Sie sind auf Seite 1von 22

Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

Contents lists available at ScienceDirect

Biocatalysis and Agricultural Biotechnology


journal homepage: www.elsevier.com/locate/bab

Marine microbes as a valuable resource for brand new industrial biocatalysts MARK
a b,⁎
Azadeh Beygmoradi , Ahmad Homaei
a
Department of Marine Biology, Faculty of Sciences, University of Hormozgan, Bandar Abbas, Iran
b
Department of Biochemistry, Faculty of Sciences, University of Hormozgan, P.O. Box 3995, Bandar Abbas, Iran

A R T I C L E I N F O A B S T R A C T

Keywords: The marine environment is one of the most important sources of bioactive compounds in the world. Marine
Marine microbes, Valuable resource, Novel microbial communities from marine environments secrete variety enzymes based on their habitat and their
industrial biocatalysts ecological roles. They are considered important ecological components in marine environments due to their
Biochemical characteristics performance in biogeochemical processes. Marine microbial enzymes including protease, lipase, collagenase,
Biotechnological applications
agarase, cellulose and Tother enzymes are important in different industrial due to their properties and appli-
cations. The biochemical diversity of marine microbial makes them reasonable sources of a wide variety of
enzymes for use in different industrial including food, medicine and other biotechnological systems. Therefore,
marine microbial enzymes preparations due to a special applications and functions in the biotechnology projects,
also these can suggestion novel catalysts of biological and biochemical reactions with unique properties and
functions. In this review, we focus on microbial enzymes from marine derived and their biochemical significance
and their increasing importance in biotechnology.

1. Introduction special lighting conditions, microorganisms in the marine environment


compared to their unique living environments and habitats genetic
The marine with more than 70% of Earth's area, not only has rich properties (Stach et al., 2003). Marine microbes of different habitats
biodiversity but also a source for the microorganisms with potential is and the habitats shelter a diverse range of microbes, including archaea,
vast. Marine environment are including diverse communities of plants, cyanobacteria, eubacteria, Actinomycetes, yeasts, filamentous fungi,
animals and microbes that marine microbial communities including microalgae, algae and protozoa. Almost all this potential source of
bacteria, fungi, viruses and etc. have several advantage in marine en- enzymes that are beneficial to their potential has not been fully un-
vironments due to their performance in biotechnological processes derstood. The main goal of modern enzyme technology and storage of
(Sowell et al., 2008). Marine environment contains huge potential of food components, effective use of raw materials, improve food quality,
microorganisms, fungi, plants and animals are a rich source of biodi- food diet, raw food use in the preparation of animal feed and process
versity, which have the capability to extract enzymes from marine optimization is to reduce costs. The oceans, huge library of unique
sources. A marine enzyme may be a unique protein molecule not found compounds and natural products and bioactive substances enzymes
in any terrestrial organism or it may be a known enzyme from a ter- promising and surprising such as protease, amylase, lipase, chitinase,
restrial source but with novel properties (Dadshahi et al., cellulose, ligninase, pectinase, xylanase, nucleases (DNAase, RNAase,
2016; Homaei, 2015b; Homaei et al., 2016a, 2016b). Microbial en- restriction enzymes), and so are the potential application, which will
zymes have more advantages than the enzymes are derived from plant never be found in a dry environment (Mohebbi et al., 2014). For ex-
or animal because these enzymes have advantages such as broad bio- ample, marine actinomycetes not only in terms of ecological and tax-
chemical diversity, the ability to mass culture, ease of genetic manip- onomical but also in terms of production and new biochemical bioac-
ulation, a more catalytic activity, lower costs, equipment and sustain- tive compounds such as antibiotics, anti-tumor, suppress the immune
ability are relatively more abundant (Bull et al., 2000). With the system, enzymes, enzyme inhibitors (Dharmaraj, 2010), antimicrobial
advancement of marine science, biotechnology and microbial fermen- compounds, including aminoglycosides, antracyclines, the glycopep-
tation technology, demand and interest researchers to identify and tide, beta-lactams, macrolides, nucleosides, peptides, made of them,
characterize microbial enzymes have been more sea (Sharifian et al., polyester, polyketoides, actinomycin and tetracyclines are important
2017; Zeinali et al., 2015). Marine environment with extreme chemical (Berdy, 2005). Marine actinomycetes are virtually unlimited sources of
conditions, including high salinity, high pressure, low temperature and novel compounds and streptomyces the most economical and the most


Corresponding author.
E-mail addresses: a.homaei@hormozgan.ac.ir, a.homaei@gmail.com (A. Homaei).

http://dx.doi.org/10.1016/j.bcab.2017.06.013
Received 22 May 2017; Received in revised form 26 June 2017; Accepted 26 June 2017
Available online 28 June 2017
1878-8181/ © 2017 Elsevier Ltd. All rights reserved.
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

Table 1
Industrial and commercial applications of numerous marine microbial enzymes.

Enzymes Applications References

Lipase Food flavoring, organic synthesis, cosmetic production, detergents, paper production, (Chi et al., 2009; Ferreira-Dias et al., 2013)
biodiesel industry and textile industry
Protease Detergent industry, leather industry, pharmaceutical, commercial liquid and solid detergents (Chanalia et al., 2011; Ilona and Zdzislaw, 2007;
Sawant and Nagendran, 2014)
Chitinase and Increase immune function, improve digestive function, drug delivery, agriculture, wound care (Je and Kim, 2006; Kumar et al., 2014; Ngo et al.,
chitosanase application, metal capture from wastewater and prevent tumor cell growth 2008)
Agarases Additive in moisturizing cosmetic (Martin et al., 2014; Oren, 2004)
Carrageenase Aoagulant, adhesives, stabilizer, gelation and emulsifier (Zhao et al., 2012)
Amylase Added to flour at the mill or bread making and (Gupta et al., 2003)
oxidized gluten for improved dough properties
Cellulase Biotextile, cotton and linen products processing and biofertilizer (Sukharnikov et al., 2011; Zhao et al., 2012)
xylanase Useful in textiles, paper and pulp industry, decrease the usage of Cl2 and ClO2, reduce (Doi, 2008; Nishiyama et al., 2002)
pollution and cleavage some polysaccharides in juice or beer industry
Tannase Usage of antioxidants in fats and oils in the food industry, wines and beer, treated green tea, (Chávez-González et al., 2012; Du-Thumm et al.,
use of tannase as an ingredient of animal feed, treatment of fruit juices to reduce the bitterness 2005; Van de Lagemaat and Pyle, 2006)
and coffee flavored, discolor the tooth surface
Phytase Use in bakery and sweeteners (Jorquera et al., 2008; Lei and Porres, 2003)
Esterase Degradation of plastic, usage of food, textile industries and deinking (Sayali et al., 2013)

valuable biotechnology are prokaryotes. Studies have shown that notably Vibrio sp., Pseudomonas stutzeri, Aeromonas sp, Cytophaga,
bioactive compounds derived from the genus streptomyces. In fact, Bacillus, Alteromonas, Pseudoalteromonas, Streptomyces (Aoki et al.,
stereptomysis genus alone is capable of producing more than 80% of 1990; Hosoda et al., 2003; Sugano et al., 1993b) and Alteromonas
natural products actinomycetes and analysis of complex biological agarlyticus GJ1B (P. Potin et al., 1993), Thalassomonas sp. JAMB-A33
polymers, and in this regard remains unrivaled in the world of microbes (Ohta and Hatada, 2006), Alteromonas sp. E-l (Kirimura et al., 1999),
(Manivasagan et al., 2014). Microscilla (Naganuma et al., 1993), Streptomyces coelicolor A3 (Buttner
According to recent problems such as high population most coun- et al., 1987), Pseudoalteromonas sp. BL-3 (Lee et al., 2005), Vibrio sp.
tries, the waste of resources and pollution, identify the marine sources AP-2 (Aoki et al., 1990), Pseudomonas atlantica (Belas, 1989), Bacillus
is important biological development. Marine microbial enzymes, par- sp. MK03 (Suzuki et al., 2003), Catenovulum agarivorans (Cui et al.,
ticularly enzymes that have the ability to survive in extreme conditions 2014), Microbulbifermaritimus (Vijayaraghavan and Rajendran, 2012),
such as heat-resistant enzymes used in PCR thermal vents near the Alteromonas sp. C-1 (Leon et al., 1992), Vibrio sp. JT0107 (Sugano et al.,
ocean floor of the bacteria obtained the importance and more appli- 1993a), Agarivorans sp. HZ105 (Hu et al., 2009), Bacillus cereus ASK202
cation (Grace, 1997). This review wills briefly discus potential uses of (Kim et al., 1999), Alteromonas sp., Cytophaga sp., Agarivorans gilvus and
properties marine microbial enzymes with novel chemical biodiversity Pseudoalteromonas sp. (Chi et al., 2012; Fu and Kim, 2010). Halococcus
for its various industrial and medical applications (Table 1). sp. is capable to production of β-agarases with molecular weight 55 kDa
and optimum pH 6 (Minegishi et al., 2013). Also gram-negative bacteria
2. Properties, importance and biotechnological applications of Microbulbifer maritimus were detected in capable of producing extra-
marine microbial agarases cellular enzymes agarases with molecular weight 75.2 kDa and Km
3 mM (Vijayaraghavan and Rajendran, 2012).
Agarases a colloid extracted from seaweed, which is used in the Agarase is an enzyme with systematic name agarose 4-glycanohy-
decomposition of agar. Agar or agar-agar is a jelly-like substance, ob- drolase that found in agarolytic microorganisms (Parro and Mellado,
tained from algae is a mixture of agarose and agaropectin which is 1994). An agarase marine bacterium are able to control many appli-
extracted from red algae Gelidium robustum. Agarose, a polysaccharide cations including red algae bloom, avoid biofouling on sea levels as a
with a molecular weight of 120 kDa, which is a duplicate of D-galactose food additive in food and cosmetics moisturizing additives, and has a
and 3,6-anhydro-L-galactose β1 → 4 and α1 → 3 connected with the high level of activity for depolimerization the complex polysaccharides
gangs have been formed (Chi et al., 2012; Fernandes, 2014; Fu and Kim, such as agar and agarose. In addition emulsification capabilities, the
2010). gelatinized and particles are stable, they also can be found in the food
Agar pectin a heterogeneous mixture of small molecules with a industry in the manufacture of soft drinks, confectionary, jelly, bread
backbone, such as agarose, but shows the ionic nature of the acid and produce some low-calorie foods used (Rasmussen and Morrissey,
groups (such as sulphate, pyruvate) is covered is as natural compounds 2007; Yaphe and Morgan, 1959). Agarase in genetic science is im-
(Delattre et al., 2011). Alpha and beta agarases has two classes that portant and necessary; also have applications in gene technology and
according to research conducted by researchers in various marine or- agarose gel electrophoresis to separate DNA fragments ( Table 2).
ganisms, most agarases been detected β-agarases category (Chi et al.,
2012; Han et al., 2013). 3. Extremozymes
In 1902, Gran isolated agar-degrading Pseudomonas galatica from
seawater (Zhang and Kim, 2010). In 1987, Mervyn found the Strepto- The sea is extremely complex environment with a variety of dif-
myces agarase gen (dagA) (Buttner et al., 1987). Marine bacteria Vibrio ferent physicochemical conditions of extreme conditions such as large
sp. (JT0107) the ability to bond hydrolysis α-l, 3 glycosidic agar is by α fluctuations in temperature, strong alkaline, strong acid, high hydro-
-Neoagaro-oligosaccharides (Sugano et al., 1993b). Rosert studied on static pressure and nutrient status is very poor. Microorganisms such as
agaA gene of Pseudomonas and agarases gene sequencing (Belas, 1989). fungi, bacteria, algae and other microorganisms that are able to grow in
In 1993 by Yasushi, agaA gene from Vibrio cloned and sequenced. extreme situations have been called extremophilic (Table 3). The ex-
Suzuki et al. (2003) purified a new type beta-agarase agaropectin of tremophilic microorganisms in biotechnology research are a topic of
bacteria Bacillus sp. MK03, enzyme hydrolysis neoagarohexaose ability interest by many researchers (Danson and Hough, 1998).
to produce neoagarotetraose and its neoagarobiose. Alteromonas, psychrotrophs and other microorganisms that are psy-
Agarases have been isolated from several marine source, most chrophilies due to resistant to low temperatures are very useful and has

132
A. Beygmoradi, A. Homaei

Table 2
Important kinetic and biochemical properties of marine microbial agarases.

B Species Optimum pH Optimum Km Activators Inhibitors Other properties References


Temprature (°C)

Saccharophagus sp. 7.5 55 Ca2+, K+,Na+ and EDTA (each 2 mM) Ca2+, Mn2+, Zn2+ and EDTA (Lee et al., 2013)
(each 2 mM)
Catenovulum agarivorans 6.0 60 3.78 mg mL−1 Na+, K+, Ca2+ and SDS (each 1 mM) Cu2+, Fe3+, Mg2+, Mn2+, EDTA Molecular mass: 46.9 kDa, (Cui et al., 2014)
(each 1 mM) and Ni2+ (10 mM) Thermostable
Catenovulum sp. 7.4 52 Ca2+ and Mg2+ (each 2 and 10 mM), Mn2+ (2 mM and 10 mM), Fe3+ (Xie et al., 2013)
Na+ and K+ (each 10 mM) and β- (2 mM), EDTA (2 mM and
mercaptoethanol (10 mM) 10 mM), SDS (2 mM) and urea
(10 mM)
Zobellia galactanivorans 6.0 2 mM Molecular mass: 13.4 kDa, (Murielle et al., 2005)
(AgaAc) Kcat: 150 s−1
Zobellia galactanivorans 7.0 1 mM Molecular mass: 60 kDa, (Murielle et al., 2005)
(AgaB) Kcat: 100 s−1
Pseudoalteromonas sp. 6.0 40 8.36 mM Ca2+ and Ba2+ Hg2+, Cu2+, Mn2+ and EDTA Molecular mass: 50.4 kDa, (Ma et al., 2007)
CY24
−1
(AgaB) Kcat: 5.16 s

133
Acinetobacter junii PS12B 7.0 35 Specific activity: 1.90+0.10 U/mg, (Roseline and
Enzyme activity: 0.37+0.12 U/mg Sachindra, 2016)
Pseudoalteromonas 37 Molecular mass: 30 kDa (Ramos et al., 2016)
hodoensis sp.a
Acinetobacter sp., AG 6.0 40 0.071 mg mL−1 Zn2+, Sn2+ and SDS Hg2+, Ag+ and Cu2+ Molecular mass: 100 kDa, (Lakshmikanth et al.,
LSL−1 Specific activity: 397 U/mg, 2009)
Vmax: 368 U/mg
Alteromonas sp.b GNUM1 7.0 40 (Seo et al., 2014)
Microscilla 6–8 24–37 10 mg/l to 500 mg/ Enzyme was stable in the presence of NaCl (Naganuma et al.,
l concentrations (0.5–0.7 M) 1993)
Alteromonas sp. E-l 7.5 40 K+, Na+ and EDTA Mn2+, Cu2+, Fe2+, Zn2+ and Molecular masses: 82 (by SDS- (Kirimura et al., 1999)
Hg2+ polyacrylamide gel electrophoresis) and
180 kDa (by Superdex 200 kDa gel
filtration),
Specific activity: 34 U/mg
Catenovulum agarivorans 6.0 60 3.78 mg mL−1 Mg2+, urea, EDTA and SDS Cu2+, Mn2+, Fe3+ and Ni2+ Molecular mass: 46.9 kDa, (Cui et al., 2014)
YM01 Vmax: 1.14 × 104 U/mg

a
Expressed in Bacillus subtilis extracellular -agarase (AgaA7).
b
Expressed in Escherichia coli (AgaG1).
Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

Table 3
Diversity of microbes in extreme marine environmental conditions.

Living environment Properties species References

Psychrophilies Extracellular protease production by the marine Rhodotorula mucilaginosa L7 (Lario et al., 2016)
antarctic yeast,
Maximum proteolytic activity 23.98 U/mL at pH
4.0 and 10 ◦C
R. mucilaginosa L7, Rhodotorula glutinis (Kamada et al., 1972)
Exiguobacterium sp. (Kasana and Yadav, 2007)
SKPB5 (MTCC 7803)
Thermophilies Aeromonas sp. (Charoenpanich et al., 2011)
Hyperthermophilic archaebacteria Pyrococcus furiosus (Adams et al., 1995)
Thermotoga sp.
Bacillus stearothermophilus, Thermocrinis ruber, (Homaei et al., 2016)
Thermus Rt41A
Sulfolobus acidocaldariusis, Methanococcus (Tsuchiya et al., 1992)
jannaschii
Thermoactinomyces sp. HS682
Pyrococcus furiosus
Thermostable and halophilic Zunongwangia profunda (Vieille and Zeikus, 2001)
Thermo- and alkali-stable Bacillus licheniformis (Liu et al., 2014)
(Songsiriritthigul et al.,2010)
Bacillus sp. GX6638 (Durham et al., 1987)
Acidophilies Can be live at pH 5 and even below 1 Aspergillus awamori BTMFW032 (Beena et al., 2010)
Alkaliphilies Bacillus sp. (Fujiwara et al., 1993)
Halophilies Planococcus rifitoensis (Essghaier et al., 2010)
Haloalkaliphilic Bacillus pumilus (Menon et al., 2010)
Bacillus Clausii (Kumar et al., 2004)
Bacillus sp. (Ve2–20–91 [HM047794]) (Raval et al., 2014)
Alkaliphilie, halophilies, and Marinimicrobium sp. (Zhao et al., 2012)
thermostable

many advantages over a simple culture, reduce costs, save energy and the microorganisms or enzymes alkaliphilic acidophilus are generally
etc. The enzymes lipase and protease derived from them, especially has enzymes. In some areas of the ocean is conditions acidophilus or al-
great potential in terms of biochemical industry and has the highest kaliphilic, microorganisms can live there. For example acidophilies
catalytic activity are most efficiently. microorganisms such as Acidianus, Desulfurolobus, Sulfolobus at pH 5
Thermophilies microorganisms in deep-sea volcanoes and hydro- and even below 1 can operate or alkaliphilies microorganisms such as
thermal vents are found that are capable of withstanding high tem- Natronococcus, Bacillus in conditions pH above 9 can actively live.
peratures even higher than 100 °C. Thermophilies enzymes derived 3.5% average salinity of sea water in the ocean, but in some areas
from microorganisms for their good performance at high temperatures, can reach more. A large number of halophiles microorganisms can
have great value in the biological sciences. For example, nucleic acid tolerate high salinity in areas with high salinity. Halophilic enzymes of
enzymes such as DNA polymerase, ligase and restriction endonuclease these microorganisms can be high levels of salinity in good shape and
are high-value applications in the molecular biology and genetics sci- optimal activity (David and Michel, 1999; Niehaus et al., 1999).
ence. Lundberg team DNA polymerase enzyme purified from thermo- A cold-active lipase isolated from bacterium Colwellia psychrery-
philic archaeo Pyrococcus furiosus, which is due to polymerization, thraea exhibited optimum activity around 25 °C and pH 7.0 (Do et al.,
reading errors and high activity at temperatures up to 100 °C, is widely 2013) and cold-active extracellular lipase isolated from psychotropic
used in polymerase reaction chain (Cowan et al., 1987; Huber et al., Yersinia enterocolitica exhibited activity at temperature ranging from
1986). 0 °C to 60 °C with optimum activity at 37 °C (Ji et al., 2014).
Non-specific nucleases isolated from a thermophilic bacteriophage
GBSV1 ability to destroy various nucleic acids, including RNA, single- 4. Production of proteases by marine microbes and their
stranded DNA and double-stranded DNA (Song and Zhang, 2008). Also industrial applications
marine bacterium Alteromonas espejiana BAL 31 is capable of producing
the enzyme endonuclease and exonuclease activity for single-stranded Proteases have been isolated from a large number of biological
DNA, which is great value in genetic engineering. Lipase isolated from sources including plants, animals and microbes that among them the
pseudomonas from marine sediments which have the highest tempera- microbes of great importance in various industries. For example, nearly
ture range 30 °C to 80 °C and pH range is from 5.0 to 9.0. The maximum two third of industrial and commercial proteases are produced by
activity for produced marine microbial lipase of 1980 U/mL, pH 7.5 marine microbial including fungi, yeasts and bacteria species (Wang
and 35 °C is the temperature (Ramani et al., 2013). et al., 2013a, 2013b).
Thermococcus genus of bacteria isolated from hydrothermal vents at Among Bacillus genus, especially Bacillus subtilis is a super source for
a concentration of 1–3% salt has been unable to work. Marine microbial protease production because of aptitude to secrete proteins into
Bacillus sonorensis 4R, Thermococcus sibiricus, Pyrococcus furiosus, medium and high growth rate (Joo and Choi, 2012).
Archaeoglobus fulgidus, Thermotoga sp., Pyrobaculus calidifontis, For the first time in 1960, alkaline protease isolated from Bacillus
Thermococcus celer and Sulfolobus sp. like S. solfataricus, S. acid- licheniformis. Then the enzyme from other marine microorganisms for
ocaldarius, and S. shibatae except hyperthermophilic that are lipase enzyme production in laboratory conditions was isolated. An alkaline
activity (Adams, 2004; Babu et al., 2008; Bhosale et al., 2016; Gantelet protease microbial obtained for the first time from Bacillus licheni-
and Duchiron, 1998; Renge et al., 2012). Species belonging to the genus formis in 1960 by Dane. Nobou Kato, 1972 isolated for the first time
Salinivibrio capable of producing thermostable lipases and activities for alkaline protease from marine psychrobacter (Zhang and Kim, 2008).
30 min at 80 °C and pH 7.5–8.0 and 0.1–3.0 M NaCl concentration is Qiu et al. selected 30 kinds of marine bacteria from the sea water,
tolerated (Dalmaso et al., 2015). Extracellular enzymes secreted from mud, fish and other samples; after UV mutagenesis they isolated the N1-

134
Table 4
Unique biochemical and catalytic properties of microbial proteases from marine organisms.

Species Optimum pH Optimum Km Activators Inhibitors Other properties References


A. Beygmoradi, A. Homaei

Temperature (°C)

Aureobasidium 4.0 28.0 (Li et al., 2007)


