Open access peer-reviewed chapter

Actinobacteria Associated with Marine Invertebrates: Diversity and Biological Significance

Written By

Vaishali R. Majithiya and Sangeeta D. Gohel

Submitted: 26 September 2021 Reviewed: 19 July 2022 Published: 18 October 2022

DOI: 10.5772/intechopen.106642

From the Edited Volume

Actinobacteria - Diversity, Applications and Medical Aspects

Edited by Wael N. Hozzein

Chapter metrics overview

144 Chapter Downloads

View Full Metrics

Abstract

The ocean harbors a wide diversity of beneficial fauna offering an enormous resource for novel compounds, and it is classified as the largest remaining reservoir of natural molecules to be evaluated for biological activity. The metabolites obtained from marine invertebrate-associated actinobacteria have different characteristics compared to terrestrial actinobacteria as marine environments are exigent and competitive. Actinobacteria produce a wide range of secondary metabolites, such as enzymes, antibiotics, antioxidative, and cytotoxic compounds. These allelochemicals not only protect the host from other surrounding pelagic microorganisms but also ensure their association with the host. The harnessing of such metabolites from marine actinobacteria assures biotechnological, agricultural, and pharmaceutical applications.

Keywords

  • actinobacteria
  • marine invertebrates
  • diversity
  • biological activity

1. Introduction

Actinobacteria are gram-positive bacteria with high G + C DNA content [1] that can live in different habitats including the marine environment. These bacteria enclose significant biotechnological potential as they produce complex biopolymers, such as polysaccharides, extracellular and intracellular enzymes, antibiotics, inhibitors, and various metabolic products [2]. Since the discovery of streptomycin from Streptomyces griseus, actinobacteria are recognized for antibiotic production. Approximately two-thirds of world bioactive compounds are isolated from these phyla. Therefore, actinobacteria are considered as a potential source for the development of new drugs [3]. It has been emphasized that actinobacteria from marine habitats may be valuable for the isolation of novel strains that can potentially produce secondary metabolites like enzymes, cosmetics, antibiotics, anti-parasitic, vitamins, nutritional material, and immunosuppressive agents having great economical and biotechnological importance [4]. Nowadays, research has been diverted toward marine niches to screen that actinobacteria can produce bioactive compounds with different metabolic characteristics as the frequency of novel bioactive compounds from terrestrial actinobacteria decreases with time [5]. Marine flora (mangroves, seaweed, seagrasses, and algae) and fauna (Porifera, coelenterates, ascidians, crustaceans, and mollusks) are part of highly productive ecosystems and are habitats of numerous bioactive compounds producing microorganisms. Bioactive compounds obtained from associated microorganisms are known for a broad range of biological effects, such as antimicrobial, antifungal, insecticides, antiprotozoal, antiparasitic, anti-inflammatory, and antitumor [6, 7, 8]. Sponges (phylum: Porifera) and corals (phylum: Cnidaria) are the most predominant source of marine natural products. Ascidians (phylum: Chordata) have been classified as the second predominant source of natural products [9, 10]. Seaweeds-associated bacterium produces a diverse range of biologically active compounds that specifically target fouling organisms and also show antifungal and antibacterial effects [11]. The rare actinobacterial species are explored from marine environments based on conventional and molecular approaches. The changes in salinity along with the nutrient value within the ocean make the marine environment a good source of novel actinobacteria. Due to the nutrient diversity present in the oceans, one can expect that the organisms that inhabit the marine environment would be very diverse. The different zones of the ocean contain different nutrients and minerals. Therefore, the microorganisms that inhabit various zones generally have metabolites as well as functions that are conducive to their specific zone. It indicates a high probability of microbial diversity that leads to the production of novel antibiotics and enzymes in the marine environment [12, 13]. The present investigation has emphasized invertebrate-associated actinobacteria as a source for novel natural products. The rationale behind this is because invertebrate-associated microorganisms are capable of producing various secondary metabolites and enzymes that can be used in predator defense, antifouling, inhibition of overgrowth, protection from ultraviolet radiation as well as acting as mediators in the competition for settling space [14].

Advertisement

2. Diversity of actinobacteria associated with marine invertebrates

Invertebrate-associated bacterial communities have a significant ability to produce bio-medically relevant micro molecules. Cultivable approaches and metagenomic analysis show that many invertebrates harbor actinobacterial species. These actinobacterial species mainly belong to genera Streptomyces, Nocardiopsis, Kocuria, Salinospora, Nocardia, Rhodococcus, Nonomuraea, Actinokinespora, and Saccharopolyspora [15, 16, 17].

2.1 Sponges

The phylum Porifera harbor dense and diverse bacterial communities. According to the literature review, about 40% of the sponge biomass was due to their associated bacteria [18]. The sponges are recognized for their potential source of bioactive metabolites. These bioactive metabolites are generally produced by their associated microbial communities which suggest that associated microorganisms might play a role in the chemical defense of their host [19]. Till today, about 60 actinobacterial genera have been isolated from marine sponges [20]. Sponge-associated actinobacteria dwell in the mesohyl matrix of sponges. They may be true halobiont or taken up from nearby water through the filtration process. About 20 actinobacterial genera belonging to genera Kocuria, Micromonospora, Nocardia, Nocardiopsis, Saccharopolyspora, Salinispora, and Streptomyces were isolated from South China sea sponges (Genera: Haliclona, Amphimedon, Phyllospongia, Agelas, Hippospongialachne, Cinachyrsina, Arenosclera, Phakellia, Cliona) [18, 20].

2.2 Coral reefs

The bacterial communities associated with coral have unique properties. Bacteria inhabit coral in three different parts of the coral body that includes the surface of the mucus layer, the interior of coral tissue, and the calcium carbonate skeleton. Each of them harbors unique beneficial properties. The skeletons of corals are porous which provides micro niches for a variety of bacterial communities for colonization [21]. Cyanobacteria in the skeleton of Oculina Patagonica provide organic compounds to the coral tissue which helps them to survive during losses of endosymbiotic algae [21, 22]. In addition, a recent study shows that bacteria isolated from the mucus of healthy Acropora palmata produce antibiotics inhibiting the growth of potentially pathogenic microorganisms. This shows that the diversity of bacterial species that are associated with a particular coral species is high, including many novel species. A number of mutualistic benefits have been suggested [23, 24]. The other microbial lives, such as bacteria are associated with coral halobiont for their nutritional requirement. In return, the associated bacteria protect the host by the production of secondary metabolites, such as antifungal, antibacterial, and antihelminth. The antimicrobial peptide damicornin produced by coral-associated bacteria was active against fungi and selective gram-positive bacteria but not against Vibrios sp. while the organic extract of Siderastrea sidereal coral showed antibacterial activity against two of the four strains of gram-positive bacterial isolates from coral surfaces [25, 26].

