Next Article in Journal
Synthesis and Antimicrobial Evaluation of Side-Chain Derivatives based on Eurotiumide A
Next Article in Special Issue
The First Genome Survey of the Antarctic Krill (Euphausia superba) Provides a Valuable Genetic Resource for Polar Biomedical Research
Previous Article in Journal
Mining Natural Product Biosynthesis in Eukaryotic Algae
Previous Article in Special Issue
Synthesis of Bioactive Silver Nanoparticles by a Pseudomonas Strain Associated with the Antarctic Psychrophilic Protozoon Euplotes focardii
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Deep Hypersaline Anoxic Basins as Untapped Reservoir of Polyextremophilic Prokaryotes of Biotechnological Interest

by
Stefano Varrella
1,
Michael Tangherlini
2 and
Cinzia Corinaldesi
1,*
1
Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, 60131 Ancona, Italy
2
Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
*
Author to whom correspondence should be addressed.
Mar. Drugs 2020, 18(2), 91; https://doi.org/10.3390/md18020091
Submission received: 28 November 2019 / Revised: 27 January 2020 / Accepted: 28 January 2020 / Published: 30 January 2020
(This article belongs to the Special Issue Bioactive Molecules from Extreme Environments)

Abstract

:
Deep-sea hypersaline anoxic basins (DHABs) are considered to be among the most extreme ecosystems on our planet, allowing only the life of polyextremophilic organisms. DHABs’ prokaryotes exhibit extraordinary metabolic capabilities, representing a hot topic for microbiologists and biotechnologists. These are a source of enzymes and new secondary metabolites with valuable applications in different biotechnological fields. Here, we review the current knowledge on prokaryotic diversity in DHABs, highlighting the biotechnological applications of identified taxa and isolated species. The discovery of new species and molecules from these ecosystems is expanding our understanding of life limits and is expected to have a strong impact on biotechnological applications.

1. Introduction

Deep-sea ecosystems (waters and seabeds of the ocean beneath 200 m depth) are the largest, most remote, and least explored biomes of the biosphere, comprising more than two-thirds of the oceanic volume [1,2,3]. They are characterized by absence of light, an average depth of approximately 4200 m, temperatures below 4 °C, and a hydrostatic pressure of about 40 MPa; taken together, these factors encompass some of the harshest environments on our planet, representing a challenge for the existence of life [2]. Over the last few decades, many deep-sea surveys have resulted in the discovery of highly diversified and peculiar habitats [2,4,5,6], including hydrothermal vents, cold seeps, mud volcanoes, and deep hypersaline anoxic basins, where life conditions are even more extreme [7]. Among these, deep hypersaline anoxic basins (DHABs) are defined as polyextreme ecosystems [8,9].
DHABs were discovered at the end of the last century on the seafloor in different deep-sea areas (at depths ranging from 630 m to 3580 m) around the globe (Figure 1), including the Mediterranean Sea [10,11], the Red Sea [12,13,14] and the Gulf of Mexico [15]. Intriguingly, the discovery of new DHABs is still ongoing, such as with the recent discovery of the new Thetis, Kyros, and Haephestus basins in the Mediterranean Sea [16,17,18]. To date, with the recent finding of these new DHABs, 35 basins have been discovered around the world. The Bannock, Tyro, Urania, L’Atalante, and Discovery basins are the deepest known DHABs, being far below the photic zone (3200–3500 m deep), and are located along the Mediterranean Ridge in the Eastern Mediterranean Sea, an accretionary complex subjected to continental collision [19]. Two of the most studied DHABs in the Red Sea are the Shaban and the Kebrit deeps. The Shaban Deep comprises four depressions at a depth of 1325 m, whereas the Kebrit Deep is a rounded basin of approximately 1 km in diameter found at a depth of 1549 m [20].
Different sampling strategies have been adopted to explore the general physical structure of DHABs. For instance, Mediterranean DHABs have been sampled through a rosette with Niskin bottles equipped with a conductivity, temperature, and depth (CTD) sensor and connected to a live camera to monitor the sampling operations [21,22,23]. Geochemical data of DHABs, such as those located in the Gulf of Mexico, have been collected through a brine-trapper, which was used to collect vertically water from different layers of the seawater–brine interface [24].
Despite the different geological features found in DHABs, most of them are derived from the re-dissolution of evaporitic minerals, like halite (NaCl-mineral) and kieserite (MgSO4-mineral), after exposition to seawater due to tectonic activity [25,26]. This determines a salt-induced stratification of the water column (Figure 2), which drives the formation of a stable, dense, hypersaline brine lake with a variable thickness, ranging from one to tens of meters; this brine lake represents a polyextreme environment because its conditions hinder oxygen exchange, creating euxinic conditions, including high hydrostatic pressure, extremely low water activity and chaotropicity, and sharp oxy-, picno-, and chemoclines at the seawater–brine interface [21,23,27]. The salt concentration progressively increases over depth in the overlying halocline interface, reaching brines values up to 7–10 times higher than those existing in seawater [28].
Chemical and physical characteristics are specific to each DHAB and greatly vary depending on how the brine was formed along with the geographic localization (Table 1). The majority of the DHABs are thalassohaline (most of the dissolved ions are represented by those composing the overlaying seawater), whereas the Discovery, Kryos, and Hephaestus basins are athalassohaline and are characterized by high Mg2+ concentrations likely deriving from the dissolution of magnesium chloride salts (i.e., bischofite [18])
Overall, DHABs can be subdivided into four different systems: the seawater–brine interface, brines, the brine–sediment interface, and the sediments underlying the brines. Each of these features is characterized by specific conditions such as the steep halocline at the water–brine interface or the anoxic conditions of the sediments beneath the brines. In addition, the geochemical characteristics of each DHAB are mostly dependent on their geological evolution and origin. The high density of the brine prevents their mixing with the overlying oxygenated seawater, thus making the DHABs completely anoxic [16]. Their different hydrochemistry and physical separation for thousands of years has made these systems greatly interesting for scientists due to their potential similarity with extraterrestrial environments [18,34,37]. Despite their extreme conditions, many studies have provided evidence of a highly active prokaryotic community and of the presence of living metazoans, greatly extending our knowledge regarding the limits of organisms’ adaptions to life [20,27,38,39,40,41,42,43,44,45,46,47,48]. These organisms require specific adaptations for withstanding numerous physicochemical stresses [49].
The complex structure and conditions of the DHABs, such as the presence of the steep halo- and oxyclines, have been found to influence the distribution, structure, and richness of the microbial communities living in these environments [19]. Many studies have been focused on the halocline, which entraps nutrients, sinking organic materials, minerals, and microbial cells, and creates environmental gradients of great interest not only for identifying and isolating novel organisms but also for clarifying their metabolic strategies employed for adapting to extreme conditions [19]. The variable accumulation of metals and nutrients, especially in the halocline, supports the presence of different ecological niches exploited by highly diverse microorganisms with peculiar features [38]. However, to date, our knowledge of how these organisms are affected and contribute to the geochemical properties of the DHABs is still limited.
The presence of life in these extreme environments has raised important questions about the molecular mechanisms that extremophiles have developed to overcome harsh conditions. Many studies have highlighted several peculiar adaptive strategies of halophilic microorganisms for maintaining stability and functionality of all their cellular components under such conditions [50,51]. Hence, microorganisms inhabiting extreme saline habitats not only have been considered useful subjects for ecological and evolutionary studies [50] but also hold an outstanding ability to produce bioactive molecules and enzymes, which can also be exploited for industrial and biotechnological purposes as well as for human wellness [52,53]. Considering the promising biotechnological potential of bacteria and archaea from DHABs due to their capability to live under extreme conditions, the present review provides an outline of the prokaryotic biodiversity in DHABs, highlighting their potential in producing enzymes and bioactive molecules for industrial, pharmaceutical, and environmental applications.

2. Prokaryotic Assemblages of DHABs

The specific characteristics and geochemical conditions of each DHAB have driven the development of different and highly-stratified communities. Brines and the seawater–brine interfaces, indeed, represent the most widely-studied domains within DHABs from both a taxonomic and ecological/functional point of view [21,23]. The halocline is a microbial “hotspot”, harboring dense microbial populations that appear to be more metabolically active than those of the adjacent layers, with the presence of unique bacterial lineages having been found [21,22,44,54,55]. Several microbial lineages have been identified within DHAB brines. In particular, many members of the new Mediterranean Sea Brine Lake lineages (MSBL1–6) have been found extensively across hypersaline basins [56] from the Mediterranean to the Red Sea (despite their name), and include Archaea (e.g., MBSL1, which are sugar-fermenting organisms capable of autotrophic growth [23,56,57] and other major divisions of bacteria (MSBL2–6 [21,38]). Interestingly, the bacterial MSBL2 lineage has shown high similarity to the SB1 division found in the Shaban Deep brine pool, located in the Red Sea, which represents a novel halophilic lineage within bacteria, with no close cultivated relatives observed so far [28,38]. Similarly to the MBSL lineages, bacteria belonging to candidate division KB1 have been identified for the first time within the Kebrit Deep basin (Red Sea [20]) and subsequently have also been found in other DHABs of the Red Sea [58], as well as in other basins (e.g., the halocline of Mediterranean Sea brine pools and pools from the Gulf of Mexico [17,21,22,31,59]). Bacteria from this division can import and/or produce glycine betaine in response to osmotic stress [59]. The KB1 glycine betaine transport systems seem to aid not only in maintaining osmotic balance but also have a role in methane production [59]. Delta- and Epsilonproteobacteria are also widely distributed across DHABs. 16S rRNA gene libraries from the Bannock, Hephaestus, and L’Atalante basins (Mediterranean Sea) have provided evidence of the presence of sulfate-reducing Deltaproteobacteria (in particular belonging to the ANME-1 clade, responsible for the anaerobic oxidation of methane [16]) and sulfur-oxidizing Gamma- and Epsilonproteobacteria [21,22]. In the GC233 basin within the Gulf of Mexico, combining geochemical data and molecular analyses, different Deltaproteobacteria sulfate-reducers (related to Desulfosarcinales, Desulfobacterium, Desulfobulbus, and Desulfocapsa) and sulfide-oxidizing Epsilonproteobacteria have been found, leading to the hypothesizing of the presence of a sulfur-cycling microbial community [15,60].
Archaea associated with the ammonia-oxidizing Thaumarchaeota Marine Group I have also been found across several DHABs worldwide [18,21,22]. In particular, they have appeared to be the most representative prokaryotic members in different Red Sea DHABs, though with different contributions: in the Atlantis II and Discovery, 99% of archaeal operational taxonomic units (OTUs) were found to belong to the phylum Thaumarchaeota, whereas in the Erba basin the percentage was about 64% [30]. Members of this phylum are capable of fixing CO2 and oxidizing methane, contributing to dark primary production [61]. Overall, the dominant thaumarchaeal lineage is closer to the genus Nitrosopumilus [62]. The adaptation of this genotype to the hostile brine–sediment interface environment can be possible not only by increasing intra-cellular salt concentrations [63] but also for the presence of “acidic tuned” membrane proteins which show optimal activity and stability at high salinity [64]. Furthermore, genomic analyses have revealed the presence of specific pathways for taking up a mixture of osmolytes and other genes encoding for the biosynthesis of ectoine/hydroxyectoine, which are not present in mesopelagic clades [30]. However, different DHAB geochemistry may shape other thaumarchaeal lineages. Genomic analyses have revealed a newly isolated methanogenic archeon from the sulfide-rich halocline of Kebrit, which holds adaptive traits (e.g., osmoprotection and oxidative stress response) for counteracting the harsh local conditions [65].
Apparently, as high salinity is one of the main features of DHABs, halophilic organisms have been found across all the basins, and most of the isolated halophilic strains also display interesting metabolic features. In particular, 33 halotolerant bacterial strains have been isolated from the halocline of the Urania, Bannock, Discovery, and L’Atalante basins [66]. For instance, Halanaeroarchaeum sulfurireducens M27-SA2 is a sulfur-reducing and acetate-oxidizing haloarcheon isolated from the Medee basin [67]. Moreover, several novel strains have been isolated from Red Sea DHABs, such as Halorhabdus tiamatea (a non-pigmented, fermenting member of the Halobacteriaceae [68]) and Haloplasma contractile (a highly unusual contractile bacterium belonging to the Haloplasmatales order, which can grow under 0.2–3.1 M NaCl conditions [69]. Two other strains of a novel species, Marinobacter salsuginis SD-14BT and SD-14C, have also been isolated from the halocline of the Shaban Deep [70].
In general, prokaryotic diversity and activity appear to be less marked in sediments under brines than in deep-sea control sediments [55]. This is likely due to the cumulative physico-chemical stressors that greatly limit the survival of microorganisms which could be better adapted to the extreme DHAB chemocline [27,55,71]. Proteobacteria, Actinobacteria, Deferribacteres, and Euryarchaeota have been found in sediments underlying either Discovery Deep or Atlantis II [72]. In the sediments of L’Atalante, OTUs belonging to the Pseudoalteromonas, Halomonas, and Pseudomonas genera have been observed to be the most represented within the abundant Gammaproteobacteria class, suggesting a mixed assemblage of halophilic and halotolerant microorganisms [55]. In addition, metatranscriptomic analyses have revealed that, in the sediments underlying the Urania basin, most transcripts are affiliated with rRNAs of the genera Pseudomonas, Rhodobacter, and Clostridium, and with sequences associated with mitomycin antibiotics typically produced by Streptomyces [71]. Prokaryotes inhabiting DHAB sediments are killed by viruses, which may represent the main mechanism of top-down control of prokaryotic dynamics in these ecosystems [42]. Since viruses are found to be well-preserved in DHAB sediments, they can shape prokaryotic assemblages [41]. Based on this information, it is possible to hypothesize that prokaryotes of DHABs can produce specific molecules against viral infections.

3. Biotechnological Potential of Prokaryotes Inhabiting DHABs

Generally, marine microorganisms represent an untapped source for the discovery and development of new biomolecules due to their rich biodiversity and genetic capacity to produce unique metabolites [73,74,75]. In this regard, it is well documented that many taxonomically novel marine species are promising sources of new bioactive compounds with noteworthy pharmaceutical activities, which can become sources of novel therapeutic agents [52,76]. In particular, marine extreme environments, like deep-sea and polar ecosystems or DHABs, have been revealed to be a rich source of secondary metabolites with novel structures and outstanding biological activities [28,52,77].
Due to the limited accessibility and remoteness of such extreme ecosystems and the need for sophisticated instruments for exploring and investigating them, they are still largely understudied and underexploited in comparison with terrestrial ecosystems.
Over the last few years, the advancement of technologies for deep-sea exploration [78] and “-omics” (e.g., environmental shotgun sequencing and metatranscriptomics) for the analysis of environmental strains of prokaryotes has revolutionized bioprospecting in extreme environments, thus increasing our knowledge of the genetic potential of microbial communities for the discovery of enzymes with a commercial value [79,80]. In addition, functional screening of extremophile metagenomes could represent a valuable approach to identify novel antibacterial and anticancer agents. In this regard, bioinformatic tools like the metabolite analysis shell (antiSMASH) have recently been used to detect from metagenomic samples collected from the Atlantis II, Discovery, and Kebrit DHABs promising specialized metabolism gene clusters (SMGCs) coding for products with reported antibacterial and anticancer effects, namely terpenes, peptides, polyketides, and phosphonates [81]. Two clones belonging to these libraries which exhibited antibacterial effects were screened by high-throughput sequencing (NGS) and bioinformatic analyses along with cytotoxicity assay (MTT) testing of the whole cell lysates against different cancer cell lines (MCF‑7, U2OS, and 1BR-hTERT) [82]. Although culture-independent approaches have radically changed microbial bioprospecting in extreme environments, the development of biotechnological applications must be accompanied by the corresponding study of pure cultures. In this regard, despite the great biodiversity-highlighted trough metagenomics in DHABs, so far less than 100 bacterial strains (Figure 3, Table S1) have been isolated and cultured for testing their extracts in few biotechnological applications [66,83,84,85]. Bioinformatic analyses on the phylogeny of the 16s rDNA sequences of those cultured strains (carried out by aligning them on the SILVA database v132 on the ACT server [86]) showed that most of these prokaryotes are affiliated with Gammaproteobacteria and Bacilli; in addition, several sequences within the same database were found to be phylogenetically related to the cultured strains (Table S2), further suggesting that more prokaryotic strains with adaptations to polyextreme ecosystems with biotechnological potential might be found within the same clades of already-cultured strains. Novel sampling and cultivation methods should be developed as alternatives to overcome culture limitations, especially in extreme environments [87].
Since the beginning of the new millennium, a number of studies have indicated the beneficial roles of extremophilic marine prokaryotes, which are a relevant but still underexplored source of bioactive molecules of commercial significance [74,88,89]. Extremophiles undoubtedly show unique capabilities and adaptations which allow them to thrive in systems characterized by harsh environmental conditions [90]. In fact, polyextremophilic microorganisms utilize alternative metabolic pathways and adaptive mechanisms which have important applications in industrial and environmental fields [50]. Since these microorganisms live in a biologically competitive environment for space and nutrients, they have developed mechanisms of defence against competitors and predators for their own survival, synthesising secondary metabolites of great value in pharmaceutical and biotechnological applications [91,92]. The advances in genome sequencing of extremophilic microorganisms have allowed us to provide a comprehensive understanding of their applications [93,94,95]. Moreover, microbes with large genomes, usually inhabiting complex harsh environments, can produce a vast array of secondary metabolites [96,97].