Pullulans
Bacillus subtilis DR8806 8.0 45 Ca2+, K+, Mg2+, Fe2+ and DMSOa Hg2+, Ba2+, Cu2+, Zn2+, H2O2, CTABb Molecular mass: 37 kDa (Farhadian et al., 2015)
and SDS
Purpureocillium lilacinum 4.0 to 9.0 65 DMSO, methanol, isopropanol, Molecular mass: 37 kDa (Cavello et al., 2013)
LPS # 876 surfactants (Triton X−100, sodium
dodecylsulfate and Tween 85)
Bacillus sp. high pH (10) 60 0.57 mg/mL Molecular mass: 71 kDa, (Jain et al., 2012)
Vmax: 445.23 U/mL
Halophilic strain SD11 10 60 EDTA, PMSFc (5 mM) Molecular mass: 45 kDa, (Cui et al., 2015)
Ca2+, Mn2+, Zn2+, Cu2+, Na+ and K+ Alkaline serine Protease
Bacillus sp. MIG 11–12 50–55 Ag+, Alkaline serine Protease (Gouda, 2006)
Hg 2+ and
PMSF
Bacillus firmus CAS 7 11 70 Mn2+, Fe2+, Co2+, Mg2+, Ca2+ and PMSF, EDTA (1 and 10 mM), Molecular mass: 21 kDa, (Annamalai et al.,
Hg2+ (0.1 and 1 mM) phenanthrolein, mercaptoethanol, DTNBd Specific activity: 473.4 U/mg 2014a, 2014b)
and SBTIe (1 and 5 mM)
Rhodotorula mucilaginosa 5.0 50 Extracellular acid protease, Halostable in (Lario et al., 2015)
L7 the presence of high concentrations of
NaCl (0.5–3.5 M)
Bacillus alveayuensis CAS 9.0 50 Mn2+, Fe2+, Co2+, Mg2+, Ca2+, Hg2+ PMSF, Thermostable, halostable alkaline (Annamalai et al.,

135
5 and EDTA (0.1 and 1 mM) mercaptoethanol, DTNB and SBTI (1 and protease, Specific activity: 518.78 U/mg 2014a, 2014b)
5 mM)
Marinobacter MBRI 7 7.0 40 Extracellular protease, (Fulzele et al., 2011)
halotolerant and high thermostabe
Aspergillus ustus 9.0 20 PMSF Serine protease, (Damare et al., 2006)
Heat-sensitive,
relatively more active than mesophilic
enzymes at a low temperature
Bacillus sp. APCMST-RS3 9.0 60 0.6666 g/l NaCl (2.5 M), surfactants (Tween 20, 40 PMSF, DTT, iodoacetamide, Haloalkalophilic. Protease, Maximum (Maruthiaha et al.,
and SDS), CaCl2, CuSO4, MnCl2, BaCl2 mercaptoethanol, EDTA (5 mM) activity in the presence of NaCl (2.5 M), 2015)
and MgCl2 (5 ppm) Molecular mass: 40 kDa,
V max: 1111.11 U/mL
Specific activity: 28.62 and 1.68 U/mg
Pseudomonas SD11 10 40 Ca2+, Mn2+, Zn2+, Cu2+, Na+, and K+ SDS, PMFS (each 5 mM) and H2O2 (1% v/ Thermostable alkaline serine protease, (Cui et al., 2015)
(each 5 mM) v) Protease activity: 171.9 U/mL
Bacillus sp. 60 (Doddapaneni et al.,
2009)
Bacillus mojavensis A21 60 (Haddar et al., 2009b)
Bacillus firmus CAS 5 9 50 Mn2+, Fe2+, Co2+, Mg2+, Ca2+, Hg2+ PMSF, EDTA, mercaptoethanol, DTNB, Thermostable and halostable alkaline (Annamalai et al.,
and EDTA (0.1 and 1 mM) SBTI (1 and 5 mM) protease 2014a, 2014b)

a
Dimethylsulfoxide.
b
Cetyl trimethyl ammonium bromide.
c
Phenyl methyl sulfonyl fluoride.
d
Dithio-bis-nitrobenzoic acid.
e
Soybean trypsin.
Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

35 strain, this strain produced protease that had significant advantages producing the collagenase is widely used in commercial industries
compared with the terrestrial ones (Graham et al., 1980). (Table 4).
Yeast Aureobasidium pullulans has a high efficiency alkaline protease
and the highest enzyme production with a value of 623.1 U/mg (7.2 U/ 5. Biochemical properties and biological functions of marine
mL) protein (Chi et al., 2007). Haddar and colleagues in 2009, alkaline microbial lipases
protease from marine bacterium Bacillus mojavensis A21 isolated, they
have two stable detergents alkaline serine – protease (BM1 and BM2) Lipase or acylglycerol acylhydrolases are ubiquitous enzymes that
were purified from this strain. Both proteases for nonionic surfactant ability to hydrolyze esters of long-chain aliphatic acids from their gly-
showed high stability. cerol (Jensen, 1983; Shahidi and JanakKamil, 2001). Lipase break-
In fact, both of them excellent stability with a wide range of liquid downs triglycerides into fatty acids and glycerol. Among the variety of
and solid showed great detergents (Haddar et al., 2009b). various lipase, including plant, animal, bacterial, microbial lipases because of
marine microorganisms, including bacteria, fungi, yeast, etc., that are the ease culture and wide as a catalyst in many reactions and synthetic
capable of producing a novel alkaline protease enzyme that in between hydrolytic most commercial applications (Saxena et al., 1999). Micro-
them yeasts including Aureobasidium pullulans (Chi et al., 2007), Can- bial lipases are also non-toxic and adaption with environment
dida olea (Nelson and Young, 1987), Yarrowia lipolytica (Matoba et al., (Boonmahome and Mongkol Thanaruk, 2013). Lipase in various in-
1988) and bacteria including Bacillus sp. (Beheshti Maal et al., 2009; dustries such as dairy, food, detergent, textile, pharmaceutical and
Nadeem et al., 2008; Sarethy et al., 2011), Bacillus mojavensis A21 cosmetics used (Patnala et al., 2016).
(Haddar et al., 2009a), Bacillus mojavensis (Khalil Beg and Gupta, 2003), The first microbial lipases in 1935 by Kirsh were found in fungi
B. stearothermophilus F1 (Rahman et al., 1994), Pseudomonas aeruginosa Aspergillus flavus and Penicillium Oxalicum (David, 1935). Bacteria
MN1 (Bayoudh et al., 2000), Brevibacterium linens ATCC 9174 (Rattray Achromobacter sp., Alcaligenes sp., Arthrobacter sp., Chromobacterium sp.,
et al., 1994) and fungi, including Aspergillus, Mucor and Rhizopus Staphylococcus sp. and Pseudomonas sp. are able to produce lipase.
(Charles et al., 2008; Devi et al., 2008; Ghasemi et al., 2011; Kuberan Major producers of fungal lipases including Aspergillus niger,
et al., 2010) the main manufacturers of these enzymes. The first time Rhizopus delemar, Rhizopus japonicus, Rhizopus niveus and Rhizopus or-
protease-resistant to heat (thermophilic) and high pressure (barophilic) yzae, Candida cylindracea, Humicola lanuginosa, Mucor miehei, M.pusillus
were isolated from Methanococcus jannaschii (Michels and Clark, 1997). of thermophilic as a manufacturer of heat-resistant extracellular lipase
Protease isolated and purified from archeo Pyrococcus abyssi is re- known. For extracting fungal lipases low cost, resistance to pH and
sistant to high temperatures (Dib et al., 1998). New extracellular serine temperature, substrate selectivity and activity in organic solvents as
proteinase of marine archeobacter Aeropyrum pernix identification has a compared to bacterial lipases more. Most bacterial lipases non-selective
half-life of 85 min in temperature 100 °C and 12 min in temperature in choosing your substrate and few of them are also resistant to heat.
110 °C (Chavez et al., 1999). According to research review paper Physical parameters such as catalytic activity, pH, temperature etc.
(Kumar and Takagi, 1999) optimum temperature (°C) of some proteases affect the rate of lipase production (Table 5). Lipase production rate in
in marine microbes including Penicillum griseofulvin is 28 °C (Dixit and different species, different strains of the fungus is reported, for example,
Verma, 1993), Bacillus sp. B21-2 is 30 °C (Fujiwara and Yamamoto, Fusarium sp., the optimal pH and temperature are 2.5 and 30 °C re-
1987), Aspergillus flavus is 32 °C (Malathi and Chakraborty, 1991), Ba- spectively, while in bacterial species such as Bacillus subtilis, the op-
cillus sp. Y is 35 °C (Shimogaki et al., 1991), Bacillus licheniformis is timum temperature and optimum pH is 30 °C and 7.0, respectively
36 °C (Mao et al., 1992), B. alcalophilus subsp. halodurans KP1239 is (Pallavi et al., 2014).
37 °C (Takii et al., 1990), Bacillus licheniformis is 39.5 °C (Hübner et al., In 2009, Mo and colleague isolated and purified a new type extra-
1993), Bacillus sp. strain B18′ is 40 °C (N. Fujiwara et al., 1993), Bacillus cellular phospholipase C from 400 a marine streptomycete that was se-
thermoruber BT2T is 45 °C (Manachini et al., 1988), Thermo- lected from approximately 400 marine bacteria. This enzyme has an
actinomycetes sp. HS682 is 52 °C (Tsuchiya et al., 1992), B. stear- optimal activity at pH 8 and temperature of 45 °C and cause the hy-
othermophilus AP-4 is 55 °C (Dhandapani and Vijayaragavan, 1994), B. drolysis of phosphatidylcholine (Mo et al., 2009). It is reported that
stearothermophilus F1 is 60 °C (Rahman et al., 1994), Aspergillus oryzae lipase production of Burkholderia cenocepacia ST8 increased in presence
MTCC 5341 is 55 °C (Vishwanatha et al., 2009). Extracellular proteases peptone as a nitrogen source (Ananthi et al., 2014). In 1935, the first
from marine bacteria Sphingomonas paucimobilis isolated from stomach isolated lipase from Penicillium oxalicum and Aspergillus flavus (Zhang
south polar krill Euphausia superba is purification and identification and Kim, 2008). The production of lipase in the thermophilic bacterium
(Turkiewicz et al., 1999). Bacillus sp. is higher when olive oil is used as carbon source. Other
Vibrio spp. is capable to produce all kinds of extracellular proteases. studies have shown that the presence of simple sugar glucose in the
V. alginolyticus able to produce 6 types serine protease-resistant de- medium inhibited catabolic enzyme production is through suppression
tergents unusual and alkaline serine exoprotease. The marine bacteria (Sarethy et al., 2011). A small number of bacterial lipase is resistant to
capable of producing the enzyme collagenase is widely used that this high temperatures. Among the microbial species Achromobacter sp.,
very useful in cosmetic production and commercial industries (Alemán Alcaligenes sp., Arthrobacter sp., Pseudomonas sp., Staphylococcus sp., and
and Martínez-Alvarez, 2008). Chromobacterium sp. are able to produce lipase (Jaeger and Eggert,
One of the most important enzymes is proteases that more than 60% 2002). This means that marine resources are bacteria extremophiles
of the total industrial and commercial industrial enzymes in the world tolerate contain data extreme conditions of temperature, pH, salinity,
are included. In modern society, proteases widely in various industries acidic or alkaline, etc. According to studies, bacteria belonging to the
including leather, industrial detergents, alcohol and beer, confectionery genus pseudomonas are able to produce lipase resistant to high tem-
and pharmaceuticals such as digestive, physiological activities in the peratures, high enantiomer sensitivity and maintain its activities in a
body, nutraceutical activity and anti-inflammatory drugs are the most wide range of kinetic parameters such as temperature and pH (Patnala
used (Alemán and Martínez-Alvarez, 2008; Kumar and Takagi, 1999). et al., 2016). Bacterial lipase from Pseudomonas otitidis of marine se-
Proteases are classified on the basis of their differences in optimal diments activity in the temperature range 30 °C to 80 °C and pH range is
conditions for specific activity, substrate, pH, temperature, stability, from 5 to 9 and maximum lipase production in pH and temperature
active site and catalytic mechanisms have been (Charles et al., 2008). have been reported at 7.5 and 35 °C respectively. It is reported that
Marine bacteria produce chemicals and enzymes are valuable source. Thermococcus genus of bacteria that have been isolated from hydro-
Proteases produced in some marine bacteria used in detergents and thermal lake's ability to produce lipase. In addition, the bacterium
cleaners. Vibrio alginolyticus produced protease 6 types that one of Thermococcus sibiricus obtained from the oil reservoir Samotlor (West
which is resistant to detergents. Also this marine bacterium capable of Siberia) is containing 15 genes to produce lipase / esterase. Other

136
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

bacteria that have been are reported include lipase activity Archae-

(Balakrishna et al., 2017)


oglobus fulgidus, Pyrobaculus calidifontis and Sulfolobus sp. S. solfataricus,

(Basheer et al., 2011)


(Bhosale et al., 2016)

(Ramakrishnan et al.,
(Sathishkumar et al.,

(Hardeman and Sara


(Vyas and Chhabra,
S. acidocaldarius and S. shibatae (Renge et al., 2012). Cold-active lipase

(Diaz et al., 2006)


(Cho et al., 2007)
(Lan et al., 2016)
from Colwellia psychrerythraea isolated from marine environment was

Sjoling, 2007)
References

observed at pH and temperature 7.0 and 25 °C, respectively (Do et al.,


2013). Cold-active extracellular lipase isolated from marine bacteria
2015)

2016)

2017)
Yersinia psychotropic showed an operating temperatures ranging from
0 °C to 60 °C, that maximum lipase production was observed at tem-

Lipase activity: 185 and 220 U/mL at 24 and


perature is 37 °C (Ji et al., 2014). One of the key factors in increasing
Optimal medium constituents: tuna powder
Maximum lipase activity: 1355.81 U/mL,

the production of lipase is metal ions. The most desirable metal ions
(14.58 g/l), olive oil (5.05 mL/l), NaCl

Ca2+ and Mg2+ are used to produce lipase. The lipase activity by these
Hyperthermostable alkaline lipase,

ions increases and partly by Cu2+, Al3+, Zn2+, Ba2+, Pb2+, Fe2+ and
Medium stability alkaline lipase,

Low-temperature-active lipase,
Mn2+ inhibited the activity of the bacterial lipase cold Pseudoalter-
Molecular mass: 21.87 kDa

omonas sp. NJ 70 isolated from Antarctic ice has been demonstrated

Molecular mass: 35.4 kDa


Molecular mass: 44 kDa

Molecular mass: 19 kDa


(Huang et al., 2004). Extracellular phospholipase C, isolated from the
Thermostable lipase,

Curcin and trypsin


marine Streptomycete is able to decompose phosphatidylcholine with
Other properties

the highest enzyme activity at a temperature of 40 °C and pH 8 (Zhang


(72.42 g/l),

and Kim, 2010). Bacteria that grow on the whales are a rich source of
lipase and esterase enzymes that are used in industry huge (Wells,
22 °C

1999). Aeromonas sp. EBB-1-a from sea mud is able to produce extra-
cellular lipase in medium heat with maximum lipase activity after 15 h
PMSF, orlistat, oleic acid, iodine, EDTA,

at temperature 25 °C and pH 8.0 in the presence of olive oil 0.5% (v / v)


CoCl2, CdCl2, HgCl2, CuCl2, Pb(NO3)2,

as the carbon source (Charoenpanich et al., 2011). Nearly to 2000


PMSF, EDTA (each 1, 5 mM) and

species fungi identified that about 600 species are found in the seas.
Since marine fungi resistance is more than physical and biological
DEPC, PMSF, EDAC and NBS
mercaptoethanol (1, 5 mM)

conditions, thus making them a good choice for the production of


secondary metabolites in front of their terrestrial counterparts. In
general they are more tolerant to physicochemical parameters such as
temperature, pH, salinity, and other conditions are associated with
ground fungi. Until now, researchers have found a wide range of mi-
Inhibitors

and urea

croorganisms that can produce lipase, including fungi such as Peni-


cillium, Aspergillus, Rhizopus, Scopulariopsis, Trichoderma, Mucor and
bacteria such as Pseudomonas, Bacillus, Staphylococcus, Aeromonas
(each 0.1, 1 mM), Tween 80, Triton X 100, SDS and

Ethanol and xylene, acetone, propanol and hexane

(Ananthi et al., 2014; Arora, 2013; Charoenpanich et al., 2011; Cho


MgSO4 (0.1 mM), CaCl2, MnSO4, CoCl2, HgCl2

et al., 2007; Ramani et al., 2013).


Lipases are important biocatalysts that break down fat and oil major
release of free fatty acids, and glycerol are the catalyst role. On the
Sodium deoxycholate (each 1,10 mM)

other hand the lipase in a variety of reactions such as esterification,


transesterification and aminolysis play an important role in industrials
Characterization and enzymatic properties of lipases as practical biocatalysts from marine microbes.

(Babu et al., 2008). Biocatalyst potential microbial lipase in aqueous


and non-aqueous as well as good resistance in alkaline environments
and high temperatures Lead to wider use in the broad fields of bio-
MgSO4 and CaSO4

technology, especially dairy, detergents, drugs, chemicals, agricultural


chemicals, oil is the synthesis of fatty substances (Mustranta et al.,
Activators

1995). Microbial lipases are widely used in commercial products and


the majority in diverse industries including food, pharmaceutical, cos-
metic and agricultural has many applications (Chi et al., 2009;
Kobayashi et al., 2008). Lipase biological catalysts are valuable because
they have done little in terms of moderation and stability is high in
Temperature (°C)

organic solvents (Davidson, 2006). According to Koop and Griffiths


these creatures can live for about 70% of organic matter and minerals.
Optimum

A lot of organic matter in marine ecosystems of compounds with high


molecular weight polymeric structure, mainly protein, starch, fat,
45

80

22

40

28

37
35

20
40
40

pectin, cellulase, chitin, nucleic acids, or lignin is formed (Arnos et al.,


Optimum pH

1998; PO Remba, 1995; Unanue et al., 1999). They must extracellular


enzymes break down into simpler compounds (Unanue et al., 1999).
Hence, the measurement and characterization of enzymes is a useful
10.8
9.0

6.0

6.0

8.0
8.0

6.0
7.0
7.5

tool to check the destruction of organic matter and nutrient cycling in


9

aquatic ecosystems provide (Boetius, 1995; Jackso et al., 1995). Lipases


Halobacillus trueperi RSK

Penicillium chrysogenum
Streptomyces sp. MAS1

Rhizopus homothallicus

are widely used in commercial products, biotechnology, organic


Bacillus sonorensis 4 R

Enterococcus faecium

Candida parapsilosis

Aspergillus awamori
uncultured bacteria

synthesis and some other industrial applications (Kempka et al., 2008).


oligophagum
MTCC5695

Due to the ease of recovery of the enzyme from the culture medium of
Cystobasidium

h1Lip1

microorganisms such as bacteria, fungi, yeasts, and actinomycetes are


CAS9
Species

known as extracellular lipase resources. Species of Bacillus, Acromo-


Table 5

bacter, Acinetobacter, Candida, Pseudomonas, Rhizopus, and Geotrichum


are known as the main commercial species (Ertugrul et al., 2007). Some

137
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

40 species are isolated of Aspergillus and Penicillium to produce lipase epimer.


broth agar medium containing olive oil as a substrate (Yadav et al., These residues are like the building blocks of homopolymer poly-M
1998). Also, many microbial lipases are very useful for heart disease (ManA) or poly G (GulA) extend sequences were created by mixing the
prevention role (Kinsella, 1986; Strom and Raa, 1993). The beneficial Heteropolymeric MG (Yamasaki et al., 2004). Based on the primary
effect is produced of polyunsaturated fatty acids (PUFA), acid eicosa- structure of the enzyme alginate lyase divided in three families with
pentaenoic (EPA), acid docosapentaenoic (DPA) and acid docosahex- names PL-5, 7, 14 (Hisano et al., 1993).
aenoic (DHA). Lipases used for commercial and industrial applications The family, in particular PL5 and PL-7 made of poly-M but PL- 14
mainly from bacterial sources. They belong to the family of serine hy- contains poly G or M (Yamasaki et al., 2004). Commercial alginate,
drolases include α / β hydrolase are in their original structure. Enzy- often with the M: G defined. Pseudomonas aeruginosa is a pathogen
matic activity of bacteria isolated from marine sediments off the opportunistic in immunocompromised hosts caused disorders including
southern coast of the Baltic, looked at Sopot beach (Mudryk and sepsis, urinary tract and respiratory tract. P. aeruginosa bacteria secrete
Podgórska, 2006). Finally, it should be noted that bacteria play a key excessive amounts of exopolysaccharide alginate, mucoid colonies
role in the processes of decomposition of organic matter accumulated in (Albrecht and Schiller, 2005).
the marine coast (Jedrzejczak, 1999). Alginate as a member of the cell wall, the brown seaweeds and some
bacteria belonging to the genus as exopolysaccharide by Azotobacter
6. Research and application of marine microbial cellulase and Pseudomonas are made. Alginate polymer is one of the most im-
portant members of biofilm of P. aeruginosa strains of the mucoid co-
Cellulose is a polysaccharide with the formula (C6H10O5) n the first lonies to be considered.
biopolymer abundant in nature. Cellulase about 50% of all plant ma- Unlike alginate which is made by algae, the bacteria produce a
terial and hemicelluloses is a polysaccharide related to cellulose make polysaccharide that most of the acetyl group in position D-mannuronate
up about 20–30% (Klemm et al., 2005; Nishiyama et al., 2002). Cel- is two or three units. Additionally, some of the references that are made
lulase enzymes that break down cellulose and polysaccharides it turn of alginate by Pseudomonas G have no production (Yamasaki et al.,
cellodextrin younger. The polysaccharide is consisting of a linear chain 2004). Type of monomer comprising alginate and alginate acetylation
with several unit D-glucose β (1 → 4). Hemicelluloses is a poly- determines sensitivity to it is destroyed (Yamasaki et al., 2004).
saccharide related to cellulose with shorter chains of sugar is about 200 Alginate lyase enzyme linked alginate glycosides by reacting beta
units, and unlike cellulose, it can be a few simple sugars such as xylose, unsaturated acid oligouronic breaks and at the end of the non-renewal
mannose, galactose and rhamnose, etc., derived (Nishiyama et al., of the production (Yamasaki et al., 2004). Related illnesses due to in-
2002). Since the connection is tightly between cellulose molecules fection of pseudomonas biofilm, the bacteria can be Fibrous cystic
thereby breaking the cellulose is relatively hard compared to other chronic lung infections and urinary tract infections associated with
polysaccharides (Brás et al., 2008). catheters pointed (Dunne and Buckmire, 1985; Henrissat et al., 2005; Li
Until now, researchers have found a wide range of marine microbes et al., 2011).
belonging to Cytophaga (C. diffluens، C. hutchinsonii), Marinobacter sp. Especially poly-M alginate acetylated form of the enzyme that de-
MS1032, Cellulomonas, Vibrio, Paenibacillus sp. BME-14, Clostridium, stroys is more significant (Yamasaki et al., 2004). Since few of alginate
Nocardia, Streptomyces, Trichoderma, Aspergillus, Fusarium, Chaetomium, lyase enzymes have this property (Hoshino and Kageyama, 1980;
Phoma, Sporotrichum, Penicillium, etc., which can generate cellulase Yamasaki et al., 2004). Thus, one of the striking points for researchers
(Dong et al., 2010; Fu et al., 2010; Vaidya et al., 2001; Zhang and Kim, to find alginate lyase with this feature is to end the destruction of
2008). bacterial biofilm is used.
Marine bacteria coexist in the gland of Deshayes cream naval ships Alginate-lyase from the marine bacterium Bacillus sp.,
and Aspergillus terreus are capable of producing cellulase. The thermo- Photobacterium and Alteromonas sp. symbiotic with Laminaria known
philic bacterium Thermotoga sp. is producing exo-1, 4- β-cellobiohy- isolated and recognition (Sawabe et al., 1997).
drolase at temperature 105 °C. According to studies reported that Alginate is a polysaccharide found in wide algae and marine and
Bacillus sp. associated with the marine sponge Axinella sp. is capable of terrestrial microorganisms. This combination of useful properties such
producing carboxy methyl cellulose (Mohapatra, 1997). as having high biocompatibility, biodegradability and, low toxicity and
Marinimicrobium sp. LS-A18 is capable of producing carbox- properties of gel formation, etc., that widely used in various industrial
ymethylcellulose with maximum activity in the temperature and pH and medical biomaterials (Gomez d′Ayala et al., 2008; Wong et al.,
55 °C and 7.0, respectively (Zhao et al., 2012). 2000; Yang et al., 2011). These enzymes have many biotechnology
Cellulase can be used in various industries including the construc- applications in various industrial such as food, cosmetics, agricultural
tion of alcohol, flavors food, paper, corn gluten, detergents, sewage and medical fields and are useful in the conversion of biomass brown
treatment, bio-textile auxiliaries, cotton, flax and bio-fertilizers pro- algae to methane (Gacesa, 1992; Rasmussen and Morrissey, 2007;
cessing (Shanmughapriya et al., 2010). Since the use of seaweed in the Wong et al., 2000). Alginate lyase activity as a therapeutic aid in the
industry can create very serious problems of environmental pollution treatment of infections caused by P. aeruginosa mucoid colonies and the
from the use of cellulase is useful to reduce waste and can easily be destruction of the biofilm can be used (Bugli et al., 2013; Laemmeli,
absorbed by the plants as bio-fertilizer. 1970). Moreover, alginate lyases removed alginate from the cell surface
mucoid colonies of Pseudomonas and inhibit the attachment of P. aer-
7. Investigation and applications of marine microbial alginate uginosa strains are mucoid colonies (Yamasaki et al., 2004). Alginate in
lyases combination with specialized additives such as chitosan, fibrin, col-
lagen and etc, to get the right combination to produce ideal for a variety
Brown algae are one of the biggest sources of marine biomass that of wounds, including chronic wounds dressing used (Willi and Sharma,
alginate has been made and it has many applications in various in- 2004).
dustries including food, pharmaceutical and so on. In recent years, huge
sea-microbial alginate lyases have been developed. 8. Importance and properties of chitinolytic enzymes isolated
Alginate lyase is linear polymer (1, 4) glycuronans that has been from marine microbes
established of residues β-D-mannosyluronic acid (M) or Apymran α-L
gulosyluronic acid (G). Alginate lyase in C5 is a β-D-mannuronate Chitin is a biopolymer with non-toxic that after cellulose is one of