The actinobacterial communities associated with corals can fix nitrogen which explains their dominance in healthy corals [21, 27]. Lampert et al. reported mucus-associated bacterial diversity among which 23% were Actinobacteria [28]. Mahmoud and Kalendar reported Brachybacterium, Brevibacterium, Cellulomonas, Dermacoccus, Devriesea, Kineococcus, Kocuria, Marmoricola, Micrococcus, Micromonospora, Ornithinimicrobium, Renibacterium, and Rhodococcus actinobacterial genera that belong to three corals (Coscinaraea columna, Platygyra daedalea, and Porites harrisoni) among which Kocuria and Brevibacterium were dominant genera [22]. Kuang et al. reported Fridmanniella and Propionibacterium as major groups associated with P. lutea while genera Demetria, Fodinicola, Friedmanniella, Geodermatohilus, Iamia, Modestobacter, Ornithinimicrobium, Tersicoccus, and Yonghaparikia were detected for the first time from P. lutea through culture-independent study [23]. The novel actinobacterial species Nocardiopsis coralli HNM0947T isolated from Hainan province, PR China inhabitants of coral Galaxeaastreata show optimum growth at pH 7, temperature 28°C, and 3% NaCl(w/v). The strain HNM0947T contained 71.3% mol G + C, MK-10(H8), MK-10(H6), and MK-10(H4) as major menaquinones, iso-C16:0, anteiso-C17:0, C18:0, C18:0 10-methyl (TBSA), and anteiso-C15:0 as major fatty acids [29]. Among the cultivable actinobacterial genera Jiangella, Micromonospora, Nocardia, Nocardiopsis, Rhodococcus, Verrucosispora, Salinispora, and Streptomyces showed potential actinobacterial activity against various test pathogens consequently contributing to coral health [23].

2.3 Shrimps

Shrimps are considered one of the most famous seafood consumed worldwide. They belong to the phylum Arthropoda, subphylum Crustacea. The exoskeleton of Shrimps contains chitin, structural proteins, and mineral deposits, and its construction is an energy-demanding process. Till today, very few studies regarding actinobacterial diversity associated with Shrimps are published. The Streptomyces californicus isolated from Shrimp farming displayed the ability to inhibit the growth of Vibrio sp., one of the disease-causing pathogens [30]. You et al. demonstrated the significance of actinobacteria in shrimp farming due to their antimicrobial, antifungal, and antioxidative ability [31]. Kumar et al. prepared extract from actinobacterial sp. and observed its in vivo effect by supplementing extract with feed to black tiger shrimp having white sport syndrome [32]. You et al. reported actinobacteria with the ability to inhibit the formation of biofilm produced by Vibrio sp. All these studies indicate the significance of actinobacteria in aquaculture [33].

2.4 Ascidians

Ascidians belong to phylum Chordata, subphylum Tunicate. More than 1000 bioactive metabolites have been isolated from ascidians and their associated microbial communities. The indolocarbazoles having anticancer ability were produced by E. toealensis associated with actinobacterial genera Salinispora and Verrucosispora [34]. Lee et al. reported actinobacterial species, including Arthrobacter rhomb, Brachybacterium muris, Micrococcus lylae, and Nocardiopsissynnemataformans from squid collected from Jumunjin, Gangwon-do, Korea [35].

Advertisement

3. Biological significance of actinobacteria associated with marine invertebrates

The actinobacteria are omnipresent in all environmental conditions. They produce a wide range of metabolites having biotechnological applications as described in Table 1. Actinobacteria is a group of organisms having the ability to produce inhibitors, immunomodifiers, biosurfactants, antioxidative, anti-inflammatory, antimicrobial, antifungal, and anticancer compounds along with intracellular and extracellular enzymes with unique characteristics in terms of substrate selectivity, stability in presence of salts, temperature tolerance, pH variation, etc. The significance of host-associated marine actinobacteria is also described in Table 1.

ActinobacteriaHostLocationSignificanceReferences
Sponge
Nocardiopsis dassonvillei MAD08D. nigraSouthwest coast, IndiaAntibacterial activity and anticandidal activity[36]
Nocardiopsis sp., Micromonospora sp., Rhodococcus sp. and Streptomyces sp.Iotrochota sp., and HymeniacidonperleveEastern Mediterranean coast, Turkey15 strains exhibited antimicrobial activity against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecium and three strains showed anticandidal activity against Candida albicans[37]
Actinoalloteichus sp. Micrococcus sp., Micromonospora sp., Nocardiopsis sp., Streptomyces sp.HalichondriapaniceaBaltic Sea, GermanyPresence of polyketide synthases (PKS) and nonribosomal peptide synthases (NRPS)[38]
Streptomyces poriferorum sp. novGeodiabarretti and AnthodichotomaTautra island, Trondheim fjord of NorwayAnti MRSA activity[39]
Salinispora arenicola and Salinispora pacificaSpongesGreat Barrier ReefN-acyl homoserine lactones[40]
]Actinobacterial strainsCallyspongia sp., Callyspongiaaerizusa, Carteriospongia contorta, Chelanoplysilla sp., and DiacarnusbismarckensisPanggang Island, Taman Nasional KepulauanSeribu, IndonesiaPlant growth promoting bioactivity: Indolacetic acid production, HCN production, phosphate solubilization, and antimicrobial activity against Xanthomonas oryzae and Pyricularia oryzae[41]
Williamsiaaurantiacus sp. novGlodiacorticostyliferaPraia Guaeca, Sao Paulo, BrazilAntimicrobial activity against S. aureus and Colletotrichum gloeosporioides[42]
Streptomyces olivaceusDysideaavaraLarak Island, Persian GulfOlivomycin A: cytotoxic activity against SW480, HepG2, and MCF7 cell line[43]
Corals
S. arenicolaP. lobata and P. panamensisTropical central PacificPromotes plant growth in salt stress conditions[44]
Micromonospora marinaSoft coralIndian Ocean coast, MozambiqueThiocoraline: anticancer activity against LoVo and SW620 human colon cancer cell lines[45]
Mycobacterium spCoralKurusadai Island, Gulf of MannarAntioxidant Activity: Scavenging of Hydrogen peroxide, Nitric Oxide Radical[46]
Actinobacterial sp.Sarcophyton glaucumRed seaAntibacterial activity against S. aureus, K. pneumonia, P. aeruginosa, V. fluvialis, antifungal activity against A. niger, Penicillium sp., and C. albicans[47]
Myceligeneranscantabricum sp. novFam. CaryophillidaeAviles Canyon, Cantabrian Sea, Asturias, SpainAntimicrobial activity against E. coli, Micrococcus luteus, and Saccharomyces cerevisiae[48]
Micromonosporaaurantiaca, Nocardiopsisdassonvillei subsp. Albirubida, Nocardiopsissynnemataformans, Streptomyces rutgersensis, Streptomyces viridodiastaticusScleractinian coralsLuhuitou fringing reefPresence of NRPS, PKS type I and II biosynthetic gene cluster[49]
Ascidians
Salinisporasp. and Verrucosispora sp.E. toealensisMicronesian Islands, ChuukAntibiotic: Indolocarbazoles and Staurosporines[34]
Mollusks
Streptomyces sp. CP32Conus pulicariusMactan Island, PhilippinesDerivatives of Pulicatins with neurological activity[50]
Nocardiopsis albaCR167C. rolani,Mactan Island, PhilippinesNocapyrones with neurological activity[51]
Streptomyces sp.Crassostrea sikameaBahia de La Paz, Baja California Sur, Mexico[52]

Table 1.

Significance of actinobacteria associated with marine invertebrates.

