins present in raw materials in fermented foods. L-Glutamic acid can also be
produced by the hydrolysis of L-glutamine in the protein hydrolysate; however, in
the absence of glutaminase, most of the released L-glutamine is transformed to
pyroglutamic acid which is tasteless (Fig. 12.5).
Soy sauce is a product known for its unique and strong umami taste, which is
created through its hydrolysis and fermentation. Umami taste induced by
monosodium L-glutamate has been widely accepted as one of the basic kinds of
tastes which is also named as savory, broth-like, or meaty taste. In fact, this taste is
linked to the taste characteristics and the chemical structure of L-glutamyl
oligopeptides. The presence of glutamic acid in di- or tripeptides is strongly
correlated with umami taste, because of the fact that the anionic L-glutamyl
oligopeptides can be present as umami taste (Zhang et al., 2017). Glutaminase
derived from Aspergillus oryzae is often used for the soy sauce fermentation, and
high salt concentrations in the soy sauce fermentation process can inhibit its activity.
Thus, salt-tolerant type of this enzyme have been studied for application in soy sauce
fermentation. Glutaminase enzymes from Micrococcus luteus, Bacillus subtilis, and
Aspergillus oryzae have been widely studied (Masuo et al., 2005). The glutaminase
of B. subtilis is commercially available. Zhuang and others (Zhuang et al., 2016)
indicated the significance of peptides’ effect on the umami taste of soy sauce. They
showed that the amino acid sequences of the peptides (Glu-Ala-Gly-Ile-Gln,
Ala-Leu-Pro-Glu-Glu-Val, Ala-Gln-Ala-Leu-Gln-Ala-Gln-Ala, Glu-Gln-Gln-Gln-
Gln, and Leu-Pro-Glu-Glu-Val), with the presence of glutamic acid/glutamine
persistent, are responsible for the umami taste in soy sauce.
Theanine (γ-glutamylethylamide) is an amino acid derived from a nonprotein
source that initially was isolated from green tea leaves (Sakato, 1949). This amino
acid is considered to be a distinctive amino acid in nature due to its limited
occurrence to the Camellia genus, especially the tea-producing plant, C. sinensis
(Nathan et al., 2006). Theanine is acknowledged for its unique umami taste. There-
fore, teas containing high theanine are normally considered to have higher quality.
Theanine can be formed from ethylamine and glutamate using sugar fermentation
reaction of baker’s yeast as an ATP regenerating system and bacterial glutamine
synthetase reaction (Nandakumar et al., 2003). There are some beneficial effects for
theanine, including improvement of learning ability, memory, and relaxation,
strengthening the immune system vascular diseases, prevention of cancer, and
weight gain (Kimura et al., 2007; Owen et al., 2008). In a mixture of γ-glutamyl
donor and γ-glutamyl acceptor, glutaminase from Pseudomonas nitroreducens has
been found to catalyze a γ-glutamyl transfer reaction. This enzymatic method has
been industrialized to produce theanine using glutaminase enzyme from
P. nitroreducens. For the enzymatic synthesis of theanine, GGT (γ-glutamyl
transpeptidase) catalyzes the hydrolysis of γ-glutamyl compounds and transmissions
of γ-glutamyl moiety from γ-glutamyl compounds, such as glutathione, to amino
acids and peptides. Also GGT was responsible for synthesizing other
physiologically significant γ-glutamyl compounds, including γ-glutamyl taurine
and γ-Glutamyl-L-3,4-dihydroxyphenylalanine (γ-glutamyl DOPA). Additionally,
bitter taste of some amino acids can be decreased by the mean of γ-glutamylization
by GGT (Suzuki et al., 2002).
Thermolysin
Synthetic sweeteners, involving anticariogenic and noncaloric sweeteners, have
been suggested to be a substitute to natural, calorie-containing sweeteners to
decrease the risk of cardiovascular diseases, diabetes, and obesity. For instance,
saccharin, aspartame, acesulfame potassium, and sucralose are examples of sugar
substitutes regularly used alone or with other natural sweeteners as food additives, in
several foods and soft drinks. Aspartame (α-aspartyl-phenylalanine methyl ester),
which is a peptide-type artificial sweetener, has been broadly used in processed and
pet foods, and beverages and drinks, such as tea, coffee, and sports drinks. The
sweetness degree of this sweetener is around 200 times higher than that of sucrose.
Aspartame thermolysin is a neutral protease from Bacillus thermoproteolyticus and
12 Enzymes in Flavor Development and Food Preservation 339
β-Galactosidase
D-glucoside glucohydrolase, also called lactase or β-Galactosidase, is able to cata-
lyze the hydrolysis of the galactosyl moiety from the nonreducing end of several
galactosyl-oligosaccharides. This enzyme is usually used in the dairy industry for
hydrolyzing lactose, Gal-(β1,4)-Glc, which is the main saccharide naturally present
in milk (Dutra Rosolen et al., 2015). Moreover, this reaction (hydrolyzing lactose to
galactose and glucose by β-galactosidase) inhibits the lactose crystallization in
condensed and frozen milk products besides strengthening the sweetness. Moreover,
β-galactosidase is capable of catalyzing transgalactosylation, in which a galactose
moiety is transmitted to the saccharides, emerging different galacto-oligosaccharides
(GOs). GOs have some advantageous effects, such as decreasing blood serum
cholesterol level, suppressing the colon cancer progress, and improving the mineral
absorption (Vulevic et al., 2013). The initial application of glycosidases was in wines
which was used to liberate flavor compounds from glycosidic precursors. The first
reason for this is that significant flavor compounds in wines from Vitis vinifera
cultivars are present as flavorless glycoconjugates in grapes, and the second reason is
that the glucose inhibitory effect of -glucosidases restricts their use to the media
containing low levels of glucose (<1 g/L), such as wine. However, due to the
presence of flavor glycoconjugates in various fruits, enzyme applications have also
been reported for fruit juices.
A. Wine Flavor Enhancement: During grape juice fermentation, there is only a
slight decrease in the amount of glycosidic flavor precursors. This can be described
by the low levels of glycosidase activities of enological yeast strains and grapes
(Hernandez-Orte et al., 2008). Therefore, there is a special interest for the use of
enzymes, such as pectinases and hemicellulases, which are commercially available
fungal enzymes (mainly derived from Aspergillus spp.) that also have glycosidase
activities. However, recent regulations do not accept including the enzymes in the
wine. The hydrolysis of monoterpenyl glycosides in muscat cultivars releases
odorous alcohols that can be identified by sensory analysis and can be analyzed by
GC/MS. Generally, the addition of glycosidase during wine making increases in the
concentration of monoterpenes, shikimate-derived compound, and
C13-norisoprenoids (Hernandez-Orte et al., 2008).
B. Fruit Juices: There are few studies about the use of glucosidases in fruit juices
which is due to the inhibition of them by glucose of the “key enzyme” glucosidase in
enzyme derived from fungi. According to a study curing passion and mango fruit
juices with a glucosidase derived from Candida cacaoi resulted in a rise in mono-
terpene concentrations (Chanprasartsuk & Prakitchaiwattana, 2015). Nevertheless,
340 F. Hashempour-Baltork and P. Farshi
this increase was pretty low rather than acid hydrolysis. Moreover, immobilization
of the enzyme on sodium alginate beads, released less terpenols rather than the free
enzyme. The addition of immobilized C. molischiana-derived glucosidase (onto
Duolite A-568 resin) to apple juices, mango, strawberry, apricot, and peach, caused
a rise in the concentration of monoterpene alcohols, such as 2-phenylethanoal,
geraniol, terpineol, benzyl alcohol, and linalool (Pogorzelski & Wilkowska, 2007).
However, there were no differences in the flavor liberations using immobilized and
free glucosidase from C. molischiana. The immobilized enzyme was stated to be
much more stable in fruit juice settings. In passion fruit juice, an immobilized (onto
cellulose or acrylic beads) endo glucosidase derived from A. niger increased the
concentration of volatile compounds, such as benzyl alcohol, benzaldehyde, and
linalool. Generally, an aroma improvement was determined in the treated juices
(Pogorzelski & Wilkowska, 2007).
Although by application of glucosidase, the levels of volatile compounds in fruit
juices have increased, there were two significant parameters restricting the effective-
ness of the process: firstly, it is known that there are lots of flavor compounds in fruit
juice, such as diglycosides, that in general, fungal glucosidases are not able to
hydrolyze them; secondly, the examined fungal glucosidases are prevented by
glucose, which might then constraint their action in fruit juices. There is a report
about the process that the amount of glycosides alters in a sweet wine, with 30 g/L
glucose, in the presence of fungal glycosidases. It is shown that the monoterpene
diglycosides’ level decreases remarkably in the enzyme-treated wine, which can be
described by the activity of exoglycosidases. The produced monoterpenyl-D
glucosides were not hydrolyzed and consequently they were accumulated in the
wine. This clearly shows that the fungal glucosidase activity is inhibited by glucose,
which significantly limits the release of volatiles as it is approved by their analysis.
In contrary, the hydrolysis of monoglucosides was very efficient though a dry wine
was made using the same conditions, such as the same enzyme dosage and grape
juice (Pogorzelski & Wilkowska, 2007).
Naringinase(α-L-Rhamnosidase)
Naringinase (EC 3.2.1.40) is mostly used for the naringin’ breakdown which is the
main bitter flavanone glycoside present in citrus fruits (Puri, 2012). Through the
activity of α-rhamnosidase and β-glucosidase, naringin is converted to a naringenin
and rhamnose. The sources for naringinase are mostly fungal isolates, such as
Circinella, Eurotium, Aspergillus niger, Rhizopus, Trichoderma, Fusarium, and
Penicillium, and bacteria, such as Pseudomonas paucimobilis, Bacillus sp.,
Thermomicrobium roseum, Bacteriodes distasonis, and Burkholderia cenocepacia
(Puri, 2012). Naringinase derived from fungi has more application rather than the
bacterial ones due to increased yield. Naringinase, as a debittering enzyme, has a
principle role in food processing when it is added to fruit juices, in both immobilized
and free forms. There are several food additives, such as sweeteners and
biopolymers, which can be produced by naringinase or rhamnosidase. Moreover,
there is another application for naringinase in addition to arabinosidase and
β-glucosidase to develop the aroma of wine (Custodio et al., 1996). It is also stated
12 Enzymes in Flavor Development and Food Preservation 341
that naringinase can be used in preparation of tomato pulp and prunin, and treatment
of kinnow peel waste (Puri, 2012).
Citrus juice is considered as one of the most commonly used juices in the world.
However, mostly the taste of it is too bitter and this bitterness is mainly because of
the presence of the naringin in the citrus fruits. Naringinase (α-L-rhamnosidase) is
used to diminish the bitterness by breaking naringin into rhamnose and prunin and it
is regularly used in the industrial production process of citrus juice (Puri et al., 2005).
β-glucosidase, which is responsible for hydrolyzing the prunin to naringenin
(a flavorless flavanone) and glucose, is usually used with naringinase, for a success-
ful debittering (Table 12.1).
12.2 Preservation
12.2.1 Introduction
It is undeniable that food is important for mankind and there is not any way of living
without eating. Therefore, it is highly important for the health of everyone to eat
food. Although the need to feed did not change during the years, the way of eating
has seen a lot of changes. Nowadays, food additives are needed to make various
foods that are meeting the progressively challenging market and legal demands
(Saltmarsh & Saltmarsh, 2013). Foods can be made in different facilities and then
can be transported to local markets that are inside the same country or even in distant
ones (Atkins & Bowler, 2016). Food transportation in suitable conditions needs
special equipment and considerations, such as controlled packaging, refrigeration, or
using some additives to prevent food spoilage and diminish food alterations. In
today’s global market which is highly competitive, it is desirable to use the cheapest
method of food preservation and food additives can be a good choice.
12.2.2 Lysozyme
Antimicrobials with animal origin are compounds, such as proteins and enzymes that
are isolated from animals. Currently, the only certified natural antimicrobial with
animal origin that is used in US and the EU is lysozyme (E-1105). This enzyme is
derived from eggs (Carocho et al., 2015), and it has high antimicrobial activity
against gram-negative bacteria. The reason for this higher activity is that gram-
negative bacteria are mostly (90%) composed of peptidoglycan, and lysozyme’s
antimicrobial activity is dependent on the hydrolysis of the β-1,4 linkage site of the
peptidoglycan in the bacterial walls. This enzyme has only a moderate effect against
gram-positive bacteria that have much less peptidoglycan in their wall, and appar-
ently it has no activity against fungi or yeasts (Barbiroli et al., 2012). It is reported
that the principal commercial use of this natural antimicrobial is in the cheese
industry, that is used for prevention of “late blowing” of cheese; also there are
studies about its application in eggs, beef (200 mg/90 mg), and milk (2 mg/mL in
342 F. Hashempour-Baltork and P. Farshi
25 mL milk) (Sung et al., 2011). Lysozyme has also been assessed for making edible
coatings and biofilms (Barbiroli et al., 2012) and its synergistic effects were studied
in corporation with other natural antimicrobials (Bayarri et al., 2014).
12.2.3 Oxidoreductase
Redox enzymes have applications for extending the shelf life of food products.
These enzymes include those that able to remove oxygen or reactive oxygen species,
such as H2O2 and superoxide anion, and those that are capable of generating
antimicrobial agents. Therefore, the stability of foods can considerably be improved
with regard to their appearance, taste, and microbial spoilage.
12.2.3.1 Lactoperoxidases
Lactoperoxidase which belongs to the superfamily of peroxidase-cyclooxygenase
enzymes is the most important enzyme present in the bovine milk in concentrations
of approximately 30 mg/L. This enzyme shows its antimicrobial activity through the
lactoperoxidase system, including lactoperoxidase, hydrogen peroxide, and thiocya-
nate, which are referred to as the lactoperoxidase system (LPS). The intermediate
compounds formed through the oxidation of thiocyanate have antimicrobial activity
(Silva et al., 2014). The main application of lactoperoxidase enzyme is to preserve
raw milk, specifically in places that it is not readily available to refrigerate it. By
344 F. Hashempour-Baltork and P. Farshi
addition of thiocyanate to the milk, the LP system will start showing its antimicrobial
activity. It is necessary to add thiocyanate, because of the low amount of thiocyanate
in the milk which is not enough to initiate the antimicrobial activity. The antimicro-
bial activity of the LP system is activated by production of thiocyanate (SCN)
oxidation products, primarily hypothiocyanate ions (OSCN), that are able to attack
the sulfhydryl groups of major metabolic enzymes of the microorganisms. More-
over, the LP system does not affect the mammalian cells. Usually barely 10–20 ppm
of lactoperoxidase is necessary for an efficient system. Also the required amount of
thiocyanate and H2O2 is pretty low which are 10–25 ppm and 10–15 ppm, respec-
tively. H2O2 is also bactericidal, but it shows its antimicrobial activity at higher
concentrations (300–900 ppm) (Martin et al., 2014). Hence, lactoperoxidase (LPO)
is usually used in combination with enzymes that generate H2O2. Moreover, in the
LP system the amounts of cofactors and also the oxidation products are toxicologi-
cally harmless. LP system has applications in dental products, veterinarian products
such as antidiarrheal, milk replacers, and antimastitis, and food products such as
functional foods, cheese, fish, meat, poultry products, and liquid milk. There are
claimed applications of this system alone or in combination with other systems for
pickled foods, fish products, Lactobacillus fermented milk products, and white mold
cheeses (Laane & Bruggeman, 2002). Moreover, there is an interest for its effect on
yogurt. By addition of LPO to yogurt, the production of unnecessary acid of LAB in
it is stifled (Masud et al., 2010). The mentioned applications are becoming accessible
now that isolating the lactoperoxidase of milk is doable with high purity on an
industrial scale. This system is commercially available now, at considerably low
costs. Latest applications recommended for lactoperoxidase are preservation of fruit
juices, and also using as coating of foodstuffs (Cissé et al., 2015).
References
Alvarez-Macarie, E., & Baratti, J. (2000). Short chain flavour ester synthesis by a new esterase from
Bacillus licheniformis. Journal of Molecular Catalysis B: Enzymatic, 10, 377–383.
Antrim, R. L., & Taylor, J. (1990). Deodorization of a water-inoil emulsion containing fish oil using
pure alcohol and aldehyde oxidases/dehydrogenase in combinations with a cofactor. Patent
US4961939.
Aravindan, R., Anbumathi, P. and Viruthagiri, T., 2007. Lipase applications in food industry.
Arnold, R., Shahani, K., & Dwivedi, B. (1975). Application of lipolytic enzymes to flavor
development in dairy products. Journal of Dairy Science, 58, 1127–1143.
Arpigny, J. L., & Jaeger, K.-E. (1999). Bacterial lipolytic enzymes: Classification and properties.
Biochemical Journal, 343, 177–183.
Asunción Longo, M., & Sanromán, M. (2005). Production of food aroma compounds: Microbial
and enzymatic methodologies. Food Technology and Biotechnology, 44, 335–353.
Atkins, P., & Bowler, I. (2016). Food in society: Economy, culture, geography. Routledge.
12 Enzymes in Flavor Development and Food Preservation 347
Baianu, I., Lozano, P., Prisecaru, V., & Lin, H. (2003). Novel techniques and their applications to
agricultural biotechnology, health foods and medical biotechnology (p. 13). University of
Illinois at Urbana, ACES College.
Baillargeon, M. W. (1990). Purification and specificity of lipases from Geotrichum candidum.
Lipids, 25, 841–848.
Barbiroli, A., Bonomi, F., Capretti, G., Iametti, S., Manzoni, M., Piergiovanni, L., & Rollini,
M. (2012). Antimicrobial activity of lysozyme and lactoferrin incorporated in cellulose-based
food packaging. Food Control, 26, 387–392.
Barrett, D., Garcia, E., Russell, G., Ramirez, E., & Shirazi, A. (2000). Blanch time and cultivar
effects on quality of frozen and stored corn and broccoli. Journal of Food Science, 65, 534–540.
Basset, F. (1990). Arômes naturels: Oui aux biotechnologies. Parfums, Cosmétiques, Arômes,
17–26.
Bayarri, M., Oulahal, N., Degraeve, P., & Gharsallaoui, A. (2014). Properties of lysozyme/low
methoxyl (LM) pectin complexes for antimicrobial edible food packaging. Journal of Food
Engineering, 131, 18–25.
Beniwal, V., Kumar, A., Sharma, J., & Chhokar, V. (2013). Recent advances in industrial applica-
tion of tannases: A review. Recent Patents on Biotechnology, 7, 228–233.
Beresford, T. P., Fitzsimons, N. A., Brennan, N. L., & Cogan, T. M. (2001). Recent advances in
cheese microbiology. International Dairy Journal, 11, 259–274.
Bhowmik, T., & Marth, E. H. (1990). Rote of Micrococcus and Pediococcus species in cheese
ripening: A review. Journal of Dairy Science, 73, 859–866.
Bigelis, R. (1992). Flavor metabolites and enzymes from filamentous fungi. Food technology
(USA).
Bills, D., Morgan, M., Libbey, L., & Day, E. (1965). Identification of compounds responsible for
fruity flavor defect of experimental Cheddar cheeses. Journal of Dairy Science, 48, 1168–1173.
Bornscheuer, U. T. (2002). Microbial carboxyl esterases: Classification, properties and application
in biocatalysis. FEMS Microbiology Reviews, 26, 73–81.
Campbell, R., & Drake, M. (2013). Invited review: The effect of native and nonnative enzymes on
the flavor of dried dairy ingredients. Journal of Dairy Science, 96, 4773–4783.
Carocho, M., Morales, P., & Ferreira, I. C. (2015). Natural food additives: Quo vadis? Trends in
Food Science & Technology, 45, 284–295.
Carunchia Whetstine, M. E., Cadwallader, K. R., & Drake, M. (2005). Characterization of aroma
compounds responsible for the rosy/floral flavor in Cheddar cheese. Journal of Agricultural and
Food Chemistry, 53, 3126–3132.
Caspia, E., Coggins, P., Schilling, M., Yoon, Y., & White, C. (2006). The relationship between
consumer acceptability and descriptive sensory attributes in cheddar cheese. Journal of Sensory
Studies, 21, 112–127.
Chambers, D. H., Esteve, E., & Retiveau, A. (2010). Effect of milk pasteurization on flavor
properties of seven commercially available French cheese types. Journal of Sensory Studies,
25, 494–511.
Chandrasekaran, M. (2012). Future prospects and the need for research, valorization of food
processing by-products (pp. 786–801). CRC Press.
Chang, R.-C., Chou, S.-J., & Shaw, J.-F. (2001). Synthesis of fatty acid esters by recombinant
Staphylococcus epidermidis lipases in aqueous environment. Journal of Agricultural and Food
Chemistry, 49, 2619–2622.
Chanprasartsuk, O.-O., & Prakitchaiwattana, C. (2015). Impacts of allochthonous and autochtho-
nous yeast starters: Case studies in fruit wine fermentations. Food Microbiology: Fundamentals,
Challenges and Health Implications, 117–160.
Charoenprasert, S., & Mitchell, A. (2012). Factors influencing phenolic compounds in table olives
(Olea europaea). Journal of Agricultural and Food Chemistry, 60, 7081–7095.
Chin, H. W., & Lindsay, R. (1993). Volatile sulfur compounds formed in disrupted tissues of
different cabbage cultivars. Journal of Food Science, 58, 835–839.
348 F. Hashempour-Baltork and P. Farshi
Chin, H.-W., & Lindsay, R. C. (1994a). Mechanisms of formation of volatile sulfur compounds
following the action of cysteine sulfoxide lyases. Journal of Agricultural and Food Chemistry,
42, 1529–1536.
Chin, H.-W., & Lindsay, R. C. (1994b). Modulation of volatile sulfur compounds in cruciferous
vegetables. ACS Publications.
Christensen, V. W. (1964). Flavor development and control in Provolone and Romano cheese.
Presented at Marschall Invitational Italian Cheese Seminar.
Cissé, M., Polidori, J., Montet, D., Loiseau, G., & Ducamp-Collin, M. N. (2015). Preservation of
mango quality by using functional chitosan-lactoperoxidase systems coatings. Postharvest
Biology and Technology, 101, 10–14.
Collar, C., Martinez, J., Andreu, P., & Armero, E. (2000). Effects of enzyme associations on bread
dough performance. A response surface analysis/Efectos de las asociaciones enzimáticas sobre
la calidad funcional de masas panarias. Análisis de superficies de respuesta. Food Science and
Technology International, 6, 217–226.
Collins, Y. F., McSweeney, P. L., & Wilkinson, M. G. (2003). Lipolysis and free fatty acid
catabolism in cheese: A review of current knowledge. International Dairy Journal, 13,
841–866.
Consultants, I. (2018). An overview of the global flavors and fragrances market, 11th edition.
Retrieved from www.ialconsultants.com.
Cousins, C., Sharpe, M., & Law, B. (1977). The bacteriological quality of milk for Cheddar
cheesemaking. Dairy Industries International.
Crow, V., Curry, B., Christison, M., & Hellier, K. (2002). Raw milk flora and NSLAB as adjuncts.
Australian Journal of Dairy Technology, 57, 99.
Crow, V., Curry, B., & Hayes, M. (2001). The ecology of non-starter lactic acid bacteria (NSLAB)
and their use as adjuncts in New Zealand Cheddar. International Dairy Journal, 11, 275–283.
Custodio, M. V. G., Otamendi, F. P., Vidal, D. R., & Alventosa, S. V. (1996). Production and
characterization of an Aspergillus terteus α-l-rhamnosidase of oenological interest. Zeitschrift
für Lebensmittel-Untersuchung und Forschung, 203, 522–527.
Custry, F., Fernander, N., & Shahani, K. (1987). Role of lipases and other enzymes in flavour
development. In Proceedings of 24th annual Miles-Marschall Italian cheese seminar.
pp. 16–17.
Czyzewska, K., & Trusek, A. (2018). Encapsulated catalase from Serratia genus for H2O2
decomposition in food applications. Polish Journal of Chemical Technology, 20, 39–43.
Das, S., Holland, R., Crow, V., Bennett, R., & Manderson, G. (2005). Effect of yeast and bacterial
adjuncts on the CLA content and flavour of a washed-curd, dry-salted cheese. International
Dairy Journal, 15, 807–815.
de Andrade Júnior, M. C., & Andrade, J. S. (2015). Changes in pectinases, dietary fibers, and
physicochemical indices related to the flavor of cubiu fruits during ripening. Acta Scientiarum
Agronomy, 37, 171–179.
Desjardins, J. J., Duby, P., Dupart, P., Wood R. D., & Zurcher, U. (1997). Producing of starchy
material with a-amylase and lipoxygenase to produce a toasted or cookielike flavor. Patent
EP772980.
Dimos, A., Urbacha, G., & Miller, A. (1996). Changes in flavour and volatiles of full-fat and
reducedfat cheddar cheeses during maturation. International Dairy Journal, 6, 981–995.
Dominguez, A., Sanroman, A., Fucinos, P., Rua, M., Pastrana, L., & Longo, M. (2004). Quantifi-
cation of intra-and extra-cellular thermophilic lipase/esterase production by Thermus
sp. Biotechnology Letters, 26, 705–708.
Dugelay, I., Gunata, Z., Sapis, J. C., Baumes, R., & Bayonove, C. (1993). Role of cinnamoyl
esterase activities from enzyme preparations on the formation of volatile phenols during
winemaking. Journal of Agricultural and Food Chemistry, 41, 2092–2096.
Dutra Rosolen, M., Gennari, A., Volpato, G., & Volken de Souza, C. F. (2015). Lactose hydrolysis
in milk and dairy whey using microbial β-galactosidases. Enzyme Research, 2015.
12 Enzymes in Flavor Development and Food Preservation 349
Dwivedi, B. K., & Kinsella, J. E. (1974). Carbonyl production from lipolyzed milk fat by the
continuous mycelial culture of Penicillium roqueforti. Journal of Food Science, 39, 83–87.
El Soda, M., Abd El Wahab, H., Ezzat, N., Desmazeaud, M., & Ismail, A. (1986). The esterolytic
and lipolytic activities of the Lactobacilli. II. Detection of the esterase system of Lactobacillus
helveticus, Lactobacillus bulgaricus. Lactobacillus lactis and Lactobacillus acidophilus. Le Lait,
66, 431–443.
Eriksson, C. (2012). Flavor modification. Flavor of Foods and Beverages: Chemistry and Technol-
ogy, 1.
Esparan, V. (2015). Fungal enzymes for the production of vanillin, divanillin & lignans. Gottfried
Wilhelm Leibniz Universität Hannover.
Ewing, T. A., Van Noord, A., Paul, C. E., & Van Berkel, W. J. (2018). A xylenol orange-based
screening assay for the substrate specificity of flavin-dependent para-phenol oxidases.
Molecules, 23, 164.
Ewis, H. E., Abdelal, A. T., & Lu, C.-D. (2004). Molecular cloning and characterization of two
thermostable carboxyl esterases from Geobacillus stearothermophilus. Gene, 329, 187–195.
Faccio, G., Nivala, O., Kruus, K., Buchert, J., & Saloheimo, M. (2011). Sulfhydryl oxidases:
Sources, properties, production and applications. Applied Microbiology and Biotechnology, 91,
957–966.
Falconnier, B., Lapierre, C., Lesage-Meessen, L., Yonnet, G., Brunerie, P., Colonna-Ceccaldi, B.,
Corrieu, G., & Asther, M. (1994). Vanillin as a product of ferulic acid biotransformation by the
white-rot fungus Pycnoporus cinnabarinus I-937: Identification of metabolic pathways. Journal
of Biotechnology, 37, 123–132.
Fenster, K., Parkin, K., & Steele, J. (2000). Characterization of an arylesterase from Lactobacillus
helveticus CNRZ32. Journal of Applied Microbiology, 88, 572–583.
Fenster, K., Parkin, K., & Steele, J. (2003a). Intracellular esterase from Lactobacillus casei LILA:
Nucleotide sequencing, purification, and characterization. Journal of Dairy Science, 86,
1118–1129.
Fenster, K., Parkin, K., & Steele, J. (2003b). Nucleotide sequencing, purification, and biochemical
properties of an arylesterase from Lactobacillus casei LILA. Journal of Dairy Science, 86,
2547–2557.
Fox, P., & Wallace, J. (1997). Formation of flavor compounds in cheese. Advances in Applied
Microbiology, 45, 17–86.
Fuciños, P., Domínguez, A., Angeles Sanromán, M., Longo, M. A., Luisa Rúa, M., & Pastrana,
L. (2005). Production of thermostable lipolytic activity by Thermus species. Biotechnology
Progress, 21, 1198–1205.
Gao, P., Jiang, Q., Xu, Y., & Xia, W. (2017). Esterase activities of autochthonous starter cultures to
increase volatile flavour compounds in Chinese traditional fermented fish (Suan yu). Interna-
tional Journal of Food Properties, 20, S663–S672.
Gao, W., Wu, K., Chen, L., Fan, H., Zhao, Z., Gao, B., Wang, H., & Wei, D. (2016). A novel
esterase from a marine mud metagenomic library for biocatalytic synthesis of short-chain flavor
esters. Microbial Cell Factories, 15, 41.
Ghosh, P., Saxena, R., Gupta, R., Yadav, R., & Davidson, S. (1996). Microbial lipases: Production
and applications. Science Progress (1933), 119–157.
Goulet, C., Mageroy, M. H., Lam, N. B., Floystad, A., Tieman, D. M., & Klee, H. J. (2012). Role of
an esterase in flavor volatile variation within the tomato clade. Proceedings of the National
Academy of Sciences, 109, 19009–19014.
Greenough, R., Perry, C., & Stavnsbjerg, M. (1996). Safety evaluation of a lipase expressed in
Aspergillus oryzae. Food and Chemical Toxicology, 34, 161–166.
Hanft, F., & Koehler, P. (2006). Studies on the effect of glucose oxidase in bread making. Journal
of the Science of Food and Agriculture, 86, 1699–1704.
Hansen, A. P., Welty, R. E., & Shen, R.-S. (1973). Free fatty acid content of cacao beans infested
with storage fungi. Journal of Agricultural and Food Chemistry, 21, 665–670.
350 F. Hashempour-Baltork and P. Farshi
Hernandez-Orte, P., Cersosimo, M., Loscos, N., Cacho, J., Garcia-Moruno, E., & Ferreira,
V. (2008). The development of varietal aroma from non-floral grapes by yeasts of different
genera. Food Chemistry, 107, 1064–1077.
Huang, S., Chang, H., & Goto, M. (1998). Preparation of surfactant-coated lipase for the esterifica-
tion of geraniol and acetic acid in organic solvents. Enzyme and Microbial Technology, 22,
552–557.
Iassonova, D. R., Johnson, L. A., Hammond, E. G., & Beattie, S. E. (2009). Evidence of an
enzymatic source of off flavors in “lipoxygenase-null” soybeans. Journal of the American Oil
Chemists’ Society, 86, 59–64.
Inouye, K., Kusano, M., Hashida, Y., Minoda, M., & Yasukawa, K. (2007). Engineering, expres-
sion, purification, and production of recombinant thermolysin. Biotechnology Annual Review,
13, 43–64.
Iwaniak, A., Minkiewicz, P., Darewicz, M., & Hrynkiewicz, M. (2016). Food protein-originating
peptides as tastants-Physiological, technological, sensory, and bioinformatic approaches. Food
Research International, 89, 27–38.
Jahadi, M., Khosravi-Darani, K., Ehsani, M., Mozafari, M., Saboury, A., Seydahmadian, F., &
Vafabakhsh, Z. (2012). Evaluating the effects of process variables on protease-loaded nano-
liposome production by Plackett-Burman design for utilizing in cheese ripening acceleration.
Asian Journal of Chemistry, 24, 3891.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., Pimentel, T., Gomes, A., & Mozafari, M. R.
(2020). Accelerating ripening of Iranian white brined cheesesusing liposome encapsulated and
free proteinases. Journal of Open Access (Biointerface Research in Applied Chemistry), 10,
4966–4971.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., Mozafari, M. R., Saboury, A. A., & Pourhosseini,
P. S. (2015). The encapsulation of flavourzyme in nanoliposome by heating method. Journal of
Food Science and Technology, 52, 2063–2072.
Jahadi, M., Khosravi-Darani, K., Ehsani, M. R., Mozafari, M. R., Saboury, A. A., Zoghi, A., &
Mohammadi, M. (2016). Modelling of proteolysis in Iranian brined cheese using proteinase-
loaded nanoliposome. International Journal of Dairy Technology, 69, 57–62.
Jayathilaka, L., Gupta, S., Huang, J.-S., Lee, J., & Lee, B.-S. (2014). Preparation of (+)-trans-
isoalliin and its isomers by chemical synthesis and RP-HPLC resolution. Journal of biomolecu-
lar techniques: JBT, 25, 67.
Jensen, F. (1970). Free fatty acids in lipase activated Samsoe cheese. In XVIII International Dairy
Congress, Sydney.
Jiang, J., Björck, L., & Fonden, R. (1998). Production of conjugated linoleic acid by dairy starter
cultures. Journal of Applied Microbiology, 85, 95–102.
Kamenetsky, R. (2007). Garlic: Botany and horticulture. Horticultural Reviews-Westport Then
New York, 33, 123.
Karami, M., Ehsani, M., Mousavi, S., Rezaei, K., & Safari, M. (2009). Microstructural properties of
fat during the accelerated ripening of ultrafiltered-Feta cheese. Food Chemistry, 113, 424–434.
Kiefl, J., Kohlenberg, B., Hartmann, A., Obst, K., Paetz, S., Krammer, G., & Trautzsch, S. (2017).
Investigation on key molecules of Huanglongbing (HLB)-induced orange juice off-flavor.
Journal of Agricultural and Food Chemistry, 66, 2370–2377.
Kimura, K., Ozeki, M., Juneja, L. R., & Ohira, H. (2007). L-Theanine reduces psychological and
physiological stress responses. Biological Psychology, 74, 39–45.
Kirk, O., Borchert, T. V., & Fuglsang, C. C. (2002). Industrial enzyme applications. Current
Opinion in Biotechnology, 13, 345–351.
Koh, M., & Kim, H.-J. (2001). The effects of metallothionein on the activity of enzymes involved in
removal of reactive oxygen species. Bulletin-Korean Chemical Society, 22, 362–366.
Kola, O., Kaya, C., Duran, H., & Altan, A. (2010). Removal of limonin bitterness by treatment of
ion exchange and adsorbent resins. Food Science and Biotechnology, 19, 411–416.
12 Enzymes in Flavor Development and Food Preservation 351
Kongor, J. E., Hinneh, M., Van de Walle, D., Afoakwa, E. O., Boeckx, P., & Dewettinck,
K. (2016). Factors influencing quality variation in cocoa (Theobroma cacao) bean flavour
profile—A review. Food Research International, 82, 44–52.
Kosikowski, F. V. (1975). Potential of enzymes in continous cheesemaking. Journal of Dairy
Science, 58, 994–1000.
Kragl, U., Kruse, W., Hummel, W., & Wandrey, C. (1996). Enzyme engineering aspects of
biocatalysis: Cofactor regeneration as example. Biotechnology and Bioengineering, 52,
309–319.
Krishna, S. H., Divakar, S., Prapulla, S., & Karanth, N. (2001). Enzymatic synthesis of isoamyl
acetate using immobilized lipase from Rhizomucor miehei. Journal of Biotechnology, 87,
193–201.
Krogerus, K., & Gibson, B. R. (2013). 125th Anniversary review: Diacetyl and its control during
brewery fermentation. Journal of the Institute of Brewing, 119, 86–97.
Kwon, D. Y., Hong, Y.-J., & Yoon, S. H. (2000). Enantiomeric synthesis of (S)-2-methylbutanoic
acid methyl ester, apple flavor, using lipases in organic solvent. Journal of Agricultural and
Food Chemistry, 48, 524–530.