138
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

the most common polysaccharides found in nature. Most of the com-

(Fu et al., 2016)


ponents of fungal cell wall structure and also one of the most abundant

(Fenice et al.,
(Gaber et al.,

(Wang et al.,
(Farag et al.,

(Yang et al.,
References
in the outer shell of insects and crustaceans such as crab, shrimp, lob-
ster and squid (Table 6).
2016)

2016)

2016)

2014)

1998)
Chitin with chemical formula (C8H13O5N) n and linear chain of
was analysed by incubating 1 mM of substrate with 1 μM of enzyme in acethylglucosamine groups and chitosan with chemical formula

The kinetic parameters, Km and Vmax values of PbChi70 for colloidal

Km and Vmax values of PbChi67 for colloidal chitin and glycol chitin
Purified by 65% ammonium sulphate precipitation, followed by gel

chitin and regenerated chitin were determined to be 7.91 mg mL−1

were 3.35 mg mL−1 and 17.1 mol/min/mg, and 2.66 mg mL−1 and
(C6H11O4N) obtained from residues removal of acetyl glucosamine.
Activity on soluble fully acetylated chitooligosaccharides (A2– A6)

filtration on Sephadex G−100 and DEAE-Sephadex A−50 ion Chitin with the scientific name β- (1 → 4) - 2-acetamido - 2 doxy - D-
glucopyranose and chitosan with the scientific name β- (1 → 4) - 2-
amino - doxy 2 - D- glucopyranose.
Chitin and chitosan history dates back to the 19th century for the
and 28.4 lmol min−1 mg−1, and 14.2 mg mL−1 and
first time in 1811 a French scientist named Braconnot chitin extracted
Product formation was analysed using HPLC.

from fungi. Then Rouget in 1859, chitosan deacetylation of the chitin in


the presence of potassium hydroxide won the game and finally in 1950
was to fully explore its structure (Khor, 2001).
21.8 lmol min−1 mg−1, respectively

High catalytic activity, even at 4 ◦C

Postoperative hydrolase, chitin and chitosan can be high perfor-


Marine psychrophilic bacterium.

15.0 mol/min/mg, respectively


mance, safety, and remove toxins from the body's digestive function
improved, even inhibiting tumor growth and other physiological
exchange chromatography

functions important to make (Ngo et al., 2008). Therefore, chitin and


chitosan hydrolyzate recently has become a hot topic and importance.
Other properties

Until now, it was found that marine microbes can produce chitinase
10 mM BisTris

and chitosanase, including Vibrio fluvialis, Vibrio parahaemolyticus,


Vibrio mimicus, Vibrio alginolyticus, Listonella anguillarum and Aeromonas
hydrophila, Aspergillus, Alteromonas, Penicillium, Rhizopus, Trichoderma,
Myxobacter, Sporocytophaga, Bacillus, Enterobacter, Klebsiella,
Pseudomonas, Serratia, Chromobacterium, Clostridium, Flavobacterium,
Co2+, Fe3+, and Mn2+
ethanol, methanol and

Arthrobacter and Streptomyces (Fukamizo, 2000; Osama and Koga, 1995;


Fe2+, Cu2+,Zn2+,

Tsujibo et al., 1992; Xia et al., 2008).


Hg+, pb, EDTA,

Chitinase genes have been cloned variety of marine fungi and bac-
Inhibitors

teria. Suolow and Jone chitinase genes including ChiB and ChiA in-
acetone

jected into E. coli and these genes transferred into Pseudomonas; finally
obtained 4 chitinase strains with high efficiency (Suolow and Jones,
1988). Meanwhile, Roberts and Cabib, chitinase genes into plant cells
Na+, Mg2+, K+, and

tobacco and are able to develop a new tobacco plant with strong disease
Metal ions such as
Ca2+, Mn2+ and

tolerance to the pathogen Alternaria Longipes (Roberts and Cabib,


1982).
Activators

Chitin and chitosan could be hypoallergenic and antibacterial


Na2+

Ca2+

properties promote digestive function and eliminate toxins from the


body, even inhibit tumor cell growth and great importance in medicine
functions and food industry (Fukamizo, 2000; Ngo et al., 2008).
Molecular mass (kDa)

67.0 kDa by SDS-PAGE

In addition, bio biological properties such as adhesion, anti-cancer,


and 67.9 kDa by gel
Novel features of chitinases from various marine microbes and their biochemical parameters.

anti-microbial, reduce inflammation and pain, antioxidants, choles-


terol-lowering and blood coagulant, have distinguished them from
other biopolymers (Kumar et al., 2004; Kurita, 2006).
filtration

Moreover, fibers made from chitin and chitosan absorbable sutures


113.5
60

70

for preparing and supplying fabrics that are highly effective for wound
healing (Jenkins and Hudson, 2001), chitin and chitosan can be used
for wastewater treatment, heavy metal ions, tissue engineering, anti-
Temperature (°C)

oxidant, anticancer, burn and wound healing, drug delivery and re-
ducer cholestrol (Kumar et al., 2004; Madhumathi et al., 2010; Seol
Optimum

et al., 2004).
Cloned and functionally expressed in Escherichia coli.
(4–30)

28.0

For example, Weltroswki and colleagues were able to use chitosan


45

50

55

20

60

derivatives in an acidic solution to remove metal ions from waste water


Optimum pH

(Weltrowski et al., 1996). A mixture of chitin and chitosan is used to


absorb arsenic in drinking waters (Elson et al., 1980). Chitosan because
of very similar structure of cellulose can be easily factories, paper use
6.0

5.6

5.5

8.0

3.5

4.0

and paper production from chitosan has a smooth surface and high
resistance to moisture much for printing and painting are suitable.
Pseudoalteromonas sp. DL−6
Paenicibacillus barengoltziia

Unlike most chitosan hydrogel properties that are anionic, cationic


Paenicibacillus barengoltzii

property that causes the same features as a preservative in cosmetics


Aspergillus terreus (EC

applied to the skin and hair. Chitosan with many compounds used in
Thermobifida fusca

janthinellum P9

cosmetics, adapted to absorb ultraviolet rays and many or reduce their


(PbChi70)
3.2.1.14)

CAU904

impact (Dutta et al., 2004). Chitosan and hair are complementary in


Penicillium
Species

terms of electric charge, as positively charged chitosan and negatively


(ChiA)
Table 6

charged hair, and a transparent solution of chitosan in the form of a


a

flexible cover is placed on the skin and hair and enhances the softness

139
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

and freshness it. Chitin and chitosan both in shampoos, hair dyes,

(Marokhazi et al., 2004)


(Rochima et al., 2016)

(Petrova et al., 2006)


emulsifiers, hair spray and amplifiers are used (Dutta et al., 2004).

(Lima et al., 2009)


Chitosan is used as a dental filling material that can kill fungi on the
surface of the teeth (Sapelli et al., 1986).
References

Chitosan has all the characteristics that should ideally be a lens such
as mechanical stability, cabn high permeability to air, especially
oxygen, moist, antimicrobial and biocompatible is being so widely In
ophthalmology for the preparation of contact lenses (Jing et al., 1996).
The substrate collagen derived of Tilapia fish skin

Chitosan is abundant in the crustaceans and crust of insects that widely


used in bone tissue engineering (Seol et al., 2004). Report on the use of
chitosan composite scaffold of calcium phosphate to be published -More
strength (Lian et al., 2009). Also, chitosan recover and rebuild cartilage
Collagenase activity: 1298 Unit mL–1

(Yamane et al., 2005), neurons (Freier et al., 2005), and liver plays an
important role (Wang et al., 2003). Also chitosan has anti-acid activity
that prevents reducing the effects of drugs in stomach (Miyazaki et al.,
1981). Chitosan-based polymer systems that have been built in the
transfer and release of proteins/peptides (Grenha et al., 2005), growth
Other properties

factors (Elcin et al., 1996), inflammatory pain medications (Aggarwal


et al., 2001), anticancer drugs (Wei et al., 2010), antibiotics (Kavaz
et al., 2010) and also are used in the treatment of genetic incompetence
waste,

(Mansouri et al., 2004).

9. Characteristics and applications of marine microbial


carrageenases
Cu2+, Zn2+, Hg2+ and

Carrageenan is family of linear sulfated polysaccharides that ex-


Mn2+ and Ca2+

tracted from marine red alga Delesseria sanguinea. In early 1943 by


Inhibitors

Mori, carrageenases extracted from marine mollusk. Sarwar et al. from


the culture medium cytophaga lk-C783, κ-carrageenase molecular
Fe2+

weight of 10 kDa extracellular won (Sarwar et al., 1987a, 1987b). In


2004, Mou et al. first isolated a new type of extracellular κ-carrageenase
Mg2+, Ca2+ and Ba2+

from Cytophaga MCA-2 from marine environment that molecular


weight determined 30 kDa (Renner and Breznak, 1998). In 2006, Yu-
Zn2+ and Co2+

kari and Yuji; λ-Carrageenan purified from bacteria Pseudoalteromonas


Activators

CL19 isolated from sediments deep-sea that molecular weight of ap-


Some properties of biophysicochemical, activators and inhibitors of collagenases from different marine microbes.

proximately 100 kDa (Ohta and Hatada, 2006). Bacterial species Pseu-
domonas, Pseudomonas elongata, Cytophaga, Alteromonas atlantica, Al-
teromonas carrageenovora are able to produce carrageenases
Molecular mass (kDa)

(Khambhaty et al., 2007; Potin et al., 1991; Roberts et al., 2007; Sarwar
et al., 1987a, 1987b).
Carrageenanases in industrial processing have many advantages.
The potential applications of carrageenanases enzymes have been re-
116. 97

ported by several authors. This enzyme can be used in food industry,


pharmaceuticals and cosmetics. For example, their potential applica-
74

tions for seafood processing are coagulant, adhesives, stabilizers and


Optimum Temperature (°C)

emulsifiers and their potential applications for medical processing are


anti-viral, anti-bacterial, anti-tumor, anti-coagulation, etc. (Roberts
et al., 2007; Sarwar et al., 1987a, 1987b).

10. Properties and applications of marine microbial xylanases

Xylanases is an enzyme that catalyzes the breakdown xylan.


50

45
37

Xylanases can produce by several microorganisms, including fungi,


bacteria, yeast and seaweed that the main commercial source of the
Optimum pH

enzyme is filamentous. Indian researchers showed alkaline xylanase


5 to 10

activity in several marine fungi. Aspergillus niger is able to produce


xylanase with high biological activity (Raghukumar et al., 2004).
8.2
7.5

7.0

Streptomyces viridochromogenes, Streptomyces M11, Trichoderma long-


Candida albicans URM3622
Bacillus subtilis ATCC 6633

ibrachiatum, Thermotoga maritima, Aureobasidium pullulans, Glaciecola


Photorhabdus luminescens

mesophila are able to produce xylanase (Guo et al., 2013; Jiang et al.,
Streptomyces strain 3B

2008; Jiang et al., 2004; Liu et al., 2014). Yin et al., 2010, Bacillus sp.
producing xylanase was isolated from seawater., and they purified this
enzyme in the optimum temperature and pH at 50 °C and 5, respec-
Species

tively (Yin et al., 2010). Xylanase is widely used in production of nat-


Table 7

ural sweeteners, dough-softening ability and paper and pulp industry


(Guo et al., 2009; Subramani and Aalbersberg, 2012).

140
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

11. Marine microbes as a potential source of collagenase enzymes Three structure-activity of superoxide dismutase (iron, copper/zinc
and an anonymous form) showed in marine Cyanobacterium synecho-
Collagen is a major protein of skin, ligaments, tendon, bones and coccus sp. (Chadd et al., 1996). Superoxide dismutase was isolated from
elastic tissues in the more organisms that composed of glysin, proline marine yeast Debaryomyces hansenii (Hernandez-Saavedra and Ochoa,
and hydroxyl proline. 1999).
Collagenolytic enzymes including cathepsin and elastase are cap- Novel glucose dehydrogenases were isolated of Cytophaga marino-
able of degrading collagen (Table 7). flava (Tsugawa et al., 1996). Glucose dehydrogenase reacts under high
Collagenase enzymes isolated and described from organisms in- salinity. New type glucose dehydrogenase was purified of marine Gram-
cluding microbes and animals (Daboor et al., 2010). Microbial col- negative bacterium Deleya sp. (Tsugawa et al., 1998).
lagenases have been obtained from microorganisms including Asper- Nitrite reductase purified by electrophoretic homogeneity of nitrite-
gillus oryzae, Clostridium histolyticum, Streptomyces parvulus, Streptomyces free extract of a marine bacterium Pseudomonas nautical (Besson et al.,
sp., actinomycete and etc. (Endo et al., 1987; Goldberg et al., 1986; 1995).
Goshev et al., 2005; Nordwig and Jahin, 1968). Pseudomonas doudoroffii strains are able to produce dimethylsulfo-
Isolated collagenases from bacteria are capable of hydrolyzing both niopropionate lyase (De Souza and Yoch, 1995).
water-insoluble native collagens and water-soluble (Mookhtiar et al., Carpenter et al. showed that marine cyanobacteria cells
1985). Trichodesmium thiebautii are able to produce Glutamine synthetase
Harper et al. (1965) reported that collagenases isolated from Clos- (Carpenter et al., 1992). Cyanobacteria Prochlorococcus sp. are able to
tridium histolyticum had molecular weights ranging from 57 to 105 kDa produce glutamine synthetase. Unusual responses were analysed and
(Harper et al., 1965). adapted to the darkness and famine mechanism of nitrogen from Pro-
In 1984 by Bond and Van Wart; six different collagenases isolated chlorococcus sp. for coping with the environment reflect light and nu-
from the same species with molecular weights ranging from 68 to trient limited (El Alaoui et al., 2001).
128 kDa (Bond and Van Wart, 1984). Afterward, collagenases isolated
from Clostridium perfringens with molecular weights ranging from 80 to 14. Characteristics and biotechnological importance of marine
120 kDa (Matsushita et al., 1994). microbial amylase
According to studies collagenases obtained from bacterial have
molecular weight > 55 kDa while molecular weights of collagenases Amylases were found in bread and can break down starch into
obtained from animal tissues tend to be lower (Daboor et al., 2010). simple sugars like compound, such as glucose, maltose and dextrin.
For the first time, collagenase was discovered from Clostridium They can classified be β -amylase, α-amylase and γ-amylase. Unlike
histolyticum. Collagenase enzymes have a several industrial applications other forms of amylase, γ-amylase is most effective in acidic environ-
in foods, medicinal products and cosmetics (Chung et al., 2004; Shahidi ments. So far, the production of amylase was reported in some microbes
and JanakKamil, 2001; Spök, 2006). Most common uses of these en- including Mucor sp., Thermotoga celer, Thermotoga maritime, Arxula
zymes seem to be in medicine. Collagenase plays an important role in adeninivorans, Pyrococcus furiosus, Lipomyces, Altermonas haloplanctis,
the successful transplantation of specific organs (Kin et al., 2007; Klock Saccharomycopsis, Schwanniomyces, Vibrio sp., Streptomyces sp., Candida
et al., 1996). japonica and Filobasidium capsuligenum (Gupta et al., 2003; Liu et al.,
2012; Manivasagan et al., 2015; Mohapatra et al., 1998). Marine Sci-
12. Marine microbial xylanases with special characteristics for ence and Technology developed by researchers at the more micro-or-
biotechnological applications ganisms of marine habitats that are capable of producing amylase have
been reported. Marine yeast Aureobasidium pullulans is capable of pro-
Xylanases is a hydrolytic enzyme that degrades xylan that can ducing exocellular amylase were isolated of depths of the Pacific Ocean
produce economic and industrial high-value products such as xylitol. sediments (Li et al., 2007). A novel α-amylase was isolated from marine
Xylanases can be used in the paper industry to improve the dissolution Streptomyces sp. and using the medium containing 2% sucrose, 0.35%
rate of lignin and reduce the use of Cl2 and ClO2 payments thus redu- and 0.15% peptone malt extract (Chakraborty et al., 2009).
cing pollution and improving the properties of the pulp. Also xylanase A novel amylase was isolated from Mucor sp. associated with marine
can damage some juice drinks and beer polysaccharides in thus helps to sponge Spirastrella sp. and showed maximum enzyme activity till 60 °C
produce better, more transparent and drinks (Doi, 2008; Klemm et al., and pH 5.0 (Mohapatra et al., 1998).
2005; Nishiyama et al., 2002; Tong et al., 1980). It was found in the Marine amylase is a subject of interest from the perspective of
some marine microbes including Aureobasidium pullulans, Trichoderma biotechnology (Table 8). Production of α-amylase identified and iso-
longibrachiatum, T. neapolitana, Thermotoga maritime, Glaciecola meso- lated by marine bacteria Pseudoalteromonas undina NKMB 0074
phila KMM 241, Nocardioides sp. KP7 and Cycloclasticus sp. A5 (Guo (Matsumoto et al., 2003). Recently reported that the enzyme α-amylase
et al., 2009; Jiang et al., 2008; Jiang et al., 2004). of marine Streptomyces sp. D1 able to hydrolysis of both α-1–4 and α-
1–6 (Chakraborty et al., 2009).
13. Oxidoreductase isolated from marine microbes Amylase in phosphate buffer systems with pH 7–8 buffers are well
within the range pH 9–11 glycine-NaOH was stable when incubated for
Chloroperoxidase isolated from a marine fungus Caldariomyces fu- 6–48 h. The production of amylase was first reported in Streptomyces
mago the peroxide is unique because it contains cysteinic thiolate as the with a wide range of pH. This strain is optimum growth temperature
fifth ligand accommodate axial ligand is imidazole. This is not only range of 37–55 °C and 45 °C maximum production of amylase is ob-
versatile enzyme peroxidase reactions but mono oxygenasis and cata- served. This enzyme is almost 50% of its activity at 85 °C is maintained.
lysis in the presence of halogen ions and H2O2 does (Colonna et al., This clearly shows the nature of thermostable enzymes.
1999). To date, the bacteria belonging to the genus Bacillus are used for
Flavodon flavus fungus isolated from seaweed degraded coral lagoon commercial production of heat-stable amylase, but such reports of
on the coast of the West Indies three vessels, extracellular enzyme is isolated heat-stable amylase from marine Streptomyces were not re-
produced which contains manganese dependent some enzymes such as ported.
peroxidase, lignin peroxidase and laccase (Raghukumar et al., 1999). Both terrestrial and marine microorganisms may be adapted to the
A manganese-dependent peroxidase activity was reported of optimum growth temperature at 80–108 °C.
Debaryomyces polymorphus, Candida tropicalis and Umbelopsis isabellina Most marine microorganisms belonging to Thermotoga sp. it has
(Yang et al., 2003). been interesting glycoside hydrolase that can be used in

141
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

transglycosylation reactions (Goyal et al., 2001a; Tramice et al., 2007).

(Santorelli et al., 2016)

(Vijayaraghavan et al.,
Pullulanase, α-amylase and α-glucosidase belonging to archaea

(Mesbah and Weigel,


(Song et al., 2016)

(Sen et al., 2016)


have been active in the high-temperature. These enzymes used for
commercial and industrial applications and used in the starch industry.
References

Pullulanases type I very rare in thermophilic microorganisms that


temperature of at 90 °C is optimal activity. This enzyme capable of

2014)

2015)
hydrolyzing α-1,6 and α-1,4, but cannot break. This enzyme has been
reported only in Fervidobacterium pullulanolyticum. This enzyme is pre-
Halophilic and alkalithermostable glucoamylopullulanase sent in the anaerobic bacteria and archaea also heat-loving bacteria

Maximal amylase production of 464 units/mL of enzyme


was observed in the presence of 100% moisture, 0.1%
have a wide distribution. Marine thermophilic bacteria, Thermococcus
litoralis (optimal temperature of 90 °C) and Pyrococcus furiosus (optimal
temperature of 98 °C) are produced Pullulanases type II. Other bacteria
from which the Pullulanases activity is reported are Pyrococcus woesei,
Molecular mass determined by SDS-PAGE.