3.1 Antimicrobial ability

The Micrococcus luteus isolated from sponge shows strong inhibition against Staphylococcus aureus,Vibrio anguillarum, and Candida albicans [53]. Mayamycin and Microluside isolated from sponge-associated Streptomyces sp. HB202 and Micrococcus sp. EG45 inhibited S. aureus with IC50 (1.16 μg/mL) and (12.42 μg/mL) respectively [54, 55]. Salinispora sp. isolated from Pseudoceratina clavate, as well as Micromonospora sp. CPI 12 and Saccharomonospora sp. CPI 13 isolated from Callyspongia diffusa showed antibacterial activity against S. epidermidis [56]. Actinobacterial genera Pseudonocardia, Streptomyces, Kocuria, Aeromicrobium, Brachybacterium, and Nocardiopsis were isolated from sponges, such as Haliclona sp., Callyspongia sp., and Desmacella sp. Among 92 isolated actinobacterial strains, 52 actinobacterial strains exhibited antibacterial activity against E. coli,P. fluorescens,V. alginolyticus, and V. splendidus. Further analysis revealed that 18% of actinobacterial strains contained NRPS gene clusters while 10% harbor PKS-KS gene and 6% have PKS-NRPS gene clusters [57]. Different actinobacterial genera of marine origin consist of nontoxic antibiotics able to inhibit the growth of Vibrio sp. resulting in a potential source for aqua culturing [58, 59].

3.2 Antioxidative ability

Ferric reducing antioxidant power (FRAP), nitric oxide (NO) scavenging, and DPPH radical scavenging activity were extensively used to measure antioxidant capacity. Nocardiopsis sp.PU3 isolated from the coral reef of the Pullivasal Island, Gulf of Mannar, and India was detected as a potential source of antioxidative agent that can be used to treat various oxidative stress-related disorders with 53.6% DPPH, 74.2% hydrogen peroxide, and 56% nitric oxide radical scavenging activity at 100 μg/mL concentration [60]. Recently, 54.50% ABTS free radical scavenging activity with 100 μg/mL was reported from coral reef-associated Saccharopolyspora sp. IMA1 [16]. While Streptomyces sp. NMF6 associated with marine sponge Diacarnus ardoukobae possessed significant phosphomolybdenum, ferric-reducing power, and DPPH free radical scavenging activity [61]. Ser et al. reported Streptomyces malaysiense sp. having 27.24% DPPH radical scavenging activity at 2 mg/mL of 0.016% DPPH solution [62].

3.3 Cytotoxic activity

Cancer treating drugs have elevated toxic effects with undesirable side effects. Therefore, the reach of new and less harmful drugs has a high demand. Progress had been made recently to reach antitumor compounds from marine actinobacteria [63]. Violapyrone H and I were isolated from Streptomyces sp. associated with starfish. Violapyrone exhibited cytotoxicity against 10 human cancer cell lines with a GI50 value from 1.10 M to 26.12 M. This is the first report of violapyrones tested for their cytotoxicity potential [64]. Streptomyces sp. isolated from Acanthaster planci collected from the Federated States of Micronesia produced violapyrone H, Iα pyrones derivatives that displayed cytotoxicity against 10 human cancer cell lines with GI50 value from 1.10 μM to 26.12 μM [63]. Donaxtrunculus anatinus-associated Actinobacterial genera Nocardioides, Kitasatosporia, and Streptomyces showed strong antitumor activity against human carcinoma of the liver (HEPG2), cervix (HELA), and breast (MCF7) cell line with IC50: 3.89, 9.4, and 10 μg/ml, respectively [65]. Steffimycin produced by Streptomyces sp. 0630 exhibited cytotoxic effect against a panel of human cancer cells MCF-7, HepG-2, and A2780 with IC50 values of 5.05, 5.57, and 1.91 μM, respectively [66]. Streptomyces sp. TMKS8 associated with sea slug exhibited cytotoxicity against murine leukemia P388 cells with IC50 9.8 μM [67, 68].

3.4 Enzymes

The significance of enzymes in food, textile, detergent, and pharmaceutical has been known for a long. The harnessing of actinobacterial-derived enzymes comprises of cost-effective and eco-friendly nature due to its mild fermentation condition, such as the use of agricultural waste as a source of nutrients, temperature, pH, agitation, and less production time [69]. Actinobacteria associated with marine hosts produce various enzymes including amylase, protease, keratinase, lipase, L asparaginase, Xylanase, chitinase, cellulase, and dextranase that embrace industrial significance [70].

Proteases hydrolyze protein molecules to peptides and eventually to free amino acids. Protease plays a significant role in the metabolic cycles of all living forms. There are several types of proteases including serine, carboxy serine, cystine, metallo, carboxy metallo, and aspartic proteases [71]. The application of protease includes animal fodder preparation, silk degumming, detergent formulation (stain remover), dehairing and dewooling (leather industry), and silver recovery from X-ray film [72, 73, 74]. Actinobacterial genera Microbacterium isolated from stony coral Pocillopora sp. and Faviia sp. produced proteases [75]. Similarly, Micrococcus sp. and Brevibacterium sp. isolated from Faviia sp. produced proteases [75, 76]. Whereas 12.6% and 10.9% of bacteria associated with marine sponge D. granulosa and S. fibulata, respectively produced proteases [77].

Amylase is one of the most demanding enzymes used mainly for scarification of starch, pulp processing, bread dough making, winery, and detergent industry whereas solvent tolerant amylase was used mainly for bioremediation and improvement of detergent [78, 79]. Meena et al. isolated 10 actinobacterial genera associated with Phallusia nigra ascidian. Among them, Kinecoccus mangrovi, Kocuria polaris, Salinospora sp., and Nocardiopsis exalbidus were amylase positive with 12.29, 8.85, 6.61, and 5.13 U/mL activity, respectively [80]. According to the earlier report, bacteria associated with sponges exhibited a higher percentage of amylase production followed by phosphatase and protease while the least urease-positive isolates were obtained from Dysidea granulose and Sigmadocia fibulata sponge [81].

Chitinase involves in hydrolyses of chitin polymer by cleaving β 1–4 linkages. Chitin is a polymer present in the cell wall of fungi, shells of marine invertebrates, and few insects. The use of chitinase involves the development of pesticides, management of marine wastes, biofuel production, and food and pharma industries [82]. Many actinobacterial genera, including Streptomyces, Micromonospora, Nocardiopsis, and Nocardia have been reported [83]. Recombinant chitinase from S. griseus showed enhanced hydrolysis of α and β chitin with a higher rate of activity using shrimp shells as substrate [84]. SaChiB chitinase isolated and cloned from S. alfalfa ACCC400021 showed maximal activity at 45°C temperature with pH 8 while SaChiB exhibited antifungal activity so considered as a biocontrol agent in agriculture [85].

The pharmaceutical demand for L-asparaginase is high due to its anti-carcinogenic ability. L-asparaginase inhibits the growth of cancerous cells by cleaving L-asparagine into ammonia and aspartic acid. Mainly L-asparaginase is produced by fungi while few actinobacterial species are also reported [86]. Streptomyces noursei MTCC 10469 associated with marine sponge Callyspongia diffusa produced 102 kDa of L-asparaginase that displayed optimum activity in pH 8 at 50°C [87]. Also, S. fradiae NEAE-82-derived L-asparaginase showed anti-proliferative activity on cancer cells (HepG2, Hep2, and Caco2) [88]. Currently, Aspergillus and Bacillus-derived L-asparaginase are available commercially while actinobacterial-derived L-asparaginase is under investigation [86].