Laane, C., & Bruggeman, Y. (2002). Applications of oxidoreductases in foods. Handbook of Food
Enzymology, 122, 161.
Laane, C., Rietjens, I., Haaker, H., & Van Berkel, W. (1997). Generation of Taste through (Redox)
Biocatalysis. Special Publication-Royal Society of Chemistry, 214, 137–152.
Laboret, F., & Perraud, R. (1999). Lipase-catalyzed production of short-chain acids terpenyl esters
of interest to the food industry. Applied Biochemistry and Biotechnology, 82, 185–198.
Lane, C., & Hammer, B. (1936). The manufacture of Blue cheese (Roquefort type) from
homogenized cows’ milk. Iowa State College Journal of Science, 10, 391.
Langrand, G., Rondot, N., Triantaphylides, C., & Baratti, J. (1990). Short chain flavour esters
synthesis by microbial lipases. Biotechnology Letters, 12, 581–586.
Larios, A., García, H. S., Oliart, R. M., & Valerio-Alfaro, G. (2004). Synthesis of flavor and
fragrance esters using Candida antarctica lipase. Applied Microbiology and Biotechnology, 65,
373–376.
Lim, T. (2012). Vanilla planifolia, edible medicinal and non-medicinal plants (pp. 106–114).
Springer.
Lin, S., Zhang, G., Liao, Y., Pan, J., & Gong, D. (2015). Dietary flavonoids as xanthine oxidase
inhibitors: Structure–affinity and structure–activity relationships. Journal of Agricultural and
Food Chemistry, 63, 7784–7794.
Liu, S.-Q., Holland, R., & Crow, V. (2003a). Ester synthesis in an aqueous environment by
Streptococcus thermophilus and other dairy lactic acid bacteria. Applied Microbiology and
Biotechnology, 63, 81–88.
Liu, S.-Q., Holland, R., & Crow, V. (2003b). Synthesis of ethyl butanoate by a commercial lipase in
aqueous media under conditions relevant to cheese ripening. The Journal of Dairy Research,
70, 359.
Lopez-Gallego, F., Betancor, L., Hidalgo, A., Dellamora-Ortiz, G., Mateo, C., Fernández-Lafuente,
R., & Guisán, J. M. (2007). Stabilization of different alcohol oxidases via immobilization and
post immobilization techniques. Enzyme and Microbial Technology, 40, 278–284.
Maehashi, K., & Huang, L. (2009). Bitter peptides and bitter taste receptors. Cellular and Molecu-
lar Life Sciences, 66, 1661–1671.
Maia, M. M. D., de Morais, M. M. C., de Morais, M. A., Jr., Melo, E. H. M., & de Lima Filho, J. L.
(1999). Production of extracellular lipase by the phytopathogenic fungus Fusarium solani FS1.
Revista de Microbiologia, 30, 304–309.
Mandal, S. (2012). Functional characterization of lipoxygenase isozymes in relation to off flavor
development in soybean (glycine max). IARI, Division of Biochemistry.
Markus, P. A., Peters, A. L. J., & Roos, R. (1991). Enzymatic process for preparation of
phenylacetaldehyde compounds especially vanillin by conversion of starting compounds in
the presence of lipoxidase. Patent EP542348.
352 F. Hashempour-Baltork and P. Farshi
Martin, N. H., Friedlander, A., Mok, A., Kent, D., Wiedmann, M., & Boor, K. J. (2014). Peroxide
test strips detect added hydrogen peroxide in raw milk at levels affecting bacterial load. Journal
of Food Protection, 77, 1809–1813.
Martinez Ortega, A. (2015). Most significant biotechnological improvements on the production
process of wine, beer and bread.
Mase, T., Sonoda, M., Morita, M., & Hirose, E. (2010). Characterization of a lipase from
Sporidiobolus pararoseus 25-A which produces cheese flavor. Food Science and Technology
Research, 17, 17–20.
Masud, T., Khalid, S., Maqsood, S., & Bilal, A. (2010). Preservation of raw buffalo’s milk by the
activation of lactoperoxidase system and its effect on yogurt preparation. Journal of Food
Processing and Preservation, 34, 241–254.
Masuda, R., Kaneko, K., & Saito, M. (2012). Accumulation of alkyl-cysteine derivatives in
Crucifers. Biology and Biotechnology of the Plant Hormone Ethylene, II, 439.
Masuo, N., Yoshimune, K., Ito, K., Matsushima, K., Koyama, Y., & Moriguchi, M. (2005).
Micrococcus luteus K-3-type glutaminase from Aspergillus oryzae RIB40 is salt-tolerant.
Journal of Bioscience and Bioengineering, 100, 576–578.
Mathew, S., & Abraham, T. E. (2005). Studies on the production of feruloyl esterase from cereal
brans and sugar cane bagasse by microbial fermentation. Enzyme and Microbial Technology,
36, 565–570.
McGugan, W., Blais, J., Boulet, M., Giroux, R., Elliott, J., & Emmons, D. (1975). Ethanol, ethyl
esters, and volatile fatty acids in fruity Cheddar cheese. Canadian Institute of Food Science and
Technology Journal, 8, 196–198.
McSweeney, P. (2016). Enzymes exogenous to milk in dairy technology: Catalase, glucose oxidase,
glucose isomerase and hexose oxidase.
Menzel, M., & Schreier, P. (2007). Enzymes and flavour biotechnology (pp. 489–505). Flavours
and Fragrances, Springer.
Mohammadi, R., Mahmoudzadeh, M., Atefi, M., Khosravi-Darani, K., & Mozafari, M. (2015).
Applications of nanoliposomes in cheese technology. International Journal of Dairy Technol-
ogy, 68, 11–23.
Molimard, P., & Spinnler, H.-E. (1996). Compounds involved in the flavor of surface mold-ripened
cheeses: Origins and properties. Journal of Dairy Science, 79, 169–184.
Moschopoulou, E. (2011). Characteristics of rennet and other enzymes from small ruminants used
in cheese production. Small Ruminant Research, 101, 188–195.
Moskowitz, G. J., Cassaigne, R., West, I. R., Shen, T., & Feldman, L. I. (1977). Hydrolysis of
animal fat and vegetable oil with Mucor miehei esterase. Properties of the enzyme. Journal of
Agricultural and Food Chemistry, 25, 1146–1150.
Nagodawithana, T., & Reed, G. (1993). Preface to the third edition, enzymes in food processing
(p. xix). Elsevier.
Nandakumar, R., Yoshimune, K., Wakayama, M., & Moriguchi, M. (2003). Microbial glutaminase:
Biochemistry, molecular approaches and applications in the food industry. Journal of Molecular
Catalysis B: Enzymatic, 23, 87–100.
Narbad, A., & Gasson, M. J. (1998). Metabolism of ferulic acid via vanillin using a novel
CoA-dependent pathway in a newly-isolated strain of Pseudomonas fluorescens. Microbiology,
144, 1397–1405.
Nathan, P. J., Lu, K., Gray, M., & Oliver, C. (2006). The neuropharmacology of L-theanine
(N-ethyl-L-glutamine) a possible neuroprotective and cognitive enhancing agent. Journal of
Herbal Pharmacotherapy, 6, 21–30.
Nunes, C. S., & Kunamneni, A. (2018). Laccases—Properties and applications, Enzymes in human
and animal nutrition (pp. 133–161). Elsevier.
Obenland, D., & Aung, L. (1996). Cystine lyase activity and anaerobically-induced sulfur gas
emission from broccoli florets. Phyton.
12 Enzymes in Flavor Development and Food Preservation 353
Ogino, H., Tsuchiyama, S., Yasuda, M., & Doukyu, N. (2010). Enhancement of the aspartame
precursor synthetic activity of an organic solvent-stable protease. Protein Engineering, Design
& Selection, 23, 147–152.
Okazawa, S., Hashimoto, M., & Hatsutori, Y. (1993). Treatment of adzuki bean with enzymes.
Patent JP0524488.
Ough, C. (1975). Further investigations with glucose oxidase-catalase enzyme systems for use with
wine. American Journal of Enology and Viticulture, 26, 30–36.
Owen, G. N., Parnell, H., De Bruin, E. A., & Rycroft, J. A. (2008). The combined effects of
L-theanine and caffeine on cognitive performance and mood. Nutritional Neuroscience, 11,
193–198.
Parker, K., Salas, M., & Nwosu, V. C. (2010). High fructose corn syrup: Production, uses and
public health concerns. Biotechnology and Molecular Biology Reviews, 5, 71–78.
Petropoulos, S., Ntatsi, G., & Ferreira, I. (2017). Long-term storage of onion and the factors that
affect its quality: A critical review. Food Reviews International, 33, 62–83.
Pico, J., Bernal, J., & Gómez, M. (2015). Wheat bread aroma compounds in crumb and crust: A
review. Food Research International, 75, 200–215.
Pogorzelski, E., & Wilkowska, A. (2007). Flavour enhancement through the enzymatic hydrolysis
of glycosidic aroma precursors in juices and wine beverages: A review. Flavour and Fragrance
Journal, 22, 251–254.
Price, E. J., Linforth, R. S., Dodd, C. E., Phillips, C. A., Hewson, L., Hort, J., & Gkatzionis,
K. (2014). Study of the influence of yeast inoculum concentration (Yarrowia lipolytica and
Kluyveromyces lactis) on blue cheese aroma development using microbiological models. Food
Chemistry, 145, 464–472.
Priefert, H., Rabenhorst, J., & Steinbüchel, A. (2001). Biotechnological production of vanillin.
Applied Microbiology and Biotechnology, 56, 296–314.
Puri, M. (2012). Updates on naringinase: Structural and biotechnological aspects. Applied Micro-
biology and Biotechnology, 93, 49–60.
Puri, M., Banerjee, A., & Banerjee, U. (2005). Optimization of process parameters for the
production of naringinase by Aspergillus niger MTCC 1344. Process Biochemistry, 40,
195–201.
Raghuvanshi, R. S., & Bisht, K. (2010). 18 uses of soybean: Products and preparation. The
Soybean, 404.
Ramírez, E. C., & Whitaker, J. R. (1999). Biochemical characterization of cystine lyase from
broccoli (Brassica oleracea Var. Italica). Journal of Agricultural and Food Chemistry, 47,
2218–2225.
Raveendran, S., Parameswaran, B., Beevi, S., Amith, U., Anil, A., Mathew, K., Madhavan, A.,
Rebello, S., & Pandey, A. (2018). Applications of microbial enzymes in food industry. Food
Technology and Biotechnology, 56(1), 16–30.
Ricard, P. B. P. (1991). Synthesis of cis-3-hexenol from unsaturated aliphatic acid—Using com-
bined action of natural enzyme system and yeast. Patent EP481147.
Richardson, G., & Nelson, J. (1968). Rapid evaluation of milk coagulating and flavor producing
enzymes for cheese manufacture. Journal of Dairy Science, 51, 1502–1503.
Röcker, J., Schmitt, M., Pasch, L., Ebert, K., & Grossmann, M. (2016). The use of glucose oxidase
and catalase for the enzymatic reduction of the potential ethanol content in wine. Food
Chemistry, 210, 660–670.
Romero, M., Calvo, L., Alba, C., Daneshfar, A., & Ghaziaskar, H. (2005). Enzymatic synthesis of
isoamyl acetate with immobilized Candida antarctica lipase in n-hexane. Enzyme and Microbial
Technology, 37, 42–48.
Ruiz-Matute, A. I., Weiss, M., Sammataro, D., Finely, J., & Sanz, M. L. (2010). Carbohydrate
composition of high-fructose corn syrups (HFCS) used for bee feeding: Effect on honey
composition. Journal of Agricultural and Food Chemistry, 58, 7317–7322.
Saerens, K., Descamps, D., & Dewettinck, K. (2008). Release of short chain fatty acids from cream
lipids by commercial lipases and esterases. Biotechnology Letters, 30, 311–315.
354 F. Hashempour-Baltork and P. Farshi
Sakato, Y. (1949). The chemical constituents of tea; III. A new amide theanine. Nippon
Nogeikagaku Kaishi, 23, 262–267.
Saltmarsh, M., & Saltmarsh, M. (2013). Essential guide to food additives. Royal Society of
Chemistry.
Salvadori, P. (1961). The use of lipolytic enzyme of microbiological origin in the manufacture of
Pecorino cheese. Latte, 35, 177.
Sánchez, M., Prim, N., Rández-Gil, F., Pastor, F. J., & Diaz, P. (2002). Engineering of baker’s
yeasts, E. coli and Bacillus hosts for the production of Bacillus subtilis lipase A. Biotechnology
and Bioengineering, 78, 339–345.
Schroeder, M., Pöllinger-Zierler, B., Aichernig, N., Siegmund, B., & Guebitz, G. (2008). Enzy-
matic removal of off-flavors from apple juice. Journal of Agricultural and Food Chemistry, 56,
2485–2489.
Schwab, W., Davidovich-Rikanati, R., & Lewinsohn, E. (2008). Biosynthesis of plant-derived
flavor compounds. The Plant Journal, 54, 712–732.
Scrimgeour, C., Gao, Y., Oh, W. Y., & Shahidi, F. (2005). Chemistry of fatty acids. Bailey’s
Industrial Oil and Fat Products, 1–40.
Seitz, E. W. (1974). Industrial application of microbial lipases: A review. Journal of the American
oil chemists’ society, 51, 12–16.
Seratlić, S. V., Miloradović, Z. N., Radulović, Z. T., & Maćej, O. D. (2011). The effect of two types
of mould inoculants on the microbiological composition, physicochemical properties and
protein hydrolysis in two Gorgonzola-type cheese varieties during ripening. International
Journal of Dairy Technology, 64, 408–416.
Sharma, R., Chisti, Y., & Banerjee, U. C. (2001). Production, purification, characterization, and
applications of lipases. Biotechnology Advances, 19, 627–662.
Shieh, C.-J., & Chang, S.-W. (2001). Optimized synthesis of lipase-catalyzed hexyl acetate in
n-hexane by response surface methodology. Journal of Agricultural and Food Chemistry, 49,
1203–1207.
Shimada, Y., Hirota, Y., Baba, T., Sugihara, A., Moriyama, S., Tominaga, Y., & Terai, T. (1999).
Enzymatic synthesis of steryl esters of polyunsaturated fatty acids. Journal of the American Oil
Chemists’ Society, 76, 713–716.
Silva, F., Gibbs, P., Nunez, H., Almonacid, S., Simpson, R., Batt, C., & Tortorello, M. (2014).
Encyclopedia of food microbiology, Thermal processes: Pasteurization (pp. 577–595). Elsevier
Netherlands.
Singh, A., & Banerjee, R. (2017). Challenges in lipase mediated synthesis of food flavor ester
compounds, developing technologies in food science (pp. 63–74). Apple Academic Press.
Singh, R., Mittal, A., Kumar, M., & Mehta, P. K. (2016). Microbial proteases in commercial
applications. Journal of Pharmaceutical, Chemical and Biological Sciences, 4, 365–374.
Small, L. E., & Asquith, T. (1989). Process for treating coffee beans with enzyme-containing
solution under pressure to reduce bitterness. Patent EP337541.
Smit, G., Smit, B. A., & Engels, W. J. (2005). Flavour formation by lactic acid bacteria and
biochemical flavour profiling of cheese products. FEMS Microbiology Reviews, 29, 591–610.
Sung, K., Khan, S. A., Nawaz, M. S., Cerniglia, C. E., Tamplin, M. L., Phillips, R. W., & Kelley,
L. C. (2011). Lysozyme as a barrier to growth of Bacillus anthracis strain Sterne in liquid egg
white, milk and beef. Food Microbiology, 28, 1231–1234.
Suzuki, H., Izuka, S., Miyakawa, N., & Kumagai, H. (2002). Enzymatic production of theanine, an
“umami” component of tea, from glutamine and ethylamine with bacterial
γ-glutamyltranspeptidase. Enzyme and Microbial Technology, 31, 884–889.
Suzzi, G., Lanorte, M., Galgano, F., Andrighetto, C., Lombardi, A., Lanciotti, R., & Guerzoni,
M. (2001). Proteolytic, lipolytic and molecular characterisation of Yarrowia lipolytica isolated
from cheese. International Journal of Food Microbiology, 69, 69–77.
Takemori, T., Ito, Y., Ito, M., & Yoshama, M. (1992). Flavor and taste improvement of cacao nib
by enzymatic treatment. Patent JP04126037.
12 Enzymes in Flavor Development and Food Preservation 355
Thorpe, C., Hoober, K. L., Raje, S., Glynn, N. M., Burnside, J., Turi, G. K., & Coppock, D. L.
(2002). Sulfhydryl oxidases: Emerging catalysts of protein disulfide bond formation in
eukaryotes. Archives of Biochemistry and Biophysics, 405, 1–12.
Tishel, M., & Mazelis, M. (1966). Enzymatic degradation of L-cystine by cytoplasmic particles
from cabbage leaves. Nature, 211, 745–746.
Tunick, M. H. (2007). Origins of cheese flavor. ACS Publications.
Urbach, G. (1993). Relations between cheese flavour and chemical composition. International
Dairy Journal, 3, 389–422.
Vafabakhsh, Z., Khosravi-Darani, K., Khajeh, K., Jahadi, M., Komeili, R., & Mortazavian, A. M.
(2013). Stability and catalytic kinetics of protease loaded liposomes. Biochemical Engineering
Journal, 72, 11–17.
Van Haandel, M. J., Saraber, F. C., Boersma, M. G., Laane, C., Fleming, Y., Weenen, H., &
Rietjens, I. M. (2000). Characterization of different commercial soybean peroxidase
preparations and use of the enzyme for N-demethylation of methyl N-methylanthranilate to
produce the food flavor methylanthranilate. Journal of Agricultural and Food Chemistry, 48,
1949–1954.
Vandamme, E. J., & Soetaert, W. (2002). Bioflavours and fragrances via fermentation and bioca-
talysis. Journal of Chemical Technology & Biotechnology: International Research in Process,
Environmental & Clean Technology, 77, 1323–1332.
Vulevic, J., Juric, A., Tzortzis, G., & Gibson, G. R. (2013). A mixture of trans-
galactooligosaccharides reduces markers of metabolic syndrome and modulates the fecal
microbiota and immune function of overweight adults. The Journal of Nutrition, 143, 324–331.
Waldmann, E., & Parhofer, K. G. (2019). Apheresis for severe hypercholesterolaemia and elevated
lipoprotein (a). Pathology, 51, 227–232.
Walker, V., & Mills, G. A. (2014). 2-Pentanone production from hexanoic acid by Penicillium
roqueforti from blue cheese: Is this the pathway used in humans? The Scientific World Journal
2014.
Welsh, F. W., Murray, W. D., Williams, R. E., & Katz, I. (1989). Microbiological and enzymatic
production of flavor and fragrance chemicals. Critical Reviews in Biotechnology, 9, 105–169.
Whitaker, J. R., Voragen, A. G., & Wong, D. W. (2002). Handbook of food enzymology, 122. CRC
Press.
Wilding, P., & Woolner, E. (1997). Reatment of tomato with eroxidase to achieve a fresh flavour
profile. Patent WO9738591.
Williams, P. J., Sefton, M. A., & Francis, I. L. (1992). Glycosidic precursors of varietal grape and
wine flavor. ACS Publications.
Wolf, I. V., Meinardi, C. A., & Zalazar, C. A. (2009). Production of flavour compounds from fat
during cheese ripening by action of lipases and esterases. Protein and Peptide Letters, 16,
1235–1243.
Wouters, A. G., Rombouts, I., Fierens, E., Brijs, K., & Delcour, J. A. (2016). Relevance of the
functional properties of enzymatic plant protein hydrolysates in food systems. Comprehensive
Reviews in Food Science and Food Safety, 15, 786–800.
Yang, H., Cai, G., Lu, J., & Gómez Plaza, E. (2020). The production and application of enzymes
related to the quality of fruit wine. Critical Reviews in Food Science and Nutrition, 1–11.
Yao, G., Chen, G., & Fu, J. (1987). Technique for brewing beer containing SOD, hedgehog
polysaccharide and theaflavin. Patent CH1145404.
Zarepour, M., Kaspari, K., Stagge, S., Rethmeier, R., Mendel, R. R., & Bittner, F. (2010). Xanthine
dehydrogenase AtXDH1 from Arabidopsis thaliana is a potent producer of superoxide anions
via its NADH oxidase activity. Plant Molecular Biology, 72, 301–310.
356 F. Hashempour-Baltork and P. Farshi
Zeng, L.-L., & Huang, H.-H. (2011). Optimization of debitterizing of kumquat juice by naringinase
with response surface method [J]. Science and Technology of Food Industry 5.
Zhang, C., Hua, Y., Li, X., Kong, X., & Chen, Y. (2020). Key volatile off-flavor compounds in peas
(Pisum sativum L.) and their relations with the endogenous precursors and enzymes using
soybean (Glycine max) as a reference. Food Chemistry, 127469.
Zhang, Y., Venkitasamy, C., Pan, Z., Liu, W., & Zhao, L. (2017). Novel umami ingredients:
Umami peptides and their taste. Journal of Food Science, 82, 16–23.
Zhuang, M., Lin, L., Zhao, M., Dong, Y., Sun-Waterhouse, D., Chen, H., Qiu, C., & Su, G. (2016).
Sequence, taste and umami-enhancing effect of the peptides separated from soy sauce. Food
Chemistry, 206, 174–181.
Enzymes from Genetically Modified
Organisms and Their Current Applications 13
in Food Development and Food Chain
Abstract
Keywords
# The Author(s), under exclusive license to Springer Nature Singapore Pte 357
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_13
358 S. Muthusamy et al.
13.1 Introduction
Enzymes are proteins (also referred to as biocatalysts) that can be extracted from
cells, which catalyze the biochemical reactions in living organisms. They can be
used to catalyze a good range of commercially important processes in paper and
pulp, textile, chemical and pharmaceutical, agriculture, and food industries. Among
food, enzymes represent a major share in the enzyme industry. For many years,
enzymes are utilized in food processes, like for the clarification and filtering of wines
and beers, for baking, the assembly of cheeses, and far more (Robinson, 2015).
Global population is accelerating and is estimated to reach ten billion by 2057. The
use of industrial enzymes is also estimated to increase in correlation with the
population from 5.8 billion USD in 2020 to 20 billion USD in 2057 (Fig. 13.1).
The preferred source of enzymes are plant and animal, which constitutes around 15%
and the remaining 85% of industrial enzymes are from microorganisms such as fungi
and bacteria. The economic and technical ease put microorganisms forefront of
enzyme synthesis.
The development of recombinant technology brought considerable improvements
in the enzyme industries by generating Genetically Modified Organisms (GMOs). In
today’s market, around 50% of the enzymes are derived from these GMOs, which
are intended to increase their yield, purity, specificity, and stability (Petersen, 2005;
Poulsen & Bucholz, 2003). As per FAO (Food and Agriculture Organization of the
United Nations). GMOs are “the organisms those not occur by natural reproduction
or natural recombination.” GMOs are the major source of enzyme production in food
industries that requires a comprehensive understanding of the integration between
enzymology, molecular biology, and bioinformatics. The enzymes from GMOs
especially from the microorganisms have several safety concerns such as contami-
nation with toxins, allergens, and other impurities (de Santis et al., 2018; Srivastava,
2019). Thus, before getting into the marketing, enzymes from GMOs need approval
from regulatory bodies such as the Food and Drug Administration (FDA; USA) and
European Food Safety Authority (EFSA; Europe) with rigorous assessment in
addition to regular ethical concerns. This chapter deals with the enzymes from
GMOs; applications in food industries and regulatory concerns for their use.
The biological variants form very often due to natural recombination and mutation
by changes in the surrounding microenvironment. More than a century, we also
exploiting the genetic materials that are naturally present in animals, plants, and
microbes. Initially, the geographic distribution of genetic variants occurred unknow-
ingly from one part of the world to another with distinct climate conditions. For
example, Asian soybean to America and American potato throughout the world. The
history of genetic engineering begins with Charles Darwin’s notions of species
variation and selection. The evolution of biotechnology leads to the development
of biological techniques that help to overcome unbeatable physiological barriers and
to exchange genetic materials among all living organisms. In late 1950s and 1960s,
the scientists discovered the double helix structure, central dogma, and genetic code
which led to the development of DNA recombination technology in 1973 by Cohen
et al. The development in the DNA recombination technology showed that the
genetically engineered DNA molecules can be transferred among same species
and in different species.
According to World Health Organization (WHO), GMOs are the organisms (i.e.,
plants, animals, or microorganisms) in which the genetic material (Deoxyribonucleic
acid) has been altered in a way that does not occur naturally by mating and/or natural
recombination. The genetic modification occurs in two ways; either mutation or
recombination, which may be spontaneous/natural or induced. The evolution of
biotechnology leads to the development of biological techniques that help to over-
come unbeatable physiological barriers and to exchange genetic materials among all
living organisms. The technology is often called modern biotechnology, recombi-
nant DNA technology, or genetic engineering.
Initially, the first-generation GMOs were developed for improving the quality and
yield by increasing their resistance to certain diseases and unfavorable environments.
In 1983, the first genetically modified plants antibiotic-resistant tobacco and petunias
were produced by three independent research teams (Bevan & Chilton, 1982; Fraley,
1983; Herrera-Estrella et al., 1983). In 1990, China first commercialized genetically
modified tobacco and in 1994 USA marketed the genetically modified tomato which
is approved by the Food and Drug Administration (FDA). Many transgenic crops,
such as Canola with modified oil composition, cotton and soybeans resistant to
herbicides, etc. have received FDA approvals. Genetically modified potatoes,
eggplants, strawberries, carrots, and many more are available in the market (Bawa
& Anilakumar, 2013). The second-generation GMOs were aimed to transfer genetic
information from one organism to the other to improve specific product quality and
quantity.
360 S. Muthusamy et al.
13.2.1.1 GM Plants
The Food and Agriculture Organization (FAO)/International Atomic Energy Agency
(IAEA) listed more than 3000 plants that have been developed using genetic
modification with physical and chemical mutagenic agents. These GM foods are
either used for the human consumption or used as feed for livestock and poultry.
The first commercialized food crop is papaya, which was genetically modified for
its resistance against papaya ringspot virus. Later, some fruit ripening characteristics
have been improved using genetic modification. Polyphenol oxidase is responsible
for the production of melanin in apples and potatoes, which cause a brown tint when
exposed to air. The restriction of polyphenol oxidase synthesis below 10% by
genetic modification allowed the apple and potato remain same after slicing. The
herbicide-tolerant varieties of alfalfa, soybean, sugar beet, sweet corn (maize), and
cotton have been developed using genetic engineering. GM Golden rice, which
contains beta-carotene (precursor of vitamin A) along with enhanced iron content
has been approved in various countries. At present, more than 170 million hectares
are under cultivation of GM crops worldwide (James, 2006).
13.2.1.2 GM Animals
Though GM animals are in the use of biomedical and pharmaceutical applications to
produce certain recombinant proteins and to study selective gene functions, the
generation of GM animals for food from a variety of target species traits is still on
bench side due to the inefficacy of the production and ethical issues. The major
causes to attain genetically modified organisms are; to increase animal health, (Tong
et al., 2011; Wheeler et al., 2001), to make them disease resistant (Lyall et al., 2011;
Richt et al., 2007), to improve growth rate (Devlin et al., 2009), to increase meat
quantity (Lai et al., 2006), to increase milk production and alter composition
(Wu et al., 2012), and to increase wool production (Damak et al., 1996). The fast-
growing Atlantic salmon is the only GM animal that has been approved for food by
health Canada and USA-FDA, in which the growth hormone gene from Chinook
salmon was transferred into the Atlantic salmon genome. The use of this fast-
growing GM fish for the food in future is still uncertain.
13.2.1.3 GM Microorganisms
Microorganisms, produce endogenous enzymes responsible for several food
products like yoghurt, cheese, beer, wine, vinegar, and many more. Humans used
these microorganisms even without knowing they are responsible for the fermenta-
tion process in various food products (Jay et al., 2005; Zhang et al., 2017). During
the eighteenth century, the discovery of the microorganisms responsible for a certain
process in food industries began to research efficient ways to use microorganisms
(Barnett, 2003). The discovery of DNA followed by recombinant technology
allowed genetic alteration to optimize the use of microorganisms in effective way
for food production. These genetic modifications either improve the production of
existing proteins or increase their potential or produce a new protein with different
characteristic features. The advancement in molecular biology led to widespread use
of GM microorganisms in food industries and is recognized as an environmentally
13 Enzymes from Genetically Modified Organisms and Their Current. . . 361
friendly and cost-effective method for food production. The yeast Saccharomyces
cerevisiae was the first GM microorganism used for enzymes synthesis in the dairy
and brewing industries. The trypsin and chymosin from the GM microorganisms
served as an alternate for animal sources such as pigs and cattle. Replacement of
certain plant-based products by GM microorganisms could meet the demand with
reduced land usage and waste generation. The GM yeast-based production of
vanillin, an alternate for vanilla extract; GM bacteria that produce riboflavin are
certain examples of the use of GM microbes in food industries (Brochado et al.,
2010; Schwechheimer et al., 2016). Other food ingredients known to be produced by
microbes are includes vitamins, amino acids, nutritional proteins, oligosaccharides,
flavoring agents, and sweeteners (Adrio & Demain, 2010).
According to legal authorities such as FDA, the European Food Safety Authority
(EFSA), the enzymes are considered nutritional additives. The major industrial
enzyme market relies on the food and beverage industry, which is expected to
reach 2.3 billion USD in 2021 (Raveendran et al., 2018). Although the majority of
the organisms including plants and animals produce enzymes, microbial sources
become more valuable. Table 13.1 shows various industrial enzymes used in the
food and animal feed industries.
The wild-type microbes are unstable and may not produce desired quality enzyme,
especially in fermenter conditions. Sometimes the feedback repression occurs that
362 S. Muthusamy et al.
Table 13.1 Industrial enzymes used in food and animal feed industries
Industry Enzyme Function
Dairy Lactase (beta Lactose reduction in milk, coagulation, faster cheese ripening,
galactosidase) debittering, flavored cheese, protein crosslinking
Aminopeptidase
catalase
Proteinase (acid
and neutral)
Lipase
Transglutaminase
Baking Amylase Bread softening, enhance lifespan, dough conditioning,
Transglutaminase dough stability, and strengthening
Lipase
Glucose oxidase
Xylanase
Beverage Pectinase Pectin digestion, starch hydrolysis fruit liquefaction, protein
Cellulase breakdown, oxygen removal, debittering, restrict mist
Amylase (Alpha formation
and Beta)
Beta Glucanase
Protease
Glucose oxidase
Pullulanase
Naringinase
Limoninase
Aminopeptidases
Animal Beta glucanase Enhance digestion
feed Phytase
Xylanase
Fig. 13.2 Major steps involved in the enzyme production from genetically modified microbes
1. Codon optimization for the heterologous expression and to limit mRNA folding
for optimal translation (Rosano & Ceccarelli, 2014).
2. In-depth understanding of enzyme sequence, corresponding amino acids, struc-
ture, and related activity. This will allow one to modify the sequence by mutation-
like site-directed mutagenesis to alter or increase the enzyme activity, specificity,
and structural stability (Hua et al., 2018).
3. Developing an efficient screening protocol for identifying effective variants
(Packer & Liu, 2015).
4. Knowledge about fusion and truncation of target sequence to produce an enzyme
with multiple functions and to eliminate the unwanted sequences those are not
important for the function (Dediu, 2015).
Fig. 13.3 Basic structure of expression vector used in recombinant enzyme synthesis
vector is shown in Fig. 13.3. An expression vector contains, at the least, an origin of
replication (ORI), a Multiple Cloning Site (MCS), in which the gene of interest will
be integrated, and a selection marker for the selection of the recombinant strain
(Fig. 13.1). Promoter and terminator regions are also present to control the expres-
sion of the gene of interest and the selection marker. In order to avoid an increase in
the metabolic load on the host organism and to avoid reduced plasmid stability, the
size of the vector is kept as small as possible (Rieder et al., 2019).
It is important to select the recombinant strain and the selection is based on the
selectable marker. A selectable marker is usually a gene that shows resistance to an
antibiotic that would otherwise kill the normal cells which lack these genes under
culture conditions with a specific antibiotic. Kanamycin (kanR), ampicillin (ampR),
and tetracycline (tetR) resistance genes are commonly used in the bacterial expres-
sion vector. Antibiotic resistance markers are associated with a public health con-
cern, which is replaced by auxotrophic strains, where it requires specific nutrients for
growth. Some auxotrophic markers are amdS, URA3, which are usually used as the
selectable marker in fungal strains (Hjort, 2007; Olempska-Beer et al., 2006).
Preparation of GM Microbes
The common method employed in the preparation of GM microbes is to cut open the
plasmid DNA; inserting the target DNA molecule (named molecular cloning) and
transform it into host microbes, which can efficiently express the target DNA.
Dissecting each part, (1) the plasmid expression vector has been chosen based on
the host organism; (2) the target DNA has been amplified from the target genome
which has restriction enzyme site overhanging on both the ends that matching to the
sites present in the multiple cloning site of the plasmid; (3) restriction digestion of
the plasmid vector and amplified target DNA molecule using restriction enzymes
(which can form either blunt or sticky ends); (4) Purify the restriction digested DNA
and plasmid; (5) Ligate the plasmid and target DNA using ligase; and (6) transform
the recombined plasmid to the host organism. The recombinant DNA molecule can
13 Enzymes from Genetically Modified Organisms and Their Current. . . 365
1. Submerged Fermentation
Submerged fermentation (SF) is the most preferred method for industrial-level
enzyme production, where the selected GM microbes (bacteria or Fungi) are
cultured in the liquid nutrient medium at a higher concentration of oxygen. In this
process, the microbes release the enzymes to break down the complex
carbohydrates, proteins, and lipids in the medium. A large fermenter with a
high capacity of around 1000 cubic meters is used to produce a large quantity
of enzymes. Simple batch reactors are used, where the reactor is filled with
selected microbe and nutrients and allowed for fermentation. After fermentation,
the contents are emptied for further enzyme purification. On the other way, the
batch-fed process is a continuous process, where the sterile nutrient mix is added
to the fermenter at the same level of fermented broth leaving for further enzyme
purification.
2. Solid-State Fermentation
Solid-state fermentation (SSF) is another method that involves the culture of the
GM microbe on a solid substrate, such as pulp, corn, wheat or rice bran, and
grains. The advantages of SSF over SF are; a high concentration of enzyme
production with less affluent in a simple fermenter. In SSF, the factors such as
particle size and moisture play a major role in enzyme production. For example,
smaller substrate particles make larger surface area microbial growth, whereas
larger particles provide efficient aeration. In SSF the water level should be
optimized for enzyme production, as moisture level greatly influences the micro-
bial activity.
From the ancient period onwards a wide variety of enzymes from a natural source
has been used for the production of numerous food products. Natural sources for
obtaining enzymes are animals, plants, and microorganism. There are about 3000
known enzymes produced by all animals, green plants, fungi, and bacteria that
catalyze about 4000 biochemical reactions (Patel et al., 2017). Among these sources,
microorganisms have been involved in the food industry for various applications like
the production of alcoholic beverages, ethyl alcohol, organic products, dairy
products, and drugs including antibiotics through fermentation (Verma et al.,
2018). For example, the usage of chymosin for cheese production and pectinases
for fruit and beverage processing (Chandrasekaran, 2015). In modern food
processing methods extreme conditions, such as high temperature, high pressure,
extreme pH, salinity, etc., are required to improve the commercialization of enzymes
and their products (Li et al., 2012). This condition may affect natural enzymes. So,
most of the enzymes produced by microbes are no longer native enzymes, but
instead engineered versions. Genetic modifications through genetic engineering
and recombinant DNA technology creates Genetically Modified Organisms
(GMOs) that have specific modifications introduced into genetic material which
enable the production of new protein or food ingredient, enhance the production, and
modify the characteristics of an existing enzyme for a new application (Maghari &
368 S. Muthusamy et al.
Ardekani, 2011). Thus, the quality and productivity of enzymes can be improved.