Molecular mass estimated by SDS- PAGE,


fructose and 0.01% ammonium sulphate.
Medium stability high-active alkalophilic
Alkali-tolerant and high-active amylase,

Thermococcus celer, Fervidobacterium pennavorans and Desulfurococcus


Extracellular gluco-amylo-pullulanase,

mucosus. Cyclomaltodextrin-glucanotransferase has potential applica-


tion in the production of cyclodextrin and alkaliphilic bacteria Bacillus
Vmax: 250 mmol/min/mL
subtilis from deep-sea mud is separated. β-Glucosidases (3.2.1.21) re-
sponsible for the breakdown terminals, without reducing residues β-D-
Thermostable amylases,
Extreme haloarchaeon.

glucosyl of alkyl and aryl β-glycosides in disaccharides and short chain


Other properties

oligosaccharides, with the release of β-D-glucose are residues (Bhatia


Km: 5.88 mg/mL,
alpha-amylase

et al., 2002). This enzymatic conversion of starch to sugar combined


with glucomylase effective are the glucose and hence a high efficiency.
Wu and Chen, Pullulanases from yeast Aureobasidium pullulans in
flower shape isolated sea (Wu and Chen, 2014). Optimal conditions for
decomposition Pullulan is 47.9◦C and pH 4.92 and incubation period
Mn2+ (each 10 mM), Fe3+ (each 10 mM),
Na+ (each 10 mM), K+ (each 100 mM),

9.40 h.
Al3+ (each 10 mM), Zn2+, Mg2+, Ca2+

Amylopullulunase isolated from high thermophilic archaea


Thermococcus hydrothermalis from deep-sea hydrothermal vents and
showed high activity around pH range 4.0–8.0 with the optimum pH
5.5 (Gantelet and Duchiron, 1998). Similarly, another Pullulanases
Cu2+, Co2+ and Zn2+

isolated from archaea Thermococcus sp. HJ21 (Xu et al., 2009). Ac-
cording efforts produce a variety of high thermophilic Pullulanases
strains or similar high thermal stability, while the optimum tempera-
ture depends on the activity profile.
Inhibitor

and SDS

Archeo Thermococcus able to produce Pullulanase, glucosidase and


EDTA
Marine microbial amylases with desirable properties to be suggested for utilizing different industrial applications.

amylase is Legin et al. (1997). Brown and Kelly (1993) purified extra-
cellular pullulanase from marine Thermococcus litoralis and Pyrococcus
PMSF, DTT, H2O2, Triton-

furiosus (Brown and Kelly, 1993). These two species have high activity
X−100, Tween 20 and

Ca2+, Mg2+ and Na+

amylopullulanases in extreme heat. Pullulanase extracellular enzyme


Na+, K+, and Mn2+

was purified from marine archeo thermostable Thermococcus hydro-


Tween 80. Ca2+

thermalis from hydrothermal vents in the east Pacific (Gantelet and


(each 1 mM)

and EDTA
Activator

Duchiron, 1998).
β-Glucosidases divided to two glycoside hydrolyze family GH1 and
Ca2+

GH3 that most enzymes β-galactosidase and β-glucosidase activity in-


dicator GH1, whereas recent activity shows GH3 enzymes (Henrissat,
Molecular mass

1991). The β-Glucosidases from marine microbes can be used in a


variety of industrial sectors including juices, wine and beer, fragrances
54.0 (SEC)
66.0 (SDS-

and flavors compounds (Kang et al., 2010; Trincone, 2013; Wang et al.,
PAGE)
(kDa)

2013). β-Glucosidases can also be linked to cellulose borers feed ad-


49

50

54

ditives for monogastric animals such as poultry or pig feed is used


Temperature (°C)

which causes gastrointestinal (Zhang et al., 1996).


Applications in Biotechnology hydrolase enzymes have high ac-
tivity, glucose tolerance, resistance to acid and heat and finally have
Optimum

transglycosylation activity (Bohlin et al., 2013). Thermotoga maritima on


marine resources has been identified as producing the enzyme β-Glu-
50

54

55

50

45

cosidases (Goyal et al., 2001b). In T. neopolitana was showed that


Optimum

produces β-glucosidase, GH3 (Turner et al., 2007).


Applications in Biotechnology hydrolase enzymes have high ac-
9.0

8.0

8.5

8.0

8.5
pH

tivity, glucose tolerance, resistance to acid and heat and finally have
transglycosylation activity (Bohlin et al., 2013). Thermotoga maritima
Amphibacillus sp. NM-

Exiguobacterium sp.

on marine resources has been identified as producing the enzyme β-


Luteimonas abyssi

Glucosidases (Goyal et al., 2001b). Marine Actinobacteria Streptomyces


turkmenica
Haloterrigena
XH031

sp. MBRC-82 for the production of secondary metabolites with biolo-


Ra2
species

gical activity such as immunosuppression and antibiotics has attracted


Table 8

considerable attention in Bio-Nano Technology (Manivasagan et al.,


2014). This species is capable of producing α-amylase is an

142
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

environmentally friendly tool. (Katikala et al., 2009). Glutaminase from marine microbes to salinity
Most studies show that about 77% of marine bacteria resistant to the and temperature is stable because of an enzyme mode is interest in
cold poles and 23% are accustomed to the cold. In Feller et al. (1994), many industries like food, pharmaceutical, etc.
showed that α-amylase obtained from polar marine bacteria Alter-
omonas haloplanktis can grow well at 4 °C and activity of α-amylase is 16. Microbial phytase in marine environment
seven times higher than the α-amylase warm-blooded animals.
α-Galactosidase in the sugar industry to increase efficiency by hy- Phytase enzyme is belonging to the family phosphatases. This en-
drolysis of sucrose, raffinose, α-galactoside is used. α-Galactosidase zyme breaks down the phytic acid to smaller parts. The enzyme to
obtained from marine Alteromonas spp. α-Galactosidase isolated from neutralize the negative effect of phytate on protein and nutrients in
sponges and algae Polysiphonia sp. and bacteria associated with muscle monogastric animal diets and increase uptake (Vats et al., 2009; Xiong
Crenomytilus grayanus and scallops Patinopecten jessoensis. et al., 2004).
Glucoamylases produced has been reported by marine microorganisms. Phytase is widely used in many industries. Are used in food, dairy,
Marine yeast Aureobasidiumpullulans N13d capable of producing glu- nutrition industries and has potential applications in other fields.
coamylase with catalytic activity temperature is more than 60 °C (Li Several species of marine microorganisms including bacteria, yeast and
et al., 2007). Also marine fungi endophytic Aspergillus sp. JAN-25 fungi are capable of producing phytase (Jorquera et al., 2008;
capable of producing glucoamylase has the maximum activity is be- Noureddini and Dang, 2009).
tween 50 and 60 °C (Matsumoto et al., 2003). Recent studies have shown that microbial phytase produced eco-
α-amylase has been used extensively in the construction industry, nomically very significant and used in industry and stake (Lei and
especially alcoholic beverages such as beer, alcohol, animal feed, de- Porres, 2003).
tergents for clothes, starch industry, confection and textile industry Now, the mushrooms are having efficient enzyme system, the bio-
(Debashish et al., 2005; Lee et al., 2010; Trincone, 2011; Venugopal, degradation of undesirable substances such as industrial and agri-
2016). α-amylase obtained from thermophilic arceo include the Pyr- cultural waste, etc., and convert them into usable products and harm-
ococcus woesei, Pyrococcus furiosus, Thermococcus celer, Fervidobacterium less, are very important. Some fungi such as Aspergillus, Penicillium,
pennavorans, Desulfurococcus mucosus and Thermotoga maritime; psico- Mucor and Rhizopus species are commonly used for the commercial
trophic vibrio isolated from the muds of the sea floor, Vibrio gazogenes, production of extracellular phytase (Pandey et al., 2001).
Alteromonas rubra and Mucor sp. α-Glucosidase use in industry, such as In 2015 by Sadati and Barghi, 6 species of fungus produces extra-
α-amylase is starch. α-Glucosidase tolerant to heat were isolated from a cellular phytase from coastal waters of the Caspian Sea in Iran was
marine thermophilic arceobacter (hyperthermophilic) include the Pyr- isolated and identified (Sadati and Barghi, 2014). These include As-
ococcus furiosus and Pyrococcus wesei. pergillus flavus, Aspergillus ochraceus, Penicillium olsonii, Penicillium digi-
tatum, Discostroma tricellulare and Cladosporium gossypiicola (Chi et al.,
15. Transglutaminase from marine microbes (TGases) 2009; Jorquera et al., 2008; Noureddini and Dang, 2009; Pandey et al.,
2001; Vats et al., 2009; Xiong et al., 2004). These three species Asper-
Transglutaminase enzyme is known with name EC 2.3.3.13, there gillus flavus, Aspergillus ochraceus and Penicillium olsonii showed the
are widespread in nature. The enzyme transglutaminase is a protein highest phytase activity (Table 9).
with a molecular weight of 37368 Da that contain 331 amino acids.
This enzyme can be between amino acids glutamine and lysine of a 17. Biochemical properties and biotechnological aspects of
protein from binding to other proteins. It is remarkable that this en- marine microbial inulinases
zyme does not adversely affect the bioavailability of lysine (Trachoo
and Mistry, 1998). Until the 1989 transglutaminase enzyme was ex- Inulinases enzyme responsible for the breakdown of inulin, fructose
tracted from the swine liver but research led to the identification of a or fructo oligosacharide produces the ultimate respectively. Inulinases a
bacterial enzyme was produced. The enzyme was extracted and purified polyfructan in some plants such as Jerusalem artichoke and chicory are
of a bacterial species called Streptoverticillium. Transglutaminase de- connected by connections β 2,1. The enzymes or probiotics as a
rived from the bacterium has a molecular weight of 40,000 Da by SAS- sweetener, has been used extensively in the food industry (Li et al.,
PAGE (Sodium Dodecyl Sulphate Poly Acrylamide Gel Electro Phoresis) 2007; Lima et al., 2011). Inulinase for endo-inulinases (EC 3.2.1.7) or
and isoelectric pH is about 9.8. The optimal pH of the enzyme activity exoinulinases (EC 3.2.1.80) ranks collapsed and have the ability to
between 5 and 9 and between 37° and 50° is the best temperature for its break the chain inulin-type oligosaccharides are smaller (Basso et al.,
operation. Generally, transglutaminase enzyme is stable in a wide 2010).
temperature range. This enzyme at 50 °C for 10 min retains its activity. The cleavage reaction can be performed in a temperature range of
Transglutaminase derived from microorganisms (MTGase) against 40–75 but most analysis is done in 45–55 °C (Kerkhoffs and, 1981;
transglutaminase derived from pig liver that is calcium-dependent en- Ricca et al., 2007). Marinimicrobium sp. LS-A18 able to produce extra-
zyme, is independent of calcium ions (Kuraishi et al., 2001). It is re- cellular inulinase is bearing alkaline conditions pH 9. In addition, in-
ported that marine yeasts, marine fungi and marine bacteria produce ulinase able to tolerate salt that has seen the most activity by 0.5% (w/
extracellular L-glutaminase (Kashyap et al., 2002; Kumar and V) NaCl and 20% NaCl is about 60% active. Inulin can be decomposed
Chandrasekaran, 2003; Sabu et al., 2000). by inulinase under alkaline conditions, temperature and pH 65◦C be at
TGases in regulating cell growth, differentiation, blood coagulation, least 10 (Li et al., 2012). In Nocardiopsis sp. DN-K15 (Lu et al., 2014)
keratin of the epidermis and tighten the red blood cell membrane plays and Bacillus cereus MU-31 (Meenakshi et al., 2013) have been identified
a role. TGases acyl transfer reaction of the γ-carboxamide groups of that are capable of producing extracellular exo-inulinase.
glutamine residues peptide bond with the ε-amino residues of lysine
and other amino groups of the protein catalyzes (Venugopal, 2016). L- 18. Marine microbial tannases
glutaminase (L-glutamine amidohydrolase, EC 3.5.1.2) is parser L-glu-
tamine. L-glutaminase has industrial applications and treatment. This Tannases (EC 3.1.1.20) breaks down glucose and tannic acid and
enzyme as anti-cancer, sour taste of food by increasing glutamic acid is gallic acid is produced in the end. Gallic acid, gallate as antioxidants,
used (Kashyap et al., 2002; Kumar and Chandrasekaran, 2003; Sabu the production of flavors of coffee and brew the tea plays an important
et al., 2000). role (Aguilar et al., 2001). Aspergillusawamori BTMFW032 is acid-
Also the enzyme can be used as biosensors for monitoring and ophilus enzyme with optimal pH 2 (Beena et al., 2010). The first time in
glutamine levels in mammalian cell cultures hybridoma to be used 2012, it was reported that marine Cyanobacteria Phormidium

143
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

valderianum BDU140441 is able to produce the enzyme Tannases

(Zhang and Kim, 2010)

(Mullaney et al., 2002)


(Palanisami et al., 2011) (Table 10).

(Balakrishna et al.,
(Casey and Walsh,

(Vats et al., 2009)


19. 1Fucoidanase isolated from marine microbes
References

Fucoidan is a sulfated polysaccharide composed of some simple

2003)

2017)
sugars. Fucoidan oligosaccharides with a molecular weight < 1000 Da
and can be used as an activator of human epidermal keratinocytes.
Enzyme activity was not significantly affected by metal ions such as Ca2+, Mg2+,

Fucoidanase obtained from marine Vibrio (Furukawa et al., 1992),


Pseudomonas atlantica and Pseudoalteromonas carrageenovora (Yaphe
and Morgan, 1959).
Marine fungi Marinimicrobium sp. LS-A18 isolated from sea sand
(Baltic Sea, Germany) is capable of producing fucoidanase with max-
Mn2+, Zn2+, but was slightly stimulated in the presence of EDTA

imum activity in the temperature and pH 50 °C and 6.0, respectively


Molecular mass determined by gel filtration and SDS–PAGE,

(Wu et al., 2011).

20. Marine microbial Glucanases

Japanese researchers focused on the extraction glucanase bacteria


Bacillus circulans. They isolated the bacteria from muds Tokyo Bay, but
when put in proper watery environment that could grow and produce
new glucanase.glucanase could α-1,3 bond and the bond α-1,6 glucan
activity in and breaks it up. Researchers isolated a new glucanase from
marine bacterium Bacillus at a temperature of 37 °C best practices
showed that it is medically appropriate for dental and other health care
(Yahata et al., 1990). So far, the production of glucanase was reported
Curcin and trypsin
Other properties

in some microbes including marine streptomycete S. xinghaiensis,


Streptomyces sp. SPB74, Micromonospora sp. ATCC 39149, Streptomyces
albogriseolus, Marinimicrobium sp. LS-A18, Saccharomyces cerevisiae and
marine fungus Chaetomium indicum (Burtseva et al., 2003; Mollers et al.,
2014; Zhao et al., 2012).
Glucanase can be used in various industries, including detergent,
textile, brewing, paper manufacturing (Sukharnikov et al., 2011)
Molecular mass

21. Other marine microbial enzymes


(kDa)

65.5

353

According to recent reports from other enzymes in the sea water and
85

84

sediment microbial enzymes lycopene k-cyclase (Stickforth et al.,


55.9 mg mL−1

2003), L-serine dehydratase (Laroche and Ulber, 2002), poly-


hydroxybutyrate depolymerase (Kita et al., 1995), polyhydroxybutyrate
Vmax

depolymerase (Kita et al., 1995), quinol oxidase (Simpson et al., 1997)


and arylsulphatase (Barbeyron et al., 1995) have been obtained.
Biological resources, including animals, plants and microbes has a
0.295 mM

huge potential for biotechnological applications and valuable products


Km

such as drug, bioactive additives and cosmetics (Lima and Porto, 2016).
Actinomycetales is a very useful bacterium in producing many
bioactive compounds, including antibiotics, probiotics, melanin, anti-
Optimum Temperature

cancer compounds and enzyme inhibitors. Therefore, research on


Actinobacteria is important in the discovery of new bioactive com-
pounds (Manivasagan et al., 2013).
Species of the genus Streptomyces is capable of producing metabo-
lites such as antibiotics, anti-cancer compounds, enzyme inhibitors,
52–55
(°C)
Marine microbes as a resource for novel phytases.

pigments, peptides, trioxacarcins, methylpyridine, macrolides, ter-


67

58

65

37

penes, polyketides, flavonoids and other ingredients (Dharmaraj,


Optimum pH

2010).
25.50–55

Halophillic actinomycetes halotolerant salts as a source of bioactive


metabolites have been less studied. The Actinomycetes capable of de-
1.3

5.0

2.5

5.0

grading organic compounds in contaminated water, soil aliphatic,


aromatic, enzyme inhibitors and metabolites such as antibiotics and
enzymes (Hamedi et al., 2013). B. subtilis SK11.004 is capable of pro-
Aspergillus niger NRRL

Aspergillus niger ATCC

Aspergillus niger van

Candida parapsilosis

ducing γ-glutamyl transpeptidase (GGT) with theanine-forming ability.


Aspergillus ficuum

This enzyme synthesis of theanine using from GGT a potential appli-


NTG−23

Teighem

cation in the production of theanine and other ingredients is γ-glutamyl


3135

9142
Species

(Shuai et al., 2010).


Table 9

Bacillus spp. coastal sediment samples can be isolated from 48 h and


pH 2.5, 35 °C, gelatinase produce. Gelatinase can cause tissue analysis

144
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

Table 10
Biochemical features of tannases extracted from different marine microbes.

Species Optimum pH Optimum Inhibitor Other properties References


Temperature (°C)

Aspergillus tubingensis 6 and 7 50 Km: 0.45 and 0.29 mM, (Wu et al., 2016)
Vmax: 0.26 µM min−1
Aspergillus niger ATTC 5.5 30 (Sabu et al., 2005)
16620
Aspergillus ruber 6.0 30 (Kumar et al., 2007)
Aspergillus fumigatus MA 5.0 25 (Manjit et al., 2008)
Penicillium variable 5.0 50 PMSF and N- ethylmaleimide Molecular mass: 158 kDa (Sharma et al., 2008)
retaining
Bacillus cereus KBR9 4.5 40 Halostable in the presence of high concentrations (Mondal et al., 2001)
of NaCl (2 M)

Table 11
Purification procedures of some microbial enzymes from marine environment.

Species Enzymes Total protein Specific activity Total activity Yield (%) Purification fold References
(mg) (U/mg) (U)

Microscilla Agarase 0.28 10.9 3.1 14 2.5 (Naganuma et al., 1993)


Alteromonas sp. E-l Agarase 0.026 34.0 0.87 1.49 40.9 (Kirimura et al., 1999)
Bacillus sonorensis 4 R Lipase 0.355 2152.08 3055.96 1.98 12.15 (Bhosale et al., 2016)
Halobacillus trueperi RSK Lipase 3.42 1193.59 4082.10 14.31 8.46 (Sathishkumar et al.,
CAS9 2015)
Aspergillus terreus Chitinase 8.79 182.08 1607.15 12 5.16 (Farag et al., 2016)
Paenicibacillus barengoltziia Chitinase 11.0 30.3 333.8 51.9 51.9 (Yang et al., 2016)
Paenicibacillus barengoltzii Chitinase 8.0 10.2 (Fu et al., 2016)
CAU904
Pseudomonas elongata κ-Carrageenase 60.7 (Khambhaty et al., 2007)
Cytophaga Carrageenase 1.69 9633 16,280 32 37 (Potin et al., 1991)
Luteimonas abyssi XH031 α-Amylase 8881 (Song et al., 2016)
Haloterrigena turkmenica α-Amylase 298 79.8 23,794 (mU) 18.9 33.1 (Santorelli et al., 2016)
Anoxybacillus beppuensis Amylase 12 10,000 120,000 20 44 (Kikani and Singh, 2012)
TSSC−1
Bacillus cereus Amylase 8.1 44 180 16 (Annamalai et al., 2011)
Streptomyces A3 Amylase 4.1 1755 7139 58 20 (Chakraborty et al., 2012)
Amphibacillus sp. NM-Ra2 Amylase 2.4 250 600 15.4 4.5 (Mesbah and Weigel,
2014)
Saccharoplyspora A9 Amylase 5.7 1641 9369 25 39 (Chakraborty et al., 2011)
Wangia sp. C52 Amylase 2.6 146 379 56 24 (Liu et al., 2011a)
Nocardiopsis sp. B2 Amylase – 1755 – 20 44 (Chakraborty et al., 2014)
Bacillus cereus IND4 Amylase 54.6 3.9 (Vijayaraghavan et al.,
2015)
Pseudomonas sp. K6–28–040 Amylase 6.5 135 875 44 5 (Liu et al., 2011b)
Zunongwangia profunda Amylase 0.7 1663 1164 20 333 (Wu et al., 2014a)
(AmyZ2)
Exiguobacterium sp. Amylase 50.5 1083 U/mL 54,668 (Sen et al., 2016)
Vibrio sp. Amylase 1.4 327 461 78 163 (Najafi and Kembhavi,
2005)
Candida parapsilosis Phytase 637.66 U/g (Balakrishna et al., 2017)
Aspergillus ficuum NTG−23 Phytase 150.1 615 4.1 mg 71.5 (Zhang et al., 2010)
Rhodotorula mucilaginosa L7 Acid protease 0.58 5218 3026 40.42 41.77 (Lario et al., 2015)
Aspergillus oryzae MTCC Acid protease 8 43,658 3,49,264 29 16.6 (Vishwanatha et al., 2009)
5341
Bacillus sp. APCMST-RS3 Protease 8.60 28.62 246.15 U/mL 22.66 8.49 (Maruthiaha et al., 2015)
Purpureocillium lilacinum LPS Serine protease 0.32 1432.7 458.5 1.3 (Cavello et al., 2013)
# 876
Bacillus subtilis DR8806 Serine protease 53 79.3 37.5 3.4 (Farhadian et al., 2015)
Bacillus alveayuensis CAS 5 Alkaline protease 7.83 518.78 4062.1 12.86 7.77 (Annamalai et al., 2014b)
Bacillus sp. Alkaline protease 1.4 657.14 920 64.11 (Jain et al., 2012)
Bacillus firmus CAS 7 Alkaline protease 7.3 473.4 3456.2 12.3 2.6 (Annamalai et al., 2014b)
Halophilic strain SD11 Alkaline protease 3.10 432.32 1340.18 68 2.38 (Cui et al., 2015)
Rhodococcus sp. YM12 Phosphinothricin N- 1.08 23.40 25.27 19.60 334.3 (Wu et al., 2014b)
acetyltransferase

a
Cloned and expressed in Escherichia coli (PbChi70).