3.1. DHABs as a Hidden Treasure for Biodiscovery of Pharmaceuticals

Over the last 50 years, the development of new multidrug-resistant pathogens, along with the consequent increase in infectious diseases, has become an important issue for human wellness [98]. Furthermore, anticancer chemotherapeutic resistance is recently becoming a biomedical challenge, arising either intrinsically or extrinsically, after therapy [99]. Thus, the need for the discovery and development of novel antimicrobial and chemotherapeutic drugs with new modes of action is nowadays becoming of fundamental importance [100,101]. Since most of the antibiotics currently available on the market have been extracted from terrestrial organisms or derived semisynthetically from fermentation products, the isolation of microorganisms from marine habitats represent an interesting possibility which can lead to the discovery of novel structures with antibiotic activity [102,103].
As such, the prokaryotic genera identified in DHABs isolated from different marine environmental sources, including extreme environments, represent an authentic treasure of many bioactive compounds useful for biomedical applications (Table 2).
For instance, in Mediterranean DHABs transcripts related to Streptomyces have been identified, thus representing an important source of bioactive natural products with clinical or pharmaceutical applications [71,155]. Additionally, Pseudoalteromonas flavipulchra recently isolated from the Nereus halocline shows great antimicrobial activity which is associated with the different metabolites and/or enzymes that this species can produce [84,156,157].
An attractive example of the potential of extremophiles in the biomedical field has been provided by Halobacteroides lacunaris TB21, which was isolated from Thetis basin [118]. This polyextremophile organism produce a lipopolysaccharide (LPS) analog which can bind to the TLR4/MD-2 complex in HEK 293 hTLR4 cells, exerting an immunostimulant activity [118]. Additionally, Pseudoalteromonas carrageenovora isolated from Erba basin sediments is able to produce an LPS whose function is still undescribed [84,122]. This halophilic bacterium can also produce low molecular weight products from carrageenans, which have been reported to hold protective effects against the human immunodeficiency virus, the yellow fever virus, the herpes simplex viruses, the vaccine virus, and the pig fever virus [158]. Another interesting species isolated from the Nereus brine-pool–sea-water interface is Bacillus halodurans, which was engineered for the production of the anti-viral therapeutic peptide Enfuvirtide, marketed by Roche under the trademark Fuzeon®, which has given rise to possibilities for pharmaceutical applications [84,110]. In addition, Zunongwangia profunda, inhabiting the same interface, produces exopolysaccharides (EPS) with antioxidant properties [154].
Innovative research was carried out for screening the bioactivity of molecules extracted from the Atlantis II, Discovery, Kebrit, Nereus, and Erba DHABs [83,85]. Extracts from 36 isolates were tested on three different human cancer cell lines: HeLa, MCF-7, and DU145 [83,85]. In particular, many extracts from Halomonas strains have been found to induce apoptotic and cytotoxic effects. For example, Halomonas meridiana collected from Nereus halocline has been observed to prompt apoptosis of MCF-7 cells [85]. Recently, it was shown that the extract of Pseudoalteromonas mariniglutinosa collected from Erba and Nereus haloclines also inhibited the growth of MCF-7 cells [84]. Halomonas species can produce EPS which have been shown to have pro-apoptotic activity towards human T-leukemia cells and breast cancer MCF-7 cells [159,160]. Other bioactive extracts derived from Chromohalobacter salexigens, Chromohalobacter israelensis, Halomonas meridiana, and Idiomarina loihiensis have been found to be able to induce more than 70% mortality in HeLa cancer cells through different caspase-mediated apoptotic pathways [83].
Intriguingly, three extracts belonging to the genus Salinivibrio have been found to specifically blocked the growth of fibrosarcoma cells (HT-1080), opening up interesting perspectives for the discovery of new bioactive compounds produced by this genus [84]. The extracts of Halomonas hamiltonii and Alcanivorax dieselolei have been observed to inhibit the proliferation of BT20 cells, whereas the Alteromonas macleodii extracts collected from Nereus and Erba halocline were found to inhibit the cell growth of HCT [84]. Alteromonas species are also well known for producing dithiolopyrrolone, a potent antibiotic approved by the Food and Drug Administration and commercialized as Bactroban® (GlaxoSmithKline) [104]. These studies emphasize the wide diversity of brine pool microorganisms capable of producing bioactive molecules, highlighting the incredible potential of DHABs as a source of novel molecules exploitable in the pharmacological industry.

3.2. DHABs as a Reservoir of Polyextreme Enzymes

Today’s society is moving toward “white” (i.e., industrial) biotechnology, which is growing for its efficiency from environmental and commercial points of view [161]. For example, natural enzyme catalysis has been utilized for application in a broader range of industrial settings, representing a valuable alternative to its chemical catalysts [162,163,164]. It is expected that 40% of the industrial application of chemical reactions that require organic solvents harmful to the environment will be replaced by enzymatic catalysis by 2030 [165]. The continuous demand for natural new enzymes that are biocompatible and non-toxic and which have high activity over a wide range of conditions, including temperature, salinity, pH, and metal concentrations, has been scaled up within pharmaceutical, food, and beverage industrial processes [166]. Hence, extremophilic microorganisms represent important sources of stable and valuable enzymes which are used as biocatalysts in industrial and biotechnological processes [53]. Enzymes from these organisms, which are called “extremozymes” due to their special features, can catalyze chemical reactions under conditions which inhibit or denature the non-extreme forms [167], including high salinity, acidic or basic pH, and high temperatures [168]. Thus, through the use of genetic engineering and/or by bioprospecting of extreme environments it is possible to discover and develop new extremozymes that can be suitable for many industrial processes [164]. Extremophilic bacteria and archaea produce enzymes which can be employed in industrial reactions using either directly living organisms or purified molecules, expanding the ranges of optimal enzyme performance and thus enabling biocatalysis under the enzymatically unfavourable conditions found in industrial processes [169]. Hence, the peculiar characteristics of extremophiles belonging to prokaryotic domains living in DHABs can represent a new source for exploitable enzymes for their capacity to operate under extreme conditions [170,171]. In fact, many of these molecules (e.g., aldehyde dehydrogenase, proteases, cellulases, esterases, ferredoxin oxidoreductase, agarase, amylases, κ-Carragenases, ketoreductases, and cyclodextrin glycosyltransferase) have been commercialized and have applications in different biotechnological areas with considerable benefits for many kinds of industries (Table 3). In particular, they are currently being employed in “red” biotechnology (i.e., biotechnology applied to pharmaceutical and medical fields). Other enzymes such as cellulase, chitinase, esterase, mercuric reductase, and β-glucosidases are exploited in “grey” (i.e., environmental) biotechnology while lipase is used in “blue” biotechnology, being applied to aquatic organisms and β-glucosidases and xylanase in biofuel production.
For instance, the production of novel thermoactive and alkali-tolerant α-amylases has been documented for many prokaryotic species such as Pontibacillus chungwhensis, Halomonas meridiana, and Zunongwangia profunda isolated taxa from DHABs. This group of enzymes has a very wide spectrum of industrial application, including in the sugar production, animal nutrition, baking, brewing, and distilling industries, in the production of digestive aids, in the pharmaceutical industries, and in the production of biofuel [239]. Amylases are consistently the most important among the enzymes of industrial interest and are forecasted to reach US$ 6.2 billion by 2020 [240]. For this reason, there is noteworthy attention paid to extremophilic α-amylases that have activity and stability characteristics suitable for the harsh conditions, including extreme salinity (2–4 M NaCl) and elevated temperature (80°C), demanded by industrial activities [241].
Interestingly, nitrilases have also been identified in DHABs, and are employed as commercial biocatalysts for the synthesis of plastics, paints, and fibers in the chemical industries and are also employed in the pharmaceutical industries for the manufacturing of (S)-ibuprofen, a widely used non-steroidal anti-inflammatory drug [242]. Moreover, nitrilases can detoxify cyanide present within wastes and degrade herbicides, representing an enzyme of extreme importance in bioremediation [243]. Biotransformation using native organisms as catalysts tends to be insufficient because the amount of nitrilases present as total cellular proteins is very low, and the reaction rate is slow and unstable [244]. Thus, the nitrilase recently identified by metagenomes mining in the Atlantis II DHAB could represent a valuable alternative not only for its thermal stability and tolerance to heavy metals compared to closely related nitrilases but also for the great number of microorganisms which could possess and produce these enzymes [211]. Another example of utilising sequence-based and activity-based metagenomics in mining for potential industrial biocatalysts is the esterase EstATII collected from the Atlantis II basin in the Red Sea, which displays a combination of extremophilic properties [197]. This enzyme is thermophilic (optimum temperature 65 °C) and halotolerant (for up to 4.5M NaCl) and maintains significant activity in the presence of a wide variety of toxic heavy metals, making it a potentially useful biocatalyst [197]. In agreement with this study, O.16 esterase was identified in the Urania basin which shoswed remarkable polyextremophilic properties (i.e., 180× enhanced activity at 2 to 4 M NaCl and functioning at 40 MPa [198]). This enzyme also displayed increased activity when dissolved in 70% ethanol or n-propanol and extraordinarily high enantioselectivity in hydrolysis and transesterification of compounds important in the pharmaceutical, cosmetic, and food industries [198,245]. Thus, DHABs seem to be a suitable habitat for mining esterases which are potentially useful for industrial biotransformation, considering the great size of the lipolytic enzyme market, which is valued at the billion-dollar mark in the world’s market [246].
A study conducted on bacterial strains isolated from haloclines of Urania, Bannock, Discovery, and L’Atalante showed that Bacillus horneckiae gave highly stereoselective reduction for racemic propyl ester of anti-2- oxotricyclo[2.2.1.0]heptan-7-carboxylic acid (R,S)-1, a key intermediate of the synthesis of D-cloprostenol (chemical analog of prostaglandin [66]). Another isolate of Halomonas aquamarina was found to enantioselectively hydrolyze this molecule, indicating the potential of DHAB extremophile microbiome and marine-derived esterases and ketoreductases in stereoselective biocatalysis [66]. The same authors also isolated Bacillus lehensis from Discovery DHABs which harness an alkali-tolerant cyclodextrin glycosyltransferase and are able to produce non-toxic products for the pharmaceutical, cosmetic, and food industries.
Overall, DHABs seem to be a suitable habitat for mining novel biocatalyst enzymes which are potentially useful for industrial biotransformations, encouraging further scientific challenges and research for fully realising the potential of DHAB extremozymes.

3.3. DHAB-Derived Prokaryotes: Promising Candidates for Enhanced Bioremediation of Oil Hydrocarbons

Petroleum hydrocarbons are among the most widespread pollutants on our planet and are becoming a severe problem because of their causing harmful damage to the environment and human health [247,248]. Oil pollution can occur in the environment following either catastrophic accidents (shipping disasters or pipeline failures) or natural oil seepages and biota [249]. Such contaminants can exert carcinogenic, neurotoxic, and mutagenic effects when organisms are exposed to them, significantly impacting the environment [250,251,252]. For these reasons, many innovative technologies have been developed for the clean-up of oil-polluted areas [247]. One of the most reliable of these is certainly bioremediation, which exploits the metabolic capabilities of microorganisms to break down recalcitrant hydrocarbons into harmless by-products, thus minimising the impact on the environment [253]. It is a more environmentally friendly alternative when compared with classical remediation techniques, which allow the reduction from the environment of a vast array of pollutants [254].
Most petroleum hydrocarbons encountered in the environment can be degraded or metabolized by indigenous bacteria which have developed specific pathways for sustaining their energetic and carbon requirements for living and blooming in the presence of these contaminants [255,256]. Indeed, many studies have focused their attention on hydrocarbon-degrading bacteria in oil-rich environments, including oil spill areas and oil reservoirs [257], and have demonstrated that their abundance is closely related to the respective types of petroleum hydrocarbons and surrounding environmental factors [258,259,260,261]. Despite this, many other normal and extreme microorganisms have been isolated and employed as biodegraders for dealing with petroleum hydrocarbons, representing a promising biotechnological alternative for achieving oil-hydrocarbon degradation [9,262]. Because of the particular physiological characteristics of microorganisms isolated from extreme environments, including DHABs, prokaryotes can be employed for enhanced bioremediation of oil hydrocarbons, especially in hypersaline environments [263,264]. For instance, members of the genera Alcanivorax and Marinobacter, which have been isolated respectively from Erba halocline and Shaban Deep [70,84], are essential marine hydrocarbonoclastic bacteria present in the active phase of oil spills, playing a significant role in the natural remediation of oil-polluted marine environments all over the world [265,266,267]. Their number increases very quickly after oil spills, although it declines only a few weeks later (see [268] as well as references therein). The outstanding bioremediation capacity of A. dieselolei has also been supported by sequencing the genome of the strain KS-293 isolated from surface seawater [269]. Its genome consistently contains multiple genes and enzymes involved in pathways associated with hydrocarbon degradation (linear and branched alkanes) and shows high similarity with A. dieselolei strain B5 [270,271,272]. This strain has been observed to preserve cell integrity under pressures of up to 10 MPa cultured with n-dodecane as a sole carbon source, downregulating 95% of its genes [273,274]. Additionally, Sass et al., 2008 demonstrated that the strain DS-1, closely related to Bacillus aquimaris, isolated from the Discovery DHAB, could grow with n-alkanes (n-dodecane and n-hexadecane) in the presence of 12–20% NaCl [275]. Furthermore, Salinisphaera shabanensis and Marinobacter salsuginis have been isolated from the Shaban Deep, displaying a high capacity to assimilate aliphatic hydrocarbons [70,276]. S. shabanensis can be cultured at a wide range of salinity and temperatures (0.2–4.8 M NaCl and 5–42 °C), on a vast array of substrates including n-alkanes (dodecane). M. salsuginis is a heterotrophic, facultative anaerobic bacterium capable of fermentation and nitrate reduction [70].
Moreover, other bacteria belonging to the genus Marinobacteria and isolated from seawater and Nereus halocline have shown to be efficient for bioremediation purposes for degrading hydrocarbons, including polycyclic aromatic hydrocarbons (PAHs), as revealed by the complete genome sequence of Marinobacter flavimaris SW-145 [277,278,279,280]. Other strains belonging to the genera Vibrio, Pseudomonas, Arthrobacter Pseudoalteromonas, Idiomarina, Halomonas, and Thalassospira identified in different DHABs have been collected and cultured from marine sediments and are able to grow on PAHs including naphthalene, dibenzothiophene, pyrene, and phenanthrene [21,39,267,281,282].
Even though many oil hydrocarbons can be easily degraded in low salinity marine habitats [267], very little is known about their fate in moderate and in hypersaline environments where microbial activity is enormously inhibited [263,283]. To this purpose, some archaea belonging to the class of Halobacteria identified in many DHABs hold great promise and have considerable potential to bioremediate hydrocarbons in high salty environments such as nearshore oil production sites, salt marshes, sabkhas, and other coastal flats, including industrial wastewaters [264,284].
Haloarchaea of the class Halobacteria identified in many DHABs located both in the Mediterranean and the Red Sea [17,44,56,189] can produce PAH-degrading enzymes, which may be exploited to remove aromatic hydrocarbons from the polluted environments safely [285]. In the hypersaline coastal areas of Kuwait, hydrocarbonoclastic haloarchaea, together with M. flavimaris, a diazotrophic strain able to grow under 1 M–3.5 M NaCl conditions, have effectively contributed to oil bioremediation [286].
Although more archaeal strains have been isolated, our information on the physiological, biochemical, and genomic basis of hydrocarbon degradation by members of the Halobacteria is still extremely scant [263,287]. Such information is crucial for designing novel and more efficient technologies employing haloarchaea for the remediation of contaminated high salinity environments.

4. Conclusions and Future Directions

Polyextremophilic bacteria and archaea are an extraordinary reservoir of novel enzymes and bioactive molecules which can provide important benefits for different biotechnological applications, ranging from medicine to environmental fields. So far, studies on DHABs are limited, and we urgently need to expand data on microbial diversity and ecology of these extreme ecosystems. The uniqueness of these habitats is able to select for highly specialized organisms which show extreme adaptions at morphological, physiological, biochemical, and genetic levels, hinting at a bright future for “blue” biotechnology. In this extensive literature review we have observed that polyextremophiles maintain several high metabolic similarities to other non-extreme prokaryotes. This important issue, which remains to be further investigated, could open new research perspectives on the production of the biomolecules’ portfolio of marine microorganisms. Such explorations are expected to provide huge rewards not only in terms of the impact on existing industries for the discovery of new products with beneficial or useful properties, but also in the “blue” economy. This scenario is also perfectly framed within the Sustainable Development Goals of the United Nations, which aims to identify actual solutions for disease outbreaks, climate change, and environmental degradation in order to have safer, cleaner, and more efficient industrial manufacturing processes in order to improve human health and wellbeing from a sustainable development perspective.
Nowadays, “blue” biotechnologies are taking advantage of increasing numbers of “-omics” tools and high-throughput screenings for unveiling the chemical diversity of the extreme environments present in the oceans. These tools are facilitating the identification of prokaryotic metabolic adaptations, which can lead to the production of novel molecules and thus can be exploited for the development of new biotechnologies. To date, novel uncultured species identified in DHABs of the genera Streptomyces, Pseuodalteromonas, and Bacillus seem to hold great potential in producing new bioactive molecules. Among the culturable species identified in DHABs, Chromohalobacter israelensis, Zunongwangia profunda, Marinobacter flavimaris, Alcanivorax dieselolei, Halomonas meridiana, Alteromonas macleodii, and Bacillus halodurans are promising species for biotechnological applications. Further innovative technologies and studies applied to DHABs will be essential to carry out in-depth investigations and to disentangle microbial assemblages, functions, and metabolites of biotechnological interest from these peculiar systems. Thus, the actual development of DHAB-derived biotechnologies will depend on technological and methodological advancements and the ability of scientists to promote research projects for the study of these ecosystems.

Supplementary Materials

The following information is available online at https://www.mdpi.com/1660-3397/18/2/91/s1, Table S1: Prokaryotic strains isolated from DHABs, Table S2: Prokaryotic strains isolated from DHABs and their closest relatives obtained from phylogenetic analyses.

Author Contributions

C.C. conceived the study. S.V. conducted literature analysis and M.T. performed bioinformatic analyses on the available data. C.C., S.V., and M.T. contributed to data interpretation. S.V., C.C., and M.T. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by University Scientific Research, the Italian Ministry for Education, University, and Research (MIUR).