Enzymes obtained from genetically modified organisms are intended to improve or
alter the enzymological properties or to increase the purity and yield of enzymes.
Although there are challenges and safety concerns for using recombinant proteins in
food processing, genetically modified enzymes are promising because of their
potential benefits for the food industry, consumers, and the environment (Zhang
et al., 2017).
Food enzymes are commonly used to perform a wide variety of applications in
food production, which include; lactase that reduce lactose in food and dairy
products, amylases that strengthen dough in bakery, mannase that helps in coffee
production, proteases that hydrolyze protiens, chymosin that used to make cheese,
glucoamylase, and transglucosidase those reduces starch into simple sugar and
phospholipase that refine vegetable oil. Considering the safety and synthesis of
other metabolites, selected bacterial, yeast, and fungal species are commonly
involved in the recombinant enzyme synthesis (Table 13.2). The list of major
enzymes derived from GM microbes and their function are given in Table 13.3.
The main industrial enzymes can be classified into three groups: carbohydrases,
proteases, and lipases (Castro et al., 2018). Microorganisms secrete a variety of
enzymes that plays an important role in the food manufacturing industries (Hanlon &
Sewalt, 2020). With the growing research on genetically modified organisms,
production, and development of these enzymes are also getting better.
Carbohydrases: Carbohydrases belonging to glycosidase family are a set of
enzymes that catalyzes hydrolysis of complex carbohydrates (polysaccharides) into
simple sugars (monosaccharides). These enzymes are used in the food industry for
baking, brewing, sweetener production, etc. Carbohydrases include amylases,
cellulases, glucanases, pectinases, xylanases, invertase, galactosidase, and others
(Castro et al., 2018).
Amylase: Amylase catalyzes the hydrolysis of glucosidic linkages of large
polysaccharides (starches) into oligosaccharides. These enzymes are used in the
13 Enzymes from Genetically Modified Organisms and Their Current. . . 369
the wild-type enzyme which makes the enzyme more suitable for maltose syrup
production (Li et al., 2018).
Glucosidase: These enzymes are involved in the breaking down of starch and
glycogen into their monomers. It is used as a flavor enzyme to enhance the flavor of
wine, tea, and fruit juice. β-glucosidases play a critical role in plant biomass
deconstruction. β-Glucosidases cause flavor liberation by catalyzing the breakdown
of β-1, 4-glycosidic linkages and producing free fermentable glucose used for the
production of glucose syrups from fruits and other plant tissues. In modern industrial
conditions, wild-type fungal β-glucosidases show weak activity at high glucose
concentrations limiting enzymatic hydrolysis of plant biomass. Genetic modification
of Trichoderma harzianum by site-directed mutagenesis efficiently improved the
functional properties of a β-glucosidase where the enzyme displayed high glucose
tolerance levels by the substitution of two amino acids that act as gatekeepers,
changing active-site accessibility and preventing product inhibition. The efficiency
of the engineered enzyme was enhanced by the yield of glucose and ethanol (Santos
et al., 2019). Genetically modifying Aspergillus aculeatus using site-saturation
mutagenesis produces β-glucosidase having enhanced hydrolytic efficiency, espe-
cially to cellobiose, and improves saccharification of alkaline-pretreated bagasse
(Baba et al., 2016).
Xylanses: These enzymes are involved in breaking down the hemicellulose in the
plant cell wall, specifically hydrolyzes xylan and arabinoxylan. It is used in the
application of the food industry for bread making, the production of corn starch,
clarification of fruit juice and wine, and alcoholic fermentation. In bread-making
process, xylanases are added to the dough for stabilizing the dough by hydrolyzing
polysaccharides in the wheat and making it more flexible, and enhancing the gluten
strength. Treating with xylanase increases moisture retention and shelf life of the
dough (Ahmad et al., 2014). It is also used in fruit juice processing where it breaks
down the polysaccharide plant cell walls to intensify the juice extraction efficiency
and thus extracting more nutrients and aroma compounds. Also, it helps in juice
clarification by decreasing cloudiness (Bajaj & Singh, 2010).
Even though xylanases have potential applications there is a need for xylanases
that can endure relevant processing conditions and industrial settings. A variant of
this enzyme generated using site-directed mutagenesis exhibited higher specific
activity and was utilized in the production of xylooligosaccharides from wheat
straw under thermal and alkaline conditions (50–65 C, pH 7–10) (Faryar et al.,
2015). Endo-b-1,4-xylanases with modification in secondary binding sites also
showed increased specificity toward water-insoluble, but not water-soluble, wheat
arabinoxylan, and its application in bread making resulted in increased loaf volumes
(Leys et al., 2016).
Glucanase: The glucanase enzymes help in the breakdown of polysaccharide
glucan by hydrolyzing the β-glycosidic bonds between glucose molecules in
glucans. This enzyme is mainly employed in the brewing industry where it
hydrolyses barley gums. It helps in beer brewing by lowering the viscosity of
warts, improves clarification by breaking the turbidity system by hydrolyzing the
beer haze, and aid in production of clear wart. β-glucanase is usually found
13 Enzymes from Genetically Modified Organisms and Their Current. . . 371
scale, the most commonly used sources of β-galactosidase are Aspergillus and
Kluyveromyces (Saqib et al., 2017). The enzyme source is selected depending on
the required reaction conditions. Bacterial β-galactosidases work with optimal pH
between 2.5–5.4 and are mainly used for acidic whey hydrolysis whereas yeast
β-galactosidase shows maximum activity at pH 6.0–7.0 which is more suitable for
the hydrolysis of milk and sweet whey. The temperature at which lactose-free milk
product is processed is relatively low. The lactase derived from the dairy yeast
Saccharomyces lactis has optimal process conditions (35–40 C, pH 6.6–6.8) that
are close to the natural temperature and pH of the milk (O’connell & Walsh, 2006). It
shows considerable activity at lower temperatures, down to about 4 C, and slows
the growth of spoilage bacteria thus it is the most widely used commercial lactase
source. Another source for lactase is fungal lactase derived from Aspergillus niger.
Its optimal process conditions are around 50 C and pH 3.5–4.5 so its application is
limited to acid (or acidified) whey or lactose.
To satisfy the growing demand for lactase the development of genetic engineer-
ing technology makes the large-scale industrial production of lactase possible
through the introduction or modification of genes to promote characteristics such
as higher enzyme activity or higher production. Modification of lactase produced by
microbes through directed evolution had high activity in industrial type conditions
for milk processing, that is, substrate lactose, buffer pH 6.75, and 8 C. The
production of recombinant strains using the lacA gene from Aspergillus niger and
Saccharomyces cerevisiae can produce a high quantity of lactase (Domingues et al.,
2010).
Proteases/peptidases: The major use of proteases in the food industry is the
hydrolysis of protein matrices to enhance flavor, texture, or functional properties in
dairy, meat, and fish products. Commonly used food proteases are derived from
animals which include trypsin, chymotrypsin, chymosin/rennet, pepsin, etc., and
papain, bromelain, and ficin from plants and from microorganisms microbial acid
protease, proteinase A, alkaline proteases, flavourzyme, etc. (Mazorra-Manzano
et al., 2018). Microbial proteases had vital roles in the production of traditional
fermented foods.
Trypsin: Trypsin belongs to the serine protease superfamily that cleaves on the
carboxyl end (C-terminal) arginine (Arg) and lysine (Lys) on peptide chains. Trypsin
is used as a baking enzyme to improve the workability of dough, in the extraction of
flavorings from vegetable or animal proteins, to control aroma formation in cheese
and milk products, to improve the texture of fish products, to tenderize meat, in the
production of hypoallergic food, i.e., proteases can break down specific allergic
components present in cow’s milk into nonallergenic peptides and diminishes the
risk of babies developing milk allergies (Bu et al., 2013).
Usually, trypsin for food uses has been obtained from purified extracts of bovine
or porcine pancreatic tissue but the trypsin from mammal sources has several
limitations, including high costs, security, and ethical issues. So microbial trypsin
is employed for easy preparation and low costs. But there have been a few strains,
including those from the genera of Fusarium, Streptomyces, and Trichoderma
reported. The reported microbial trypsins show poor specific activity and operational
13 Enzymes from Genetically Modified Organisms and Their Current. . . 373
Lipases: Lipases are the enzymes that catalyses the hydrolysis of long-chain
triglycerides (fats and oils). These are used for the following applications: dairy,
baking, cocoa butter substitutes, human milk fat substitutes, egg processing, and
edible oil production. In baking and dairy applications, the addition of lipases
enhances and accelerates the development of aromatic notes (Goncalves et al.,
2019). In vegetal oil processing, lipases allow a significant increase in oil yield
and enhance the appearance of end product, i.e., improves their texture and softness.
Also, lipases are used to enhance the emulsification properties of egg yolk lipids.
These enzymes help in developing new functional ingredients and functional foods,
such as cocoa butter equivalents or human milk fat equivalents. Lipases help in the
breakdown of milk fats and provide characteristic flavors to cheeses. The flavor is
due to the release of free fatty acids produced during the hydrolysis of milk fats.
Microbial lipases are produced by fungal, yeast, and bacterial species. Candida sp.,
Aspergillus sp., Rhizomucor sp., Rhizopus sp., Humicola sp., Yarrowia lipolytica,
and Pseudomonas sp., etc. are microbial sources for lipase (Tan et al., 2003).
Lipases are used widely for different applications in the food industry and thus it
is necessary to meet the increasing demand and enzymes capable of tolerating
industrial conditions. Lipase secretion by Rhizopus oryzae was improved by rational
design of the N-glycosylation sites especially for the use in the edible oil and fat
industries (Yu et al., 2017). Genetically modified Candida rugosa by site-directed
mutagenesis produces lipases having high catalytic efficiency for producing fatty
acid methyl esters and diglycerides as food emulsifiers (Chang et al., 2014).
The Generally Recognized as Safe (GRAS) is well-known process that gives general
supporting materials such as methodology involved and safety of the enzymes used
in food industries from genetically modified organisms. Section 201(s) of the Act
[21 U.S.C. 321(s)] exempts the use(s) of a substance that is GRAS from the
definition “food additive.” In general, the enzyme preparations should confirm the
purity and specifications according to “Enzyme Preparations” of the 6th (2008) or
the current edition of the Food Chemicals Codex (FCC). According to the USA, if an
enzyme is produced to use in meat and poultry products, FDA has to approve it in
consultation with the Food Safety and Inspection Service (FSIS) of the U.-
S. Department of Agriculture. The Food Allergen Labeling and Consumer Protec-
tion Act of 2004 (FALCPA) is used to prevent food allergens contamination from
enzyme. Genetically modified food enzymes, which are produced from genetically
modified organisms fall under the regulation, (Regulation (EC) No 1829/2003)
according to these regulation food enzymes produced from a part or whole of a
genetically modified organism (GMO) but it does not contain GMO. Processing aid
of the enzymes is not included in this regulation because food produced with GMO
are not included in this regulation. Genetically modified microorganisms (GMM)
which are producing food enzymes and are available in commercialized preparations
need authorization according to the regulations (EC) No. 1332/2008 and (EC) No.
13 Enzymes from Genetically Modified Organisms and Their Current. . . 375
1829/2003. The European regulation (EC) No. 1829/2003 is only regulation for the
material which is produced from genetically modified sources is used in enzyme
preparation or food production. Food enzymes fall under two categories either an
ingredient or processing aid. The risk assessment performed by the European Food
Safety Agency (EFSA) for food additives requires European regulation (EC) No
1129/2011.
There are many other European regulations for the safety evaluation for use of
food enzymes in terms of food additives, enzymes, flavorings they are (1) (EC) No.
1332/2008: Food Enzymes (2) (EC) No. 1334/2008: Food Flavourings (3) (EC) No.
1331/2008: Common Authorization Procedure (4) (EC) No. 1333/2008: Food
Additives (The European Parliament and the Council of the European Union,
2008). Some specific food enzymes had already been given authorization for use
according to regulation (EU) No. 231/2012 (The European Parliament and the
Council of the European Union 2008), which allows the specifications according
to regulations (EC) No. 1333/2008 and (EC) No. 1493/1999 lysozyme and invertase
are authorized for use in food additives. Lysozyme, Urease, and β-glucanase are
authorized for use in wine production according to regulation (Deckers et al., 2020).
Deriving food enzymes from Genetically Modified organisms have raised concerns
regarding the chance of contaminating the food with bacterial toxins or mycotoxins,
or some uncharacterized extraneous substances that may act as allergens (de Santis
et al., 2018). Since the organisms itself is genetically modified there is also a chance
for modifying their allergenic properties which increases the safety concerns
(Olempska-Beer et al., 2006) There are situations in which industrial workers
exposed to environments using genetically modified enzymes have developed type
I sensitization (Budnik et al., 2017). The regulation of enzymes including those
produced by genetic engineering is a major concern in the food industry. So before
bringing genetically modified food enzymes into the market, it is necessary to obtain
approval from various regulatory bodies such as the US Food and Drug Administra-
tion, the Association of Manufacturers and Formulators of Enzyme Products, the
European Food Safety Authority, and these processes vary in different countries.
Also, ethical and religious concerns are raised for genetically modified enzymes,
such as the raw materials used in the fermentation of microorganisms for the
production of enzymes should be halal (Ermis, 2017).
Genetically modified food enzymes are typically better suited for specific indus-
trial applications than their native counterparts, and research on their enzymological
properties in comparison to wild-type enzymes enables us to better understand how
to optimize structure-function relationships. Currently, novel genetically modified
food enzymes are mostly used for applications in food processing involving
carbohydrates, followed by lipids. Although there are challenges for safe use,
genetic modification strategies for the development of novel food enzymes are
highly promising for the future, and we expect that more genetically modified
376 S. Muthusamy et al.
Fig. 13.6 Improved technologies currently involved in genetically modified enzyme production
13 Enzymes from Genetically Modified Organisms and Their Current. . . 377
major improvements that GM microbes could provide. For example, the use of
enzyme Amino peptidases to modify by debituminize proteins which helps in the
large scale production of food products such as milk, cheese meat, beverages and
flavoring (Molinaro et al., 2005; Raksakulthai & Haard, 2003; Izawa et al., 1997).
Pectinolytic enzymes are also widely used in the food industry for large-scale
production of wine and juice production (Semenova et al., 2006).
It has been more than 15 years, since the use of genetically modified enzymes in the
food industry. With the development of genetic engineering and recombinant DNA
technology, the production of enzymes enhances the natural responsive production
and purity of enzymes. It also offers safe handling and cost-effective production of
enzymes in the commercial market. The prime factor to be considered before
commercialization of a food enzyme is the host organism that expresses an enzyme.
A microbe used in enzyme production should be well characterized, which should
not make any adverse effects by synthesizing byproducts like toxins. The safety and
hazardous risks of the enzymes from genetically modified organisms have to be
evaluated similarly to other enzymes those in use.
References
Adrio, J. L., & Demain, A. L. (2010). Recombinant organisms for production of industrial products.
Bioeng Bugs, 1, 116–131.
Ahmad, Z., Butt, M. S., Ahmed, A., Riaz, M., Sabir, S. M., Farooq, U., & Rehman, F. U. (2014).
Effect of aspergillus Niger xylanase on dough characteristics and bread quality attributes.
Journal of Food Science and Technology-Mysore, 51, 2445–2453.
Baba, Y., Sumitani, J., Tanaka, K., Tani, S., & Kawaguchi, T. (2016). Site-saturation mutagenesis
for beta-glucosidase 1 from Aspergillus aculeatus to accelerate the saccharification of alkaline-
pretreated bagasse. Applied Microbiology and Biotechnology, 100, 10495–10507.
378 S. Muthusamy et al.
Bajaj, B. K., & Singh, N. P. (2010). Production of xylanase from an alkali tolerant Streptomyces
sp. 7b under solid-state fermentation, its purification, and characterization. Applied Biochemis-
try and Biotechnology, 162, 1804–1818.
Barnett, J. A. (2003). Beginnings of microbiology and biochemistry: The contribution of yeast
research. Microbiology-Sgm, 149, 557–567.
Bawa, A. S., & Anilakumar, K. R. (2013). Genetically modified foods: Safety, risks and public
concerns-a review. Journal of Food Science and Technology-Mysore, 50, 1035–1046.
Bevan, M. W., & Chilton, M. D. (1982). Multiple transcripts of T-DNA detected in nopaline crown
gall tumors. Journal of Molecular and Applied Genetics, 1, 539–546.
Brochado, A. R., Matos, C., Moller, B. L., Hansen, J., Mortensen, U. H., & Patil, K. R. (2010).
Improved vanillin production in baker’s yeast through in silico design. Microbial Cell
Factories, 9, 84.
Bu, G. H., Luo, Y. K., Chen, F. S., Liu, K. L., & Zhu, T. W. (2013). Milk processing as a tool to
reduce cow’s milk allergenicity: A mini-review. Dairy Science & Technology, 93, 211–223.
Budnik, L. T., Scheer, E., Burge, P. S., & Baur, X. (2017). Sensitising effects of genetically
modified enzymes used in flavour, fragrance, detergence and pharmaceutical production:
Cross-sectional study. Occupational and Environmental Medicine, 74, 39–45.
Castro, A. M. D., Santos, A. F. D., Kachrimanidou, V., Koutinas, A. A., & Freire, D. M. G. (2018).
Solid-state fermentation for the production of proteases and amylases and their application in
nutrient medium production. In A. Pandey, C. Larroche, & C. R. Soccol (Eds.), Current
developments in biotechnology and bioengineering. Elsevier.
Chandrasekaran, M. (2015). Enzymes in food and beverage processing.
Chang, M., Chu, X., Lv, J., Li, Q., Tian, J., & Wu, N. (2016). Improving the thermostability of
acidic pullulanase from Bacillus naganoensis by rational design. PLoS One, 11, e0165006.
Chang, S. W., Huang, M., Hsieh, Y. H., Luo, Y. T., Wu, T. T., Tsai, C. W., Chen, C. S., & Shaw,
J. F. (2014). Simultaneous production of fatty acid methyl esters and diglycerides by four
recombinant Candida rugosa lipase’s isozymes. Food Chemistry, 155, 140–145.
Damak, S., Su, H. Y., Jay, N. P., & Bullock, D. W. (1996). Improved wool production in transgenic
sheep expressing insulin-like growth factor 1. Bio-Technology, 14, 185–188.
De Santis, B., Stockhofe, N., Wal, J. M., Weesendorp, E., Lalles, J. P., Van Dijk, D. J., Kok, E., De
Giacomo, M., Einspanier, R., Onori, R., Brera, C., Bikker, P., Van Der Meulen, J., & Kleter,
G. (2018). Case studies on genetically modified organisms (GMOs): Potential risk scenarios and
associated health indicators. Food and Chemical Toxicology, 117, 36–65.
De Souza, P. M., & Magalhaes, P. D. E. (2010). Application of microbial alpha-amylase in
industry—A review. Brazilian Journal of Microbiology, 41, 850–861.
Deckers, M., Deforce, D., Fraiture, M. A., & Roosens, N. H. C. (2020). Genetically modified micro-
organisms for industrial food enzyme production: An overview. Food, 9, 326.
Dediu, D. (2015). An introduction to genetics for language scientists: Current concepts, methods
and findings.
Dekker, P. J. T., Koenders, D., & Bruins, M. J. (2019). Lactose-free dairy products: Market
developments, production, nutrition and health benefits. Nutrients, 11, 551.
Demain, A. L., & Vaishnav, P. (2009). Production of recombinant proteins by microbes and higher
organisms. Biotechnology Advances, 27, 297–306.
Devlin, R. H., Sakhrani, D., Tymchuk, W. E., Rise, M. L., & Goh, B. (2009). Domestication and
growth hormone transgenesis cause similar changes in gene expression in coho salmon
(Oncorhynchus kisutch). Proceedings of the National Academy of Sciences of the United States
of America, 106, 3047–3052.
Domingues, L., Guimaraes, P. M., & Oliveira, C. (2010). Metabolic engineering of Saccharomyces
cerevisiae for lactose/whey fermentation. Bioeng Bugs, 1, 164–171.
Ermis, E. (2017). Halal status of enzymes used in food industry. Trends in Food Science &
Technology, 64, 69–73.
Ersoz, F., & Inan, M. (2019). Large-scale production of yak (Bos grunniens) chymosin A in Pichia
pastoris. Protein Expression and Purification, 154, 126–133.
13 Enzymes from Genetically Modified Organisms and Their Current. . . 379
Faryar, R., Linares-Pasten, J. A., Immerzeel, P., Mamo, G., Andersson, M., Stalbrand, H.,
Mattiasson, B., & Karlssona, E. N. (2015). Production of prebiotic xylooligosaccharides from
alkaline extracted wheat straw using the K80R-variant of a thermostable alkali-tolerant
xylanase. Food and Bioproducts Processing, 93, 1–10.
Fraley, R. T. (1983). Liposome-mediated delivery of tobacco mosaic virus RNA into petunia
protoplast: Improved conditions for liposome-protoplast incubations. Plant Molecular Biology,
2, 5–14.
Goncalves, D., Silva, A. G., & Guidini, C. Z. (2019). Lipases: Sources, immobilization methods,
and industrial applications. Applied Microbiology and Biotechnology, 103, 7399–7423.
Hanlon, P., & Sewalt, V. (2020). GEMs: Genetically engineered microorganisms and the regulatory
oversight of their uses in modern food production. Critical Reviews in Food Science and
Nutrition.
Headon, D. R., & Walsh, G. (1994). The industrial-production of enzymes. Biotechnology
Advances, 12, 635–646.
Herrera-Estrella, L., Block, M. D., Messens, E., Hernalsteens, J. P., Montagu, M. V., & Schell,
J. (1983). Chimeric genes as dominant selectable markers in plant cells. The EMBO Journal, 2,
987–995.
Hjort, C. (2007). Industrial enzyme production for food applications. Novel enzyme technology for
food applications. Woodhead Publishing.
Hua, W., Sheikha, A. F. E., & Xu, J. (2018). Molecular techniques for making recombinant
enzymes used in food processing. In A. F. E. Sheikha, R. Levin, & J. Xu (Eds.), Molecular
techniques in food biology: Safety, biotechnology, authenticity and traceability. John Wiley
& Sons.
Izawa, N., Tokuyasu, K., & Hayashi, K. (1997). Debittering of protein hydrolysates using
Aeromonas caviae aminopeptidase. Journal of Agricultural and Food Chemistry, 45, 543–545.
James, C. (2006). Global status of commercialized biotech/GM crops, 2006. Ithaca N.Y. New
Delhi, International Service for the Acquisition of Agri-Biotech Applications; Publication
orders, ISAAA South Asia Office.
Jay, J. M., Loessner, M. J., & Golden, D. A. (2005). Modern food microbiology. Springer.
Ke, Y., Huang, W. Q., Li, J. Z., Xie, M. Q., & Luo, X. C. (2012). Enzymatic characteristics of a
recombinant neutral protease I (rNpI) from Aspergillus oryzae expressed in Pichia pastoris.
Journal of Agricultural and Food Chemistry, 60, 12164–12169.
Kotwal, S. M., & Shankar, V. (2009). Immobilized invertase. Biotechnology Advances, 27,
311–322.
Lai, L. X., Kang, J. X., Li, R. F., Wang, J. D., Witt, W. T., Yong, H. Y., Hao, Y. H., Wax, D. M.,
Murphy, C. N., Rieke, A., Samuel, M., Linville, M. L., Korte, S. W., Evans, R. W., Starzl, T. E.,
Prather, R. S., & Dai, Y. F. (2006). Generation of cloned transgenic pigs rich in omega-3 fatty
acids. Nature Biotechnology, 24, 435–436.
Lee, J. M., Moon, S. Y., Kim, Y. R., Kim, K. W., Lee, S. J., & Kong, I. S. (2017). Improvement of
thermostability and halostability of beta-1,3-1,4-glucanase by substituting hydrophobic residue
for Lys(48). International Journal of Biological Macromolecules, 94, 594–602.
Leys, S., Pauly, A., Delcour, J. A., & Courtin, C. M. (2016). Modification of the secondary binding
site of xylanases illustrates the impact of substrate selectivity on bread making. Journal of
Agricultural and Food Chemistry, 64, 5400–5409.
Li, S., Yang, Q., Tang, B., & Chen, A. (2018). Improvement of enzymatic properties of Rhizopus
oryzae alpha-amylase by site-saturation mutagenesis of histidine 286. Enzyme and Microbial
Technology, 117, 96–102.
Li, S., Yang, X., Yang, S., Zhu, M., & Wang, X. (2012). Technology prospecting on enzymes:
Application, marketing and engineering. Computational and Structural Biotechnology Journal,
2, e201209017.
Lievens, A., Petrillo, M., Querci, M., & Patak, A. (2015). Genetically modified animals: Options
and issues for traceability and enforcement. Trends in Food Science & Technology, 44,
159–176.
380 S. Muthusamy et al.
Liu, Y. H., Zhang, T., Zhang, Z. M., Sun, T. Y., Wang, J. L., & Lu, F. P. (2014). Improvement of
cold adaptation of Bacillus alcalophilus alkaline protease by directed evolution. Journal of
Molecular Catalysis B-Enzymatic, 106, 117–123.
Lyall, J., Irvine, R. M., Sherman, A., Mckinley, T. J., Nunez, A., Purdie, A., Outtrim, L., Brown,
I. H., Rolleston-Smith, G., Sang, H., & Tiley, L. (2011). Suppression of avian influenza
transmission in genetically modified chickens. Science, 331, 223–226.
Maghari, B. M., & Ardekani, A. M. (2011). Genetically modified foods and social concerns.
Avicenna Journal of Medical Biotechnology, 3, 109–117.
Mazorra-Manzano, M. A., Ramirez-Suarez, J. C., & Yada, R. Y. (2018). Plant proteases for
bioactive peptides release: A review. Critical Reviews in Food Science and Nutrition, 58,
2147–2163.
Meyer, V. (2008). Genetic engineering of filamentous fungi—Progress, obstacles and future trends.
Biotechnology Advances, 26, 177–185.
Molinaro, G., Carmona, A. K., Juliano, M. A., Juliano, L., Malitskaya, E., Yessine, M. A.,
Chagnon, M., Lepage, Y., Simmons, W. H., Boileau, G., & Adam, A. (2005). Human recombi-
nant membrane-bound aminopeptidase P: Production of a soluble form and characterization
using novel, internally quenched fluorescent substrates. Biochemical Journal, 385, 389–397.
Murthy, P. S., & Kusumoto, K.-I. (2015). Acid protease production by Aspergillus oryzae on potato
pulp powder with emphasis on glycine releasing activity: A benefit to the food industry. Food
and Bioproducts Processing, 96, 180–188.
Nandan, A., & Nampoothiri, K. M. (2020). Therapeutic and biotechnological applications of
substrate specific microbial aminopeptidases. Applied Microbiology and Biotechnology, 104,
5243–5257.
O’connell, S., & Walsh, G. (2006). Physicochemical characteristics of commercial lactases relevant
to their application in the alleviation of lactose intolerance. Applied Biochemistry and Biotech-
nology, 134, 179–191.
Olempska-Beer, Z. S., Merker, R. I., Ditto, M. D., & Dinovi, M. J. (2006). Food-processing
enzymes from recombinant microorganisms—A review. Regulatory Toxicology and Pharma-
cology, 45, 144–158.
Packer, M. S., & Liu, D. R. (2015). Methods for the directed evolution of proteins. Nature Reviews.
Genetics, 16, 379–394.
Patel, A. K., Ania, R. R. S., & Pandey, A. (2017). Production, purification, and application of
microbial enzymes. In G. Brahmachari (Ed.), Biotechnology of microbial enzymes. Academic
Press.
Petersen, S. B. (2005). Protein engineering: Design and engineering on the nano scale. In
A. J. J. Straathof & P. Adlercreutz (Eds.), Applied biocatalysis. Harwood Academic
Publications.
Poulsen, P. B., & Bucholz, K. (2003). History of enzymology with emphasis on food production. In
J. R. Whitaker, A. Voragen, & D. W. S. Wong (Eds.), Handbook of food enzymology. Marcel
Dekker.
Puetz, J., & Wurm, F. M. (2019). Recombinant proteins for industrial versus pharmaceutical
purposes: A review of process and pricing. PRO, 7, 476.
Raksakulthai, R., & Haard, N. F. (2003). Exopeptidases and their application to reduce bitterness
in food: A review. Critical Reviews in Food Science and Nutrition, 43, 401–445.
Raveendran, S., Parameswaran, B., Ummalyma, S. B., Abraham, A., Mathew, A. K., Madhavan,
A., Rebello, S., & Pandey, A. (2018). Applications of microbial enzymes in food industry. Food
Technology and Biotechnology, 56, 16–30.
Richt, J. A., Kasinathan, P., Hamir, A. N., Castilla, J., Sathiyaseelan, T., Vargas, F., Sathiyaseelan,
J., Wu, H., Matsushita, H., Koster, J., Kato, S., Ishida, I., Soto, C., Robl, J. M., & Kuroiwa,
Y. (2007). Production of cattle lacking prion protein. Nature Biotechnology, 25, 132–138.
Rieder, L., Teuschler, N., Ebner, K., & Glieder, A. (2019). Eukaryotic expression systems for
industrial enzymes. In A. Vogel & O. May (Eds.), Industrial enzyme applications. Wiley.
13 Enzymes from Genetically Modified Organisms and Their Current. . . 381
Wheeler, M. B., Bleck, G. T., & Donovan, S. M. (2001). Transgenic alteration of sow milk to
improve piglet growth and health. Reproduction (Cambridge, England) Supplement, 58,
313–324.
Wheelwright, S. M. (1989). The design of downstream processes for large-scale protein-purifica-
tion. Journal of Biotechnology, 11, 89–102.
Wu, X., Ouyang, H. S., Duan, B. A., Pang, D. X., Zhang, L., Yuan, T., Xue, L., Ni, D. B., Cheng,
L., Dong, S. H., Wei, Z. Y., Li, L., Yu, M., Sun, Q. Y., Chen, D. Y., Lai, L. X., Dai, Y. F., & Li,
G. P. (2012). Production of cloned transgenic cow expressing omega-3 fatty acids. Transgenic
Research, 21, 537–543.
Yu, X. W., Yang, M., Jiang, C. H., Zhang, X. F., & Xu, Y. (2017). N-glycosylation engineering to
improve the constitutive expression of rhizopus oryzae lipase in Komagataella phaffii. Journal
of Agricultural and Food Chemistry, 65, 6009–6015.
Zhang, Y. H. P., Sun, J. B., & Ma, Y. H. (2017). Biomanufacturing: History and perspective.
Journal of Industrial Microbiology & Biotechnology, 44, 773–784.
Zhao, H. Y., & Feng, H. (2018). Engineering Bacillus pumilus alkaline serine protease to increase
its low-temperature proteolytic activity by directed evolution. BMC Biotechnology, 18, 34.
Current Applications of Enzymes in GM
(Genetically Modified) Food Development 14
and Food Chain
Abstract
New applications are introduced for food enzymes with regard to their impact on
the improvement of organoleptic attributes and quality of foods other than their
cost-effectiveness compared to chemical alternatives. Along with technological
advances, novel and specified enzymes have been widely used in food industries.
The enzymes may be extracted from plants and animals or be produced through
biotechnological processes. In this regard, microbial fermentation is preferred
compared to the other sources to some extent because it is a convenient process,
needs lower cost, and is conducted consistently in the laboratory. Moreover, the
fermentation process can be optimized by the use of genetically modified
(GM) microorganisms or the addition of recombinant enzymes through which
the production yield will be increased significantly. This chapter overviews the
use of GM microorganisms in order to produce food enzymes. Advantages of the
biological systems including recombinant DNA and metabolic engineering for
the production of food enzymes are also discussed. Due to the importance of
changes that occurred in the structure of GM enzymes, their halal status is further
studied. In addition, the main regulations set by countries and the safety
evaluations by risk assessment agencies on GM enzymes are addressed.
N. S. Naghavi
Department of Microbiology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
F. Mahmoodsaleh
Department of Biology, Faculty of Sciences, Shahid Chamran University, Ahvaz, Iran
M. Moslemi (*)
Halal Research Center of IRI., Ministry of Health and Medical Education, Tehran, Iran
# The Author(s), under exclusive license to Springer Nature Singapore Pte 383
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_14
384 N. S. Naghavi et al.
Keywords
14.1 Introduction
It was predicted that the market volume of industrial enzymes will have a significant
growth in the early future years (Robinson, 2015). The utilization of enzymes
especially for the production of food and beverages represents a wide industrial
enzyme market. For industrial use, it is possible to extract enzymes from plant and
animal sources or obtained by fermentation using Wild-Type (WT) or Genetically
Modified (GM) strains of bacteria or fungi for which both have been manipulated to
enhance protein production and limit the production of unfavorable secondary
metabolites. In addition to genetic modifications of the desired strain, it is also
possible to modify the enzyme itself for the purpose of obtaining an improved
yield and improve the characteristics of enzymes (Singh et al., 2016). On the other
hand, by genetic modification of producing microorganisms, the enzyme can be
modified to have better performance and improved properties.
The reasonable host strain is selected on a case-by-case analysis. The most used host
are discussed below.
14.2.1.1 Bacteria
A simple and inexpensive host organisms are bacteria, with many expression
systems suitable for recombination, leading to an easy combination of modifications.
However, because of protein folding issues, only simple proteins are made in
bacteria. In addition, bacteria are unable to make posttranslational modifications
(Deckers et al., 2020).
Commonly used bacterial host strains are including Escherichia coli, Bacillus
subtilis, B. licheniformis, and B. amyloliquefaciens. For E. coli, many molecular
toolboxes are identified to ease the construction of expression systems available for
high-yield enzyme production. However, proteins are produced in the cytosol,
leading to the requirement for extra purification steps. Nevertheless, various species
of Bacillus can secrete higher concentrations of the enzymes such as amylase and
proteases into the medium (Zhang et al., 2019; Trono, 2019).
14.2.1.2 Fungi
Fungal host strains contain a subcellular organization, which lets proteins be folded
properly and perform posttranslational modifications. Moreover, fungi secrete few
amounts of their own secondary metabolites into the medium, easing the subsequent
purificationsteps. Any other way, filamentous fungi generally produce low numbers
of non-fungal recombinant proteins (Deckers et al., 2020).
Filamentous fungi that commonly used for the production of recombinant
proteins are Aspergillus niger, A. oryzae, and Trichoderma reesei. A. oryzae, for
instance, has been utilized for decades to produce fermented foods, but the produc-
tion of the mycotoxin, aflatoxin is also known. To solve this problem, the mutant
strain BECh2 was built, in which the cluster of the aflatoxin gene and the
cyclopiazonic acid coding gene was removed and the production of Kojic acid
was declined (Fernandes, 2010; Deckers et al., 2020).
Expression vectors are used for introducing modifications indifferent host strains.
The expression vector usually includes at least One Origin of Replication (ORI), a
Multiple Cloning Site (MCS), where the gene is integrated, and a marker for
selecting the modified strain (Fig. 14.1). Besides, the promoter and terminator
regions are available to control the expression of the desired gene and selector
marker. To prevent an increase in metabolic expressions in the host organism and
a decrease in plasmid stability, small size is considered for the vector (Patel et al.,
2017).