Actinobacteria are a distinctive group of prokaryotes having similarities to both fungi and bacteria. The actinobacterial-derived cellulase has unique features in terms of adaptation to extreme environmental parameters and degradation of plant-based biomass. Cellulase is a carbohydrate degrading enzyme that hydrolyzes cellulose into mono- and oligosaccharides [89]. The Kineococcus mangrove, Kocuria Polaris, Nocarciopsis exalbidus, and Salinispora sp. belonging to the phylum Actinobacteria were isolated from marine ascidian Phallusia nigra exhibited cellulase production [80]. Most terrestrial microbial-derived cellulase showed an inhibitory effect in presence of glucose whereas marine halophilic Streptomyces-derived cellulase retained more than 60% activity in presence of 0.5 M glucose which makes the enzyme feasible to conduct high biomass saccharification [90]. Also, Actinoalloteichus cyanogriseus strain MHA15 isolated from marine habitat showed higher cellulase activity 14.378 U/mL in carboxy methyl cellulose medium suggesting ideal bacteria for cellulose bioconversion [89].

3.5 Helometabolites

Halometabolites produced by marine organisms play a significant role in host defense mechanisms by quorum sensing and production of toxins, growth hormones, or antibiotics. Helometabolites, such as chloride, bromide, and iodine are omnipresent in marine environments whereas fluoride is present in Earth’s crust [91]. Micromonospora echinospora-derived Calicheamicin is a group of enediyne metabolites with iodine and has remarkable anticancer activity [92]. Whereas, halogenated glycopeptide Vancomycin produced by S. lavendulae showed antibacterial activity against various test pathogens [93] followed by the production of indimicine A-E chlorinated bisindolie alkaloid from deep-sea Streptomyces sp. showed strong antimicrobial activity along with cytotoxic activity against MCF-7 cell line with IC50 10 μM [94]. FU et al. reported chlorinated streptochlorides derived from coral-associated Streptomyces sp. with antimicrobial activity against proteobacteria and cytotoxicity against breast cancer cell line MCF-7 [95]. The research of halo metabolites through genome mining resulted in 26 FADH2dependanthalogenase positive actinobacterial strains associated with mangroves [96]. The antifungal kutzneride involves in dichlorination at C6 and C7 position of tryptophan has been isolated from Kutzneria sp. whereas pyrroindomycin isolated from S. rugosporus involves tryptophan dichlorination at position 5 [97]. Further, three novel halogenase gene clusters were identified from sponge-associated actinobacteria that suggest the importance of actinobacteria as a remarkable source for harnessing halometabolites [98].

3.6 Enzymes inhibitors

Enzyme inhibitors have a pivotal position in agriculture to protect crops from predators. Allosamidin derived from Streptomyces sp. can inhibit chitinase and shows potent insecticidal activity against Bombyx mori and Silkworm [99]. Several α-glucosidase and α-amylase inhibitor has been reported from marine habitat, for instance, Streptomyces sp.PW638-derived Acarviostatin 103 inhibit α-amylase with IC50 value 12.23 μg/mL and α-glucosidase with IC50 1.25 μg/mL [100]. Whereas amino-oligosaccharide α-glucosidase inhibitors produced by Streptomyces sp. CKD-711 showed potent inhibitory activity against Comamonas terrigena [101]. Protease inhibitor has valuable antiviral activity against Zika, Dengue, hepatitis C virus, and many more. Kamarudheen et al. reported protease inhibitor from marine sponge Callyspongiasp-associated S. griseoincarnatus HK12 having antiviral activity against Chikungunya [102]. Leupeptin, Pepstatin, Antipain, Phosphoramidon, Talopeptin, and Diketopiperazine are well-known actinobacteria-derived protease inhibitors having remarkable pharmaceutical significance [103, 104].

Advertisement

4. Conclusion

The marine actinobacterial provides vast scope for therapeutically active macromolecules, such as antibiotics and halometabolites with the addition of industrially significant enzymes, such as amylase, protease, asparaginase, xylanase, cellulase, chitinase, and lipase. The presence of actinobacteria in marine habitats plays a pivotal role in their associated organisms by providing protection against harmful moieties. Harnessing host-specific actinobacterial diversity from the marine ecosystem will result in novel species with the ability to produce new and diverse secondary metabolites which will be beneficial for detergent, food, medicine, agriculture, cosmetics, paper, and pulp industries. Furthermore, the exploitation of bioactive compounds from marine microorganisms will fulfill the current demand for drug-resistant microorganisms as many more marine niches are still unexplored.

Advertisement

Acknowledgments

Ms. Vaishali R. Majithiya acknowledges SHODH (ScHeme Of Developing High quality research- MYSY), Gujarat, India for a research fellowship. The authors are grateful to DST-SERB, New Delhi, India for financial support (Sanction order No. ECR/2016/000928 Dated: 20.03.2017). The authors are also thankful to Saurashtra University, Gujarat, India for infrastructural facilities.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Acronyms and abbreviations