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Danovaro, R.; Corinaldesi, C.; Dell’Anno, A.; Snelgrove, P.V.R. The deep-sea under global change. Curr. Biol. 2017, 27, R461–R465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Danovaro, R.; Snelgrove, P.V.R.; Tyler, P. Challenging the paradigms of deep-sea ecology. Trends Ecol. Evol. 2014, 29, 465–475. [Google Scholar] [CrossRef] [PubMed]
  3. Herring, P.J. The biology of the deep ocean; Oxford University Press: New York, NY, USA, 2001; pp. 1–25. [Google Scholar]
  4. Bartlett, D.H. Microbial Life in the Trenches. Mar. Technol. Soc. J. 2009, 43, 128–131. [Google Scholar] [CrossRef]
  5. Jørgensen, B.B.; Boetius, A. Feast and famine - Microbial life in the deep-sea bed. Nat. Rev. Microbiol. 2007, 5, 770–781. [Google Scholar] [CrossRef]
  6. Ramirez-Llodra, E.; Brandt, A.; Danovaro, R.; De Mol, B.; Escobar, E.; German, C.R.; Levin, L.A.; Martinez Arbizu, P.; Menot, L.; Buhl-Mortensen, P.; et al. Deep, diverse and definitely different: Unique attributes of the world’s largest ecosystem. Biogeosciences 2010, 7, 2851–2899. [Google Scholar] [CrossRef] [Green Version]
  7. Merino, N.; Aronson, H.S.; Bojanova, D.P.; Feyhl-Buska, J.; Wong, M.L.; Zhang, S.; Giovannelli, D. Living at the extremes: Extremophiles and the limits of life in a planetary context. Front. Microbiol. 2019, 10, 1–25. [Google Scholar] [CrossRef] [Green Version]
  8. Merlino, G.; Barozzi, A.; Michoud, G.; Ngugi, D.K.; Daffonchio, D. Microbial ecology of deep-sea hypersaline anoxic basins. FEMS Microbiol. Ecol. 2018, 94, 1–15. [Google Scholar] [CrossRef]
  9. Barone, G.; Varrella, S.; Tangherlini, M.; Rastelli, E.; Dell’Anno, A.; Danovaro, R.; Corinaldesi, C. Marine Fungi: Biotechnological Perspectives from Deep-Hypersaline Anoxic Basins. Diversity 2019, 11, 113. [Google Scholar] [CrossRef] [Green Version]
  10. Jongsma, D.; Fortuin, A.R.; Huson, W.; Troelstra, S.R.; Klaver, G.T.; Peters, J.M.; Van Harten, D.; De Lange, G.J.; Ten Haven, L. Discovery of an anoxic basin within the strabo trench, eastern mediterranean. Nature 1983, 305, 795–797. [Google Scholar] [CrossRef]
  11. Camerlenghi, A. Anoxic basins of the eastern Mediterranean: Geological framework. Mar. Chem. 1990, 31, 1–19. [Google Scholar] [CrossRef]
  12. Hartmann, M.; Scholten, J.C.; Stoffers, P.; Wehner, F. Hydrographic structure of brine-filled deeps in the Red Sea - new results from the Shaban, Kebrit, Atlantis II, and Discovery Deep. Mar. Geol. 1998, 144, 311–330. [Google Scholar] [CrossRef]
  13. Pautot, G.; Guennoc, P.; Coutelle, A.; Lyberis, N. Discovery of a large brine deep in the northern Red Sea. Nature 1984, 310, 133–136. [Google Scholar] [CrossRef]
  14. Backer, H.; Schoell, M. New Deeps with Brines and Metalliferous Sediments in the Red Sea. Nat. Phys. Sci. 1972, 240, 153–158. [Google Scholar] [CrossRef]
  15. Shokes, R.F.; Trabant, P.K.; Presley, B.J.; Reid, D.F. Anoxic, hypersaline basin in the northern Gulf of Mexico. Science 1977, 4297, 1443–1446. [Google Scholar] [CrossRef] [PubMed]
  16. La Cono, V.; Smedile, F.; Bortoluzzi, G.; Arcadi, E.; Maimone, G.; Messina, E.; Borghini, M.; Oliveri, E.; Mazzola, S.; L’Haridon, S.; et al. Unveiling microbial life in new deep-sea hypersaline Lake Thetis. Part I: Prokaryotes and environmental settings. Environ. Microbiol. 2011, 13, 2250–2268. [Google Scholar] [CrossRef] [PubMed]
  17. Yakimov, M.M.; La Cono, V.; Spada, G.L.; Bortoluzzi, G.; Messina, E.; Smedile, F.; Arcadi, E.; Borghini, M.; Ferrer, M.; Schmitt-Kopplin, P.; et al. Microbial community of the deep-sea brine Lake Kryos seawater-brine interface is active below the chaotropicity limit of life as revealed by recovery of mRNA. Environ. Microbiol. 2015, 17, 364–382. [Google Scholar] [CrossRef] [Green Version]
  18. La Cono, V.; Bortoluzzi, G.; Messina, E.; La Spada, G.; Smedile, F.; Giuliano, L.; Borghini, M.; Stumpp, C.; Schmitt-Kopplin, P.; Harir, M.; et al. The discovery of Lake Hephaestus, the youngest athalassohaline deep-sea formation on Earth. Sci. Rep. 2019, 9, 1679. [Google Scholar] [CrossRef]
  19. Mapelli, F.; Barozzi, A.; Michoud, G.; Merlino, G.; Crotti, E.; Borin, S.; Daffonchio, D. An updated view of the microbial diversity in deep hypersaline anoxic basins. In Adaption of Microbial Life to Environmental Extremes; Stan-Lotter, H., Fendrihan, S., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 23–40. ISBN 9783319483276. [Google Scholar]
  20. Eder, W.; Ludwig, W.; Huber, R. Novel 16S rRNA gene sequences retrieved from highly saline brine sediments of Kebrit Deep, Red Sea. Arch. Microbiol. 1999, 172, 213–218. [Google Scholar] [CrossRef]
  21. Daffonchio, D.; Borin, S.; Brusa, T.; Brusetti, L.; van der Wielen, P.W.J.J.; Bolhuis, H.; Yakimov, M.M.; D’Auria, G.; Giuliano, L.; Marty, D.; et al. Stratified prokaryote network in the oxic–anoxic transition of a deep-sea halocline. Nature 2006, 440, 203–207. [Google Scholar] [CrossRef]
  22. Yakimov, M.M.; La Cono, V.; Denaro, R.; D’Auria, G.; Decembrini, F.; Timmis, K.N.; Golyshin, P.N.; Giuliano, L. Primary producing prokaryotic communities of brine, interface and seawater above the halocline of deep anoxic lake L’Atalante, Eastern Mediterranean Sea. ISME J. 2007, 1, 743–755. [Google Scholar] [CrossRef] [Green Version]
  23. Borin, S.; Brusetti, L.; Mapelli, F.; D’Auria, G.; Brusa, T.; Marzorati, M.; Rizzi, A.; Yakimov, M.; Marty, D.; De Lange, G.J.; et al. Sulfur cycling and methanogenesis primarily drive microbial colonization of the highly sulfidic Urania deep hypersaline basin. Proc. Natl. Acad. Sci. USA 2009, 106, 9151–9156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Joye, S.B.; MacDonald, I.R.; Montoya, J.P.; Peccini, M. Geophysical and geochemical signatures of Gulf of Mexico seafloor brines. Biogeosciences 2005, 2, 295–309. [Google Scholar] [CrossRef] [Green Version]
  25. Cita, M.B. Exhumation of Messinian evaporites in the deep-sea and creation of deep anoxic brine-filled collapsed basins. Sediment. Geol. 2006, 188–189, 357–378. [Google Scholar] [CrossRef]
  26. Hsü, K.J.; Cita, M.B.; Ryan, W.B.F. The origin of the Mediterranean evaporites. Initial Reports Deep Sea Drill. Proj. 1973, 13, 1203–1231. [Google Scholar]
  27. van der Wielen, P.W.J.J.; Bolhuis, H.; Borin, S.; Daffonchio, D.; Corselli, C.; Giuliano, L.; D’Auria, G.; de Lange, G.J.; Huebner, A.; Varnavas, S.P.; et al. The enigma of prokaryotic life in deep hypersaline anoxic basins. Science 2005, 307, 121–123. [Google Scholar] [CrossRef]
  28. Barozzi, A.; Mapelli, F.; Michoud, G.; Crotti, E.; Merlino, G.; Molinari, F.; Borin, S.; Daffonchio, D. Microbial Diversity and Biotechnological Potential of Microorganisms Thriving in the Deep-Sea Brine Pools. In Extremophiles From Biology to Biotechnology; Durvasula, R., Rao, D.S., Eds.; CRC Press: Boca Raton, FL, USA; Taylor & Francis, a CRC title, part of the Taylor & Francis imprint, a member of the Taylor & Francis Group, the academic division of T&F Informa plc: Boca Raton, FL, USA, 2018; pp. 19–32. ISBN 9781498774925. [Google Scholar]
  29. Swift, S.A.; Bower, A.S.; Schmitt, R.W. Vertical, horizontal, and temporal changes in temperature in the Atlantis II and Discovery hot brine pools, Red Sea. Deep. Res. Part I Oceanogr. Res. Pap. 2012, 64. [Google Scholar] [CrossRef]
  30. Ngugi, D.K.; Blom, J.; Alam, I.; Rashid, M.; Ba-Alawi, W.; Zhang, G.; Hikmawan, T.; Guan, Y.; Antunes, A.; Siam, R.; et al. Comparative genomics reveals adaptations of a halotolerant thaumarchaeon in the interfaces of brine pools in the Red Sea. ISME J. 2015, 9, 396–411. [Google Scholar] [CrossRef] [Green Version]
  31. Yakimov, M.M.; La Cono, V.; Slepak, V.Z.; La Spada, G.; Arcadi, E.; Messina, E.; Borghini, M.; Monticelli, L.S.; Rojo, D.; Barbas, C.; et al. Microbial life in the Lake Medee, the largest deep-sea salt-saturated formation. Sci. Rep. 2013, 3, 1–9. [Google Scholar] [CrossRef]
  32. Karbe, L. Hot Brines and the Deep Sea Environment. In Red Sea; Edwards, A.J., Head, S.J., Eds.; Elsevier: Oxford, UK, 1987; pp. 70–89. [Google Scholar]
  33. MacDonald, I.R.; Guinasso, N.L.; Reilly, J.F.; Brooks, J.M.; Callender, W.R.; Gabrielle, S.G. Gulf of Mexico hydrocarbon seep communities: VI. Patterns in community structure and habitat. Geo-Marine Lett. 1990, 10, 244–252. [Google Scholar] [CrossRef]
  34. De Lange, G.J.; Middelburg, J.J.; Van der Weijden, C.H.; Catalano, G.; Luther, G.W.; Hydes, D.J.; Woittiez, J.R.W.; Klinkhammer, G.P. Composition of anoxic hypersaline brines in the Tyro and Bannock Basins, eastern Mediterranean. Mar. Chem. 1990, 31, 63–88. [Google Scholar] [CrossRef]
  35. Van Cappellen, P.; Viollier, E.; Roychoudhury, A.; Clark, L.; Ingall, E.; Lowe, K.; Dichristina, T. Biogeochemical cycles of manganese and iron at the oxic-anoxic transition of a stratified marine basin (Orca Basin, Gulf of Mexico). Environ. Sci. Technol. 1998, 32, 2931–2939. [Google Scholar] [CrossRef]
  36. Schmidt, M.; Al-Farawati, R.; Botz, R. Geochemical Classification of Brine-Filled Red Sea Deeps. In The Red Sea; RasulIan, N.M.A., Stewart, C.F., Eds.; Springer Berlin Heidelberg: Cham, Switzerland, 2015; pp. 219–233. [Google Scholar]
  37. Stock, A.; Filker, S.; Yakimov, M.; Stoeck, T. Deep Hypersaline Anoxic Basins as Model Systems for Environmental Selection of Microbial Plankton. In Polyextremophiles Life Under Multiple Forms of Stress Life Under Multiple Forms of Stress; Springer Netherlands: Dordrecht, The Netherlands, 2013; pp. 499–515. [Google Scholar]
  38. Eder, W.; Schmidt, M.; Koch, M.; Garbe-Schönberg, D.; Huber, R. Prokaryotic phylogenetic diversity and corresponding geochemical data of the brine-seawater interface of the Shaban Deep, Red Sea. Environ. Microbiol. 2002, 4, 758–763. [Google Scholar] [CrossRef] [PubMed]
  39. Sass, A.M.; Sass, H.; Coolen, M.J.L.; Cypionka, H.; Overmann, J. Microbial Communities in the Chemocline of a Hypersaline Deep-Sea Basin (Urania Basin, Mediterranean Sea). Appl. Environ. Microbiol. 2001, 67, 5392–5402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. van der Wielen, P.W.J.J.; Heijs, S.K. Sulfate-reducing prokaryotic communities in two deep hypersaline anoxic basins in the Eastern Mediterranean deep sea. Environ. Microbiol. 2007, 9, 1335–1340. [Google Scholar] [CrossRef]
  41. Danovaro, R.; Corinaldesi, C.; Dell’Anno, A.; Fabiano, M.; Corselli, C. Viruses, prokaryotes and DNA in the sediments of a deep-hypersaline anoxic basin (DHAB) of the Mediterranean Sea. Environ. Microbiol. 2005, 7, 586–592. [Google Scholar] [CrossRef]
  42. Corinaldesi, C.; Tangherlini, M.; Luna, G.M.; Dell’Anno, A. Extracellular DNA can preserve the genetic signatures of present and past viral infection events in deep hypersaline anoxic basins. Proc. R. Soc. B Biol. Sci. 2014, 281, 1–10. [Google Scholar] [CrossRef]
  43. Danovaro, R.; Gambi, C.; Dell’Anno, A.; Corinaldesi, C.; Pusceddu, A.; Neves, R.C.; Kristensen, R.M. The challenge of proving the existence of metazoan life in permanently anoxic deep-sea sediments. BMC Biol. 2016, 14, 1–7. [Google Scholar] [CrossRef] [Green Version]
  44. Pachiadaki, M.G.; Yakimov, M.M.; Lacono, V.; Leadbetter, E.; Edgcomb, V. Unveiling microbial activities along the halocline of Thetis, a deep-sea hypersaline anoxic basin. ISME J. 2014, 8, 2478–2489. [Google Scholar] [CrossRef]
  45. Edgcomb, V.P.; Orsi, W.; Breiner, H.W.; Stock, A.; Filker, S.; Yakimov, M.M.; Stoeck, T. Novel active kinetoplastids associated with hypersaline anoxic basins in the Eastern Mediterranean deep-sea. Deep. Res. Part I Oceanogr. Res. Pap. 2011, 58, 1040–1048. [Google Scholar] [CrossRef]
  46. Alexander, E.; Stock, A.; Breiner, H.W.; Behnke, A.; Bunge, J.; Yakimov, M.M.; Stoeck, T. Microbial eukaryotes in the hypersaline anoxic L’Atalante deep-sea basin. Environ. Microbiol. 2009, 11, 360–381. [Google Scholar] [CrossRef]
  47. Danovaro, R.; Dell’Anno, A.; Pusceddu, A.; Gambi, C.; Heiner, I.; Kristensen, R.M. The first metazoa living in permanently anoxic conditions. BMC Biol. 2010, 8, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Eder, W.; Jahnke, L.L.; Schmidt, M.; Huber, R. Microbial Diversity of the Brine-Seawater Interface of the Kebrit Deep, Red Sea, Studied via 16S rRNA Gene Sequences and Cultivation Methods. Appl. Environ. Microbiol. 2001, 67, 3077–3085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Cavalazzi, B.; Barbieri, R.; Gómez, F.; Capaccioni, B.; Olsson-Francis, K.; Pondrelli, M.; Rossi, A.P.; Hickman-Lewis, K.; Agangi, A.; Gasparotto, G.; et al. The Dallol Geothermal Area, Northern Afar (Ethiopia)—An Exceptional Planetary Field Analog on Earth. Astrobiology 2019, 19, 553–578. [Google Scholar] [CrossRef] [PubMed]
  50. Poli, A.; Finore, I.; Romano, I.; Gioiello, A.; Lama, L.; Nicolaus, B. Microbial Diversity in Extreme Marine Habitats and Their Biomolecules. Microorganisms 2017, 5, 25. [Google Scholar] [CrossRef] [Green Version]
  51. Gunde-Cimerman, N.; Plemenitaš, A.; Oren, A. Strategies of adaptation of microorganisms of the three domains of life to high salt concentrations. FEMS Microbiol. Rev. 2018, 42, 353–375. [Google Scholar] [CrossRef]
  52. Giddings, L.-A.; Newman, D.J. Bioactive Compounds from Extremophiles. In Bioactive Compounds from Extremophiles; Springer: Cham, Switzerland, 2015; pp. 1–44. [Google Scholar]
  53. Coker, J.A. Extremophiles and biotechnology: Current uses and prospects. F1000Research 2016, 5, 396–403. [Google Scholar] [CrossRef]
  54. Sorokin, D.Y.; Kublanov, I.V.; Gavrilov, S.N.; Rojo, D.; Roman, P.; Golyshin, P.N.; Slepak, V.Z.; Smedile, F.; Ferrer, M.; Messina, E.; et al. Elemental sulfur and acetate can support life of a novel strictly anaerobic haloarchaeon. ISME J. 2016, 10, 240–252. [Google Scholar] [CrossRef] [Green Version]
  55. Kormas, K.A.; Pachiadaki, M.G.; Karayanni, H.; Leadbetter, E.R.; Bernhard, J.M.; Edgcomb, V.P. Inter-comparison of the potentially active prokaryotic communities in the halocline sediments of Mediterranean deep-sea hypersaline basins. Extremophiles 2015, 19, 949–960. [Google Scholar] [CrossRef]
  56. Mwirichia, R.; Alam, I.; Rashid, M.; Vinu, M.; Ba-Alawi, W.; Anthony Kamau, A.; Kamanda Ngugi, D.; Goker, M.; Klenk, H.P.; Bajic, V.; et al. Metabolic traits of an uncultured archaeal lineage-MSBL1-from brine pools of the Red Sea. Sci. Rep. 2016, 6, 1–14. [Google Scholar] [CrossRef] [Green Version]
  57. La Cono, V.; Arcadi, E.; Spada, G.; Barreca, D.; Laganà, G.; Bellocco, E.; Catalfamo, M.; Smedile, F.; Messina, E.; Giuliano, L.; et al. A Three-Component Microbial Consortium from Deep-Sea Salt-Saturated Anoxic Lake Thetis Links Anaerobic Glycine Betaine Degradation with Methanogenesis. Microorganisms 2015, 3, 500–517. [Google Scholar] [CrossRef]