386 N. S. Naghavi et al.
Fig. 14.1 Schematic representation of the expression vector. (a) Expression vectors include the
origin of replication (ORI), the multiple cloning site (MCS) in which the desired gene is integrated,
and a Selection Marker (SM). (b) An episomal plasmid and an integrated plasmid, containing a
flanked integration site can be formed. ARS is an independent replication sequence and IS
represents the integration site (Patel et al., 2017)
Bacterial expression vectors can either be used in the form of integration into the
host genome (integrative plasmid) or they can be extra-chromosomal, utilizing
independent replicating plasmids (episomal vector). Yeast and fungal expression
vectors are usually integrative. In addition, shuttle vectors are commonly used for
eukaryotes. These vectors can inter both prokaryotes and eukaryotes, and contain
selective markers that work in both MB groups. To insert the host genome, the
integrative plasmid consists of two Integration Sites (IS) that flanks via the Multiple
Cloning Site (MCS), allowing the cleavage of the vector and then consolidation by
recombination in the host genome containing homologous ordering to the IS. The
integration usually takes place at a unique locus. In such a case, an extension
homologous to a specific desired site needed to be added to the expression cassette
(Patel et al., 2017).
Episomal plasmids contain an origin of replication (ORI) that is homologous via
the ORI of the host type. Autonomous replication in eukaryotes utilizes an Autono-
mous Replication Sequence (ARS), which is the same as the ORI found in
prokaryotes. Different methods of transformation are employed to introduce the
vector DNA into the host organism. For bacterial host strains, this method contains
conjugation that needs cell-to-cell contact, electroporation that is done by using
competent cells, or vector incubation by constructing protoplasts. Protoplast fusion
is mainly utilized for gene transfer into eukaryotes (Deckers et al., 2020).
In order to the purpose of catch a high enzyme yield, an effective expression of the
interested gene is required. One of the most common strategies used to upgrade the
expression level is done by mix integrations (Patel et al., 2017). For this purpose,
14 Current Applications of Enzymes in GM (Genetically Modified). . . 387
various procedures are used, such as the use of high quantities of DNA in the
transformation process; high antibiotic concentration pressure that putting to use;
or simultaneous or sequential integration of multiple expression vectors that involve
different markers for the selection of transforms or via applying copies of two
preferred genes in a bidirectional vector (Deckers et al., 2020).
Other important factor affecting gene expression is the promoter strength. The
expression is usually conducted by a group of inducers and suppressors that distin-
guish constitutive promoters from tunable promoters. Constitutive promoters are
independent of the transcription factors that brought about environmental agents and
regulate the transcription from basal genes. Tunable promoters, as opposed to
constitutive promoters, are induced or suppressed by the biotic or abiotic factors in
the environment (Fitz et al., 2018). For example, amyL and amyMare, respectively
used as promoters for B. stearothermophilus maltogenic amylase and
B. licheniformis α-amylase genes (Deckers et al., 2020), and TAKA amylase
promoter is usually utilized in A. oryzae (Vieille & Zeikus, 2001). The last tunable
promoter is activated via the transcription induceramyR (amylolytic gene expres-
sion) and is suppressed via the regulator creA (carbon catabolite suppressor)
(Nedovic et al., 2011). Another promoter that is commonly used by T. reesei or
some other fungi, is the promoter of cellobiohydrolase I gene (cbh1), and activated
by cellulose (Olempska-Beer et al., 2006; Trono, 2019).
A simple solve for tandem repetition of one-directional expression cassettes is to
apply bidirectional promoters (BDPs). These primers lead to the bidirectional gene
transcription from one or more than one gene (Hui et al., 2006). Combination
expression of the desired gene and the selective marker is also available (Rieder
et al., 2019) while in order to use industrially, larger groups of accessed promoters
are required. These promoters control variable expression rates as well as combining
various regulatory profiles that are required in each expression direction (Yan & Wu,
2017).
et al., 2020). Using this method, the production of extracellular pullulanase has been
induced in a strain of B. subtilis. Because proteases can debase other enzymes, like
pullulanase, the genes involved in protease generation have been removed (Dijck
et al., 2003). The genes responsible for the production of unfavorable products
during the fermentation such as foam, or involved in spore formation may also be
disrupted to construct a more suitable strain for food enzyme production (Clyne &
Kelly, 1997). CRISPR/Cas can also be requested for the transformation of
eukaryotes. For instance, a strain of Penicillium subrubescens has been constructed
in which the ku70 gene was deleted. This gene has participated in the
non-homologous end-joining (NHEJ) DNA rebuild system. With this deletion, the
homologous repair (HR) system is preferred, and for this reason, the subjunction of a
wanted DNA sequence into the leakage site is performed (Zou et al., 2012).
The origin of enzymes usage in industries of food processing dates back to 1874,
when Christian Hansen obtained rennin (chymosin) from calves’ stomach and used
it for cheese production (Petersen et al., 2006). Chymosin is now made up of
microorganisms that involve the bovine prochymosin gene, which is imported by
recombinant deoxyribonucleic acid (rDNA) technology. The bovine chymosin,
expressed in E. coli K-12, is the first recombinant enzyme approved by the Food
and Drug Administration (FDA) for use in the food industry (Flamm, 1991).
Different enzymes that are utilized in food staffs are developed in recombinant
microorganisms. Enzyme producers use new genetic technologies to obtain enzymes
through desired properties. These kinds of enzymes most of the time are produced by
microorganisms that are fastidious in growing on laboratory or industrial scales. By
selecting suspicious host microorganisms, it is possible to produce recombinant
strains and produce enzymes that are significantly free of unwanted enzymes or
other microbial metabolites. Advanced enhancement of food processing resulted in a
request for a wide range of food processing enzymes via properties reasonable for
food processing conditions (Beilen & Li, 2002). For instance, usually applied
sweeteners like glucose or fructose syrups are usually made from corn starch
utilizing hydrolyzing enzymes. In the first stage of starch hydrolysis, the starch is
recognized by using α-amylase for 2–5 min via heating at 105 C and next put at
90–100 C for around 1–2 h. Advances in rDNA technology have made it catchable
to engineer α-amylases by improving the stability of heat and high compatibility
with other parameters of the liquefaction stage. These improvements have been
made by understanding the α-amylase amino acid sequences via the DNA sequence
of the α-amylase genes. Another enzyme recently used in food processing has also
been cached using rDNA technology. These enzymes are well-known commercially
as enzyme ministration. An enzyme preparation usually contains the interested
enzyme and varied additional substances, such as preservatives, diluents, and
stabilizers. These extra substances are usually well-known ingredients that are
accepted for utilization in food staffs (Olempska-Beer et al., 2006).
14 Current Applications of Enzymes in GM (Genetically Modified). . . 389
Halal term is a familiar concept in Muslim countries which have been interested in
non-Muslim populations in recent decades. Today, it is globally accepted that a halal
certificate refers to both religiously permitted and safe/qualified commodities. There-
fore, the halal logo facilitates international trade (Ab Talib, 2017). However, it
makes some difficulties for non-Muslim countries’ exporters such as European
392 N. S. Naghavi et al.
Fig. 14.2 Various steps for the progression of synthetic biology and metabolic engineering
procedures
because of their lower knowledge about halal (Tieman, 2017) As a global rule, foods
and their ingredients should be free of compounds that originated/are made from
haram sources or exclude alcohol. On the other hand, the animals should be
slaughtered in a way that is accepted by Islam (named as Dhabihah or Zabiha)
(Alzeer & Abou Hadeed, 2020).
Halal commitment is a critical issue in the whole food chain at the international
level. In 2001 a Japanese company was condemned due to the production of
non-halal monosodium glutamate as a flavor enhancer to achieve better economical
productivity. The process was included using BactoSoytoneas a culture media
prepared by the enzyme derived from porcine that is not acceptable in the halal
concept. This violation was further conceded by the manufacturers that were given
the halal certificate by Indonesian authorities for their products. The event obliged
other enzyme manufacturers to be approved by the halal certification bodies in the
world (Fischer, 2011). There are two types of certificating for halal foods including
1) site registration to evaluate the ability of suppliers in fulfilling the requirements
and 2) certifying the imported products (Hanzaee & Ramezani, 2011). Unfortu-
nately, the halal certifiers grow slowly compared to other scientific associations in
the world due to the disagreements on the halal concept (Halim & Salleh, 2012). On
the one hand, the term Istahala has different understanding and interpretations
among communities. There is not a consensus on the final products produced from
the non-halal origin when they underwent chemical transformation up to
microstructures or molecular level. On the other hand, there is a doubt about the
synthetic genes designed in the laboratory which are modeled from haram sources
such as pork (Riaz & Chaudry, 2019).
Enzymes are food components that are traded worldwide and their halal assur-
ance is a concern among Muslims. Food enzymes are produced from different
14 Current Applications of Enzymes in GM (Genetically Modified). . . 393
sources of plants, animals, and microbes. Several points are considered in their
eligibilities by Islamic traders. The enzymes should be extracted from halal sources
and produced in media containing halal raw materials. Furthermore, fermentation
media should contain the approved organisms other than halal ingredients if a
microbial source is used for enzyme production. It will be important when geneti-
cally modified (GM) microorganisms activate for enzyme synthesis. Based on the
World Health Organization’s definition, “genetically modified organisms can be
defined as organisms (i.e., plants, animals, or microorganisms) in which the genetic
material (DNA) has been altered in a way that does not occur naturally by mating
and/or natural recombination” (WHO, 2014). The source of genes and method of
preparation determines the fate of GM products. For example, accordance of GM
foods with religious remarks is the most important factor for Malaysians in prepara-
tion of their food basket (Oz et al., 2017). It is also a deterministic factor in Islamic
countries such as Iran through which the GM foods and components such as
enzymes first checked whether they are halal followed by safety evaluation. In this
way, the genome of host cells that is manipulated to encode the enzyme should be
originated from a halal source (Ermis, 2017). Some steps are generally monitored in
halal control points of enzymes extraction and production including the source
(animal or microbial), cleanliness of the production utilities, the agents released
into the environment, acceptable processing-aid, substrate or raw materials, and
packing process. However, up to 0.5% of alcohol may be acceptable when an
optimum activity is required (Riaz & Chaudry, 2019).
Use of animal-based enzymes is limited in some cases such as restrictions on the
dietary intake of vegans other than halal uncertainties (Nugraha et al., 2015). In
comparison, the enzymes extracted from conventional plants are not doubtful
(Khattak et al., 2011) and there is no concern about plant-derived genes inserted in
transgenic microbes (Riaz & Chaudry, 2019). However, microbial sources are used
extensively for enzyme production because of their cost-effectiveness and ease of
process control for optimization. Furthermore, it eliminates the concerns of animal
origin and slaughtering methods to meet halal criteria in enzyme production (Riaz &
Chaudry, 2019). Microbial enzymes are commonly produced by submerged fermen-
tation (Khattak et al., 2011). Several recombinant food enzymes have been
engineered to promote resistance of the wild type against harsh conditions of
temperature/pH and better reactivity of the GM enzyme with substrates in the
environment. Pichia pastoris (which was recognized as safe by US Food and
Drug Administration) (Espinoza-Molina et al., 2016) and E. coli are two species
commonly used as a host cells in this regard (Espinoza-Molina et al., 2016; Zhang
et al., 2019). In addition, GM Streptococcus thermophiles, as a common starter
culture of yogurt, capable of antioxidant enzymes’ expression was evaluated by
Carmen et al. in vitro and positive results were observed in the suppression of
oxidative stress in the colon cancer model (del Carmen et al., 2017). The bacteria
was manipulated by insertion of the gene responsible for antioxidant enzyme’
production isolated from Lactobacillus caseiBL23 as an anticancer and anti-
inflammatory probiotic bacterium (del Carmen et al., 2014; Jacouton et al., 2017)
that is considered as a halal source. Moreover, transfer of the encoding gene of
394 N. S. Naghavi et al.
chymosin from camel (Aboulnaga, 2019) and bovine (Ulusu et al., 2016) to E. coli
and from calve to E. coli, Kluyveromyces marxianus and A. niger (Fernandez-
Salguero et al., 2003) in coagulation of milk protein are other examples. At the
end of fermentation, the enzyme will be in accordance with halal merits if the
aforesaid factor is met in the processing. AbdLatip and Nordin in 2018 listed
endemic psychrophilic bacteria of South Pole as a halal source able to produce
enzymes resistant against the cold condition. In their opinion, bioengineering of
these microorganisms toward specific characteristics would be of interest in the
future because their low-temperature resistance limits the side reactions and lowers
the required activation energy (Abd Latip & Hadry Nordin, 2018).
Compared to foods, therapeutic agents may be exempted from halal regulations,
occasionally. For example, Alzeer and Hadeed in 2020 listed the orally administered
capsule of “Nutrizym 22” containing the pancreatic enzymes for pancreatitis treat-
ment as a highly critical pharmaceutical in view of halal because of its porcine origin
(Alzeer & Abou Hadeed, 2020). It is expected that there would be less restrictions on
therapeutic compounds when there are no halal alternatives.
A big deal in halal assurance is that labeling regulations in some countries suffer
from adequate liabilities. In this case, there is no obligation on listing the ingredients
of GM foods that might be originated from non-halal sources (Alhariri, 2020). With
regard to the enzymes, it is due to the fact that they may decompose during the
process, and no residue is left in the final products. Moreover, some regulatory
bodies believe that the labeling (if the product does not include significant changes in
the composition or make an adverse impact on the consumers) may threaten the
products’ sales because of consumers’ misconception about health risks arising from
GM ingredients (Uzogara, 2000).
GM foods were firstly released to the market in 1994 in the United States by
introducing a GM tomato containing the gene expressing an enzyme responsible
for modulation of the ripening process (Ujj, 2016). Since then, the countries have
had different strategies in accepting or withdrawal of such products. In general,
European countries have more restrictions on GM foods than America, and no GM
products are approved and marketed without thorough risk assessment or clear
labeling. Although, the products processed by GM enzymes or the animals fed by
GM feeds may be considered lenient (Yuen-Ting Wong & Wai-Kit Chan, 2016). For
example, the bakery products prepared by GM amylase do not require labeling in the
EU (European Commission, 2003a). In 2019, a member of the European Commis-
sion explained the EU regulation on GM foods. Other than the risk assessment,
market surveillance, and labeling, the regulation includes risk communication of the
results of safety evaluation performed by the European Food Safety Authority
(EFSA) which is responsible for risk assessment in the EU. The regulation was
implemented by the authorities for the engineered targeted gene not conventional
breeding with a long history of safety in humans, animals, and the environment.
14 Current Applications of Enzymes in GM (Genetically Modified). . . 395
However, like many other populations, people are not interested in GM foods in
Europe and the most quantities of authorized GM products are used for animal
feeding (Bruetschy, 2019). The list of authorized GM cops by the EU is available on
the European Commission website (European Commission, 2020a) and EU regula-
tion on genetically modified food enzymes is addressed in the directives of EC
No. 1332/2008 and EC No. 1829/2003 (European Commission, 2003b, 2008). As
mentioned, safety evaluations of GM food enzymes in Europe are done by EFSA
and the results are freely published to be available within the EU and for other
countries. The approved enzyme by EFSA can be commercialized under the rule of
the two above directives. The EU directive on GM modified food and feed, which is
in accordance with the Cartagena Protocol on Biosafety, clearly mentioned that the
processing aids are out of the scope of the directive and considered differently
(European Commission, 2003b). Therefore, if the food enzyme is used as a
processing aid so that irreversibly denatures, degrades, or removed before consump-
tion, is not covered by the directive. On the contrary, if the food enzyme is an
ingredient or additive in the food, it will fall under the scope of the directive of GM
food and feed that should be mandatorily listed on the labeling. Importantly, there is
an exception about the enzymes are or possibly contain allergenic or intolerant
compounds that should be mentioned on the labeling every time (European Com-
mission, 2014).
The United States has the biggest share in the production of GM foods in the
world (Oz et al., 2017). It was estimated that the arable land in the United States is
1.4 times more than that Europe but the GM crops are cultured approximately
600 times in the United States (Ujj, 2016).
Three governmental bodies in the US are involved in the safety evaluation of GM
foods from farm to fork that in turn included the Department of Agriculture (USDA),
Environmental Protection Agency (US EPA), and Food and Drug Administration
(US FDA). The first one engages in the protection of GM plants in the farm from
pests and diseases and also protects the environment against possible risks arisen by
GM crops. The second body is responsible for the safety of both environment and
humans faced with the GM traits. The last administration is responsible for the safety
of GM foods and feeds taken by humans and animals (Schiemann et al., 2019). The
US regulatory bodies focus on the GM characteristics rather than the method of
product’s synthesis. The USDA classified the GM seeds to regulated and deregulated
articles which are those that require direct- and exempted from overseeing, respec-
tively. It is based on the known safety level of the seeds. A joint collaboration of
USDA and US EPA is required for the safety evaluation of GM edible crops
containing pesticides. In this way, the producer should examine and present the
amount of pesticides existed in the final food to US EPA. The producer is allowed by
US EPA to conduct a field trial of the edible pest-resistant GM crops on a farm of 0.5
hectares or larger under US EPA consultation. The USDA studies source of the gene
producing the toxin, its characteristics, the environmental impact, and its impact on
nontarget organisms while the US EPA is responsible for toxicological,
allergenicity, and gastrointestinal fate studies on the toxin in the GM corps (Craig
et al., 2012). Other than plant-sourced biotechnological products, the safety
396 N. S. Naghavi et al.
Agriculture and the duties are delegated by the ministry to other bodies. They are
included in Inter-Ministerial Joint Meeting for integration and coordination of GM
issues, National Biosafety Committee for safety evaluation of GM products, the
Standardization Committee to amend the safety standards, testing institutions
responsible for characterization and safety examination of GM products, and other
peer governmental departments responsible for a comprehensive inspection of final
products (Jian-ping et al., 2015). There are several steps of evaluation of GM foods
before their commercialization in China. The first one is biosafety assessment which
covers laboratory to field investigations. The committee in charge of biosafety
evaluation categorized the products into 1 to 4 risk groups based on their threat to
the life cycle (Yuen-Ting Wong & Wai-Kit Chan, 2016). In this regard, no or
negligible risk, low risk, moderate risk, and high risk refer to classes 1 to 4 respec-
tively (European Commission, 2020b). Obviously, a restricted regulation is set for
classes 3 and 4. China makes an obligation on GM foods’ labeling through which the
GM ingredients or those additives degraded under processing (such as an enzyme)
should be named on the label (Yuen-Ting Wong & Wai-Kit Chan, 2016). To the
contrary, Feng and Yang stated that adequate specification has not been made in
Chinese law about labeling GM food (Feng & Yang, 2019). Safety regulation on
GM foods in China undertakes three issues of risk assessment (back to the early
twenty-first century for GM agricultural foods), labeling of GM foods to alarm the
consumers about the content, and risk communication (Sun, 2019). However, the
results of a survey showed that the Chinese population tended to lower its intake of
GM foods in 2013 compared to 2002. It may be due to their increased knowledge
about these types of foods. The authors believed that the oriented advertisement of
Chinese media (that are managed by the government) against GM foods might be an
important reason. However, the increased significant rate of neutral citizens on GM
foods in 10 years along with strict regulations and fast-growing research on GM
sources in China are effective factors on GM food development in the country
(Zheng et al., 2017).
Genetic engineering is a polar issue in the world so there are several advocates and
critics for and against the technology, respectively. The advocates are interested in
the promising events evolved in new products by the activity of recombinant genes
compared to native ones. Today, other technologies are served to increase the
functionality of recombinant proteins. For example, recombinant dextransucrase
and dextranase were protected by encapsulation techniques to be more efficient
under intended use. The enzymes were coated with edible polymers of alginate
and pectin (Sharma et al., 2019). Such polymers which are nontoxic, biodegradable,
well-defined film-former, and resistant to higher temperatures and gastrointestinal
conditions (Moslemi et al., 2018; Cheraghali et al., 2018; Sharma et al., 2019),
398 N. S. Naghavi et al.
some explanations are presented against the concern. The fans of genetic engineer-
ing believe that a chain of reactions should be occurred to transfer the target gene
into an unintended organism. They are included in excision of the gene, its resistance
within the digestive tract, its insertion into the second organism, its compatibility and
stable joining to the host, and active expression within the unwanted cell. In addition,
the specialists stated that the normal flora within the digestive tract already has the
gene encoding antibiotics and no additional worries would be expected if the known
genes are used for this purpose (Dadgarnejad et al., 2017).
It is believed that glycoprotein provokes the immune response directed by
immunoglobulin E and glycosylation process is a key factor in vital systems toward
the formation of allergenic proteins. Allergenicity of transgenic protein is evaluated
by several methods. At first, bioinformatics tests that examine homology of the new
protein and the known allergens are conducted (structural information of the
allergens is available at databases). Then, the serological tests consisting of immu-
noglobulin E binding assay are done when more than 35% similarity in a sequence of
80 amino acids in the new protein with the allergens is detected by bioinformatics
assay. Pepsin resistance test together with quantity evaluation which is important in
risk assessment studies is another way of safety evaluation. In this way, a quantified
protein is examined because less allergenic foods may be more harmful than the
others due to their high frequency of consumption. Moreover, the heat stability and
in vivo trials referred to as animal studies are common examinations. Although,
in vivo trials may not be reliable enough because it is species-specific and could not
be strictly extrapolated to humans in some cases (Yavari et al., 2016).
A guidance has been presented by EFSA about allergenicity assessment of new
proteins stimulated a non-IgE mediated response like what is observed in coeliac
patients. It includes finding out the source of protein and the population exposure
followed by amino acid sequencing by which its similarity to the known allergens is
determined. Performing in silico tests and examining the binding possibility of the
protein residues to the receptors overexpressed in coeliac disease (HLA-DQ2- and
HLA-DQ8) to evoke immunogenicity is a further step. Finally, in vitro digestibility
test is conducted (Naegeli et al., 2017).
Food enzymes that are commonly used by the industry are mainly produced by
microorganisms and their full characterization especially on the transgene organisms
is done by EFSA. The first step is collecting the taxonomic information up to strain
level for both bacteria and fungi. The whole-genome sequencing of chromosomes
and plasmid is further required. Third, an antimicrobial resistance test is done on the
producing microorganism to avoid propagation of the resistance gene among other
viable organisms. If the microbe comprises antimicrobial-resistant gene, the enzyme
must be free of genetic materials of the producing organism (Silano et al., 2019).
EFSA developed a Qualified Presumption of Safety (QPS) list containing the
microorganisms subjected to and passed the safety evaluation for food enzyme
production. Filamentous fungi are excluded from the list because of the possible
production of toxic secondary metabolites, occasionally (Koutsoumanis et al., 2020).
The QPS document also includes a safety assessment approach for enzymes produc-
ing microorganisms for additional minor evaluation, if required. Food enzymes
400 N. S. Naghavi et al.
calculating the dietary exposure of 1.701 mg/kg body weight per day in Europe. The
low MOE might be due to overestimation of dietary exposure as the scientists
presumed its intake from all brewing products and extrapolated the data of restricted
time to longtime exposure. As a conclusion, they reported that the enzyme can be
used but a low concern would not be avoidable under the defined condition of oral
consumption if no residue remained in the foods (Silano et al., 2019a).
α-amylase from GM A. niger supplied by Novozymes Company was not
genotoxic and MOE for bakeries that contain the GM enzyme accounted for
370 in Europe. Two similar segments in amino acid sequences were observed in
the allergenicity test which was matched to respiratory, not oral allergens and
resulted in a safe status of the GM enzyme for oral intake (Silano et al., 2018).
The enzyme glucan 1,4-α-glucosidase produced by two different GM organisms
of A. niger and T. reesei for conversion of starch to glucose syrup, baking, and
brewing purposes was not genotoxic. NOAEL of 1244 and 1149 mg/kg body weight
per day was detected for the enzyme produced by recombinant A. niger and T. reesei,
respectively. But, one segment of 80 amino acid in the enzyme of both origins
showed more than 35% similarity with a respiratory allergen. Although, the EFSA
panel did deny the low safety concern with regard to the oral intake of the enzyme,
particularly in those susceptible to the known respiratory allergen, but the enzyme
was finally considered safe for oral intake. It is due to the fact that more than 99% of
the enzyme is removed in glucose syrup and variable dietary exposure would be
expected for dough and beer at different countries (Silano et al., 2018a, 2020a).
Same results were observed for glucoamylase produced by GM A. niger for conver-
sion of starch to glucose syrup and brewing purposes. A NOAEL of 1360 mg/kg
body weight per day and MOE of 375 were determined for European countries.
Similarity of the enzyme with the known respiratory allergen was neglected
according to the fact mentioned above for glucan 1,4-α-glucosidase. Therefore,
GM glucoamylase was considered as safe for dietary consumption (Silano et al.,
2018b).
Three domains of GM maltogenic amylase produced by recombinant B. subtilis
(produced by Novozymes A/S) showed more than 35% similarity with the known
respiratory allergens but the GM enzyme was reported as safe by EFSA for bakery,
brewing and starch processing because no evidence was found about their
allergenicity under dietary exposure. No genotoxicity and acceptable MOE for
Europe was also reported (Silano et al., 2018c). In agreement, the same results and
conclusion on genotoxicity (negative), oral toxicity (acceptable risk), and
allergenicity (three similar domains with the known allergens) were further made
by EFSA for maltogenic amylase derived from GM B. licheniformis (Danisco US
Inc) (Silano et al., 2020b).
Xylanase is a commonly used enzyme in the cereal industry for brewing and
starch/gluten production. Its counterpart produced by T. reesei (AB Enzymes
GmbH) showed no genotoxicity, high MOE in Europe (more than 43,000), and no
allergenicity. GM xylanase is removed at the end of grain processing but a trace
amount of the enzyme delivered from brewing products might pose the consumer at
low risk. However, the EFSA considered it safe under the defined oral condition of
402 N. S. Naghavi et al.
14.8 Conclusion
Enzymes found in nature have been beneficial for the generation of fermented foods
for thousands of years. The enzymes producing microorganisms that obtained from
natural sources date back to the late-nineteenth century. Furthermore, the progress in
molecular genetics and cell biology over the past four decades have altered enzyme
generation processes. It is possible to choose and extract enzyme-encoding genes
and show them in host microorganisms that are well compatible with large-scale
industrial fermentation. In order to utilize effective promoters and introducing
variant copies of the enzyme coding genes, enzyme performance can be significantly
increased. Also, it became possible to adopt enzyme characteristics to food
processing conditions by using recombinant DNA technology. The evolution of
metabolic engineering in relation to systems biology in recent years and use them for
designation of biological systems promises to produce more sufficient food enzymes
in the early future. Currently, USA, EU, and China are the main players in GM foods
worldwide. The GM enzymes approval is based on the results of widespread risk
assessments. Safety evaluation includes homology assessment of the GM enzymes
to the known allergens, immunology evaluation, pepsin resistance, heat resistance,
and in vivo trials. With regard to labeling of the GM foods, the US FDA is more
lenient than the two others because it believes that the approved GM products do not
represent an additional risk to the consumers compared to the traditional foods.
However, all GM enzymes should be listed on the labels of food packages in China.
Upon the fate of the enzymes under processing, the EU exempts the GM enzymes
added as processing aid from labeling while those additives should be mandatorily
listed on the labels. Besides the all-positive points addressed about the GM enzymes;
the improvements have introduced a new controversial issue in Islamic countries. As
a rule, halal food enzymes should be derived from halal sources, produced in
environments containing halal ingredients, and exhausted from alcohol up to the
acceptable level in the final product. In addition, the gene responsible for encoding
recombinant enzymes should be extracted from halal origin.
References
Ab Talib, M. S. (2017). Motivations and benefits of halal food safety certification. Journal of
Islamic Marketing, 8(4), 605–624.
Abd Latip, M. A., & Hadry Nordin, N. F. (2018). Antarctica as new source for halal industrial
enzymes. In International conference on halal innovations in products and services, pp. 58–60.
Aboulnaga, E. A. (2019). Cloning and expression of camel pro-chymosin encoding gene in E. coli
and characterization of the obtained active enzyme. Journal of Food and Dairy Sciences, 10(3),
71–78.
404 N. S. Naghavi et al.
Alhariri, M. (2020). Halal and genetically modified ingredients. In Y. R. Al-Teinaz, S. Spear, &
I. H. A. Abd El-Rahim (Eds.), The halal food handbook (pp. 169–182). Wiley.
Alzeer, J., & Abou Hadeed, K. (2020). Halal certification of food, nutraceuticals, and
pharmaceuticals in the Arab world. In I. Laher (Ed.), Handbook of healthcare in the Arab
world (pp. 1–22). Springer.
Atsumi, S., Cann, A. F., Connor, M. R., Shen, C. R., Smith, K. M., Brynildsen, M. P., et al. (2008a).
Metabolic engineering of Escherichia coli for 1-butanol production. Metabolic Engineering,
10(6), 305–311.
Atsumi, S., Hanai, T., & Liao, J. C. (2008b). Non-fermentative pathways for synthesis of branched-
chain higher alcohols as biofuels. Nature, 451(7174), 86–89.
Beilen, J. B. V., & Li, Z. (2002). Enzyme technology: An overview. Current Opinion in Biotech-
nology, 13(4), 338–344.
Boyle, P. M., & Silver, P. A. (2012). Parts plus pipes: Synthetic biology approaches to metabolic
engineering. Metabolic Engineering, 14(3), 223–232.
Bruetschy, C. (2019). The EU regulatory framework on genetically modified organisms (GMOs).
Transgenic Research, 28, 169–174.
Budnik, L. T., Scheer, E., Burge, P. S., & Baur, X. (2017). Sensitising effects of genetically
modified enzymes used in flavour, fragrance, detergence and pharmaceutical production:
Cross-sectional study. Occupational and Environmental Medicine, 74, 39–45.
Cameron, D. E., Bashor, C. J., & Collins, J. J. (2014). A brief history of synthetic biology. Nature
Reviews Microbiology, 12(5), 381–390.
Casini, A., Storch, M., Baldwin, G. S., & Ellis, T. (2015). Bricks and blueprints: Methods and
standards for DNA assembly. Nature Reviews Molecular Cell Biology, 16(9), 568–576.
Cheraghali, F., Shojaee-Aliabadi, S., Hosseini, S. M., Mirmoghtadaie, L., Mortazavian, A. M.,
Ghanati, K., Abedi, A. S., & Moslemi, M. (2018). Characterization of microcapsule containing
walnut (Juglansregia L.) green husk extract as preventive antioxidant and antimicrobial agent.
International Journal of Preventive Medicine, 9, 101.
Choi, C. W., Choi, Y. H., Cha, M. R., Yoo, D. S., Kim, Y. S., Yon, G. H., et al. (2010). A new
glycoside of resveratrol dimer from stem bark of Vitisvinifera. Bulletin of the Korean Chemical
Society, 31(11), 3448–3450.
Choi, O., Lee, J. K., Kang, S. Y., Pandey, R. P., Sohng, J. K., Ahn, J. S., et al. (2014). Construction
of artificial biosynthetic pathways for resveratrol glucoside derivatives. Journal of Microbiology
and Biotechnology, 24(5), 614–618.
Clyne, R. K., & Kelly, T. J. (1997). Identification of autonomously replicating sequence (ARS)
elements in eukaryotic cells. Methods, 13(3), 221–233.
Craig, W., Vanga, S. R., & Medaglia, J. C. (2012). Commercialisation of GM crops: Comparison of
regulatory frameworks (pp. 2286–2319). Springer. https://doi.org/10.1007/978-1-4614-5797-
8_837
Dadgarnejad, M., Kouser, S., & Moslemi, M. (2017). Genetically modified foods: Promises,
challenges and safety assessments. Applied Food Biotechnology, 4(4), 193–202.
Deckers, M., Deforce, D., Fraiture, M. A., & Roosens, N. H. (2020). Genetically modified
microorganisms for industrial food enzyme production: An overview. Food, 9(3), 326.
del Carmen, S., de Moreno de LeBlanc, A., Levit, R., Azevedo, V., Langella, P., Bermudez-
Humaran, L. G., & LeBlanc, J. G. (2017). Anti-cancer effect of lactic acid bacteria expressing
antioxidant enzymes or IL-10 in a colorectal cancer mouse model. International
Immunopharmacology, 42, 122–129.
del Carmen, S., de Moreno de LeBlanc, A., Martin, R., Chain, F., Langella, P., Bermudez-
Humaran, L. G., & LeBlanc, J. G. (2014). Genetically engineered immunomodulatory Strepto-
coccus thermophilus strains producing antioxidant enzymes exhibit enhanced anti-inflammatory
activities. Applied and Environmental Microbiology, 80(3), 869–877.
Dhamankar, H., & Prather, K. L. (2011). Microbial chemical factories: Recent advances in pathway
engineering for synthesis of value added chemicals. Current Opinion in Structural Biology,
21(4), 488–494.
14 Current Applications of Enzymes in GM (Genetically Modified). . . 405
Dijck, P. W. M. V., Selten, G. C. M., & Hempenius, R. A. (2003). On the safety of a new generation
of DSM Aspergillus niger enzyme production strains. Regulatory Toxicology and Pharmacol-
ogy, 38(1), 27–35.
Donohoue, P. D., Barrangou, R., & May, A. P. (2018). Advances in industrial biotechnology using
CRISPR-Cas systems. Trends in Biotechnology, 36(2), 134–146.
Ermis, E. (2017). Halal status of enzymes used in food industry. Trends in Food Science &
Technology, 64, 69–73.
Espinoza-Molina, J. A., Acosta-Muniz, C. H., Sepulveda, D. R., Zamudio-Flores, P. B., & Rios-
Velasco, C. (2016). Codon optimization of the “Bos Taurus Chymosin” gene for the production
of recombinant chymosin in Pichia pastoris. Molecular Biotechnology, 58, 657–664.
Esvelt, K. M., & Wang, H. H. (2013). Genome-scale engineering for systems and synthetic biology.
Molecular Systems Biology, 9(1), 641.
European Commission. (2003a). Question and answers on the regulation of GMOs in the EU.
MEMO/02/160REV.
European Commission. (2003b). Regulation no. 1829/2003. Genetically modified food and feed.
Official Journal of the European Union.
European Commission. (2008). Regulation no. 1332/2008. Food enzymes. Official Journal of the
European Union.
European Commission. (2014). Guidance document on criteria for categorisation of food enzymes,
pp. 1–14.
European Commission. (2020a). Contained use. Retrieved from https://ec.europa.eu/food/plant/
gmo/authorisation/contained_en.
European Commission. (2020b). Geneticallymodified organisms, community register of GM food
and feed. Retrieved from https://webgate.ec.europa.eu/dyna/gm_register/index_en.cfm.
European Food Safety Authority. (2020). Analysis of JECFA’s draft guideline on “Evaluation of
enzyme preparations used in the manufacture of foods”. EFSA Supporting Publication, pp. 1–8.
Feng, J., & Yang, F. (2019). The regulation of genetically modified food in China. Biotechnology
Law Report, 38(5), 289–293.
Fernandes, P. (2010). Enzymes in food processing: A condensed overview on strategies for better
biocatalysts. Enzyme Research, 2010, 1–19.
Fernandez-Salguero, J., Prados, F., Calixto, F., Vioque, M., Sampaio, P., & Tejada, L. (2003). Use
of recombinant cyprosin in the manufacture of ewe’s milk cheese. Journal of Agricultural and
Food Chemistry, 51, 7426–7430.
Fischer, J. (2011). The halal frontier, Muslim consumers in a globalized market. United States,
Martin’s Press LLC. https://doi.org/10.1007/978-0-230-11978-9
Fitz, E., Wanka, F., & Seiboth, B. (2018). The promoter toolbox for recombinant gene expression in
Trichoderma reesei. Frontiers in Bioengineering and Biotechnology, 6, 135.
Flamm, E. L. (1991). How FDA approved chymosin: A case history. Nature Biotechnology, 9(4),
349–351.
Goodwin, K. D., Thompson, L. R., Duarte, B., Kahlke, T., Thompson, A. R., Marques, J. C., et al.