FRAP

Ferric reducing antioxidant power

NO

Nitric oxide

VCPO

Vanadium dependant chloroperoxidase

MRSA

Methicillin-resistant Staphylococcus aureus

PKS

Polyketide synthases

NRPS

Nonribosomal peptide synthases

IC50

Half maximal inhibitory concentration

References

  1. 1. Goodfellow M. Phylum XXVI. Actinobacteria phyl. nov. In: Goodfellow M, Kampfer P, Trujillo ME, Suzuki K, Ludwig W, Whitman WB, editors. Bergey’s Manual of Systematic Bacteriology. second ed. Vol. 5. New York: Springer; 2012. pp. 33-34
  2. 2. Hauhan JV, Gohel SD. Molecular diversity and pharmaceutical applications of free-living and Rhizospheric marine Actinobacteria. In: Marine Niche: Applications in Pharmaceutical Sciences. Singapore: Springer; 2020. pp. 111-131
  3. 3. Sivakumar K, Sahu MK, Thangaradjou T, Kannan L. Research on marine actinobacteria in India. Indian Journal of Microbiology. 2007;47(3):186-196. DOI: 10.1007/s12088-007-0039-1
  4. 4. Gohel SD, Singh SP. Molecular phylogeny and diversity of the salt-tolerant alkaliphilic actinobacteria inhabiting coastal Gujarat. India. Geomicrobiology Journal. 2018;35(9):775-789. DOI: 10.1080/01490451.2018.1471107
  5. 5. Quinn GA, Banat AM, Abdelhameed AM, Banat IM. Streptomyces from traditional medicine: Sources of new innovations in antibiotic discovery. Journal of Medical Microbiology. 2020;69(8):1040. DOI: 10.1099/jmm.0.001232
  6. 6. Egan S, James S, Holmström C, Kjelleberg S. Inhibition of algal spore germination by the marine bacterium Pseudoalteromonastunicata. FEMS Microbiology Ecology. 2001;35(1):67-73. DOI: 10.1111/j.1574-6941.2001.tb00789.x
  7. 7. Egan S, Thomas T, Kjelleberg S. Unlocking the diversity and biotechnological potential of marine surface associated microbial communities. Current Opinion in Microbiology. 2008;11(3):219-225. DOI: 10.1016/j.mib.2008.04.001
  8. 8. Penesyan A, Tebben J, Lee M, Thomas T, Kjelleberg S, Harder T, et al. Identification of the antibacterial compound produced by the marine epiphytic bacterium Pseudovibrio sp. D323 and related sponge-associated bacteria. Marine Drugs. 2011;9(8):1391-1402. DOI: 10.3390/md9081391
  9. 9. Gohel SD, Sharma AK, Thakrar FJ, Singh SP. Endophytic actinobacteria and their interactions with plant host systems. In: Understanding Host-microbiome Interactions-an Omics Approach. Singapore: Springer; 2017. pp. 247-246
  10. 10. Shady NH, Tawfike AF, Yahia R, Fouad MA, Brachmann AO, Piel J, et al. Cytotoxic activity of actinomycetes Nocardia sp. and Nocardiopsi s sp. associated with marine sponge Amphimedon sp. Natural Product Research. 2021;19:1-6. DOI: 10.1080/14786419.2021.1931865
  11. 11. Hindu SV, Chandrasekaran N, Mukherjee A, Thomas J. A review on the impact of seaweed polysaccharide on the growth of probiotic bacteria and its application in aquaculture. Aquaculture International. 2019;27(1):227-238. DOI: 10.1007/s10499-018-0318-3
  12. 12. De Voogd NJ, Cleary DF. An analysis of sponge diversity and distribution at three taxonomic levels in the Thousand Islands/Jakarta Bay reef complex, West-Java, Indonesia. Marine Ecology. 2008;29(2):205-215. DOI: 10.1111/j.1439-0485.2008.00238.x
  13. 13. Baker PW, Kennedy J, Morrissey J, O’Gara F, Dobson AD, Marchesi JR. Endoglucanase activities and growth of marine-derived fungi isolated from the sponge Haliclonasimulans. Journal of Applied Microbiology. 2010;108(5):1668-1675. DOI: 10.1111/j.1365-2672.2009.04563.x
  14. 14. Ziegler M, Seneca FO, Yum LK, Palumbi SR, Voolstra CR. Bacterial community dynamics are linked to patterns of coral heat tolerance. Nature Communications. 2017;8(1):1-8. DOI: 10.1038/ncomms14213
  15. 15. Hifnawy M, Hassan HM, Mohammed R, Fouda M, Sayed AM, Hamed A, et al. Induction of Antibacterial Metabolites by Co-Cultivation of Two Red-Sea-Sponge-Associated Actinomycetes Micromonospora sp. UR56 and Actinokinespora sp. EG49. Marine Drugs. 2020;18(5):243
  16. 16. Krishnamoorthy M, Dharmaraj D, Rajendran K, Karuppiah K, Balasubramanian M, Ethiraj K. Pharmacological activities of coral reef associated actinomycetes, Saccharopolyspora sp. IMA1. Biocatalysis and Agricultural Biotechnology. 2020;28:101748. DOI: 10.1016/j.bcab.2020.101748
  17. 17. Ramesh C, Tulasi BR, Raju M, Thakur N, Dufossé L. Marine natural products from tunicates and their associated microbes. Marine Drugs. 2021;19(6):308. DOI: 10.3390/md19060308
  18. 18. Campana S, Busch K, Hentschel U, Muyzer G, de Goeij JM. DNA-stable isotope probing (DNA-SIP) identifies marine sponge-associated bacteria actively utilizing dissolved organic matter (DOM). Environmental Microbiology. 2021;2022. DOI: 10.1111/1462-2920.15642
  19. 19. Ravikumar S, Krishnakumar S, Inbaneson SJ, Gnanadesigan M. Antagonistic activity of marine actinomycetes from Arabian Sea coast. Archives of Applied Science Research. 2010;2(6):273-280
  20. 20. Li Z, Sun W, Zhang F, He L, Loganathan K. Actinomycetes from the South China Sea sponges: Isolation, diversity and potential for aromatic polyketides discovery. Frontiers in Microbiology. 2015;6:1048
  21. 21. Sun W, Liu C, Zhang F, Zhao M, Li Z. Comparative genomics provides insights into the marine adaptation in sponge-derived Kocuriaflava S43. Frontiers in Microbiology. 2018;9:1257. DOI: 10.3389/fmicb.2015.01048
  22. 22. Mahmoud HM, Kalendar AA. Coral-associated Actinobacteria: Diversity, abundance, and biotechnological potentials. Frontiers in Microbiology. 2016;7:204. DOI: 10.3389/fmicb.2016.00204
  23. 23. Kuang W, Li J, Zhang S, Long L. Diversity and distribution of Actinobacteria associated with reef coral Porites lutea. Frontiers in Microbiology. 2015;6:1094. DOI: 10.3389/fmicb.2015.01094
  24. 24. Nithyanand P, Manju S, Karutha PS. Phylogenetic characterization of culturable actinomycetes associated with the mucus of the coral Acropora digitifera from Gulf of Mannar. FEMS Microbiology Letters. 2011;314(2):112-118. DOI: 10.1111/j.1574-6968.2010.02149.x
  25. 25. Tremblay P, Maguer JF, Grover R, Ferrier-Pagès C. Trophic dynamics of scleractinian corals: Stable isotope evidence. The Journal of experimental biology. 2015;218(8):1223-1234. DOI: 10.1242/jeb.115303
  26. 26. Little AF, Van Oppen MJ, Willis BL. Flexibility in algal endosymbioses shapes growth in reef corals. Science. 2004;304(5676):1492. DOI: 10.1126/science.1095733