  58. Guan, Y.; Hikmawan, T.; Antunes, A.; Ngugi, D.; Stingl, U. Diversity of methanogens and sulfate-reducing bacteria in the interfaces of five deep-sea anoxic brines of the Red Sea. Res. Microbiol. 2015, 166, 688–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Nigro, L.M.; Hyde, A.S.; MacGregor, B.J.; Teske, A. Phylogeography, Salinity Adaptations and Metabolic Potential of the Candidate Division KB1 Bacteria Based on a Partial Single Cell Genome. Front. Microbiol. 2016, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Joye, S.B.; Samarkin, V.A.; Orcutt, B.N.; MacDonald, I.R.; Hinrichs, K.U.; Elvert, M.; Teske, A.P.; Lloyd, K.G.; Lever, M.A.; Montoya, J.P.; et al. Metabolic variability in seafloor brines revealed by carbon and sulphur dynamics. Nat. Geosci. 2009, 2, 349–354. [Google Scholar] [CrossRef]
  61. Stahl, D.A.; de la Torre, J.R. Physiology and Diversity of Ammonia-Oxidizing Archaea. Annu. Rev. Microbiol. 2012, 66, 83–101. [Google Scholar] [CrossRef]
  62. Könneke, M.; Bernhard, A.E.; de la Torre, J.R.; Walker, C.B.; Waterbury, J.B.; Stahl, D.A. Isolation of an autotrophic ammonia-oxidizing marine archaeon. Nature 2005, 437, 543–546. [Google Scholar] [CrossRef]
  63. Oren, A. Molecular ecology of extremely halophilic Archaea and Bacteria. FEMS Microbiol. Ecol. 2002, 39, 1–7. [Google Scholar] [CrossRef]
  64. Oren, A. Life at High Salt Concentrations. In The Prokaryotes; Rosenberg, E., DeLong, E.F., Lory, S., Stackebrandt, E., Thompson, F., Eds.; Springer Berlin Heidelberg: Berlin, Germany, 2013; pp. 421–440. [Google Scholar]
  65. Guan, Y.; Ngugi, D.K.; Vinu, M.; Blom, J.; Alam, I.; Guillot, S.; Ferry, J.G.; Stingl, U. Comparative Genomics of the Genus Methanohalophilus, Including a Newly Isolated Strain From Kebrit Deep in the Red Sea. Front. Microbiol. 2019, 10, 1–11. [Google Scholar] [CrossRef]
  66. De Vitis, V.; Guidi, B.; Contente, M.L.; Granato, T.; Conti, P.; Molinari, F.; Crotti, E.; Mapelli, F.; Borin, S.; Daffonchio, D.; et al. Marine Microorganisms as Source of Stereoselective Esterases and Ketoreductases: Kinetic Resolution of a Prostaglandin Intermediate. Mar. Biotechnol. 2015, 17, 144–152. [Google Scholar] [CrossRef] [Green Version]
  67. Messina, E.; Sorokin, D.Y.; Kublanov, I.V.; Toshchakov, S.; Lopatina, A.; Arcadi, E.; Smedile, F.; La Spada, G.; La Cono, V.; Yakimov, M.M. Complete genome sequence of “Halanaeroarchaeum sulfurireducens” M27-SA2, a sulfur-reducing and acetate-oxidizing haloarchaeon from the deep-sea hypersaline anoxic lake Medee. Stand. Genomic Sci. 2016, 11, 1–15. [Google Scholar] [CrossRef] [Green Version]
  68. Antunes, A.; Taborda, M.; Huber, R.; Moissl, C.; Nobre, M.F.; da Costa, M.S. Halorhabdus tiamatea sp. nov., a non-pigmented extremely halophilic archaeon from a deep-sea hypersaline anoxic basin of the Red Sea, and emended description of the genus Halorhabdus. Int. J. Syst. Evol. Microbiol. 2008, 58, 215–220. [Google Scholar] [CrossRef] [Green Version]
  69. Antunes, A.; Rainey, F.A.; Wanner, G.; Taborda, M.; Pätzold, J.; Nobre, M.F.; da Costa, M.S.; Huber, R. A New Lineage of Halophilic, Wall-Less, Contractile Bacteria from a Brine-Filled Deep of the Red Sea. J. Bacteriol. 2008, 190, 3580–3587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Antunes, A.; Francça, L.; Rainey, F.A.; Huber, R.; Fernanda Nobre, M.; Edwards, K.J.; da Costa, M.S. Marinobacter salsuginis sp. nov., isolated from the brine-seawater interface of the Shaban Deep, Red Sea. Int. J. Syst. Evol. Microbiol. 2007, 57, 1035–1040. [Google Scholar] [CrossRef] [PubMed]
  71. Edgcomb, V.P.; Pachiadaki, M.G.; Mara, P.; Kormas, K.A.; Leadbetter, E.R.; Bernhard, J.M. Gene expression profiling of microbial activities and interactions in sediments under haloclines of E. Mediterranean deep hypersaline anoxic basins. ISME J. 2016, 10, 2643–2657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Siam, R.; Mustafa, G.A.; Sharaf, H.; Moustafa, A.; Ramadan, A.R.; Antunes, A.; Bajic, V.B.; Stingl, U.; Marsis, N.G.R.; Coolen, M.J.L.; et al. Unique Prokaryotic Consortia in Geochemically Distinct Sediments from Red Sea Atlantis II and Discovery Deep Brine Pools. PLoS ONE 2012, 7, e42872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Tortorella, E.; Tedesco, P.; Esposito, F.P.; January, G.G.; Fani, R.; Jaspars, M.; De Pascale, D.; Palma Esposito, F.; January, G.G.; Fani, R.; et al. Antibiotics from deep-sea microorganisms: Current discoveries and perspectives. Mar. Drugs 2018, 16, 355. [Google Scholar] [CrossRef] [Green Version]
  74. Corinaldesi, C.; Barone, G.; Marcellini, F.; Dell’Anno, A.; Danovaro, R. Marine microbial-derived molecules and their potential use in cosmeceutical and cosmetic products. Mar. Drugs 2017, 15, 118. [Google Scholar] [CrossRef]
  75. Lo Giudice, A.; Rizzo, C. Bacteria Associated with Marine Benthic Invertebrates from Polar Environments: Unexplored Frontiers for Biodiscovery? Diversity 2018, 10, 80. [Google Scholar] [CrossRef] [Green Version]
  76. Andryukov, B.; Mikhailov, V.; Besednova, N. The Biotechnological Potential of Secondary Metabolites from Marine Bacteria. J. Mar. Sci. Eng. 2019, 7, 176. [Google Scholar] [CrossRef] [Green Version]
  77. Núñez-Montero, K.; Barrientos, L. Advances in Antarctic Research for Antimicrobial Discovery: A Comprehensive Narrative Review of Bacteria from Antarctic Environments as Potential Sources of Novel Antibiotic Compounds Against Human Pathogens and Microorganisms of Industrial Importance. Antibiotics 2018, 7, 90. [Google Scholar] [CrossRef] [Green Version]
  78. Corinaldesi, C. New perspectives in benthic deep-sea microbial ecology. Front. Mar. Sci. 2015, 2, 1–12. [Google Scholar] [CrossRef] [Green Version]
  79. Kim, S.K. Handbook of Marine biotechnology; Springer-Verlag Berlin Heidelberg: Berlin, Germany, 2015; pp. 307–326. ISBN 9783642539718. [Google Scholar]
  80. Ferrer, M.; Martínez-Martínez, M.; Bargiela, R.; Streit, W.R.; Golyshina, O.V.; Golyshin, P.N. Estimating the success of enzyme bioprospecting through metagenomics: Current status and future trends. Microb. Biotechnol. 2016, 9, 22–34. [Google Scholar] [CrossRef] [PubMed]
  81. Ziko, L.; Adel, M.; Malash, M.N.; Siam, R. Insights into red sea brine pool specialized metabolism gene clusters encoding potential metabolites for biotechnological applications and extremophile survival. Mar. Drugs 2019, 17, 273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ziko, L.; Saqr, A.-H.A.; Ouf, A.; Gimpel, M.; Aziz, R.K.; Neubauer, P.; Siam, R. Antibacterial and anticancer activities of orphan biosynthetic gene clusters from Atlantis II Red Sea brine pool. Microb. Cell Fact. 2019, 18, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Sagar, S.; Esau, L.; Holtermann, K.; Hikmawan, T.; Zhang, G.; Stingl, U.; Bajic, V.B.; Kaur, M. Induction of apoptosis in cancer cell lines by the Red Sea brine pool bacterial extracts. BMC Complement. Altern. Med. 2013, 13, 344. [Google Scholar] [CrossRef] [Green Version]
  84. Esau, L.; Zhang, G.; Sagar, S.; Stingl, U.; Bajic, V.B.; Kaur, M. Mining the deep Red-Sea brine pool microbial community for anticancer therapeutics. BMC Complement. Altern. Med. 2019, 19. [Google Scholar] [CrossRef]
  85. Sagar, S.; Esau, L.; Hikmawan, T.; Antunes, A.; Holtermann, K.; Stingl, U.; Bajic, V.B.; Kaur, M. Cytotoxic and apoptotic evaluations of marine bacteria isolated from brine-seawater interface of the Red Sea. BMC Complement. Altern. Med. 2013, 13, 1–8. [Google Scholar] [CrossRef] [Green Version]
  86. Pruesse, E.; Peplies, J.; Glöckner, F.O. SINA: Accurate high-throughput multiple sequence alignment of ribosomal RNA genes. Bioinformatics 2012, 28, 1823–1829. [Google Scholar] [CrossRef]
  87. Lewis, K.; Epstein, S.; D’Onofrio, A.; Ling, L.L. Uncultured microorganisms as a source of secondary metabolites. J. Antibiot. 2010, 63, 468–476. [Google Scholar] [CrossRef]
  88. Ksouri, R.; Ksouri, W.M.; Jallali, I.; Debez, A.; Magné, C.; Hiroko, I.; Abdelly, C. Medicinal halophytes: Potent source of health promoting biomolecules with medical, nutraceutical and food applications. Crit. Rev. Biotechnol. 2012, 32, 289–326. [Google Scholar] [CrossRef]
  89. Raddadi, N.; Cherif, A.; Daffonchio, D.; Neifar, M.; Fava, F. Biotechnological applications of extremophiles, extremozymes and extremolytes. Appl. Microbiol. Biotechnol. 2015, 99, 7907–7913. [Google Scholar] [CrossRef] [Green Version]
  90. Singh, O.V. Extremophiles; Singh, O.V., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2012; pp. 1–429. ISBN 9781118394144. [Google Scholar]
  91. Rampelotto, P.H.; Trincone, A. Grand Challenges in Marine Biotechnology; Springer International Publishing: Cham, Switzerland, 2016; pp. 1–260. ISBN 978-3-319-69075-9. [Google Scholar]
  92. Babu, P.; Chandel, A.K.; Singh, O.V. Extremophiles and Their Applications in Medical Processes; SpringerBriefs in Microbiology: Cham, Switzerland, 2015; pp. 1–54. ISBN 978-3-319-12807-8. [Google Scholar]
  93. Shin, D.S.; Pratt, A.J.; Tainer, J.A. Archaeal genome guardians give insights into eukaryotic DNA replication and damage response proteins. Archaea 2014, 2014, 206735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Majhi, M.C.; Behera, A.K.; Kulshreshtha, N.M.; Mahmooduzafar, D.; Kumar, R.; Kumar, A. ExtremeDB: A Unified Web Repository of Extremophilic Archaea and Bacteria. PLoS ONE 2013, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Cárdenas, J.P.; Valdés, J.; Quatrini, R.; Duarte, F.; Holmes, D.S. Lessons from the genomes of extremely acidophilic bacteria and archaea with special emphasis on bioleaching microorganisms. Appl. Microbiol. Biotechnol. 2010, 88, 605–620. [Google Scholar] [CrossRef] [PubMed]
  96. Land, M.; Hauser, L.; Jun, S.R.; Nookaew, I.; Leuze, M.R.; Ahn, T.H.; Karpinets, T.; Lund, O.; Kora, G.; Wassenaar, T.; et al. Insights from 20 years of bacterial genome sequencing. Funct. Integr. Genomics 2015, 15, 141–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Baltz, R.H. Molecular beacons to identify gifted microbes for genome mining. J. Antibiot. 2017, 70, 639–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Brown, E.D.; Wright, G.D. Antibacterial drug discovery in the resistance era. Nature 2016, 529, 336–343. [Google Scholar] [CrossRef]
  99. Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef]
  100. Ventola, C.L. The antibiotic resistance crisis: Part 1: Causes and threats. P T 2015, 40, 277–283. [Google Scholar]
  101. Housman, G.; Byler, S.; Heerboth, S.; Lapinska, K.; Longacre, M.; Snyder, N.; Sarkar, S. Drug resistance in cancer: An overview. Cancers 2014, 6, 1769–1792. [Google Scholar] [CrossRef] [Green Version]
  102. Trindade, M.; van Zyl, L.J.; Navarro-Fernández, J.; Elrazak, A.A. Targeted metagenomics as a tool to tap into marine natural product diversity for the discovery and production of drug candidates. Front. Microbiol. 2015, 6, 1–14. [Google Scholar] [CrossRef]
  103. Malve, H. Exploring the ocean for new drug developments: Marine pharmacology. J. Pharm. Bioallied Sci. 2016, 8, 83–91. [Google Scholar] [CrossRef] [PubMed]
  104. Li, B.; Wever, W.J.; Walsh, C.T.; Bowers, A.A. Dithiolopyrrolones: Biosynthesis, synthesis, and activity of a unique class of disulfide-containing antibiotics. Nat. Prod. Rep. 2014, 31, 905–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Imada, C.; Maeda, M.; Hara, S.; Taga, N.; Simidu, U. Purification and characterization of subtilisin inhibitors ‘Marinostatin’ produced by marine Alteromonas sp. J. Appl. Bacteriol. 1986, 60, 469–476. [Google Scholar] [CrossRef]
  106. Gustafson, K.; Roman, M.; Fenical, W. The Macrolactins, a Novel Class of Antiviral and Cytotoxic Macrolides from a Deep-Sea Marine Bacterium. J. Am. Chem. Soc. 1989, 111, 7519–7524. [Google Scholar] [CrossRef]
  107. Nagao, T.; Adachi, K.; Sakai, M.; Nishijima, M.; Sano, H. Novel macrolactins as antibiotic lactones from a marine bacterium. J. Antibiot. 2001, 54, 333–339. [Google Scholar] [CrossRef] [Green Version]
  108. Jaruchoktaweechai, C.; Suwanborirux, K.; Tanasupawatt, S.; Kittakoop, P.; Menasveta, P. New macrolactins from a marine Bacillus sp. Sc026. J. Nat. Prod. 2000, 63, 984–986. [Google Scholar] [CrossRef]
  109. Xue, C.; Tian, L.; Xu, M.; Deng, Z.; Lin, W. A new 24-membered lactone and a new polyene δ-lactone from the marine bacterium Bacillus marinus. J. Antibiot. 2008, 61, 668–674. [Google Scholar] [CrossRef] [Green Version]
  110. Berger, E.; Crampton, M.C.; Nxumalo, N.P.; Louw, M.E. Extracellular secretion of a recombinant therapeutic peptide by Bacillus halodurans utilizing a modified flagellin type III secretion system. Microb. Cell Fact. 2011, 10. [Google Scholar] [CrossRef] [Green Version]
  111. Gerard, J.M.; Haden, P.; Kelly, M.T.; Andersen, R.J. Loloatins A-D, cyclic decapeptide antibiotics produced in culture by a tropical marine bacterium. J. Nat. Prod. 1999, 62, 80–85. [Google Scholar] [CrossRef]
  112. Rahman, H.; Austin, B.; Mitchell, W.J.; Morris, P.C.; Jamieson, D.J.; Adams, D.R.; Spragg, A.M.; Schweizer, M. Novel anti-infective compounds from marine bacteria. Mar. Drugs 2010, 8, 498–518. [Google Scholar] [CrossRef]
  113. Mondol, M.A.M.; Shin, H.J.; Islam, M.T. Diversity of secondary metabolites from marine Bacillus species: Chemistry and biological activity. Mar. Drugs 2013, 11, 2846–2872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhang, H.L.; Hua, H.M.; Pei, Y.H.; Yao, X.S. Three New Cytotoxic Cyclic Acylpeptides from Marine Bacillus sp. Chem. Pharm. Bull. 2004, 52, 1029–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Oku, N.; Adachi, K.; Matsuda, S.; Kasai, H.; Takatsuki, A.; Shizuri, Y. Ariakemicins A and B, novel polyketide-peptide antibiotics from a marine gliding bacterium of the genus Rapidithrix. Org. Lett. 2008, 10, 2481–2484. [Google Scholar] [CrossRef] [PubMed]
  116. Hu, Y.; MacMillan, J.B. Erythrazoles A-B, cytotoxic benzothiazoles from a marine-derived Erythrobacter sp. Org. Lett. 2011, 13, 6580–6583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Hu, Y.; Legako, A.G.; Espindola, A.P.D.M.; MacMillan, J.B. Erythrolic acids A-E, meroterpenoids from a marine-derived Erythrobacter sp. J. Org. Chem. 2012, 77, 3401–3407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Di Lorenzo, F.; Palmigiano, A.; Paciello, I.; Pallach, M.; Garozzo, D.; Bernardini, M.-L.L.; Cono, V.L.; Yakimov, M.M.; Molinaro, A.; Silipo, A.; et al. The deep-sea polyextremophile Halobacteroides lacunaris TB21 rough-type LPS: Structure and inhibitory activity towards toxic LPS. Mar. Drugs 2017, 15, 201. [Google Scholar] [CrossRef]
  119. Homann, V.V.; Sandy, M.; Tincu, J.A.; Templeton, A.S.; Tebo, B.M.; Butler, A. Loihichelins A-F, a suite of amphiphilic siderophores produced by the marine bacterium Halomonas LOB-5. J. Nat. Prod. 2009, 72, 884–888. [Google Scholar] [CrossRef] [Green Version]
  120. Wang, L.; Große, T.; Stevens, H.; Brinkhoff, T.; Simon, M.; Liang, L.; Bitzer, J.; Bach, G.; Zeeck, A.; Tokuda, H.; et al. Bioactive hydroxyphenylpyrrole-dicarboxylic acids from a new marine Halomonas sp.: Production and structure elucidation. Appl. Microbiol. Biotechnol. 2006, 72, 816–822. [Google Scholar] [CrossRef]