during tumor metastasis and swollen joints (Balan et al., 2012). Araki macrura ability to synthesize extracellular β-galactosidase is adapted to
et al., 117 marine bacteria belonging to Pseudomonas, Alcaligenes, cold conditions (Turkiewicz et al., 2003).
Klebsiella, Enterobacter, Vibrio, Aeromonas, Moraxella, Bacillus, etc., each
of which can be detected and isolated β-mannanase were produced
22. Conclusions
(Amki, 1981).
Pseudoalteromonas sp. isolated from the Antarctic krill Thyssanoessa
Our growing knowledge and technique improvement regarding

145
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

protein extraction and purifi cation has led to the production of many simultaneous detoxification by Candida parapsilosis isolated from poultry garbage.
Bioresour. Technol. 225, 215–224.
enzymes at analytical-grade purity for research and biotechnological Balan, S.S., Nethaji, R., Sankar, S., Jayalakshmi, S., 2012. Production of gelatinase en-
applications (A. Homaei, 2015a; Homaei et al., 2014; Homaei and zyme from Bacillus spp. isolated from the sediment sample of Porto Novo coastal
Etemadipour, 2015; Sharifian et al., 2017; Zeinali et al., 2015). Recent sites. Asian Pac. J. Trop. Biomed. 2 (3).
Barbeyron, T., Potin, P., Richard, C., Collin, O., Kloareg, B., 1995. Arylsulphatase from
advances in biotechnology, particularly in protein engineering, have Alteromonas carrageenovora. Microbiology 141 (11), 2897–2904.
provided the basis for the efficient development of enzymes with im- Basheer, S.M., Chellappan, S., Beena, P.S., Sukumaran, R.K., Elyas, K.K., Chandrasekaran,
proved properties. This has led to establishment of novel, tailor-made M., 2011. Lipase from marine Aspergillus awamori BTMFW032: production, partial
purification and application in oil effluent treatment. New Biotechnol. 28 (6),
enzymes for completely new applications (Dadshahi et al., 627–638.
2016; Homaei, 2015b; Homaei and Saberi, 2015; Homaei and Samari, Basso, A., Spizzo, P., Ferrario, V., Knapic, L., Savko, N., Braiuca, P., Ebert, C., Ricca, E.,
2017; Homaei et al., 2017; Homaei et al., 2013a; Homaei et al., Calabro, V., Gardossi, L., 2010. Endo- and exo-inulinases: enzyme-substrate interac-
tion and rational immobilization. Biotechnol. Prog. 26 (2), 397–405.
2010; Homaei et al., 2013b; Rouzbahan et al., 2016; Rouzbehan et al.,
Bayoudh, A., Gharsallah, N., Chamkha, M., Dhouib, A., Ammar, S., Nasri, M., 2000.
2017). Studies related to the prospecting of microbial enzymes from Purification and characterization of an alkaline protease from Pseudomonas aerugi-
marine environments that are different from their terrestrial counter- nosa MN1. J. Ind. Microbiol. Biotechnol. 24, 291–295.
parts and also increase our understanding about the function, applica- Beena, P.S., Soorej, M.B., Elyas, K.K., Sarita, G.B., Chandrasekaran, M., 2010. Acidophilic
tannase from marine Aspergillus awamori BTMFW032. J. Microbiol. Biotechnol. 20
tion, diversity and ecology of this microbial group. (10), 1403–1414.
Microbes and microorganisms from marine environments secret Beheshti Maal, K., Emtiazi, G., Nahvi, I., 2009. Production of alkaline protease by Bacillus
different enzymes based on their function and their habitat. In the cereus and Bacillus polymixa in new industrial culture mediums and its immobiliza-
tion. Afr. J. Biotech. 3 (9), 491–497.
present review, several marine microbial enzymes were isolated from Belas, R., 1989. Sequence analysis of the agrA gene encoding beta-agarase from
seawater and marine sediments, purified and characterized for this Pseudomonas atlantica. J. Bacteriol. 171 (1), 602–605.
properties and applications (Table 11). Marine microbial enzymes, Berdy, J., 2005. Bioactive microbial metabolites. J. Antibiot. (Tokyo) 58 (1), 1–26.
Besson, S., Carneiro, C., Moura, J.J., Moura, I., Fauque, G., 1995. A cytochrome cd1-type
especially extremophiles, have become more and more important in nitrite reductase isolated from the marine denitrifier Pseudomonas nautica 617: pur-
applications. Clinical and industrial application of marine microbial ification and characterization. Anaerobe 1 (4), 219–226.
enzymes has been developing also. Collectively, this article has pro- Bhatia, Y., Mishra, S., Bisaria, V.S., 2002. Microbial beta-glucosidases: cloning, proper-
ties,and applications. Crit. Rev. Biotechnol. 22, 375–407.
vided many examples of marine microbial from marine sources which
Bhosale, H., Shaheen, U., Kadam, T., 2016. Characterization of a Hyperthermostable al-
are being used as successful biocatalysts. Their use highlights a pros- kaline lipase from Bacillus sonorensis 4R. Enzym. Res. 2016, 11.
pering and exciting area of industrial biotechnology. Boetius, A., 1995. Microbial hydrolytic enzyme activities in deep-sea sediments. Hel.
Meer 49, 177.
Bohlin, C., Praestgaard, E., Baumann, M.J., Borch, K., Praestgaard, J., Monrad, R.N.,
Acknowledgements Westh, P., 2013. A comparative study of hydrolysis and transglycosylation activities
of fungal β-glucosidases. Appl. Microbiol. Biotechnol. 97 (1), 159–169.
The authors express their gratitude to the research council of the Bond, M.D., Van Wart, H.E., 1984. Characterization of the individual collagenases from
Clostridium histolyticum. Biochemistry 23 (13), 3085–3091.
University of Hormozgan for financial support during the course of this Boonmahome, P., Mongkol thanaruk, W., 2013. Lipase-producing bacterium and its en-
project. zyme characterization. J. Life Sci. Technol. 1 (4), 196–200.
Brás, N.F., Cerqueira, N.M.F.S.A., Fernandes, P.A., Ramos, M.J., 2008. Carbohydrate
binding modules from family 11: understanding the binding mode of polysaccharides.
References Int. J. Quantum Chem. 108, 2030–2040.
Brown, S.H., Kelly, R.M., 1993. Characterization of amylolytic enzymes, having both
Adams, D.J., 2004. Fungal cell wall chitinases and glucanases. Microbiology 150 (Pt 7), alpha- 1, 4 and alph- 1, 6 hydrolytic activity, from the thermophilic archaea pyr-
2029–2035. ococcus furiosus and thermococcus litorlis. Appl. Environ. Microbiol. 59, 2614–2621.
Adams, M.W., Perler, F.B., Kelly, R.M., 1995. Extremozymes: expanding the limits of Bugli, F., Posteraro, B., Papi, M., Torelli, R., Maiorana, A., Sterbini, F., Posteraro, P.,
biocatalysis. Biotechnology (N.Y.) 13 (7), 662–668. Sanguinetti, M., De Spirito, M., 2013. In vitro interaction between Alginate lyase
Aggarwal, A., Kaur, S., Tiwary, A.K., Gupta, S., 2001. Chitosan microspheres prepared by fumigatus and Amphotericin B against Aspergillus biofilm determined by different
an aqueous process: release of indomethacin. J. Microencapsul. 18 (6), 819–823. methods. Antimicrob. Agents Chemother. 57, 1275–1282.
Aguilar, C.N., Augur, C., Favela-Torres, E., Viniegra-Gonzalez, G., 2001. Production of Bull, A.T., Ward, A.C., Goodfellow, M., 2000. Search and discovery strategies for bio-
tannase by Aspergillus niger Aa-20 in submerged and solid-state fermentation: in- technology: the paradigm shift. Microbiol. Mol. Biol. Rev. 64 (3), 573–606.
fluence of glucose and tannic acid. J. Ind. Microbiol. Biotechnol. 26 (5), 296–302. Burtseva, Y.V., Verigina, N.S., Sova, V.V., Pivkin, M.V., Zvyagintseva, T.N., 2003. O-
Albrecht, M.T., Schiller, N.L., 2005. Alginate Lyase (AlgL) activity is required for alginate Glycosylhydrolases of marine filamentous fungi: β-1,3-glucanases of Trichoderma
biosynthesis in Pseudomonas aeruginosa. J. Bacteriol. 187, 3869–3872. aureviride. Appl. Biochem. Microbiol. 39 (5), 475–481.
Alemán, A., & Martínez-Alvarez, O. (2008). Marine collagen as a source of bioactive Buttner, M.J., Fearnley, I.M., Bibb, M.J., 1987. The agarase gene (dagA) of Streptomyces
molecules. coelicolor A3(2): nucleotide sequence and transcriptional analysis. Mol. Gen. Genet.
Amki, T., 1981. β-mannanse of bacteria isolated from natural habitats. Bull. Jpn. Soc. Sci. 209 (1), 101–109.
Fish. 47, 753–760. Carpenter, E.J., Bergman, B., Dawson, R., Siddiqui, P.J., Söderbäck, E., Capone, D.G.,
Ananthi, S., Ramasubburayan, R., Palavesam, A., Immanuel, G., 2014. Optimisation and 1992. Glutamine synthetase and nitrogen cycling in colonies of the marine diazo-
purification of lipase through solid state fermentation by Bacillus cereus MSU as trophic cyanobacteria Trichodesmium spp. Appl. Environ. Microbiol. 58 (9),
isolated from the gut of a marine fish Sardinella longiceps. Int. J. Pharm. Pharm. Sci. 6 3122–3129.
(5), 291–298. Casey, A., Walsh, G., 2003. Purification and characterization of extracellular phytase from
Annamalai, N., Rajeshwari, M.V., Sahu, S.K., Balasubramanian, T., 2014a. Purification Aspergillus niger ATCC 9142. Bioresour. Technol. 86 (2), 183–188.
and characterization of solvent stable, alkaline protease from Bacillus firmus CAS7 by Cavello, I.A., Hours, R.A., Rojas, N.L., Cavalitto, S.F., 2013. Purification and character-
microbial conversion of marine wastes and molecular mechanism underlying solvent ization of a keratinolytic serine protease from Purpureocillium lilacinum LPS# 876.
stability. Process Biochem. 49, 1012–1019. Process Biochem. 48 (5), 972–978.
Annamalai, N., Rajeswari, M.V., Balasubramanian, T., 2014b. Extraction, purification and Chadd, H.E., Newman, J., Mann, N.H., Carr, N.G., 1996. Identification of iron superoxide
application of thermostable and halostable alkaline protease from Bacillus al- dismutase and a copper/zinc Superoxide dismutase enzyme activity within the
veayuensis CAS 5 using marine wastes. Food Bioprod. Process. 92, 335–342. marine cyanobacterium Synechococcus sp. WH 7803. FEMS Microbiol. Lett. 138
Annamalai, N., Thavasi, R., Vijayalakshmi, S., Balasubramanian, T., 2011. Extraction, (2–3), 161–165.
purification and characterization of thermostable, alkaline tolerant α-amylase from Chakraborty, S., Jana, S., Gandhi, A., Sen, K.K., Zhiang, W., Kokare, C., 2014. Gellan gum
Bacillus cereus. Indian J. Microbiol. 51 (4), 424–429. microspheres containing a novel α-amylase from marine Nocardiopsis sp. strain B2 for
Aoki, T., Araki, T., Kitamikado, M., 1990. Purification and characterization of a novel immobilization. Int. J. Biol. Macromol. 70, 292–299.
beta-agarase from Vibrio sp. AP-2. Eur. J. Biochem. 187 (2), 461–465. Chakraborty, S., Khopade, A., Biao, R., Jian, W., Liu, X.Y., Mahadik, K., Chopade, B.,
Arnos, T.C., Jorgensen, B.B., Sagemann, J., Tramdrup, T., 1998. Temperature dependence Zhang, L., Kokare, C., 2011. Characterization and stability studies on surfactant,
of microbial degradation of organic matter in marine sediment: polysaccharide hy- detergent and oxidant stable -amylase from marine haloalkaliphilic Saccharopolyspora
drolysis, oxygen consumption, and sulphate reduction. Mar. Ecol. Prog. Ser. 165, 59. sp. A9. J. Mol. Catal. B: Enzym. 68, 52–58.
Arora, P.K., 2013. Staphylococcus lipolyticus sp. nov., a new cold-adapted lipase producing Chakraborty, S., Khopade, A., Kokare, C., Mahadika, K., Chopade, B., 2009. Isolation and
marine species. Ann. Microbiol. 63 (3), 913–922. characterization of novel α-amylase from marine Streptomyces sp. D1. J. Mol. Catal. B
Babu, J., Pramod, W.R., George, T., 2008. Cold active microbial lipases: some hot issues Enzym 58, 17–23.
and recent developments. Biotechnol. Adv. 26, 457–470. Chakraborty, S., Raut, G., Khopade, A., Mahadik, K., Kokare, C., 2012. Study on calcium
Balakrishna, K., Swaruparani, G., Hanumalal, N., Bindu, S., Bhima, B., 2017. Plausible ion independent α-amylase from haloalkaliphilic marine. Streptomyces Strain A3.
exploitation of Jatropha de-oiled seed cake for lipase and phytase production and Chanalia, P., Gandhi, D., Jodha, D., Singh, J., 2011. Applications of microbial proteases in

146
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

pharmaceutical industry: an over view. Rev. Med. Microb. 22 (4), 96–101. 34H. Struct. Biol. Cryst. Commun. 69 (8), 920–924.
Charles, P., Devanathan, V., Anbu, P., Ponnuswamy, M.N., Kalaichelvan, P.T., Hur, B.K., Doddapaneni, K.K., Tatineni, R., Vellanki, R.N., Rachcha, S., Anabrolu, N., Narakuti, V.,
2008. Purification, characterization and crystallization of an extracellular alkaline Mangamoori, L.N., 2009. Purification and characterization of a solvent and de-
protease from Aspergillus nidulans HA-10. J. Basic Microbiol. 48 (5), 347–352. tergent-stable novel protease from Bacillus cereus. Microbiol. Res. 164 (4), 383–390.
Charoenpanich, J., Suktanarag, S., Toobbucha, N., 2011. Production of a thermostable Doi, R.H., 2008. Cellulases of mesophilic microorganisms: cellulosome and noncellulo-
lipase by Aeromonas sp. EBB-1 isolated from marine sludge in Angsila, Thailand. Sci. some producers. Ann. N.Y Acad. Sci. 1125, 267–279.
Asia 37, 105–114. Dong, J., Hong, Y., Shao, Z., Liu, Z., 2010. Molecular cloning, purification, and char-
Chávez-González, M., Rodríguez-Durán, L.V., Balagurusamy, N., Prado-Barragán, A., acterization of a novel, acidic, pH-stable endoglucanase from Martelella medi-
Rodríguez, R., Contreras, J.C., Aguilar, C.N., 2012. Biotechnological advances and terranea. J. Microbiol. 48 (3), 393–398.
challenges of tannase. Food Bioprocess Technol. 5 (2), 445–459. Du-Thumm., L., Szeles, L. H., Sullivan, R. J., Masters, J. G., & Robinson, R. S. (2005).
Chavez Croocker, P., Sako, Y., A., U, 1999. Purification and characterization of an in- Chewable antiplaque confectionery dental composition. US20050008582.
tracellular heat-stable proteinase (pernilase) from the marine hyperthermophilic ar- Dunne, W.M., Buckmire, F., 1985. Partial purification and characterization of a poly-
chaeon Aeropyrum pernix K1. Extremophiles 3, 3–9. mannuronic acid depolymerase produced by a mucoid strain of Pseudomonas aeru-
Chi, W.J., Chang, Y.K., Hong, S.K., 2012. Agar degradation by microorganisms and agar- ginosa isolated from a patient with cystic fibrosis. Appl. Environ. Microbiol. 50,
degrading enzymes. Appl. Microbiol. Biotechnol. 94, 917–930. 562–567.
Chi, Z., Chi, Z., Zhang, T., Liu, G., Li, J., Wang, X., 2009. Production, characterization and Durham, D.R., Stewart, D.B., Stellwag, E.J., 1987. Novel alkaline- and heat-stable serine
gene cloning of the extracellular enzymes from the marine-derived yeasts and their proteases from alkalophilic Bacillus sp. strain GX6638. J. Bacteriol. 169 (6),
potential applications. Biotechnol. Adv. 27 (3), 236–255. 2762–2768.
Chi, Z., Ma, C., Wang, P., Li, H.F., 2007. Optimization of medium and cultivation con- Dutta, P.K., Dutta, J., Tripathi, V.S., 2004. Chitin and chitosan: Chemistry, properties and
ditions for alkaline protease production by the marine yeast Aureobasidium pull- applications. J. Sci. Ind. Res. 63 (1), 20–31.
ulans. Bioresour. Technol. 98 (3), 534–538. El Alaoui, S.E., Diez, J., Humanes, L., Toribio, F., Partensky, F., García-Fernández, J.M.,
Cho, H.Y., Bancerz, R., Ginalska, G., Leonowicz, A., Cho, N.S., Ohga, S., 2007. Culture 2001. In vivo regulation of glutamine synthetase activity in the marine chlorophyll b-
conditions of psychrotrophic fungus, Penicillium chrysogenum and its lipase char- containing cyanobacterium Prochlorococcus sp. strain PCC 9511 (Oxyphotobacteria).
acteristics. J. Fac. Agric. 52 (2), 281–286. Appl. Environ. Microbiol. 67, 2202–2207.
Chung, L., Dinakarpandian, D., Yoshida, N., Lauer Fields, J.L., Fields, G.B., al., e, 2004. Elcin, Y.M., Dixit, V., Gitnick, G., 1996. Controlled release of endothelial cell growth
Collagenase unwinds triple-helical collagen prior to peptide bond hydrolysis. Eur. factor from chitosan-albumin microspheres for localized angiogenesis: in vitro and in
Mol. Biol. Organ. J. 23, 3020–3030. vivo studies. Artif. Cells Blood Substit. Immobil. Biotechnol. 24 (3), 257–271.
Colonna, S., Gaggero, N., Richelmi, C., Pasta, P., 1999. Recent biotechnological devel- Elson, C.M., Davies, D.H., Hayes, E.R., 1980. Removal of arsenic from contaminated
opments in the use of peroxidases. Trends Biotechnol. 17, 163–168. drinking water by a chitosan/chitin mixture. Water Res. 14 (9), 1307–1311.
Cowan, D.A., Smolenski, K.A., Daniel, R.M., Morgan, H.W., 1987. An extremely ther- Endo, A., Murakawa, S., Shimizu, H., Shiraishi, Y., 1987. Purification and properties of
mostable extracellular proteinase from a strain of the archaebacterium collagenase from a Streptomyces Species. J. Biochem. 102, 163–170.
Desulfurococcus growing at 88 °C. Biochem. J. 247, 121–133. Ertugrul, S., Donmez, G., Takac, S., 2007. Isolation of lipase producing Bacillus sp. from
Cui, F., Dong, S., Shi, X., Zhao, X., Zhang, X.H., 2014. Overexpression and character- olive mill wastewater and improving its enzyme activity. J. Hazard Mater. 149 (3),
ization of a novel thermostable β-agarase YM01–3, from marine bacterium 720–724.
Catenovulum agarivorans YM01T. Mar. Drugs 12 (5), 2731–2747. Essghaier, B., Rouaissi, M., Boudabous, A., Jijakli, H., Sadfi-Zouaoui, N., 2010. Production
Cui, H., Wang, L., Yu, Y., 2015. Production and characterization of alkaline protease from and partial characterization of chitinase from a halotolerant Planococcus rifitoensis
a high yielding and moderately halophilic strain of SD11 marine bacteria. J. Chem. strain M2-26. World J. Microbiol. Biotechnol. 26 (6), 977–984.
2015, 8. Farag, A.M., Abd-Elnabey, H.M., Ibrahim, H.A.H., El-Shenawy, M., 2016. Purification,
Daboor, S.M., Budge, S.M., Ghaly, A.E., Brooks, S.L., Dave, D., 2010. Extraction and characterization and antimicrobial activity of chitinase from marine-derived
purification of collagenase enzymes: aa critical review. Am. J. Biochem. Biotechnol. 6 Aspergillus terreus. Egypt. J. Aquat. Res. 42, 185–192.
(4), 239–263. Farhadian, S., Asoodeh, A., Lagzian, M., 2015. Purification, biochemical characterization
Dadshahi, Z., Homaei, A., Zeinali, F., Sajedi, R.H., Khajeh, K., 2016. Extraction and and structural modeling of a potential htrA-like serine protease from Bacillus subtilis
purification of a highly thermostable alkaline caseinolytic protease from wastes DR8806. J. Mol. Catal. B: Enzym. 115, 51–58.
Penaeus vannamei suitable for food and detergent industries. Food Chem. 202, Feller, G., Narinx, E., L., A.J., Zekhnini, Z., Swing, J., Gerday, C., 1994. Temperature
110–115. dependence of growth, enzyme secretion and activity of psychrophilic Antarctic
Dalmaso, G.Z., Ferreira, D., Vermelho, A.B., 2015. Marine extremophiles: a source of bacteria. Appl. Microbiol. Biotechnol. 41 (40), 477–479.
hydrolases for biotechnological applications. Mar. Drugs 13 (4), 1925–1965. Fenice, M., Leuba, J., Federici, F., 1998. Chitinolytic enzyme activity of Penicillium jan-
Damare, S., Raghukumar, C., Raghukumar, S., 2006. Fungi in deep-sea sediments of the thinellum P9 in bench-top bioreactor. J. Ferment. Bioeng. 86, 620–623.
Central Indian Basin. Deep Sea Res. 53 (1), 14–27. Fernandes, P., 2014. Marine enzymes and food industry: insight on existing and potential
Danson, M.J., Hough, D.W., 1998. Structure, function and stability of enzymes from the interactions. Front. Mar. Sci. 1.
Archaea. Trends Microbiol. 6 (8), 307–314. Ferreira-Dias, S., Sandoval, G., Plou, F., Valero, F., 2013. The potential use of lipases in
David, K., 1935. Lipase production by Penicillium oxalicum and Aspergillus flavus. Int. J. the production of fatty acid derivatives for the food and nutraceutical industries.
Plant Sci. 97, 2. Electron. J. Biotechnol. 16 (3), 12.
David, W., Michel, J., 1999. Extremozymes. Curr. Opin. Chem. Biol. 3, 39–46. Freier, T., Montenegro, R., Shan Koh, H., Shoichet, M.S., 2005. Chitin-based tubes for
Davidson, M.H., 2006. Mechanisms for the hypotriglyceridemic effect of marine omega-3 tissue engineering in the nervous system. Biomaterials 26 (22), 4624–4632.
fatty acids. Am. J. Cardiol. 98 (4A), 27i–33i. Fu, X., Liu, P., Lin, L., Hong, Y., Huang, X., Meng, X., Liu, Z., 2010. A novel endoglucanase
De Souza, M.P., Yoch, D.C., 1995. Comparative physiology of dimethyl sulfide production (Cel9P) from a marine bacterium Paenibacillus sp. BME-14. Appl. Biochem.
by dimethylsulfoniopropionate lyase in Pseudomonas doudoroffii and Alcaligenes sp. Biotechnol. 160 (6), 1627–1636.
strain M3A. Appl. Environ. Microbiol. 61 (11), 3986–3991. Fu, X., Yan, Q., Wang, J., Yang, S., Jiang, Z., 2016. Purification and biochemical char-
Debashish, G., Malay, S., Barindra, S., Joydeep, M., 2005. Marine enzymes. Adv. acterization of novel acidic chitinase from Paenicibacillus barengoltzii. Int. J. Biol.
Biochem. Eng. Biotechnol. 96, 189–218. Macromol. 91, 973–979.
Delattre, C., Fenoradosoa, T.A., Michaud, P., 2011. Galactans: an overview of their most Fu, X.T., Kim, S.M., 2010. Agarase: review of major sources, categories, purification
important sourcing and applications as natural polysaccharides. Braz. Arch. Biol. method, enzyme characteristics and applications. Mar. Drugs 26, 200–218.
Technol. 54, 1075–1092. Fujiwara, N., Masui, A., Imanaka, T., 1993. Purification and properties of the highly
Devi, M.K., Banu, A.R., Gnanaprabhal, G.R., Pradeep, B.V., Palaniswamy, M., 2008. thermostable alkaline protease from an alkaliphilic and thermophilic Bacillus sp. J.
Purification characterization of alkaline protease enzyme from native isolate Biotechnol. 30 (2), 245–256.
Aspergillus niger and compatibility with commercial detergent. Indian J. Sci. Technol. Fujiwara, N., Yamamoto, K., 1987. Production of alkaline protease in a low-cost medium
1 (7), 250–259. by alkalophilic Bacillus sp. and properties of the enzyme. J. Ferment. Technol. 65,
Dhandapani, R., Vijayaragavan, R., 1994. Production of a thermophilic, extracellular 345–348.
alkaline protease by Bacillus stearothermophilus AP-4. World J. Microbiol. Fukamizo, T., 2000. Chitinolytic enzymes: catalysis, substrate binding, and their appli-
Biotechnol. 10 (1), 33–35. cation. Curr. Protein Pept. Sci. 1 (1), 105–124.
Dharmaraj, S., 2010. Marine Streptomyces as a novel source of bioactive substances. Fulzele, R., DeSa, E., Yadav, A., Shouche, V., Bhadekar, R., 2011. Characterization of
World J. Microbiol. Biotechnol. 26 (12), 2123–2139. novel extracellular protease produced by marine bacterial isolate from the Indian
Diaz, J.M., Rodríguez, J.A., Roussos, S., Cordova, J., Abousalham, A., Carriere, F., Baratti, ocean. Braz. J. Microbiol. 42, 1364–1373.
J., 2006. Lipase from the thermotolerant fungus Rhizopus homothallicus is more Furukawa, S.I., Fujikawa, T., Koga, D., Ide, A., 1992. Purification and some properties of
thermostable when produced using solid state fermentation than liquid fermentation exo-type fucoidanases from Vibrio sp. N-5. Biosci. Biotech. Biochem. 56 (11),
procedures. Enzym. Microb. Technol. 39 (5), 1042–1050. 1829–1834.
Dib, R., Chobert, J.M., Dalgalarrondo, M., Barbier, G., Haertle, T., 1998. Purification, Gaber, Y., Mekasha, S., Vaaje-Kolstad, G., Eijsink, V.G., Fraaije, M.W., 2016.
molecular properties and specificity of a thermoactive and thermostable proteinase Characterization of a chitinase from the cellulolytic actinomycete Thermobifida fusca.
from Pyrococcus abyssi, strain st 549, hyperthermophilic archaea from deep-sea hy- Biochim. Et. Biophys. Acta (BBA)-Proteins Proteom. 1864 (9), 1253–1259.
drothermal ecosystem. FEBS Lett. 431, 279–284. Gacesa, P., 1992. Enzymic degradation of alginates. Int. J. Biochem. 24 (4), 545–552.
Dixit, G., Verma, S.C., 1993. Production of alkaline proteases by Penicillium griseofulvin. Gantelet, H., Duchiron, F., 1998. Purification and properties of a thermoactive and
Indian J. Microbiol. 33, 257–260. thermostable pullulanase from Thermococcus hydrothermalis, a hyperthermophilic
Do, H., Lee, J.H., Kwon, M.H., Song, H.E., An, J.Y., Eom, S.H., Kim, H.J., 2013. archaeon isolated from a deep-sea hydrothermal vent. Appl. Microbiol. Biotechnol.
Purification, characterization and preliminary X-ray diffraction analysis of a cold- 49 (6), 770–777.
active lipase (CpsLip) from the psychrophilic bacterium Colwellia psychrerythraea Ghasemi, Y., Rasoul-Amini, S., Ebrahiminezhad, A., Kazemi, A., Shahbazi, M., Talebnia,