  27. 27. Lesser MP, Morrow KM, Pankey SM, Noonan SH. Diazotroph diversity and nitrogen fixation in the coral Stylophorapistillata from the Great Barrier Reef. The ISME Journal. 2018;12(3):813-824. DOI: 10.1038/s41396-017-0008-6
  28. 28. Lampert Y, Kelman D, Dubinsky Z, Nitzan Y, Hill RT. Diversity of culturable bacteria in the mucus of the Red Sea coral Fungiascutaria. FEMS Microbiology Ecology. 2006;58(1):99-108. DOI: 10.1111/j.1574-6941.2006.00136.x
  29. 29. Li F, Xie Q , Zhou S, Kong F, Xu Y, Ma Q , et al. Nocardiopsiscoralli sp. nov. a novel actinobacterium isolated from the coral Galaxeaastreata. International Journal of Systematic and Evolutionary Microbiology. 2021;71(6):004817
  30. 30. Gozari M, Mortazavi MS, Bahador N, Rabbaniha M. Isolation and screening of antibacterial and enzyme producing marine actinobacteria to approach probiotics against some pathogenic vibrios in shrimp Litopenaeusvannamei. Iranian Journal of Fisheries Sciences. 2016;15(2):630-644
  31. 31. You JL, Cao LX, Liu GF, Zhou SN, Tan HM, Lin YC. Isolation and characterization of actinomycetes antagonistic to pathogenic Vibrio spp. from nearshore marine sediments. World Journal of Microbiology and Biotechnology. 2005;21(5):679-682. DOI: 10.1007/s11274-004-3851-3
  32. 32. Kumar SS, Philip R, Achuthankutty CT. Antiviral property of marine actinomycetes against white spot syndrome virus in penaeid shrimps. Current Science. 2006;25:807-811. DOI: stable/24093913
  33. 33. You J, Xue X, Cao L, Lu X, Wang J, Zhang L, et al. Inhibition of Vibrio biofilm formation by a marine actinomycete strain A66. Applied Microbiology and Biotechnology. 2007;76(5):1137-1144. DOI: 10.1007/s00253-007-1074-x
  34. 34. Steinert G, Taylor MW, Schupp PJ. Diversity of actinobacteria associated with the marine ascidian Eudistomatoealensis. Marine Biotechnology. 2015;17(4):377-385. DOI: 10.1007/s10126-015-9622-3
  35. 35. Lee NY, Cha CJ, Im WT, Kim SB, Seong CN, Bae JW, et al. A report of 42 unrecorded actinobacterial species in Korea. Journal of Species Research. 2018;7(1):36-49
  36. 36. Selvin J, Shanmughapriya S, Gandhimathi R, Kiran GS, Ravji TR, Natarajaseenivasan K, et al. Optimization and production of novel antimicrobial agents from sponge associated marine actinomycetes Nocardiopsisdassonvillei MAD08. Applied Microbiology and Biotechnology. 2009;83(3):435-445. DOI: 10.1007/s00253-009-1878-y
  37. 37. Jiang S, Li X, Zhang L, Sun W, Dai S, Xie L, et al. Culturable actinobacteria isolated from marine sponge Iotrochota sp. Marine Biology. 2008;153(5):945-952. DOI: 10.1007/s00227-007-0866-y
  38. 38. Ekiz G, Hames EE, Demir V, Can F, Yokes MB, Uzel A, et al. Cultivable sponge-associated actinobacteria from coastal area of Eastern Mediterranean Sea. Advances in Microbiology. 2014;2014
  39. 39. Schneemann I, Nagel K, Kajahn I, Labes A, Wiese J, Imhoff JF. Comprehensive investigation of marine Actinobacteria associated with the sponge Halichondriapanicea. Applied and Environmental Microbiology. 2010;76(11):3702-3714. DOI: 10.1128/AEM.00780-10
  40. 40. Sandoval-Powers M, Králová S, Nguyen GS, Fawwal DV, Degnes K, Lewin AS, et al. Streptomyces poriferorum sp. nov., a novel marine sponge-derived Actinobacteria species expressing anti-MRSA activity. Systematic and Applied Microbiology. 2021;44(5):126244
  41. 41. Bose U, Ortori CA, Sarmad S, Barrett DA, Hewavitharana AK, Hodson MP, et al. Production of N-acyl homoserine lactones by the sponge-associated marine actinobacteria Salinisporaarenicola and Salinisporapacifica. FEMS Microbiology Letters. 2017;1:364. DOI: 10.1093/femsle/fnx002
  42. 42. Retnowati D, Solihin DD, Ghulamahdi M, Lestari Y. New information on the potency ofthe potency of sponge-associated actinobacteria as producer of plant growth-promoting bioactive compounds. Malaysian Applied Biology. 2018;47(6):127-135
  43. 43. De Menezes CB, Afonso RS, de Souza WR, Parma MM, de Melo IS, Fugita FL, et al. Williamsiaaurantiacus sp. nov. a novel actinobacterium producer of antimicrobial compounds isolated from the marine sponge. Archives of Microbiology. 2019;201(5):691-698. DOI: 10.1007/s00203-019-01633-z
  44. 44. Ocampo-Alvarez H, Meza-Canales ID, Mateos-Salmón C, Rios-Jara E, Rodríguez-Zaragoza FA, Robles-Murguía C, et al. Diving into reef ecosystems for land-agriculture solutions: Coral microbiota can alleviate salt stress during germination and photosynthesis in terrestrial plants. Frontiers in Plant Science. 2020;25(11):648. DOI: 10.3389/fpls.2020.00648
  45. 45. Erba E, Bergamaschi D, Ronzoni S, Faretta M, Taverna S, Bonfanti M, et al. Mode of action of thiocoraline, a natural marine compound with anti-tumour activity. British Journal of Cancer. 1999;80(7):971-980. DOI: 10.1038/sj.bjc.6690451
  46. 46. Poongodi SU, Karuppiah VA, Sivakumar KA, Kannan LA. Marine actinobacteria of the coral reef environment of the gulf of Mannar biosphere reserve, India: A search for antioxidant property. International Journal of Pharmacy and Pharmaceutical Sciences. 2012;4:316-321
  47. 47. ElAhwany AM, Ghozlan HA, ElSharif HA, Sabry SA. Phylogenetic diversity and antimicrobial activity of marine bacteria associated with the soft coral Sarcophyton glaucum. Journal of Basic Microbiology. 2015;55(1):2-10. DOI: 10.1002/jobm.201300195
  48. 48. Sarmiento Vizcaíno A, González Iglesias V, Fernández Braña AJ, Molina Ramírez A, Acuña Fernández JL, García Díaz LA, et al. Myceligeneranscantabricum sp. nov., a barotolerant actinobacterium isolated from a deep cold-water coral. International Journal of Systematic and Evolutionary Microbiology. 2015;65(4)
  49. 49. Li J, Dong JD, Yang J, Luo XM, Zhang S. Detection of polyketide synthase and nonribosomal peptide synthetase biosynthetic genes from antimicrobial coral-associated actinomycetes. Antonie Van Leeuwenhoek. 2014;106(4):623-635. DOI: 10.1007/s10482-014-0233-1
  50. 50. Lin Z, Antemano RR, Hughen RW, Tianero MD, Peraud O, Haygood MG, et al. Pulicatins A− E, neuroactive thiazoline metabolites from cone snail-associated bacteria. Journal of Natural Products. 2010;73:1922-1926. DOI: 10.1021/np100588c
  51. 51. Lin Z, Torres JP, Ammon MA, Marett L, Teichert RW, Reilly CA, et al. A bacterial source for mollusk pyrone polyketides. Chemistry & Biology. 2013;20(1):73-81. DOI: 10.1016/j.chembiol.2012.10.019