  121. Bitzer, J.; Große, T.; Wang, L.; Lang, S.; Beil, W.; Zeeck, A. New aminophenoxazinones from a marine Halomonas sp.: Fermentation, structure elucidation, and biological activity. J. Antibiot. 2006, 59, 86–92. [Google Scholar] [CrossRef] [Green Version]
  122. Silipo, A.; Lanzetta, R.; Parrilli, M.; Sturiale, L.; Garozzo, D.; Nazarenko, E.L.; Gorshkova, R.P.; Ivanova, E.P.; Molinaro, A. The complete structure of the core carbohydrate backbone from the LPS of marine halophilic bacterium Pseudoalteromonas carrageenovora type strain IAM 12662T. Carbohydr. Res. 2005, 340, 1475–1482. [Google Scholar] [CrossRef]
  123. Mitova, M.; Tutino, M.L.; Infusini, G.; Marino, G.; De Rosa, S. Exocellular peptides from antarctic psychrophile Pseudoalteromonas haloplanktis. Mar. Biotechnol. 2005, 7, 523–531. [Google Scholar] [CrossRef] [PubMed]
  124. Shiozawa, H.; Shimada, A.; Takahashi, S. Thiomarinols D, E, F and G, new hybrid antimicrobial antibiotics produced by a marine bacterium. Isolated, structure, and antimicrobial activity. J. Antibiot. 1997, 50, 449–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Shiozawa, H.; Kagasaki, T.; Takahashi, S.; Kinoshita, T.; Haruyama, H.; Domon, H.; Utsuib, Y.; Kodama, K. Thiomarinol, a new hybrid antimicrobial antibiotic produced by a marine bacterium fermentation, isolation, structure, and antimicrobial activity. J. Antibiot. 1993, 46, 1834–1842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Mitchell, S.S.; Nicholson, B.; Teisan, S.; Lam, K.S.; Potts, B.C.M. Aureoverticillactam, a novel 22-atom macrocyclic lactam from the marine actinomycete Streptomyces aureoverticillatus. J. Nat. Prod. 2004, 67, 1400–1402. [Google Scholar] [CrossRef]
  127. Pesic, A.; Baumann, H.I.; Kleinschmidt, K.; Ensle, P.; Wiese, J.; Süssmuth, R.D.; Imhoff, J.F. Champacyclin, a new cyclic octapeptide from Streptomyces strain C42 isolated from the Baltic Sea. Mar. Drugs 2013, 11, 4834–4857. [Google Scholar] [CrossRef] [Green Version]
  128. Kasanah, N.; Triyanto, T. Bioactivities of Halometabolites from Marine Actinobacteria. Biomolecules 2019, 9, 225. [Google Scholar] [CrossRef] [Green Version]
  129. Martin, G.D.A.; Tan, L.T.; Jensen, P.R.; Dimayuga, R.E.; Fairchild, C.R.; Raventos-Suarez, C.; Fenical, W. Marmycins A and B, cytotoxic pentacyclic C-glycosides from a marine sediment-derived actinomycete related to the genus Streptomyces. J. Nat. Prod. 2007, 70, 1406–1409. [Google Scholar] [CrossRef]
  130. Zhou, X.; Huang, H.; Li, J.; Song, Y.; Jiang, R.; Liu, J.; Zhang, S.; Hua, Y.; Ju, J. New anti-infective cycloheptadepsipeptide congeners and absolute stereochemistry from the deep sea-derived Streptomyces drozdowiczii SCSIO 10141. Tetrahedron 2014, 70, 7795–7801. [Google Scholar] [CrossRef]
  131. Bruntner, C.; Binder, T.; Pathom-Aree, W.; Goodfellow, M.; Bull, A.T.; Potterat, O.; Puder, C.; Hörer, S.; Schmid, A.; Bolek, W.; et al. Frigocyclinone, a novel angucyclinone antibiotic produced by a Streptomyces griseus strain from Antarctica. J. Antibiot. 2005, 58, 346–349. [Google Scholar] [CrossRef] [Green Version]
  132. El-Gendy, M.M.A.; Shaaban, M.; Shaaban, K.A.; El-Bondkly, A.M.; Laatsch, H. Essramycin: A first triazolopyrimidine antibiotic isolated from nature. J. Antibiot. 2008, 61, 149–157. [Google Scholar] [CrossRef] [Green Version]
  133. Song, Y.; Huang, H.; Chen, Y.; Ding, J.; Zhang, Y.; Sun, A.; Zhang, W.; Ju, J. Cytotoxic and antibacterial marfuraquinocins from the deep south china sea-derived Streptomyces niveus scsio 3406. J. Nat. Prod. 2013, 76, 2263–2268. [Google Scholar] [CrossRef] [PubMed]
  134. Song, Y.; Li, Q.; Liu, X.; Chen, Y.; Zhang, Y.; Sun, A.; Zhang, W.; Zhang, J.; Ju, J. Cyclic hexapeptides from the deep South China sea-derived Streptomyces scopuliridis SCSIO ZJ46 active against pathogenic gram-positive bacteria. J. Nat. Prod. 2014, 77, 1937–1941. [Google Scholar] [CrossRef] [PubMed]
  135. Takahashi, A.; Kurasawa, S.; Ikeda, D.; Okami, Y.; Takeuchi, T. Altemicidin, a new acaricidal and antitumor substance. I. Taxonomy, fermentation, isolation and physico-chemical and biological properties. J. Antibiot. 1989, 42, 1556–1561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Pan, H.Q.; Zhang, S.Y.; Wang, N.; Li, Z.L.; Hua, H.M.; Hu, J.C.; Wang, S.J. New spirotetronate antibiotics, lobophorins H and I, from a South China Sea-derived Streptomyces sp. 12A35. Mar. Drugs 2013, 11, 3891–3901. [Google Scholar] [CrossRef] [Green Version]
  137. Moon, K.; Ahn, C.H.; Shin, Y.; Won, T.H.; Ko, K.; Lee, S.K.; Oh, K.B.; Shin, J.; Nam, S.I.; Oh, D.C. New Benzoxazine Secondary Metabolites from an Arctic Actinomycete. Mar. Drugs 2014, 12, 2526–5238. [Google Scholar] [CrossRef] [Green Version]
  138. Schultz, A.W.; Oh, D.C.; Carney, J.R.; Williamson, R.T.; Udwary, D.W.; Jensen, P.R.; Gould, S.J.; Fenical, W.; Moore, B.S. Biosynthesis and structures of cyclomarins and cyclomarazines, prenylated cyclic peptides of marine actinobacterial origin. J. Am. Chem. Soc. 2008, 130, 4507–4516. [Google Scholar] [CrossRef]
  139. Renner, M.K.; Shen, Y.C.; Cheng, X.C.; Jensen, P.R.; Frankmoelle, W.; Kauffman, C.A.; Fenical, W.; Lobkovsky, E.; Clardy, J. Cyclomarins A-C, new antiinflammatory cyclic peptides produced by a marine bacterium (Streptomyces sp.). J. Am. Chem. Soc. 1999, 121, 11273–11276. [Google Scholar] [CrossRef]
  140. Hughes, C.C.; Prieto-Davo, A.; Jensen, P.R.; Fenical, W. The marinopyrroles, antibiotics of an unprecedented structure class from a marine Streptomyces sp. Org. Lett. 2008, 10, 629–631. [Google Scholar] [CrossRef] [Green Version]
  141. Hughes, C.C.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Structures, Reactivities, and Antibiotic Properties of the Marinopyrroles A–F. J. Org. Chem. 2010, 75, 3240–3250. [Google Scholar] [CrossRef] [Green Version]
  142. Asolkar, R.N.; Jensen, P.R.; Kauffman, C.A.; Fenical, W. Daryamides A-C, weakly cytotoxic polyketides from a marine-derived actinomycete of the genus Streptomyces strain CNQ-085. J. Nat. Prod. 2006, 69, 1756–1759. [Google Scholar] [CrossRef]
  143. Hughes, C.C.; MacMillan, J.B.; Gaudêncio, S.P.; Jensen, P.R.; Fenical, W. The ammosamides: Structures of cell cycle modulators from a marine-derived Streptomyces species. Angew. Chemi. Int. Ed. 2009, 48, 725–727. [Google Scholar] [CrossRef] [Green Version]
  144. Hughes, C.C.; Fenical, W. Total synthesis of the ammosamides. J. Am. Chem. Soc. 2010, 132, 2528–2529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Pan, E.; Jamison, M.; Yousufuddin, M.; MacMillan, J.B. Ammosamide D, an oxidatively ring opened ammosamide analog from a marine-derived Streptomyces variabilis. Org. Lett. 2012, 14, 2390–2393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Li, F.; Maskey, R.P.; Qin, S.; Sattler, I.; Fiebig, H.H.; Maier, A.; Zeeck, A.; Laatsch, H. Chinikomycins A and B: Isolation, structure elucidation, and biological activity of novel antibiotics from a marine Streptomyces sp. isolate M045. J. Nat. Prod. 2005, 68, 349–353. [Google Scholar] [CrossRef] [PubMed]
  147. Pérez, M.; Crespo, C.; Schleissner, C.; Rodríguez, P.; Zúñiga, P.; Reyes, F. Tartrolon D, a cytotoxic macrodiolide from the marine-derived actinomycete Streptomyces sp. MDG-04-17-069. J. Nat. Prod. 2009, 72, 2192–2194. [Google Scholar] [CrossRef] [PubMed]
  148. Hawas, U.W.; Shaaban, M.; Shaaban, K.A.; Speitling, M.; Maier, A.; Kelter, G.; Fiebig, H.H.; Meiners, M.; Helmke, E.; Laatsch, H. Mansouramycins A-D, cytotoxic isoquinolinequinones from a marine Streptomycete. J. Nat. Prod. 2009, 72, 2120–2124. [Google Scholar] [CrossRef] [PubMed]
  149. Nachtigall, J.; Schneider, K.; Bruntner, C.; Bull, A.T.; Goodfellow, M.; Zinecker, H.; Imhoff, J.F.; Nicholson, G.; Irran, E.; Süssmuth, R.D.; et al. Benzoxacystol, a benzoxazine-type enzyme inhibitor from the deep-sea strain Streptomyces sp. NTK 935. J. Antibiot. 2011, 64, 453–457. [Google Scholar] [CrossRef] [Green Version]
  150. Zhang, W.; Liu, Z.; Li, S.; Yang, T.; Zhang, Q.; Ma, L.; Tian, X.; Zhang, H.; Huang, C.; Zhang, S.; et al. Spiroindimicins A-D: New bisindole alkaloids from a deep-sea-derived actinomycete. Org. Lett. 2012, 14, 3364–3367. [Google Scholar] [CrossRef]
  151. Song, Y.; Liu, G.; Li, J.; Huang, H.; Zhang, X.; Zhang, H.; Ju, J. Cytotoxic and antibacterial angucycline- and prodigiosin-analogues from the deep-sea derived Streptomyces sp. SCSIO 11594. Mar. Drugs 2015, 13, 1304–1316. [Google Scholar] [CrossRef]
  152. You, Z.Y.; Wang, Y.H.; Zhang, Z.G.; Xu, M.J.; Xie, S.J.; Han, T.S.; Feng, L.; Li, X.G.; Xu, J. Identification of two novel anti-fibrotic benzopyran compounds produced by engineered strains derived from Streptomyces xiamenensis M1-94P that originated from deep-sea sediments. Mar. Drugs 2013, 11, 4035–4049. [Google Scholar] [CrossRef]
  153. Miller, E.D.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Piperazimycins: Cytotoxic hexadepsipeptides from a marine-derived bacterium of the genus Streptomyces. J. Org. Chem. 2007, 72, 323–330. [Google Scholar] [CrossRef] [PubMed]
  154. Sun, M.-L.; Liu, S.-B.; Qiao, L.-P.; Chen, X.-L.; Pang, X.; Shi, M.; Zhang, X.-Y.; Qin, Q.-L.; Zhou, B.-C.; Zhang, Y.-Z.; et al. A novel exopolysaccharide from deep-sea bacterium Zunongwangia profunda SM-A87: Low-cost fermentation, moisture retention, and antioxidant activities. Appl. Microbiol. Biotechnol. 2014, 98, 7437–7445. [Google Scholar] [CrossRef] [PubMed]
  155. Chater, K.F. Recent advances in understanding Streptomyces. F1000Research 2016, 5, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Hettiarachchi, S.A.; Lee, S.-J.; Lee, Y.; Kwon, Y.-K.; De Zoysa, M.; Moon, S.; Jo, E.; Kim, T.; Kang, D.-H.; Heo, S.-J.; et al. A rapid and efficient screening method for antibacterial compound-producing bacteria. J. Microbiol. Biotechnol. 2017, 27, 1441–1448. [Google Scholar] [PubMed]
  157. Andreo-Vidal, A.; Sanchez-Amat, A.; Campillo-Brocal, J. The Pseudoalteromonas luteoviolacea L-amino Acid Oxidase with Antimicrobial Activity Is a Flavoenzyme. Mar. Drugs 2018, 16, 499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Kalitnik, A.A.; Byankina Barabanova, A.O.; Nagorskaya, V.P.; Reunov, A.V.; Glazunov, V.P.; Solov’eva, T.F.; Yermak, I.M. Low molecular weight derivatives of different carrageenan types and their antiviral activity. J. Appl. Phycol. 2013, 25, 65–72. [Google Scholar] [CrossRef]
  159. Queiroz, E.A.I.F.; Fortes, Z.B.; da Cunha, M.A.A.; Sarilmiser, H.K.; Barbosa Dekker, A.M.; Öner, E.T.; Dekker, R.F.H.; Khaper, N. Levan promotes antiproliferative and pro-apoptotic effects in MCF-7 breast cancer cells mediated by oxidative stress. Int. J. Biol. Macromol. 2017, 102, 565–570. [Google Scholar] [CrossRef]
  160. Ruiz-Ruiz, C.; Srivastava, G.K.; Carranza, D.; Mata, J.A.; Llamas, I.; Santamaría, M.; Quesada, E.; Molina, I.J. An exopolysaccharide produced by the novel halophilic bacterium Halomonas stenophila strain B100 selectively induces apoptosis in human T leukaemia cells. Appl. Microbiol. Biotechnol. 2011, 89, 345–355. [Google Scholar] [CrossRef]
  161. Lorenz, P.; Eck, J. Metagenomics and industrial applications. Nat. Rev. Microbiol. 2005, 3, 510–516. [Google Scholar] [CrossRef]
  162. Prasad, S.; Roy, I. Converting Enzymes into Tools of Industrial Importance. Recent Pat. Biotechnol. 2018, 12, 33–56. [Google Scholar] [CrossRef]
  163. Bommarius, A.S.; Paye, M.F. Stabilizing biocatalysts. Chem. Soc. Rev. 2013, 42, 6534–6565. [Google Scholar] [CrossRef] [PubMed]
  164. Madhavan, A.; Sindhu, R.; Binod, P.; Sukumaran, R.K.; Pandey, A. Strategies for design of improved biocatalysts for industrial applications. Bioresour. Technol. 2017, 245, 1304–1313. [Google Scholar] [CrossRef] [PubMed]
  165. Martínez-Martínez, M.; Bargiela, R.; Ferrer, M. Metagenomics and the Search for Industrial Enzymes. In Biotechnology of Microbial Enzymes: Production, Biocatalysis and Industrial Applications; Brahmachari, G., Demain, A.L., Adrio, J.L., Eds.; Academic Press: Cambridge, MA, USA, 2016; pp. 1–608. [Google Scholar]
  166. Chapman, J.; Ismail, A.; Dinu, C. Industrial Applications of Enzymes: Recent Advances, Techniques, and Outlooks. Catalysts 2018, 8, 238. [Google Scholar] [CrossRef] [Green Version]
  167. Dumorné, K.; Córdova, D.C.; Astorga-Eló, M.; Renganathan, P. Extremozymes: A potential source for industrial applications. J. Microbiol. Biotechnol. 2017, 27, 649–659. [Google Scholar] [CrossRef] [PubMed]
  168. Dalmaso, G.; Ferreira, D.; Vermelho, A. Marine Extremophiles: A Source of Hydrolases for Biotechnological Applications. Mar. Drugs 2015, 13, 1925–1965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Sarmiento, F.; Peralta, R.; Blamey, J.M. Cold and Hot Extremozymes: Industrial Relevance and Current Trends. Front. Bioeng. Biotechnol. 2015, 3, 1–15. [Google Scholar] [CrossRef] [Green Version]
  170. Di Donato, P.; Buono, A.; Poli, A.; Finore, I.; Abbamondi, G.R.; Nicolaus, B.; Lama, L. Exploring marine environments for the identification of extremophiles and their enzymes for sustainable and green bioprocesses. Sustainability 2019, 11, 149. [Google Scholar] [CrossRef] [Green Version]
  171. Bruno, S.; Coppola, D.; di Prisco, G.; Giordano, D.; Verde, C. Enzymes from Marine Polar Regions and Their Biotechnological Applications. Mar. Drugs 2019, 17, 544. [Google Scholar] [CrossRef] [Green Version]
  172. Yamanaka, Y.; Kazuoka, T.; Yoshida, M.; Yamanaka, K.; Oikawa, T.; Soda, K. Thermostable aldehyde dehydrogenase from psychrophile, Cytophaga sp. KUC-1: Enzymological characteristics and functional properties. Biochem. Biophys. Res. Commun. 2002, 298, 632–637. [Google Scholar] [CrossRef]
  173. Anburajan, L.; Meena, B.; Narendar Sivvaswamy, S. First report on molecular characterization of novel betaine aldehyde dehydrogenase from the halotolerant eubacteria, Bacillus halodurans. Gene Reports 2017, 9, 131–135. [Google Scholar] [CrossRef]
  174. Kim, H.J.; Joo, W.A.; Cho, C.W.; Kim, C.W. Halophile aldehyde dehydrogenase from Halobacterium salinarum. J. Proteome Res. 2006, 5, 192–195. [Google Scholar] [CrossRef] [PubMed]
  175. Akal, A.L.; Karan, R.; Hohl, A.; Alam, I.; Vogler, M.; Grötzinger, S.W.; Eppinger, J.; Rueping, M. A polyextremophilic alcohol dehydrogenase from the Atlantis II Deep Red Sea brine pool. FEBS Open Bio 2019, 9, 194–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Borchert, E.; Knobloch, S.; Dwyer, E.; Flynn, S.; Jackson, S.A.; Jóhannsson, R.; Marteinsson, V.T.; O’Gara, F.; Dobson, A.D.W.W.; O’Gara, F.; et al. Biotechnological Potential of Cold Adapted Pseudoalteromonas spp. Isolated from ‘Deep Sea’ Sponges. Mar. Drugs 2017, 15, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Izotova, L.S.; Strongin, A.Y.; Chekulaeva, L.N.; Sterkin, V.E.; Ostoslavskaya, V.I.; Lyublinskaya, L.A.; Timokhina, E.A.; Stepanov, V.M. Purification and properties of serine protease from Halobacterium halobium. J. Bacteriol. 1983, 155, 826–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Venugopal, M.; Saramma, A.V. An alkaline protease from Bacillus circulans BM15, newly isolated from a mangrove station: Characterization and application in laundry detergent formulations. Indian J. Microbiol. 2007, 47, 298–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Wu, S.; Liu, G.; Zhang, D.; Li, C.; Sun, C. Purification and biochemical characterization of an alkaline protease from marine bacteria Pseudoalteromonas sp. 129-1. J. Basic Microbiol. 2015, 55, 1427–1434. [Google Scholar] [CrossRef]