147
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

N., 2011. Screening and isolation of extracellular protease producing bacteria from Homaei, A.A., Sajedi, R.H., Sariri, R., Seyfzadeh, S., Stevanato, R., 2010. Cysteine en-
the Maharloo salt lake. Iran. J. Pharm. Sci. 7, 175–180. hances activity and stability of immobilized papain. Amino Acids 38 (3), 937–942.
Goldberg, G.I., Wilhelm, S.M., Kronberger, A., Bauer, E.A., Grant, G.A., Eisen, A.Z., 1986. Homaei, A.A., Sariri, R., Vianello, F., Stevanato, R., 2013b. Enzyme immobilization: an
Human fibroblast collagenase. Complete primary structure and homology to an on- update. J. Chem. Biol. 6 (4), 185–205.
cogene transformation-induced rat protein. J. Biol. Chem. 261 (14), 6600–6605. Hosoda, A., Sakai, M., Kanazawa, S., 2003. Isolation and characterization of agar-de-
Gomez d′Ayala, G., Malinconico, M., Laurienzo, P., 2008. Marine derived polysaccharides grading Paenibacillus spp. associated with the rhizosphere of spinach. Biosci.
for biomedical applications: chemical modification approaches. Molecules 13, Biotechnol. Biochem. 67 (5), 1048–1055.
2069–2106. Hoshino, T., Kageyama, M., 1980. Purification and properties of a binding protein for
Goshev, I., Gousterova, A., Vasileva-Tonkova, E., Nedkov, P., 2005. Characterization of branched-chain amino acids in Pseudomonas aeruginosa. J. Bacteriol. 141 (3),
the enzyme complexes produced by two newly isolated thermophylic actinomycete 1055–1063.
strains during growth on collagen-rich materials. Proc. Biochem. 40, 1627–1631. Hu, Z., Lin, B.K., Xu, Y., Zhong, M.Q., Liu, G.M., 2009. Production and purification of
Gouda, M.K., 2006. Optimization and purification of alkaline proteases produced by agarase from a marine agarolytic bacterium Agarivorans sp. HZ105. J. Appl.
marine Bacillus sp. MIG newly isolated from eastern harbour of Alexandria. Pol. J. Microbiol. 106, 181–190.
Microbiol. 55, 19–126. Huang, Y., Locy, R., Weete, J.D., 2004. Purification and characterization of an extra-
Goyal, K., Selvakumar, P., Hayashi, K., 2001a. Characterization of a thermostable β- cellular lipase from Geotrichum marinum. Lipids 39 (3), 251–257.
glucosidase (BglB) from Thermotoga maritima showing transglycosylation activity. J. Huber, R., Langworthy, T.A., Konig, H., Thomm, M., Woese, C.R., Sleytr, U.B., Stetter,
Mol. Catal. B Enzym. 15, 45–53. K.O., 1986. Thermotoga maritima sp. nov. represents a new genus of unique ex-
Goyal, K., Selvakumar, P., Hayashi, K., 2001b. Characterization of a thermostable β- tremely thermophilic eubacteria growing up to 90 °C. Arch. Microbiol. 144, 324–333.
glucosidase (BglB) from Thermotoga maritima showing transglycosylation activity. J. Hübner, U., Bock, U., Schügerl, K., 1993. Production of alkaline serine protease subtilisin
Mol. Catal. B Enzym. 15, 45–53. Carlsberg by Bacillus licheniformis on complex medium in a stirred tank reactor.
Grace, E.S., 1997. Biotechnology in Seas and Trees. Biotechnology Unzipped: Promises Appl. Microbiol. Biotechnol. 40, 182–188.
and Realities. National Academies Press, Washington, pp. 170. Ilona, K., Zdzislaw, E.S., 2007. Neutral and alkaline muscle proteases of marine fish and
Graham, C.R., David, R.W., Frank, T.R., 1980. Peptone induction and Rifampin-in- invertebrates. Artic. J. Food Biochem. 20 (3), 349–364.
sensitive collagenase production by Vibrio alginolyticus. J. Bacteriol. 142, 447–454. Jackso, N.C.R., Foreman, C.M., Sinsabaugh, R.L., 1995. Microbial enzyme activities as
Grenha, A., Seijo, B., Remunan-Lopez, C., 2005. Microencapsulated chitosan nano- indicator of organic matter processing rates in a lake Erie coastal wetland. Fresh Biol.
particles for lung protein delivery. Eur. J. Pharm. Sci. 25 (4–5), 427–437. 34, 329.
Guo, B., Chen, X.L., Sun, C.Y., Zhou, B.C., Zhang, Y.Z., 2009. Gene cloning, expression Jaeger, K.E., Eggert, T., 2002. Lipases for biotechnology. Curr. Opin. Biotechnol. 13 (4),
and characterization of a new cold-active and salt-tolerant endobeta-1,4-xylanase 390–397.
from marine Glaciecola mesophila KMM 241. Appl. Microbiol. Biotechnol. 84, Jain, D., Pancha, I., Mishra, S.K., Shrivastav, A., Mishra, S., 2012. Purification and
1107–1115. characterization of haloalkaline thermoactive, solvent stable and SDS-induced pro-
Guo, B., Li, P.Y., Yue, Y.S., Zhao, H.L., Dong, S., Song, X.Y., al., e, 2013. Gene cloning, tease from Bacillus sp.: a potential additive for laundry detergents. Bioresour.
expression and characterization of a novel xylanase from the marine bacterium, Technol. 115, 228–236.
Glaciecola mesophila KMM241. Mar. Drugs 11, 1173–1187. Je, J.Y., Kim, S.K., 2006. Antimicrobial action of novel chitin derivative. Biochim.
Gupta, R., Gigras, P., Mohapatra, H., Goswami, V.K., Chauhan, B., 2003. Microbial α- Biophys. Acta 1760 (1), 104–109.
amylases: a biotechnological perspective. Process Biochem. 38, 1599–1616. Jedrzejczak, M.F., 1999. The degradation of stranded carrion on a Baltic Sea sandy beach.
Haddar, A., Agrebi, R., Bougatef, A., Hmidet, N., Sellami-Kamoun, A., Nasri, M., 2009a. Oceanol. Stud. 3, 109.
Two detergent stable alkaline serine-proteases from Bacillus mojavensis A21: pur- Jenkins, D.W., Hudson, S.M., 2001. Review of vinyl graft copolymerization featuring
ification, characterization and potential application as a laundry detergent additive. recent advances toward controlled radical-based reactions and illustrated with
Bioresour. Technol. 100 (13), 3366–3373. chitin/chitosan trunk polymers. Chem. Rev. 101 (11), 3245–3273.
Haddar, A., Agrebi, R., Bougatef, A., Hmidet, N., Sellami-Kamoun, A., Nasri, M., 2009b. Jensen, R.G., 1983. Detection and determination of lipase (acylglycerol hydrolase) ac-
Two detergent stable alkaline serine-proteases from Bacillus mojavensis A21: pur- tivity from various sources. Lipids 18 (9), 650–657.
ification, characterization and potential application as a laundry detergent additive. Ji, C., Cao, X., Yao, C., Xue, S., Xiu, Z., 2014. Protein-protein interaction network of the
Bioresour. Technol. 100, 3366–3373. marine microalga Tetraselmis subcordiformis: prediction and application for starch
Hamedi, J., Mohammadipanah, F., Ventosa, A., 2013. Systematic and biotechnological metabolism analysis. J. Ind. Microbiol. Biotechnol. 41 (8), 1287–1296.
aspects of halophilic and halotolerant actinomycetes. Extremophiles 17 (1), 1–13. Jiang, Z., Le Bail, A., Wu, A., 2008. Effect of the thermostable xylanase B (XynB) from
Han, W.J., Gu, J.Y., Liu, H.H., Li, F.C., Wu, Z.H., Li, Y.Z., 2013. An extra peptide within Thermotoga maritima on the quality of frozen partially baked bread. J. Cereal Sci. 47,
the catalytic module of a β-agarase affects the agarose degradation pattern. J. Biol. 172–179.
Chem. 288, 9519–9531. Jiang, Z.Q., Deng, W., Zhu, Y.P., Li, L.T., Sheng, Y.J., Hayashi, K., 2004. The recombinant
Hardeman, F., Sara Sjoling, S., 2007. Metagenomic approach for the isolation ofa novel xylanase B of Thermotoga maritima is highly xylan specific and produces exclusively
low-temperature-active lipase from uncultured bacteria of marine sediment. FEMS xylobiose from xylans, a unique character for industrial applications. J. Mol. Catal. B
Microbiol. Ecol. 59, 524–534. Enzym. 27, 207–213.
Harper, E., Seifter, S., Hospelhorn, V.D., 1965. Evidence for subunits in bacterial col- Jing, H., Su, W., Caracci, S., Bunning, T.G., Cooper, T., Adams, W., 1996. Optical wa-
lagenase. Biochem. Biophys. Res Commun. 18, 627–632. veguiding and morphology of chitosan thin film. J. Appl. Polym. Sci. 61 (7),
Henrissat, B., 1991. A classification of glycosyl hydrolases based on amino acid sequence 1163–1171.
similarities. Biochem. J. 280, 309–316. Joo, H.S., Choi, J.W., 2012. Purification and characterization of a novel alkaline protease
Henrissat, B., Coutinho, P., Deleury, E., 2005. CAZy-Carbohydrate- Active Enzymes: from Bacillus horikoshii. J. Microbiol. Biotechnol. 22 (1), 58–68.
Polysaccharide Lyase Families. fr/_cazy/CAZY/index. Html. Jorquera, M., Martinez, O., Maruyama, F., Marschner, P., de la Luz Mora, M., 2008.
Hernandez-Saavedra, N., Ochoa, J.L., 1999. Copper-zinc superoxide dismutase from the Current and future biotechnological applications of bacterial phytases and phytase-
marine yeast Debaryomyces hansenii. Yeast 15 (8), 657–668. producing bacteria. Microb. Environ. 23 (3), 182–191.
Hisano, T., Nishimura, M., Yonemoto, Y., Abe, S., Yamashita, T., Sakaguchi, K., Kimura, Kamada, M., Oda, K., Murao, S., 1972. The purification of the extracellular acid protease
A., Murata, K., 1993. Bacterial Alginate lyase highly active on acetylated alginates. J. of Rhodotorula glutinis K-24 and its general properties. Agric. Biol. Chem. 36 (7),
Ferment. Bioeng. 75, 332–335. 1095–1101.
Homaei, A., 2015a. Enhanced activity and stability of papain immobilized on CNBr-ac- Kang, W., Xu, Y., Qin, L., Wang, Y., 2010. Effects of Different β-D-glycosidases on bound
tivated sepharose. Int. J. Biol. Macromol. 75, 373–377. aroma compounds in muscat grape determined by HS- SPME and GC-MS. J. Inst.
Homaei, A., 2015b. Purification and biochemical properties of highly efficient alkaline Brew. 116, 70–77.
phosphatase from Fenneropenaeus merguiensis brain. J. Mol. Catal. B: Enzym. 118, Kasana, R.C., Yadav, S.K., 2007. Isolation of a psychrotrophic Exiguobacterium sp. SKPB5
16–22. (MTCC 7803) and characterization of its alkaline protease. Curr. Microbiol. 54 (3),
Homaei, A., Barkheh, H., Sariri, R., Stevanato, R., 2014. Immobilized papain on gold 224–229.
nanorods as heterogeneous biocatalysts. Amino Acids 46 (7), 1649–1657. Kashyap, P., Sabu, A., Pandey, A., Szakacs, G., Soccol, C.R., 2002. Extra-cellular L-glu-
Homaei, A., Etemadipour, R., 2015. Improving the activity and stability of actinidin by taminase production by Zygosaccharomyces rouxii under solid-state fermentation.
immobilization on gold nanorods. Int. J. Biol. Macromol. 72, 1176–1181. Process Biochem. 38 (3), 307–312.
Homaei, A., Ghanbarzadeh, M., Monsef, F., 2016a. Biochemical features and kinetic Katikala, P.K., Bobbarala, V., Tadimalla, P., Guntuku, G.S., 2009. Screening of L-gluta-
properties of α-amylases from marine organisms. Int. J. Biol. Macromol. 83, 306–314. minase producing marine bacterial cultures for extracellular production of L-gluta-
Homaei, A., Lavajoo, F., Sariri, R., 2016b. Development of marine biotechnology as a minase. Int. J. ChemTech Res. 1, 1232–1235.
resource for novel proteases and their role in modern biotechnology. Int. J. Biol. Kavaz, D., Odabas, S., Güven, E., Demirbilek, M., Denkbas, E., 2010. Bleomycin loaded
Macromol. 88, 542–552. magnetic chitosan nanoparticles as multifunctional nanocarriers. J. Bioact. Compat.
Homaei, A., Saberi, D., 2015. Immobilization of α-amylase on gold nanorods: an ideal Polym. 25 (3), 305–318.
system for starch processing. Process Biochem. 50 (9), 1394–1399. Kempka, A.P., Lipke, N.L., da Luz Fontoura Pinheiro, T., Menoncin, S., Treichel, H.,
Homaei, A., Samari, F., 2017. Investigation of activity and stability of papain by ad- Freire, D.M., Di Luccio, M., de Oliveira, D., 2008. Response surface method to opti-
sorption on multi-wall carbon nanotubes. Int. J. Biol. Macromol. mize the production and characterization of lipase from Penicillium verrucosum in
Homaei, A., Stevanato, R., Etemadipour, R., Hemmati, R., 2017. Purification, catalytic, solid-state fermentation. Bioprocess Biosyst. Eng. 31 (2), 119–125.
kinetic and thermodynamic characteristics of a novel ficin from Ficus johannis. Kerkhoffs, P.L., 1981. & . Preparation of fructose. In 4277563 (Ed.), (US Patent).
Biocatal. Agric. Biotechnol. 10, 360–366. Khalil Beg, Q., Gupta, R., 2003. Purification and characterization of an oxidation-stable,
Homaei, A.A., Mymandi, A.B., Sariri, R., Kamrani, E., Stevanato, R., Etezad, S.-M., Khajeh, thiol-dependent serine alkaline protease from Bacillus mojavensis. Enzym. Microb.
K., 2013a. Purification and characterization of a novel thermostable luciferase from Technol. 32, 294–304.
Benthosema pterotum. J. Photochem. Photobiol. B: Biol. 125, 131–136. Khambhaty, Y., Mody, K., Jha, B., 2007. Purification and characterization of k-

148
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

carrageenase from a novel g-Proteobacterium, Pseudomonas elongata (MTCC 5261) Leon, O., Quintana, L., Peruzzo, G., Slebe, J.C., 1992. Purification and properties of an
syn Microbulbifer elongates comb. nov. Biotechnol. Bioprocess Eng. 12, 668–675. extracellular agarase from Alteromonas sp. strain C-1. Appl. Environ. Microbiol 58
Khor, E., 2001. Chitin: fulfilling a biomaterials promise. (12), 4060–4063.
Kikani, B.A., Singh, S.P., 2012. The stability and thermodynamic parameters of a very Li, H., Chi, Z., Duan, X., Wang, L., Sheng, J., Wu, L., 2007. Glucoamylase production by
thermostable and calcium-independent -amylase from a newly isolated bacterium. the marine yeast Aureobasidium pullulans N13d and hydrolysis of potato starch
Anoxybacillus beppuensis TSSC-1. Process Biochem. 47, 1791–1798. granules by the enzyme. Process Biochem. 42, 462–465.
Kim, B.J., Kim, H.J., Ha, S.D., Hwang, S.H., Byun, D.S., Lee, T.H., Kong, J.Y., 1999. Li, H.F., Chi, Z.M., Wang, X.H., Ma, C.L., 2007. Amylase production by the marine yeast
Purification and characterization of -agarase from marine bacterium Bacillus cereus Aureobasidium pullulans N13d. J. Ocean Univ. Chin. 6, 61–66.
ASK202. Biotechnol. Lett. 21, 1011–1015. Li, J., Yang, J., Zhu, W.Y., He, J., Tian, X.P., Xie, Q., al., e, 2012. Nocardiopsis coralliicola
Kin, T., Johnson, P.R., Shapiro, A.M., Lakey, J.R., 2007. Factors influencing the col- sp. nov., isolated from the gorgonian coral, Menella praelonga. Int. J. Syst. Evol.
lagenase digestion phase of human islet isolation. Transplantation 83 (1), 7–12. Microbiol. 62, 1653–1658.
Kinsella, L.E., 1986. Food components with potential therapeutic benefits: the n-3 poly- Li, J.W., Dong, S., Song, J., Li, C.B., Chen, X.L., Xie, B.B., Zhang, Y.Z., 2011. Purification
unsaturated fatty acids with fish oils. Food Technol. 40 (2), 89–97. and characterization of a bifunctional alginate lyase from Pseudoalteromonas sp.
Kirimura, K., Masuda, N., Iwasaki, Y., Nakagawa, H., Kobayashi, R., Usami, S., 1999b. SM0524. Mar. Drugs 9 (1), 109–123.
Purification and characterization of a novel β-agarase from an alkalophilic bac- Lian, Q., Li, D., Jin, Z., Wang, J., Li, A., Wang, Z., Jin, Z., 2009. Fabrication and in vitro
terium, Alteromonas sp. E-1. J. Biosci. Bioeng. 87 (4), 436–441. evaluation of calcium phosphate combined with chitosan fibers for scaffold struc-
Kita, K., Ishimaru, K., Teraoka, M., Yanase, H., Kato, N., 1995. Properties of poly (3- tures. J. Bioact. Compat. Polym. 24 (1), 113–124.
hydroxybutyrate) depolymerase from a marine bacterium. Alcaligenes faecalis AE122. Lima, C.A., Rodrigues, P.M.B., Porto, T.S., Viana, D.A., Lima Filho, J.L., Porto, A.L.F.,
Appl. Environ. Microbiol. 61 (5), 1727–1730. Cunha, M.G.C., 2009. Production of a collagenase from Candida albicans URM362.
Klemm, D., Heublein, B., Fink, H.P., Bohn, A., 2005. Cellulose: fascinating biopolymer Biochem. Eng. J. 43, 315–320.
and sustainable raw material. Angew. Chem. Int Ed. Engl. 44 (22), 3358–3393. Lima, D.M., Fernandes, P., Nascimento, D.S., Ribeiro, R.C.L.F., de Assis, S.A.A., 2011.
Klock, G., Kowalski, M.B., Hering, B.J., Eiden, M.E., Weidemann, A., Langer, S., Fructose syrup: a biotechnology asset. Food Technol. Biotechnol. 49, 424–434.
Zimmermann, U., Federlin, K., Bretzel, R.G., 1996. Fractions from commercial col- Lima, R.N., Porto, A.L.M., 2016. Chapter eight-recent advances in marine enzymes for
lagenase preparations: use in enzymic isolation of the islets of Langerhans from biotechnological processes. Adv. Food Nutr. Res. 78, 153–192.
porcine pancreas. Cell Transplant. 5 (5), 543–551. Liu, J., Zhang, Z., Dang, H., Jianren Lu, J., Cui, Z., 2011a. Isolation and characterization
Kobayashi, T., Koide, O., Mori, K., Shimamura, S., Matsuura, T., Miura, T., Takaki, Y., of a cold-active amylase from marine Wangia sp. C52. Afr. J. Microbiol. Res. 5 (10),
Morono, Y., Nunoura, T., Imachi, H., Inagaki, F., Takai, K., Horikoshi, K., 2008. 1156–1162.
Phylogenetic and enzymatic diversity of deep subseafloor aerobic microorganisms in Liu, J., Zhang, Z., Zhu, H., Dang, H., Lu, J., Cui, Z., 2011b. Isolation and characterization
organics- and methane-rich sediments off Shimokita Peninsula. Extremophiles 12 (4), of α-amylase from marine Pseudomonas sp. K6-28-040. Afr. J. Biotechnol. 10 (14),
519–527. 2733–2740.
Kuberan, T., Sangaralingam, S., Thirumalaiarasu, V., 2010. Isolation and optimization of Liu, X., Huang, Z., Zhang, X., Shao, Z., Liu, Z., 2014. Cloning, expression and char-
protease producing bacteria from halophilic soil. J. Biol. Sci. Res. 1, 163–174. acterization of a novel cold-active and halophilic xylanase from Zunongwangia
Kumar, C., Joo, H.S., Koo, Y.M., Paik, S., Chang, C.S., 2004. Thermostable alkaline pro- profunda. Extremophiles 18 (2), 441–450.
tease from a novel marine haloalkalophilic Bacillus Clausii isolate. World J. Microbiol. Liu, Y., Lei, Y., Zhang, Z., Gao, Y., Xiao, Y., Peng, H., 2012. Identification and phyloge-
Biotechnol. 20, 351–357. netic characterization of a new subfamily of α-amylase enzymes from marine mi-
Kumar, C.G., Takagi, H., 1999. Microbial alkaline proteases: from a bioindustrial view- croorganisms. Mar. Biotechnol. 14 (3), 253–260.
point. Biotechnol. Adv. 17 (7), 561–594. Lu, W.D., Li, A.X., Guo, Q.L., 2014. Production of novel alkalitolerant and thermostable
Kumar, M.N.V.R., Muzarelli, R.A.A., Muzarelli, C., Sashiwa, H., Domb, A.J., 2004. inulinase from marine actinomycete Nocardiopsis sp. DN-K15 and inulin hydrolysis
Chitosan chemistry and pharmaceutical perspectives. Chem. Rev. 104 (12), by the enzyme. Ann. Microbiol. 64, 441–449.
6017–6084. Ma, C., Lu, X., Shi, C., Li, J., Gu, Y., Ma, Y., Yu, W., 2007. Molecular cloning and char-
Kumar, R., Sharma, J., Singh, R., 2007. Production of tannase from Aspergillus ruber under acterization of a novel β-agarase, AgaB, from marine Pseudoalteromonas sp. CY24. J.
solid-state fermentation using jamun (Syzygium cumini) leaves. Microbiol. Res. 162, Biol. Chem. 282 (6), 3747–3754.
384–390. Madhumathi, K., Sudheesh Kumar, P.T., Abilash, S., Sreeja, V., Tamura, H., Manzoor, K.,
Kumar, S.R., Chandrasekaran, M., 2003. Continuous production of L-glutaminase by an al., e, 2010. Development of novel chitin/nanosilver composite scaffolds for wound
immobilized marine Pseudomonas sp. BTMS-51 in a packed bed reactor. Process dressing applications. J. Mater. Sci.: Mater. Med. 21 (2), 807–813.
Biochem. 38 (10), 1431–1436. Malathi, S., Chakraborty, R., 1991. Production of alkaline protease by a new Aspergillus
Kuraishi, C., Yamazaki, K., Susa, Y., 2001. Transglutaminase: its utilization in the food flavus isolate under solidsubstrate fermentation conditions for use as a depilation
industry. Food Rev. Int 17, 221–246. agent. Appl. Environ. Microbiol. 57, 712–716.
Kurita, K., 2006. Chitin and chitosan: functional biopolymers from marine crustaceans. Manachini, P., Fortina, M., Parini, C., 1988. Thermostable alkaline protease produced by
Mar. Biotechnol. (NY) 8 (3), 203–226. Bacillus thermoruber a new species of Bacillus. Appl. Microbiol. Biotechnol. 28,
Laemmeli, U., 1970. Cleavage of structural proteins during the assemble of the head of 409–413.
bacteriophage T4. Nature 227, 680–685. Manivasagan, P., Venkatesan, J., Kang, K.H., Sivakumar, K., Park, S.J., Kim, S.K., 2015.
Lakshmikanth, M., Manohar, S., Lalitha, J., 2009. Purification and characterization of β- Production of α-amylase for the biosynthesis of gold nanoparticles using
agarase from agar-liquefying soil bacterium Acinetobacter sp., AG LSL-1. Process Streptomyces sp. MBRC-82. Int. J. Biol. Macromol. 72, 71–78.
Biochem. 44 (9), 999–1003. Manivasagan, P., Venkatesan, J., Sivakumar, K., Kim, S.K., 2013. Marine actinobacterial
Lan, D., Qu, M., Yang, B., Wang, Y., 2016. Enhancing production of lipase MAS1 from metabolites: current status and future perspectives. Microbiol. Res. 168 (6), 311–332.
marine Streptomyces sp. strain in Pichia pastoris by chaperones co-expression. Manivasagan, P., Venkatesan, J., Sivakumar, K., Kim, S.K., 2014. Pharmaceutically active
Electron. J. Biotechnol. 22, 62–67. secondary metabolites of marine actinobacteria. Microbiol. Res. 169 (4), 262–278.
Lario, L.D., Chaud, L., das Graças Almeida, M., Converti, A., Sette, L.D., Pessoa, A., 2015. Manjit, A.Y., Aggarwal, N.K., Kumar, K., Kumar, A., 2008. Tannase production by
Production, purification, and characterization of an extracellular acid protease from Aspergillus fumigatus MA under solid-state fermentation. World J. Microbiol.
the marine Antarctic yeast Rhodotorula mucilaginosa L7. Fungal Biol. 119 (11), Biotechnol. 24 (12), 3023–3030.
1129–1136. Mansouri, S., Lavigne, P., Corsi, K., Benderdour, M., Beaumont, E., Fernandes, J.C., 2004.
Lario, L.D., Malpiedi, L.P., Pereira, J.F.B., Sette, L.D., Pessoa-Junior, A., 2016. Liquid- Chitosan-DNA nanoparticles as non-viral vectors in gene therapy: strategies to im-
liquid extraction of protease from cold-adapted yeast Rhodotorula mucilaginosa L7 prove transfection efficacy. Eur. J. Pharm. Biopharm. 57 (1), 1–8.
using biocompatible and biodegradable aqueous two-phase systems. Sep. Sci. Mao, W., Pan, R., Freedman, D., 1992. High production of alkaline protease by Bacillus
Technol. 51 (1), 57–67. licheniformis in a fed-batch fermentation using a synthetic medium. J. Ind. Microbiol.
Laroche, M., Ulber, R., 2002. Natural Products from Marine Microorganisms. 11, 1–6.
(International Symposiumon Natural Products from Marine Microorganisms). Marokhazi, J., Koczan, G., Hudecz, F., Graf, L., Fodor, A., Venekei, I., 2004. Enzymic
Greifswald, Germany. characterization with progress curve analysis of a collagen peptidase from an en-
Lee, H.S., Kwon, K.K., Kang, S.G., Cha, S.S., Kim, S.J., Lee, J.H., 2010. Approaches for thomopathogenic bacterium, Photorhabdus luminescens. Biochem. J. 379 (3),
novel enzyme discovery from marine environments. Curr. Opin. Biotechnol. 21 (3), 633–640.
353–357. Martin, M., Portetelle, D., Michel, G., Vandenbol, M., 2014. Microorganisms living on
Lee, Y., Oh, C., De Zoysa, M., Kim, H., Wickramaarachchi, W.D., Whang, I., Lee, J., 2013. macroalgae: diversity, interactions, and biotechnological applications. Appl.
Molecular cloning, overexpression and enzymatic characterization of glycosyl hy- Microbiol. Biotechnol. 98 (7), 2917–2935.
drolase family 16 β-agarase from marine bacterium Saccharophagus sp. AG21 in Maruthiaha, T., Somanathb, B., Immanuela, G., Palavesam, A., 2015. Deproteinization
Escherichia coli. J. Microbiol. Biotechnol. 23 (7), 913–922. potential and antioxidant property of haloalkalophilic organic solvent tolerant pro-
Lee, Y.H., Jun, S.E., Shin, H.D., 2005. Low molecular weight agarose-specific alpha- tease from marine Bacillus sp. APCMST-RS3 using marine shell wastes. Biotechnol.
agarase from agarolytic marine microorganism Pseudoalteromonas sp. BL-3 which Rep. 8, 124–132.
hydrolyzes alpha-1,3-glycoside bond of agar or agarose to produce agarobiose and Matoba, S., Fukayama, J., Wing, R.A., Ogrydziak, D.M., 1988. Intracellular precursors and
agarotetraose. In,vol.KR2005079035–A). secretion of alkaline extracellular protease of Yarrowia lipolytica. Mol. Cell Biol. 8
Legin, E., Ladrat, C., Godfroy, A., Barbier, G., Duchiron, F., 1997. Thermostable amylo- (11), 4904–4916.
lytic enzymes of thermophilic microorganisms from deep-sea hydrothermal vents. Matsumoto, M., Yokouchi, H., Suzuki, N., Ohata, H., Matsunaga, T., 2003.
Comptes rendus de l'Academiel′Academie des sciences Ser. 3, Sci. De. la vie 320 (11), Saccharification of marine microalgae using marine bacteria for ethanol production.
893–898. Appl. Biochem. Biotechnol. 105–108, 247–254.
Lei, X.G., Porres, J.M., 2003. Phytase enzymology, applications, and biotechnology. Matsushita, O., Yoshihara, K., Katayama, S., Minami, J., Okabe, A., 1994. . Purification
Biotechnol. Lett. 25 (21), 1787–1794. and characterization of Clostridium perfringens 120-kilodalton collagenase and

149
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

nucleotide sequence of the corresponding gene. J. Bacteriol. 176 (1), 149–156. Parro, V., Mellado, R.P., 1994. Effect of glucose on agarase overproduction by
Meenakshi, S., Umayaparvathi, S., Manivasagan, P., Arumugam, M., Balasubramanian, T., Streptomyces. Gene 145 (1), 49–55.
2013. Purification and characterization of inulinase from marine bacterium Bacillus Patnala, H.S., Kabilan, U., Gopalakrishnan, L., Rao, R.M., Kumar, D.S., 2016. Marine
cereuscereus MU. Indian J. Geo-Mar. Sci. 31. fungal and bacterial isolates for lipase production: a comparative study. Adv. Food
Menon, G., Mody, K., Keshri, J., Jha, B., 2010. Isolation, purification, and characteriza- Nutr. Res 78, 71–94.
tion of haloalkaline xylanase from a marine Bacillus pumilus strain, GESF-1. Petrova, D., Derekova, A., Vlahov, S., 2006. Purification and properties of individual
Biotechnol. Bioprocess Eng. 15 (6), 998–1005. collagenases from Streptomyces sp. strain 3B. Folia Microbiol. (Praha) 51 (2), 93–98.
Mesbah, N.M., Weigel, J., 2014. Purification and biochemical characterization of halo- PO, Remba, K., 1995. Hydrolytic enzymatic activity in deep-sea sediments. FEMS Microb.
philic, alkalithermophilic protease AbCP from Alkalibacillus sp. NM-Fa4. J. Mol. Ecol. 16, 213.
Catal. B: Enzym. 105, 74–81. Potin, P., Richard, C., Rochas, C., Kloareg, B., 1993. Purification and characterization of
Michels, P.C., Clark, D.S., 1997. Pressure enhanced activity and stability of a hy- the α-agarase from Alteromonas agarlyticus (Cataldi) comb. nov., strain GJ1B. Eur. J.
perthermophilic protease from a deep sea methanogen. Appl. Environ. Microbiol. 63, Biochem. 214, 599–607.
3985–3991. Potin, P., Sanseau, A., Le Gall, Y., Rochas, C., Kloareg, B., 1991. Purification and char-
Minegishi, H., Shimane, Y., Echigo, A., Ohta, Y., Hatada, Y., Kamekura, M., al., e, 2013. acterization of a new kappacarrageenase from a marine Cytophaga-like bacterium.
Thermophilic and halophilic β-agarase from a halophilic archaeon Halococcus sp. Eur. J. Biochem. 201, 241–247.
197A. Extremophiles 17, 931–939. Raghukumar, C., D’Souza, T.M., Thorn, R.G., Reddy, C.A., 1999. Lignin modifying en-
Miyazaki, S., Ishii, K., Nadai, T., 1981. The use of chitin and chitosan as drug carriers. zymes of Flavodon flavus, a basidiomycete isolated from a coastal marine environ-
Chem. Pharm. Bull. (Tokyo) 29 (10), 3067–3069. ment. Appl. Environ. Microbiol. 65, 2103–2111.
Mo, S., Kim, J.H., Cho, K.W., 2009. Enzymatic properties of an extracellular phospholi- Raghukumar, C., Muraleedharan, U., Gaud, V.R., Mishra, R., 2004. Xylanases of marine
pase C purified from a marine streptomycete. Biosci. Biotechnol. Biochem. 73 (9), fungi of potential use for biobleaching of paper pulp. J. Ind. Microbiol. Biotechnol. 31
2136–2137. (9), 433–441.
Mohapatra, B.R., 1997. Properties of carboxymethylcellulase (endo-1,4-β-glucanase) Rahman, R.N.Z.A., Razak, C.N., Ampon, K., Basri, M., Yunus, W.M.Z.W., Salleh, A.B.,
from Bacillus sp. isolated from the marine sponge (Axinella sp.). Indian J. Mar. Sci. 26, 1994. Purification and characterization of a heat-stable alkaline protease from
292–296. Bacillus stearothermophilus F1. Appl. Microbiol. Biotechnol. 40, 822–827.
Mohapatra, B.R., Banerjee, U.C., Bapuji, M., 1998. Characterization of a fungal amylase Ramakrishnan, V., Goveas, L.C., Suralikerimath, N., Jampani, C., Halami, P.M., Narayan,
from Mucor sp. associated with the marine sponge Spirastrella sp. J. Biotechnol. 60, B., 2016. Extraction and purification of lipase from Enterococcus faecium MTCC5695
113–117. by PEG/phosphate aqueous-two phase system (ATPS) and its biochemical char-
Mohebbi, G.H., Nabipour, I., Vazirizadeh, A., 2014. The sea, the future pharmacy. Iran. acterization. Biocatal. Agric. Biotechnol. 6, 19–27.
South Med. J. 17 (4), 748–788. Ramani, K., Saranya, P., Jain, S.C., Sekaran, G., 2013. Lipase from marine strain using
Mollers, K.B., Cannella, D., Jorgensen, H., Frigaard, N.U., 2014. Cyanobacterial biomass cooked sunflower oil waste: production optimization and application for hydrolysis
as carbohydrate and nutrient feedstock for bioethanol production by yeast fermen- and thermodynamic studies. Bioprocess Biosyst. Eng. 36 (3), 301–315.
tation. Biotechnol. Biofuels 7, 64. Ramos, K.R.M., Valdehuesa, K.N.G., Cabulong, R.B., Moron, L.S., Nisola, G.M., Hong, S.K.,
Mondal, K.C., Banerjee, D., Banerjee, R., Pati, B.R., 2001. Production and characterization Chung, W.J., 2016. Overexpression and secretion of AgaA7 from Pseudoalteromonas
of tannase from Bacillus cereus KBR9. J. General. Appl. Microbiol. 47 (5), 263–267. hodoensis sp. nov in Bacillus subtilis for the depolymerization of agarose. Enzym.
Mookhtiar, K.A., Steinbrink, D.R., Van Wart, H.E., 1985. Mode of hydrolysis of collagen- Microb. Technol. 90, 19–25.
like peptides by class I and class II Clostridium histolyticum collagenases: evidence Rasmussen, R.S., Morrissey, M.T., 2007. Marine biotechnology for production of food
for both endopeptidase and tripeptidylcarboxypeptidase activities. Biochemistry 24 ingredients. Adv. Food Nutr. Res. 52, 237–292.
(23), 6527–6533. Rattray, F.P., Bockelmann, W., Fox, P.F., 1994. Purification and characterization of an
Mudryk, Z.J., Podgórska, B., 2006. Enzymatic activity of bacterial strains isolated from extracellular proteinase from Brevibacterium linens ATCC 9174. Appl. Environ.
marine beach sediments. Pol. J. Environ. Stud. 15 (3), 441–448. Microbiol. 61, 3454–3456.
Mullaney, E.J., Daly, C.B., Kim, T., Porres, J.M., Lei, X.G., Sethumadhavan, K., Ullah, Raval, V.H., Pillai, S., Rawal, C.M., Singh, S.P., 2014. Biochemical and structural char-
A.H., 2002. Site-directed mutagenesis of Aspergillus niger NRRL 3135 phytase at re- acterization of a detergent-stable serine alkaline protease from seawater haloalk-
sidue 300 to enhance catalysis at pH 4.0. Biochem. Biophys. Res. Commun. 297 (4), aliphilic bacteria. Process Biochem. 49 (6), 955–962.
1016–1020. Renge, V.C., Khedkar, S.V., Nandurkar, N.R., 2012. Enzyme synthesis by fermentation
Murielle, J.A.M., Flament, D., Allouch, J., Potin, P., Thion, L., Kloareg, B., Barbeyron, T., method. Sci. Rev. Chem. Commun. 2, 585–590.
2005. The endo-β-agarases AgaA and AgaB from the marine bacterium Zobellia ga- Renner, M.J., Breznak, J.A., 1998. Purification and properties of ArfI, an alpha-L-arabi-
lactanivorans: two paralogue enzymes with different molecular organizations and nofuranosidase from Cytophaga xylanolytica. Appl. Environ. Microbiol. 64 (1),
catalytic behaviours. Biochem. J. 385 (3), 703–713. 43–52.
Mustranta, A., Forssell, P., Poutanen, K., 1995. Comparison of lipases and phospholipases Ricca, E., Calabro, V., Curcio, S., Iorio, G., 2007. The state of the art in the production of
in the hydrolysis of phospholipids. Process Biochem. 30, 393–401. fructose from inulin enzymatic hydrolysis. Crit. Rev. Biotechnol. 27 (3), 129–145.
Nadeem, M., Qazi, J.I., Baij, S., Syed, Q.A., 2008. Effect of medium composition on Roberts, J.N., Christopher, B.B., Cynthia, D.T., Rhonda, K., Marcelino, B., Peter, L.C.,
commercially important alkaline protease production by Bacillus licheniformis N-2. Douglas, R.L., John, T., 2007. Genital transmission of HPV in a mouse model is po-
Food Technol. Biotech. 46 (4), 388–394. tentiated by nonoxynol-9 and inhibited by carrageenan. Nat. Med. 13, 857–861.
Naganuma, T., Coury, D.A.M.P.A.G., Horikoshi, K., 1993. Characterization of agarolytic Roberts, R.L., Cabib, E., 1982. Serratia marcescens chitinase: one-step purification and
microscilla isolates and their extracellular agarases. Syst. Appl. Microbiol. 16, use for the determination of chitin. Anal. Biochem. 127, 402–412.
183–190. Rochima, E., Sekar, N., Buwono, I.D., Afrianto, E., Pratama, R.I., 2016. Isolation and
Najafi, M.F., Kembhavi, A., 2005. One step purification and characterization of an ex- Characterization of Collagenase from Bacillus subtilis (Ehrenberg, 1835); ATCC 6633
tracellular α-amylase from marine Vibrio sp. Enzym. Microb. Technol. 36 (4), for Degrading Fish Skin Collagen Waste from Cirata Reservoir.
535–539. Roseline, T.L., Sachindra, N.M., 2016. Characterization of extracellular agarase produc-
Nelson, G., Young, W.T., 1987. Extracellular acid and alkaline protease from Candida olea. tion by Acinetobacter junii PS12B, isolated from marine sediments. Biocatal. Agric.
J. General. Microbiol. 133, 1461–1469. Biotechnology 6, 219–226.
Ngo, D.N., Kim, M.M., Kim, S.K., 2008. Chitin oligosaccharides inhibit oxidative stress in Rouzbahan, S., Moein, S., Homaei, A., 2016. Total antioxidant capacities and reduction of
live cells. Carbohydr. Polym. 74, 228–234. ferric ions by extracts of three medicinal plants and their fractions. Bimon. J.
Niehaus, F., Bertoldo, C., Kähler, M., Antranikian, G., 1999. Extremophiles as a source of Hormozgan Univ. Med. Sci. 20 (4), 245–254.
novel enzymes for industrial application. Appl. Microbiol. Biotechnol. 51, 711–729. Rouzbehan, S., Moein, S., Homaei, A., Moein, M.R., 2017. Kinetics of α-glucosidase in-
Nishiyama, Y., Langan, P., Chanzy, H., 2002. Crystal structure and hydrogen-bonding hibition by different fractions of three species of Labiatae extracts: a new diabetes
system in cellulose Ibeta from synchrotron X-ray and neutron fiber diffraction. J. Am. treatment model. Pharm. Biol. 55 (1), 1483–1488.
Chem. Soc. 124 (31), 9074–9082. Sabu, A., Keerthi, T.R., Kumar, S.R., Chandrasekaran, M., 2000. L-Glutaminase produc-
Nordwig, A., Jahin, W.F., 1968. A collagenolytic enzyme from aspergillus oryzae pur- tion by marine Beauveria sp. under solid state fermentation. Process Biochem. 35,
ification and properties. Eur. J. Biochem. 3, 519–529. 705–710.
Noureddini, H., Dang, J., 2009. Degradation of phytates in distillers' grains and corn Sabu, A., Pandey, A., Daud, M.J., G, S, 2005. Tamarind seed powder and palm kernel
gluten feed by Aspergillus niger phytase. Appl. Biochem. Biotechnol. 159 (1), 11–23. cake: two novel agro residues for the production of tannase under solid state fer-
Ohta, Y., Hatada, Y., 2006. A novel enzyme, lambda-carrageenase, isolated from a deep- mentation by Aspergillus niger ATCC 16620. Bioresour. Technol. 96 (11), 1223–1228.
sea bacterium. J. Biochem. 140 (4), 475–481. Sadati, R., Barghi, A., 2014. Urmia hypersaline lake: a potential source of actinomycets
Oren, A., 2004. Prokaryote diversity and taxonomy: current status and future challenges. possessing. Iran. J. Public Health 43 (2), 149.
Philos. Trans. R. Soc. Lond. B Biol. Sci. 359 (1444), 623–638. Santorelli, M., Maurelli, L., Pocsfalvi, G., Fiume, I., Squillaci, G., La Cara, F., del Monaco,
Osama, R., Koga, T., 1995. An investigation of aquatic bacteria capable of utilizing chitin G., Morana, A., 2016. Isolation and characterisation of a novel alpha-amylase from
as the sole source of nutrients. Lett. Appl. Microbiol. 21, 288–291. the extreme haloarchaeon Haloterrigena turkmenica. Int. J. Biol. Macromol. 92,
Palanisami, S., Kannan, K., Lakshmanan, U., 2011. Tannase activity from the marine 174–184.
bacterium Phormidium valderium BDU 140441. J. Appl. Phycol. Sapelli, P.L., Baldassarre, V., Muzzarelli, R.A.A., Emanuelli, M., 1986. Chitosan in den-
Pallavi, P., Chandra, S.J., Reddy, V.K., Reddy, S.R., 2014. Optimization of cultural tistry. In: Muzzarelli, R., Jeuniaux, G., Gooday, G.W. (Eds.), Chitin in Nature and
parameters for lipase production by Bacillus subtilis Y-IVI. Int. J. Curr. Microbiol. Technology. Springer, US, pp. 507–512.
Appl. Sci. 3 (12), 194–200. Sarethy, I.P., Saxena, Y., Kapoor, A., Sharma, M., Sharma, S.K., Gupta, V., Gupta, S., 2011.
Pandey, A., Szakacs, G., Soccol, C.R., Rodriguez-Leon, J.A., Soccol, V.T., 2001. Alkaliphilic bacteria: applications in industrial biotechnology. J. Ind. Microbiol.
Production, purification and properties of microbial phytases. Bioresour. Technol. 77 Biotechnol. 38 (7), 769–790.
(3), 203–214. Sarwar, G., Matayoshi, S., Oda, H., 1987a. Purification of a kappa-carrageenase from