  52. 52. García Bernal M, Trabal Fernández N, Saucedo Lastra PE, Medina Marrero R, Mazón-Suástegui JM. Streptomyces effect on the bacterial microbiota associated to Crassostrea sikamea oyster. Journal of Applied Microbiology. 2017;122(3):601-614. DOI: 10.1111/jam.13382
  53. 53. Indraningrat AA, Smidt H, Sipkema D. Bioprospecting sponge-associated microbes for antimicrobial compounds. Marine Drugs. 2016;14(5):87. DOI: 10.3390/md14050087
  54. 54. Schneemann I, Kajahn I, Ohlendorf B, Zinecker H, Erhard A, Nagel K, et al. Mayamycin, a cytotoxic polyketide from a Streptomyces strain isolated from the marine sponge Halichondriapanicea. Journal of Natural Products. 2010;73(7):1309-1312. DOI: 10.1021/np100135b
  55. 55. Eltamany EE, Abdelmohsen UR, Ibrahim AK, Hassanean HA, Hentschel U, Ahmed SA. New antibacterial xanthone from the marine sponge-derived Micrococcus sp. EG45. Bioorganic & Medicinal Chemistry Letters. 2014;24(21):4939-4942
  56. 56. Gandhimathi R, Arunkumar M, Selvin J, Thangavelu T, Sivaramakrishnan S, Kiran GS, et al. Antimicrobial potential of sponge associated marine actinomycetes. Journal de MycologieMedicale. 2008;18(1):16-22. DOI: 10.1016/j.mycmed.2007.11.001
  57. 57. Liu T, Wu S, Zhang R, Wang D, Chen J, Zhao J. Diversity and antimicrobial potential of Actinobacteria isolated from diverse marine sponges along the Beibu Gulf of the South China Sea. FEMS Microbiology Ecology. 2019;95(7):fiz089
  58. 58. Jagannathan SV, Manemann EM, Rowe SE, Callender MC, Soto W. Marine actinomycetes, new sources of biotechnological products. Marine Drugs. 2021;19(7):365. DOI: 10.3390/md19070365
  59. 59. Mahapatra GP, Raman S, Nayak S, Gouda S, Das G, Patra JK. Metagenomics approaches in discovery and development of new bioactive compounds from marine actinomycetes. Current Microbiology. 2020;77(4):645-656. DOI: 10.1007/s00284-019-01698-5
  60. 60. Poongodi S, Karuppiah V, Sivakumar K, Kannan L. Antioxidant activity of Nocardiopsis sp., a marine actinobacterium, isolated from the Gulf of Mannar Biosphere Reserve, India. National Academy Science Letters. 2014;37(1):65-70. DOI: 10.1007/s40009-013-0193-4
  61. 61. Fahmy NM, Abdel-Tawab AM. Isolation and characterization of marine sponge–associated Streptomyces sp. NMF6 strain producing secondary metabolite (s) possessing antimicrobial, antioxidant, anticancer, and antiviral activities. Journal, Genetic Engineering & Biotechnology. 2021;19(1):1-4. DOI: 10.1186/s43141-021-00203-5
  62. 62. Webb HE, Brichta-Harhay DM, Brashears MM, Nightingale KK, Arthur TM, Bosilevac JM, et al. Salmonella in peripheral lymph nodes of healthy cattle at slaughter. Frontiers in Microbiology. 2017;9(8):2214. DOI: 10.3389/fmicb.2017.02214
  63. 63. Hassan SS, Anjum K, Abbas SQ , Akhter N, Shagufta BI, Shah SA, et al. Emerging biopharmaceuticals from marine actinobacteria. Environmental Toxicology and Pharmacology. 2017;49:34-47. DOI: 10.1016/j.etap.2016.11.015
  64. 64. Shin HJ, Lee HS, Lee JS, Shin J, Lee MA, Lee HS, et al. Violapyrones H and I, new cytotoxic compounds isolated from Streptomyces sp. associated with the marine starfish Acanthasterplanci. Marine Drugs. 2014;12(6):3283-3291. DOI: 10.3390/md12063283
  65. 65. El-Shatoury SA, El-Shenawy NS, Abd El-Salam IM. Antimicrobial, antitumor and in vivo cytotoxicity of actinomycetes inhabiting marine shellfish. World Journal of Microbiology and Biotechnology. 2009;25(9):1547-1555. DOI: 10.1007/s11274-009-0040-4
  66. 66. Liu CY, Li YL, Lu JH, Qian LL, Xu K, Wang NN, et al. A new steffimycin-type derivative from the lichen-derived actinomycetes steptomyces sp. Journal of Molecular Structure. 2021;1227:129352. DOI: 10.1016/j.molstruc.2020.129352
  67. 67. Zhang Z, Sibero MT, Kai A, Fukaya K, Urabe D, Igarashi Y. TMKS8A, an antibacterial and cytotoxic chlorinated α-lapachone, from a sea slug-derived actinomycete of the genus Streptomyces. The Journal of Antibiotics. 2021;74(7):464-469. DOI: 10.1038/s41429-021-00415-4
  68. 68. Zhang X, Song C, Bai Y, Hu J, Pan H. Cytotoxic and antimicrobial activities of secondary metabolites isolated from the deep-sea-derived Actinoalloteichuscyanogriseus 12A22. 3. Biotech. 2021;11(6):1-8. DOI: 10.1007/s13205-021-02846-0
  69. 69. Gohel SD, Sharma AK, Dangar KG, Thakrar FJ, Singh SP. Biology and applications of halophilic and haloalkaliphilic actinobacteria. In: Extremophiles. CRC Press; 2018. pp. 103-136
  70. 70. Majithiya V, Gohel S. Isolation and screening of extracellular enzymes producing Actinobacteria associated with sea weed. In: Proceedings of the National Conference on Innovations in Biological Sciences (NCIBS). 2020
  71. 71. Gohel SD, Sharma AK, Dangar KG, Thakrar FJ, Singh SP. Antimicrobial and biocatalytic potential of haloalkaliphilic actinobacteria. In: Halophiles. Cham: Springer; 2015. pp. 29-55
  72. 72. Singh SP, Thumar JT, Gohel SD, Kikani B, Shukla R, Sharma A, et al. Actinomycetes from marine habitats and their enzymatic potential. In: Marine Enzymes for Biocatalysis. Woodhead Publishing; 2013. pp. 191-214
  73. 73. Gohel SD, Singh SP. Thermodynamics of a Ca2+−dependent highly thermostable alkaline protease from a haloalkliphilic actinomycete. International Journal of Biological Macromolecules. 2015;72:421-429. DOI: 10.1016/j.ijbiomac.2014.08.008
  74. 74. Gohel SD, Singh SP. Cloning and expression of alkaline protease genes from two salt-tolerant alkaliphilic actinomycetes in E. coli. International Journal of Biological Macromolecules. 2012;50(3):664-671. DOI: 10.1016/j.ijbiomac.2012.01.039
  75. 75. Bourne DG, Munn CB. Diversity of bacteria associated with the coral Pocilloporadamicornis from the Great Barrier Reef. Environmental Microbiology. 2005;7(8):1162-1174. DOI: 10.1111/j.1462-2920.2005.00793.x
  76. 76. Feby A, Nair S. Sponge-associated bacteria of Lakshadweep coral reefs, India: Resource for extracellular hydrolytic enzymes. Advances in Bioscience and Biotechnology. 2010;1:330-337. DOI: 10.4236/abb.2010.14043
  77. 77. Su H, Xiao Z, Yu K, Huang Q , Wang G, Wang Y, et al. Diversity of cultivable protease-producing bacteria and their extracellular proteases associated to scleractinian corals. PeerJ. 2020;8:e9055
  78. 78. Kikani BA, Singh SP. Amylases from thermophilic bacteria: Structure and function relationship. Critical Reviews in Biotechnology. 2021;20:1-7. DOI: 10.1080/07388551.2021.1940089