(2017). DNA sequencing as a tool to monitor marine ecological status. Frontiers in Marine
Science, 4, 107.
Halim, M. A. A., & Salleh, M. M. M. (2012). The possibility of uniformity on halal standards in
organization of Islamic countries (OIC) country. World Applied Sciences Journal, 17(17), 6–10.
Hanzaee, K. H., & Ramezani, M. R. (2011). Intention to halal products in the world markets.
Interdisciplinary Journal of Research in Business, 1(5), 1–7.
Hui, Y.H., Nip, W.K., Nollet, L.M., Paliyath, G. & Simpson, B.K. (2006). Food biochemistry and
food processing. Blackwell Publishing. First edition.
Jacobson, M. (2000). The genetically modified food fight. Western Journal of Medicine, 172(4),
220–221.
Jacouton, E., Chain, F., Sokol, H., Langella, P., & Bermudez-Humaran, L. G. (2017). Probiotic
strain Lactobacillus casei BL23 prevents colitis-associated colorectal cancer. Frontiers in
Immunology, 8, 1–10.
406 N. S. Naghavi et al.
Jamali, A., Moslemi, M., Akramzadeh, N., Mousavi Khaneghah, A., & Dadgarnejad, M. (2020).
Safety factors of oils marketed in Iran and applicable strategies in control of food derived
cardiovascular diseases. Current Nutrition & Food Science, 16(4), 571–577.
Jian-ping, K., Qiao-ling, T., & Xian-fa, Z. (2015). Agricultural GMO safety administration in
China. Journal of Integrative Agriculture, 14(11), 2157–2165.
Khattak, J. Z. K., Mir, A., Anwar, Z., Mustatab Wahedi, H., Abbas, G., Khattak, H. Z. K., &
Ismatullah, H. (2011). Concept of halal food and biotechnology. Advance Journal of Food
Science and Technology, 3(5), 385–389.
Khersonsky, O., & Tawfik, D. S. (2010). Enzyme promiscuity: A mechanistic and evolutionary
perspective. Annual Review of Biochemistry, 79, 471–505.
Kim, G. B., Kim, W. J., Kim, H. U., & Lee, S. Y. (2020). Machine learning applications in systems
metabolic engineering. Current Opinion in Biotechnology, 64, 1–9.
Koutsoumanis, K., Allende, A., Alvarez-Ordonez, A., Bolton, D., Bover-Cid, S., Chemaly, M.,
et al. (2020). Update of the list of QPS-recommended biological agents intentionally added to
food or feed as notified to EFSA 12: Suitability of taxonomic units notified to EFSA until March
2020. EFSA Journal, 18(7), 1–42.
Liu, D., Evans, T., & Zhang, F. (2015). Applications and advances of metabolite biosensors for
metabolic engineering. Metabolic Engineering, 31, 35–43.
Liu, X., & Kokare, C. (2017). Microbial enzymes of use in industry. In G. Brahmachari (Ed.),
Biotechnology of microbial enzymes (pp. 267–298). Elsevier.
Marchant, G. E., & Cardineau, G. A. (2013). The labeling debate in the United States. GM Crops
& Food: Biotechnology in Agriculture and the Food Chain, 4(3), 126–134.
Meyer, V. (2008). Genetic engineering of filamentous fungi-progress, obstacles and future trends.
Biotechnology Advances, 26(2), 177–185.
Moslemi, M., Hosseini, H., Neyestani, T. R., Akramzadeh, N., & Mazaheri Nezhad Fard, R. (2018).
Effects of non-digestive polymers used in iron encapsulation on calcium and iron apparent
absorption in rats fed by infant formula. Journal of Trace Elements in Medicine and Biology, 50,
393–398.
Moslemi, M., Kheirandish, M., Mazaheri, N. F. R., Hosseini, H., Jannat, B., Mofid, V., et al. (2020).
National food policies in the Islamic Republic of Iran aimed at prevention of noncommunicable
diseases. Eastern Mediterranean Health Journal, 26(12), 1556–1564.
Naegeli, H., Birch, A. N., Casacuberta, J., De Schrijver, A., Gralak, M. A., Guerche, P., et al.
(2017). Guidance on allergenicity assessment of genetically modified plants. EFSA Journal,
15(6), 1–49.
Nedovic, V., Kalusevic, A., Manojlovic, V., Levic, S., & Bugarski, B. (2011). An overview of
encapsulation technologies for food applications. Procedia Food Science, 1, 1806–1815.
Nugraha, W. T., Murti, T. W., Novitasari, I. S., Sari, T. K., Murcita, G., & Wijakangka,
G. R. T. (2015). Development of halal goat cheese using rennet like from vegetable source to
replace commercial rennet source. In The 6th international seminar on tropical animal produc-
tion, integrated approach in developing sustainable tropical animal production, pp. 733–737.
Olempska-Beer, Z. S., Merker, R. I., Ditto, M. D., & DiNovi, M. J. (2006). Food-processing
enzymes from recombinant microorganisms—A review. Regulatory Toxicology and Pharma-
cology, 45(2), 144–158.
Oz, B., Unsal, F., & Movassaghi, H. (2017). Consumer attitudes toward genetically modified food
in the United States: Are Millennials different? Journal of Transnational Management, 1–19.
https://doi.org/10.1080/15475778.2017.1373316
Patel, A. K., Singhania, R. R., & Pandey, A. (2017). Production, purification, and application of
microbial enzymes. In G. Brahmachari (Ed.), Biotechnology of microbial enzymes (pp. 13–41).
Elsevier.
Petersen, S. G., Rahbek-Nielsen, H., Hellmuth, K., & Harboe, M. (2006). Increased production of
chymosin by glycosylation. Journal of Biotechnology, 125(2), 304–310.
14 Current Applications of Enzymes in GM (Genetically Modified). . . 407
Pugh, S., McKenna, R., Moolick, R., & Nielsen, D. R. (2011). Advances and opportunities at the
interface between microbial bioenergy and nanotechnology. The Canadian Journal of Chemical
Engineering, 89(1), 2–12.
Rajamanickam, V., Metzger, K., Schmid, C., & Spadiut, O. (2017). A novel bi-directional promoter
system allows tunable recombinant protein production in Pichia pastoris. Microbial Cell
Factories, 16(1), 152.
Riaz, M. N., & Chaudry, M. M. (2019). Handbook of halal food production. CRC Press.
Rieder, L., Teuschler, N., Ebner, K., & Glieder, A. (2019). Eukaryotic expression systems for
industrial enzymes. Industrial Enzyme Applications, 47–69.
Robinson, P. K. (2015). Enzymes: Principles and biotechnological applications. Essays in Bio-
chemistry, 59, 1–41.
Schiemann, J., Dietz-Pfeilstetter, A., Hartung, F., Kohl, C., Romeis, J., & Sprink, T. (2019). Risk
assessment and regulation of plants modified by modern biotechniques: Current status and
future challenges. Annual Review of Plant Biology, 70, 699–726.
Scott, S. E., Inbar, Y., & Rozin, P. (2016). Evidence for absolute moral opposition to genetically
modified food in the United States. Perspectives on Psychological Science, 11(3), 315–324.
Sharma, M., Sangwan, R. S., Khatkar, B. S., & Singh, S. P. (2019). Alginate-pectin
co-encapsulation of dextransucrase and dextranase for oligosaccharide production from sucrose
feedstocks. Bioprocess and Biosystems Engineering, 42, 1681–1693.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018). Safety evaluation of the food enzyme α-amylase from a genetically modified
Aspergillus niger (strain NZYM-MC). EFSA Journal, 16(10), 1–17.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018a). Safety evaluation of the food enzyme glucan1,4-α-glucosidase from a genetically
modified Aspergillus niger (strain NZYM-BW). EFSA Journal, 16(10), 1–13.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018b). Safety of the food enzyme glucoamylase from a genetically modified Aspergillus
niger (strain NZYM-BF). EFSA Journal, 16(10), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018c). Safety evaluation of the food enzyme maltogenic amylase from a genetically
modified Bacillus subtilis (strain NZYM-SO). EFSA Journal, 16(11), 1–17.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018d). Safety evaluation of the food enzyme endo-1,4-β-xylanase from a genetically
modified Bacillus subtilis (strain LMG S-24584). EFSA Journal, 16(10), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018e). Safety evaluation of the food enzyme endo-1,4-β-xylanase from a genetically
modified Trichodermareesei (strain DP-Nzd22). EFSA Journal, 16(11), 1–15.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2018f). Safety evaluation of the food enzyme acetolactate decarboxylase from a geneti-
cally modified Bacillus licheniformis (strain NZYM-JB). EFSA Journal, 16(11), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2019). Characterization of microorganisms used for the production of food enzymes.
EFSA Journal, 17(6), 1–13.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2019a). Safety evaluation of the food enzyme alpha-amylase from a genetically modified
Trichodermareesei (strain DP-Nzb48). EFSA Journal, 17(1), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Bruschweiler, B. J., Cocconcelli, P. S., Crebelli, R.,
et al. (2019b). Safety evaluation of the food enzyme pullulanase from a genetically modified
Bacillus licheniformis (strain DP-Dzp39). EFSA Journal, 17(1), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020). Safety evaluation of the food enzyme α-amylase from the genetically modified Pseudo-
monas fluorescens strain BD15754. EFSA Journal, 18(3), 1–12.
408 N. S. Naghavi et al.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020a). Safety evaluation of the food enzyme glucan 1,4-alpha-glucosidase from the geneti-
cally modified Trichodermareesei strain DP-Nzh38. EFSA Journal, 18(5), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020b). Safety evaluation of the food enzyme maltogenic amylase from the genetically
modified Bacillus licheniformis strain DP-Dzr50. EFSA Journal, 18(1), 1–17.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020c). Safety evaluation of the food enzyme xylanase from the genetically modified
Trichodermareesei strain RF5703. EFSA Journal, 18(1), 1–16.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020d). Safety evaluation of the food enzyme β-galactosidase from the genetically modified
Escherichia coli NCIMB 30325. EFSA Journal, 18(1), 1–13.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020f). Safety evaluation of the food enzyme lysophospholipase from the genetically modified
Aspergillus niger strain NZYM-LP. EFSA Journal, 18(5), 1–14.
Silano, V., Baviera, J. M. B., Bolognesi, C., Cocconcelli, P. S., Crebelli, R., Gott, D. M., et al.
(2020g). Safety evaluation of the food enzyme triacylglycerol lipase from the genetically
modified Ogataeapolymorpha strain DP-Jzk33. EFSA Journal, 18(3), 1–15.
Silano, V., Baviera, J.M.B., Bolognesi, C., Cocconcelli, P.S., Crebelli, R., Gott, DM. et al. (2020e).
Safety evaluation of the food enzyme xylose isomerase from the genetically modified Strepto-
myces rubiginosusstrain DP-Pz n37. EFSA Journal, 18(1): 1–12.
Silano, V., Bolognesi, C., Castle, L., Chipman, K., Cravedi, J. P., Fowler, P., et al. (2018g). Safety
evaluation of the food enzyme alpha-amylase from a genetically modified Bacillus licheniformis
(strain NZYM-AN). EFSA Journal, 16(7), 1–14.
Silano, V., Bolognesi, C., Castle, L., Chipman, K., Cravedi, J. P., Fowler, P., et al. (2018h). Safety
evaluation of food enzyme xylanase from a genetically modified Bacillus subtilis (strain LMG
S-27588). EFSA Journal, 16(5), 1–19.
Silano, V., Bolognesi, C., Castle, L., Cravedi, J. P., Fowler, P., Franz, R., et al. (2017). Safety
evaluation of the food enzyme endo-1,4-β-xylanase from genetically modified Aspergillus niger
strain XYL. EFSA Journal, 15(5), 1–20.
Silano, V., Bolognesi, C., Castle, L., Cravedi, J. P., Fowler, P., Franz, R., et al. (2017a). Safety
evaluation of the food enzyme pullulanase from genetically modified Bacillus subtilis strain
NZYM-AK. EFSA Journal, 15(8), 1–12.
Singh, R., Kumar, M., Mittal, A., & Mehta, P. K. (2016). Microbial enzymes: Industrial progress in
21st century. 3 Biotech, 6(2), 174.
Song, R., Zhai, Q., Sun, L., Huang, E., Zhang, Y., Zhu, Y., et al. (2019). CRISPR/Cas9 genome
editing technology in filamentous fungi: Progress and perspective. Applied Microbiology and
Biotechnology, 103(17), 6919–6932.
Stephanopoulos, G. (2012). Synthetic biology and metabolic engineering. ACS Synthetic Biology,
1(11), 514–525.
Sun, J. (2019). Genetically modified foods in China: Regulation, deregulation, or governance? In
K. C. Liu & U. S. Racherla (Eds.), Innovation, economic development, and intellectual property
in India and China (pp. 347–366). Springer.
Tieman, M. (2017). Halal Europe: Apremium halal-tayyib brand? Islam and Civilisational Renewal
(ICR), 8(2), 260–263.
Trono, D. (2019). Recombinant enzymes in the food and pharmaceutical industries. In R. S. Singh,
R. R. Singhania, A. Pandey, & C. Larroche (Eds.), Advances in enzyme technology
(pp. 349–387). Elsevier.
Tseng, H. C., Martin, C. H., Nielsen, D. R., & Prather, K. L. J. (2009). Metabolic engineering of
Escherichia coli for enhanced production of (R)-and (S)-3-hydroxybutyrate. Applied and
Environmental Microbiology, 75(10), 3137–3145.
Ujj, O. (2016). European and American views on genetically modified foods. The New Atlantis,
77–92.
14 Current Applications of Enzymes in GM (Genetically Modified). . . 409
Ulusu, Y., Senturk, S. B., Kudug, H., & Gokce, I. (2016). Expression, purification, and characteri-
zation of bovine chymosin enzyme using an inducible pTOLT system. Preparative Biochemis-
try & Biotechnology, 46(6), 596–601.
US Food and Drug Administration. (2019). New plant variety consultations. Retrieved from https://
www.cfsanappsexternal.fda.gov/scripts/fdcc/index.cfm?set¼NewPlantVarietyConsultations&
sort¼fda_letter_dt&order¼desc&startrow¼151&type¼basic&search¼.
US Food and Drug Administration. (2020a). Agricultural biotechnology. Retrieved from https://
www.fda.gov/food/consumers/agricultural-biotechnology.
US Food and Drug Administration. (2020b). GRAS notices. Retrieved from https://www.
cfsanappsexternal.fda.gov/scripts/fdcc/index.cfm?set¼GRASNotices&sort¼GRN_No&
order¼DESC&showAll¼true&type¼basic&search¼.
Uzogara, S. G. (2000). The impact of genetic modification of human foods in the 21st century: A
review. Biotechnology Advances, 18, 179–206.
Vieille, C., & Zeikus, G. J. (2001). Hyperthermophilic enzymes: Sources, uses, and molecular
mechanisms for thermostability. Microbiology and Molecular Biology Reviews, 65(1), 1–43.
Vogl, T., Kickenweiz, T., Pitzer, J., Sturmberger, L., Weninger, A., Biggs, B. W., et al. (2018).
Engineered bidirectional promoters enable rapid multi-gene co-expression optimization. Nature
Communications, 9(1), 1–13.
Wang, Y., Halls, C., Zhang, J., Matsuno, M., Zhang, Y., & Yu, O. (2011). Stepwise increase of
resveratrol biosynthesis in yeast Saccharomyces cerevisiae by metabolic engineering. Metabolic
Engineering, 13(5), 455–463.
World Health Organization. (2014). Frequently asked questions on genetically modified foods.
Retrieved from https://www.who.int/foodsafety/areas_work/food-technology/faq-genetically-
modified-food/en/.
Wozniak, C. A., McClung, G., Gagliardi, J., Segal, M., & Matthews, K. (2012). Regulation of
genetically engineered microorganisms under FIFRA, FFDCA and TSCA. In C. A. Wozniak &
A. McHughen (Eds.), Regulation of agricultural biotechnology: The United States and Canada
(pp. 57–94). Springer.
Xia, X. X., Qian, Z. G., Ki, C. S., Park, Y. H., Kaplan, D. L., & Lee, S. Y. (2010). Native-sized
recombinant spider silk protein produced in metabolically engineered Escherichia coli results in
a strong fiber. Proceedings of the National Academy of Sciences, 107(32), 14059–14063.
Yan, S., & Wu, G. (2017). Bottleneck in secretion of α-amylase in Bacillussubtilis. Microbial Cell
Factories, 16(1), 124.
Yang, S., Sleight, S. C., & Sauro, H. M. (2013). Rationally designed bidirectional promoter
improves the evolutionary stability of synthetic genetic circuits. Nucleic Acids Research,
41(1), 1–7.
Yavari, B., Sarami, S., Shahgaldi, S., Athari, S. S., & Sharma, A. (2016). If there is really a notable
concern about allergenicity of genetically modified foods? Journal of Food Quality and
Hazards Control, 3, 3–9.
Yuen-Ting Wong, A., & Wai-Kit Chan, A. (2016). Genetically modified foods in China and the
United States: A primer of regulation and intellectual property protection. Food Science and
Human Wellness, 5, 124–140.
Zhang, Y., Geary, T., & Simpson, B. K. (2019). Genetically modified food enzymes: A review.
Current Opinion in Food Science, 25, 14–18.
Zheng, Z., Gao, Y., Zhang, Y., & Henneberry, S. (2017). Changing attitudes toward genetically
modified foods in urban China. China Agricultural Economic Review, 9(3), 397–414.
Zou, G., Shi, S., Jiang, Y., den Brink, J. V., de Vries, R. P., Chen, L., & el. (2012). Construction of a
cellulase hyper-expression system in Trichodermareesei by promoter and enzyme engineering.
Microbial Cell Factories, 11(1), 1–12.
Enzyme Immobilization and Its Application
Strategies in Food Products 15
Nafiseh Sadat Naghavi, Nazanin Sanei, and Martin Koller
Abstract
Keywords
Food enzymes · Immobilization · Reuse · Cost efficiency
N. S. Naghavi
Department of Microbiology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
N. Sanei
Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
M. Koller (*)
Research Management and Service, c/o Institute of Chemistry, NAWI Graz, University of Graz,
Graz, Austria
ARENA - Association for Resource Efficient and Sustainable Technologies, Graz, Austria
e-mail: martin.koller@uni-graz.at
# The Author(s), under exclusive license to Springer Nature Singapore Pte 411
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_15
412 N. S. Naghavi et al.
15.1 Introduction
Fig. 15.1 Examples for the applications of different supports for immobilization enzymes used in
food processing
enzyme that was the first to be industrially utilized already in the 1970s was amino
acylase (EC 3.5.1.14), a hydrolase which was implemented for the production of
racemic mixtures of D- and L-amino acids from N-acyl-amino acids as substrates.
Industrially, this reaction took and still takes place in columns carrying the
immobilized enzyme, while the substrate solution is washed thoroughly. Two
other effectually immobilized enzymes were the glycosidase invertase
(EC 3.2.1.26), an approved food additive (E 1103) which is used in fructose-rich
corn syrup production, and lipases (EC 3.1.1.x), a versatile group of amphiphilic that
are applied in the hydrolysis and transesterification of oily products, which, beyond
food industry, is also a novel route toward biodiesel production from waste lipids.
The advantages of immobilized enzymes in food industries are increased productiv-
ity, reduction of product recovery cost, and ultimately increased yields for diverse
food products in the future (DiCosimo et al., 2013; Homaei et al., 2013).
E
Adsorption
E Method for immobilization Covalent E
of enzymes binding E
-+
E E E
lonic binding E
E
E Irreversible Crosslinking E
E
-+ Reversible E
E
By disulfide
SS E
bonds Physical E
E
entrapment E E
E Affinity
binding
E
In irreversible enzyme immobilization, the biocatalyst that binds to the matrix cannot
be detached without causing an effect on the biological enzyme activity or the
structural properties of the support. The most usual methods of irreversible enzyme
immobilization encompass covalent binding, cross-linking, and entrapment or
encapsulation.
15.2.1.2 Cross-Linking
Crosslinking resorts to covalent linking between the enzyme and active molecules,
and is used to generate biocatalytically active polymeric particles, which can conve-
niently be used for given reactions. This mechanism is also known as “copolymeri-
zation”. Enzymes bind to each other with the aid of bi-functional reagents,
such as glutaraldehyde, glutardialdehyde, glyoxal, diisocyanates, hexamethylene
15 Enzyme Immobilization and Its Application Strategies in Food Products 415
15.2.2.1 Adsorption
The adsorption of enzymes on supports, such as activated charcoal, alumina, and ion
exchange resins is among the simplest techniques used to limit enzyme mobility
(Brady & Jordaan, 2009). Depending on the nature of amino acids present on the
surface of enzymes and the chemical nature of the support, the enzyme is fixed by
non-covalent binding trough ionic and hydrophobic interactions, or by formation of
hydrogen bonds. This method can be achieved by mixing an aqueous enzyme
solution with a matrix for a defined time, followed by a washing step to remove
the remaining free enzyme from the immobilization matrix. This method of immo-
bilization is simple and has little effect on enzyme activity and can be repeatedly
applied by adding fresh enzyme solution (“recharging” of the support with biocata-
lyst). However, in this method, shortcomings, like fast enzyme desorption from the
support, or loss of activity by changing pH-value, temperature, solvent, and ionic
strength of the surrounding environment, can hardly be avoided (Costa et al., 2005).
alterations in the temperature, solvent polarity, and ionic strength conditions (Nisha
et al., 2012).
The reaction occurring between the enzyme and a carrier generates an immobilized
enzyme with particular structural, biochemical, physical, and kinetic characteristics.
Carriers can be divided into different groups based on their appearance or their
chemical components. The support can be a synthetic or biological organic polymer,
or an inorganic solid. The support must display certain features, like extended
surface-to-volume ratio, high permeability (mass transference), acceptable func-
tional groups for enzyme binding under non-denaturing conditions, hydrophilic
moieties, insolubility in water, chemical and thermal stability, mechanical strength,
high recalcitrance, applicable particle shape, resistance to microbial attack,
regenerability, biological safety, and low or acceptable price (Sheldon, 2007b;
Garcia-Galan et al., 2011). Furthermore, multi-enzyme biocatalysis on only one
support, especially processes resorting to multi-enzyme cascades, is an emerging
approach to generate high-value chemicals on an industrial scale (Xu et al., 2020).
15 Enzyme Immobilization and Its Application Strategies in Food Products 417
Polymers
A recent method of enzyme immobilization is based on covalent linkage of such
enzymes to polymers that undergo significant structural conformation changes in
response to even minor environmental changes in terms of pH-value, temperature,
and ionic strength (Klouda & Mikos, 2008). A studied sample is poly (N-isopropyl
acrylamide) (poly NIPAM), a thermo- and bio-compatible polymer (Klis et al.,
2009). Aqueous poly NIPAM has its critical solution temperature (CST) at around
32 C. Above the CST, it becomes dissolvable because of release of water molecules
from the polymer fibers. Thus, the bio-conversion can take place under states that
maintain the enzyme solubility, thereby minimizing diffusional restriction. Subse-
quently, an increase in temperature above the CST leads to detachment of the
immobilized enzyme, thus the enzyme is recovered and can be reused (Virtanen &
Tenhu, 2000; Virtanen et al., 2000; Lozinsky et al., 2003). Polyurethane has recently
been proposed as an entrapping polymer that retains the bioactivity of biocatalysts
for long times. The usage of this polymer resulted in a remaining crude oil degrada-
tion capacity of 44.31% by a microbial consortium after more than 6 months
(Kazemzadeh et al., 2020).
Polymers that possess electrical conductivity have already been successfully
synthesized and utilized in different areas, including biotechnology. Recently, a
new class of polymers has been proposed as novel electro-active conjugated
polymers. This kind of supports exhibit interesting electrical and optical
characteristics previously reported only for inorganic systems. Electronically
directing polymers are different from all the familiar inorganic crystalline
semiconductors, such as silicon. They are molecular in nature and long chains are
absent in them. Immobilization of enzymes and biosensor construction are two
applications of these polymers (Cirpan et al., 2003). Lots of theoretical models
418 N. S. Naghavi et al.
Nanomaterials
The headway of nanotechnology in the 1990s was preceded by the quick evolvement
of nanobiotechnology including the construction of nanobiocatalysts. In the early
methods applied in nanobiocatalysis, enzymes were immobilized on various
nanostructured materials utilizing conventional procedures, like simple adsorption
and covalent linkage. This method attracted attention for immobilizing enzymes on a
wide range of nanostructured matrices, like porous nanomaterials, electroconductive
nanofibers, and magnetic nanoparticles (MNPs). This large area provides better
enzyme loading, which in turn improves the enzyme mobility in comparison to
immobilized enzyme systems on conventional matrices. One of the special
advantages of nanostructured materials is that the pore size in nanopores, nanofibers,
or nanotubes can be controlled at nanometer scale (Homaei et al., 2013).
Recently, nanobiocatalytic methods have evolved from simple strategies for
immobilization of enzymes (Homaei et al., 2013). By rapid advancement in nano-
technology, MNPs are presently extremely interesting. The physico-chemical
characteristics of MNPs can widely differ from the properties of the bulk
material from which the nanoparticle is made; this has attracted attention in these
materials also for enzyme immobilization. For instance, magnetizing a particle in a
particular direction by magnetic anisotropy is usually done on the surface of a
particle (Schellenberger et al., 2002). Nanoparticles constructed by ultra-small
superparamagnetic iron oxide (Kooi et al., 2003; Keller et al., 2004), cross-linked
iron oxide (CLIO), and mono-crystalline iron oxide (Krause et al., 2004) all were
fabricated as imaging elements in magnetic resonance imaging (MRI). Magnetic
particles are used for enzyme immobilization for the purpose of increasing the
stability of the biocatalyst, to maintain the stability of the catalyst, and, importantly,
they can conveniently be separated from the interaction environment and recovered
by applying an external magnetic field. (Bilal & Iqbal, 2019). MNPs perform best at
typical sizes ranging between 10 and 20 nm, where superparamagnetism emerges
(Netto et al., 2013). Such magnetic particles have been suggested for biotechnologi-
cal applications (Kluchova et al., 2009; Defaei et al., 2018) or for developing
analytical systems, like biosensors (Bilal & Iqbal, 2019; Kouassi et al., 2005).
Nanostructured metal oxides (NMOs) recently became of interest in the area of
enzyme immobilization because these materials have the best structural revising and
high bioactivity, which leads to elevated sensing properties (Antony et al., 2016). As
a part of NMOs, MNPs have been widely utilized in enzyme immobilization because
of their advantageous properties, such as their size, magnitude, higher safety levels,
better reusability, wide surface, and large capacity of enzyme loading. Because they
have an inactive surface that limits direct binding to enzymes, protective molecules
must be coated on MNPs to supply dynamic functional groups for immobilization of
enzymes (Amirbandeh & Taheri-Kafrani, 2016; Mehnati-Najafabadi et al., 2018).
With all these benefits in mind, MNPs are highly inhibited in acidic and oxidative
15 Enzyme Immobilization and Its Application Strategies in Food Products 419
conditions. Therefore, coating of the outer protective surface is so vital to sustain the
consistency of MNPs (Landarani-Isfahani et al., 2015). In a recent study, Defaei
et al. (2018) immobilized the hydrolase α-amylase (EC 3.2.1.1) onto naringin-
functionalized MNPs by ionic reactions. The MNPs were covered with naringin,
which is a biocompatible flavonoid. The appearance, structure, and features of
functionalized MNPs and the immobilization status of the nanocomposite were
determined by analytical instruments, like thermogravimetry (TGA), vibrating sam-
ple magnetometer (VSM), Fourier transform infrared spectroscopy (FTIR), scanning
electron microscopy with energy dispersive X-ray (SEM-EDX), and transmission
electron microscopy (TEM). In addition, the optimum conditions of temperature,
pH-value, interaction time, and enzyme tendency for better immobilization were
evaluated. The results evaluated the optimum conditions for α-amylase immobiliza-
tion on the synthesized nanocarrier at pH 6.5 and a temperature of 55 C. Reuse
experiments showed high maintenance of immobilized α-amylase activity even after
10 reaction repeats. Furthermore, the storage consistency of enzyme immobilization
was repaired by immobilization compared to that of the free enzyme and it retained
60% of its original activity even after 6 weeks of storage at 4 C. Improving the
catalytic properties of enzymes through immobilization has made this
nanobiocatalyst a feasible tool in bio-industrial systems.
Another field of application of MNPs as supports for immobilized enzymes is the
pharma sector; here, it was shown that the enzyme penicillin G acylase (PGA; EC
3.5.1.11) can be immobilized to functionalized MNPs (epoxy-activated magnetic
cellulose beads) due to the cavity and affinity forces in the matrix of activated
cellulose, and applied for hydrolytic removal of the side chain of penicillin G
molecules, generating 6-aminopenicillanic acid as product. Improved biocatalytic
activity and stability of the enzyme were reported for this process in comparison to
the use of free enzyme (Luo & Zhang, 2010). In recent years, this process was
improved by various research groups globally, such as by Liu and colleagues, who
covalently immobilized PGA on hydroxy- and aldehyde-functionalized magnetic
Fe2O3/Fe3O4 nanoparticles (Liu et al., 2020), or Zhaoyu et al., who used novel
di-functional magnetic “nanoflowers,” equipped with epoxy groups and hydrophilic
catechol as well as with phthaloquinone groups enabling the covalent coupling of
penicillin G acylase (Zhaoyu et al., 2020).
stable in the entire pH-range of 0 to 14 and are not inactivated even at a sudden and
tremendous pH-change. A basic disadvantage of hydrophilic resins are diffusion
limitations that have been shown in kinetically monitored procedures. Immobiliza-
tion by covalent binding to acrylic resins has been successfully utilized for a variety
of enzymes in industrial operations (Katchalski-Katzir & Kraemer, 2000; Boller
et al., 2002).
Enzymes may also be immobilized in biological or artificial hydro- or cryo-gels in
an insoluble environment. For example, poly (vinyl alcohol) (PVA) crucibles made
by melting ice are frequently used for whole-cell immobilization (Tripathi et al.,
2010). Also, because of their minor size, free enzymes can spread from the gel
matrix and get dissolved in an aqueous medium. To trap free enzymes, the enzyme
size should be enlarged by mechanisms, such as cross-linking. Another option to
increase the biocatalytic enzyme size is to design a composite material with a
polyelectrolyte. Because of their ampholytic nature, proteins are released either as
polycations or polyanions, based on the environmental pH-value. Therefore, the
typically can form complexes with polyelectrolytes of opposite charges (Homaei
et al., 2013).
Fig. 15.3 Schematic illustration for multi-enzymes co-immobilization using supports, such as
polymers, graphene, silica, MOFs, DNA, and CNTs
which notably addressable DNA nanocomposites can ease the suitable self-
aggregation of varied enzymes and optimize the substrate penetration (Xu et al.,
2020).
Unlike pharmaceutical industries and some chemical industries, the food industry
requires the production of vast quantities of commercial products. For this purpose,
the cost of the biocatalyst should be lowered, thus, using immobilized enzymes show
acceptable operational consistency that allows lots of repetitive production cycles to
be carried out. In the food sector, continuous fermentation processes are preferred to
15 Enzyme Immobilization and Its Application Strategies in Food Products 423
parallel with maintenance of the properties obtained by the sugar ingredient, such as
browning, bulking, texture, and sweetness. Allulose is also supposed to display
potential antihyperglycemic effects, and was shown to prevent postprandial hyper-
glycemia in humans. More than 0.2 g per day human allulose intake is estimated
when consuming naturally found materials, such as processed cane and beet molas-
ses, coffee treated via steam, wheat products, and HFCS. In 2012, allulose was
labeled as generally recognized as safe (GRAS) by the FDA and approved as
sweetener in food products (not yet approved in, e.g., the European Union!), so it
is commercially utilized as food additive in some parts of the world. Moreover,
ketose-3-epimerase (EC 5.1.3.31), an isomerase which is found in various
microorganisms, can interconvert fructose to allulose and vice versa (Basso &
Serban, 2019).
Another emerging sweetener is the ketohexose tagatose, which can be isolated from
animal origins. Tagatose is a kind of sweetener with 92% fructose sweetness, but
contains only 38% of fructose calories. Its catabolic route is different from sucrose;
therefore, it has insignificant effect on insulin and blood glucose levels. Moreover,
tagatose is considered a “tooth-friendly” compound for dental care products.
Tagatose can be gained from lactose with only 16% of the sweetening of the later.
The disaccharide lactose (β-D-galactopyranosyl-(1 ! 4)-D-glucose) is a natural sugar
found in milk and normally makes up 2–8% of its total mass. The process of tagatose
production utilizing the immobilized glucosidase β-galactosidase (3.2.1.23) has been
reported in 2014. This process suggests preparation and valorization of lactose
present in whey at a concentration of 18 wt.-%. It is possible to obtain tagatose by
lactose hydrolysis using immobilized β-galactosidase to produce the monomers
glucose and galactose. Then glucose is removed from the mixture by
deglycosylation using baker’s yeast; now, tagatose is obtained by epimerization of
galactose with aerated Ca(OH)2 (Basso & Serban, 2019). There have also been
reports on the utilization of immobilized L-arabinose isomerase (EC 5.3.1.4) to
obtain tagatose in stirred tank reactors or continuous flow systems (Lim et al.,
2008; Oh, 2007).
Another sweetener for food and beverage industry, also accessible by means of
biocatalysis, is the canonic amino acid L-aspartate (2-aminobutanedioic acid). Aspar-
tate production is performed by amination of fumaric acid; this reaction is catalyzed
by the lyase enzyme aspartase (aspartate ammonia-lyase, EC 4.3.1.1). Already in
1973, Tosa et al. reported the application of aspartase from Escherichia coli for
aspartate production from ammonium fumarate; these authors immobilized the
enzyme by different methods: ionic binding on cellulose derivatives or sephadex,
15 Enzyme Immobilization and Its Application Strategies in Food Products 425
The polyphenol naringin, a flavonoid, is responsible for the bitter taste of citrus
fruits. There is increased interest in fruit juice industry by using highly efficient
immobilized enzymes for debittering of citrus fruit juices (Puri et al., 2008). Already
in 1979, Olson and co-workers reported the immobilization of commercially avail-
able naringinase (mixture of the hydrolases α-rhamnosidase, EC 3.2.1.40, and
β-glucosidase, EC 3.2.1.21; hydrolyzes naringin to naringenin, glucose, and rham-
nose) in a reactor system consisting of polysulfone hollow fibers; immobilization
426 N. S. Naghavi et al.
took place by ultrafiltration of the enzymes into the sponge region of the hollow
fibers. After 210 min of continuous operation, 50% of naringin contained in grape-
fruit juice was hydrolyzed at 25 C and a flow rate of 300 mL/min (Olson et al.,
1979). Other approaches to immobilize naringinase encompass entrapping in algi-
nate, which resulted in 60% debittering of kinnow juice after 3 h when using a total
enzyme activity of 30 U (Puri et al., 1996), immobilization on electrospun cellulose
acetate nanofibers (Huang et al., 2017), on chitin (Tsen, 1984) or chitosan
microspheres (Bodakowska-Boczniewicz & Garncarek, 2019) by linking with glu-
taraldehyde, or adsorption of the enzyme on mesoporous molecular sieves via
glutaraldehyde for naringin hydrolysis in white grapefruit juice (Lei et al., 2011).