  180. Ibrahim, A.S.S.S.; Al-Salamah, A.A.; El-Badawi, Y.B.; El-Tayeb, M.A.; Antranikian, G. Detergent-, solvent- and salt-compatible thermoactive alkaline serine protease from halotolerant alkaliphilic Bacillus sp. NPST-AK15: Purification and characterization. Extremophiles 2015, 19, 961–971. [Google Scholar] [CrossRef]
  181. Raval, V.H.; Pillai, S.; Rawal, C.M.; Singh, S.P. Biochemical and structural characterization of a detergent-stable serine alkaline protease from seawater haloalkaliphilic bacteria. Process Biochem. 2014, 49, 955–962. [Google Scholar] [CrossRef]
  182. Mothe, T.; Sultanpuram, V.R. Production, purification and characterization of a thermotolerant alkaline serine protease from a novel species Bacillus caseinilyticus. 3 Biotech 2016, 6, 1–10. [Google Scholar] [CrossRef] [Green Version]
  183. Sanchez-Porro, C.; Martin, S.; Mellado, E.; Ventosa, A. Diversity of moderately halophilic bacteria producing extracellular hydrolytic enzymes. J. Appl. Microbiol. 2003, 94, 295–300. [Google Scholar] [CrossRef]
  184. Sánchez-Porro, C.; Mellado, E.; Pugsley, A.P.; Francetic, O.; Ventosa, A. The haloprotease CPI produced by the moderately halophilic bacterium Pseudoalteromonas ruthenica is secreted by the type II secretion pathway. Appl. Environ. Microbiol. 2009, 75, 4197–4201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Lama, L.; Romano, I.; Calandrelli, V.; Nicolaus, B.; Gambacorta, A. Purification and characterization of a protease produced by an aerobic haloalkaliphilic species belonging to the Salinivibrio genus. Res. Microbiol. 2005, 156, 478–484. [Google Scholar] [CrossRef] [PubMed]
  186. Behera, B.C.; Sethi, B.K.; Mishra, R.R.; Dutta, S.K.; Thatoi, H.N. Microbial cellulases—Diversity & biotechnology with reference to mangrove environment: A review. J. Genet. Eng. Biotechnol. 2017, 15, 197–210. [Google Scholar] [PubMed]
  187. Maki, M.; Leung, K.T.; Qin, W. The prospects of cellulase-producing bacteria for the bioconversion of lignocellulosic biomass. Int. J. Biol. Sci. 2009, 5, 500–516. [Google Scholar] [CrossRef] [PubMed]
  188. Dos Santos, Y.Q.; de Veras, B.O.; de França, A.F.J.; Gorlach-Lira, K.; Velasques, J.; Migliolo, L.; Dos Santos, E.A. A new salt-tolerant thermostable cellulase from a marine bacillus sp. Strain. J. Microbiol. Biotechnol. 2018, 28, 1078–1085. [Google Scholar] [CrossRef]
  189. Werner, J.; Ferrer, M.; Michel, G.; Mann, A.J.; Huang, S.; Juarez, S.; Ciordia, S.; Albar, J.P.; Alcaide, M.; La Cono, V.; et al. Halorhabdus tiamatea: Proteogenomics and glycosidase activity measurements identify the first cultivated euryarchaeon from a deep-sea anoxic brine lake as potential polysaccharide degrader. Environ. Microbiol. 2014, 16, 2525–2537. [Google Scholar] [CrossRef] [Green Version]
  190. Zhang, T.; Datta, S.; Eichler, J.; Ivanova, N.; Axen, S.D.; Kerfeld, C.A.; Chen, F.; Kyrpides, N.; Hugenholtz, P.; Cheng, J.F.; et al. Identification of a haloalkaliphilic and thermostable cellulase with improved ionic liquid tolerance. Green Chem. 2011, 13, 2083–2090. [Google Scholar] [CrossRef] [Green Version]
  191. Zhang, C.; Wang, X.; Zhang, W.; Zhao, Y.; Lu, X. Expression and characterization of a glucose-tolerant β-1,4-glucosidase with wide substrate specificity from Cytophaga hutchinsonii. Appl. Microbiol. Biotechnol. 2017, 101, 1919–1926. [Google Scholar] [CrossRef]
  192. Makhdoumi, A.; Dehghani-Joybari, Z.; Mashreghi, M.; Jamialahmadi, K.; Asoodeh, A. A novel halo-alkali-tolerant and thermo-tolerant chitinase from Pseudoalteromonas sp. DC14 isolated from the Caspian Sea. Int. J. Environ. Sci. Technol. 2015, 12, 3895–3904. [Google Scholar] [CrossRef] [Green Version]
  193. Roman, D.L.; Roman, M.; Sletta, H.; Ostafe, V.; Isvoran, A. Assessment of the properties of chitin deacetylases showing different enzymatic action patterns. J. Mol. Graph. Model. 2019, 88, 41–48. [Google Scholar] [CrossRef]
  194. Zhao, Y.; Park, R.D.; Muzzarelli, R.A.A. Chitin deacetylases: Properties and applications. Mar. Drugs 2010, 8, 24–46. [Google Scholar] [CrossRef] [PubMed]
  195. Kim, S.B.; Lee, W.; Ryu, Y.W. Cloning and characterization of thermostable esterase from Archaeoglobus fulgidus. J. Microbiol. 2008, 46, 100–107. [Google Scholar] [CrossRef] [PubMed]
  196. Ghati, A.; Paul, G. Purification and characterization of a thermo-halophilic, alkali-stable and extremely benzene tolerant esterase from a thermo-halo tolerant Bacillus cereus strain AGP-03, isolated from ‘Bakreshwar’ hot spring, India. Process Biochem. 2015, 50, 771–781. [Google Scholar] [CrossRef]
  197. Mohamed, Y.M.; Ghazy, M.A.; Sayed, A.; Ouf, A.; El-Dorry, H.; Siam, R. Isolation and characterization of a heavy metal-resistant, thermophilic esterase from a Red Sea Brine Pool. Sci. Rep. 2013, 3, 3358. [Google Scholar] [CrossRef] [Green Version]
  198. Ferrer, M.; Golyshina, O.V.; Chernikova, T.N.; Khachane, A.N.; Martins, V.A.P.; Santos, D.; Yakimov, M.M.; Timmis, K.N.; Golyshin, P.N. Microbial Enzymes Mined from the Urania Deep-Sea Hypersaline Anoxic Basin. Chem. Biol. 2005, 12, 895–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Zhang, S.; Wu, G.; Liu, Z.; Shao, Z.; Liu, Z. Characterization of EstB, a novel cold-active and organic solvent-tolerant esterase from marine microorganism Alcanivorax dieselolei B-5(T). Extremophiles 2014, 18, 251–259. [Google Scholar] [CrossRef]
  200. Rahman, M.A.; Culsum, U.; Tang, W.; Zhang, S.W.; Wu, G.; Liu, Z. Characterization of a novel cold active and salt tolerant esterase from Zunongwangia profunda. Enzyme Microb. Technol. 2016, 85, 1–11. [Google Scholar] [CrossRef]
  201. Garczarek, F.; Dong, M.; Typke, D.; Witkowska, H.E.; Hazen, T.C.; Nogales, E.; Biggin, M.D.; Glaeser, R.M. Octomeric pyruvate-ferredoxin oxidoreductase from Desulfovibrio vulgaris. J. Struct. Biol. 2007, 159, 9–18. [Google Scholar] [CrossRef]
  202. Bock, A.K.; Schönheit, P.; Teixeira, M. The iron-sulfur centers of the pyruvate:ferredoxin oxidoreductase from Methanosarcina barkeri (Fusaro). FEBS Lett. 1997, 414, 209–212. [Google Scholar]
  203. Chen, C.K.M.; Lee, G.C.; Ko, T.P.; Guo, R.T.; Huang, L.M.; Liu, H.J.; Ho, Y.F.; Shaw, J.F.; Wang, A.H.J. Structure of the Alkalohyperthermophilic Archaeoglobus fulgidus Lipase Contains a Unique C-Terminal Domain Essential for Long-Chain Substrate Binding. J. Mol. Biol. 2009, 390, 672–685. [Google Scholar] [CrossRef]
  204. Akbari, E.; Beheshti-Maal, K.; Nayeri, H. A novel halo-alkalo-tolerant bacterium, Marinobacter alkaliphilus ABN-IAUF-1, isolated from Persian Gulf suitable for alkaline lipase production. Int. J. Environ. Sci. Technol. 2018, 15, 1767–1776. [Google Scholar] [CrossRef]
  205. Jeon, J.H.; Kim, J.-T.; Kim, Y.J.; Kim, H.-K.; Lee, H.S.; Kang, S.G.; Kim, S.-J.; Lee, J.-H. Cloning and characterization of a new cold-active lipase from a deep-sea sediment metagenome. Appl. Microbiol. Biotechnol. 2009, 81, 865–874. [Google Scholar] [CrossRef] [PubMed]
  206. Loperena, L.; Soria, V.; Varela, H.; Lupo, S.; Bergalli, A.; Guigou, M.; Pellegrino, A.; Bernardo, A.; Calviño, A.; Rivas, F.; et al. Extracellular enzymes produced by microorganisms isolated from maritime Antarctica. World J. Microbiol. Biotechnol. 2012, 28, 2249–2256. [Google Scholar] [CrossRef] [PubMed]
  207. Kiran, G.S.; Lipton, A.N.; Kennedy, J.; Dobson, A.D.W.; Selvin, J. A halotolerant thermostable lipase from the marine bacterium Oceanobacillus sp. PUMB02 with an ability to disrupt bacterial biofilms. Bioeng. Bugs 2014, 5, 305–318. [Google Scholar]
  208. Ciok, A.; Dziewit, L. Exploring the genome of Arctic Psychrobacter sp. DAB_AL32B and construction of novel Psychrobacter-specific cloning vectors of an increased carrying capacity. Arch. Microbiol. 2019, 201, 559–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Sayed, A.; Ghazy, M.A.; Ferreira, A.J.S.; Setubal, J.C.; Chambergo, F.S.; Ouf, A.; Adel, M.; Dawe, A.S.; Archer, J.A.C.; Bajic, V.B.; et al. A novel mercuric reductase from the unique deep brine environment of Atlantis II in the Red sea. J. Biol. Chem. 2014, 289, 1675–1687. [Google Scholar] [CrossRef] [Green Version]
  210. Zhou, P.; Huo, Y.Y.; Xu, L.; Wu, Y.H.; Meng, F.X.; Wang, C.S.; Xu, X.W. Investigation of mercury tolerance in Chromohalobacter israelensis DSM 6768T and Halomonas zincidurans B6T by comparative genomics with Halomonas xinjiangensis TRM 0175T. Mar. Genomics 2014, 19, 15–16. [Google Scholar] [CrossRef]
  211. Sonbol, S.A.; Ferreira, A.J.S.; Siam, R. Red Sea Atlantis II brine pool nitrilase with unique thermostability profile and heavy metal tolerance. BMC Biotechnol. 2016, 16, 1–13. [Google Scholar] [CrossRef] [Green Version]
  212. Hii, S.L.; Tan, J.S.; Ling, T.C.; Ariff, A. Bin Pullulanase: Role in Starch Hydrolysis and Potential Industrial Applications. Enzyme Res. 2012, 2012, 1–14. [Google Scholar] [CrossRef] [Green Version]
  213. Chakdar, H.; Kumar, M.; Pandiyan, K.; Singh, A.; Nanjappan, K.; Kashyap, P.L.; Srivastava, A.K. Bacterial xylanases: Biology to biotechnology. 3 Biotech 2016, 6, 150. [Google Scholar] [CrossRef] [Green Version]
  214. Araki, T.; Tani, S.; Maeda, K.; Hashikawa, S.; Nakagawa, H.; Morishita, T. Purification and Characterization of β-1,3-Xylanase from a Marine Bacterium, Vibrio sp. XY-214. Biosci. Biotechnol. Biochem. 1999, 63, 2017–2019. [Google Scholar] [CrossRef] [PubMed]
  215. Wejse, P.L.; Ingvorsen, K.; Mortensen, K.K. Purification and characterisation of two extremely halotolerant xylanases from a novel halophilic bacterium. Extremophiles 2003, 7, 423–431. [Google Scholar] [CrossRef] [PubMed]
  216. Kim, J.J.; Kwon, Y.K.; Kim, J.H.; Heo, S.J.; Lee, Y.; Lee, S.J.; Shim, W.B.; Jung, W.K.; Hyun, J.H.; Kwon, K.K.; et al. Effective microwell plate-based screening method for microbes producing cellulase and xylanase and its application. J. Microbiol. Biotechnol. 2014, 24, 1559–1565. [Google Scholar] [CrossRef] [PubMed]
  217. Møller, M.F.; Kjeldsen, K.U.; Ingvorsen, K. Marinimicrobium haloxylanilyticum sp. nov., a new moderately halophilic, polysaccharide-degrading bacterium isolated from Great Salt Lake, Utah. Antonie van Leeuwenhoek, Int. J. Gen. Mol. Microbiol. 2010, 98, 553–565. [Google Scholar] [CrossRef]
  218. Rattu, G.; Joshi, S.; Satyanarayana, T. Bifunctional recombinant cellulase–xylanase (rBhcell-xyl) from the polyextremophilic bacterium Bacillus halodurans TSLV1 and its utility in valorization of renewable agro-residues. Extremophiles 2016, 20, 831–842. [Google Scholar] [CrossRef]
  219. Kim, J.; Hong, S.-K. Isolation and Characterization of an Agarase-Producing Bacterial Strain, Alteromonas sp. GNUM-1, from the West Sea, Korea. J. Microbiol. Biotechnol. 2012, 22, 1621–1628. [Google Scholar] [CrossRef] [Green Version]
  220. Fu, X.T.; Kim, S.M. Agarase: Review of major sources, categories, purification method, enzyme characteristics and applications. Mar. Drugs 2010, 8, 200–218. [Google Scholar] [CrossRef] [Green Version]
  221. Han, X.; Lin, B.; Ru, G.; Zhang, Z.; Liu, Y.; Hu, Z. Gene Cloning and Characterization of an α-Amylase from Alteromonas macleodii B7 for Enteromorpha Polysaccharide Degradation. J. Microbiol. Biotechnol. 2014, 24, 254–263. [Google Scholar] [CrossRef]
  222. Sewalt, V.J.; Reyes, T.F.; Bui, Q. Safety evaluation of two α-amylase enzyme preparations derived from Bacillus licheniformis expressing an α-amylase gene from Cytophaga species. Regul. Toxicol. Pharmacol. 2018, 98, 140–150. [Google Scholar] [CrossRef]
  223. Amoozegar, M.A.; Siroosi, M.; Atashgahi, S.; Smidt, H.; Ventosa, A. Systematics of haloarchaea and biotechnological potential of their hydrolytic enzymes. Microbiology 2017, 163, 623–645. [Google Scholar] [CrossRef] [Green Version]
  224. Qin, Y.; Huang, Z.; Liu, Z. A novel cold-active and salt-tolerant α-amylase from marine bacterium Zunongwangia profunda: Molecular cloning, heterologous expression and biochemical characterization. Extremophiles 2014, 18, 271–281. [Google Scholar] [CrossRef] [PubMed]
  225. Amoozegar, M.A.; Malekzadeh, F.; Malik, K.A. Production of amylase by newly isolated moderate halophile, Halobacillus sp. strain MA-2. J. Microbiol. Methods 2003, 52, 353–359. [Google Scholar] [CrossRef]
  226. de Lourdes Moreno, M.; Pérez, D.; García, M.; Mellado, E.; De Lourdes Moreno, M.; Pérez, D.; García, M.T.; Mellado, E. Halophilic Bacteria as a Source of Novel Hydrolytic Enzymes. Life 2013, 3, 38–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Kiran, K.; Koteswaraiah, P.; Chandra, T.S. Production of Halophilic α-Amylase by Immobilized Cells of Moderately Halophilic Bacillus sp. Strain TSCVKK. Br. Microbiol. Res. J. 2012, 2, 146–157. [Google Scholar] [CrossRef]
  228. Uotsu-Tomita, R.; Tonozuka, T.; Sakai, H.; Sakano, Y. Novel glucoamylase-type enzymes from Thermoactinomyces vulgaris and Methanococcus jannaschii whose genes are found in the flanking region of the α-amylase genes. Appl. Microbiol. Biotechnol. 2001, 56, 465–473. [Google Scholar] [CrossRef]
  229. Kim, J.W.; Flowers, L.O.; Whiteley, M.; Peeples, T.L. Biochemical confirmation and characterization of the family-57-like α-amylase of Methanococcus jannaschii. Folia Microbiol. 2001, 46, 467–473. [Google Scholar] [CrossRef]
  230. Aghajari, N.; Feller, G.; Gerday, C.; Haser, R. Structures of the psychrophilic Alteromonas haloplanctis α-amylase give insights into cold adaptation at a molecular level. Structure 1998, 6, 1503–1516. [Google Scholar] [CrossRef] [Green Version]
  231. Mageswari, A.; Subramanian, P.; Chandrasekaran, S.; Sivashanmugam, K.; Babu, S.; Gothandam, K.M. Optimization and immobilization of amylase obtained from halotolerant bacteria isolated from solar salterns. J. Genet. Eng. Biotechnol. 2012, 10, 201–208. [Google Scholar] [CrossRef]
  232. Coronado, M.J.; Vargas, C.; Mellado, E.; Tegos, G.; Drainas, C.; Nieto, J.J.; Ventosa, A. The α-amylase gene amyH of the moderate halophile Halomonas meridiana: Cloning and molecular characterization. Microbiology 2000, 146, 861–868. [Google Scholar] [CrossRef] [Green Version]
  233. Singh, G.; Verma, A.K.; Kumar, V. Catalytic properties, functional attributes and industrial applications of β-glucosidases. 3 Biotech 2016, 6, 1–14. [Google Scholar] [CrossRef] [Green Version]