  79. 79. Strahler J, Skoluda N, Kappert MB, Nater UM. Simultaneous measurement of salivary cortisol and alpha-amylase: Application and recommendations. Neuroscience & Biobehavioral Reviews. 2017;83:657-677. DOI: 10.1016/j.neubiorev.2017.08.015
  80. 80. Meena B, Anburajan L, Nitharsan K, Vinithkumar NV, Dharani G. Existence in cellulose shelters: Industrial and pharmaceutical leads of symbiotic actinobacteria from ascidian Phallusia nigra, Andaman Islands. World Journal of Microbiology and Biotechnology. 2021;37(7):1-22. DOI: 10.1007/s11274-021-03090-7
  81. 81. Feby A, Nair S. Sponge-associated bacteria of Lakshadweep coral reefs, India. Enzymes. 2010;ct1:330-337. DOI: 10.4236/abb.2010.14043
  82. 82. Ober C, Chupp GL. The chitinase and chitinase-like proteins: A review of genetic and functional studies in asthma and immune-mediated diseases. Current Opinion in Allergy and Clinical Immunology. 2009;9(5):401. DOI: 10.1097%2FACI.0b013e3283306533
  83. 83. Zhang W, Liu Y, Ma J, Yan Q , Jiang Z, Yang S. Biochemical characterization of a bifunctional chitinase/lysozyme from Streptomyces sampsonii suitable for N-acetyl chitobiose production. Biotechnology Letters. 2020;1:42
  84. 84. Nakagawa YS, Kudo M, Onodera R, Ang LZ, Watanabe T, Totani K, et al. Analysis of four chitin-active lytic polysaccharide monooxygenases from Streptomyces griseus reveals functional variation. Journal of Agricultural and Food Chemistry. 2020;68(47):13641-13650. DOI: 10.1021/acs.jafc.0c05319
  85. 85. Lv C, Gu T, Ma R, Yao W, Huang Y, Gu J, et al. Biochemical characterization of a GH19 chitinase from Streptomyces alfalfae and its applications in crystalline chitin conversion and biocontrol. International Journal of Biological Macromolecules. 2021;167:193-201. DOI: 10.1016/j.ijbiomac.2020.11.178
  86. 86. Chand S, Mahajan RV, Prasad JP, Sahoo DK, Mihooliya KN, Dhar MS, et al. A comprehensive review on microbial l-asparaginase: Bioprocessing, characterization, and industrial applications. Biotechnology and Applied Biochemistry. 2020;67(4):619-647. DOI: 10.1002/bab.1888
  87. 87. Dharmaraj S. Study of L-asparaginase production by Streptomyces noursei MTCC 10469, isolated from marine sponge Callyspongiadiffusa. Iranian Journal of Biotechnology. 2011;9(2):102-108
  88. 88. Meena B, Anburajan L, Sathish T, Raghavan RV, Dharani G, Vinithkumar NV, et al. L-asparaginase from Streptomyces griseus NIOT-VKMA29: Optimization of process variables using factorial designs and molecular characterization of l-asparaginase gene. Scientific Reports. 2015;5(1):1-2. DOI: 10.1038/srep12404
  89. 89. Rajagopal G, Kannan S. Systematic characterization of potential cellulolytic marine actinobacteria Actinoalloteichus sp. MHA15. Biotechnology Reports. 2017;13:30-36. DOI: 10.1016/j.btre.2016.12.003
  90. 90. Bhattacharya AS, Bhattacharya A, Pletschke BI. Synergism of fungal and bacterial cellulases and hemicellulases: A novel perspective for enhanced bio-ethanol production. Biotechnology Letters. 2015;37(6):1117-1129. DOI: 10.1007/s10529-015-1779-3
  91. 91. Kasanah N, Triyanto T. Bioactivities of halometabolites from marine actinobacteria. Biomolecules. 2019;9(6):225. DOI: 10.3390/biom9060225
  92. 92. Menon BR, Richmond D, Menon N. Halogenases for biosynthetic pathway engineering: Toward new routes to naturals and non-naturals. Catalysis Reviews. 2020;16:1-59. DOI: 10.1080/01614940.2020.1823788
  93. 93. Barka EA, Vatsa P, Sanchez L, Gaveau-Vaillant N, Jacquard C, Klenk HP, et al. Taxonomy, physiology, and natural products of Actinobacteria. Microbiology and Molecular Biology Reviews. 2016;80:1-43. DOI: 10.1128/MMBR.00019-15
  94. 94. Zhang W, Ma L, Li S, Liu Z, Chen Y, Zhang H, et al. Indimicins A–E, bisindole alkaloids from the deep-sea-derived Streptomyces sp. SCSIO 03032. Journal of Natural Products. 2014;77(8):1887-1892. DOI: 10.1021/np500362p
  95. 95. Fu P, Kong F, Wang Y, Wang Y, Liu P, Zuo G, et al. Antibiotic metabolites from the coral-associated actinomycete Streptomyces sp. OUCMDZ-1703. Chinese Journal of Chemistry. 2013;31(1):100-104. DOI: 10.1002/cjoc.201201062
  96. 96. Li XG, Tang XM, Xiao J, Ma GH, Xu L, Xie SJ, et al. Harnessing the potential of halogenated natural product biosynthesis by mangrove-derived actinomycetes. Marine Drugs. 2013;11:3875-3890. DOI: 10.3390/md11103875
  97. 97. Ismail M, Frese M, Patschkowski T, Ortseifen V, Niehaus K, Sewald N. Flavin-dependent Halogenases from Xanthomonas campestris pv. Campestris B100 prefer bromination over chlorination. Advanced Synthesis and Catalysis. 2019;361(11):2475-2486. DOI: 10.1002/adsc.201801591
  98. 98. Bayer K, Scheuermayer M, Fieseler L, Hentschel U. Genomic mining for novel FADH 2-dependent halogenases in marine sponge-associated microbial consortia. Marine Biotechnology. 2013;15(1):63-72. DOI: 10.1007/s10126-012-9455-2
  99. 99. Sakuda S, Isogai A, Matsumoto S, Suzuki A, Koseki K. The structure of allosamidin, a novel insect chitinase inhibitor, produced by Streptomyces sp. Tetrahedron Letters. 1986;27(22):2475-2478. DOI: 10.1016/S0040-4039(00)84560-8
  100. 100. Meng P, Xie C, Geng P, Qi X, Zheng F, Bai F. Inhibitory effect of components from Streptomyces species on α-glucosidase and α-amilase of different origin. Applied Biochemistry and Microbiology. 2013;49(2):160-168. DOI: 10.1134/S0003683813020099
  101. 101. Xu JL, Liu ZF, Zhang XW, Liu HL, Wang Y. Microbial oligosaccharides with biomedical applications. Marine Drugs. 2021;19(6):350. DOI: 10.3390/md19060350
  102. 102. Siddharth S, Vittal RR. Evaluation of antimicrobial, enzyme inhibitory, antioxidant and cytotoxic activities of partially purified volatile metabolites of marine Streptomyces sp. S2A. Microorganisms. 2018;6(3):72
  103. 103. Kamarudheen N, Khaparde A, Gopal DS, Rao KB. Unraveling a natural protease inhibitor from marine Streptomyces griseoincarnatus HK12 active against chikungunya virus. Microbiological Research. 2021;31:126858. DOI: 10.1016/j.micres.2021.126858
  104. 104. Kamarudheen N, Rao B. An overview of protease inhibitors from Actinobacteria. Research Journal of Biotechnology. 2018;13:1

Written By

Vaishali R. Majithiya and Sangeeta D. Gohel

Submitted: 26 September 2021 Reviewed: 19 July 2022 Published: 18 October 2022