150
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

marine Cytophaga species. Microbiol. Immunol. 31 (9), 869–877. Tramice, A., Pagnotta, E., Romano, I., Gambacorta, A., Trincone, A., 2007.
Sarwar, G., Matoyoshi, S., Oda, H., 1987b. Purification of a α-carrageenan from marine Transglycosylation reactions using glycosyl hydrolases from Thermotoga neapolitana,
Cytophaga species. Microbiol. Immunol. 31, 869–877. a marine hydrogen-producing bacterium. J. Mol. Catal. B Enzym. 47, 21–27.
Sathishkumar, R., Ananthan, G., Iyappan, K., Stalin, C., 2015. A statistical approach for Trincone, A., 2011. Marine biocatalysts: enzymatic features and applications. Mar. Drugs
optimization of alkaline lipase production by ascidian associated—Halobacillus true- 9 (4), 478–499.
peri RSK CAS9. Biotechnol. Rep. 8, 64–71. Trincone, A., 2013. Angling for uniqueness in enzymatic preparation of glycosides.
Sawabe, T., Ohtsuka, M., Ezura, Y., 1997. Novel alginate lyases from marine bacterium Biomolecules 3 (2), 334–350.
Alteromonas sp. strain H-4. Carbohydr. Res. 304, 69–76. Tsuchiya, K., Nakamura, Y., Sakashita, H., Kimura, T., 1992. Purification and char-
Sawant, R., S, N, 2014. Protease: an enzyme with multiple industrial applications. World acterization of a thermostable alkaline protease from alkalophilic
J. Pharm. Pharm. Sci. 3 (6), 568–579. Thermoactinomyces sp. HS682. Biosci. Biotechnol. Biochem. 56 (2), 246–250.
Saxena, R.K., Ghosh, P.K., Gupta, R., Davidson, W.S., Bradoo, S., Gulati, R., 1999. Tsugawa, W., Horiuchi, S., Tanaka, M., Wake, H., Sode, K., 1996. Purification of a marine
Microbial lipases: potential biocatalysts for the future industry. Curr. Sci. 77 (1), bacterial glucose dehydrogenase from Cytophaga marinoflava and its application for
101–115. measurement of 1,5-anhydro-d-glucitol. Appl. Biochem. Biotechnol. 56 (3), 301–310.
Sayali, K., Sadichha, P., Surekha, S., 2013. Microbial esterases. Int. J. Curr. Microbiol. Tsugawa, W., Ogasawara, N., Sode, K., 1998. Fluorescent measurement of 1, 5-anhydro-
Appl. Sci. 2 (7), 135–146. D-glucitol based on a novel marine bacterial glucose dehydrogenase. Enzym. Microb.
Sen, S.K., Jana, A., Bandyopadhyay, P., Das Mohapatra, P.K., Raut, S., 2016. Technol. 22 (4), 269–274.
Thermostable amylase production from hot spring isolate Exiguobacterium sp: a Tsujibo, H., Yoshida, Y., Miyamoto, K., Imada, C., Okami, Y., Inamori, Y., 1992.
promising agent for natural detergents. Sustain. Chem. Pharm. 3, 59–68. Purification, properties, and partial amino acid sequence of chitinase from a marine
Seo, Y.B., Lu, Y., Chi, W.J., Park, H.R., Jeong, K.J., Hong, S.K., Chang, Y.K., 2014. Alteromonas sp. strain O-7. Can. J. Microbiol. 38, 891–897.
Heterologous expression of a newly screened -agarase from Alteromonas sp. GNUM1 Turkiewicz, M., Gromek, E., Kalinowska, H., Zielińska, M., 1999. Biosynthesis and
in Escherichia coli and its application for agarose degradation. Process Biochem. 49, properties of an extracellular metalloprotease from the Antarctic marine bacterium
430–436. Sphingomonas paucimobilis. J. Biotechnol. 70, 53–60.
Seol, Y.J., Lee, J.Y., Park, Y.J., Lee, Y.M., Young, K., Rhyu, I.C., Lee, S.J., Han, S.B., Turkiewicz, M., Kur, J., Białkowska, A., Cieśliński, H., Kalinowska, H., Bielecki, S., 2003.
Chung, C.P., 2004. Chitosan sponges as tissue engineering scaffolds for bone for- Antarctic marine bacterium Pseudoalteromonas sp. 22b as a source of cold-adapted β-
mation. Biotechnol. Lett. 26 (13), 1037–1041. galactosidase. Biomol. Eng. 20, 317–324.
Shahidi, F., JanakKamil, Y.V.A., 2001. Enzymes from fish and aquatic invertebrates and Turner, P., Svensson, D., Adlercreutz, P., Karlsson, E.N., 2007. A novel variant of
their application in the food industry. Trends Food Sci. Technol. 12, 435–464. Thermotoga neapolitana beta-glucosidase B is an efficient catalyst for the synthesis of
Shanmughapriya, S., Kiran, G.S., Selvin, J., Thomas, T.A., Rani, C., 2010. Optimization, alkyl glucosides by transglycosylation. J. Biotechnol. 130 (1), 67–74.
purification, and characterization of extracellular mesophilic alkaline cellulase from Unanue, M., Ayo, B., Agis, M., Slezak, D., Herndl, G.J., Iriberri, J., 1999. Ectoenzymatic
sponge-associated Marinobacter sp. MSI032. Appl. Biochem. Biotechnol. 162 (3), activity and uptake of monomers in marine bacterioplankton described by a biphasic
625–640. kinetic model. Microb. Ecol. 37 (1), 36–48.
Sharifian, S., Homaei, A., Hemmati, R., Khajeh, K., 2017. Light emission miracle in the sea Vaidya, S.Y., Vala, A.K., Dube, H.C., 2001. Bactrial indicators of fecal pollution at
and preeminent applications of bioluminescence in recent new biotechnology. J. Bhavnagar coast. Indian J. Microb. 41, 37–39.
Photochem. Photobiol. B: Biol. 172, 115–128. Van de Lagemaat, J., Pyle, D.L., 2006. Tannase. Enzyme Technology. Springer, New York.
Sharma, S., Agarwal, L., Saxena, R.K., 2008. Purification, immobilization and char- Vats, P., Bhushan, B., Banerjee, U.C., 2009. Studies on the dephosphorylation of phytic
acterization of tannase from Penicillium variable. Bioresour. Technol. 99, 2544–2551. acid in livestock feed using phytase from Aspergillus niger van Teighem. Bioresour.
Shimogaki, H., Takeuchi, K., Nishino, T., Ohdera, M., Kudo, T., Ohba, K., Iwama, M., Irie, Technol. 100 (1), 287–291.
M., 1991. Purification and properties of a novel surface-active agent- and alkaline- Venugopal, V., 2016. Enzymes from seafood processing waste and their applications in
resistant protease from Bacillus sp. Y. Agric. Biol. Chem. 55 (9), 2251–2258. seafood processing. Adv. Food Nutr. Res. 78, 47–69.
Shuai, Y., Zhang, T., Jiang, B., Mu, W., 2010. Development of efficient enzymatic pro- Vieille, C., Zeikus, G.J., 2001. Hyperthermophilic enzymes: sources, uses, and molecular
duction of theanine by gamma-glutamyltranspeptidase from a newly isolated strain of mechanisms for thermostability. Microbiol. Mol. Biol. Rev. 65 (1), 1–43.
Bacillus subtilis, SK11.004. J. Sci. Food Agric. 90 (15), 2563–2567. Vijayaraghavan, P., Kalaiyarasi, M., Vincent, S.G.P., 2015. Cow dung is an ideal fer-
Simpson, H., Denis, M., Malatesta, F., 1997. The terminal oxidase in the marine bacterium mentation medium for amylase production in solid-state fermentation by Bacillus
Pseudomonas nautica 617. Biosci. Rep. 17 (3), 343–346. cereus. J. Genet. Eng. Biotechnol. 13 (2), 111–117.
Song, Q., Wang, Y., Yin, C., H., Z.X., Laa, A., 2016. A novel high-active alkalophilic alpha- Vijayaraghavan, R., Rajendran, S., 2012. Identification of a novel agarolytic gamma
amylase from deep-sea bacterium Luteimonas abyssi XH031. Enzym. Microb. -Proteobacterium microbulbifer maritimus and characterization of its agarase. J.
Technol. 90, 83–92. Basic Microbiol. 52 (6), 705–712.
Song, Q., Zhang, X., 2008. Characterization of a novel non-specific nuclease from ther- Vishwanatha, K.S., Appu-Rao, A.G., Singh, S.A., 2009. Characterization of acid protease
mophilic bacteriophage GBSV1. BMC Biotechnol. 8, 43. expressed from Aspergillus oryzae MTCC 5341. Food Chem. 114, 402–407.
Songsiriritthigul, C., Buranabanyat, B., Haltrich, D., Yamabhai, M., 2010. Efficient re- Vyas, S., Chhabra, M., 2017. Isolation, identification and characterization of
combinant expression and secretion of a thermostable GH26 mannan endo-1,4-beta- Cystobasidium oligophagum JRC1: a cellulase and lipase producing oleaginous yeast.
mannosidase from Bacillus licheniformis in Escherichia coli. Microb. Cell Fact. 9, 20. Bioresour. Technol. 223, 250–258.
Sowell, S.M., Norbeck, A.D., Lipton, M.S., Nicora, C.D., Callister, S.J., Smith, R.D., Wang, J., Xu, A., Wan, Y., Li, Q., 2013. Purification and characterization of a new metallo-
Barofsky, D.F., Giovannoni, S.J., 2008. Proteomic analysis of stationary phase in the neutral protease for beer brewing from Bacillus amyloliquefaciens SYB-001. Appl.
marine bacterium "Candidatus Pelagibacter ubique". Appl. Environ. Microbiol. 74 Biochem. Biotechnol. 170, 2021–2033.
(13), 4091–4100. Wang, X., Zhao, Y., Tan, H., Chi, N., Zhang, Q., Du, Y., Yin, H., 2014. Characterisation of a
Spök, A., 2006. Safety regulations of food enzymes. Food Technol. Biotechnol. 44, chitinase from Pseudoalteromonas sp. DL-6, a marine psychrophilic bacterium. Int. J.
197–209. Biol. Macromol. 70, 455–462.
Stach, J.E., Maldonado, L.A., Ward, A.C., Goodfellow, M., Bull, A.T., 2003. New primers Wang, X.H., Li, D.P., Wang, W.J., Feng, Q.L., Cui, F.Z., Xu, Y.X., Song, X.H., van der Werf,
for the class Actinobacteria: application to marine and terrestrial environments. M., 2003. Crosslinked collagen/chitosan matrix for artificial livers. Biomaterials 24
Environ. Microbiol. 5 (10), 828–841. (19), 3213–3220.
Stickforth, P., Steiger, S., Hess, W.R., Sandmann, G., 2003. A novel type of lycopene ε- Wang, Y., Zhang, C., Li, J., Xu, Y., 2013. Different influences of beta-glucosidases on
cyclase in the marine cyanobacterium Prochlorococcus marinus MED4. Arch. volatile compounds and anthocyanins of Cabernet Gernischt and possible reason.
Microbiol. 179 (6), 409–415. Food Chem. 140 (1–2), 245–254.
Strom, T., Raa, J., 1993. Marine biotechnology in Norway. J. Mar. Biotechnol. 1, 3–7. Wei, X.H., Niu, Y.P., Xu, Y.Y., Du, Y.Z., Hu, F.Q., Yuan, H., 2010. Salicylic acid-grafted
Subramani, R., Aalbersberg, W., 2012. Marine actinomycetes: an ongoing source of novel chitosan oligosaccharide nanoparticle for paclitaxel delivery. J. Bioact. Compat.
bioactive metabolites. Microbiol. Res. 167 (10), 571–580. Polym. 25 (3), 319–335.
Sugano, Y., Matsumoto, T., Kodama, H., Noma, M., 1993a. Cloning and sequencing of Wells, W., 1999. Extreme chemistry. AB/BA/1297xtremo.html accessed.
agaA, a unique agarase 0107 gene from a marine bacterium, Vibrio sp. strain JT0107. Weltrowski, M., Martel, B., Morcellent, M., 1996. Chitosan N-benzyl sulfonate derivatives
Appl. Environ. Microbiol. 59 (11), 3750–3756. as sorbents for removal of metal ions in an acidic medium. J. Appl. Polym. Sci. 59 (4),
Sugano, Y., Terada, I., Arita, M., Noma, M., Matsumoto, T., 1993b. Purification and 647–654.
characterization of a new agarase from a marine bacterium, Vibrio sp. strain JT0107. Willi, P., Sharma, P., 2004. Chitosan and alginate wound dressings. Trends Biomater.
Appl. Environ. Microbiol. 59 (5), 1549–1554. Artif. Organs 18 (1), 18–23.
Sukharnikov, L.O., Cantwell, B.J., Podar, M., Zhulin, I.B., 2011. Cellulases: ambiguous Wong, T.Y., Preston, L.A., Schiller, N.L., 2000. Alginate lyase: review of major sources and
nonhomologous enzymes in a genomic perspective. Trends Biotechnol. 29, 473–479. enzyme characteristics, structure-function analysis, biological roles, and applications.
Suolow, T.V., Jones, J., 1988. Chitinase-producing bacteria. In US Patent. Ann. Rev. Microbiol. 54, 289–340.
Suzuki, H., Sawai, Y., Suzuki, T., Kawai, K., 2003. Purification and characterization of an Wu, C., Xu, C., Ni, H., Yang, Q., Cai, H., Xiao, A., 2016. Preparation and characterization
extracellular beta-agarase from Bacillus sp. MK03. J. Biosci. Bioeng. 95 (4), 328–334. of tannase immobilized onto carboxylfunctionalized superparamagnetic ferroferric
Takii, Y., Kuriyama, N., Suzuki, Y., 1990. Alkaline serine protease produced from citric oxide nanoparticles. Bioresour. Technol. 205, 67–74.
acid by Bacillus alcalophilus subsp. halodurans KP 1239. Appl. Microbiol. Biotechnol. Wu, G., Qin, Y., Cheng, Q., Liu, Z., 2014a. Characterization of a novel alkalistable and
34 (1), 57–62. salt-tolerant α-amylasefrom marine bacterium Zunongwangia profunda. J. Mol. Catal.
Tong, C.C., Cole, A.L., Shepherd, M.G., 1980. Purification and properties of the cellulases B: Enzym. 110, 8–15.
from the thermophilic fungus Thermoascus aurantiacus. Biochem. J. 191 (1), 83–94. Wu, G., Yuan, M., Wei, L., Zhang, Y., Lin, Y., Zhang, L., al., e, 2014b. Characterization of a
Trachoo, N., Mistry, V.V., 1998. Application of ultrafiltered sweet buttermilk and sweet novel cold-adapted phosphinothricin N-acetyltransferase from the marine bacterium
buttermilk powder in the manufacture of nonfat and low fat yogurts. J. Dairy Sci. 81, Rhodococcus sp. strain YM12. J. Mol. Catal. B: Enzym. 104, 23–28.
3163–3171. Wu, Q., Ma, S., Xiao, H., Zhang, M., Cai, J., 2011. Purification and the secondary structure

151
A. Beygmoradi, A. Homaei Biocatalysis and Agricultural Biotechnology 11 (2017) 131–152

of Fucoidanase from Fusarium sp. LD8. Evid.-Based Complement. Altern. Med. Chem. 279 (30), 31863–31872.
2011, 8. Yang, J.S., Xie, Y.J., He, W., 2011. Research progress on chemical modification of algi-
Wu, S.J., Chen, J., 2014. Preparation of maltotriose from fermentation broth by hydro- nate: a review. Carbohydr. Polym. 84, 33–39.
lysis of pullulan using pullulanase. Carbohydr. Polym. 107, 94–97. Yang, Q., Yang, M., Pritsch, K., Yediler, A., Hagn, A., Schloter, M., Kettrup, A., 2003.
Xia, W., Liu, P., Liu, J., 2008. Advance in chitosan hydrolysis by non-specific cellulases. Decolorization of synthetic dyes and production of manganese-dependent peroxidase
Bioresour. Technol. 15, 6751–6762. by new fungal isolates. Biotechnol. Lett. 25 (9), 709–713.
Xie, W., Lin, B., Zhou, Z., Lu, G., Lun, J., Xia, C., Li, S., Hu, Z., 2013. Characterization of a Yang, S., Fu, X., Yan, Q., Jiang, Z., Wang, J., 2016. Biochemical characterization of a
novel beta-agarase from an agar-degrading bacterium Catenovulum sp. X3. Appl. novel acidic exochitinase from Rhizomucor miehei with antifungal activity. J. Agric.
Microbiol Biotechnol. 97, 4907–4915. Food Chem. 64, 461–469.
Xiong, A.S., Yao, Q.H., Peng, R.H., Li, X., Fan, H.Q., Guo, M.J., Zhang, S.L., 2004. Yaphe, W., Morgan, K., 1959. Enzymic hydrolysis of fucoidin by Pseudomonas atlantica
Isolation, characterization, and molecular cloning of the cDNA encoding a novel and Pseudomonas carrageenovora. Nature 183 (4663), 761–762.
phytase from Aspergillus niger 113 and high expression in Pichia pastoris. J. Yin, L.J., Lin, H.H., Chiang, Y.I., Jiang, S.T., 2010. Bioproperties and purification of xy-
Biochem. Mol. Biol. 37 (3), 282–291. lanase from Bacillus sp. YJ6. J. Agric. Food Chem. 58 (1), 557–562.
Xu, J.L., Lu, M.S., Wang, S.J., Li, H.Z., Sun, Y.Y., Fang, Y.W., 2009. Production and Zeinali, F., Homaei, A., Kamrani, E., 2015. Sources of marine superoxide dismutases:
characterization of pullulanase from hyperthermophilic archaeon Thermococcus sp. characteristics and applications. Int. J. Biol. Macromol. 79, 627–637.
HJ21. J. Food Sci. Biotechnol. 2, 243–249. Zhang, C., Kim, S., 2008. Research and application of marine microbial enzymes: status
Yadav, R.P., Saxena, R.K., Gupta, R., Davidson, W.S., 1998. Purification and character- and prospects. Mar. Drugs 8 (6), 1920–1934.
ization of a regiospecific lipase from Aspergillus terreus. Biotechnol. Appl. Biochem. Zhang, C., Kim, S.K., 2010. Research and application of marine microbial enzymes: status
28 (Pt 3), 243–249. and prospects. Mar. Drugs 8 (6), 1920–1934.
Yahata, N., Watanabe, T., Nakamura, Y., Yamamoto, Y., Kamimiya, S., Tanaka, H., 1990. Zhang, G.Q., Dong, X.F., Wang, Z.H., Zhang, Q., Wang, H.X., Tong, J.M., 2010.
Structure of the gene encoding beta-1,3-glucanase A1 of Bacillus circulans WL-12. Purification, characterization, and cloning of a novel phytase with low pH optimum
Gene 86 (1), 113–117. and strong proteolysis resistance from Aspergillus ficuum NTG-23. Bioresour. Technol.
Yamane, S., Iwasaki, N., Majima, T., Funakoshi, T., Masuko, T., Harada, K., Minami, A., 101 (11), 4125–4131.
Monde, K., Nishimura, S., 2005. Feasibility of chitosan-based hyaluronic acid hybrid Zhang, Z., Marquardt, R.R., Wang, G., Guenter, W., Crow, G.H., Han, Z., Bedford, M.R.,
biomaterial for a novel scaffold in cartilage tissue engineering. Biomaterials 26 (6), 1996. A simple model for predicting the response of chicks to dietary enzyme sup-
611–619. plementation. J. Anim. Sci. 74 (2), 394–402.
Yamasaki, M., Moriwaki, S., Miyake, O., Hashimoto, W., Murata, K., Mikami, B., 2004. Zhao, K., Guo, L.Z., Lu, W.D., 2012. Extracellular production of novel halotolerant,
Structure and function of a hypothetical Pseudomonas aeruginosa protein PA1167 thermostable, and alkali-stable carboxymethyl cellulase by marine bacterium
classified into family PL-7: a novel alginate lyase with a beta-sandwich fold. J. Biol. Marinimicrobium sp. LS-A18. Appl. Biochem. Biotechnol. 168 (3), 550–567.

152

Das könnte Ihnen auch gefallen