  234. Sun, J.; Wang, W.; Yao, C.; Dai, F.; Zhu, X.; Liu, J.; Hao, J. Overexpression and characterization of a novel cold-adapted and salt-tolerant GH1 β-glucosidase from the marine bacterium Alteromonas sp. L82. J. Microbiol. 2018, 56, 656–664. [Google Scholar] [CrossRef] [PubMed]
  235. Hebbale, D.; Bhargavi, R.; Ramachandra, T.V. Saccharification of macroalgal polysaccharides through prioritized cellulase producing bacteria. Heliyon 2019, 5, e01372. [Google Scholar] [CrossRef] [Green Version]
  236. Zhu, B.; Ning, L. Purification and Characterization of a New k-Carrageenase from the Marine Bacterium Vibrio sp. NJ-2. J. Microbiol. Biotechnol. 2016, 26, 255–262. [Google Scholar] [CrossRef] [PubMed]
  237. Xiao, A.; Zeng, J.; Li, J.; Zhu, Y.; Xiao, Q.; Ni, H. Molecular cloning, characterization, and heterologous expression of a new κ-carrageenase gene from Pseudoalteromonas carrageenovora ASY5. J. Food Biochem. 2018, 42. [Google Scholar] [CrossRef]
  238. Blanco, K.C.; De Lima, C.J.B.; Monti, R.; Martins, J.; Bernardi, N.S.; Contiero, J. Bacillus lehensis—An alkali-tolerant bacterium isolated from cassava starch wastewater: Optimization of parameters for cyclodextrin glycosyltransferase production. Ann. Microbiol. 2012, 62, 329–337. [Google Scholar] [CrossRef]
  239. Suriya, J.; Bharathiraja, S.; Krishnan, M.; Manivasagan, P.; Kim, S.-K. Marine Microbial Amylases. In Advances in food and nutrition research; Elsevier Inc. Academic Press: Cambridge, MA, USA, 2016; Volume 79, pp. 161–177. ISBN 978-0-12-804714-9. [Google Scholar]
  240. Mehta, D.; Satyanarayana, T. Bacterial and archaeal α-amylases: Diversity and amelioration of the desirable characteristics for industrial applications. Front. Microbiol. 2016, 7. [Google Scholar] [CrossRef] [Green Version]
  241. Jabbour, D.; Sorger, A.; Sahm, K.; Antranikian, G. A highly thermoactive and salt-tolerant α-amylase isolated from a pilot-plant biogas reactor. Appl. Microbiol. Biotechnol. 2013, 97, 2971–2978. [Google Scholar] [CrossRef] [Green Version]
  242. Cowan, D.; Cramp, R.; Pereira, R.; Graham, D.; Almatawah, Q. Biochemistry and biotechnology of mesophilic and thermophilic nitrile metabolizing enzymes. Extremophiles 1998, 2, 207–216. [Google Scholar] [CrossRef]
  243. Gupta, N.; Balomajumder, C.; Agarwal, V.K. Enzymatic mechanism and biochemistry for cyanide degradation: A review. J. Hazard. Mater. 2010, 176, 1–13. [Google Scholar] [CrossRef]
  244. Nigam, V.K.; Arfi, T.; Kumar, V.; Shukla, P. Bioengineering of Nitrilases Towards Its Use as Green Catalyst: Applications and Perspectives. Indian J. Microbiol. 2017, 57, 131–138. [Google Scholar] [CrossRef]
  245. Kuddus, M. Cold-active enzymes in food biotechnology: An updated mini review. J. Appl. Biol. Biotechnol. 2018, 6, 58–63. [Google Scholar]
  246. Hasan, F.; Shah, A.A.; Hameed, A. Industrial applications of microbial lipases. Enzyme Microb. Technol. 2006, 39, 235–251. [Google Scholar] [CrossRef]
  247. Mahjoubi, M.; Cappello, S.; Souissi, Y.; Jaouani, A.; Cherif, A. Microbial Bioremediation of Petroleum Hydrocarbon–Contaminated Marine Environments. In Recent Insights in Petroleum Science and Engineering; Zoveidavianpoor, M., Ed.; InTechOpen: London, UK, 2018; pp. 325–350. [Google Scholar]
  248. Hassanshahian, M.; Cappello, S. Crude Oil Biodegradation in the Marine Environments. In Biodegradation—Engineering and Technology; Rolando Chamy, Ed.; InTechOpen: London, UK, 2013; pp. 101–135. [Google Scholar]
  249. Floodgate, G.D. Biodegradation of hydrocarbons in the sea. In Water Pollution Microbiology; Mitchell, R., Ed.; Wiley-Interscience: New York, NY, USA, 1972; pp. 153–171. [Google Scholar]
  250. Jarvis, I.W.H.; Dreij, K.; Mattsson, Å.; Jernström, B.; Stenius, U. Interactions between polycyclic aromatic hydrocarbons in complex mixtures and implications for cancer risk assessment. Toxicology 2014, 321, 27–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Moreno, R.; Jover, L.; Diez, C.; Sardà, F.; Sanpera, C. Ten Years after the Prestige Oil Spill: Seabird Trophic Ecology as Indicator of Long-Term Effects on the Coastal Marine Ecosystem. PLoS ONE 2013, 8. [Google Scholar] [CrossRef]
  252. Sikkema, J.; de Bont, J.A.; Poolman, B. Mechanisms of Membrane Toxicity of Hydrocarbons. Microbiol. Rev. 1995, 59, 201–222. [Google Scholar] [CrossRef]
  253. Catania, V.; Santisi, S.; Signa, G.; Vizzini, S.; Mazzola, A.; Cappello, S.; Yakimov, M.M.; Quatrini, P. Intrinsic bioremediation potential of a chronically polluted marine coastal area. Mar. Pollut. Bull. 2015, 99, 138–149. [Google Scholar] [CrossRef]
  254. Durval, I.J.B.; Resende, A.H.M.; Figueiredo, M.A.; Luna, J.M.; Rufino, R.D.; Sarubbo, L.A. Studies on Biosurfactants Produced using Bacillus cereus Isolated from Seawater with Biotechnological Potential for Marine Oil-Spill Bioremediation. J. Surfactants Deterg. 2018, 22, 349–363. [Google Scholar] [CrossRef]
  255. Hazen, T.C.; Dubinsky, E.A.; DeSantis, T.Z.; Andersen, G.L.; Piceno, Y.M.; Singh, N.; Jansson, J.K.; Probst, A.; Borglin, S.E.; Fortney, J.L.; et al. Deep-sea oil plume enriches indigenous oil-degrading bacteria. Science 2010, 330, 204–208. [Google Scholar] [CrossRef] [Green Version]
  256. Kleindienst, S.; Paul, J.H.; Joye, S.B. Using dispersants after oil spills: Impacts on the composition and activity of microbial communities. Nat. Rev. Microbiol. 2015, 13, 388–396. [Google Scholar] [CrossRef]
  257. Speight, J.G.; El-Gendy, N.S. Introduction to petroleum biotechnology; Elsevier: Amsterdam, The Netherlands, 2017; ISBN 9780128051511. [Google Scholar]
  258. Fuentes, S.; Méndez, V.; Aguila, P.; Seeger, M. Bioremediation of petroleum hydrocarbons: Catabolic genes, microbial communities, and applications. Appl. Microbiol. Biotechnol. 2014, 98, 4781–4794. [Google Scholar] [CrossRef]
  259. Fuentes, S.; Barra, B.; Gregory Caporaso, J.; Seeger, M. From rare to dominant: A fine-tuned soil bacterial bloom during petroleum hydrocarbon bioremediation. Appl. Environ. Microbiol. 2016, 82, 888–896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Varjani, S.J.; Gnansounou, E. Microbial dynamics in petroleum oilfields and their relationship with physiological properties of petroleum oil reservoirs. Bioresour. Technol. 2017, 245, 1258–1265. [Google Scholar] [CrossRef] [PubMed]
  261. Coulon, F.; McKew, B.A.; Osborn, A.M.; McGenity, T.J.; Timmis, K.N. Effects of temperature and biostimulation on oil-degrading microbial communities in temperate estuarine waters. Environ. Microbiol. 2007, 9, 177–186. [Google Scholar] [CrossRef] [PubMed]
  262. Xu, X.; Liu, W.; Tian, S.; Wang, W.; Qi, Q.; Jiang, P.; Gao, X.; Li, F.; Li, H.; Yu, H. Petroleum Hydrocarbon-Degrading Bacteria for the Remediation of Oil Pollution Under Aerobic Conditions: A Perspective Analysis. Front. Microbiol. 2018, 9, 1–11. [Google Scholar] [CrossRef]
  263. Fathepure, B.Z. Recent studies in microbial degradation of petroleum hydrocarbons in hypersaline environments. Front. Microbiol. 2014, 5. [Google Scholar] [CrossRef] [Green Version]
  264. Oren, A. Aerobic Hydrocarbon-Degrading Archaea. In Taxonomy, Genomics and Ecophysiology of Hydrocarbon-Degrading Microbes; Springer International Publishing: Cham, Switzerland, 2017; pp. 1–12. [Google Scholar]
  265. Gutierrez, T.; Singleton, D.R.; Berry, D.; Yang, T.; Aitken, M.D.; Teske, A. Hydrocarbon-degrading bacteria enriched by the Deepwater Horizon oil spill identified by cultivation and DNA-SIP. ISME J. 2013, 7, 2091–2104. [Google Scholar] [CrossRef]
  266. Yakimov, M.M.; Timmis, K.N.; Golyshin, P.N. Obligate oil-degrading marine bacteria. Curr. Opin. Biotechnol. 2007, 18, 257–266. [Google Scholar] [CrossRef]
  267. McGenity, T.J.; Folwell, B.D.; McKew, B.A.; Sanni, G.O. Marine crude-oil biodegradation: A central role for interspecies interactions. Aquat. Biosyst. 2012, 8, 10. [Google Scholar] [CrossRef] [Green Version]
  268. Head, I.M.; Jones, D.M.; Röling, W.F.M. Marine microorganisms make a meal of oil. Nat. Rev. Microbiol. 2006, 4, 173–182. [Google Scholar] [CrossRef]
  269. Barbato, M.; Mapelli, F.; Magagnini, M.; Chouaia, B.; Armeni, M.; Marasco, R.; Crotti, E.; Daffonchio, D.; Borin, S. Hydrocarbon pollutants shape bacterial community assembly of harbor sediments. Mar. Pollut. Bull. 2016, 104, 211–220. [Google Scholar] [CrossRef]
  270. Barbato, M.; Mapelli, F.; Chouaia, B.; Crotti, E.; Daffonchio, D.; Borin, S. Draft genome sequence of the hydrocarbon-degrading bacterium Alcanivorax dieselolei KS-293 isolated from surface seawater in the Eastern Mediterranean Sea. Genome Announc. 2015, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Wang, W.; Shao, Z. The long-chain alkane metabolism network of Alcanivorax dieselolei. Nat. Commun. 2014, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. Li, A.; Shao, Z. Biochemical characterization of a haloalkane dehalogenase DadB from Alcanivorax dieselolei B-5. PLoS ONE 2014, 9. [Google Scholar] [CrossRef] [PubMed]
  273. Scoma, A.; Barbato, M.; Hernandez-Sanabria, E.; Mapelli, F.; Daffonchio, D.; Borin, S.; Boon, N. Microbial oil-degradation under mild hydrostatic pressure (10 MPa): Which pathways are impacted in piezosensitive hydrocarbonoclastic bacteria? Sci. Rep. 2016, 6, 1–14. [Google Scholar] [CrossRef] [Green Version]
  274. Barbato, M.; Scoma, A.; Mapelli, F.; De Smet, R.; Banat, I.M.; Daffonchio, D.; Boon, N.; Borin, S. Hydrocarbonoclastic alcanivorax isolates exhibit different physiological and expression responses to N-dodecane. Front. Microbiol. 2016, 7, 1–14. [Google Scholar] [CrossRef]
  275. Sass, A.M.; McKew, B.A.; Sass, H.; Fichtel, J.; Timmis, K.N.; McGenity, T.J. Diversity of Bacillus-like organisms isolated from deep-sea hypersaline anoxic sediments. Saline Systems 2008, 4, 1–11. [Google Scholar] [CrossRef] [Green Version]
  276. Antunes, A.; Eder, W.; Fareleira, P.; Santos, H.; Huber, R. Salinisphaera shabanensis gen. nov., sp. nov., a novel, moderately halophilic bacterium from the brine-seawater interface of the Shaban Deep, Red Sea. Extremophiles 2003, 7, 29–34. [Google Scholar] [CrossRef]
  277. Palau, M.; Boujida, N.; Manresa, À.; Miñana-Galbis, D. Complete genome sequence of Marinobacter flavimaris LMG 23834T, which is potentially useful in bioremediation. Genome Announc. 2018, 6, 1–2. [Google Scholar] [CrossRef] [Green Version]
  278. Marquez, M.C. Marinobacter hydrocarbonoclasticus Gauthier et al. 1992 and Marinobacter aquaeolei Nguyen et al. 1999 are heterotypic synonyms. Int. J. Syst. Evol. Microbiol. 2005, 55, 1349–1351. [Google Scholar] [CrossRef] [Green Version]
  279. Handley, K.M.; Lloyd, J.R. Biogeochemical implications of the ubiquitous colonization of marine habitats and redox gradients by Marinobacter species. Front. Microbiol. 2013, 4, 1–10. [Google Scholar] [CrossRef] [Green Version]
  280. Gomes, M.B.; Gonzales-Limache, E.E.; Sousa, S.T.P.; Dellagnezze, B.M.; Sartoratto, A.; Silva, L.C.F.; Gieg, L.L.M.; Valoni, E.; Souza, R.S.; Torres, A.P.R.; et al. Exploring the potential of halophilic bacteria from oil terminal environments for biosurfactant production and hydrocarbon degradation under high-salinity conditions. Int. Biodeterior. Biodegradation 2018, 126, 231–242. [Google Scholar] [CrossRef]
  281. Kodama, Y.; Stiknowati, L.I.; Ueki, A.; Ueki, K.; Watanabe, K. Thalassospira tepidiphila sp. nov., a polycyclic aromatic hydrocarbon-degrading bacterium isolated from seawater. Int. J. Syst. Evol. Microbiol. 2008, 58, 711–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Budiyanto, F.; Thukair, A.; Al-Momani, M.; Musa, M.M.; Nzila, A. Characterization of Halophilic Bacteria Capable of Efficiently Biodegrading the High-Molecular-Weight Polycyclic Aromatic Hydrocarbon Pyrene. Environ. Eng. Sci. 2018, 35, 616–626. [Google Scholar] [CrossRef] [Green Version]
  283. Castillo-Carvajal, L.C.; Sanz-Martín, J.L.; Barragán-Huerta, B.E. Biodegradation of organic pollutants in saline wastewater by halophilic microorganisms: A review. Environ. Sci. Pollut. Res. 2014, 21, 9578–9588. [Google Scholar] [CrossRef] [PubMed]
  284. Paniagua-Michel, J.; Babu, Z. Fathepure Microbial Consortia and Biodegradation of Petroleum Hydrocarbons in Marine Environments. In Microbial Action on Hydrocarbons; Kumar, V., Kumar, M., Prasad, R., Eds.; Springer Nature: Singapore, 2019; pp. 1–20. [Google Scholar]
  285. Erdoǧmuş, S.F.; Mutlu, B.; Korcan, S.E.; Güven, K.; Konuk, M. Aromatic hydrocarbon degradation by halophilic archaea isolated from ÇamaltI Saltern, Turkey. Water. Air. Soil Pollut. 2013, 224. [Google Scholar] [CrossRef]
  286. Al-Mailem, D.M.; Eliyas, M.; Radwan, S.S. Oil-bioremediation potential of two hydrocarbonoclastic, diazotrophic Marinobacter strains from hypersaline areas along the Arabian Gulf coasts. Extremophiles 2013, 17, 463–470. [Google Scholar] [CrossRef] [PubMed]
  287. Edbeib, M.F.; Wahab, R.A.; Huyop, F. Halophiles: Biology, adaptation, and their role in decontamination of hypersaline environments. World J. Microbiol. Biotechnol. 2016, 32, 1–23. [Google Scholar] [CrossRef]
Figure 1. Global distribution of deep hypersaline anoxic basins (DHABs) (a). Locations and corresponding names of DHABs identified in the Red Sea (b1b2), the Gulf of Mexico (c), and the Mediterranean Sea (d).
Figure 1. Global distribution of deep hypersaline anoxic basins (DHABs) (a). Locations and corresponding names of DHABs identified in the Red Sea (b1b2), the Gulf of Mexico (c), and the Mediterranean Sea (d).
Marinedrugs 18 00091 g001
Figure 2. Simplified vertical section of a DHAB. The transition from the overlying seawater to the brine is commonly referred to as the halocline or brine–seawater interface, which is characterized by gradients of temperature, salinity, pH, and dissolved oxygen (A); the main biogeochemical processes taking place within the halocline are shown in (B). From left to right, the manganese cycle, the sulfate reduction and sulfide oxidation cycle, the methanogenesis and aerobic (anaerobic) methane oxidation cycle, and the anammox and denitrification cycle that occur in the halocline are shown [8,21,23,28,29,30]. DOC: Dissolved Organic Carbon.
Figure 2. Simplified vertical section of a DHAB. The transition from the overlying seawater to the brine is commonly referred to as the halocline or brine–seawater interface, which is characterized by gradients of temperature, salinity, pH, and dissolved oxygen (A); the main biogeochemical processes taking place within the halocline are shown in (B). From left to right, the manganese cycle, the sulfate reduction and sulfide oxidation cycle, the methanogenesis and aerobic (anaerobic) methane oxidation cycle, and the anammox and denitrification cycle that occur in the halocline are shown [8,21,23,28,29,30]. DOC: Dissolved Organic Carbon.
Marinedrugs 18 00091 g002
Figure 3. Phylogenetic tree of bacterial and archaeal strains isolated from DHABs. The tree was built using 16S rRNA gene sequences from [66,83,84,85] and phylogenetically close 16s rRNA sequences from the SILVA database v132.