A rather bizarre protocol for naringinase immobilization was developed by Puri and
colleagues, who attached the enzyme on chicken egg white beads obtained by cross-
linking the protein with glutaraldehyde; debittering of Kinnow juice achieved an
efficiency of 68% (Puri et al., 2001). In addition, Busto et al. immobilized thermo-
philic Aspergillus niger naringinase by entrapping it into a PVA hydrogel matrix,
which was cryostructured in liquid nitrogen, to generate beads biocatalytically active
for naringin hydrolysis. Authors reported high stability of the beads; after storage at
4 C for 2 months, they retained 75% of initial activity (Busto et al., 2007).
One of the main water-soluble natural antioxidants is L-ascorbic acid (vitamin C). L-
ascorbic acid and its derivatives act as free radical scavengers, reacting with oxygen,
and destroying it. Moreover, hydrophobic long-chain fatty acid ester derivatives of
L-ascorbic acid are used as antioxidants in fat-rich food because of their higher
ability to dissolve in fats in comparison to the typical hydrophilic compound
vitamin C, which is insoluble in oils (Burham et al., 2009). In this context, ascorbic
palmitate and stearate are currently prepared by reaction between ascorbic acid with
sulfuric acid, followed by re-esterification with the corresponding fatty acid; finally,
a purification step by re-crystallization is carried out (Ferreira-Dias et al., 2013). In a
biocatalytic approach, immobilization of Candida antarctica B lipase (CalB) was
utilized for generation of ascorbyl esters. The biocatalytic conversion can reach a
yield of approximately 95%, depending on process temperature, the level of removal
of the side product water, and fatty acid chain length. In spite of the fact that
enzymatic synthesis suggests some benefits to the current chemical procedures,
such as interaction in the lower temperatures than chemical reactions temperatures,
higher material purity, and decreased downstream processing expenditure, many of
the manufactures of ascorbyl esters still carry out this synthesis by chemical pro-
cesses, because of the long interaction time needed by the enzymatic procedure and
the high price of the immobilized enzymes in contrast to the chemical catalysts
(Villeneuve, 2007).
The source of vegetable oils, such as palm, rapeseed, canola, and sunflower,
specifies the physical characteristics of fats and oils present in food products,
15 Enzyme Immobilization and Its Application Strategies in Food Products 427
because each oil has a different arrangement and type of saturated mono- and
polyunsaturated fatty acids in the 1, 2, and 3 locations of triacylglycerides. To obtain
the suitable melting properties of fats and oils, especially in the generation of
margarine and baking fat, chemical hydrogenation, fractioning, and esterification
have been applied. The enzymatic transesterification of food oils and fats is one of
the benefits because of the option to better monitor the product composition com-
pared to chemically transesterified products due to the removal of the hydrogenated
trans fats that have important health challenges (Marangoni & Rousseau, 1998; Asif,
2011).
Enzymatic transesterification was first investigated to produce an equivalent of
cocoa that used the sn-1,3 specificity of different fungal lipases. Cocoa butter
homologs are semisolid oils that commonly have a melting temperature of 37 C.
They are obtained from more cost-effective origins than cocoa, like palm, sunflower,
or rapeseed oil. A variety of commercial processes have been developed to produce
the equivalent of cocoa butter with elevated amount of the demanded triglycerides,
1(3)-palmitoyl-3(1)-stearoyl-2-monooleine, and 1,3-distearoyl-2-monooleine,
required for chocolate production. Most systems are made by using fungal lipases
immobilized by surface adsorption or encapsulation in liposomes (Basso & Serban,
2019).
Bovine milk contains 4.3–4.5 wt.-% lactose that exposes 38–40% of the whole milk
solids. Lactose in milk and milk products is not hydrolyzed in the stomach or in the
initial part of the small intestine; it enters to other parts of the intestine and gets
hydrolyzed into the monosaccharides D-galactose and D-glucose by the glycosidase
β-galactosidase (lactase, EC 3.2.1.23) excreted by the intestinal microflora. About
65% of the entire human population (up to 90% in some Asian countries) are unable
to secrete sufficient quantities of β-galactosidase, causing many health disorders.
Elimination of lactose from milk and milk products makes them suitable for con-
sumption by people with lactose intolerance (hypolactasia), so the dairy industry has
demonstrated great interest to develop advanced lactose hydrolysis processes based
on β-galactosidase. Because the sweetening potential of lactose, glucose, and galac-
tose is 20, 70, and 58%, respectively, of sucrose, lactose-hydrolyzed milk is sweeter
than pristine milk (Panesar et al., 2010).
The simplest but most expensive solution for this problem is to add free
β-galactosidase to whole milk. Enzyme activity is stopped after complete substrate
hydrolysis, typically combined with pasteurization. Another procedure is the usage
of immobilized β-galactosidase for processing of skimmed milk; after completion of
hydrolysis, the fat fraction is added again to the hydrolyzed milk to reassemble its
nutritious components. This technique, of course, displays the benefits of recycling
and reusing the immobilized enzyme in contrast to adding free enzyme, and the final
product is free from additional ingredients, like enzymes or components or the
enzyme formulation that can constitute putative allergens.
428 N. S. Naghavi et al.
A notable amount (approximately 40%) of all types of food are disposed as waste
(Godfray et al., 2010), and these losses not only lead to environmental pollution but
also affect the entire food chain. This amount varies between different geographic
regions, and one should differentiate between food waste sensu stricto and agricul-
tural waste. These waste streams are responsible for a major global challenge both in
15 Enzyme Immobilization and Its Application Strategies in Food Products 429
causing environmental pollution and ethical concern considering the huge number of
people starving worldwide. With the global population expected to increase to 9.8
billion until 2050, suitable technological solutions should be developed to solve this
problem. Some technical proposals are represented at the food processing level.
Liquid food processing waste contains numerous organic carbonaceous compounds;
therefore, it has high biological oxygen demand (BOD) that causes problems for
direct disposal of them to wastewater removal plants. Here, the disposal of about one
million liters of lactose-rich whey per day only on the Northern Italian region
constitutes a prime example (Koller et al., 2016). Hence, the lipid, carbohydrate,
and protein contents of food and agricultural waste liquids are leading to high BOD;
however, at the same time, they have the potential to be converted to valuable
products, thus upgrading waste liquids into potential recoverable sources. Examples
of such conversions include oxidation, hydrolysis, acylation, and phosphorylation of
carbohydrates as well as glycosylation and deamination of amino acids, and esterifi-
cation and hydrogenation of lipids. In particular, esterification processes are widely
used for production of different value-added food and agricultural products. Waste
oils and animal waste lipids from the slaughtering and rendering industry can be
transesterified with alcohol to generate biofuels (Koller et al., 2018). Esterified
sugars can be applied as surfactants, and esterified starch may be used as biodegrad-
able plastics and adhesives. Esterification of flavonoids was reported to increase their
life time, health, and acceptance characteristics (Walle, 2009). Traditional processes
to these transformations require significant amounts of chemical catalysts and energy
resources that have limited reactivity, and lead to formation of by-products, espe-
cially when done in complex matrices, like food waste liquids (Alissandratos &
Halling, 2012; Fang et al., 2002).
Food processing waste streams which are carbohydrate-rich can easily be converted
by the enzymatic valorization catalyzed by hydrolases and isomerases into more
valuable products, like sweeteners and prebiotics. In fact, some of the best accepted,
well-known procedures in food and agricultural systems begin with the use of
carbohydrate substrates. In this context, immobilized thermophilic enzymes
(“thermozymes”), which have been studied for the production of high fructose
corn syrup, could be progressed and used for valorization of food waste liquids
that are carbohydrate rich (Andler & Goddard, 2018). Emtiazi et al. (2001) used
immobilized cellulase enzymes from Aspergillus terreus to decrease chemical oxy-
gen demand (COD) by cellulose removal (40–80%) from pulp manufacturing waste.
surfactants, and lubricants, by the use of enzymes. Lipases, like most other enzymes,
can be mined from different microbial sources with different performance properties.
For instance, lipases produced by Thermomyces lanuginosus and Candida
antarctica were used for lipid hydrolysis and esterification, respectively, yielding
valuable products, such as biofuel, from waste cooking oil. More than 90% conver-
sion was obtained after 10 h of hydrolysis and 10 h of esterification reactions.
Noteworthy, after 5 catalysis repeats, the lipase from C. antarctica retained its
activity, while the lipase from T. lanuginosus lost some of its activity after each
use (Vescovi et al., 2016). In another study, Rhizomucor miehei lipase and
C. antarctica lipases were immobilized on silica particles that were epoxy-
functionalized and used to enhance the performance of biofuel generation from
waste cooking oil. A 91.5% conversion rate was achieved during 10 h (Babaki
et al., 2017).
Other examples for immobilization of C. antarctica lipase for conversion of lipid
substrates to value-added products encompass the use of support materials as diverse
as core–shell MNPs for conversion of waste cooking oil to biodiesel (Mehrasbi et al.,
2017), immobilization by adsorption on poly (styrene) nanoparticles (Miletić et al.,
2010), covalent attachment on chitosan-based hydrogels (Silva et al., 2012), or
adsorption to green coconut fibers (Brígida et al., 2007).
Proteinaceous food waste may stem from different origins like dairy products (whey
retentate), grains (Zhi et al., 2017), oilseeds (Doshi et al., 2014), soybeans (Laskar
et al., 2018), eggs (Hong et al., 2019), or even poultry feathers (Pernicova et al.,
2019). Proteases are used for hydrolyzing proteins from waste streams and
converting them to biological peptides or useful chemicals, such as the monomers
that build up polymers. Here, enzymatic pathways are more favorable than chemical
reactions, which are not easily controllable, as observed for the degradation of
tryptophan via acid hydrolysis (Kumar et al., 2015). Immobilized trypsin has been
used for hydrolysis of dairy waste, such as whey protein (retentate fraction
remaining after ultrafiltration of full whey), as an alternative to well-established
acidic hydrolysis (Koller et al., 2019). Immobilization of bovine pancreas trypsin on
porous polymethacrylate with a pore size of 2.1 μm has led to 9.68% hydrolysis. The
hydrolysis degree had reached ~6% under the same conditions when using free
trypsin, which indicates the need for optimization of the immobilization process.
Most significantly, the peptide analysis differed between the immobilized and free
trypsin that showed the effect of immobilization methods on enzyme selectivity for
hydrolysis of amino acid sequences (Mao et al., 2017). Trypsin has also been
immobilized on reusable matrices containing spent grain and lignocellulose for
hydrolysis of whey protein (Tavano, 2013; Bassan et al., 2016).
15 Enzyme Immobilization and Its Application Strategies in Food Products 431
15.9 Conclusions
Enzymes usage in soluble form for food processing is well established. Although a
high variety of enzymes have already been immobilized and used in different food
manufacturing industries, only few procedures have become practical and economi-
cal, and succeeded in getting established on the long term. Numerous recent
concepts have been attempted or are being used in this field. The future of such
applications and operations will depend mainly on their cost and also political
decisions. Despite the fact that not all established transformations involved in food
processing and food production can to date be replaced by biocatalytic techniques
resorting to immobilized enzymes, and although many immobilization processes
holding promise in lab-scale experiments are not yet scalable to industrial
dimensions, the outlook for immobilized enzymes in food industry is indeed
promising considering current food industry trends to become more efficient and
sustainable, combined with the rapid progression of immobilized enzyme
techniques. In any case, due to the growing human population on Earth and a
remarkable decrease in limited natural food resources, the future use of immobilized
enzymes may elevate significantly in order to produce higher amounts of food
products, and even to unlock alternative food sources for enhanced global food
security.
432 N. S. Naghavi et al.
References
Albayrak, N., & Yang, S. T. (2002). Immobilization of β-galactosidase on fibrous matrix by
polyethyleneimine for production of galacto-oligosaccharides from lactose. Biotechnology
Progress, 18(2), 240–251.
Alissandratos, A., & Halling, P. J. (2012). Enzymatic acylation of starch. Bioresource Technology,
115, 41–47.
Alkan, S., Gür, A. Y. C. A. N., Ertan, M., Savran, A., Gür, T., & Genel, Y. (2009). Immobilization
of catalase via adsorption into natural and modified active carbon obtained from walnut in
various methods. African Journal of Biotechnology, 8(11).
Amirbandeh, M., & Taheri-Kafrani, A. (2016). Immobilization of glucoamylase on triazine-
functionalized Fe3O4/graphene oxide nanocomposite: Improved stability and reusability. Inter-
national Journal of Biological Macromolecules, 93, 1183–1191.
Andler, S. M., & Goddard, J. M. (2018). Transforming food waste: How immobilized enzymes can
valorize waste streams into revenue streams. NPJ Science of Food, 2(1), 1–11.
Antony, N., Balachandran, S., & Mohanan, P. V. (2016). Immobilization of diastase α-amylase on
nano zinc oxide. Food Chemistry, 211, 624–630.
Asgher, M., Shahid, M., Kamal, S., & Iqbal, H. M. N. (2014). Recent trends and valorization of
immobilization strategies and ligninolytic enzymes by industrial biotechnology. Journal of
Molecular Catalysis B: Enzymatic, 101, 56–66.
Asif, M. (2011). Process advantages and product benefits of interesterification in oils and fats.
International Journal of Nutrition, Pharmacology, Neurological Diseases, 1(2), 134.
Axelsson, A., & Zacchi, G. (1990). Economic evaluation of the hydrolysis of lactose using
immobilized β-galactosidase. Applied Biochemistry and Biotechnology, 24(1), 679.
Babaki, M., Yousefi, M., Habibi, Z., & Mohammadi, M. (2017). Process optimization for biodiesel
production from waste cooking oil using multi-enzyme systems through response surface
methodology. Renewable Energy, 105, 465–472.
Bassan, J. C., de Souza Bezerra, T. M., Peixoto, G., Da Cruz, C. Z. P., Galán, J. P. M., Vaz,
A. B. D. S., et al. (2016). Immobilization of trypsin in lignocellulosic waste material to produce
peptides with bioactive potential from whey protein. Materials, 9(5), 357.
Basso, A., & Serban, S. (2019). Industrial applications of immobilized enzymes—A review.
Molecular Catalysis, 479, 110607.
Bhosale, S. H., Rao, M. B., & Deshpande, V. V. (1996). Molecular and industrial aspects of glucose
isomerase. Microbiological Reviews, 60(2), 280–300.
Bilal, M., & Iqbal, H. M. (2019). Sustainable bioconversion of food waste into high-value products
by immobilized enzymes to meet bio-economy challenges and opportunities–A review. Food
Research International, 123, 226–240.
Blanco, R. M., Terreros, P., Fernández-Pérez, M., Otero, C., & Díaz-González, G. (2004).
Functionalization of mesoporous silica for lipase immobilization: Characterization of the
support and the catalysts. Journal of Molecular Catalysis B: Enzymatic, 30(2), 83–93.
Bodakowska-Boczniewicz, J., & Garncarek, Z. (2019). Immobilization of naringinase from Peni-
cillium decumbens on chitosan microspheres for debittering grapefruit juice. Molecules,
24(23), 4234.
Boller, T., Meier, C., & Menzler, S. (2002). Eupergit oxirane acrylic beads: How to make enzymes
fit for biocatalysis. Organic Process Research & Development, 6(4), 509–519.
Brady, D., & Jordaan, J. (2009). Advances in enzyme immobilization. Biotechnology Letters,
31(11), 1639.
Brígida, A. I., Pinheiro, Á. D., Ferreira, A. L., & Gonçalves, L. R. (2007). Immobilization of
Candida antarctica lipase B by adsorption to green coconut fiber. In Biotechnology for fuels and
chemicals (pp. 293–307). Humana Press.
Burham, H., Rasheed, R. A. G. A., Noor, N. M., Badruddin, S., & Sidek, H. (2009). Enzymatic
synthesis of palm-based ascorbyl esters. Journal of Molecular Catalysis B: Enzymatic, 58(1–4),
153–157.
15 Enzyme Immobilization and Its Application Strategies in Food Products 433
Busto, M. D., Meza, V., Ortega, N., & Perez-Mateos, M. (2007). Immobilization of naringinase
from Aspergillus niger CECT 2088 in poly (vinyl alcohol) cryogels for the debittering of juices.
Food Chemistry, 104(3), 1177–1182.
Chen, Y., Yu, B., Lin, J., Naidu, R., & Chen, Z. (2016). Simultaneous adsorption and biodegrada-
tion (SAB) of diesel oil using immobilized Acinetobacter venetianus on porous material.
Chemical Engineering Journal, 289, 463–470.
Cirpan, A., Alkan, S., Toppare, L. E. V. E. N. T., Hepuzer, Y., & Yagci, Y. (2003). Immobilization
of invertase in conducting copolymers of 3-methylthienyl methacrylate. Bioelectrochemistry,
59(1–2), 29–33.
Costa, S. A., Azevedo, H. S., & Reis, R. L. (2005). Enzyme immobilization in biodegradable
polymers for biomedical applications. In R. L. Reis & J. S. Roma (Eds.), Biodegradable systems
in tissue engineering and regenerative medicine (pp. 301–323). CRC Press, LLC.
Crabb, W. D., & Shetty, J. K. (1999). Commodity scale production of sugars from starches. Current
Opinion in Microbiology, 2(3), 252–256.
Deere, J., Magner, E., Wall, J. G., & Hodnett, B. K. (2002). Mechanistic and structural features of
protein adsorption onto mesoporous silicates. The Journal of Physical Chemistry B, 106(29),
7340–7347.
Defaei, M., Taheri-Kafrani, A., Miroliaei, M., & Yaghmaei, P. (2018). Improvement of stability and
reusability of α-amylase immobilized on naringin functionalized magnetic nanoparticles: A
robust nanobiocatalyst. International Journal of Biological Macromolecules, 113, 354–360.
De Maio, A., El-Masry, M. M., Portaccio, M., Diano, N., Di Martino, S., Mattei, A., et al. (2003).
Influence of the spacer length on the activity of enzymes immobilised on nylon/polyGMA
membranes: Part 1. Isothermal conditions. Journal of Molecular Catalysis B: Enzymatic,
21(4–6), 239–252.
Dey, G., Nagpal, V., & Banerjee, R. (2002). Immobilization of α-amylase from Bacillus circulans
GRS 313 on coconut fiber. Applied Biochemistry and Biotechnology, 102(1–6), 303–313.
DiCosimo, R., McAuliffe, J., Poulose, A. J., & Bohlmann, G. (2013). Industrial use of immobilized
enzymes. Chemical Society Reviews, 42(15), 6437–6474.
Doshi, P., Srivastava, G., Pathak, G., & Dikshit, M. (2014). Physicochemical and thermal charac-
terization of nonedible oilseed residual waste as sustainable solid biofuel. Waste Management,
34(10), 1836–1846.
Emtiazi, G., Naghavi, N., & Bordbar, A. (2001). Biodegradation of lignocellulosic waste by
Aspergillus terreus. Biodegradation, 12(4), 257–261.
Fang, J. M., Fowler, P. A., Tomkinson, J., & Hill, C. A. S. (2002). The preparation and
characterisation of a series of chemically modified potato starches. Carbohydrate Polymers,
47(3), 245–252.
Fernández-Lorente, G., Palomo, J. M., Mateo, C., Munilla, R., Ortiz, C., Cabrera, Z., et al. (2006).
Glutaraldehyde cross-linking of lipases adsorbed on aminated supports in the presence of
detergents leads to improved performance. Biomacromolecules, 7(9), 2610–2615.
Ferreira-Dias, S., Sandoval, G., Plou, F., & Valero, F. (2013). The potential use of lipases in the
production of fatty acid derivatives for the food and nutraceutical industries. Electronic Journal
of Biotechnology, 16(3), 12–12.
Gaikwad, S. M., Rao, M. B., & Deshpande, V. V. (1992). d-Glucose/xylose isomerase from
Streptomyces: Differential roles of magnesium and cobalt ions. Enzyme and Microbial Tech-
nology, 14(4), 317–320.
Gardossi, L., Poulsen, P. B., Ballesteros, A., Hult, K., Švedas, V. K., Vasić-Rački, Đ., et al. (2010).
Guidelines for reporting of biocatalytic reactions. Trends in Biotechnology, 28(4), 171–180.
Garcia-Galan, C., Berenguer-Murcia, Á., Fernandez-Lafuente, R., & Rodrigues, R. C. (2011).
Potential of different enzyme immobilization strategies to improve enzyme performance.
Advanced Synthesis & Catalysis, 353(16), 2885–2904.
Gerard, M., Chaubey, A., & Malhotra, B. D. (2002). Application of conducting polymers to
biosensors. Biosensors and Bioelectronics, 17(5), 345–359.
Godfray, H. C. J., Beddington, J. R., Crute, I. R., Haddad, L., Lawrence, D., Muir, J. F., et al.
(2010). Food security: The challenge of feeding 9 billion people. Science, 327(5967), 812–818.
434 N. S. Naghavi et al.
Gonzalez-Saiz, J. M., & Pizarro, C. (2001). Polyacrylamide gels as support for enzyme immobili-
zation by entrapment. Effect of polyelectrolyte carrier, pH and temperature on enzyme action
and kinetics parameters. European Polymer Journal, 37(3), 435–444.
Herzog, G., Gorgy, K., Gulon, T., & Cosnier, S. (2005). Electrogeneration and characterization of
photoactivable films and their application for enzyme grafting. Electrochemistry
Communications, 7(8), 808–814.
Hirohara, H., Yamamoto, H., Kawano, E., & Nabeshima, S. (1981) Immobilized lactase, its
preparation and use. EP 0037667B1.
Homaei, A. A., Sariri, R., Vianello, F., & Stevanato, R. (2013). Enzyme immobilization: An update.
Journal of Chemical Biology, 6(4), 185–205.
Hong, Y. G., Moon, Y. M., Hong, J. W., Choi, T. R., Jung, H. R., Yang, S. Y., Jang, D.-W., Park,
Y.-R., Brigham, C. J., Kim, J.-S., Lee, Y.-K., & Lee, Y. K. (2019). Discarded egg yolk as an
alternate source of poly (3-hydroxybutyrate-co-3-hydroxyhexanoate). Journal of Microbiology
and Biotechnology, 29(3), 382–391.
Huang, W., Zhan, Y., Shi, X., Chen, J., Deng, H., & Du, Y. (2017). Controllable immobilization of
naringinase on electrospun cellulose acetate nanofibers and their application to juice debittering.
International Journal of Biological Macromolecules, 98, 630–636.
Hudson, S., Cooney, J., & Magner, E. (2008). Proteins in mesoporous silicates. Angewandte
Chemie International Edition, 47(45), 8582–8594.
Hwang, E. T., & Lee, S. (2019). Multienzymatic cascade reactions via enzyme complex by
immobilization. ACS Catalysis, 9(5), 4402–4425.
Ispas, C., Sokolov, I., & Andreescu, S. (2009). Enzyme-functionalized mesoporous silica for
bioanalytical applications. Analytical and Bioanalytical Chemistry, 393(2), 543–554.
Kahraman, M. V., Bayramoğlu, G., Kayaman-Apohan, N., & Güngör, A. (2007). α-Amylase
immobilization on functionalized glass beads by covalent attachment. Food Chemistry,
104(4), 1385–1392.
Katchalski-Katzir, E., & Kraemer, D. M. (2000). Eupergit® C, a carrier for immobilization of
enzymes of industrial potential. Journal of Molecular Catalysis B: Enzymatic, 10(1–3),
157–176.
Kazemzadeh, S., Naghavi, N. S., Emami-Karvani, Z., Fouladgar, M., & Emtiazi, G. (2020). Gas
chromatography-mass spectrometry analyses of crude oil bioremediation by the novel Klebsi-
ella variicola SKV2 immobilized in polyurethane polymer scaffold and two-layer
microcapsulation. Bioremediation Journal, 24(2–3), 129–149.
Keller, T. M., Michel, S. C., Fröhlich, J., Fink, D., Caduff, R., Marincek, B., & Kubik-Huch, R. A.
(2004). USPIO-enhanced MRI for preoperative staging of gynecological pelvic tumors: Prelim-
inary results. European Radiology, 14(6), 937–944.
Klein, M. P., Scheeren, C. W., Lorenzoni, A. S. G., Dupont, J., Frazzon, J., & Hertz, P. F. (2011).
Ionic liquid-cellulose film for enzyme immobilization. Process Biochemistry, 46(6),
1375–1379.
Klis, M., Karbarz, M., Stojek, Z., Rogalski, J., & Bilewicz, R. (2009). Thermoresponsive poly
(N-isopropylacrylamide) gel for immobilization of laccase on indium tin oxide electrodes. The
Journal of Physical Chemistry B, 113(17), 6062–6067.
Klouda, L., & Mikos, A. G. (2008). Thermoresponsive hydrogels in biomedical applications.
European Journal of Pharmaceutics and Biopharmaceutics, 68(1), 34–45.
Kluchova, K., Zboril, R., Tucek, J., Pecova, M., Zajoncova, L., Safarik, I., et al. (2009).
Superparamagnetic maghemite nanoparticles from solid-state synthesis–Their functionalization
towards peroral MRI contrast agent and magnetic carrier for trypsin immobilization.
Biomaterials, 30(15), 2855–2863.
Koh, W. G., & Pishko, M. (2005). Immobilization of multi-enzyme microreactors inside
microfluidic devices. Sensors and Actuators B: Chemical, 106(1), 335–342.
Koller, M., Hesse, P., & Braunegg, G. (2019). Application of whey retentate as complex nitrogen
source for growth of the polyhydroxyalkanoate producer Hydrogenophaga pseudoflava strain
DSM1023. The EuroBiotech Journal, 3(2), 78–89.
15 Enzyme Immobilization and Its Application Strategies in Food Products 435
Koller, M., Marsalek, L., & Braunegg, G. (2016). PHA Biopolyester production from surplus whey:
Microbiological and engineering aspects. Recent Advances in Biotechnology, 1, 100–172.
Koller, M., Shahzad, K., & Braunegg, G. (2018). Waste streams of the animal-processing industry
as feedstocks to produce polyhydroxyalkanoate biopolyesters. Applied Food Biotechnology,
5(4), 193–203.
Kooi, M. E., Cappendijk, V. C., Cleutjens, K. B. J. M., Kessels, A. G. H., Kitslaar, P. J. E. H. M.,
Borgers, M., et al. (2003). Accumulation of ultrasmall superparamagnetic particles of iron oxide
in human atherosclerotic plaques can be detected by in vivo magnetic resonance imaging.
Circulation, 107(19), 2453–2458.
Koszelewski, D., Müller, N., Schrittwieser, J. H., Faber, K., & Kroutil, W. (2010). Immobilization
of ω-transaminases by encapsulation in a sol–gel/celite matrix. Journal of Molecular
Catalysis B: Enzymatic, 63(1–2), 39–44.
Kouassi, G. K., Irudayaraj, J., & McCarty, G. (2005). Activity of glucose oxidase functionalized
onto magnetic nanoparticles. BioMagnetic Research and Technology, 3(1), 1.
Krause, M. H., Kwong, K. K., Gragoudas, E. S., & Young, L. H. (2004). MRI of blood volume with
superparamagnetic iron in choroidal melanoma treated with thermotherapy. Magnetic Reso-
nance Imaging, 22(6), 779–787.
Kumar, M. B., Gao, Y., Shen, W., & He, L. (2015). Valorisation of protein waste: An enzymatic
approach to make commodity chemicals. Frontiers of Chemical Science and Engineering, 9(3),
295–307.
Landarani-Isfahani, A., Taheri-Kafrani, A., Amini, M., Mirkhani, V., Moghadam, M., Soozanipour,
A., & Razmjou, A. (2015). Xylanase immobilized on novel multifunctional hyperbranched
polyglycerol-grafted magnetic nanoparticles: An efficient and robust biocatalyst. Langmuir,
31(33), 9219–9227.
Laskar, I. B., Rajkumari, K., Gupta, R., Chatterjee, S., Paul, B., & Rokhum, L. (2018). Waste snail
shell derived heterogeneous catalyst for biodiesel production by the transesterification of
soybean oil. RSC Advances, 8(36), 20131–20142.
Lei, S., Xu, Y., Fan, G., Xiao, M., & Pan, S. (2011). Immobilization of naringinase on mesoporous
molecular sieve MCM-41 and its application to debittering of white grapefruit. Applied Surface
Science, 257(9), 4096–4099.
Li, Z., Zhang, Y., Su, Y., Ouyang, P., Ge, J., & Liu, Z. (2014). Spatial co-localization of multi-
enzymes by inorganic nanocrystal–protein complexes. Chemical Communications, 50(83),
12465–12468.
Lim, B. C., Kim, H. J., & Oh, D. K. (2008). Tagatose production with pH control in a stirred tank
reactor containing immobilized L-arabinose isomerase from Thermotoga neapolitana. Applied
Biochemistry and Biotechnology, 149(3), 245–253.
Liu, R., Huang, W., Pan, S., Li, Y., Yu, L., & He, D. (2020). Covalent immobilization and
characterization of penicillin G acylase on magnetic Fe2O3/Fe3O4 heterostructure
nanoparticles prepared via a novel solution combustion and gel calcination process. Interna-
tional Journal of Biological Macromolecules, 162, 1587–1596.
Lozinsky, V. I., Simenel, I. A., Kulakova, V. K., Kurskaya, E. A., Babushkina, T. A., Klimova,
T. P., et al. (2003). Synthesis and studies of N-vinylcaprolactam/N-vinylimidazole copolymers
that exhibit the “proteinlike” behavior in aqueous media. Macromolecules, 36(19), 7308–7323.
Luo, X., & Zhang, L. (2010). Immobilization of penicillin G acylase in epoxy-activated magnetic
cellulose microspheres for improvement of biocatalytic stability and activities.
Biomacromolecules, 11(11), 2896–2903.
Marangoni, A. G., & Rousseau, D. (1998). Chemical and enzymatic modification of butterfat and
butterfat-canola oil blends. Food Research International, 31(8), 595–599.
Marguet, M., Bonduelle, C., & Lecommandoux, S. (2013). Multicompartmentalized polymeric
systems: Towards biomimetic cellular structure and function. Chemical Society Reviews, 42(2),
512–529.
436 N. S. Naghavi et al.
Puri, M., Seth, M., Marwaha, S. S., & Kothari, R. M. (2001). Debittering of kinnow mandarin juice
by covalently bound naringinase on hen egg white. Food Biotechnology, 15(1), 13–23.
Rana, M., Kumari, A., Chauhan, G. S., & Chauhan, K. (2014). Modified chitosan microspheres in
non-aggregated amylase immobilization. International Journal of Biological Macromolecules,
66, 46–51.
Rathner, R., Petz, S., Tasnádi, G., Koller, M., & Ribitsch, V. (2017). Monitoring the kinetics of
biocatalytic removal of the endocrine disrupting compound 17α-ethinylestradiol from differ-
ently polluted wastewater bodies. Journal of Environmental Chemical Engineering, 5(2),
1920–1926.
Ren, S., Li, C., Jiao, X., Jia, S., Jiang, Y., Bilal, M., & Cui, J. (2019). Recent progress in
multienzymes co-immobilization and multienzyme system applications. Chemical Engineering
Journal, 373, 1254–1278.
Reshmi, R., Sanjay, G., & Sugunan, S. (2006). Enhanced activity and stability of α-amylase
immobilized on alumina. Catalysis Communications, 7(7), 460–465.
Roy, I., & Gupta, M. N. (2003). Lactose hydrolysis by Lactozym™ immobilized on cellulose beads
in batch and fluidized bed modes. Process Biochemistry, 39(3), 325–332.
Sardar, M., Roy, I., & Gupta, M. N. (2000). Simultaneous purification and immobilization of
Aspergillus niger xylanase on the reversibly soluble polymer EudragitTM L-100. Enzyme and
Microbial Technology, 27(9), 672–679.
Schellenberger, E. A., Bogdanov Jr, A., Högemann, D., Tait, J., Weissleder, R., & Josephson,
L. (2002). Annexin V–CLIO: A nanoparticle for detecting apoptosis by MRI. Molecular
Imaging, 1(2), 15353500200202103.
Sheldon, R. A. (2007a). Cross-linked enzyme aggregates (CLEA® s): Stable and recyclable
biocatalysts. Biochemical Society Transactions, 35(6), 1583–1587.
Sheldon, R. A. (2007b). Enzyme immobilization: The quest for optimum performance. Advanced
Synthesis & Catalysis, 349(8–9), 1289–1307.
Sheldon, R. A., & Woodley, J. M. (2018). Role of biocatalysis in sustainable chemistry. Chemical
Reviews, 118(2), 801–838.
Silva, J. A., Macedo, G. P., Rodrigues, D. S., Giordano, R. L. C., & Gonçalves, L. R. B. (2012).
Immobilization of Candida antarctica lipase B by covalent attachment on chitosan-based
hydrogels using different support activation strategies. Biochemical Engineering Journal, 60,
16–24.
Rodrigues, R. C., Berenguer-Murcia, Á., & Fernandez-Lafuente, R. (2011). Coupling chemical
modification and immobilization to improve the catalytic performance of enzymes. Advanced
Synthesis & Catalysis, 353(13), 2216–2238.
Rodrigues, R. C., Ortiz, C., Berenguer-Murcia, Á., Torres, R., & Fernández-Lafuente, R. (2013).
Modifying enzyme activity and selectivity by immobilization. Chemical Society Reviews,
42(15), 6290–6307.
Tavano, O. L. (2013). Protein hydrolysis using proteases: An important tool for food biotechnology.
Journal of Molecular Catalysis B: Enzymatic, 90, 1–11.
Tipton, K. F., Armstrong, R. N., Bakker, B. M., Bairoch, A., Cornish-Bowden, A., Halling, P. J.,
et al. (2014). Standards for reporting enzyme data: The STRENDA consortium: What it aims to
do and why it should be helpful. Perspectives in Science, 1(1–6), 131–137.
Torres, R., Mateo, C., Fuentes, M., Palomo, J. M., Ortiz, C., Fernández-Lafuente, R., & Guisan,
J. M. (2002). Reversible immobilization of invertase on Sepabeads coated with
polyethyleneimine: Optimization of the biocatalyst’s stability. Biotechnology Progress, 18(6),
1221–1226.
Tosa, T., Sato, T., Mori, T., & Chibata, I. (1974). Basic studies for continuous production of
L-aspartic acid by immobilized Escherichia coli cells. Applied Microbiology, 27(5), 886–889.
Tosa, T., Sato, T., Mori, T., Matuo, Y., & Chibata, I. (1973). Continuous production of L-aspartic
acid by immobilized aspartase. Biotechnology and Bioengineering, 15(1), 69–84.
438 N. S. Naghavi et al.
Tripathi, A., Sami, H., Jain, S. R., Viloria-Cols, M., Zhuravleva, N., Nilsson, G., Jungvid, H., &
Kumar, A. (2010). Improved bio-catalytic conversion by novel immobilization process using
cryogel beads to increase solvent production. Enzyme and Microbial Technology, 47(1–2),
44–51.
Tsen, H. Y. (1984). Factors affecting the inactivation of naringinase immobilized on chitin during
debittering of fruit juice. Journal of Fermentation Technology, 62(3), 263–267.
Van de Velde, F., Lourenço, N. D., Pinheiro, H. M., & Bakker, M. (2002). Carrageenan: A food-
grade and biocompatible support for immobilisation techniques. Advanced Synthesis & Cataly-
sis, 344(8), 815–835.
Vescovi, V., Rojas, M. J., Baraldo, A., Botta, D. C., Santana, F. A. M., Costa, J. P., et al. (2016).
Lipase-catalyzed production of biodiesel by hydrolysis of waste cooking oil followed by
esterification of free fatty acids. Journal of the American Oil Chemists’ Society, 93(12),
1615–1624.
Vianello, F., Ragusa, S., Cambria, M. T., & Rigo, A. (2006). A high sensitivity amperometric
biosensor using laccase as biorecognition element. Biosensors and Bioelectronics, 21(11),
2155–2160.
Villeneuve, P. (2007). Lipases in lipophilization reactions. Biotechnology Advances, 25(6),
515–536.
Virtanen, J., Baron, C., & Tenhu, H. (2000). Grafting of poly (N-isopropylacrylamide) with poly
(ethylene oxide) under various reaction conditions. Macromolecules, 33(2), 336–341.
Virtanen, J., & Tenhu, H. (2000). Thermal properties of poly (N-isopropylacrylamide)-g-poly
(ethylene oxide) in aqueous solutions: Influence of the number and distribution of the grafts.
Macromolecules, 33(16), 5970–5975.
Walle, T. (2009). Methylation of dietary flavones increases their metabolic stability and
chemopreventive effects. International Journal of Molecular Sciences, 10(11), 5002–5019.
Xu, K., Chen, X., Zheng, R., & Zheng, Y. (2020). Immobilization of multi-enzymes on support
materials for efficient biocatalysis. Frontiers in Bioengineering and Biotechnology, 8.
Zhaoyu, Z., Ping, X., Keren, S., Weiwei, Z., Chunmiao, H., & Peng, L. (2020). Di-functional
magnetic nanoflowers: A highly efficient support for immobilizing penicillin G acylase. Journal
of the Chinese Chemical Society, 67, 1591–1601.
Zhi, Y., Wu, Q., & Xu, Y. (2017). Production of surfactin from waste distillers’ grains by co-culture
fermentation of two Bacillus amyloliquefaciens strains. Bioresource Technology, 235, 96–103.
Prospects and Challenges in Food-Grade
Enzymes Industrial Production 16
Musliu Olushola Sunmonu and Mayowa Saheed Sanusi
Abstract
Keywords
Industrial application · Food-grade enzymes · Food industry · Novel
technologies · Safety
# The Author(s), under exclusive license to Springer Nature Singapore Pte 439
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_16
440 M. O. Sunmonu and M. S. Sanusi
16.1 Introduction
The emergence of the food-grade enzyme for industrial production can be tagged as
green revolution approach due to its catalytic biological nature in transforming food
into various value-added products in different food industries, such as in beverages
and juices, baking, development of functional foods, meat, wine, dairy, fats and oils.
The food-grade enzyme has been projected as a potential replacement for chemical
additives or harsh physical conditions (temperature, pressure, pH) used in industrial
production, therefore guarantees sustainable production for safe and nutritious food
supply. Also, with an increase in the awareness on the need to consume a healthier
diet which has increased consumers’ preference for new and convenient food
products manufactured from natural food additives has further increased the use of
food-grade enzymes. Presently, food-grade enzymes are being used in the food
industry to increase diversity, quality and varieties of food produced. The food-
grade enzymes can be generated from the plants, animals and microbial sources.
However, out of the entire sources used for enzymes generation, microbial is the
most preferred choice in the food industry. Enzymes from microbial sources can be
produced in control conditions at a faster rate with minimal harmful by-products and
in larger quantities. Large-scale Industrial production has explored the use of
Saccharomyces cerevisiae, Aspergillus oryzae, Fungi Aspergillus niger and Bacillus
subtilis bacteria as sources for microbial enzymes production. Some of the commer-
cially important food-grade enzymes produced from this sources are proteases,
rennet, pectinases, invertases, cellulases, lipases, amylases, catalase, lactase,
raffinase, pullulanase, glucose oxidase, etc. Most of naturally occurring enzymes
from plants, animals or microbial sources are not often suitable for economical
production processes and sustainability. Therefore, there is a need to adopt novel
technological approaches that can be used to improve the industrial production of
food-grade enzymes. Uzuner and Cekmecelioglu (2019) reported that industrial
enzymes market is competitive with minimal profit margin and the enzymes are
currently manufactured by few companies in the United States, Denmark and
Germany with approximately twenty (20) enzymes being commercially produced
at present. Hydrolase represents 75% of food-grade enzymes in the market. Carbo-
hydrase (i.e. tannase, phytases, amylases, pectinases, cellulases, β-galactosidases),
lipase and proteases are family of hydrolase usually employed as food-grade
additives or processing aids in the food industry. This chapter provides an overview
of the prospect and challenges of a novel application of techniques for improving the
industrial production of food-grade enzymes through screening, enzyme immobili-
zation, solid-state fermentation, recombinant DNA technology, protein engineering,
and use of bioinformatics tool in enzyme engineering. The potential application of
some selected food-grade enzymes used for industrial production and the safety of
food-grade enzyme were discussed.
16 Prospects and Challenges in Food-Grade Enzymes Industrial Production 441
The use of two or more substrates has been reported as a way of increasing the yield
of the enzymes in solid-state fermentation (Aguilar et al., 2008).
According to the Food Drug Administration (FDA) in the USA, Escherichia coli
K-12 also known as bovine chymosin is the first recombinant enzyme. The recom-
binant deoxyribonucleic acid (rDNA) techniques or genetic engineering has now
been used to produce chymosin by transforming microorganisms with bovine
prochymosin gene. At least there are thirty-six (36) enzymes which are genetically
modified microorganisms (GMOs) among the one hundred and sixty (160) enzymes
produced for use in the food industry based on Association of Manufacturers and
Formulators of Enzyme Products (AMFEP) enzymes lists. To achieve the synthesis
of efficient enzymes that is free of undesirable enzymes or metabolites of microbes
using rDNA, appropriate selection of host microorganisms as recombinant strains
with potential DNA sequences that can code for the defined enzyme amino acid
sequence is needed. For instance, engineering microbial host stains were applied to
some fungi strain to manipulate or eliminate their capacity to release metabolites that
are toxic to the formulation of the synthesis enzyme (Olempska-Beer et al., 2006).
The application of rDNA has immensely increased the availability of food-grade
enzymes with distinct properties to meet the food process and food matrices require-
ment. Also, it has assisted in synthesizing enzymes that have desirable sensitivity
and specificity at a low cost of production.
Rastogi and Bhatia (2019) stated that the application of rDNA can be
implemented in batch or continuous operations using cheaper raw material, therefore
lessens the criteria for enzyme extraction and purification. The use of food-grade
enzymes from genetic engineering has minimized the cost connected with upstream
product processing and also increases productivity (Panesar, 2010; Rastogi &
Bhatia, 2019). ɑ-amylases, proteases, lipases and pullulanases used in food
applications, such as in hydrolysis of starch, are produced by using improve strains
(Ramos et al., 2011; Rastogi & Bhatia, 2019). Despite this entire prospect, the main
challenge of genetically improved strain is that is not yet certified safe for consump-
tion by the FDA and European Union due to the rise in public consciousness
regarding the non-consumption of genetic modified processed foods. Also, the
lack of standard in its production and purification procedures has caused a restriction
in its application food technology.
In order to analyse the information in enzymes data within a limited time either
online or off the network, the novel application of bioinformatics technique in
enzyme engineering is the recent trend useful in performing sophisticated
computations analysis (Alderson et al., 2012). The enormous of structural data and
sequencing coming out of nuclear magnetic resonance spectroscopy, X-ray crystal-
lography, mass spectrometry and NGS next-generation sequencing (NGS) for
enzyme informatics are now better understood by the application of bioinformatics
technique (Rastogi & Bhatia, 2019).
The use of food-grade enzymes for industrial production has widened their applica-
tion in the development of different food products. The application of some selected
food-grade enzymes for industrial production of food are as follows.
16 Prospects and Challenges in Food-Grade Enzymes Industrial Production 445
ɑ-amylase and β-amylase have been used over years for the industrial production of
maltose and glucose by applying the enzyme on starch. The application of ɑ-amylase
has paved way for the production of glucose syrup and has replaced the chemical
hydrolysis of starch in most starch manufacturing companies (Reddy et al., 2003).
The sources used for commercial production of ɑ-amylase are some selected strains
of bacteria (Bacillus spp.: B. amyloliquefaciens, B. licheniformis, B. subtilis,
B. cereus, B. stearothermophilus, B. licheniformis; halophilic bacteria:
Chromohalobacter sp., Halobacillus sp., Haloarcula hispanica, Halomonas
meridiana; fungi: Aspergillus species and few species of Penicillium:
P. brunneum, P. fellutanum) (Konsoula & Liakopoulou-Kyriakides, 2007; Erdal &
Taskin, 2010). ɑ-amylase had been used in the baking industry to play an important
role in enhancing the quantity, aroma and taste of baked products. Jegannathan and
Nielsen (2013) reported that the addition of amylase and lipase enzymes in bread
helps in extending its shelf life and reduces crystallization. Antistaling in bread and
improved softness can also be achieved by the addition of ɑ-amylase (Gupta et al.,
2008). However, more research still needs to be done on the application of
ɑ-amylase in the baking industry as a small overdose of ɑ-amylase can result in
sticky bread (Singh et al., 2019). For the fermentation of raw starch into ethanol, a
combination of ɑ-amylase produced from Streptococcus bovis and glucoamylase
from Rhizopus oryzae has been reported to improve the ethanol production (Singh et
al., 2019).
Protease enzymes can be the source from plant, animal, humans and microorganisms
(bacteria, fungi and virus) and are known to hydrolyse peptide bones of proteins.
Protease enzymes produced from microorganisms’ source are the most preferred for
industrial production due to ease of genetic manipulation and extensive diversity,
respectively. They are also known to favour low manufacturing cost, extensive
chemical and physical characteristics, lack of seasonal variation, large production
in industrial fermentators and quick culture development (Singh et al., 2019).
Protease enzymes of a wide variety have been exploited for industrial production
in food and pharmaceutical industries because of their specificity in action (Kalpana
et al., 2008). Almost 60% of the total food-grade enzymes in the market fall under
the category of protease enzymes (Mala et al., 1998). Alkaline and neutral proteases
produced for commercial purpose are usually generated from Bacillus genus. Pow-
dered bacteria proteases derived from Bacillus can be stable and remain 95% active
for a year. In the food industry, proteases can be applied in cheese production, baked
products making, soy hydrolysates production and meat tenderization. Proteases
from A. oryzae can improve bread colour and texture when mixed with flour dough.
More so it also aids in the reduction of mixing time. Heredia-Sandoval et al. (2016)
stated that the gluten content of bakery products can be decreased and degraded
446 M. O. Sunmonu and M. S. Sanusi
using proteases. In the dairy industry during cheese manufacturing, vegetable rennet,
animal rennet, genetically engineered chymosin and microbial milk coagulants are
milk coagulating enzymes used. Paracasein and micropeptides are generated from
the hydrolysis of peptide bone using protease enzymes in cheese making. However,
chymosin is good for generating casein. In beer production, application of protease
and amylase enzymes have been used as an efficient alternative way of achieving
malting process, thus saves energy and agricultural land that would have been used if
the conventional malting that involves drying was to be used (Jegannathan &
Nielsen, 2013). Microbial proteases have been reported to hydrolyse proteinaceous
in beer that causes haze formation, while production of wort can be achieved by the
use of B. subtilis proteases to solubilize proteins from barley adjuncts (Singh et al.,
2019). For commercial purpose, improved tenderness of meat which is the organo-
leptic quality of meat as desired by the consumers and for easy marketability can be
achieved by using thermophile protease derived from Bacillus strain, caldolysin
from Thermus strain, collagenase from Clostridium histolyticum, and aspartic prote-
ase from A. oryzae (Bekhit et al., 2014). For industrial production of aspartame
which is known as nutrasweet (zero-calorie sweetener), enzymatic synthesis of
aspartame has been preferred over chemical method due to their ability to maintain
stereospecificity. Proteases have been reported to be used to synthesize aspartame by
catalysing the reverse reaction in two amino acids, thus maintain stereospecificity
and reduce the cost of production (Singh et al., 2019).
Rennet is enzyme made up of chymosin, lipase and pepsin and it is usually referred
to as complex enzymes. Rennet can be generated from the plant, animal and
microbial sources. Rennet has been extensively applied in the commercial produc-
tion of different types of cheese. However, rennet from the microbial source is
mostly used for cheese production worldwide. For instance, R. miehei, Irpex lactis,
A. oryzae and R. pusillus are the good source of microorganism that can be used to
generate rennet for cheese production (Neelakantan et al., 1999; Singh et al., 2019).
Also, microbial rennet has served as a replacement for mammalian rennet used in
cheese production due to consumers’ demand of non-animal derivative. Unlimited
availability, low cost of production, bulk production and no risk of disease transmis-
sion are some of the advantages of microbial rennet over animal rennet (Singh et al.,
2019). The application of rennet enzyme is one of the most important uses of
enzymes in the food industry.
pumpkin (Eising et al., 1990; Kandukuri et al., 2012). Catalase found industrial
application in the removal of hydrogen peroxide used in sterilizing, oxidizing and
bleaching agents (Ertaş et al., 2000). The combination of catalase and glucose
oxidase has been used for the preservation of some selected foods. The enzyme
can also be used in egg processing but has limited usage in cheese production.
This enzyme is very important in the dairy industry, most especially in the produc-
tion of low-lactose or lactose-free food, such as flavoured milk drinks, ice cream,
yoghurt and frozen dessert. Lactase can be the source and isolate from animal
organs, plants, yeasts, bacteria or molds. For industrial production of lactase, it is
usually source from A. niger, A. oryzae and Kluyveromyces lactis due to their safety
of use history and the numerous safety tests they undergo before use. The source
used for generating the lactase and the method of preparation determines its
properties, such as the temperature and pH. For optimal performance of immobilized
lactases, the procedure of immobilization and the type of carrier are important.
Acidic pH of 2.5 to 4.5 is favourable for fungi lactase production, while neural
region pH is favourable for bacterial and yeast lactase with ranges of 6.5–7.5 and
6–7, respectively. The prospects in the utilization of lactase, hydrolase and
β-galactosidase have been extensive research due to the discovery of enzymes
immobilization techniques (Mehaia & Cheryan, 1987). The application of lactase
in milk has assisted in preventing diarrhea and severe tissue dehydration which affect
lactose intolerance people. It has also been reported to increase digestibility, sweet-
ness and scoop and creaminess of ice cream, yoghurt and frozen dessert. Increase in
sweetness and sugar addition level reduction has also been ascribed as another
advantage of lactase-treated milk in the production of flavoured milk drinks.
Cellulases are enzymes used for extracting constituents from soy protein, essential
oils, aromatic products, green tea and sweet potato in the food industry. Dillon et al.
(2004) described cellulase enzymes as enzymes that break the glucosidic bond of
cellulose microfibrils and release oligosaccharides, cellobiose and glucose.
Cellulases can be produced from filamentous fungi (Aspergillus, Chaetomium,
Fusarium, Penicillium, Phoma and Trichoderma), anaerobic bacteria (Acetovibrio,
Bacteroides, Butyrivibrio, Caldocellum, Clostridium, Eubacterium, Erwinia,
Pseudonocardia, Ruminococcus and Thermoanaerobacter) and aerobic bacteria
(Acidothermus, Bacillus, Cellvibrio, Pseudomonas, Streptomyces, Staphylococcus,
and Xanthomonas) (Soares et al., 2012; Singh et al., 2019). Among all the sources of
producing cellulolytic enzymes, Aspergillus has been reported to be the most
outstanding (Singh et al., 2019). Combination of cellulases, hemicellulase and
16 Prospects and Challenges in Food-Grade Enzymes Industrial Production 449
pectinase is currently being used in the food industry to extract and clarify fruits
juice. Cellulase can also be used to degrade solid phase, thus enhances liquefaction.
This enzyme has a great commercial significance in the production of inverted sugar
and is known for its sweetness effect. Invertase can catalyse sucrose to glucose and
fructose through hydrolysis. Temperature and pH within the range of 40–60 C and
3–5, respectively, are more favourable for more active invertase. At the industrial
level, invertase is mostly isolated from yeast, such as S. cerevisiae, and there is
ongoing research for the use of high yielding filamentous fungi for production of
invertase. Examples of the filamentous fungi are Rhizopus sp. And Aspergillus
casiellus. The filamentous fungi Rhizopus sp. was cultivated in the medium of
wheat bran to obtain invertase (Soares et al., 2012); Aspergillus casiellus was
inoculated in a medium of soybean meal and the invertase was isolated after 72 h,
while in yeast, invertase (S-b D-fructofuranosidase) is isolated from S. cerevisiae
(Singh et al., 2019).
In the food industry glucose isomerase has found extensive application in the
production of fructose-rich corn syrup. The enzyme is also known as d-xylose
ketol isomerase which aids in catalysing reversible isomerase from d-glucose and
d-xylose into d-xylulose and d-fructose, respectively. Protein engineering technique
is being explored to generate thermostable glucose isomerase enzymes (Hartley
et al., 2000). Glucose isomerase has also been used for bioconversion of hemicellu-
lose into ethanol (Singh et al., 2019). For commercialization of glucose isomerase,
the application of biotechnology in isolating mutants of promising prospect is
important.
Glucose oxidase is majorly used in the food industry to eliminate harmful oxygen in
food product containing probiotic microorganisms. The enzyme allows catalysing
oxidation–reductions reaction through the prosthetic group flavin adenine dinucleo-
tide (FAD). High level of glucose oxidase can be isolated from filamentous fungi,
such as Aspergillus versicolor and Rhizopus stolonifera (Guimarães et al., 2006).
Singh et al. (2019) suggested that the use of glucose oxidase through biotechnology
can be used to improve the stability of probiotic bacteria in yoghurt without adding
chemical additives.
450 M. O. Sunmonu and M. S. Sanusi
Industrial food-grade enzymes are generally considered safe and are known as food
additives, while some considered them as food processing aids. The safety regulation
on the use of food-grade industrial enzymes differs among countries, premarket
approval of the enzymes and the information provided by the manufacturer. The
prominent occupational food safety risks that have been associated with the use of
industrial food enzymes are mainly allergic reaction (in the respiratory tract), irrita-
tion risks, residual microbiological activity, chemical toxicity and oral toxicity
among consumers (Spök, 2006; Ramos et al., 2011; Singh et al., 2019). Ramos
et al. (2011) stated that enzymes are sensitizers and can cause allergic reaction in the
respiratory tract (asthma) upon inhalation and also irritation upon contact with the
skin. However, aside from the occupational risk, the final consumers of food
products containing food-grade enzymes rarely report allergic reaction because
low levels of the enzymes are usually used in food formulation which would have
eventually be deactivated upon processing and before reaching the consumers. The
future challenges that might be associated with the safety concern of food-grade
enzymes are toxic substance arising from the by-product of contaminants present in
the preparation of the enzyme. Adequate attention must be paid towards fermenta-
tion broth contaminate or environmental conditions of some phylogenetical strains
used in enzyme production. Although with the advent of genetic engineering
technique, toxicity problems can easily be eliminated. Food industry and national
or European regulatory bodies must develop strategies to harmonize and enforce
legislation on the available food enzymes that are categorized as a novel or those
with unusual properties and their food safety assessment practice to eradicate
occupational risks that may arise from their use. One of the current issues related
to food enzymes legislation is that in the USA, Japan and Canada food-grade
enzymes are considered as food additives, while Australia regards food enzymes
as food processing aids. WHO/FAO, joint committee of food additives, does not
differentiate food-grade enzymes either as a food processing aids or food additives
(Ramos et al., 2011). However, EU food legislation considered more than 160 food-
grade enzymes as food processing aids, while invertase and lysozyme enzymes are
the only two food-grade enzymes considered as food additives. National legislation
of France, Hungary, Denmark and Poland do subject food-grade enzymes that are
referred to as food processing aids to authorization before use, while in the UK the
authorization is voluntary (Ramos et al., 2011). Therefore, it is imperative for
legislation of each country worldwide to define the ranges of the permitted enzymes
and their applications.
16.10 Conclusions
For the future purpose, there is need to explore the benefits of novel approaches, such
as recombinant DNA technology, screening, protein engineering, enzyme immobi-
lization, solid-state fermentation and use of bioinformatics tool in enzyme
16 Prospects and Challenges in Food-Grade Enzymes Industrial Production 451
engineering in generating food-grade enzymes, and to adapt the enzymes for sus-
tainable and economical industrial production processes. Understanding the novel
approaches can aid the industrial production of food-grade enzymes which can be
useful in improving food product quality and consistency, preventing potential
harmful by-products in the food, reducing raw material dependence during
processing, replacing chemical additives, reducing process time, increasing shelf
life and creating more opportunities for enhancing the aromatic, structural and
textural properties of food. In addition, application of food-grade enzymes for
industrial processes should be able to lead to the development of new or improve
healthy food products and food properties within the food processing industry;
bakery, dairy, beverages, juice, alcoholic, meat, fats and oils, vegetables and other
industries. Proper selection of enzymes source, enzymes isolating method and
purification technique that is safe and non-toxic must be carefully selected as they
influence the cost of production of food-grade enzymes and yield. Solid-state
fermentation produces higher enzymes than submerged fermentation at a lower
cost. National legislation of countries should make provision for permitting and
regulating products from food-grade enzymes and their applications. More research
should be channel towards the application of enzymes technologies in food
processing to broaden our knowledge and future use because consumers are now
demanding for food products or additives that are free of chemicals.
References
Alderson, R. G., Ferrari, L. D., Mavridis, L., McDonagh, J. L., Mitchell, J. B. O., & Nath,
N. (2012). Enzyme informatics. Current Topics in Medicinal Chemistry, 12(17), 1911–1923.
Aguilar, C. N., Gutiérrez-Sánchez, G., & Rado-Barragán, P. A. (2008). Perspectives of solid state
fermentation for production of food enzymes. American Journal of Biochemistry and Biotech-
nology, 4, 354–366.
Aravindan, R., Anbumathi, P., & Viruthagiri, T. (2007). Lipase applications in food industry.
Indian Journal of Biotechnology (IJBT), 6(02), 141–158.
Bekhit, A. A., Hopkins, D. L., Geesink, G., Bekhit, A., & Franks, P. (2014). Exogenous proteases
for meat tenderization. Critical Reviews in Food Science and Nutrition, 54(8), 1012–1031.
Chelikani, P., Fita, I., & Loewen, P. C. (2004). Diversity of structures and properties among
catalases. Cellular and Molecular Life Sciences, 61(2), 192–208.
Dillon, A., Said, S., & Pietro, R. C. L. (2004). Celulases. In L. Summa (Ed.), Enzimas como agentes
biotecnológicos (pp. 243–270). Legis Summa.
Dipasquale, L., Gambacorta, A., Siciliano, R. A., Mazzeo, M. F., & Lama, L. (2009). Purification
and biochemical characterization of a native invertase from the hydrogen-producing
Thermotoga neapolitana (DSM 4359). Extremophiles, 13(2), 345–354.
Eising, R., Trelease, R. N., & Ni, W. (1990). Biogenesis of catalase in glyoxysomes and leaf-type
peroxisomes of sunflower cotyledons. Archives of Biochemistry and Biophysics, 278(1),
258–264.
Erdal, S., & Taskin, M. (2010). Production of alpha-amylase by Penicillium expansum MT-1 in
solid-state fermentation using waste Loquat (Eriobotrya japonica Lindley) kernels as substrate.
Romanian Biotechnological Letters, 15(3), 5342–5350.
Ertaş, N., Timur, S., Akyılmaz, E., & Dinfikaya, E. (2000). Specific determination of hydrogen
peroxide with a catalase biosensor based on mercury thin film electrodes. Turkish Journal of
Chemistry, 24(1), 95–99.
452 M. O. Sunmonu and M. S. Sanusi
Guimarães, L. H. S., Peixoto-Nogueira, S. C., Michelin, M., Rizzatti, A. C. S., Sandrim, V. C., &
Zanoelo, F. F. (2006). Screening of filamentous fungi for production of enzymes of biotechno-
logical interest. Brazilian Journal of Microbiology, 37, 474–480.
Gulati, K., & Poluri, K. M. (2016). Mechanistic and therapeutic overview of glycosaminoglycans:
The unsung heroes of biomolecular signaling. Glycoconjugate Journal, 33(1), 1–17.
Gupta, A., Gupta, V. K., Modi, D. R., & Yadava, L. P. (2008). Production and characterization of
a-amylase from Aspergillus niger. Biotechnology, 7(3), 551–556.
Gupta, R., Gigras, P., Mohapatra, H., Goswami, V. K., & Chauhan, B. (2003). Microbial α-
amylases: A biotechnological perspective. Process Biochemistry, 38(11), 1599–1616.
Hartley, B. S., Hanlon, N., Robin, J., Rangarajan, J., & Ragaranjan, M. (2000). Glucose isomerase:
Insights into protein engineering for increased thermostability. Biochimica et Biophysica Acta
(BBA) - Protein Structure and Molecular Enzymology, 1543(2), 294–335.
Hasan, F., Shah, A. A., & Hameed, A. (2006). Industrial applications of microbial lipases. Enzyme
and Microbial Technology, 39, 235–251.
Heredia-Sandoval, N. G., Valencia-Tapia, M. Y., Calderón de la Barca, A. M., & Islas-Rubio, A. R.
(2016). Microbial proteases in baked goods: Modification of gluten and effects on immunoge-
nicity and product quality. Food, 5(3), 59.
Jackel, C., Kast, P., & Hilvert, D. (2008). Protein design by directed evolution. Annual Review of
Biophysics, 37, 153–173.
Jegannathan, K. R., & Nielsen, P. H. (2013). Environmental assessment of enzyme use in industrial
production—A literature review. Journal of Cleaner Production, 42, 228–240.
Kalpana, D. M., Banu, R. A., Gnanaprabhal, G. R., Pradeep, B. V., & Palaniswamy, M. (2008).
Purification, characterization of alkaline protease enzyme from native isolate Aspergillus niger
and its compatibility with commercial detergents. Indian Journal of Science and Technology, 1,
1–6.
Kandukuri, S. S., Noor, A., Ranjini, S. S., & Vijayalakshm, M. A. (2012). Purification and
characterization of catalase from sprouted black gram (Vignamungo) seeds. Journal of
Chromatography B, 889-890, 50–54.
Konsoula, Z., & Liakopoulou-Kyriakides, M. (2007). Co-production of alpha-amylase and beta-
galactosidase by Bacillus subtilis in complex organic substrates. Bioresource Technology,
98(1), 150–157.
Lin, L.-L., Liu, J.-S., Wang, W.-C., Chen, S.-H., Huang, C.-C., & Lo, H.-F. (2008). Glutamic acid
219 is critical for the thermostability of a truncated α-amylase from alkaliphilic and thermophilic
Bacillus sp. strain TS-23. World Journal of Microbiology and Biotechnology, 24(5), 619–626.
Mao, X., Hong, Y., Shao, Z., Zhao, Y., & Liu, Z. (2010). A novel cold-active and alkali-stable beta-
glucosidase gene isolated from the marine bacterium Martelella mediterranea. Applied Bio-
chemistry and Biotechnology, 162(8), 2136–2148.
Mala, R. B., Tanksale, A. M., Ghatge, M. S., & Deshpande, V. V. (1998). Molecular and
biotechnological aspects of microbial proteases. Microbiology and Molecular Biology Reviews,
62(3), 597–635.
Mehaia, M. A., & Cheryan, M. (1987). Production of lactic acid from sweet whey permeates
concentrates. Process Biochemistry, 22(6), 185–188.
Nakagawa, T., Nagaoka, T., Taniguchi, S., Miyaji, T., & Tomizuka, N. (2004). Isolation and
characterization of psychrophilic yeasts producing cold-adapted pectinolytic enzymes. Letters
in Applied Microbiology, 38(5), 383–387.
Nakagawa, T., Ikehata, R., Uchino, M., Miyaji, T., Takano, K., & Tomizuka, N. (2006). Cold-
active acid β-galactosidase activity of isolated psychrophilic-basidiomycetous yeast
Guehomyces pullulans. Microbiological Research, 161(1), 75–79.
Neelakantan, S., Mohanty, A. K., & Kaushik, J. K. (1999). Production and use of microbial
enzymes for dairy processing. Current Science, 77(1), 143–148.
Olempska-Beer, Z. S., Merker, R. I., Ditto, M. D., & DiNovi, M. J. (2006). Food-processing
enzymes from recombinant microorganisms—A review. Regulatory Toxicology and Pharma-
cology, 45(2), 144–158.
16 Prospects and Challenges in Food-Grade Enzymes Industrial Production 453
Abstract
Public concerns about food safety and its health, on the one hand, and the rapid
advancement in emerging technologies and the green ones, on the other hand,
have led to less interest in chemical interactions occurring in the food industry.
Enzymes are proteins and consequently sensitive to environmental effects such as
pH and temperature. Additionally, they are less likely to be reused and the cost of
their recovery is exorbitant. Therefore, their application as natural catalysts has
been limited. Having tackled the problems associated with free enzymes, enzyme
immobilization made it possible to reuse and store enzymes. Then, nanotechnol-
ogy implemented this technique to end in nanobiocatalysis which merges nano-
technology and biotechnology and stabilizes enzymes accordingly. Nano carriers
improve enzymes’ activity and efficiency, stability, and storage ability as they
possess unique features such as nanoscale size, high surface/volume ratio, and
variety in composition and structure. Nanobiosensors are another breakthrough in
bio and nanotechnologies which can substitute chemical and electrochemical
Z. B. Mohammadi (*)
Department of Food Science and Technology, North Tehran Branch, Islamic Azad University,
Tehran, Iran
e-mail: z.beigmohammadi@iau-tnb.ac.ir
K. Khoshtinat
Institute of Nutrition and Food Technology Research Institute, Nutrition and Food Technology
Faculty, Shahid Beheshti Medical Science of University, Tehran, Iran
S. Ghasemi
Faculty of Food Science and Technology, Isfahan University of Technology, Isfahan, Iran
Z. Ahmadi
Food Hydrocolloids Research Center, Department of Food Science and Technology, Ferdowsi
University of Mashhad (FUM), Mashhad, Iran
# The Author(s), under exclusive license to Springer Nature Singapore Pte 455
Ltd. 2022
A. Dutt Tripathi et al. (eds.), Novel Food Grade Enzymes,
https://doi.org/10.1007/978-981-19-1288-7_17
456 Z. B. Mohammadi et al.
sensors for analysis, quality control, and food safety measurement. Recently,
nanomaterials-based biosensors have acted as the focal point in food contamina-
tion detection, namely pesticides, antibiotics, and heavy metals, due to their
variety, highly specialized activity, and high precision. In this chapter, we
investigate nanotechnology and food enzymes, probable risks, and legal aspects
of nanomaterial application in food bioprocess.
Keywords
Nanotechnology · Enzyme · Nanobiocatalysis · Nanobiosensor · Food analysis ·
Food safety
17.1 Introduction
The prefix “nano” comes from the Greek word for dwarf which means “one-
billionth” and the nanometer is equal to one-billionth of a meter that is extremely
small to see even by a conventional light microscope (Ravichandran, 2010;
Sundarraj, 2019). The late Norio Taniguchi initially applied the prefix “nanotech-
nology” in 1974 and Richard Feynman first gave its concepts in 1959 (Handford
et al., 2014; He & Hwang, 2016).
Nanomaterials are tiny particles at least ranging from 1 to 100 nm in size in one
dimension, bio-persistent or insoluble in nature, synthesized through numerous
ways. Nanotechnology is a branch of science that manipulates the materials at
nanoscale which are synthesized in various routes, and are used in different fields
including electronics, medicine, agriculture, and food industries (He & Hwang,
2016; Ghasemi et al., 2017).
This reduction in size results in highly impressive features and functions.
Nanomaterials are able to be applied in different forms such as nanotubes, nano
rods, nanoparticles, nano sheets, and nanofibers in various technologies and
industries of food, electronics, catalysts, energy, and agriculture (Jafarizadeh-
Malmiri et al., 2019; Handford et al., 2014). Comparing nanomaterials and materials
in bulk form, it can be concluded that changing the particle sizes of materials into
nanoscale increases the ratio of surface area to volume, leading to unique physical,
mechanical, chemical, optical, and biological properties such as solubility, absorp-
tion of the cells, delivery properties, and the residence time in the cells of body. On
the other hand, it affects the bioavailability, safety, nutritional value, and efficiency
properties (McClements & Xiao, 2017). This factor plays a critical role in creating
unique features, so engineered nanomaterials have raised more attention in the
industries of agro-food, medicine, sewage water treatments, and others (Haroon &
Ghazanfar, 2016).
Recently, the industries of food and agricultural have had significant investments
in nanotechnology which enhance the quality of product and reduce total prices
(Yu et al., 2018a). Firstly, Pasteur introduced nanotechnology by using it in pasteur-
ization process in food industries to omit the bacteria of spoilage (1000 nm) which
17 Nanotechnology and Food Grade Enzymes 457
made the first step to transform the food processing and improve food quality.
Watson and Crick first built a model for the structure of DNA at almost 2.5 nm
which enabled the applications in biomedicine, biotechnology, production pro-
cesses, and agriculture (Hansen et al., 2013). Nanotechnology has immense potential
in the postharvest food processing. The main applications of nanotechnology in the
food and agriculture structures are food processing and security improvements,
production, packaging of food which enhances its functionality, food bioavailability,
nutrition and flavor, pathogen detection, protection of the environment, nutrients
absorption of plants, and the cost-effectiveness of storage and distribution. It also
modifies the particle size, size distribution, surface charge, possible cluster forma-
tion, and delivery methods (Rashidi & Khosravi-Darani, 2011; Ghasemi et al.,
2018). Study in nanotechnology has high potential to advantage humanity in food-
related industries (Fig. 17.1).
Generally the potential of applying nanotechnology in food industries has been in
nanocarriers (such as nanoparticles, nanoemulsion, nanocomposites,
nanobiocomposites, and nanolaminates), nanosensors and nanobiosensors (for the
control of safety and quality of the food), processing (such as nanofiltration, nano-
scale enzymatic reactor, heat and mass transfer, nanofabrication and nanocapsules
for modification of absorption), delivery, packaging, formulation, DNA recombinant
technology, evaluation, etc. (Ghasemi et al., 2018; Jafarizadeh-Malmiri et al., 2019),
but the main nanotechnology applications in food industries are food nanostructured
components and food nano sensing to increase the safety and quality of food.
Improvements in the production of inorganic nanoparticle have permitted the prepa-
ration of effective nanosensors to identify pathogens or pesticides rapidly in food
(Neethirajan & Jayas, 2011; Nile et al., 2020).
Our main goal in this chapter was to provide a brief overview of the use of
nanotechnology and nanobiotechnology, namely enzymes, in/on nanocarriers on the
food industry improvements especially food packaging. Also, the potential
applications and future perspectives of nanobiotechnology on food safety were
discussed. Also this chapter overviews the information usually accessible about
nanomaterials’ risk assessments, marketing concerns, safety and toxicity aspects,
and regulatory aspects.
Request for extra innovative improvements and technologies in valuable tools and
materials for the life study are supported by the technologies of nanotechnology and
biotechnology which are among the promising technologies in the twenty-first
century (McClements & Xiao, 2017). Nanotechnology is a technology that uses
materials and devices possessing at least one dimension sized in nanoscale, which
plays an essential role in the food and agriculture parts, contributes to improve the
crop, progresses the food quality and safety, and supports the health of humans
through innovative and novel approaches (Jafarizadeh-Malmiri et al., 2019).
458
NanoTechnology
Animals Plants Nutrition & Feed Processing Equipment Smart Packaging Active Packaging
Nutrient delivery
accessible about nanomaterials’ risk assessments, marketing concerns, safety and toxicity aspects, and
regulatory aspects.
Biotechnology
Silver oxide Apple slice Delays microbial spoilage Zhou et al. (2011)
Edible Silica in situ improved Cherries Significant reduction in the Yu et al. (2018b)
nanocomposite the biodegradable films permeability of moisture and
film of PVA/chitosan (CS) oxygen by 10.2% and 25.6%,
respectively, and extending the
preservation time of cherries
Edible film with chitosan Ready-to-eat (RTE) roast Chitosan exhibited antimicrobial Beverlya et al. (2008)
beef activity against Listeria
monocytogenes
Nanosensors Cadmium telluride 2,4-D (herbicide) The pesticide of Vinayaka et al. (2009)
quantum dot nanoparticle 2,4dichlorophenoxyacetic acid was
(CdTe QD) detected
Hetero-structured silicon/ Suspension of salmonella Au/Si nanorods based biosensor Park et al. (2007)
gold nanorod bacterial in 1% BSA could detect Salmonella
solution
461
462 Z. B. Mohammadi et al.
• High prices
• The need of various enzymes
• Low stability of the enzyme
• High residual enzyme remained in the product and difficulties for their recovery
and reuse
• Incompatibility of the continuous process
• Low activity
• Higher labor costs
The protein and sensitive nature of enzymes justifies the need to select appropriate
immobilization methods to maintain their structure and activity. Significant
advances in the recognition of enzymes and their immobilization have led to the
use of NMs in various structures and compounds as a support to immobilize
enzymes. Enzyme carriers, in general, should have a large surface area, functional
groups, as well as biodegradability. NMs have a much higher surface-to-volume
ratio than conventional carriers and can provide more and better enzymatic immobi-
lization (Jia et al., 2014; Saallah et al., 2016).
Enzyme immobilization methods are generally divided into three main
categories: entrapment or encapsulation, binding to a carrier or matrix, and cross-
linking. Immobilization may be reversible or irreversible, involving direct adsorp-
tion to the surface. Different bonds on the surface of enzymes lead to various
interactions, such as hydrophobic, electrostatic, and van der Waals forces and
hydrogen bonds. Types of irreversible immobilization include immobilizing the
enzyme by covalent bonding to the carrier surface, entrapment in the matrix, or
464 Z. B. Mohammadi et al.
encapsulating the enzyme. In this case, it prevents the enzyme from being washed
during the reaction, but these supports cannot be reused after finishing the work and
reducing the activity of the enzyme and should be discarded. In contrast, reversible
immobilization allows recovery of enzymes after inactivation of the enzyme, but
some of the enzyme may be lost during washing. Cross-linking agents (such as
glutaraldehyde) are commonly used in cross-linking method. These agents have very
low molecular weight relative to the enzyme, so in this method the enzyme remains
almost unchanged and is also called a carrier-free biocatalyst with 100% activity. It
is undeniable that in order to achieve optimal immobilization, a proper understand-
ing of support and enzyme properties is required. On the one hand, changes in the
three-dimensional structure of enzymes and the amphiphilic nature of proteins
(hydrophobic inner part and hydrophilic outer part), the functional groups of
supports, and the type of bonds they can form should be carefully considered
(Netto et al., 2013).
Advanced developments in nanotechnology have created different modes for
enzyme immobility in different nanomaterials (Husain, 2010). The combination of
enzymes with nanomaterials as nanobiocatalysts (NBs) provides a better option than
a variety of chemical catalysts that can be referred to green technology. Enzyme
immobilization has been used by various nanostructures such as nanofibers,
nanotubes, and nanoparticles. These supports are preferred because of their large
specific surface areas (great surface area-to-volume ratios) which can provide
extreme immobilization enzymes (Agustian et al., 2016; Beig Mohammadi et al.,
2016). However, some nanostructured materials also have their own disadvantages.
For example, mesoporous silica typically binds enzyme molecules to its inner
surface, which restricts the diffusion of the substrate to the enzyme and reduces
activity. Nanoparticles and nanotubes also dramatically reduce mass transfer
limitations, while their dispersing and recycling is more challenging. Conversely,
electrospun nanofibers have a high ability to solve these problems and can be
favorable supports for enzyme immobilization (Beig Mohammadi et al., 2016).
17.3.1.1 Nanofibers
Nanofiber membranes with special properties such as continuous, porous, and nano-
open structure allow easy access of the substrate to the active sites of enzyme
molecules. Therefore, compared to other nanostructures, substrate can more easily
diffuse in this continuous porous space and perform the necessary reactions. In
general, nanocarriers for enzyme immobilization can be solid or liquid, spherical or
non-spherical, fibers, tubes, and network characteristics (Wen et al., 2017). During
the last few years, materials based on one-dimensional nanostructures (nanofibers
and nanotubes) have created a subject of substantial interest due to their unique
properties. Nanoelectrospun fibers, for instance, have achieved more attention com-
pared to other thicker fibers made of the equal material, and their applications in
different industries are relatively new. Immobilization of enzymes and encapsulation
of nutraceutical compounds or probiotics in/on nanofibers for maintaining the
quality and effectivity of foods have gained due to the specific properties of
nanofibers. In addition, nanofibers produced by electrospinning method have many
17 Nanotechnology and Food Grade Enzymes 465
Enzymes are actually biocatalysts that have many applications in the commercial
and industrial regions of food processing, bioreactors, and biosensors. Commercial
and recyclable use of biocatalysts depends on the degree of efficiency of the
immobilization technique and the residual activity of the enzymes. Methods of
enzyme immobilization are divided into different categories: carrier-free.
466 Z. B. Mohammadi et al.
Enzyme immobilization on these nanofibers can enhance and retain their activity
and binding of enzymes to the nanofibers as well as enzyme separation for recycling.
Enzyme encapsulation in nanofibers can also be used as an efficient method to
preserve the third structure and activity of the enzyme. The main limitation for
immobilized enzymes on nanoelectrospun fibers is the nature of a liquid system for
dissolving enzymes and water-insoluble polymers that can remain resistant in a
variety of environments and are easily separated. Fiber materials are generally the
most desirable supports due to their inherent properties such as good mechanical
strength, high specific surface area, and inter-fiber porosity (Gabrielczyk et al., 2018;
Rojas-Mercado et al., 2018).
Enzymes can be immobilized after electrospinning by covalent or noncovalent
bonding on the fibers, or encapsulated coelectrospinning methods within polymer
fibers. Encapsulation of ficin and papain enzymes in nanofiber polyvinyl alcohol
(PVA) electrospun and glutaraldehyde (GA) as cross-linking agents were studied.
The results showed phycin and papain remained stable 92% (after nine consecutive
use periods) and 40% (after four consecutive use periods) of their initial activity,
respectively (Rojas-Mercado et al., 2018).
Phospholipase A1 was successfully immobilized on modified surface
(MS) chitosan:polyethylene oxide (CS:PEO; 90:10) nanofibrous mat (NFM) with
atmospheric plasma (AP) at different times (2, 6 and 10 min). Scanning electron
microscopy (SEM) images showed that the membrane structures maintained the
nanofiber porous structures and uniformity before and after AP treatments. PLA1
was successfully immobilized by covalent bonding with functional groups of
chitosan NFM. The results showed 80% of initial activity of immobilized PLA1
on APSM CS: PEO NFM was still maintained after ten consecutive use periods
(Beig Mohammadi et al., 2016).
17 Nanotechnology and Food Grade Enzymes 467
17.3.1.2 Nanoparticles
Nanoparticles are generally unique and discrete particles with three dimensions with
a size of 100 nm or less and have very different properties from bulk materials.
Magnetic nanoparticles are composed of various elements, especially iron, nickel,
cobalt, and their various chemical compounds. In the food industry, due to the
existing safety limitations, nontoxicity of good biocompatibility, nanoparticles com-
monly used include iron oxides such as Fe3O4 magnetic nanoparticles.
Like other nanoscale materials, they perform better than bulk magnetic materials
in sizes of 10–20 nm. Magnetic nanoparticles are very efficient as they can be easily
separated and recovered using a magnetic field during the process. Since surface
energies play an important role, surface interactions between magnetic particles and
proteins must be carefully considered. Bates noted that the adsorption capacity of
proteins is highly correlated with the activation of chemical bonds on the particle and
the protein surface. The absorption capacity of proteins is highly related to the
activation of chemical bonds on the particle and the surface of the protein.
Nanoporous materials have a lower enzyme load as support but no restriction on
substrate diffusion. In contrast, porous materials have a lower restriction of substrate
diffusion while showing a high enzymatic load (Netto et al., 2013). Immobilization
of α-amylase on carboxylated magnetic nanoparticles, lipase on magnetic Fe3O4
chitosan to synthesize ascorbyl palmitate, invertase on silica-modified magnetic
nanoparticles in order to hydrolyze the potato starch to glucose and fructose are
successful examples of the application of nanoparticle-immobilized enzymes in the
food industry (Cao et al., 2016; Wang et al., 2015; Bayramoglu et al., 2017).
17.3.1.3 Nanosheets
The carbon family is one of the most widely used materials to immobilize enzymes.
For this reason, different methods of producing carbon-derived nanomaterials have
been studied. Among these materials, graphene is used in the immobilization of
enzymes due to its remarkable optical, thermal, electronic, and mechanical
properties. It is composed of carbon atoms with a dense two-dimensional structure
similar to a honeycomb network. Graphene, in addition to successful drug and gene
delivery, has been able to immobilize enzymes due to maintaining the natural protein
structure and activity of enzymes (Singh et al., 2014).
Immobilization of Laccase obtained from Aspergillus oryzae (for ethanol produc-
tion, clarification of wine and tea) and α-galactosidase (for hydrolysis of raffinose
oligosaccharides (RFOs) cause of flatulence in soybeans) on graphene nanosheets
are significant examples of the application of nanosheets in food industries (Singh
et al., 2014; Skoronski et al., 2017).
17.3.1.4 Nanotubes
In recent years, carbon nanotubes (CNTs) have been considered for enzyme immo-
bilization due to their special surface properties along with their desirable structural
and mechanical properties as well as their very specific surface. The main types of
CNT, including single-walled (SWCNTs) and multiwalled (MWCNTs), are used to
immobilize enzymes which provide higher enzyme loading and lower mass transfer
468 Z. B. Mohammadi et al.
• Palladium, platinum, and gold based nanosensors to detect any sort of color
changes in food, any gases produced by spoilage, any changes in light, heat,
472 Z. B. Mohammadi et al.
humidity, and chemicals into electrical signals, and toxins such as aflatoxin
in milk.
• Single-walled carbon nanotubes and DNA to monitor soil condition needed to
crop growth and detect pesticides on fruit and vegetable surface.
• Carbon black and polyaniline to detect carcinogens and pathogens in food.
• Array biosensors, electronic noses, nano-test strips, and nanocantilevers to moni-
tor color changes resulting from food spoilage.
• Nano-smart dust in order to detect any type of environmental pollution.
• Nanobarcodes to detect agricultural products quality.
• Nanobiosensors in order to detect viruses and bacteria.
• Biomimetic sensors (protein and biomimetic membranes) and smart biosensors to
determine mycotoxins and some toxins, act as pseudo cell surfaces to assist
pathogens detection and removal.
• Surface plasmon-coupled emission biosensors (containing gold) to detect
pathogens.
• Cerium oxide immunosensors and chitosan-based nanocomposites to detect some
toxins like ochratoxin A.
• Carbon nanotubes and silicon nanowire transistors to detect staphylococcal
enterotoxin B and cholera toxin.
• iSTrip of time-temperature indicator/integrator to detect the spoilage of food
based on the history of temperature.
• Abuse indicators to detect desired temperature has been achieved or not.
• Partial temperature history indicator, when the temperature exceeds a certain
pre-determined value.
• Full-temperature history indicator to register continuous temperature changes
considering time and detect frozen food temperature changes.
• Reflective interferometry in order to detect E. coli contamination in packaged
foods (Pradhan et al., 2015).
To keep food quality and safety during storage and transport, food packaging
plays a key role, by extending the shelf life. Therefore, the type of food packaging
materials is very important, as well as being biodegradable and using natural
polymer instead of petroleum-based plastics. Some biodegradable polymers used
in food packaging are:
Measurable signals
Electrical signals
Biosensors
Bioreceptor Transducer
Electrochemical, Potentiometric,
Molecular Imprinted Polymer Conductometric, Impedimetric
have their own importance. E. coli and Salmonella are examples which have been
detected by amperometric and electrochemical DNA biosensors. Classifications and
methods of biosensors can be seen in Fig. 17.5.
Transducers, the main part of biosensors, cause physical responses, which may
include heat absorption (calorimetric biosensor); variation of charge distribution
(potentiometric biosensor); motion of electrons made by a redox reaction (ampero-
metric biosensor); or total effects of reactants (piezo-electric biosensor).
478 Z. B. Mohammadi et al.
biochemical signal into an electrical signal), and finally an electronic system includ-
ing amplifier, processor, or recorders. The biosensors could be classified into
electrochemical, potentiometric, calorimetric, amperometric, resonant,
ion-sensitive, and optical. Nanobiosensors are included to optical, bio, chemical,
physical, and sol-gel types. Nanosensors matrix would protect food from effects of
enzyme or dye toxicity. Of course, their usage in food industries depends on the
detection type. For example, chemical or bio detection for optical nanosensors, in
which their color changes by reaction with chemical or biological reagents. In
biological nanosensors, antibody/antigen; DNA; enzyme interactions would happen.
While, chemical composition or molecular concentration may occur in chemical
type, or pressure, force, mass, and displacement in physical ones.
Knowing the mechanism of nanobiosensors is very important, for example, by
spraying a nano bioluminescent on a food, visual glow could indicate microbial
contamination, or released nano particles, such as nano-silver used in active packag-
ing will react with microorganisms. In amperometric nanosensors, organophospho-
rus pesticides could be detected by mechanism of dual enzyme electrode system.
Technology of biosensors effects on many sections, including pharmacology,
medical care, and food industry, in which safety has an important role, as well as
easy, quick, and precise control of nutrients and pollutants.
In fact, enzymatic biosensors are electrochemical ones for food analysis, in which
substrates would be detected either from their transformed products obtaining by
enzymatic catalysis or by the detection of enzyme inhibitors.
To detect xanthine in fish, immobilized xanthine oxidase (MWCNTs) on
multiwalled carbon nanotubes and a copolymer to act as a biosensor were used. It
was shown that MWCNTs could have increased sensitivity more than biosensor
when was applied to immobilize alone. By immobilization of glutamate dehydroge-
nase in MWCNTs, detection limit of monosodium glutamate was improved.
Nanotechnology has an important role in the agri-food industries for very dis-
tinctive properties of nanomaterials. Recently, they are used to detect food
adulterants, pathogens, and toxins. Enzymatic nanobiosensor is made of an enzyme
immobilized on the surface of a transducer.
Enzymes in nanobiosensor adjust the sensitivity and stability of them; therefore,
more substrate specificity makes more enzyme turnover number and thermostability,
the main necessity for a biosensor. Bioconjugation is a biosensing method with cost-
effectiveness, based on the nature of interaction between nanomaterials and the
enzyme surface functional groups. In nanobioconjugation technique, physical
(adsorption, entrapment), covalent (covalent binding, cross-linking), and bioaffinity
types are used to plan a strong nanobiosensor. Weak bioconjugation could have a
little activity of biosensor, following the change of the enzyme active site conforma-
tion. Therefore, by more understanding functionality of a biomolecule at interface,
better choice to plan and improve a bioconjugation would happen.
Nanobiosensors have been used to detect the pathogens in foods, and the optical
assays like fluorescence and surface plasmon resonance are the most usable
techniques. It was demonstrated that by using covalently co-immobilized enzymes
choline oxidase and peroxidase on zinc oxide nanorod films, detection limit would
480 Z. B. Mohammadi et al.
17.4.2 Toxicity
17.6 Conclusion
The latest attentions to nanomaterials and their wide applications have made it
possible to immobilize enzymes, maintain their enzymatic activity, reuse, and finally
recycle them. Various nanostructures such as nanofibers, nanoparticles, nanotubes,
nanosheets, and others have been proven to act as efficient media for enzyme
stabilization. If produced under good manufacturing practice and according to health
regulations, these particles are preferred in comparison with their chemical
counterparts. Enzymatic biocatalysts have become the center of attention as sensors
in order to ensure food quality and safety. They are used as indicators of pesticides’
residues, pathogenic microorganism, and their toxic metabolites. Nanotechnology
and stabilization on nanostructures have been widely used in the production process
of these biosensors.
References
Agustian, J., Kamaruddin, A. H., & Aboul-Enein, H. Y. (2016). Enantio-conversion and-selectivity
of racemic atenolol kinetic resolution using free Pseudomonas fluorescens lipase (Amano)
conducted via transesterification reaction. RSC Advances, 6(31), 26077–26085.
Alehosseini, A., Ghorani, B., Sarabi-Jamab, M., & Tucker, N. (2017). Principles of electrospraying:
A new approach in protection of bioactive compounds in foods. Critical Reviews in Food
Science and Nutrition, 8398, 1–18.
An, J., Zhang, M., Wang, S., & Tang, J. (2008). Physical, chemical and microbiological changes in
stored green asparagus spears as affected by coating of silver nanoparticles-PVP. LWT-Food
Science and Technology, 41(6), 1100–1107.
Athinarayanan, J., Periasamy, V. S., Alsaif, M. A., Al-Warthan, A. A., & Alshatwi, A. A. (2014).
Presence of nanosilica (E551) in commercial food products: TNF-mediated oxidative stress and
altered cell cycle progression in human lung fibroblast cells. Cell Biology and Toxicology, 30(2),
89–100.
17 Nanotechnology and Food Grade Enzymes 483
Bajpai, V. K., Kamle, M., Shukla, S., Mahato, D. K., Chandra, P., Hwang, S. K., Kumar, P., Huh,
Y. S., & Han, Y. K. (2018). Prospects of using nanotechnology for food preservation, safety,
and security. Journal of Food and Drug Analysis, 26(4), 1201–1214.
Baskar, G., Banu, N. A., Leuca, G. H., Gayathri, V., & Jeyashree, N. (2015). Magnetic immobili-
zation and characterization of α-amylase as nanobiocatalyst for hydrolysis of sweet potato
starch. Biochemical Engineering Journal, 102, 18–23.
Bayramoglu, G., Doz, T., Ozalp, V. C., & Arica, M. Y. (2017). Improvement stability and
performance of invertase via immobilization on to silanized and polymer brush grafted magnetic
nanoparticles. Food Chemistry, 221, 1442–1450.
Beig Mohammadi, Z., Hamidi Esfahani, Z., Sahari, M., & Khosravi Darani, K. (2020). Enzymatic
degumming of soybean oil by immobilized phospholipase A1 on plasma surface modified
chitosan nanofibrous membrane. Iranian Journal of Chemistry and Chemical Engineering
(IJCCE), 40, 1421–1430. https://doi.org/10.30492/ijcce.2020.40607
Beig Mohammadi, Z., Hamidi-Esfahani, Z., Sahari, M., & Khosravi-Darani, K. (2016). Optimiza-
tion of phospholipase A1 immobilization on plasma surface modified chitosan nanofibrous mat.
Applied Food Biotechnology, 3(1), 25–34.
Beverlya, R. L., Janes, M. E., Prinyawiwatkula, W., & No, H. K. (2008). Edible chitosan films on
ready-to-eat roast beef for the control of Listeria monocytogenes. Food Microbiology, 25(3),
534–537.
Buyukhatipoglu, K., & Clyne, A. M. (2011). Superparamagnetic iron oxide nanoparticles change
endothelial cell morphology and mechanics via reactive oxygen species formation. Journal of
Biomedical Materials Research Part A, 96(1), 186–195.
Cao, Y., Wen, L., Svec, F., Tan, T., & Lv, Y. (2016). Magnetic AuNP@ Fe3O4 nanoparticles as
reusable carriers for reversible enzyme immobilization. Chemical Engineering Journal, 286,
272–281.
Cushen, M., Kerry, J., Morris, M., Cruz-Romero, M., & Cummins, E. (2012). Nanotechnologies in
the food industry—Recent developments, risks and regulation. Trends in Food Science &
Technology, 24(1), 30–46.
Dwivedi, C., Pandey, I., Misra, V., Giulbudagian, M., Jungnickel, H., Laux, P., Luch, A., Ramteke,
P. W., & Singh, A. V. (2018). The prospective role of nanobiotechnology in food and food
packaging products. Integrative Food, Nutrition and Metabolism (IFNM), 5, 1–5.
Emamifar, A., Kadivar, M., Shahedi, M., & Soleimanian-Zad, S. (2010). Evaluation of
nanocomposite packaging containing Ag and ZnO on shelf life of fresh orange juice. Innovative
Food Science & Emerging Technologies, 11(4), 742–748.
Emamifar, A., Kadivar, M., Shahedi, M., & Soleimanian-Zad, S. (2012). Effect of nanocomposite
packaging containing Ag and ZnO on reducing pasteurization temperature of orange juice.
Journal of Food Processing and Preservation, 36(2), 104–112.
Faramarzi, S., Anzabi, Y., & Jafarizadeh-Malmiri, H. (2020). Nanobiotechnology approach in
intracellular selenium nanoparticle synthesis using Saccharomyces cerevisiae—Fabrication
and characterization. Archives of Microbiology, 1–7.
Fathi, M., Karim, M., Khoigani, S. R., & Mosayebi, V. (2018). Use of nanotechnology for
immobilization and entrapment of food applicable enzymes. In Bioactive molecules in food
(pp. 1–25). Springer.
Fatima Mustafa, F., & Andreescu, S. (2018). Chemical and biological sensors for food-quality
monitoring and smart packaging. Food, 7, 168. https://doi.org/10.3390/foods7100168
Fernández, A., Picouet, P., & Lloret, E. (2010). Cellulose-silver nanoparticle hybrid materials to
control spoilage-related microflora in absorbent pads located in trays of fresh-cut melon.
International Journal of Food Microbiology, 142(1-2), 222–228.
Fernandez, A., Picouet, P., & Lloret, E. (2010). Reduction of the spoilage-related microflora in
absorbent pads by silver nanotechnology during modified atmosphere packaging of beef meat.
Journal of Food Protection, 73(12), 2263–2269.
484 Z. B. Mohammadi et al.
Fernández, A., Soriano, E., López-Carballo, G., Picouet, P., Lloret, E., Gavara, R., &
HernándezMuñoz, P. (2009). Preservation of aseptic conditions in absorbent pads by using
silver nanotechnology. Food Research International, 42(8), 1105–1112.
Fuertes, G., Soto, I., Carrasco, R., Vargas, M., Sabattin, J., & Lagos, C. (2016). Intelligent
packaging systems: Sensors and nanosensors to monitor food quality and safety. Journal of
Sensors, 2016, 4046061.
Gabrielczyk, J., Duensing, T., Buchholz, S., Schwinges, A., & Jordening, H. J. (2018). A compara-
tive study on immobilization of fructosyltransferase in biodegradable polymers by
electrospinning. Applied Biochemistry and Biotechnology, 185(3), 847–862.
Ghasemi, S., Jafari, S. M., Assadpour, E., & Khomeiri, M. (2017). Production of pectin-whey
protein nano-complexes as carriers of orange peel oil. Carbohydrate Polymers, 177, 369–377.
Ghasemi, S., Jafari, S. M., Assadpour, E., & Khomeiri, M. (2018). Nanoencapsulation of
d-limonene within nanocarriers produced by pectin-whey protein complexes. Food
Hydrocolloids, 77, 152–162.
Ghorani, B., Alehosseini, A., & Tucker, N. (2017). 8. Nanocapsule formation by electrospinning. In
S. M. Jafari (Ed.), Nanoencapsulation technologies for the food and nutraceutical industries
(pp. 264–319). Academic Press.
Ghosal, K., Chandra, A., Praveen, G., Snigdha, S., Roy, S., Agatemor, C., et al. (2018).
Electrospinning over solvent casting: Tuning of mechanical properties of membranes. Scientific
Reports, 8(1), 5058.
Ghouri, M. Z., Khan, Z., Khan, S. H., Ismail, M., Aftab, S. O., Sultan, Q., & Ahmad, A. (2020).
Nanotechnology: Transformation of agriculture and food security. Bioscience, 3, 19.
Handford, C. E., Dean, M., Spence, M., Henchion, M., Elliott, C. T., & Campbell, K. (2014).
Nanotechnology in the agri-food industry on the island of Ireland: Applications, opportunities
and challenges. Institute for Global Food Security at Queen’s University, Belfast, and the
Teagasc Ashtown Food Research Centre.
Hansen, S. F., Maynard, A., Baun, A., Tickner, J. A., & Bowman, D. M. (2013).
22 Nanotechnologyearly lessons from early warnings. In Late lessons from early warnings:
Science, precaution, innovation (Vol. 32).
Haroon, F., & Ghazanfar, M. (2016). Applications of food biotechnology. Journal of Ecosystem
and Ecography, 6(215), 2.
He, X., & Hwang, H. M. (2016). Nanotechnology in food science: Functionality, applicability, and
safety assessment. Journal of Food and Drug Analysis, 24(4), 671–681.
Higashisaka, K., Yoshioka, Y., & Tsutsumi, Y. (2015). Applications and safety of nanomaterials
used in the food industry. Food Safety, 3(2), 39–47.
Homaei, A., & Samari, F. (2017). Investigation of activity and stability of papain by adsorption on
multi-wall carbon nanotubes. International Journal of Biological Macromolecules, 105,
1630–1635.
Husain, Q. (2010). β Galactosidases and their potential applications: A review. Critical Reviews in
Biotechnology, 30(1), 41–62.
Jafari, S. M., Fathi, M., & Mandala, I. (2015). Chapter 13-Emerging product formation. In C. M.
Galanakis (Ed.), Food waste recovery (pp. 293–317). Academic Press.
Jafari, S. M. (2019). Nanoencapsulation of food ingredients by specialized equipment: Volume 3 in
the Nanoencapsulation in the Food Industry series (Vol. 3). Academic Press.
Jafarizadeh-Malmiri, H., Sayyar, Z., Anarjan, N., & Berenjian, A. (2019). Nanobiotechnology
in food: Concepts, applications and perspectives. Springer.
James, J., Simpson, B. K., & Marshall, M. R. (1996). Application of enzymes in food processing.
Critical Reviews in Food Science & Nutrition, 36(5), 437–463.
Jia, F., Narasimhan, B., & Mallapragada, S. (2014). Materialsbased strategies for multienzyme
immobilization and colocalization: A review. Biotechnology and Bioengineering, 111(2),
209–222.
17 Nanotechnology and Food Grade Enzymes 485
Jin, T., & Gurtler, J. B. (2011). Inactivation of Salmonella in liquid egg albumen by antimicrobial
bottle coatings infused with allyl isothiocyanate, nisin and zinc oxide nanoparticles. Journal of
Applied Microbiology, 110(3), 704–712.
Kharazmi, S., Taheri-Kafrani, A., & Soozanipour, A. (2020). Efficient immobilization of pectinase
on trichlorotriazine-functionalized polyethylene glycol-grafted magnetic nanoparticles: A stable
and robust nanobiocatalyst for fruit juice clarification. Food Chemistry, 325, 126890.
Kumar, H., & Neelam, R. (2016). Enzyme-based electrochemical biosensors for food safety: A
review. Nanobiosensors in Disease Diagnosis, 5, 29–39.
Li, H., Li, F., Wang, L., Sheng, J., Xin, Z., Zhao, L., Xiao, H., Zheng, Y., & Hu, Q. (2009). Effect of
nano-packing on preservation quality of Chinese jujube (Ziziphus jujuba Mill. var. inermis
(Bunge) Rehd). Food Chemistry, 114(2), 547–552.
McClements, D. J., & Xiao, H. (2017). Is nano safe in foods? Establishing the factors impacting the
gastrointestinal fate and toxicity of organic and inorganic food-grade nanoparticles. NPJ Science
of Food, 1(1), 1–13.
McClements, D. J., Decker, E. A., Park, Y., & Weiss, J. (2009). Structural design principles for
delivery of bioactive components in nutraceuticals and functional foods. Critical Reviews in
Food Science and Nutrition, 49(6), 577–606.
Morais, M. G. D., Martins, V. G., Steffens, D., Pranke, P., & da Costa, J. A. V. (2014). Biological
applications of nanobiotechnology. Journal of Nanoscience and Nanotechnology, 14(1),
1007–1017.
Mosafa, L., Moghadam, M., & Shahedi, M. (2013). Papain enzyme supported on magnetic
nanoparticles: Preparation, characterization and application in the fruit juice clarification.
Chinese Journal of Catalysis, 34(10), 1897–1904.
Mukhopadhyay, A., Bhattacharyya, T., Dasgupta, A. K., & Chakrabarti, K. (2015). Nanotechnol-
ogy based activation-immobilization of psychrophilic pectate lyase: A novel approach towards
enzyme stabilization and enhanced activity. Journal of Molecular Catalysis B: Enzymatic, 119,
54–63.
Mustafa, F., & Andreescu, S. (2020). Nanotechnology-based approaches for food sensing and
packaging applications. RSC (Royal Society of Chemistry) Advances, 10, 19309–19336. https://
doi.org/10.1039/d0ra01084g
Neethirajan, S., & Jayas, D. S. (2011). Nanotechnology for the food and bioprocessing industries.
Food and Bioprocess Technology, 4(1), 39–47.
Neo, Y. P., Ray, S., & Perera, C. O. (2018). Fabrication of functional electrospun nanostructures for
food applications. In Role of materials science in food bioengineering (pp. 109–146). Academic
Press.
Netto, C. G., Toma, H. E., & Andrade, L. H. (2013). Superparamagnetic nanoparticles as versatile
carriers and supporting materials for enzymes. Journal of Molecular Catalysis B: Enzymatic, 85,
71–92.
Nile, S. H., Baskar, V., Selvaraj, D., Nile, A., Xiao, J., & Kai, G. (2020). Nanotechnologies in food
science: Applications, recent trends, and future perspectives. Nano-Micro Letters, 12(1), 45.
Park, B., Fu, J., Zhao, Y., Siragusa, G. R., Cho, Y. J., Lawrence, K. C., & Windham, W. R. (2007).
Bio-functional Au/Si nanorods for pathogen detection. In Nanosensing: Materials, devices, and
systems III (Vol. 6769, p. 67690). International Society for Optics and Photonics.
Park, Y. W., Kim, S. M., Lee, J. Y., & Jang, W. (2015). Application of biosensors in smart
packaging. Molecular & Cellular Toxicology, 11(3), 277–285.
Patel, S. N., Singh, V., Sharma, M., Sangwan, R. S., Singhal, N. K., & Singh, S. P. (2018).
Development of a thermo-stable and recyclable magnetic nanobiocatalyst for bioprocessing of
fruit processing residues and D-allulose synthesis. Bioresource Technology, 247, 633–639.
Pradhan, N., Singh, S., Ojha, N., Shrivastava, A., Barla, A., Rai, V., & Bose, S. (2015). Facets of
nanotechnology as seen in food processing, packaging, and preservation industry. BioMed
Research International, 2015, 365672.
486 Z. B. Mohammadi et al.
Raghu, P., Reddy, T. M., Reddaiah, K., Swamy, B. K., & Sreedhar, M. (2014). Acetylcholinester-
ase based biosensor for monitoring of malathion and acephate in food samples: A voltammetric
study. Food Chemistry, 142, 188–196.
Rashidi, L., & Khosravi-Darani, K. (2011). The applications of nanotechnology in food industry.
Critical Reviews in Food Science and Nutrition, 51(8), 723–730.
Ravichandran, R. (2010). Nanotechnology applications in food and food processing: Innovative
green approaches, opportunities and uncertainties for global market. International Journal of
Green Nanotechnology: Physics and Chemistry, 1(2), P72–P96.
Rojas-Mercado, A. S., Moreno-Cortez, I. E., Lucio-Porto, R., & Pavo’n, L. L. (2018). Encapsula-
tion and immobilization of ficin extract in electrospun polymeric nanofibers. International
Journal of Biological Macromolecules, 118(Pt B), 2287–2295.
Ruane, J., & Sonnino, A. (2011). Agricultural biotechnologies in developing countries and their
possible contribution to food security. Journal of Biotechnology, 156(4), 356–363.
Saallah, S., Naim, M. N., Lenggoro, I. W., Mokhtar, M. N., Bakar, N. F. A., & Gen, M. (2016).
Immobilisation of cyclodextrin glucanotransferase into polyvinyl alcohol (PVA) nanofibres via
electrospinning. Biotechnology Reports, 10, 44–48.
Shahiri Tabarestani, H., & Jafari, S. M. (2019). Production of food bioactive-loaded nanofibers by
electrospinning. In Nanoencapsulation of food ingredients by specialized equipment
(pp. 31–105). Academic Press.
Shahrestani, H., Taheri-Kafrani, A., Soozanipour, A., & Tavakoli, O. (2016). Enzymatic
clarification of fruit juices using xylanase immobilized on 1,3,5-triazine-functionalized
silicaencapsulated magnetic nanoparticles. Biochemical Engineering Journal, 109, 51–58.
Sharma, C., Dhiman, R., Rokana, N., & Panwar, H. (2017). Nanotechnology: An untapped resource
for food packaging. Frontiers in Microbiology, 8, 1735.
Sheldon, R. A. (2007). Enzyme immobilization: The quest for optimum performance. Advanced
Synthesis & Catalysis, 349(8-9), 1289–1307.
Singh, N., Srivastava, G., Talat, M., Raghubanshi, H., Srivastava, O. N., & Kayastha, A. M. (2014).
Cicer α-galactosidase immobilization onto functionalized graphene nanosheets using response
surface method and its applications. Food Chemistry, 142, 430–438.
Skoronski, E., Souza, D. H., Ely, C., Broilo, F., Fernandes, M., Junior, A. F., & Ghislandi, M. G.
(2017). Immobilization of laccase from Aspergillus oryzae on graphene nanosheets. Interna-
tional Journal of Biological Macromolecules, 99, 121–127.
Sojitra, U. V., Nadar, S. S., & Rathod, V. K. (2016). A magnetic tri-enzyme nanobiocatalyst for
fruit juice clarification. Food Chemistry, 213, 296–305.
Sundarraj, A. A. (2019). Nano-agriculture in the food industry. In Plant nanobionics (pp. 183–200).
Springer.
Tavares, A. P., Silva, C. G., Dražić, G., Silva, A. M., Loureiro, J. M., & Faria, J. L. (2015). Laccase
immobilization over multi-walled carbon nanotubes: Kinetic, thermodynamic and stability
studies. Journal of Colloid and Interface Science, 454, 52–60.
Thakur, M. S., & Ragavan, K. V. (2013). Biosensors in food processing. Journal of Food Science
and Technology, 50(4), 625–641.
Vinayaka, A. C., Basheer, S., & Thakur, M. S. (2009). Bioconjugation of CdTe quantum dot for the
detection of 2,4-dichlorophenoxyacetic acid by competitive fluoroimmunoassay based biosen-
sor. Biosensors and Bioelectronics, 24(6), 1615–1620.
Wang, X. Y., Jiang, X. P., Li, Y., Zeng, S., & Zhang, Y. W. (2015). Preparation Fe3O4@ chitosan
magnetic particles for covalent immobilization of lipase from Thermomyces lanuginosus.
International Journal of Biological Macromolecules, 75, 44–50.
Wen, P., Zong, M. H., Linhardt, R. J., Feng, K., & Wu, H. (2017). Electrospinning: A novel
nanoencapsulation approach for bioactive compounds. Trends in Food Science & Technology,
70, 56–68.
Wong, D. E. (2016). Enzyme stabilization in hierarchical biocatalytic food packaging and
processing materials. Doctoral dissertations, p. 672.
17 Nanotechnology and Food Grade Enzymes 487
Xie, W., & Ma, N. (2010). Enzymatic transesterification of soybean oil by using immobilized lipase
on magnetic nano-particles. Biomass and Bioenergy, 34(6), 890–896.
Yu, Z., Li, B., Chu, J., & Zhang, P. (2018a). Silica in situ enhanced PVA/chitosan biodegradable
films for food packages. Carbohydrate Polymers, 184, 214–220.
Yu, H., Park, J. Y., Kwon, C. W., Hong, S. C., Park, K. M., & Chang, P. S. (2018b). An overview of
nanotechnology in food science: Preparative methods, practical applications, and safety. Jour-
nal of Chemistry, 2018.
Zhou, L., Lv, S., He, G., He, Q., & Shi, B. I. (2011). Effect of PE/AG2O nano packaging on the
quality of apple slices. Journal of Food Quality, 34(3), 171–176.