Figure 3. Phylogenetic tree of bacterial and archaeal strains isolated from DHABs. The tree was built using 16S rRNA gene sequences from [66,83,84,85] and phylogenetically close 16s rRNA sequences from the SILVA database v132.
Marinedrugs 18 00091 g003
Table 1. Minimum and maximum values of the main physicochemical variables observed among DHABs.
Table 1. Minimum and maximum values of the main physicochemical variables observed among DHABs.
Environmental ParametersRangesDHABsLocationReferences
TemperatureMin: 14 °C
Max: 68 °C
La Medee
Atlantis II
Mediterranean Sea
Red Sea
[31]
[32]
DepthMin 630 m
Max: 3580 m
GC233
Discovery
Gulf of Mexico
Mediterranean Sea
[33]
[17]
Na+Min: 1751 mM
Max 5300 mM
GC233
Tyro
Gulf of Mexico
Mediterranean Sea
[24]
[34]
Cl-Min: 2092 mM
Max: 10,154.3 mM
GC233
Discovery
Gulf of Mexico
Mediterranean Sea
[24]
[17]
Mg2+Min: 8.7 mM
Max: 5143 mM
GB425
Discovery
Gulf of Mexico
Mediterranean Sea
[24]
[17]
K+Min: 17.2 mM
Max: 471 mM
Orca
La Medee
Gulf of Mexico
Mediterranean Sea
[35]
[31]
Ca2+Min: 1 mM
Max: 150 mM
Discovery, Kyros
Atlantis II
Mediterranean Sea
Red Sea
[17]
[36]
SO42−Min: <1 mM
Max: 333.1 mM
GB425; GC233
L’Atalante
Gulf of Mexico
Mediterranean Sea
[24]
[17]
SulfideMin: 0.002 mM
Max: 16 mM
GC233
Urania
Gulf of Mexico
Mediterranean Sea
[24]
[16]
Table 2. Bioactive molecules for pharmaceutical use produced by prokaryotes directly isolated from DHABs and promising bioactive molecules produced by prokaryotic taxa which have been identified in DHABs and isolated from other marine systems.
Table 2. Bioactive molecules for pharmaceutical use produced by prokaryotes directly isolated from DHABs and promising bioactive molecules produced by prokaryotic taxa which have been identified in DHABs and isolated from other marine systems.
Marine ProkaryotesProductBioactivityEnvironmental SourcesRef.
Alteromonas macleodiiDithiolopyrroloneAntibiotic and antitumorErba and Nereus DHABs[84,104]
Alteromonas sp. B-10-31Marinostatins B-1, C1, and C2Serine protease inhibitorCoastal seawater[105]
Bacillus sp.Macrolactins A–FCytotoxic, antimicrobial, antiviral Deep sea[106,107,108,109]
Bacillus haloduransEnfuvirtideAntiviralNereus DHAB[84,110]
Bacillus MK-PNG-276ALoloatins A–D AntimicrobialGreat barrier reef[111]
Bacillus sp.Bogorol AAntimicrobialSeawater [112]
Bacillus sp. CND-914Halobacillin AntitumorDeep-sea sediments[113]
Bacillus sp. MIX-62 Mixirins A–C Antitumor [114]
Bacteroidetes rapidithrix HC35 Ariakemicins A and BAntimicrobial, cytotoxicSea mud[115]
Erythrobacter sp.Erythrazoles A and B
Erythrolic acids A–E
CytotoxicMangrove sediments[116,117]
Halobacteroides lacunaris TB21R-LPS ImmunomodulatorThetis DHAB[118]
Halomonas LOB-5Loihichelins A–Fn.a.Deep sea hydrothermal vents [119]
Halomonas meridianan.a.AntitumorNereus DHAB[84]
Halomonas sp. GWS-BW-H8hM3-(4′-Hydroxyphenyl)-4-phenylpyrrole-2,5-dicarboxylic acid (HPPD-1 and HPPD-2)CytotoxicSeawater[120]
Halomonas sp. GWS-BW-H8hM2-Amino-6-hydroxyphenoxazin-3-one2-Amino-8-benzoyl-phenoxazin-3-one2-Amino-8-(4-hydroxybenzoyl)-6-hydroxyphenoxazin-3-oneAntimicrobial, cytotoxicSeawater[121]
Pseudoalteromonas carrageenovora IAM 12662LPSAntiviral Erba DHAB[84,122]
Pseudoalteromonas haloplanktis TAC125PeptidesAntioxidantAntarctic coastal sea water[123]
Pseudoalteromonas mariniglutinosan.a.AntitumorErba and Nereus DHABs[84]
Pseudoalteromonas rava SANK 73390Thiomarinols A–H and JAntimicrobialSeawater[124,125]
Streptomyces aureoverticillatus (NPS001583)AureoverticillactamAntitumorMarine sediments[126]
Streptomyces C42ChampacyclinAntimicrobialDeep sea [127]
Streptomyces CNH-990Marmycins A and B CytotoxicSeawater[128,129]
Streptomyces drozdowiczii SCSIO 10141Marfomycins A, B, and EAnti-infectiveDeep sea[130]
Streptomyces drozdowiczii NTK 97FrigocyclinoneAntimicrobialAntarctica[131]
Streptomyces Merv 8102EssramycinAntimicrobialMarine animals, plants, and sediments[132]
Streptomyces niveus SCSIO 3406MarfuraquinocinsCytotoxic antimicrobialDeep sea [133]
Streptomyces scopuliridis SCSIO ZJ46Desotamide BAntimicrobialDeep-sea sediments[134]
Streptomyces sioyaensis SA-1758Altemicidin Cytotoxic, antimicrobialSea mud[135]
Streptomyces sp. 12A35Lobophorins H and IAntimicrobialDeep sea [136]
Streptomyces sp. ART5ArticosideCytotoxic, Arctic deep sea[137]
Streptomyces sp. CNB-982Cyclomarins A–C anti-inflammatoryMarine sediments[138,139]
Streptomyces sp. CNQ-418Marinopyrroles A–FAntimicrobial, cytotoxic, anti-apoptoticDeep-sea sediments[140,141]
Streptomyces sp. CNQ-85Daryamides A–C
(2E,4E)-7-Methylocta-2,4-dienoic acid amide 26
Antitumor, antifungalSeawater[142]
Streptomyces sp. CNR-698Ammosamides A–DCytotoxicDeep sea[143,144,145]
Streptomyces sp. M045Chinikomycins A and BAntitumorSeawater[146]
Streptomyces sp. MDF-04-17-069Tartrolon DCytotoxicMarine sediments[147]
Streptomyces sp. Mei37Mansouramycins A–D Antimicrobial, cytotoxicMarine sediments [148]
Streptomyces sp. NTK 935BenzoxacystolAntiproliferativeDeep sea[149]
Streptomyces sp. SCSIO 03032Spiroindimicins A–DAntitumorDeep sea[150]
Streptomyces sp. SCSIO 11594Dehydroxyaquayamycin, Marangucycline B Antibacterial, antitumorDeep sea [151]
Streptomyces xiamenensis M1-94PXiamenmycin C and DAnti-fibroticDeep-sea sediments[152]
Streptomycete sp.Piperazimycins A–CAntitumorMarine sediments[153]
Zunongwangia profunda SM-A87EPSAntioxidantNereus DHAB[154]
Table 3. DHAB microbiome as a source of polyextremozymes. The bacterial and archaeal species marked with an asterisk have been isolated from DHABs, whereas the other genera are potentially producers of extremozymes because these have identified from DHABs (but not cultured thus, being isolated from marine and/or other extreme environments).
Table 3. DHAB microbiome as a source of polyextremozymes. The bacterial and archaeal species marked with an asterisk have been isolated from DHABs, whereas the other genera are potentially producers of extremozymes because these have identified from DHABs (but not cultured thus, being isolated from marine and/or other extreme environments).
EnzymeBiological SourceSpecific AdaptationsFunction and/or ApplicationsRef.
Aldehyde dehydrogenase
(EC 1.2.1.3–7)
Bacillus halodurans from Nereus interface; Atlantis II Red Sea brine pool; Cytophaga sp. KUC-1 from Antarctic seawater and Halobacterium salinarumSlight halophile; thermo- and psychrophilicBiotransformation of a large number of drugs and other xenobiotics generates aldehydes as intermediates or as products resulting from oxidative deaminations[172,173,174,175]
Protease
(EC 3.4.21–25)
Salinivibrio costicola* and Pseudoalteromonas ruthenica* from Erba DHAB. Bacillus circulans BM15 and PseudoAlteromonas sp. 129-1. Bacillus sp. NPST-AK1, Halobacterium halobium (ATCC 43214), Bacillus licheniformis, Bacillus halophilus, Pseudoalteromonas strain EB27, Halomonas meridiana DSM 5425, Bacillus sp. (Ve2-20-91 (HM047794)), and Bacillus caseinilyticus Haloalkaliphilic and thermotolerant alkalineProtein hydrolysis finds a broad variety of potential applications in diverse biotechnological processes such as in the feed, food, pharmacology (anticancer and antihemolytic activity) and cosmetic (keratin-based preparation) industries, and cleaning processes (e.g., detergent additive)[176,177,178,179,180,181,182,183,184,185]
Cellulase
(EC 3.2.1.4)
Cytophaga hutchinsonii, Halorhabdus tiamatea from Shaban DHAB, Bacillus sp. SR22 from seawater, Bacillus sp., Vibrio sp., Rhodococcus sp., Clostridium and Streptomyces from mangrove Halorhabdus utahensis from Great Salt LakeHalo-alkali tolerant and thermotolerantBreakdown of cellulose-producing polysaccharides; potential application in the food, animal feed, beer and wine, textile and laundry, and pulp and paper industries, agriculture, biofuel, pharmaceutical industries, and waste management[186,187,188,189,190,191]
Chitinase (EC 3.2.1.14), chitin deacetylase (EC 3.5.1.41)Bacillus thuringiensis HBK-51 from soil. PseudoAlteromonas sp. DC14, Vibrio cholerae, Vibrio parahaemolyticus, and Arthrobacter sp. AW19M34-1 from seawaterHalo-alkali tolerant and thermotolerantHydrolysis of chitin and hence N-acetyl chitobiose production which in turn can be useful in fermentation research and biomedicine. There have also been applications in the cosmetic and pharmaceutic fields [192,193,194]
Esterase
(EC 3.1.1.1)
Zunongwangia profunda* from Atlantis II and Nereus interface and brine pools. Alcanivorax dieselolei B5(T) from Erba interface. Bacillus cereus AGP-03 from hot spring. Archaeoglobus fulgidusThermo-halotolerant and metal resistant; cold-active and organic solvent-tolerantLeather manufacturing, flavor development in the dairy industry, oil biodegradation, and the synthesis of pharmaceuticals and chemicals[195,196,197,198,199,200]
Ferredoxin oxidoreductase
(EC 1.2.7.1)
Halorhabdus tiamatea SARL4BT* from Shaban DHAB. Desulfovibrio sp. from Atlantis II DHAB. Methanosarcina barkeriLow-oxygen tolerantOxidation/reduction processes which are applied in the asymmetric oxyfunctionalization of steroids and other pharmaceuticals, synthesis and modification of polymers, oxidative degradation of pollutants, oxyfunctionalization of hydrocarbons, and the construction of biosensors for diverse clinical applications[189,201,202]
Lipase
(EC 3.1.1.3)
Idiomarina sp. W33, HaloBacillus sp., and Archaeoglobus fulgidus. Marinobacter alkaliphilus ABN-IAUF-1. Bacillus sp., Arthrobacter sp., Pseudomonas sp., and Psychrobacter sp. from Antarctic marine sediments. Oceano Bacillus sp. PUMB02 from seawaterHalo- alkalitolerant and hyperthermophilicHydrolysis of acylglycerols to release fatty acids and lower acylglycerols or glycerol. Lipase enzymes are exploited in the food, beverage, detergent, biofuel production, animal feed, textiles, leather, paper processing, and cosmetic industries[203,204,205,206,207,208]
Mercuric reductase
(EC 1.16.1.1)
Atlantis II deep-sea brine. Chromohalobacter israelensis* from Erba and Atlantis II DHABs. Bacillus firmus* from Discovery DHABExtreme halophilic and thermophilicThis enzyme can convert toxic mercury ions into relatively inert elemental mercury. It is very useful in waste-water treatments[209,210]
Nitrilase
(EC 3.5.5.1)
Red Sea Atlantis II brine Thermostable and heavy metal tolerant Nitrilase can hydrolyze a single cyano group in dinitriles or polynitriles, yelding cyanocarboxilic acids, which are used in different kinds of industries, including the food and pharmacology industries; also used for bioremediative purposes[211]
Pullulanase
(EC 3.2.1.41)
Bacillus sp. and Streptomyces sp.AlkaliphilicUtilized to hydrolyze the α-1,6 glucosidic linkages in starch, enabling a complete and efficient conversion of the branched polysaccharides into small fermentable sugars during the saccharification process [212]
Xylanase (EC 3.2.1.8) and β-Xylosidase
(EC 3.2.1.37)
Staphylococcus sp., Arthrobacter sp., Streptomyces sp., and Vibrio sp. XY-214 from seawater. Oceanospirillum linum CL8 and Halorhabdus utahensis from Great Salt Lake. Halorhabdus tiamatea SARL4BT* from Shaban DHAB. Pseudoalteromonas mariniglutinosa* from Erba and Nereus DHAB. Marinimicrobium haloxylanilyticum* from Kebrit DHAB. Zunongwangia profunda* from Nereus and Atlantis II DHABs. Halomonas meridiana* from Bannock, Erba, and Nereus DHABs. Bacillus halodurans* from Nereus interface Alkali-halotolerant and psychrophilicCommercial exploitation in the areas of the food, feed, and paper and pulp industries; also used to increase sugar recovery from agricultural residues for biofuel production[189,213,214,215,216,217,218]
α-agarase
(EC 3.2.1.158) and
β-agarase
(EC 3.2.1.81)
Alteromonas macleodii* from Erba, Discovery, and Nereus DHABs. Alteromonas sp. GNUM-1, Alteromonas agarlyticus, Alteromonas sp. strain C-1, Vibrio sp. PO-303, Altermonas sp. SY37-12, and Cytophaga flevensis from seawater and marine sedimentsModerate halophileDegradation of agar-degrading bacteria used as oriental food; wide applications in the food industry, cosmetics, and medical fields, and as a tool enzyme for biological, physiological, and cytological studies[219,220,221]
α-amylase
(EC 3.2.1.1)
PontiBacillus chungwhensis* from Discovery DHAB. Halomonas meridiana* from Nereus, Erba, and Bannock DHABs. Zunongwangia profunda* from Atlantis II and Nereus DHABs. Cytophaga sp. HaloBacillus sp., Bacillus sp. GM8901, Bacillus sp. TSCVKK, and Methanococcus jannaschii. Halobacterium sp. from hypersaline environment. Alteromonas haloplanctis from Antarctic seawaterModerate halophile and alkali- tolerant; hyperthermophilic α-amylase has implications in the food, pharmaceutical, and chemical industries; multifunctional amylase exhibits transglycosylation and hydrolysis activities to produce isomaltooligosaccharides, maltooligosaccharides and glucose[222,223,224,225,226,227,228,229,230,231,232]
β-glucosidases
(EC 3.2.1.21)
Halorhabdus tiamatea SARL4BT* from Shaban DHAB. Alteromonas sp. L82 from the Mariana Trench. Cytophaga hutchinsoniiLow-oxygen tolerant, cold-adapted, and salt-tolerantβ-glucosidases convert cellobiose and short cellodextrins into glucose. β-glucosidases are widely used in the production of biofuels and ethanol from cellulosic agricultural wastes, in the production of wine, and in the flavor industry. They can cleave phenolic and phytoestrogen glucosides from fruits and vegetables for extracting medicinally important compounds and enhancing the quality of beverages[189,191,233,234]
κ-Carragenases
(EC 3.2.1.83)
Pseudoalteromonas carrageenovora* from Erba sediments. Bacillus sp. Alteronomonas sp., Cytophaga sp., and PseudoAlteromonas sp. Pseudomonas sp., Vibrio sp. NJ-2, and Vibrio parahaemolyticus from seawaterAlkali-halotolerantProduction of oligosaccharides with potential applications in the biomedical field, in bioethanol production, in the textile industry, and as a detergent additive[235,236,237]
Cyclodextrin glycosyltransferase
(EC 2.4.1.19)
Bacillus lehensis* from Discovery DHABAlkali-halotolerantCyclodextrins produced by this enzyme have broad, non-toxic applications in the pharmaceutical, cosmetic, and food industries[66,238]

Share and Cite

MDPI and ACS Style

Varrella, S.; Tangherlini, M.; Corinaldesi, C. Deep Hypersaline Anoxic Basins as Untapped Reservoir of Polyextremophilic Prokaryotes of Biotechnological Interest. Mar. Drugs 2020, 18, 91. https://doi.org/10.3390/md18020091

AMA Style

Varrella S, Tangherlini M, Corinaldesi C. Deep Hypersaline Anoxic Basins as Untapped Reservoir of Polyextremophilic Prokaryotes of Biotechnological Interest. Marine Drugs. 2020; 18(2):91. https://doi.org/10.3390/md18020091

Chicago/Turabian Style

Varrella, Stefano, Michael Tangherlini, and Cinzia Corinaldesi. 2020. "Deep Hypersaline Anoxic Basins as Untapped Reservoir of Polyextremophilic Prokaryotes of Biotechnological Interest" Marine Drugs 18, no. 2: 91. https://doi.org/10.3390/md18020091

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop