Next Article in Journal
Anthracenedione Derivatives as Anticancer Agents Isolated from Secondary Metabolites of the Mangrove Endophytic Fungi
Next Article in Special Issue
Biological Activities of Aqueous and Organic Extracts from Tropical Marine Sponges
Previous Article in Journal / Special Issue
Synthesis of the Marine Pyrroloiminoquinone Alkaloids, Discorhabdins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Marine Drugs from Sponge-Microbe Association—A Review

by
Tresa Remya A. Thomas
,
Devanand P. Kavlekar
and
Ponnapakkam A. LokaBharathi
*
Biological Oceanography, National Institute of Oceanography, Dona Paula, Goa, Pin-403004, India
*
Author to whom correspondence should be addressed.
Mar. Drugs 2010, 8(4), 1417-1468; https://doi.org/10.3390/md8041417
Submission received: 24 February 2010 / Revised: 13 April 2010 / Accepted: 19 April 2010 / Published: 22 April 2010
(This article belongs to the Special Issue Bioactive Compound from Marine Sponges)

Abstract

:
The subject of this review is the biodiversity of marine sponges and associated microbes which have been reported to produce therapeutically important compounds, along with the contextual information on their geographic distribution. Class Demospongiae and the orders Halichondrida, Poecilosclerida and Dictyoceratida are the richest sources of these compounds. Among the microbial associates, members of the bacterial phylum Actinobacteria and fungal division Ascomycota have been identified to be the dominant producers of therapeutics. Though the number of bacterial associates outnumber the fungal associates, the documented potential of fungi to produce clinically active compounds is currently more important than that of bacteria. Interestingly, production of a few identical compounds by entirely different host-microbial associations has been detected in both terrestrial and marine environments. In the Demospongiae, microbial association is highly specific and so to the production of compounds. Besides, persistent production of bioactive compounds has also been encountered in highly specific host-symbiont associations. Though spatial and temporal variations are known to have a marked effect on the quality and quantity of bioactive compounds, only a few studies have covered these dimensions. The need to augment production of these compounds through tissue culture and mariculture has also been stressed. The reviewed database of these compounds is available at www.niobioinformatics.in/drug.php.

1. Introduction

Sponges (Phylum: Porifera) are evolutionarily ancient metazoans that have existed for 700–800 million years. They not only populate the tropical oceans in great abundance but also occur in temperate waters and even in freshwater [1,2]. Marine sponges are widely distributed from intertidal zones to thousands of meters deep in the ocean [3]. They are simple multicellular invertebrates attached to solid substrates in benthic habitats. Sponges are filter feeders, having numerous tiny pores on their surface, which allow water to enter and circulate through a series of canals where microorganisms and organic particles are filtered out and eaten [4]. There are mainly three classes of sponges, namely the Calcarea (five orders and 24 families), Demospongiae (15 orders and 92 families) and Hexactinellida (six orders and 20 families). So far about 15,000 species of sponges have been described, but their true diversity may be higher [5]. Most of them occur in the marine environment and only about 1% inhabit freshwater [6]. Most of the species are placed under the class Demospongiae. Since sponges are simple and sessile organisms; during evolution they have developed potent chemical defensive mechanism to protect themselves from competitors and predators as well as infectious microorganisms. Studies show that secondary metabolites in sponges play a crucial role in their survival in the marine ecosystem [7]. These natural products have interesting biomedical potential, pharmaceutical relevance and diverse biotechnological applications [4,813]. The biomedical and pharmaceutical importances of these compounds are attributed to their antiviral, antitumor, antimicrobial and general cytotoxic properties [14]. Interestingly, out of the 13 marine natural products that are currently under clinical trials as new drug candidates, 12 are derived from invertebrates. Among them, Porifera remains the most important phylum, as it provides a greater number of natural products, especially novel pharmacologically active compounds [15,16]. Biochemical characteristics seem to be useful taxonomic markers and good indicators of sponge phylogeny [17]. The diversity of biochemical properties of sponges has been demonstrated by the continued discovery of novel compounds, having pharmacological properties [18]. These investigations started with the pioneering work of Bergmann on the extraction of novel bioactive nucleosides from the sponge Tectitethya crypta (formerly Cryptotethya crypta) [19]. The chemical diversity of secondary metabolites isolated from sponges includes amino acids, nucleosides, macrolides, porphyrins, terpenoids, aliphatic cyclic peroxides and sterols [7]. Sponges are well known to be hosts for a large community of microorganisms, which comprise a significant percentage (up to 50–60%) of the biomass of the sponge host [20,21]. The role of these diverse microbes in sponge biology varies from source of nutrition to mutualistic symbiosis with the sponge [22]. Based on bacterial community studies employing molecular methods such as Denaturing Gradient Gel Electrophoresis (DGGE), 16S rRNA gene sequencing and Fluorescence In Situ Hybridization (FISH), it has been recognized that the sponge-associated bacterial community consists of at least ten bacterial phyla such as Proteobacteria, Nitrospira, Cyanobacteria, Bacteriodetes, Actinobacteria, Chloroflexi, Planctomycetes, Acidobacteria, Poribacteria and Verrucomicrobia besides members of the domain Archaea [1,2330]. Other symbiotic microbial populations that inhabit sponges are fungi and microalgae. Little is known about viruses in sponges, although virus-like particles have been observed in cell nuclei of Aplysina (Verongia) cavernicola [32]. There are two pathways through which a developing sponge acquires bacterial symbionts. The first one is by selective absorption of specific bacteria from the large diversity of bacteria in the surrounding water column that passes through the sponge during filter feeding. The second one is by vertical transmission of symbionts through the gametes of the sponge by inclusion of the bacteria in the oocytes or larvae [33].
Symbiotic functions that have been attributed to microbial associates include nutrient acquisition, stabilization of sponge skeleton, processing of metabolic waste and secondary metabolite production [1]. It is hypothesized that symbiotic marine microorganism harboured by sponges are the original producers of these bioactive compounds [12,3335]. The first experimental evidence supporting this hypothesis was derived from the work of Faulkner et al. [27], who investigated the localization of natural products within sponge-microorganism association. For this purpose, cell populations within sponge samples were separated by differential centrifugation and the fractions obtained were analyzed chemically. By this approach, it was possible to locate the cytotoxic macrolide swinholide A and the peptide theopalauamide in the heterotrophic unicellular bacteria and in the filamentous heterotrophic bacteria, respectively. Both the bacterial strains were isolated from the sponge Theonella swinhoei.
Microbial associates of sponges gained significance as source of bioactive compounds only when a remarkable similarity was found between those compounds isolated predominantly from sponges and those found in terrestrial organism of entirely different taxa [36]. Likewise, similarities between the structures of mycalamide A & B from the marine sponge Mycale hentscheli, collected in Dunedin Harbour (New Zealand) and pederin, a toxin originally isolated from the Paederus beetle in South America was recognized by Perry et al. [37]. Mycalamides have been reported to be potent inhibitors of protein synthesis and were recently found to cause apoptosis [38]. Thus, it indicates that at least some of the bioactive secondary metabolites isolated from sponges are produced by functional enzyme clusters, originated from the sponges and/or their associated microorganisms [39]. It is now known that polybrominated biphenyl ether antibiotics isolated from the sponge Dysidea herbacea (Demospongiae) are actually produced by the endosymbiotic cyanobacterium Oscillatoria spongeliae [35,40]. Molecular methods (e.g; rDNA, DGGE and FISH) have revealed the association of a variety of unculturable bacteria and Archaea in sponges. It has recently been demonstrated that sponge isolates with antimicrobial activity are numerically very abundant in the genus Pseudoalteromonas and the group of α-Proteobacteria [7,41] and Actinobacteria [42]. As infectious microorganisms evolve and develop resistance to existing pharmaceuticals, marine sponges provide novel leads against bacterial, viral, fungal and parasitic diseases [39]. Thus, it is extremely relevant to highlight the therapeutic properties of various secondary metabolites synthesized by the microbial flora inhabiting sponges. In this review, an effort has been made to relate the biomedical significance of secondary metabolites of sponge-microbial association, which were discovered so far and their richness in different sponge taxa. It is also important to understand their ecological distribution in space and time so as to enable harnessing these compounds in an optimal and sustainable manner.
No bioactive compounds have been reported from microbes associated with sponge families such as Agelasidae, Astroscleridae, Calthropellidae, Geodiidae, Pachastrellidae, Thrombidae, Dictyodendrillidae, Acanthochaetetidae, Alectonidae, Hemiasterellidae, Placospongiidae, Polymastiidae, Spirastrellidae, Stylocordylidae, Tethyidae, Timeidae, Trachycladidae, Bubaridae, Dictyonellidae, Heteroxyidae, Halisarcidae, Calcifibrospongiidae, Phloeodictyidae, Lubomirskiidae, Malawispongiidae, Metaniidae, Metschnikowiidae, Palaeospongillidae, Potamolepiidae, Spongillidae, Spongillina incertae sedis, Plakinidae, Azoricidae, Corallistidae, Desmanthidae, Isoraphiniidae, Lithistida incertae sedis, Macandrewiidae, Phymaraphiniidae, Phymatellidae, Pleromidae, Scleritodermidae, Siphonidiidae, Vetulinidae, Latrunculiidae, Microcionidae, Rhabderemiidae, Cladorhizidae, Desmacellidae, Esperiopsidae, Guitarridae, Hamacanthidae, Merliidae, Podospongiidae, Chondropsidae, Coelosphaeridae, Crambeidae, Crellidae, Dendoricellidae, Desmacididae, Hymedesmiidae, Iotrochotidae, Phellodermidae, Tedaniidae, Samidae and Spirasigmidae of the class Demospongiae; Baeriidae, Lepidoleuconidae, Trichogypsiidae, Achramorphidae, Amphoriscidae, Grantiidae, Heteropiidae, Jenkinidae, Lelapiidae, Leucosoleniidae, Sycanthidae, Sycettidae, Minchinellidae, Petrobionidae, Clathrinida incertae sedis, Clathrinidae, Leucaltidae, Leucascidae, Levinellidae, Soleneiscidae, Lelapiellidae, Murrayonidae, Paramurrayonidae of the class Calcarea. There are no reports of microbially originated bioactive compounds from the class Hexactinellida.

2. Sponges and Associated Microbes Involved in Drug Production

2.1. Class: Demospongiae

2.1.1. Order: Astrophorida

Family: Ancorinidae
l,l-Diketopiperazine known as cyclo-(l-Pro-l-Phe), showing moderate antimicrobial activity was isolated from the bacterium Alcaligenes faecalis A72, which was found in association with the South China Sea sponge Stelletta tenuis [42]. The sponge Stelletta tenuis is known for harbouring large number of cultivable bacterial diversity, including α-, γ-, δ-Proteobacteria, Bacteroidetes, Firmicutes and Actinobacteria [23,42]. A marine fungus of the class Hyphomycetes was isolated from the Indo-Pacific sponge Jaspis aff. johnstoni. Fermentation of this marine culture led to the isolation of the tricyclic sesquiterpenes coriolin B, dihydrocoriolin C as well as the novel chloriolines A-C. Coriolin B and dihydrocoriolin C were earlier isolated from the terrestrial wood-rotting basidiomycete Coriolus consors. Coriolin B exhibited strong inhibition of human breast and CNS cell lines with IC50 values of 0.7 μg (breast) and 0.5 μg (neuroblastoma) [9,4345].

2.1.2. Order: Chondrosida

Family: Chondrillidae
Seven new fungal polyketides were isolated from the mycelium extract of the fungus Penicillium rugulosum, derived from the sponge Chondrosia reniformis (Elba, Italy). They include prugosenes A1–A3, B1, B2, C1 and C2. These compounds can be used as templates for new anti-infectives [4648].

2.1.3. Order: Dendroceratida

Family: Darwinellidae
The sponge Dendrilla nigra is a rich source of cultivable marine actinomycetes. Investigations on a sponge specimen collected from the Vizhinjam coast (west coast of India) revealed that Micromonospora-Saccharomonospora-Streptomyces group was the major cultivable actinobacteria found in the sponge [49]. The species Streptomyces dendra sp. nov. MSI051 isolated from Dendrilla nigra from the same coast exhibited a broad spectrum of antibacterial activity. The host sponge, as well as the associated bacterial symbiont MSI051, contained high levels of PLA2 (Phospholipase A2) [50]. Since PLA2 is a well-established antibacterial protein in the defense system of higher animals, its presence in the sponge-associated bacteria may indicate an integrated functional role in the host defense system [51]. Another strain, Streptomyces sp. BLT7 isolated from Dendrilla nigra obtained from Kanyakumari (south east coast of India) also showed potential antibacterial activity in their extracellular products [52,53]. A number of actinobacterial strains were also obtained from Dendrilla nigra, collected from the southwest coast of India. Among eleven heterotrophic actinobacteria isolated from one specimen, Nocardiopsis dassonvillei MAD08 was prominent in its antibacterial and anticandidal activity against the multidrug resistant pathogenic microbial strains. The antibacterial activity was assigned to the presence of 11 compounds and the anticandidal activity to a single protein. The uniqueness of this strain is reflected in the expression of both organic solvent (antibacterial) and water soluble (antifungal) antimicrobial compounds. In future, this may lead the way towards large-scale profitable production of antimicrobials from Nocardiopsis dassonvillei MAD08 [53]. The above studies reflect the consistent production of antimicrobial compounds by the actinobacteria harbouring individuals of Dendrilla nigra from south west coast of India.

2.1.4. Order: Dictyoceratida

2.1.4.1. Family: Dysideidae

Many marine sponges, especially the tropical ones, form symbioses with algae and often become net primary producers. Although associations with cyanobacteria are the most common, such partnership has also been observed with chlorophytes, rhodophytes, dinoflagellates and diatoms [54,55]. A variety of marine sponges hold cyanobacteria as autotrophic symbionts, which are known to contribute to nutrition of host through extracellular lysis and phagocytosis, with possible glycogen reutilization by sponge cells. Cyanobacteria transfer glycerol and organic phosphate to sponge tissue, as derivatives of these compounds are known to support several basic metabolic pathways. Moreover, symbiotic cyanobacteria appear to be capable of fixing nitrogen [55]. The tropical marine shallow water sponge Lamellodysidea herbacea (formerly Dysidea herbacea) which is common throughout the Indo-Pacific, is always found to harbour filamentous non-heterocystous cyanobacterium Oscillatoria spongeliae. It occurs intercellularly in large numbers up to 20% of the symbiotic associations’ volume and 30–50% of the sponge tissue volume [10,57]. These cyanobacterial symbionts have been reported to be responsible for the production of a wide array of secondary metabolites by the sponge [55]. Nuclear magnetic resonance analysis of the symbiont cell preparations from the specimen of Lamellodysidea herbacea obtained from Great Barrier Reef, Australia showed that they usually contain the chlorinated diketopiperazines, dihydrodysamide C and didechlorodihydrodysamide C, which are characteristic metabolites of the sponge-symbiont association [56,57]. Since diketopiperazines (DKPs) are a common motif in various biologically active natural products, they may be useful scaffolds for the rational design of receptor probes and therapeutic agents [58]. Symbiotic microorganisms of Dysidea sp. can synthesize physiologically active compounds which belong to the group of brominated diphenyl ethers. Vibrio sp. associated with Dysidea specimen collected near the islands of Tutuila and Ofu (Eastern Samoa) synthesize cytotoxic and antibacterial tetrabromodiphenyl ethers [59]. A specimen of Lamellodysidea herbacea collected from the Republic of Palau (Caroline Island, Western Pacific Ocean) yielded a polybrominated biphenyl ether such as 2-(2′,4′-dibromophenyl)-4,6-dibromophenol. The compound was deposited as conspicuous crystals throughout the sponge tissue. The cyanobacteria Oscillatoria spongeliae was also observed as endosymbiont in the sponge mesohyl. They were separated from the sponge cells and heterotrophic bacteria by flow cytometry. Coupled gas chromatography-mass spectrometry and protein nuclear magnetic resonance revealed that the real source of the compound was the cyanobacteria Oscillatoria spongeliae. The polybrominated metabolites produced by the cyanobacteria are excreted into the surrounding aqueous medium in which they are not soluble, and therefore crystallize. Thus considerable amount of brominated metabolites are seen as crystalline material in the sponge mesohyl, with only a relatively small amount in the cyanobacteria. Polybrominated biphenyl ethers from Lamellodysidea herbacea are active against both Gram-, Gram+ bacteria and unicellular marine cyanobacteria. The compound, 2-(2′,4′-dibromophenyl)-4,6-dibromophenol showed antibacterial activity against Staphylococcus aureus, Escherichia coli, Bacillus subtilis etc. The apparent general toxicity of polybrominated compounds particularly to prokaryotes is beneficial to the association Lamellodysidea herbacea-Oscillatoria spongeliae. This association is more resistant to these compounds [40]. Lamellodysidea herbacea is one of the established model systems for addressing the question as to whether sponge metabolites are produced by the symbiotic bacterium or the host itself [60]. An unknown bacterium associated with the marine sponge Dysidea avara, collected from Adriatic Sea was found to produce the compound 2-methylthio-1,4-naphthoquinone. This compound showed strong antiangiogenic and antimicrobial properties [61]. 16S rDNA analysis revealed that the bacterial strain shares 99% identity to the α-Proteobacteria MBIC3368 [62].

2.1.4.2. Family: Irciniidae

Marine sponges in the genus Ircinia are known to be good sources of secondary metabolites having biological activities [61,63,64]. The species Ircinia fasciculata, collected from the shallow coastal habitats of the Mediterranean Sea (~15 m depth) showed antimicrobial activity in the agar media inoculated with different indicator organisms such as Escherichia coli, Staphylococcus lentus, Candida sp., Bacillus subtilis and Mycobacterium sp. The sponge specimen was chosen for the isolation of bacteria, on the basis of the accumulated evidence that microorganism could well be the true source for some of the metabolites produced by sponges. γ-Proteobacteria was detected in the sponge isolate [32]. An antileukemic marine natural product, sorbicillactone A was isolated from the salt water culture of the fungus Penicillium chrysogenum obtained from another Mediterranean specimen of Ircinia fasciculata. It possesses a unique bicyclic lactone structure, seemingly derived from sorbicillin. The compound exhibited promising activities in several mammalian and viral test systems, particularly in a highly selective cytostatic activity against murine leukemic lymphoblasts (L5178y) and also showed the ability to protect human T cells against the cytopathic effects of HIV-1. These properties qualify sorbicillactone A for future therapeutic human trials [64,65].

2.1.4.3. Family: Spongiidae

An antibacillus compound, which was chemically identified as the peptide antibiotic andrimid was detected in the extract of the sponge Hyatella sp. A bacterial isolate M22-1, belonging to the genus Vibrio was also isolated from the homogenate of the same sponge. The bacterium when cultured in marine agar also produced the same compound. This suggests that the origin of andrimid in the sponge is from the bacterium [66]. Andrimid previously isolated from the cultures of an Enterobacter sp. which is an intracellular symbiont of the brown plant hopper Nilaparvata lugens and was found to exhibit potent activity against Xanthomonas campestris pv. oryzae [67]. It has also been isolated from marine Pseudomonas fluorescens, which was active against methicillin-resistant Staphylococcus aureus. Due to the diversity of the microorganism producing this toxin, one can speculate that the production of this compound might be encoded by genes transferable on a plasmid [68]. The culture broth extracts of the fungus, Myrothecium verrucaria 973023 which was separated from Spongia sp. of Hawaii, showed potent activity against murine lymphocytic leukemia L1210 and human colon tumor H116 cell lines in the soft agar-based bioassay system. Further studies indicated the presence of three new trichothecenes, viz. 3-hydroxyroridin E, 13′-acetyltrichoverrin B, miophytocen C and nine known related compounds such as roridin A, L, M, isororidin A, epiroridin E, verrucarin A, M, trichoverrin A and B in the extract. All the compounds except miophytocen C showed significant cytotoxicity against murine and human tumor cell lines [69].

2.1.4.4. Family: Thorectidae

A new antibiotic trisindole derivative, viz. trisindoline, has been characterized from a marine Vibrio sp., which was separated from the fresh marine okinawan sponge Hyrtios altum. Trisindoline was shown to exhibit potential antibiotic activity against Escherichia coli, Bacillus subtilis and Staphylococcus aureus [7072]. An antileukemic compound, asperazine was isolated from the saltwater culture of the fungus Aspergillus niger obtained from a caribbean Hyrtios sponge by Crews et al. [73]. Asperazine is a member of a large family of diketopiperazine alkaloids. Asperazine displayed remarkable cytotoxicity and an interesting leukemia selectivity [45,74]. Culture extract of another strain of Aspergillus niger from the sponge Hyrtios proteus (Dry Tortugas National Park, Florida) displayed broad chemodiversity and five compounds belonging to a wide range of biosynthetic classes were isolated. Among them, malformin C and asperazine displayed tumor and leukemia selective bioactivity [75]. From the above findings, it can be deduced that Aspergillus niger associated with two different species of Hyrtios inhabiting different geographical locations is capable of producing asperazine. It also gives insight in to the adaptability of a particular microbial associate to a particular sponge genus. An epibiotic bacterial strain Pseudoalteromonas maricaloris KMM 636T, isolated from the Great Barrier Reef sponge Fascaplysinopsis reticulata was the source of two brominated chromopeptides such as bromoalterochromide A and bromoalterochromide A. They showed moderate cytotoxicity to the eggs of the sea urchin Strongylocentrotus intermedius [48,76].

2.1.5. Order: Hadromerida

2.1.5.1. Family: Spirastrellidae

A polyketide, 14,15-secocurvularin was isolated from the saltwater culture of an unidentified fungus obtained from an Indonesian encrusting sponge Spirastrella vagabunda [77]. It was described as being mildly antibiotic against Bacillus subtilis when compared to tetracycline [78].

2.1.5.2. Family: Suberitidae

Suberites domuncula is yet another excellent source for the recovery of bacteria having bioactive potential. This sponge typically grows on snail shells and has a compact, smooth, waxy and colorful surface. Bacteria were isolated from the sponge surface as well as from the laboratory-developed primmorphs of Suberites domuncula collected from northern Adriatic Sea. Two bacteria isolated from the sponge surface were identified as α-Proteobacterium MBIC3368 by using 16S rDNA sequences [79]. This bacterium has also been isolated from several other sponges (e.g., Rhopaloeides odorabile, Aplysina aerophoba) regardless of their taxonomic identity, geographic location or natural product profile [30,40]. Another bacterial isolate from the sponge surface showed 98.8% species level similarity to Idiomarina loihiensis (Alteromonadaceae). The bacteria on primmorph represented unidentified novel species of Pseudomonas [79]. Bioactive extracts of α-Proteobacterial strains from the sponge surface as well as Pseudomonas sp. associated with primmorph exhibited antiangiogenic, antimicrobial, hemolytic and cytotoxic properties. These bacterial extracts were strongly active against multidrug-resistant clinical strains of Staphylococcus aureus and Staphylococcus epidermidis, isolated from hospital patients. Extracts from Idiomarina species also showed hemolytic activity [15].

2.1.6. Order: Halichondrida

2.1.6.1. Family: Axinellidae

A cyclic depsipeptide, majusculamide C has been isolated from the metabolites of the sponge Ptilocaulis trachys collected at the Enewetak Atoll (Marshall Island, Pacific Ocean). It was originally isolated from the toxic blue-green alga Lyngbya majuscula obtained from the same site. Majusculamide C exhibited antifungal activity against pathogens of commercially important plants. This discovery proved that accumulation of cyanobacteria in sponges is diet derived [81,82]. A symbiotic fungal strain Myrothecium sp. JS9 in the marine sponge Axinella sp. from South China Sea was found to be an efficient producer of most effective antifungal metabolites roridin A and D (macrocyclic trichothecenes). Biologically, this class of compounds was reported to possess antileukemic, antimalarial, antimicrobial, phytotoxic and cytotoxic properties [83]. Structurally unique steroids, isocyclocitrinols A and 22-acetylisocyclocitrinol A were isolated from the extract of a saltwater culture of sponge derived fungus Penicillium citrinum, separated from the sponge Axinella sp., collected in Papua New Guinea. Both the steroid compounds exhibited weak antibacterial activity against Staphylococcus epidermidis and Enterococcus durans [84]. The ethyl acetate extract of Penicillium sp., derived from the Mediterranean sponge Axinella verrucosa, yielded the known compound communesin B and its new congeners communesins C and D, and the known compounds oxaline, griseofulvin and dechlorogriseofulvin. Oxaline is an antiproliferative agent which inhibits microtubule protein/purified tubulin polymerization, resulting in arresting cell cycle at the M-phase [85]. Griseofulvin is a widely used antifungal agent for the treatment of superficial dermatomycoses [86]. In several bioassays performed on different leukemia cell lines, the communesins exhibited moderate antiproliferative activity [87]. From a static culture of the fungal strain Aspergillus niger isolated from the Mediterranean sponge Axinella damicornis, eight secondary metabolites belonging to four entirely different structural classes were obtained. Among these, the new compound 3,3′-bicoumarin (bicoumanigrin A) showed moderate cytotoxicity against human cancer cell lines in vitro. Another compound, aspernigrin B displayed a strong neuroprotective effect by significantly reducing the increase of intracellular calcium concentration in rat cortical neurons stimulated with glutamic acid or quisqualic acid [88]. A crude extract from a small-scale culture of the fungus Acremonium sp. 021172C cultured from an Axinella sp. collected from Milne Bay (Papua New Guinea) displayed potent cytotoxicity in a primary screening using leukemia and solid tumor murine and human cancer cell lines. This prompted the growth of a larger-scale culture of the fungus to facilitate the purification of potential therapeutic metabolites which resulted in four new related linear octapeptides, RHM1, 2, 3 and 4, and the known peptaibiotic efrapeptins E, F, G, new efrapeptins Eα and H, known cyclic N-methylated scytalidamides A and B. Efrapeptins displayed antibacterial activity and potent cytotoxicity against murine and human cancer cell lines. RHM1 and RHM2 showed only weak cytotoxicity against murine cancer cell lines but RHM1 exhibited antibacterial activity [89,90]. These studies further confirm the potentiality of fungal metabolites from marine environment.

2.1.6.2. Family: Halichondriidae

The halichondrids form the most important members of demosponges. They are of particular interest because the composition of secondary metabolites is influenced by the presence of prokaryotic symbionts [91]. Sponges of the genus Halichondria such as Halichondria okadai and Halichondria melanodocia provide good examples for the importance of microalgal association in the production of natural compounds recovered from these invertebrates. Both species of Halichondria contain the protein phosphatase inhibitor okadaic acid [14]. It was first isolated from the sponge Halichondria okadai, but, later it was found out that a dinoflagellate Prorocentrum lima produced the inhibitor [17]. Two unidentified bacteria of the genera Pseudomonas and Alteromonas have been isolated from Halichondria Okadai homogenates. The Pseudomonas sp. KK10206C produced a novel C50-carotenoid, okadaxanthine. It turned out to be a potent singlet oxygen quencher and a well known source of okadaic acid [61,92,93]. Alteromonas sp. was responsible for the production of a well-known lactam alteramide A. The genus Alteromonas was found commonly associated with marine sponges that produce macrolactam and amide ester compounds with cytotoxic and antimicrobial properties. The tetracyclic alkaloid alteramide A exhibited cytotoxic activity against leukemia P-388, lymphoma L-1210 and epidermal carcinoma KB cells [9395]. A fungal strain, Trichoderma harzianum OUPS-N115, isolated from the Japanese specimen of Halichondria okadai yielded novel cytotoxic compounds such as trichodenone A, B and C. They exhibited significant cytotoxicity against leukemia P388 cell line [79,96,97]. A Gram-bacterial strain Rubritalea squalenifasciens HOact23T obtained from Halichondria okadai yielded potent red pigmented antioxidants acyl glycol-carotenoic acids such as diapolycopenedioic acid xylosyl esters A, B and C [30,48,98]. Another Halichondria species, Halichondria panacea, which occurs abundantly in the Adriatic Sea, North Sea and Baltic Sea, was colonized by bacteria in its mesohyl compartment. Moreover, different specimens of Halichondria panacea collected from all the three seas harboured bacteria of same genera and indicated the dominance of the genus Rhodobacter, suggesting the symbiotic relationship of these bacteria with the sponge. Evidence has been presented to support that growth of bacteria in Halichondria panacea is maintained by a lectin produced from eukaryotic host. The organic extracts prepared from the sponge samples displayed cytotoxicity against leukemia cells, which supports the possibility of toxic bacteria in the sponges [99]. Bacteria synthesizing neuroactive compounds were also isolated from Halichondria panacea. Two such bacterial species were identified from this sponge which displayed the highest identity to Antarcticum vesiculatum and Psychroserpens burtonensis [100]. An actinobacterium Microbacterium sp. isolated from the sponge Halichondria panacea (Adriatic coast, Croatia) produced four glycoglycerolipids and one diphosphatidylglycerol when grown on marine broth and artificial sea water. The glycoglycerolipid, 2 (1-O-acyl-3-[R-glucopyranosyl-(1–3)-(6-O-acyl-R-mannopyranosyl)]glycerol), showed positive results for antitumor activities in the initial studies [101]. Novel cytotoxic compounds, designated as gymnastatins A-H, Q and R, cytotoxic ergastanoids such as gymnasterone A, B, C and D, novel class of steroid dankasterones A and B, and dankastatins A and B were isolated from an ascomycete fungal strain Gymnascella dankaliensis OUPS-N134, derived from the sponge Halichondria japonica. Gymnastatins A, B, C, F, G, Q and R, dankastatins A and B exhibited potent cytotoxicity and growth inhibition in a P388 lymphocytic leukemia test system in cell culture. Gymnastatin Q was equally active against breast and human cancer cell lines [78,102107]. Gymnasterones B, C and D, and dankasterone A showed significant cytotoxic activity in P388 lymphocytic leukemia test system in cell culture. Dankasterone A was also active against human cancer cell lines [79,104,108,109]. Again from Halichondria Japonica, a fungal strain Phoma sp. Q60596 was obtained, which gave rise to the new antifungal antibiotic, YM-202204. It exhibited potent antifungal activities against Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus [110]. Novel antibiotics, YM-266183 and YM-266184, were found in the culture broth of Bacillus cereus QN03323, which was isolated from Halichondria japonica. They exhibited potent antibacterial activities against staphylococci and enterococci including multiple drug resistant strains, whereas they were inactive against Gram-bacteria [111113]. The antifungal macrolid halichondramide from another Halichondria sp. showed resemblance to the compound scytophycin B, which was extracted earlier from the cyanobacterium Scytonema pseudohofmanni, and therefore halichondramide is speculated to be of microbial origin [114]. Halichondramide also showed in vitro antimalarial activity [115]. The marine bacterial strain Bacillus pumilus AAS3 isolated from the Mediterranean sponge Acanthella acuta, produced a diglucosyl-glycerolipid, GGL11. Lipase catalyzed modification of this native substance led to the deacylated parent compound GG11. Antitumor promoting studies showed that the diglucosyl-glycerol GG11 strongly inhibited the growth of the tumor cell lines HM02 and Hep G2. Thus, it indicates the potential inhibitory activity of the compound with carbohydrate/glycerol backbone [116]. Twenty nine marine bacterial strains were isolated from the sponge Hymeniacidon perlevis at Nanji Island (China Sea), and the antimicrobial screening showed that eight strains inhibited the growth of terrestrial microorganisms. Among them, the strain NJ6-3-1 with wide antimicrobial spectrum was identified as Pseudoalteromonas piscida based on its 16S rRNA sequence analysis. The major antimicrobial metabolite isolated from this bacterium was norhman [43,117]. Another specimen of Hymeniacidon perlevis from the intertidal zone of Fujiazhuang coastline (China) was identified to be a good source of large amount of culturable and active epi/endophytic fungal strains. Of the various fungal isolates obtained from Hymeniacidon perlevis, the extracts of epiphytic fungus Fusarium oxysporum DLFP2008005 exhibited effective antibacterial and antifungal activities against Gram+ Staphylococcus epidermidis, Bacillus subtilis, Gram-Pseudomonas fluorescens, Pseudomonas aeruginosa and the yeast Candida albicans. Several terrestrial as well as marine Fusarium species have been reported to produce structurally diversified antimicrobial compounds. The potential of fungi of the genus Fusarium as producers of novel antibiotics is therefore quite evident [44].

2.1.7. Order: Haplosclerida

2.1.7.1. Family: Callyspongiidae

An antimicrobial fungal metabolite known as acetyl Sumiki’s acid was isolated from a seawater-based fermentation of the fungal isolate Cladosporium herbarum, obtained from the marine sponge Callyspongia aerizusa in Indonesia. Both Sumiki’s acid and its acetyl derivative showed activity against Bacillus subtilis and Staphylococcus aureus at 5 μg/disc [118]. The tropical sponge Callyspongia vaginalis from the Caribbean Sea, yielded a new tyrosine kinase inhibitor and the antimicrobial compound ulocladol together with the antifungal agent 1-hydroxy-6-methyl-8-(hydroxyl-methyl)xanthone. These compounds have been extracted from the culture of sponge-derived fungi Ulocladium botrylis 193A4 [119,120].

2.1.7.2. Family: Chalinidae

The marine sponge genus Haliclona has been extensively examined, and at least 190 metabolites exhibiting anti-fouling, antimicrobial, antifungal, antimalarial and cytotoxic activities have been isolated [121]. A fungal strain isolated from the sponge Haliclona valliculata collected from Elba, Italy and identified as Emericella variecolor showed a remarkable diversity of secondary metabolites. However, strains of the fungus Emericella variecolor have been the source of a variety of natural products. The culture of Emericella variecolor isolated from Haliclona valliculata proved to be chemically prolific. Among various compounds isolated, the novel anthraquinone, evariquinone revealed a strong antiproliferative activity against KB (ATCC CCL17, human cervix carcinoma) and NCI-H460 (NCI 503473, non-small cell lung cancer) cells [122]. Associated with Haliclona simulans from the west coast of Ireland, 52 bacteria isolated belonged to the genera Pseudoalteromonas, Pseudomonas, Halomonas, Psychrobacter, Marinobacter, Sulfitobacter, Pseudovibrio, Salegentibacter, Bacillus, Cytophaga, Rhodococcus and Streptomyces [23]. These strains were found to be rich sources of biological activities with over 50% exhibiting antimicrobial activities. Twelve Streptomyces and one Bacillus strain were found to produce substance active against drug-resistant pathogenic bacteria. PKS (polyketide synthase) and NRPS (nonribosomal peptide synthetase) genes found in Actinobacteria, Bacillus, Sulfitobacter and Pseudovibrio, suggest a high potential for secondary metabolite production by these organisms. Detection of wide spectrum antibiotic activities from Streptomyces isolates SM2 and SM4 is another evidence to support that culturable sponge microbiota is an important source of biologically active compounds. The saltwater culture of an unidentified fungus obtained from the sponge Haliclona sp. was found to produce several new hirsutane sesquiterpenes such as hirsutanols A–C and ent-gloeosteretriol. Hirsutanols are biosynthetically related to several compounds reported from the terrestrial fungus Coriolus consors. Hirsutanol A and ent-gloeosteretriol exhibited mild antibiotic activity against Bacillus subtilis [123]. Potent bacterial strains from Haliclona sp. (Bandangan water, North Java Sea, Indonesia) exhibiting antibacterial activity against the pathogenic bacteria such as Vibrio parahaemolyticus, Aeromonas hydrophila and Staphylococcus aureus were identified using rep-PCR followed by the construction of dendrogram and subsequent DNA sequencing. The active strains showed closest similarity to Vibrio parahaemolyticus, Pseudovibrio denitrificans, Pseudoalteromonas sp., α-Proteobacterium and uncultured bacterium clone [2].

2.1.7.3. Family: Niphatidae

The fungus Curvularia lunata isolated from the marine sponge Niphates olemda from Indonesia yielded two antibacterial anthraquinones such as lunatin and cytoskyrin A. Both of them were found to be active against Staphylococcus aureus, Escherichia coli and Bacillus subtilis [124,125].

2.1.7.4. Family: Petrosiidae

Genus Petrosia has been recognized as a source of diverse metabolites [126,127]. Petrosia ficiformis is a common Mediterranean sponge living in hard substrata between 5 and 45 m depth. Its colour mainly due to symbiotic cyanobacteria, ranges from violet to brown according to the illumination of environment. Petrosia ficiformis hosts a variety of heterotrophic bacteria, most of which live together with cyanobacteria within specialized cells called bacteriocytes [128]. Antimicrobial activity in several epibiotic bacterial isolates from Petrosia ficiformis has been observed by Chelossi et al. [129]. Two of these were identified as Rhodococcus sp. and Pseudomonas sp. by partial 16S rRNA gene sequencing. A strain of Penicillium brevicompactum derived from the specimen of Petrosia ficiformis provided two new cyclopentadepsipeptides, petrosifungins A and B along with the known fungal metabolites brevianamide A, mycophenolic acid (a well known immunosuppressive agent) and asperphenamate. Since cyclodepsipeptides constitute new class of potential drugs, petrosifungins A and B, may serve as lead compounds for more pharmacologically potent and toxicologically safe derivatives [130,131]. A strain of Aspergillus insuetus obtained from the surface of Petrosia ficiformis yielded two new compounds, terretonins E and F. They are potent inhibitors of mammalian mitochondrial respiratory chain [132]. One of the most potent antibacterial activities was detected in the crude extracts of a bacterial strain Micrococcus luteus R-1588-10, isolated from the surface of the sponge Xestospongia sp. collected from off Noumea (New Caledonia, southwest Pacific). Micrococcus luteus is an ubiquitous Gram+ bacteria. Two antimicrobial compounds such as 2,4,4′-trichloro-2′-hydroxydiphenylether (triclosan) and acyl-1-(acyl-6′-mannobiosyl)-3-glycerol (lutoside) have been isolated from Micrococcus luteus [133]. Fungal isolates of Penicillium cf. montanense obtained from the sponge Xestospongia exigua from Bali Sea (Indonesia) has yielded three novel decalactone metabolites, xestodecalactones A, B, and C [134]. Among these, xestodecalactone B showed antifungal activity against Candida albicans [79]. An antibacterial compound, aspergillitine was also isolated from Xestospongia exigua in association with the fungus Aspergillus versicolor. It showed moderate antibacterial activity against Bacillus subtilis [124,135]. An anti-infective alkaloid manzamine A was successfully obtained from the culture of the actinobacterium Micromonospora sp. harbouring the deep water Indonesian sponge Acanthostrongylophora sp. [82]. Manzamine alkaloids were reported earlier from several unrelated and geographically separated sponges, which suggest the microbial origin for the biosynthesis of these compounds [13,136]. Manzamine A was initially described as an antitumor agent against mouse leukemia cells [137] and recently shown to possess antimalarial properties that inhibit in vivo the growth of the rodent malarial parasite Plasmodium berghei [138]. Large scale culture of the sponge derived Micromonospora sp. has since been achieved in 20-litre fermentations, maintaining the manzamine production [13]. The fungus Aspergillus versicolor, isolated from Petrosia sp. (Jeju Island, Korea) yielded three known polyketides such as decumbenones A, B and versiol, and the cytotoxic lipopeptide fellutamide C. The same polyketides have been also reported from soil associated fungus Penicillium decumbens. Decumbenone A is a good inhibitor of melanin [139141].

2.1.8. Order: Lithistida

2.1.8.1. Family: Neopeltidae

Lithistid sponges are renowned among marine organisms for their ability to produce a diverse array of biologically active metabolites [142], including novel peptides characterized by a high proportion of D and/or N-methylated amino acids. The similarity between lithistid peptides and those from microorganisms leads to the speculation that lithistid peptides might arise from symbiotic microbes [143]. A Gram-strain, 1537–E7 was identified as new Pseudomonas species from the surface of the sponge Homophymia sp. collected from off Touho (New Caledonia). Among the five compounds isolated from this bacterium, compound 1 (2-undecyl-4-quinolone) was active against the malarial parasite Plasmodium falciparum and HIV-1. Compound 2 (2-undecen-1′-yl-4-quinolone) displayed mild toxicity and compound 4 (2-nonyl-4-hydroxyquinoline N-oxide) showed antimicrobial activity against Staphylococcus aureus as well as cytotoxicity [142].

2.1.8.2. Family: Theonellidae

The marine sponge Theonella swinhoei from Palau contains a cytotoxic polyketide, swinholide A and the bicyclic glycopeptide antifungal compound theopalauamide [144]. Bacteria associated with this sponge include unicellular cyanobacteria, unicellular bacteria and filamentous bacteria. Swinholide A is likely to be a bacterial metabolite because this compound was associated with fractions from unicellular bacteria in Theonella swinhoei [145]. A single morphotype of a filamentous bacterium was present in a separate fraction that contained the antifungal compound theopalauamide [146]. Subsequent application of molecular approaches identified this filamentous bacterium as novel δ-Proteobacterium related to myxobacteria. According to 16S rDNA data, the filamentous strain is a previously unknown δ-Proteobacterium with close association to the myxococcales and designated as ‘Candidatus Entotheonella palauensis’ [26]. An antifungal glycopeptide known as theonegramide was previously isolated from Theonella swinhoei, collected from Philippines at a depth of 20 m [147]. Interestingly, 16S sequences which showed 98% identity to that of the filamentous δ-Proteobacterium, Entotheonella palauensis were detected in Theonella swinhoei specimens containing the closely related metabolites theonegramide (from the Philippines) and theonellamide F (from Japan), while they were absent in sponges with different metabolites [148]. Theopalauamide-type compounds therefore, seem to be chemical markers for symbiosis of Entotheonella palauensis in sponges [145]. Discovery of onn genes encoding the biosynthesis of onnamide A in the microbial metagenome of the sponge Theonella swinhoei was made by Piel et al. [149]. This polyketide exhibited extremely potent antitumor activities. This provides the first experimental proof for bacterial origin of marine sponge derived natural compounds [150].

2.1.9. Order: Poecilosclerida

2.1.9.1. Family: Acarnidae

Three novel cytotoxic polyketides, brocaenols A-C were produced by Penicillium brocae obtained from a tissue sample of the Fijian sponge Zyzzya sp. When tested against HCT-116 cell line, all three compounds showed cytotoxicity [151,152].

2.1.9.2. Family: Isodictyidae

An antibacterial compound known as cyclo-(l-proline-l-methionine) has been isolated from the culture broth of a symbiotic bacterium Pseudomonas aeruginosa, obtained from the Antarctic sponge Isodictya setifera. It showed antimicrobial activity against Bacillus subtilis, Staphylococcus aureus, and Micrococcus luteus [153].

2.1.9.3. Family: Raspailiidae

A fungal strain Coniothyrium sp. 193477, isolated from the sponge Ectyoplasia ferox from the waters around the Caribbean Islands of Dominica, yielded novel antimicrobial compounds such as (3S)-(3′,5′-dihydroxyphenyl)butan-2-one and 2-(1′(E)-propenyl)-octa-4(E),6(Z)-diene-1,2-diol together with known fungal metabolites such as (3R)-6-methoxymellein, (3R)-6-methoxy-7-chloromellein and cryptosporiopsinol. Among these, cryptosporiopsinol demonstrated significant antimicrobial activity [154]. Potent cytotoxic compounds, epoxyphomalin A and B were discovered from Phoma sp., associated with Ectyoplasia ferox collected from the same region. The former one showed superior activity against various human tumor cell lines [155]. Another fungus Spicellum roseum 193H15, derived from Ectyoplasia ferox was found to produce trichothecenes such as trichodermol and 8-deoxytrichothecin. They considerably inhibited the activity of LacCer synthase (role in oncogene expression and cell proliferation) in neuroblastoma cells [156,157]. The fungus also yielded two cyclohexadepsipeptides, spicellamides A and B [48,158].

2.1.9.4. Family: Mycalidae

An actinobacterium strain Saccharopolyspora sp. nov. associated with the sponge Mycale plumose from Qingdao coast (China) showed cytotoxic activities against temperature sensitive mutant cell lines of mouse (tsFT210). This led to the isolation of two prodigiosins analogs-metacycloprodigiosin and undecylprodigiosin. Prodigiosins are a family of naturally occurring polypyrrole red pigments produced by a restricted group of microorganisms including Streptomyces and Serratia strains. They are known to exhibit a wide range of biological activities. Both the above mentioned prodigiosin analogs exhibited potent in vitro cytotoxic activity against cancer cell lines such as P388, HL60, A-549, BEL-7402 and SPCA4 [159]. The fungus Penicillium auratiogriseum was also isolated from the specimen of Mycale plumose taken from the same geographical area. A new cytotoxic compound (S)-2,4-dihydroxy-1-butyl-(4-hydroxy) benzoate and a known compound fructigenine A were obtained from the fungus. Both the compounds were tested for their antitumor activity and exhibited potent cytotoxic effects [160]. Besides these, two new quinazoline alkaloids such as aurantiomides B and C showing moderate cytotoxic activities were isolated from another strain of Penicillium auratiogriseum associated with Mycale plumose from China [161]. Exophilin A, a new antibacterial compound, was discovered in the culture of the fungus Exophiala pisciphila NI10102, that was isolated from a marine sponge Mycale adhaerens. Exophilin A showed antimicrobial activity against Gram+ bacteria [48,162].

2.1.9.5. Family: Myxillidae

A new antimicrobial fungal metabolite known as microsphaeropsisin together with the known compounds (R)-mellein, (3R,4S)-hydroxymellein, (3R,4R)-hydroxymellein and 4,8-dihydroxy-3,4-dihydro-2H-naphthalen-1-one were obtained from the fungal strain Microsphaeropsis sp. H5-50 associated with the marine sponge Myxilla incrustance, collected from Helgoland, Germany [154]. Microsphaeropsin, an eremophilane derivative showed antifungal activity at the 50 μg level [79].

2.1.10. Order: Spirophorida

2.1.10.1. Family: Tetillidae

A chitinase exhibiting antifungal activity was isolated from marine Streptomyces sp. DA11 associated with south China sponge Craniella australiensis. Compared with chitinase derived from terrestrial organisms, marine chitinase with higher pH and salinity tolerance may contribute to special biotechnological applications. Therefore, novel marine chitinase could be of great importance [163].

2.1.11. Order: Verongida

2.1.11.1. Family: Aplysinellidae

Ten strains of marine actinobacteria belonging to the genus Salinospora were isolated from the Great Barrier Reef sponge Suberea clavata (formerly Pseudoceratina clavata) [164]. The Salinospora group, a relatively newly discovered group of actinobacteria, has great applied potential. The Salinospora strains previously isolated from marine sediments showed significant cancer cell cytotoxicities as well as antifungal and antibiotic activities [165]. Significantly, Salinospora forms a potential new source of rifamycins and polyketide synthesis gene clusters specific to rifamycin synthesis. Salinospora isolate from Suberea clavata was found to produce compounds of the rifamycin class, including rifamycin B and rifamycin SV [166]. Other culturable symbiotic bacterial communities isolated from Suberea clavata include α-, γ-Proteobacteria, Bacteriodetes and Firmicutes [167].

2.1.11.2. Family: Aplysinidae

Sponges of the Aplysinidae family are abundant in the subtropical and tropical waters of the Mediterranean Sea, Pacific and Atlantic Oceans [168]. Aplysina sponges harbour large amounts of microorganisms with antimicrobial activities that are embedded within the mesohyl [41,168]. The Mediterranean sponge Aplysina aerophoba is especially rich in bacteria. The amount of bacteria present in the sponge tissue matrix exceeds the microbial concentration of the seawater by two to three orders of magnitude [169]. One of the studies conducted using FISH on Aplysina aerophoba and its sibling species Aplysina cavernicola showed that the bacterial profiles of both species was very similar. Up to 40% of the sponge biomass consisted of bacteria and cyanobacteria. A large fraction of the microbial community was specific to and permanently associated with the host sponge [41,168,169]. The similarity of the bacterial communities in Aplysina aerophoba and Aplysina cavernicola corresponds to similarities in the natural product profiles of both sponges which are characterized by brominated alkaloids with cytotoxic activities and repellent properties against predators [12,41]. Among the bacterial isolates obtained from these species those which showed antimicrobial activity were numerically the most abundant in the genus Pseudoalteromonas and the class α-Proteobacteria. A general pattern was observed in that Gram+ bacteria inhibited Gram+ strains while Gram-bacteria inhibited Gram− isolates. Antimicrobial activities were also found against clinical isolates, i.e., multi-drug resistant Staphylococcus aureus and Staphylococcus epidermidis strains isolated from hospital patients. The high recovery of strains with antimicrobial activity suggests that marine sponges represent an ecological niche which harbours largely uncharacterized microbial diversity and yet undiscovered metabolic potential [41]. Antimicrobial activity of bacterial isolates from Aplysina aerophoba collected from the Mediterranean coast of France has been tested against a set of standard Gram+, Gram− and eukaryotic microorganisms. The results showed that Bacillus subtilis strains A184, A190 and A202 exhibited strong activity against the fungus Candida albicans [170]. It is generally accepted that a combination of fungicidal and hemolytic activity in Bacillus is a valid indicator for the presence of lipopeptide from the iturin or surfactin class [171173]. For the Bacillus subtilis strains A184, A190 and A202, these features are consistent. The results of MALDI MS which was applied to study the production of secondary metabolites by Bacillus species showed that strain A184 produced surfactins, iturins and fengycins while strain A190 produced surfactin and strain A202 produced iturin. The highly versatile strain Bacillus subtilis A184 was highly active against the multidrug resistant pathogenic Staphylococcus aureus and Staphylococcus epidermidis. Another species, Bacillus pumilus A586 demostrated high activity against Staphylococcus aureus and produced plumilacidin containing β-hydroxy fatty acid (surfactin like compound) [170]. An undescribed fungus of the genus Microsphaeropsis, isolated from the Mediterranean specimen of Aplysina aerophoba, was shown to produce a Protein Kinase C inhibitor known as 10-Hydroxy-18-methoxylbetaenone [174]. Since PKC plays an important role in neoplastic transformation, carcinogenesis and tumour cell invasion, those agents which inhibit the action of PKC are therapeutically very important [175].

2.1.11.3. Family: Pseudoceratinidae

Extract of Metarrhizium sp. 001103 from Pseudoceratina purpurea (Fiji), yielded six known N-methylated cyclic depsipeptides of the destruxin family. They include destruxins A, B, B2, desmethyl B, E and E2 chlorohydrin. Destruxins A, B2, desmethyl B and E chlorohydrin displayed selective inhibition of human tumor cell lines. E2 chlorohydrin showed cytotoxicity towards murine c38 cell line. E chlorohydrin was the most potent among the group [90].

2.2. Class: Calcarea

Order: Clathrinida
Family: Leucettidae
Nonribosomal cyclic peptide leucamide A was isolated from the sponge Leucetta microraphis, obtained from the Great Barrier Reef of Australia. The compound was found to inhibit the growth of three tumor cell lines (stomach carcinoma, liver carcinoma and liver carcinoma with mutated p53). Leucamide A closely resembles the compound albeit, which is found frequently in cyanobacteria. Scanning electromicrographs of Leucetta microraphis revealed the presence of microbial symbionts, including cyanobacteria in the tissue. The sponge-derived leucamide A might, therefore be produced by cyanobacteria associated with it and not by the invertebrate itself [120].

2.3. Unidentified sponges

A marine-derived strain of the fungus Emericella variecolor, obtained from a Venezuelan sponge, yielded new compounds along with a group of known metabolites. Some of the novel compounds such as varitriol and varixanthone exhibited potent pharmaceutical activities. Varitriol displayed increased potency towards selected renal, CNS and breast cancer cell lines, whereas varixanthone showed antimicrobial activity [176]. Two novel antimycin antibiotics viz. urauchimycins A and B, were isolated from a fermentation broth of Streptomyces sp. Ni-80. The strain was isolated from an unidentified sponge. They are the first antimycin antibiotics which possess a branched side chain moiety. They exhibited inhibitory activity against morphological differentiation of Candida albicans [177]. A strain of the fungus Microascus longirostris SF-73 from a marine sponge collected at Harrington Point (Otago Harbour, New Zealand) was found to produce secondary metabolites such as cathestatin A, B and C, which strongly inhibited cystein proteases. Since specific and selective protease inhibitors are potentially powerful tools in clinical therapy, these inhibitors could be used in inactivating the target proteases in the pathogenic processes of human diseases such as emphysema, arthritis, pancreatitis, thrombosis, high blood pressure, muscular dystrophy, cancers, AIDS and many others [178]. Three antibacterial compounds were isolated from the fungus Aspergillus ostianus 01F313, derived from an unidentified sponge collected at Pohnpei (The federated state of Micronesia). They include 8-chloro-9-hydroxy-8,9-deoxyasperlactone, 9-chloro-8-hydroxy-8,9-deoxyasperlactone and 9-chloro-8-hydroxy-8,9-deoxyaspyrone [179]. From the same strain, five cytotoxic compounds such as aspinorene, dihydroaspyrone, aspergillides A, B and C were also obtained when cultured in a brominated medium. They exhibited cytotoxicity against lymphocytic leukemia cells (L1210) [48,180,181]. Cultivation of the fungus Cryptosphaeria eunomia, obtained from an unidentified sponge at Pohnpei yielded the antimycobacterial compounds diaporthein A and B. These compounds have been previously isolated from the terrestrial fungus Diaporthe sp. BCC 6140 [48,182,183].

3. Discussion

Sponge-microbial associations which synthesize clinically significant bioactive compounds have been discovered so far from geographically different regions such as Great Barrier Reef of Australia, South China Sea, Mediterranean Sea, Indonesia, Papua New Guinea, Indo-Pacific region etc. (Table 2). The review brings out the fact that members of the class Demospongiae are the richest producer of pharmacologically significant bioactive compounds in association with microbes. Out of 92 families under class Demospongiae, 26 familes have been identified to produce medicinally important bioactive compounds of microbial origin. They includes Ancorinidae, Chondrillidae, Darwinellidae, Dysideidae, Irciniidae, Spongiidae, Thorectidae, Spirastrellidae, Suberitidae, Axinellidae, Halichondriidae, Callyspongiidae, Chalinidae, Niphatidae, Petrosiidae, Neopeltidae, Theonellidae, Acarnidae, Raspailiidae, Isodictyidae, Mycalidae, Myxillidae, Tetillidae, Aplysinellidae, Aplysinidae and Pseudoceratinidae. The major orders which contribute maximum to the compound production are Halichondrida, Dictyoceratida and Poecilosclerida. Families which belong to the order Halichondrida such as Axinellidae and Halichondriidae are more influenced by microbes in the production of secondary metabolites. The microbial associates of halichondrid comprises broad spectrum of bacteria, actinobacteria, fungi and micro algae. Association of these microbes with different species of halichondrid sponges have been shown to be the real source of bioactive compounds exhibiting significant therapeutic effects. These compounds include alteramide, trichodenone A-C, gymnastatins A-C (antileukemic) YM-202204, YM-266183 and YM-266184 (antibiotics). Apart from these, species belonging to the families Chalinidae and Petrosiidae of the order Haplosclerida, Darwinellidae of the order Dendroceratida are also rich sources of bioactive compounds of microbial origin. Only one family from the class Calcarea has been identified as a source of pharmacologically significant bioactive compounds of microbial origin. There are no reports in the literature regarding isolation of microbial originated therapeutic compounds from the class Hexactinellida.
Some of the compounds produced by microbes in association with sponge orders such as Hadromerida, Haplosclerida and Verongida have not been characterized and therefore have not been included in the above figure.
The major groups of microorganisms recognized from this review as possible contributors of pharmacologically relevant secondary metabolites of sponges includes α, β, γ, δ- Proteobacteria, Firmicutes, Actinobacteria, Cyanobacteria and Fungi. Interestingly, the members of the fungal genus Aspergillus, which is ubiquitous in terrestrial, is also the principle source of bioactive compounds in marine sponges. Out of more than 680 fungal strains isolated worldwide from 16 sponge species, majority belong to the genera Aspergillus and Penicillium [184].The Fusarium genus is also considered as a potential candidate for the production of novel antibiotics [44]. Even though most of the the sponge-microbial association is very specific for the production of a particular compound, a few compounds have also been isolated from free living and associated microbes in marine and terrestrial ecosystems. Tricyclic sesquiterpene coriolin B (anticancer) has been isolated from a marine fungus of the class Hyphomycetes in Jaspis. aff. johnstoni as well as from the terrestrial wood rotting basidiomycete Coriolus consors [45]. Both the antimycobacterial compounds diaporthein A and B were isolated from the terrestrial fungus Diaporthe sp. and the marine fungus Cryptosphaeria eunomia associated with an unidentified sponge [48,182,183]. Polyketides such as decumbenones A and B were earlier isolated from the soil fungus Penicillium decumbens and later from Aspergillus versicolor associated with Petrosia sp. [139141]. The antibacillus peptide antibiotic, associated with Petrosia sp. [139141]. The antibacillus peptide antibiotic, andrimid was isolated from Vibrio sp. M 22-1 associated with the sponge Hyatella sp. and also from a symbiotic Enterobacter sp. of the brown plant-hopper Nilaparvata lugens [68]. Some sponges always harbour a particular genera or species of microorganism and consistently produce specific group of compounds. The association of the tropical marine shallow water sponge Lamellodysidea herbacea with cyanobacterium Oscillatoria spongeliae, is one such example which produces chlorinated diketopiperazines [58]. Similarly, the symbiotic microbes of Dysidea sp. consistently synthesize brominated diphenyl ethers [60]. Likewise, irrespective of the geographical region the antileukemic compound asperazine is produced by Aspergillus niger from two different Hyrtios species. Also, the antileukemic and antitumor compound roridin A is produced by Myrothecium sp. present in Spongia sp. of Hawaii and Axinella sp. of South China Sea [70,74,76,83].
Figures 2, 3 and 4 show the percentage distribution of clinically active compounds obtained from bacteria and fungi. Even though the number of bacterial isolates exhibiting clinical activities are more than fungi, many of the compounds produced by bacteria are not yet characterized. Figures 2 and 3 could be altered later once those compounds are characterized. Phylum Actinobacteria dominates in the production of therapeutic compounds followed by Proteobacteria. Bioactive potential of firmicutes and cyanobacteria is yet to be explored. Among fungi, Ascomycota is a predominant producer of bioactive molecules and Deuteromycota is also a potential group exhibiting bioactivity.
A wide range of chemical and functional diversity has been observed among bioactive compounds. Of the various chemical classes of compounds, polyketides, alkaloids, fatty acids, peptides and terpenes are the most abundant ones. Majority of them show antimicrobial, antitumor and anticancer properties. Bacterial and fungal associates in the order Dictyoceratida are found to synthesize antiangiogenic, anticancer, antiHIV, antitumor as well as antimicrobial compounds [15,40,57,59,61,62,6466,69]. Another noticeable fact is the discovery of an actinobacterial strain (Nocardiopsis dassonvillei MAD08) from the sponge Dendrilla nigra of the family Darwinellidae from southwest coast of India (Table 3). This particular strain was able to produce compounds exhibiting antimicrobial, antioxidant, hypocholesterolemic, nematicidal, antiandrogenic, hemolytic, anti-inflammatory and anticancer properties. Of the various compounds produced by this strain, hexadecanoic acid- methyl ester, n-hexadecanoic acid, hexadecanoic acid-ethyl ester, 9-octadecenoic acid (Z)-methyl ester, oleic acid and (E)-9-octadecenoic acid-ethyl ester have been shown to be multifunctional [54]. Similarly the fungal strain Gymnascella dankaliensis OUPS-N134 from Halichondria japonica was very potent and produced 12 antileukemic compounds [78,102109].
Some of the drugs available in the market, which were previously isolated from various terrestrial microbial genera were also detected in the marine counterparts associated with the sponges. A fungistatic drug, griseofulvin used for dermatophytoses has been isolated from various terrestrial and marine strains of Penicillium. This drug has also been reported from Penicillium symbiont of the Mediterranean sponge Axinella verrucosa [85,186]. Similarly, a well known immunosuppressive and antibiotic drug, mycophenolic acid which was produced by various strains of Penicillium including Penicillium stoloniferum and Penicillium roqueforti has also been isolated from Penicillium brevicompactum associated with the sponge Petrosia ficiformis [130,187,188]. Thus, the bioactive potential of the genus Penicillium either marine or terrestrial origin, free living or symbiotic makes it a worthy candidate for understanding the microbe-sponge association and harnessing the bioactive compounds.

4. Ecological and Cultural Aspects of Sponge Symbionts

To date, the primary target for marine bioprospecting has been tropical seas particularly coral reefs and other highly diverse ecosystems such as mangroves and seagrass because they host a high level of biodiversity and often face intense competition for space, leading to a chemical warfare among the sessile organisms. It was proven extremely difficult and in some cases impossible to provide sufficient quantity of these substances from invertebrates. The reason was due to the limited quantity of the compound, or still due to limited number of organisms producing the compound. Geographical, seasonal or sexual variations in the amount and nature of secondary metabolites could also be the other reasons for not consistently getting the required quantity of the compound. Marine invertebrates which are abundant in the Indo-Pacific regions, are rich in secondary metabolites and are becoming targets of continuing search for bioactive compounds [189]. The China Sea has become an important source of marine natural compounds since 2001 [190]. Among metazoans, the phylum Porifera contains the taxa which produce the highest diversity of secondary metabolites [191].
With some exceptions, sponge-associated microbial communities appear to be relatively stable with time and space [192]. With respect to temporal variability, the fluctuation of microbial communities in Aplysina aerophobha (an aquarium maintained specimen), Geodia barrette (Cultivated explant), temperate Australian sponges such as Callyspongia sp., Stylinos sp. and Cymbastela concentrica were detected to be low with no evidence of major seasonal changes [169,193,194]. In contrast to these studies, the bacterial community abundant in the North Sea sponge Halichondria panacea was found to vary considerably over a 10 month period [195]. Spatial variability could be ascribed to difference in microbiota within and among individuals which are separated by geographical barriers [194196]. Marked differences were evident between the microbial communities inhabiting the outer (cortex) and inner (endosome) tissue in the Mediterranean sponge Tethya aurantium [197]. Contrary to this, Antarctic sponges such as Homaxinella balfourensis, Kirkpatrickia varialosa, Latrunculia apicalis, Mycale acerata and Sphaerotylus antarcticus collected from different sampling sites separated by 10 km were found to possess highly consistent bacterial communities. It highlights that site variability does not affect bacterial community composition in Antarctic sponges, but is highly consistent within a particular species [195]. Another study by Taylor et al. [198], showed that bacterial communities associated with temperate and tropical population of Cymbastela concentrica along the eastern Australian coast vary substantially.
Seasonal changes in the production of bioactive compounds by sponges are poorly understood. Seasonal fluctuations occurring in temperate seas impose significant alterations on the biology of the organism [199]. Seasonal changes, both qualitative and quantitative, have been observed in bioactive compound production in the sponge Crambe crambe from Mediterranean Sea. More importantly, high intra individual variability has also been observed. High toxicity in the producer organism during autumn may be a defense mechanism to counter increased growth of competing animal species at the end of summer. A decrease in toxicity in the months preceding April could be due to the reproductive rhythm. Energy diversion towards reproduction may explain the decrease in toxic metabolite production [200]. It was also found that non-polar fraction of the crude extract obtained from associated bacteria of the sponge Ircinia ramosa possessed strong antibacterial activity in summer. During winter season, activity was detected in the polar fraction and it was comparatively weaker than the observed activity in non-polar fraction during summer. This give insight in to the assumption that chemistry and production rate of metabolites from sponges or associated bacteria could be governed by environmental conditions [201]. More studies are being done to show that microbes are the real source of many of the bioactive compounds in sponges. Future efforts may throw more light on seasonal effects of bioactive compound production by these associates.
The occurrence of important metabolites within sponge-associated bacteria opens up the possibility of providing a continuous supply of the biologically active compounds by laboratory cultivation of the producer [202]. It would seem a logical step trying to isolate and cultivate putative bacterial producers outside invertebrate hosts in order to set up a sustainable and manageable source of pharmacologically active compounds. Even if microbial populations can be successfully separated from the hosts, the undefined metabolic factors of the host may render it difficult for the symbiont to grow ex hospite [82]. Many bacterial inhabitants in sponges appear to be highly selective with regard to culture media and conditions which probably reflect their evolutionary adaptation to the conditions provided by the host. Attempts to culture the theopaulamide producing bacteria from the sponge Theonella swinhoei have failed so far [203]. A notable exception is an anti-infective alkaloid manzamine A, which was successfully obtained from the culture of bacterium Micromonospora sp. of the deep-water Indonesian sponge Acanthostrongylophora sp. [13]. Another possibility is to grow the entire sponge and its microbial community in self-contained aquaculture systems for the economic, sustainable supply of important metabolites. The advantage of the latter strategy compared with growth of sponges in the wild or in open-water mariculture system is the possibility of better control of environmental conditions such as temperature, light, food supply and possibly precursors of important bioactive metabolites. In addition, aquaculture of sponges may provide less perturbation of the bacterium-host association over growth of bacterial ‘producers’ strains in pure culture which could be very important for maintaining production of compounds of interest [62].
It is hypothesized that antagonism, polyketide synthase genes and PLA2 are the key functional precursors of secondary metabolite synthesis and/or host defense of marine sponges. The study of metabolite-related genes of microorganisms associated with sponges may give insight into the origin of sponge-derived natural products. Polyketides, comprising a large and structural diverse family of bioactive natural products are one of the most important classes of marine natural compounds [190]. Polyketide synthase genes of host sponge and associated bacteria are predicted to be biosynthetic modules of polyketide analogues as well as phospholipases [51]. The PKS gene-based molecular approach can be applied to efficient screening of strains of pharmaceutical value and prediction of related compounds. This strategy has been employed to discover the efficiency of polyketide production in Firmicutes especially Bacillus, Actinobacteria and Proteobacteria isolated from sponges of the South China Sea [190]. Isolation and culture of symbiotic microorganisms as producers of secondary metabolites as well as transfer of symbiont biosynthetic genes into cultivable bacteria are subjects of ongoing research [149,204]. Even if compounds or compound groups appear exclusive for a particular taxon, they are not necessarily homologous and derived from a common ancestor and therefore do not necessarily reflect a genealogical relationship. They might originate from different precursors and biochemical pathways [203]. Rajdasa et al. [2], highlighted the repetitive PCR method as a powerful tool in estimating the richness of secondary metabolite producers among colonizers of sponge Haliclona sp. and this approach may be useful in studying the diversity of other sponge-associated microorganisms.

5. Conclusions

Sponge-microbial associations are found to be very specific in the production of particular bioactive compounds. However, the mutual mechanism between host and the microbial associate, in compound production is not well understood. The easiest and best way for commercial production of these compounds are either by culturing the host and/or the associated microbe under controlled conditions. But, the ability of the symbiont to produce the compound consistently for several generation in culture media has to be tested and standardized. Moreover, there is a need for quantifying the role of sponge ecology in orchestrating the production of specific compounds. Metagenomic approaches are also being increasingly used for targeting putative genes encoding potential metabolites in uncultured microbial biota. These approaches would help in delineating the contribution of either the host or microbial associate or both partners in the production of metabolites. A few compounds have been found to be produced both in terrestrial and marine ecosystems by different groups of host-symbiont association. This suggests the possibility of horizontal gene transfer through evolution. Discovery of potent microbial associates producing therapeutic compounds has opened up a new era in marine pharmacology. Understanding the optimum ecological conditions which drives the sustainable production of bioactive compounds from sponges and their microbial associates would help in formulating various production strategies. Adopting different cultivation strategies and metagenomic approaches would be the need of the hour in discovering new genes, enzymes and natural products and in enhancing the commercial production of marine drugs.

Acknowledgments

Aknowledgements
The authors thank the Director, NIO, Goa for providing facilities. This review was benefited from the discussions with N. Thakur of Bioorganic laboratory, NIO, Goa. Contributions of Michelle Rodrigues (Information Technology Group), in the development of the related database ‘Marassoc.drug’ is appreciated. The authors also thank C. T. Achuthankutty, Visiting Scientist, NCAOR, Goa for critically reading the manuscript and suggesting improvements. They also thank the two anonymous reviewers for offering useful suggestions for improving the quality of the paper. This work was carried out under BTIS (Biotechnology Information System) funded by DBT (Department of Biotechnology, New Delhi). The NIO contribution no. 4730.
  • Samples Availability: Available from the authors.

References

  1. Hentschel, U; Hopke, J; Horn, M; Friedrich, AB; Wagner, M; Hacker, J; Moore, BS. Molecular evidence for a uniform microbial community in sponges from different oceans. Appl Environ Microbiol 2002, 68, 4431–4440. [Google Scholar]
  2. Radjasa, OK; Sabdono, A; Junaidi; Zocchi, E. Richness of secondary metabolite producing marine bacteria associated with sponge Haliclona sp. Int J Pharm 2007, 3, 275–279. [Google Scholar]
  3. Fusetani, N; Matsunaga, S. Bioactive sponge peptides. Chem Rev 1993, 93, 1793–1806. [Google Scholar]
  4. Lee, YK; Lee, JH; Lee, HK. Microbial symbiosis in marine sponges. J Microbiol 2001, 39, 254–264. [Google Scholar]
  5. Fieseler, L; Horn, M; Wagner, M; Hentschel, U. Discovery of the novel candidate Phylum “Poribacteria” in marine sponges. Appl Environ Microbiol 2004, 70, 3724–3732. [Google Scholar]
  6. Belarbi, EH; Gomez, AC; Chisti, Y; Camacho, FG; Grima, EM. Producing drugs from marine sponges. Biotechnol Adv 2003, 21, 585–598. [Google Scholar]
  7. Thakur, NL; Müller, WEG. Biotechnological potential of marine sponges. Curr Sci 2004, 86, 1506–1512. [Google Scholar]
  8. Jensen, PR; Fenical, W. Strategies for the discovery of secondary metabolites from marine bacteria: ecological perspectives. Annu Rev Microbiol 1994, 48, 559–584. [Google Scholar]
  9. Bernan, VS; Greenstein, M; Maise, WM. Marine microorganisms as a source of new natural products. Adv Appl Microbiol 1997, 43, 57–89. [Google Scholar]
  10. Haygood, MG; Schmidt, EW; Davidson, SK; Faulkner, DJ. Microbial symbionts of marine invertebrates: opportunities for microbial biotechnology. J Molec Microbiol Biotechnol 1999, 1, 33–43. [Google Scholar]
  11. Osinga, R; Armstrong, E; Burgess, JG; Hoffmann, F; Reitner, J; Schumann-Kindel, G. Sponge microbe associations and their importance for sponge bioprocess engineering. Hydrobiologia 2001, 461, 55–62. [Google Scholar]
  12. Proksch, P; Edrada, RA; Ebel, R. Drugs from the seas: current status and microbiological imblications. Appl Microbiol Biotechnol 2002, 59, 125–134. [Google Scholar]
  13. Taylor, MW; Radax, R; Steger, D; Wagner, M. Sponge associated microorganisms: Evolution, ecology and biotechnological potential. Microbiol Mol Biol Rev 2007, 71, 295–347. [Google Scholar]
  14. Wang, G. Diversity and biotechnological potential of the sponge-associated microbial consortia. J Ind Microbiol Biotechnol 2006, 33, 545–551. [Google Scholar]
  15. Thakur, AN; Thakur, NL; Indap, MM; Pandit, RA; Datar, VV; Müller, WEG. Antiangiogenic, antimicrobial and cytotoxic potential of sponge-associated bacteria. Mar Biotechnol 2005, 7, 245–252. [Google Scholar]
  16. Gunasekera, AS; Sfanos, KS; Harmody, DK; Pomponi, SA; McCarthy, PJ; Lopez, JV. An enhanced database of the microorganisms associated with deeper water marine invertebrates. Appl Microbiol Biotechnol 2004, 66, 373–376. [Google Scholar]
  17. Kobayashi, J; Ishibashi, M. Bioactive metabolites of symbiotic marine microorganism. Chem Rev 1993, 93, 1753–1769. [Google Scholar]
  18. Ridley, CP; Faulkner, DJ; Haygood, MG. Investigation of Oscillatoria spongeliae dominated bacterial communities in four dictyoceratid sponges. Appl Environ Microbiol 2005, 71, 7366–7375. [Google Scholar]
  19. Alvarez, B; Crisp, MD; Driver, F; Hooper, JNA; Soest, RWMV. Phylogenetic relationships of the family Axinellidae (Porifera: Demospongiae) using morphological and molecular data. Zool Scripta 2000, 29, 169–198. [Google Scholar]
  20. Bergmann, W; Burke, DC. Contributions to the study of marine products. XXXIX. The nucleosides of sponges. III. Spongothymidine and spongouridine. J Org Chem 1955, 20, 1501–1507. [Google Scholar]
  21. Wang, G. Diversity and biotechnological potential of the sponge-associated microbial consortia. J Ind Microbiol Biotechnol 2006, 33, 545–551. [Google Scholar]
  22. Kennedy, J; Baker, P; Piper, C; Cotter, PD; Walsh, M; Mooij, MJ; Bourke, MB; Rea, MC; O’Connor, PM; Ross, RP; Hill, C; O’Gara, F; Marchesi, JR; Dobson, ADW. Isolation and analysis of bacteria with antimicrobial activities from the marine sponge Haliclona simulans collected from Irish waters. Mar Biotechnol 2009, 11, 384–396. [Google Scholar]
  23. Li, Z; He, L; Miao, X. Cultivable bacterial community from South China Sea sponge as revealed by DGGE fingerprinting and 16S rDNA phylogenetic analysis. Curr Microbiol 2007, 55, 465–472. [Google Scholar]
  24. Head, IM; Saunders, JR; Pickup, RW. Microbial Evolution, Diversity, and Ecology: A Decade of Ribosomal RNA Analysis of Uncultivated Microorganisms. Microb Ecol 1998, 35, 1–21. [Google Scholar]
  25. Juretschko, S; Timmermann, G; Schmid, M; Schleifer, K; Pommerening-Röser, A; Koops, H; Wagner, M. Combined Molecular and Conventional Analyses of Nitrifying Bacterium Diversity in Activated Sludge: Nitrosococcus mobilis and Nitrospira-Like Bacteria as Dominant Populations. Appl Environ Microbiol 1998, 64, 3042–3051. [Google Scholar]
  26. Schmidt, EW; Obraztsova, AY; Davidson, SK; Faulkner, DJ; Haygood, MG. Identification of the antifungal peptide-containing symbiont of the marine sponge Theonella swinhoei as a novel δ-Proteobacterium Candidatus Entotheonella palauensis. Mar Biol 2000, 136, 969–977. [Google Scholar]
  27. Vacelet, J; Gallissian, M. Virus-like particles in cells of the sponge Virongia cavernicola (demospongiae, dictyoceratida) and accompanying tissue changes. J Invertebr Pathol 1978, 31, 246–254. [Google Scholar]
  28. Olson, JB; Mccarthy, PJ. Associated bacterial communities of two deep-water sponges. Aquat Microb Ecol 2005, 39, 47–55. [Google Scholar]
  29. Hill, M; Hill, A; Lopez, N; Harriott, O. Sponge-specific bacterial symbionts in the Caribbean sponge, Chondrilla nucula (Demospongiae, Chondrosida). Mar Biol 2006, 148, 1221–1230. [Google Scholar]
  30. Kasai, H; Katsuta, A; Sekiguchi, H; Matsuda, S; Adachi, K; Shindo, K; Yoon, J; Yokota, A; Shizuri, Y. Rubritalea squalenifaciens sp. nov., a squalene-producing marine bacterium belonging to subdivision 1 of the phylum ‘Verrucomicrobia’. Int J Syst Evol Microbiol 2007, 57, 1630–1634. [Google Scholar]
  31. Enticknap, JJ; Kelly, M; Peraud, O; Hill, RT. Characterization of a culturable alphaproteobacterial symbiont common to many marine sponges and evidence for vertical transmission via sponge larvae. Appl Environ Microbiol 2006, 72, 3724–3732. [Google Scholar]
  32. Thiel, V; Imhoff, JF. Phylogenetic identification of bacteria with antimicrobial activities isolated from Mediterranean sponges. Biomol Eng 2003, 20, 421–423. [Google Scholar]
  33. Radjasa, OK; Martens, T; Grossart, H; Brinkhoff, T; Sabdono, A; Simmon, M. Antagonistic activity of a marine bacterium Pseudoalteromonas luteoviolacea TAB4.2 associated with coral Acropora sp. J Biol Sci 2007, 7, 239–246. [Google Scholar]
  34. Zhang, L; An, R; Wang, J; Sun, N; Zhang, S; Hu, J; Kuai, J. Exploring novel bioactive compounds from marine microbes. Curr Opin Microbiol 2005, 8, 276–281. [Google Scholar]
  35. Newman, DJ; Hill, RT. New drugs from marine microbes: the tide is turning. J Ind Microbiol Biotechnol 2006, 33, 539–544. [Google Scholar]
  36. Perry, NB; Blunt, JW; Munro, MHG. Mycalamide A, an antiviral compound from a New Zealand sponge of the genus Mycale. J Am Chem Soc 1988, 110, 4850–4851. [Google Scholar]
  37. Hood, KA; West, LM; Northcote, PT; Berridge, MV; Miller, JH. Induction of apoptosis by the marine sponge (Mycale) metabolites, mycalamide A and pateamine. Apoptosis 2001, 6, 207–219. [Google Scholar]
  38. Laport, MS; Santos, OCS; Muricy, G. Marine sponges: Potential sources of new antimicrobial drugs. Curr Pharmaceut Biotechnol 2009, 10, 86–105. [Google Scholar]
  39. Unson, MD; Holland, ND; Faulkner, DJ. A brominated secondary metabolite synthesized by the cyanobacterial symbiont of a marine sponge and accumulation of the crystalline metabolite in the sponge tissue. Mar Biol 1994, 119, 1–11. [Google Scholar]
  40. Hentschel, U; Schmid, M; Wagner, M; Fieseler, L; Gernert, C; Hacker, J. Isolation and phylogenetic analyses of bacteria with antimicrobial activities from the Mediterranean sponges Aplysina aerophoba and Aplysina cavernicula. FEMS Microbiol Ecol 2001, 35, 305–312. [Google Scholar]
  41. Zheng, Z; Zeng, W; Huang, Y; Yang, Z; Li, J; Cai, H; Su, W. Detection of antitumor and antimicrobial activities in marine organism associated actinomycetes isolated from the Taiwan Strait, China. FEMS Microbiol Lett 2000, 188, 87–91. [Google Scholar]
  42. Li, Z. Advances in marine microbial symbionts in the China Sea and related pharmaceutical metabolites. Mar Drugs 2009, 7, 113–129. [Google Scholar]
  43. Zhang, Y; Mu, J; Feng, Y; Kang, Y; Zhang, J; Gu, P; Wang, Y; Ma, L; Zhu, Y. Broad-spectrum antimicrobial epiphytic and endophytic fungi from marine organisms: Isolation, bioassay and taxonomy. Mar Drugs 2009, 7, 97–112. [Google Scholar]
  44. Biabani, MAF; Laatsch, H. Advances in chemical studies on low-molecular weight metabolites of marine fungi. J Prakt Chem 1998, 340, 589–607. [Google Scholar]
  45. Cheng, X; Varoglu, M; Abrell, L; Crews, P; Lobkovsky, E; Clardy, J. Chloriolins A-C, chlorinated sesquiterpenes produced by fungal cultures separated from a Jaspis marine sponge. J Org Chem 1994, 59, 6344–6348. [Google Scholar]
  46. Sufrin, JR; Finckbeiner, S; Oliver, CM. Marine-Derived Metabolites of S-Adenosylmethionine as Templates for New Anti-Infectives. Mar Drugs 2009, 7, 401–434. [Google Scholar]
  47. Lang, G; Wiese, J; Schmaljohann, R; Imhoff, JF. New pentaenes from the sponge-derived marine fungus Penicillium rugulosum: structure determination and biosynthetic studies. Tetrahedron 2007, 63, 11844–11849. [Google Scholar]
  48. Blunt, JW; Copp, BR; Hu, W; Munro, MHG; Northcote, PT; Prinsep, MR. Marine natural products. Nat Prod Rep 2009, 26, 170–244. [Google Scholar]
  49. Selvin, J; Gandhimathi, R; Seghal Kiran, G; Shanmugha Priya, S; Ravji, TR; Hema, TA. Culturable heterotrophic bacteria from the marine sponge Dendrilla nigra: Isolation and phylogenetic diversity of actinobacteria. Helgol Mar Res 2009, 63, 239–247. [Google Scholar]
  50. Selvin, J. Exploring the antagonistic producer Streptomyces MSI051: Implications of polyketide synthase gene type II and a ubiquitous defense enzyme phospholipase A2 in the host sponge Dendrilla nigra. Curr Microbiol 2009, 58, 459–463. [Google Scholar]
  51. Selvin, J; Lipton, AP. Biopotentials of secondary metabolites isolated from marine sponges. Hydrobiologia 2004, 513, 231–238. [Google Scholar]
  52. Selvin, J; Joseph, S; Asha, KRT; Manjusha, WA; Sangeetha, VS; Jayaseema, DM; Antony, MC; Vinitha, AJD. Antibacterial potential of antagonistic Streptomyces sp. isolated from marine sponge Dendrilla nigra. FEMS Microbiol Ecol 2004, 50, 117–122. [Google Scholar]
  53. Selvin, J; Shanmughapriya, S; Gandhimathi, R; Kiran, GS; Ravji, TR; Natarajaseenivasan, K; Hema, TA. Optimization and production of novel antimicrobial agents from sponge associated marine actinomycetes Nocardiopsis dassonvillei MAD08. Appl Microbiol Biotechnol 2009, 83, 435–445. [Google Scholar]
  54. Hinde, R; Pironet, F; Borowitzka, MA. Isolation of Oscillatoria spongeliae, the filamentous cyanobacterial symbiont of the marine sponge Dysidea herbacea. Mar Biol 1994, 119, 99–104. [Google Scholar]
  55. Arillo, A; Bavestrello, G; Burlando, B; Sara, M. Metabolic integration between symbiotic cyanobacteria and sponges: a possible mechanism. Mar Biol 1993, 117, 159–162. [Google Scholar]
  56. Unson, MD; Faulkner, DJ. Cyanobacterial symbiont biosynthesis of chlorinated metabolites from Dysidea herbacea (Porifera). Cell Mol Life Sci 1993, 49, 349–353. [Google Scholar]
  57. Flowers, AE; Garson, MJ; Webb, RI; Dumdei, EJ; Charan, RD. Cellular origin of chlorinated diketopiperazines in the dictyoceratid sponge Dysidea herbacea (Keller). Cell Tissue Res 1998, 292, 597–607. [Google Scholar]
  58. Besada, P; Mamedova, L; Thomas, CJ; Costanzi, S; Jacobson, KA. Design and synthesis of new bicyclic diketopiperazines as scaffolds for receptor probes of structurally diverse functionality. Org Biomol Chem 2005, 3, 2016–2025. [Google Scholar]
  59. Elyakov, GB; Kuznetsova, T; Mikhailov, VV; Maltsev, II; Voinov, VG; Fedoreyev, SA. Brominated diphenyl ethers from a marine bacterium associated with the sponge Dysidea sp. Cell Mol Life Sci 47, 632–633.
  60. Scheuermayer, M; Pimentel-Elardo, S; Fieseler, L; Grozdanov, L; Hentschel, U. Proksch, P, Müller, WEG, Eds.; Microorganisms of sponges: Phylogenetic diversity and biotechnological potential. In Frontiers in Marine Biotechnology; Horizon Bioscience: Norfolk, UK, 2006; pp. 289–312. [Google Scholar]
  61. Thakur, NL; Müller, WEG. Sponge-bacteria association: A useful model to explore symbiosis in marine invertebrates. Symbiosis 2005, 39, 109–116. [Google Scholar]
  62. Müller, WEG; Thakur, NL; Ushijima, H; Thakur, AN; Krasko, A; Pennec, G; Indap, MM; Perovic-Ottstadt, S; Schröder, HC; Lang, G; Bringmann, G. Matrix-mediated canal formation in primmorphs from the sponge Suberites domuncula involves the expression of a CD36 receptor-ligand system. J Cell Sci 2004, 117, 2579–2590. [Google Scholar]
  63. Mohamed, NM; Rao, V; Hamann, MT; Kelly, M; Hill, RT. Monitoring bacterial diversity of the marine sponge Ircinia strobilina upon transfer to aquaculture. Appl Environ Microbiol 2008, 74, 4133–4143. [Google Scholar]
  64. Bringmann, G; Lang, G; Muhlbacher, J; Schaumann, K; Steffens, S; Rytik, PG; Hentschel, U; Morschhauser, J; Müller, WEG. Sorbicillactone A: A structurally unprecedented bioactive novel-type alkaloid from a sponge-derived fungus. Prog Mol Subcell Biol 2003, 37, 231–253. [Google Scholar]
  65. Bringmann, G; Gulder, TAM; Lang, G; Schmitt, S; Stöhr, R; Wiese, J; Nagel, K; Imhoff, JF. Large–scale biotechnological production of the antileukemic marine natural product sorbicillactone A. Mar Drugs 2007, 5, 23–30. [Google Scholar]
  66. Oclarit, JM; Okada, H; Ohta, S; Kaminura, K; Yamaoka, Y; Iizuka, T; Miyashiro, S; Ikegami, S. Anti-bacillus substance in the marine sponge, Hyatella species, produced by an associated Vibrio species bacterium. Microbios 1994, 78, 7–16. [Google Scholar]
  67. Fredenhagen, A; Tamura, SY; Kenny, PTM; Komura, H; Naya, Y; Nakanishi, K; Nishiyama, K; Sugiura, M; Kita, H. Andrimid, a new peptide antibiotic produced by an intracellular bacterial symbiont isolated from a brown planthopper. J Am Chem Soc 1987, 109, 4409–4411. [Google Scholar]
  68. Needham, J; Kelly, MT; Ishige, M; Andersen, RJ. Andrimid and moiramides A-C, metabolites produced in culture by a marine isolate of the bacterium Pseudomonas fluorescens: Structure elucidation and biosynthesis. J Org Chem 1994, 59, 2058–2063. [Google Scholar]
  69. Amagata, T; Rath, C; Rigot, JF; Tarlov, N; Tenney, K; Valeriote, FA; Crews, P. Structures and cytotoxic properties of trichoverroids and their macrolide analogues produced by saltwater culture of Myrothecium verrucaria. J Med Chem 2003, 46, 4342–4350. [Google Scholar]
  70. Kobayashi, M; Kitagawa, I. Bioactive substances isolated from marine sponge, a miniature conglomerate of various organisms. Pure Appl Chem 1994, 66, 819–826. [Google Scholar]
  71. Kobayashi, M; Aoki, S; Gato, K; Matsunami, K; Kurosu, M; Kitagawa, I. Marine natural products. XXXIV. Trisindoline, a new antibiotic indole trimer, produced by a bacterium of Vibrio sp. separated from the marine sponge Hyrtios altum. Chem Pharm Bull 1994, 42, 2449–2451. [Google Scholar]
  72. Braekman, J; Daloze, D. Chemical and biological aspects of sponge secondary metabolites. Phytochem Rev 2004, 3, 275–283. [Google Scholar]
  73. Varoglu, M; Corbett, TH; Valeriote, FA; Crews, P. Asperazine, a selective cytotoxic alkaloid from a sponge-derived culture of Aspergillus niger. J Org Chem 1997, 62, 7078–7079. [Google Scholar]
  74. Govek, SP; Overman, LE. Total synthesis of asperazine. J Am Chem Soc 2001, 123, 9468–9469. [Google Scholar]
  75. Varoglu, M; Crews, P. Biosynthetically diverse compounds from a saltwater culture of sponge derived Aspergillus niger. J Nat Prod 2000, 63, 41–43. [Google Scholar]
  76. Speitling, M; Smetanina, OF; Kuznetsova, TA; Laatsch, H. Bromoalterochromides A and A′, unprecedented chromopeptides from a marine Pseudoalteromonas maricaloris strain KMM 636T. J Antibiot 2007, 60, 36–42. [Google Scholar]
  77. Abrell, LM; Borgeson, B; Crews, P. A new polyketide, secocurvularin, from the salt water culture of a sponge derived fungus. Tetrahedon Lett 1996, 37, 8983–8984. [Google Scholar]
  78. Bugni, TS; Ireland, CM. Marine derived fungi: a chemically and biologically diverse group of microorganisms. Nat Prod Rep 2004, 21, 143–163. [Google Scholar]
  79. Thakur, NL; Hentschel, U; Krasko, A; Pabel, CT; ANR, AC; Müller, WEG. Antibacterial activity of the sponge Suberites domuncula and its primmorphs: potential basis for epibacterial chemical defense. Aquat Microb Ecol 2003, 31, 77–83. [Google Scholar]
  80. Webster, NS; Hill, RT. The culturable microbial community of the Great Barrier Reef sponge Rhopaloeides odorabile is dominated by an α-Proteobacterium. Mar Biol 2001, 138, 843–851. [Google Scholar]
  81. Williams, DE; Burgoyne, DL; Rettig, SJ; Andersen, RJ; Fathi-Afshar, ZR; Allen, TM. The isolation of Majusculamide C from the sponge Ptilocaulis trachys collected in Enewetak and determination of the absolute configuration of the 2-methyl-3-aminopentanoic acid residue. J Nat Prod 1993, 56, 545–551. [Google Scholar]
  82. Dunlap, WC; Battershill, CN; Liptrot, CH; Cobb, RE; Bourne, DG; Jaspars, M; Long, PF; Newman, DJ. Biomedicinals from the phytosymbionts of marine invertebrates: A molecular approach. Methods 2007, 42, 358–376. [Google Scholar]
  83. Xie, LW; Jiang, SM; Zhu, HHl; Sun, W; Ouyang, YC; Dai, SK; Li, X. Potential inhibitors against Sclerotinia sclerotiorum, produced by the fungus Myrothecium sp. associated with the marine sponge Axinella sp. Eur J Plant Pathol 2008, 122, 571–578. [Google Scholar]
  84. Amagata, T; Amagata, A; Tenney, K; Valeriote, FA; Lobkovsky, E; Clardy, J; Crews, P. Unusual C25 steroids produced by a sponge-derived Penicillium citrinum. Org Lett 2003, 5, 4393–4396. [Google Scholar]
  85. Koizumi, Y; Arai, M; Tomoda, H; Omura, S. Oxaline, a fungal alkaloid, arrests the cell cycle in M phase by inhibition of tubulin polymerization. Biochim Biophys Acta 2004, 1693, 47–55. [Google Scholar]
  86. Kolachana, P; Smith, MT. Induction of kinetochore-positive micronuclei in human lymphocytes by the anti-fungal drug griseofulvin. Mutat Res 1994, 322, 151–159. [Google Scholar]
  87. Jadulco, R; Edrada, RA; Ebel, R; Berg, A; Schaumann, K; Wray, V; Steube, K; Proksch, P. New communesin derivatives from the fungus Penicillium sp. derived from the Mediterranean sponge Axinella verrucosa. J Nat Prod 2004, 67, 78–81. [Google Scholar]
  88. Hiort, J; Maksimenka, K; Reichert, M; Perovic-Ottstadt, S; Lin, WH; Wray, V; Steube, K; Schaumann, K; Weber, H; Proksch, P; Ebel, R; Müller, WEG; Bringmann, G. New natural products from the sponge-derived fungus Aspergillus niger. J Nat Prod 2004, 67, 1532–1543. [Google Scholar]
  89. Boot, CM; Tenney, K; Valeriote, FA; Crews, P. Highly N-methylated linear peptides produced by an atypical sponge-derived Acremonium sp. J Nat Prod 2006, 69, 83–92. [Google Scholar]
  90. Boot, CM; Amagata, T; Tenney, K; Compton, JE; Pietraszkiewicz, H; Valeriote, FA; Crews, P. Four classes of structurally unusual peptides from two marine-derived fungi: structures and bioactivities. Tetrahedon 2007, 63, 9903–9914. [Google Scholar]
  91. Erpenbeck, D; Breeuwer, JAJ; van der Velde, HC; van Soest, RWM. Unravelling host and symbiont phylogenies of halichondrid sponges (Demospongiae, Porifera) using mitochondrial marker. Mar Biol 2002, 141, 377–386. [Google Scholar]
  92. Miki, W; Otaki, N; Yokoyama, A; Izumida, H; Shimidzu, N. Okadaxanthin, a novel C50-carotenoid from a bacterium Pseudomonas sp. KK10206C associated with a marine sponge Halichondria okadai. Experientia 1994, 50, 684–686. [Google Scholar]
  93. Kelecom, A. Secondary metabolites from marine microorganisms. An Acad Bras Cienc 2002, 74, 151–170. [Google Scholar]
  94. Shigemori, H; Bae, MA; Yazawa, K; Sasaki, T; Kobayashi, J. Alteramide A, a new tetracyclic alkaloid from a bacterium Alteromonas sp. associated with the marine sponge Halichondria okadai. J Org Chem 1992, 57, 4317–4320. [Google Scholar]
  95. Bhalla, TC; Sharma, M; Sharma, NN. Satyanarayana, T, Chand, S, Eds.; Microbial production of flavours and fragrances; fats and oils; dyes; bioplastics (PHAS); polysaccharides; pharmacologically active substances from marine microbes; anticancer agents and microbial transformation. In Applied Microbiology; National Science Digital Library NISCAIR: New Delhi, India, 2008; Volume 7, pp. 1–34. [Google Scholar]
  96. Amagata, T; Usami, Y; Minoura, K; Ito, T; Numata, A. Cytotoxic substances produced by a fungal strain from a sponge: physico-chemical properties and structures. J Antibiot 1998, 51, 33–40. [Google Scholar]
  97. Usami, Y; Ikura, T; Amagata, T; Numata, A. First total syntheses and configurational assignments of cytotoxic trichodenones A–C. Tetrahedron-Asymmetry 2000, 11, 3711–3725. [Google Scholar]
  98. Shindo, K; Asagi, E; Sano, A; Hotta, E; Minemura, N; Mikami, K; Tamesada, E; Misawa, N; Maoka, T. Diapolycopenedioic Acid Xylosyl Esters A, B, and C, Novel Antioxidative Glyco-C30-carotenoic Acids Produced by a New Marine Bacterium Rubritalea squalenifaciens. J Antibiot 2008, 61, 185–191. [Google Scholar]
  99. Althoff, K; Schutt, C; Steffen, R; Batel, R; Müller, WEG. Evidence for a symbiosis between bacteria of the genus Rhodobacter and the marine sponge Halichondria panicea: Harbor also for putatively toxic bacteria? Mar Biol 1998, 130, 529–536. [Google Scholar]
  100. Perovic, S; Wichels, A; Schutt, C; Gerdts, G; Pahler, S; Steffen, R; Müller, WEG. Neuroactive compounds produced by bacteria from the marine sponge Halichondria panicea: activation of the neuronal NMDA receptor. Environ Toxicol Pharmacol 1998, 6, 125–133. [Google Scholar]
  101. Wicke, C; Hners, M; Wray, V; Nimtz, M; Bilitewski, U; Lang, S. Production and structure elucidation of glycoglycerolipids from a marine sponge associated Microbacterium species. J Nat Prod 2000, 63, 621–626. [Google Scholar]
  102. Numata, A; Amagata, T; Minoura, K; lto, T. Gymnastatins, novel cytotoxic metabolites produced by a fungal strain from a sponge. Tetrahedon Lett 1997, 38, 5675–5678. [Google Scholar]
  103. Amagata, T; Doi, M; Ohta, T; Minoura, K; Numata, A. Absolute stereostructures of novel cytotoxic metabolites, gymnastatins A–E, from a Gymnascella species separated from a Halichondria sponge. J Chem Soc Perkin Trans 1 1998, 1, 3585–3599. [Google Scholar]
  104. Amagata, T; Doi, M; Tohgo, M; Minoura, K; Numata, A. Dankasterone, a new class of cytotoxic steroid produced by a Gymnascella species from a marine sponge. Chem Commun 1999, 1321–1322. [Google Scholar]
  105. Mayer, AMS. Marine Pharmacology in 1998: Antitumor and Cytotoxic Compounds. Pharmacologist 1999, 41, 159–164. [Google Scholar]
  106. Amagata, T; Minoura, K; Numata, A. 2006. Gymnastatins F-H, Cytostatic Metabolites from the Sponge-Derived Fungus Gymnascella dankaliensis. J Nat Prod 2006, 69, 1384–1388. [Google Scholar]
  107. Amagata, T; Tanaka, M; Yamada, T; Minoura, K; Numata, A. Gymnastatins and Dankastatins, Growth Inhibitory Metabolites of a Gymnascella Species from a Halichondria Sponge. J Nat Prod 2008, 71, 340–345. [Google Scholar]
  108. Amagata, T; Tanaka, M; Yamada, T; Doi, M; Minoura, K; Ohishi, H; Yamori, T; Numata, A. Variation in Cytostatic Constituents of a Sponge-Derived Gymnascella dankaliensis by Manipulating the Carbon Source. J Nat Prod 2007, 70, 1731–1740. [Google Scholar]
  109. Amagata, T; Minoura, K; Numata, A. Gymnasterones, novel cytotoxic metabolites produced by a fungal strain from a sponge. Tetrahedron Lett 1998, 39, 3773–3774. [Google Scholar]
  110. Nagai, K; Kamigiri, K; Matsumoto, H; Kawano, Y; Yamaoka, M; Shimoi, H; Watanabe, M; Suzuki, K. YM-202204, a new antifungal antibiotic produced by marine fungus Phoma sp. J Antibiot 2002, 55, 1036–1041. [Google Scholar]
  111. Nagai, K; Kamigiri, K; Arao, N; Suzumura, K; Kawano, Y; Yamaoka, M; Zhang, H; Watanabe, M; Suzuki, K. YM-266183 and YM-266184, novel thiopeptide antibiotics produced by Bacillus cereus isolated from a marine sponge. I. Taxonomy, fermentation, isolation, physico-chemical properties and biological properties. J Antibiot 2003, 56, 123–128. [Google Scholar]
  112. Suzumura, K; Yokoi, T; Funatsu, M; Nagai, K; Tanaka, K; Zhang, H; Suzuki, K. YM-266183 and YM-266184, novel thiopeptide antibiotics produced by Bacillus cereus isolated from a marine sponge II. Structure elucidation. J Antibiot 2003, 56, 129–134. [Google Scholar]
  113. Laatsch, H. Proksch, P, Müller, WEG, Eds.; Marine bacterial metabolite. In Frontiers in Marine Biotechnology; Horizon Bioscience: Norfolk, UK, 2006; pp. 225–288. [Google Scholar]
  114. Hildebrand, M; Waggoner, LE; Lim, GE; Sharp, KH; Ridley, CP; Haygood, MG. Approaches to identify, clone, and express symbiont bioactive metabolite genes. Nat Prod Rep 2003, 21, 122–142. [Google Scholar]
  115. El Sayed, KA; Dunbar, DC; Perry, TL; Wilkins, SP; Hamann, MT. Marine natural products as prototype insecticidal agents. J Agric Food Chem 1997, 45, 2735–2739. [Google Scholar]
  116. Ramm, W; Schatton, W; Wagner-Dobler, I; Wray, V; Nimtz, M; Tokuda, H; Enjyo, F; Nishino, H; Beil, W; Heckmann, R; Lurtz, V; Lang, S. Diglucosyl-glycerolipids from the marine sponge-associated Bacillus pumilus strain AAS3: their production, enzymatic modification and properties. Appl Microbiol Biotechnol 2004, 64, 497–504. [Google Scholar]
  117. Zheng, L; Chen, H; Han, X; Line, W; Yan, X. Antimicrobial screening and active compound isolation from marine bacterium NJ6-3-1 associated with the sponge Hymeniacidon perleve. World J Microbiol Biotechnol 2005, 21, 201–206. [Google Scholar]
  118. Jadulco, R; Proksch, P; Wray, V; Sudarsono; Berg, A; Grafe, U. New macrolides and furan carboxylic acid derivative from the sponge derived fungus Cladosporium herbarum. J Nat Prod 2001, 64, 527–530. [Google Scholar]
  119. Höller, U; König, GM; Wright, AD. A new tyrosine kinase inhibitor from a marine isolate of Ulocladium botrytis and new metabolites from the marine fungi Asteromyces cruciatus and Varicosporina ramulosa. Eur J Org Chem 1999, 1999, 2949–2955. [Google Scholar]
  120. König, GM; Kehraus, S; Seibert, SF; Abdel-Lateff, A; Müller, D. Natural products from marine organisms and their associated microbes. Chem Bio Chem 2005, 7, 229–238. [Google Scholar]
  121. Yu, S; Deng, Z; Proksch, P; Lin, W. Oculatol, oculatolide and A-nor sterols from the sponge Haliclona oculata. J Nat Prod 2006, 69, 1330–1334. [Google Scholar]
  122. Bringmann, G; Lang, G; Steffens, S; Gunther, E; Schaumann, K. Evariquinone, isoemericellin, and stromemycin from a sponge derived strain of the fungus Emericella variecolor. Phytochemistry 2003, 63, 437–443. [Google Scholar]
  123. Wang, G; Abrell, LM; Avelar, A; Borgeson, BM; Crews, P. New hirsutane based sesquiterpenes from salt water cultures of a marine sponge derived fungus and the terrestrial fungus Coriolus consors. Tetrahedron 1998, 54, 7335–7342. [Google Scholar]
  124. Bhadury, P; Mohammad, BT; Wright, PC. The current status of natural products from marine fungi and their potential as anti-infective agents. J Ind Microbiol Biotechnol 2006, 33, 325–337. [Google Scholar]
  125. Jadulco, R; Brauers, G; Edrada, RU; Ebel, R; Wray, V; Sudarsono; Proksch, P. New metabolites from sponge derived fungi Curvularia lunata and Cladosporium herbarum. J Nat Prod 2002, 65, 730–733. [Google Scholar]
  126. Kim, JS; Im, KS; Jung, JH. New bioactive polyacetylenes from the marine sponge Petrosia sp. Tetrahedron 1998, 54, 3151–3158. [Google Scholar]
  127. Lim, YJ; Park, HS; Im, KS; Lee, CO; Hong, J; Lee, M; Kim, D; Jung, JH. Additional cytotoxic polyacetylenes from the marine sponge Petrosia sp. J Nat Prod 2001, 64, 46–53. [Google Scholar]
  128. Vacelet, J; Donadey, C. Electron microscope study of the association between some sponges and bacteria. J Exp Mar Bio Ecol 1977, 30, 301–314. [Google Scholar]
  129. Chelossi, E; Milanese, M; Milano, A; Pronzato, R; Riccardi, G. Characterisation and antimicrobial activity of epibiotic bacteria from Petrosia ficiformis (Porifera, Demospongiae). J Exp Mar Biol Ecol 2004, 309, 21–33. [Google Scholar]
  130. Bringmann, G; Lang, G; Steffens, S; Schaumann, K. Petrosifungins A and B, novel cyclodepsipeptides from a sponge-derived strain of Penicillium brevicompactum. J Nat Prod 2004, 67, 311–315. [Google Scholar]
  131. Lemmens-Gruber, R; Kamyar, MR; Dornetshuber, R. Cyclodepsipeptides - Potential drugs and lead compounds in the drug development process. Curr Med Chem 2009, 16, 1122–1137. [Google Scholar]
  132. López-Gresa, MP; Cabedo, N; González-Mas, MC; Ciavatta, MA; Avila, C; Primo, J. Terretonins E and F, Inhibitors of the Mitochondrial Respiratory Chain from the Marine-Derived Fungus Aspergillus insuetus. J Nat Prod 2009, 72, 1348–1351. [Google Scholar]
  133. Bultel-Poncé, V; Debitus, C; Berge, J; Cerceau, C; Guyot, M. Metabolites from the sponge-associated bacterium Micrococcus luteus. J Mar Biotechnol 1998, 6, 233–236. [Google Scholar]
  134. Edrada, RA; Heubes, M; Brauers, G; Wray, V; Berg, A; Grafe, U; Wohlfarth, M; Muhlbacher, J; Schaumann, K; Sudarsono, S; Bringmann, G; Proksch, P. Online analysis of xestodecalactones A-C, novel bioactive metabolites from the fungus Penicillium cf. montanense and their subsequent isolation from the sponge Xestospongia exigua. J Nat Prod 2002, 65, 1598–1604. [Google Scholar]
  135. Lin, W; Brauers, G; Ebel, R; Wray, V; Berg, A; Sudarsono; Proksch, P. Novel chromone derivatives from the fungus Aspergillus versicolor isolated from the marine sponge Xestospongia exigua. J Nat Prod 2002, 66, 57–61. [Google Scholar]
  136. El Sayed, KA; Kelly, M; Kara, UAK; Ang, KKH; Katsuyama, T; Dumbar, DC; Khan, AA; Hamann, MT. New manzamine alkaloids with potent activity against infectious diseases. J Am Chem Soc 2001, 123, 1804–1808. [Google Scholar]
  137. Sakai, R; Higa, T; Jefford, CW; Bernardinelli, G. Manzamine A, a novel antitumor alkaloid from a sponge. J Am Chem Soc 1986, 108, 6404–6405. [Google Scholar]
  138. Ang, KKH; Holmes, MJ; Higa, T; Hamann, MT; Kara, UAK. In vivo antimalarial activity of the beta-carboline alkaloid manzamine A. Antimicrob Agents Chemother 2000, 44, 1645–1649. [Google Scholar]
  139. Lee, YM; Mansoor, TA; Hong, J; Lee, C-O; Bae, KS; Jung, JH. Polyketides from a Sponge-Derived Fungus, Aspergillus versicolor. Nat Prod Sci 2007, 13, 90–96. [Google Scholar]
  140. Lee, YM; Dang, HT; Hong, J; Lee, C-O; Bae, KS; Kim, DK; Jung, JH. A Cytotoxic Lipopeptide from the Sponge-Derived Fungus Aspergillus versicolor. Bull Korean Chem Soc 2010, 31, 205–208. [Google Scholar]
  141. Fujii, Y; Asahara, M; Ichinoe, M; Nakajima, H. Fungal melanin inhibitor and related compounds from Penicillium decumbens. Phytochemistry 2002, 60, 703–708. [Google Scholar]
  142. Bultel-Poncé, V; Berge, J; Debitus, C; Nicolas, J; Guyot, M. Metabolites from the sponge associated bacterium Pseudomonas species. Mar Biotechnol 1999, 1, 384–390. [Google Scholar]
  143. Capon, RJ; Ford, J; Lacey, E; Gill, JH; Heiland, K; Friedel, T. Phoriospongin A and B: Two new nematocidal depsipeptides from the Australian marine sponges Phoriospongia sp. and Callyspongia bilamellata. J Nat Prod 2002, 65, 358–363. [Google Scholar]
  144. Bewley, CA; Faulkner, DJ. Lithistid sponges: Star performers or hosts to the stars. Angew Chem Int Ed 1998, 37, 2162–2178. [Google Scholar]
  145. Bewley, CA; Holland, ND; Faulkner, DJ. Two classes of metabolites from Theonella swinhoei are localized in distinct populations of bacterial symbionts. Experientia 1996, 52, 716–722. [Google Scholar]
  146. Schmidt, EW; Bewley, CA; Faulkner, DJ. Theopalauamide, a bicyclic glycopeptide from filamentous bacterial symbionts of the lithistid sponge Theonella swinhoei from Palau and Mozambique. J Org Chem 1998, 63, 1254–1258. [Google Scholar]
  147. Bewley, CA; Faulkner, DJ. Theonegramide, an antifungal glycopeptide from the Philippine lithistid sponge Theonella swinhoei. J Org Chem 1994, 59, 4849–4852. [Google Scholar]
  148. Piel, J. Metabolites from symbiotic bacteria. Nat Prod Rep 2004, 21, 519–538. [Google Scholar]
  149. Piel, J; Hui, D; Wen, G; Butzke, D; Platzer, M; Fusetani, N; Matsunaga, S. Antitumor polyketide biosynthesis by an uncultivated bacterial symbiont of the marine sponge Theonella swinhoei. Proc Natl Acad Sci 2004, 101, 16222–16227. [Google Scholar]
  150. Grozdanov, L; Hentschel, U. An environmental genomics perspective on the diversity and function of marine sponge-associated microbiota. Curr Opinion Microbiol 2007, 10, 215–220. [Google Scholar]
  151. Bugni, TS; Bernan, VS; Greenstein, M; Janso, JE; Maiese, WM; Mayne, CL; Ireland, CM. Brocaenols A–C: Novel polyketides from a marine-derived Penicillium brocae. J Org Chem 2003, 68, 2014–2017. [Google Scholar]
  152. Ebel, R. Proksch, P, Müller, WEG, Eds.; Secondary metabolites from marine-derived fungi. In Frontiers in Marine Biotechnology; Horizon Scientific Press: Norwich, UK, 2006; pp. 73–143. [Google Scholar]
  153. Jayatilake, GS; Thornton, MP; Leonard, AC; Grimwade, JE; Baker, BJ. Metabolites from an Antarctic sponge associated bacterium Pseudomonas aeruginosa. J Nat Prod 1996, 59, 293–296. [Google Scholar]
  154. Höller, U; König, GM; Wright, AD. Three new metabolites from marine derived fungi of the genera Coniothyrium and Microsphaeropsis. J Nat Prod 1999, 62, 114–118. [Google Scholar]
  155. Mohamed, IE; Gross, H; Pontius, A; Kehraus, S; Krick, A; Kelter, G; Maier, A; Fiebig, H; König, GM. Epoxyphomalin A and B, Prenylated Polyketides with Potent Cytotoxicity from the Marine-Derived Fungus Phoma sp. Org Lett 2009, 11, 5014–5017. [Google Scholar]
  156. Kralj, A; Gurgui, M; König, GM; van Echten-Deckert, G. Trichothecenes induce accumulation of glucosylceramide in neural cells by interfering with lactosylceramide synthase activity. Toxicol Appl Pharmacol 2007, 225, 113–122. [Google Scholar]
  157. Chatterjee, S; Kolmakova, A. Lactosylceramide synthase: From molecular biochemistry to biological function. In Lipids (sphingolipid metabolizing enzymes 2004); Research Signpost: Trivandrum, India, 2004; pp. 33–41. [Google Scholar]
  158. Kralj, A; Kehraus, S; Krick, A; van Echten-Deckert, G; König, GM. Two new depsipeptides from the marine fungus Spicellum roseum. Planta Med 2007, 73, 366–371. [Google Scholar]
  159. Liu, R; Cui, C; Duan, L; Gu, Q; Zhu, W. Potent in Vitro anticancer activity of metacycloprodigiosin and undecylprodigiosin from a sponge-derived actinomycete Saccharopolyspora sp nov. Arch Pharm Res 2005, 28, 1341–1344. [Google Scholar]
  160. Xin, ZH; Zhu, WM; Gu, QQ; Fang, LD; Cui, CB. A new cytotoxic compound from Penicillium auratiogriseum, symbiotic or epiphytic fungus of sponge Mycale plumose. Chin Chem Lett 2005, 16, 1227–1229. [Google Scholar]
  161. Xin, ZH; Fang, Y; Du, L; Zhu, T; Duan, L; Chen, J; Gu, Q; Zhu, W. Aurantiomides A C, quinazoline alkaloids from the sponge-derived fungus Penicillium aurantiogriseum SP0-19. J Nat Prod 2007, 70, 853–855. [Google Scholar]
  162. Doshida, J; Hasegawa, H; Onuki, H; Shimidzu, N. Exophilin A, a new antibiotic from a marine microorganism Exophiala pisciphila. J Antibiot 1996, 49, 1105–1109. [Google Scholar]
  163. Han, Y; Yang, B; Zhang, F; Miao, X; Li, Z. Characterization of antifungal chitinase from marine Streptomyces sp. DA11 associated with south China sea sponge Craniella australiensis. Mar Biotechnol 2009, 11, 132–140. [Google Scholar]
  164. Kim, TK; Garson, MJ; Fuerst, JA. Marine actinomycetes related to the ‘Salinospora’ group from the Great Barrier Reef sponge Pseudoceratina clavata. Environ Microbiol 2005, 7, 509–518. [Google Scholar]
  165. Mincer, TJ; Jensen, PR; Kauffman, CA; Fenical, W. Widespread and persistent populations of a major new marine Actinomycete taxon in ocean sediments. Appl Environ Microbiol 2002, 68, 5005–5011. [Google Scholar]
  166. Kim, TK; Hewavitharana, AK; Shaw, PN; Fuerst, JA. Discovery of a new source of rifamycin antibiotics in marine sponge Actinobacteria by phylogenetic prediction. Appl Environ Microbiol 2006, 72, 2118–2125. [Google Scholar]
  167. Lafi, FF; Garson, MJ; Fuerst, JA. Culturable bacterial symbionts isolated from two distinct sponge species (Pseudoceratina clavata and Rhabdastrella globostellata) from the Great Barrier Reef of Australia. Microb Ecol 2005, 50, 213–220. [Google Scholar]
  168. Friedrich, AB; Merkert, H; Fendert, T; Hacker, J; Proksch, P; Hentschel, U. Microbial diversity in the marine sponge Aplysina cavernicola (formerly Verongia cavernicola) analyzed by fluorescence in situ hybridization (FISH). Mar Biol 1999, 134, 461–470. [Google Scholar]
  169. Friedrich, AB; Fischer, I; Proksch, P; Hacker, J; Hentschel, U. Temporal variation of the microbial community associated with the Mediterranean sponge Aplysina aerophoba. FEMS Microbiol Ecol 2001, 38, 105–113. [Google Scholar]
  170. Pabel, CT; Vater, J; Wilde, C; Franke, P; Hofemeister, J; Adler, B; Bringmann, G; Hacker, J; Hentschel, U. Antimicrobial activities and Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry of Bacillus Isolates from the marine sponge Aplysina aerophoba. Mar Biotechnol 2003, 5, 424–434. [Google Scholar]
  171. Maget-Dana, R; Peypoux, F. Iturins, a special class of pore-forming lipopeptides: biological and physicochemical properties. Toxicology 1994, 87, 151–174. [Google Scholar]
  172. Besson, F; Peypoux, F; Michel, G; Delcambe, L. Mode of action of iturin A, an antibiotic isolated from Bacillus subtilis on Micrococcus luteus. Biochem Biophys Res Commun 1978, 81, 297–304. [Google Scholar]
  173. Klich, MA; Lax, AR; Bland, JM. Inhibition of some mycotoxigenic fungi by iturin A, a peptidolipid produced by Bacillus subtilis. Mycopathologia 2004, 116, 77–80. [Google Scholar]
  174. Brauers, G; Edrada, RA; Ebel, R; Proksch, P; Wray, V; Berg, A; Grafe, U; Schachtele, C; Totzke, F; Finkenzeller, G; Marme, D; Kraus, J; Munchbach, M; Michel, M; Bringmann, G; Schaumann, K. Anthraquinones and betaenone derivatives from the sponge-associated fungus Microsphaeropsis species: Novel inhibitors of protein kinases. J Nat Prod 2000, 63, 739–745. [Google Scholar]
  175. Mackay, HJ; Twelves, CJ. Protein kinase C: a target for anticancer drugs. Endocr Relat Cancer 2003, 10, 389–396. [Google Scholar]
  176. Malmstroem, J; Christophersen, C; Barrero, AF; Oltra, JE; Justicia, J; Rosales, A. Bioactive metabolites from a marine-derived strain of the fungus Emericella Variecolor. J Nat Prod 2002, 65, 364–367. [Google Scholar]
  177. Imamura, N; Nishijima, M; Adachi, K; Sano, H. Novel antimycin antibiotics, urauchimycins A and B, produced by marine actinomycete. J Antibiot 1993, 46, 241–246. [Google Scholar]
  178. Yu, C; Curtis, JM; Walter, JA; Wright, JLC; Ayer, SW; Kaleta, J; Querengesser, L; Fathi-Afshar, ZR. Potent inhibitors of cysteine proteases from the marine fungus Microascus longirostris. J Antibiot 1996, 49, 395–397. [Google Scholar]
  179. Namikoshi, M; Negishi, R; Nagai, H; Dmitrenok, A; Kobayashi, H. Three new chlorine containing antibiotics from a marine-derived fungus Aspergillus ostianus collected in Pohnpei. J Antibiot 2003, 56, 755–761. [Google Scholar]
  180. Kito, K; Ookura, R; Yoshida, S; Namikoshi, M; Ooi, T; Kusumi, T. Pentaketides Relating to Aspinonene and Dihydroaspyrone from a Marine-Derived Fungus Aspergillus ostianus. J Nat Prod 2007, 70, 2022–2025. [Google Scholar]
  181. Kito, K; Ookura, R; Yoshida, S; Namikoshi, M; Ooi, T; Kusumi, T. New Cytotoxic 14-Membered Macrolides from Marine-Derived Fungus Aspergillus ostianus. Org Lett 2008, 10, 225–228. [Google Scholar]
  182. Yoshida, S; Kito, K; Ooi, T; Kanoh, K; Shizuri, Y; Kusumi, T. 2008. Four Pimarane Diterpenes from Marine Fungus: Chloroform Incorporated in Crystal Lattice for Absolute Configuration Analysis by X-Ray. Chem Lett 2007, 36, 1386. [Google Scholar]
  183. Dettrakul, S; Kittakoop, P; Isaka, M; Nopichai, S; Suyarnsestakorn, C; Tanticharoen, M; Thebtaranonth, Y. Antimycobacterial pimarane diterpenes from the Fungus Diaporthe sp. Bioorg Med Chem Lett 2003, 13, 1253–1255. [Google Scholar]
  184. Höller, U; Wright, AD; Matthee, GF; König, GM; Draeger, S; Aust, H; Schulz, B. Fungi from marine sponges: Diversity, biological activity and secondary metabolites. Mycol Res 2000, 104, 1354–1365. [Google Scholar]
  185. Müller, WEG; Grebenjuk, VA; Pennec, G; Schröder, H; Brummer, F; Hentschel, U; Müller, IM; Breter, H. Sustainable production of bioactive compounds by sponges-cell culture and gene cluster approach: A review. Mar Biotechnol 2004, 6, 105–117. [Google Scholar]
  186. Petit, KE; Mondeguer, F; Roquebert, MF; Biard, JF; Pouches, YF. Detection of griseofulvin in a marine strain of Penicillium waksmanii by ion trap mass spectrometry. J Microbiol Methods 2004, 58, 59–65. [Google Scholar]
  187. Muth, WL; Nash, CH, III. Biosynthesis of Mycophenolic acid: Purification and characterization of S-Adenosyl-L-Methionine: Demethylmycophenolic Acid O-Methyltransferase. Antimicrob Agents Chemother 1975, 8, 321–327. [Google Scholar]
  188. Engel, G; Milczewski, KE; Prokopek, D; Teuber, M. Strain-specific synthesis of mycophenolic acid by P. roqueforti in blue-veined cheese. Appl Environ Microbiol 1982, 43, 1034–1040. [Google Scholar]
  189. Sabdono, A; Radjasa, OK. Microbial symbionts in marine sponges: Marine natural product factory. J Coast Dev 2008, 11, 57–61. [Google Scholar]
  190. Zhang, W; Zhang, F; Li, Z; Miao, X; Meng, Q; Zhang, X. Investigation of bacteria with polyketide synthase genes and antimicrobial activity isolated from South China Sea sponges. J Appl Microbiol 2009, 107, 567–575. [Google Scholar]
  191. Müller, WEG; Brummer, F; Batel, R; Müller, IM; Schröder, HC. Molecular biodiversity. Case study: Porifera (Sponges). Naturwissenschaften 2003, 90, 103–120. [Google Scholar]
  192. Hentschel, U; Usher, KM; Taylor, MW. Marine sponges as microbial fermenters. FEMS Microbiol Ecol 2006, 55, 167–177. [Google Scholar]
  193. Hoffmann, F; Rapp, HT; Reitner, J. Monitoring microbial community composition by Fluorescence In situ Hybridization during cultivation of the marine cold-water sponge Geodia barretti. Mar Biotechnol 2006, 8, 373–379. [Google Scholar]
  194. Taylor, MW; Schupp, PJ; Dahllof, I; Kjelleberg, S; Steinberg, PD. Host specificity in marine sponge-associated bacteria, and potential implications for marine microbial diversity. Environ Microbiol 2004, 6, 121–130. [Google Scholar]
  195. Webster, NS; Negri, AP; Munro, MMHG; Battershill, N. Diverse microbial communities inhabit Antarctic sponges. Environ Microbiol 2004, 6, 288–300. [Google Scholar]
  196. Wichels, A; Wurtz, S; Dopke, H; Schutt, C; Gerdts, G. Bacterial diversity in the breadcrumb sponge Halichondria panacea (Pallas). FEMS Microbiol Ecol 2006, 56, 102–118. [Google Scholar]
  197. Thiel, V; Neulinger, SC; Staufenberger, T; Schmaljohann, R; Imhoff, JF. Spatial distribution of sponge-associated bacteria in the Mediterranean sponge Tethya aurantium. FEMS Microbiol Ecol 2006, 59, 47–63. [Google Scholar]
  198. Taylor, MW; Schupp, PJ; Nys, R; Kjelleberg, S; Steinberg, PD. Biogeography of bacteria associated with the marine sponge Cymbastela concentrica. Environ Microbiol 2005, 7, 419–433. [Google Scholar]
  199. Giese, AC; Pearse, JS. Giese, AC, Pearse, JS, Eds.; Introduction: general principles. In Reproduction of Marine Invertebrates. Vol I. Acoelomate and Pseudocoelomate Metazoans; Academic Press: New York, USA, 1974; pp. 1–49. [Google Scholar]
  200. Turon, X; Becerro, MA; Uriz, MJ. Seasonal patterns of toxicity in benthic invertebrates: The encrusting sponge Crambe crambe (Pecilosclerida). Oikos 1996, 75, 33–46. [Google Scholar]
  201. Thakur, NL; Anil, AC. Antibacterial activity of the sponge Ircinia ramosa: Importance of its surface-associatedbacteria. J Chem Ecol 2000, 26, 57–71. [Google Scholar]
  202. Imhoff, JF; Stohr, R. Sponge-associated bacteria: general overview and special aspects of bacteria associated with Halichondria panicea. Prog Mol Subcell Biol 2003, 37, 35–57. [Google Scholar]
  203. Erpenbeck, D; van Soest, RWM. Status and perspective of sponge chemosystematics. Mar Biotechnol 2006, 9, 2–19. [Google Scholar]
  204. Proksch, P; Edrada-Ebel, R; Ebel, R. Drugs from the Sea-Opportunities and obstacles. Mar Drugs 2003, 1, 5–17. [Google Scholar]
Figure 1. Percentage distribution of compounds produced by different orders of Demospongiae in association with microbes.
Figure 1. Percentage distribution of compounds produced by different orders of Demospongiae in association with microbes.
Marinedrugs 08 01417f1
Figure 2. Percentage distribution of compounds produced by bacterial and fungal associates in sponges.
Figure 2. Percentage distribution of compounds produced by bacterial and fungal associates in sponges.
Marinedrugs 08 01417f2
Figure 3. Percentage distribution of compounds produced by associated bacteria- phylum wise.
Figure 3. Percentage distribution of compounds produced by associated bacteria- phylum wise.
Marinedrugs 08 01417f3
Figure 4. Percentage distribution of compounds produced by associated fungi-division wise.
Figure 4. Percentage distribution of compounds produced by associated fungi-division wise.
Marinedrugs 08 01417f4
Table 1. Current status of species producing clinically active compounds in association with microbes.
Table 1. Current status of species producing clinically active compounds in association with microbes.
Class: Demospongiae
OrderFamilySpeciesReference

AstrophoridaAncorinidaeStelletta tenuis[23,42]

Jaspis aff. johnstoni[4345]

ChondrosidaChondrillidaeChondrosia reniformis[4648]

DendroceratidaDarwinellidaeDendrilla nigra[5053]

DictyoceratidaDysideidaeLamellodysidea herbacea[40,57]
Dysidea sp.[59]
Dysidea avara[61,62]
IrciniidaeIrcinia fasciculata[33,64,65]
SpongiidaeHyatella sp.[66]
Spongia sp.[69]
ThorectidaeHyrtios altum[7072]
Hyrtios sp.[45,73]
Hyrtios proteus[75]
Fascaplysinopsis reticulata[48,76]

HadromeridaSpirastrellidaeSpirastrella vagabunda[77,78]
SuberitidaeSuberites domuncula[15,79]

HalichondridaAxinellidaePtilocaulis trachys[81,82]
Axinella sp. 1[82]
Axinella sp. 2[84]
Axinella verrucosa[8587]
Axinella damicornis[88]
Axinella sp. 3[89]
HalichondriidaeHalichondria okadai[78,9397]
Halichondria panacea[100,101]
Halichondria japonica[79,102,104,109113]
Acanthella acuta[116]
Hymeniacidon perlevis[43,44,117]

HaploscleridaCallyspongiidaeCallyspongia aerizusa[118]
Callyspongia vaginalis[119,120]
ChalinidaeHaliclona valliculata[121]
Haliclona simulans[25]
Haliclona sp. 1[123]
Haliclona sp. 2[2]
NiphatidaeNiphates olemda[124,125]
PetrosiidaePetrosia ficiformis[129131]
Xestospongia sp.[133]
Xestospongia exigua[79,124,134,135]
Acanthostrongylohpora sp.[82]
Petrosia sp.[139141]

LithistidaNeopeltidaeHomophymia sp.[142]
TheonellidaeTheonella swinhoei[145148,150]

PoeciloscleridaAcarnidaeZyzzya sp.[151,152]
IsodictyidaeIsodictya setifera[153]
RaspailiidaeEctyoplasia ferox[154]
MycalidaeMycale plumose[159,160]
Mycale adhaerens[48,162]
MyxillidaeMyxilla incrustance[79,158]

SpirophoridaTetillidaeCraniella australiensis[163]

VerongidaAplysinellidaeSuberea clavata[167]
AplysinidaeAplysina aerophoba[170,174]
Aplysina cavernicola[41]
PseudoceratinidaePseudoceratina purpurea[90]

Class: Calcarea
LeucettidaeLeucetta microraphis[120]
Table 2. Clinically important bioactive compounds from sponge-microbe associations.
Table 2. Clinically important bioactive compounds from sponge-microbe associations.
Class: Demospongiae
Order
Astrophorida
Family
Ancorinidae
SpongeSymbiontCompoundPropertyReference

Stelletta tenuis (South China Sea)Alcaligenes faecalis A72 (β-Proteobacteria)Cyclo-(L-Pro-L-Phe)Antimicrobial[42]
Jaspis aff. johnstoni (Indo-Pacific)Hyphomycete fungus (Deuteromycota (fungus))Chloriolin BAntitumor[4345]

Order
Chondrosida
Family
Chondrillidae
Chondrosia reniformis (Elba, Italy)Penicillium rugulosum (Ascomycota (fungus))Prugosene A1Anti-infective[4648]
Prugosene A2Anti-infective[4648]
Prugosene A3Anti-infective[4648]
Prugosene B1Anti-infective[4648]
Prugosene B2Anti-infective[4648]
Prugosene C1Anti-infective[4648]
Prugosene C2Anti-infective[4648]

Order
Dendroceratida
Family
Darwinellidae
Dendrilla nigra (Vizhinjam, India)Streptomyces dendra sp. nov. MSI051 (Actinobacteria)Unidentified compoundAntibacterial[51]

Dendrilla nigra (Kanyakumari, India)Streptomyces sp. BLT7 (Actinobacteria)Unidentified compoundAntibacterial[52,53]

Dendrilla nigra (South east coast, India)Nocardiopsis dassonvillei MAD08 (Actinobacteria)Acetic acid,-butyl-esterAntimicrobial[53]
Ethanol, 2-(octyloxy)-Antimicrobial[53]
Oxalic acid, allyl-nonyl esterAntimicrobial[54]
2-Isopropyl-5-methyl-1-heptanolAntimicrobial[53]
Butylated-hydroxytolueneAntimicrobial[53]
Cyclohexane-carboxylic acid, hexyl esterAntimicrobial[53]
Diethyl-phthalateAntimicrobial[53]
Pentadecanal-Antimicrobial[53]
1-TridecanolAntimicrobial[53]
9-OctadecenalAntimicrobial[53]
Hexadecanoic acid, methyl-esterAntioxidant, hypo-cholesterolemic, nematicide, antiandrogenic, hemolytic[53]
n-Hexadecanoic-acidAntioxidant, hypo-cholesterolemic, nematicide, antiandrogenic, hemolytic[53]
Hexadecanoic-acid, ethyl esterAntioxidant, hypo-cholesterolemic, nematicide, antiandrogenic, hemolytic[53]
9-Octadecenoic-acid-(Z)-, methyl-esterAnti-inflammatory, antiandrogenic, cancer-preventive, dermatitigenic, hypo-cholesterolemic, anemiagenic[53]
Oleic AcidAnti-inflammatory, antiandrogenic, cancer-preventive, dermatitigenic, hypo-cholesterolemic, anemiagenic[53]
(E)-9-Octadecenoic-acid ethyl esterAnti-inflammatory, antiandrogenic, cancer-preventive, dermatitigenic, hypo-cholesterolemic, anemiagenic[53]
9-Octa-decenamide-(Z)-Anti-inflammatory, antiandrogenic, cancer-preventive, dermatitigenic, hypo-cholesterolemic, anemiagenic[53]

Order
Dictyoceratida
Family
Dysideidae
Lamellodysidea herbacea (Great Barrier Reef, Australia)Oscillatoria spongeliae (Cyanobacteria)Dihydrodysamide CTherapeutic (unknown action)[57]
Didechloro-dihydrodysamide CTherapeutic (unknown action)[57]

Dysidea sp. (Eastern Samoa)Vibrio sp. (γ-Proteobacteria)Tetrabromo-diphenyl ethersCytotoxic, antibacterial[59]

Lamellodysidea herbacea (Republic of Palau)Oscillatoria spongeliae (Cyanobacteria)2-(2′,4′-dibromo-phenyl)-4,6-dibromophenolAnibacterial[40]

Dysidea avara (Adriatic Sea)Unidentified bacterium2-methylthio-1,4-naphthoquinoneAntiangiogenic, antimicrobial[61,62]

IrciniidaeIrcinia fasciculata (Mediterranean Sea)Penicillium chrysogenum (Ascomycota (fungus))Sorbicillactone AAntileukemic, anti HIV[64,65]

SpongiidaeHyatella sp.Vibrio sp. M22-1 (γ-Proteobacteria)AndrimidAntibiotic[66]

Spongia sp. (Hawaii)Myrothecium verrucaria 973023 (Deuteromycota (fungus))3-hydroxyroridin EAntileukemic, antitumor[69]
13′-acetyl-trichoverrin BAntileukemic, antitumor[69]
Roridin AAntileukemic, antitumor[69]
Roridin LAntileukemic, Antitumor[69]
Roridin MAntileukemic, Antitumor[69]
Verrucarin MAntileukemic, antitumor[69]
Verrucarin AAntileukemic, antitumor[69]
Isororidin AAntileukemic, antitumor[69]
Epiroridin EAntileukemic, antitumor[69]
Trichoverrin AAntileukemic, antitumor[69]
Trichoverrin BAntileukemic, antitumor[69]

ThorectidaeHyrtios altum (Okinawa)Vibrio sp. (γ-Proteobacteria)TrisindolineAntibiotic[7072]

Hyrtios sp. (Caribbean Sea)Aspergillus niger (Ascomycota (fungus))AsperazineAntileukemic, cytotoxic[45,74,75]

Hyrtios proteus (Dry Tortugas National Park, Florida )Aspergillus niger (Ascomycota (fungus))AsperazineAntileukemic, cytotoxic[76]
Malformin CAntitumor[76]
Fascaplysinopsis reticulate (Great Barrier Reef, Australia)Pseudo-alteromonas maricaloris KMM 636T (γ-Proteobacteria)Bromo-alterochromide ACytotoxic[48,76]

Bromo-alterochromide ACytotoxic[48,76]

Order
Hadromerida
Family
Spirastrellidae
Spirastrella vagabunda (Indonesia)Unidentified fungus14,15-secocurvularinAntibiotic[77,78]

SuberitidaeSuberites domuncula (Northern Adriatic Sea)α-Proteobacterium MBIC3368 (isolate 1)Unidentified compoundAntiangiogenic, antimicrobial, hemolytic, cytotoxic[15,79]
α-Proteobacterium MBIC3368 (isolate 2)Unidentified compoundAntimicrobial, hemolytic[15,80]
Idiomarina sp. (γ-Proteobacteria)Unidentified compoundHemolytic[15,80]
Pseudomonas sp. (isolate 1) (γ-Proteobacteria)Unidentified compoundHemolytic, cytotoxic[15,80]
Pseudomonas sp. (isolate 2) (γ-Proteobacteria)Unidentified compoundAntiangiogenic, antimicrobial, hemolytic, cytotoxic[15,80]

Order
Halichondrida
Family
Axinellidae
Ptilocaulis trachys (Enewetak Atoll, Marshall Island, Pacific Ocean)Lyngbya majuscula (Cyanobacteria)Majusculamide CAntifungal[81,82]

Axinella sp. (South China Sea)Myrothecium sp. JS9 (Deuteromycota (fungus))Roridin AAntifungal[83]
Roridin DAntifungal[83]

Axinella sp. (Papaua New Guinea)Penicillium citrinum (Ascomycota (fungus))Isocyclocitrinol AAntibacterial[84]
22-acetyl-isocyclocitrinol AAntibacterial[84]

Axinella verrucosa (Mediterranean Sea)Penicillium sp. (Ascomycota (fungus))OxalineAnti-proliferative[85]
GriseofulvinAntifungal[85,86]
Communesin BAntileukemic[85,87]
Communesin CAntileukemic[85,87]
Communesin DAntileukemic[85,87]

Axinella sp. (Papua New Guinea)Acremonium sp. (Ascomycota (fungus))Efrapeptin ECytotoxic, antibacterial[90]
Efrapeptin FCytotoxic, antibacterial[90]
Efrapeptin EαCytotoxic, antibacterial[90]
Efrapeptin GCytotoxic, antibacterial[89]
Efrapeptin HCytotoxic, antibacterial[90]
RHM1Antibacterial[89]

Axinella damicornis (Mediterranean Sea)Aspergillus niger (Ascomycota (fungus))BicoumanigrinAnticancer, cytotoxic[88]
Aspernigrin BNeuroprotective[88]

HalichondriidaeHalichondria okadaiAlteromonas sp. (γ-Proteobacteria)Alteramide AAnticancer, cytotoxic[9395]

Halichondria okadai (Japan)Trichoderma harzianum OUPS-N115 (Ascomycota (fungus))Trichodenone AAntileukemic, cytotoxic[78,96,97]
Trichodenone BAntileukemic, cytotoxic[78,96,97]
Trichodenone CAntileukemic, cytotoxic[78,96,97]

Halichondria okadaiRubritalea squalenifasciens HOact23T (Verrucomicrobiae)Dia-polycopenedioic acid xylosyl esters AAntioxidant[30,48,98]
Dia-polycopenedioic acid xylosyl esters BAntioxidant[30,48,98]
Dia-polycopenedioic acid xylosyl esters CAntioxidant[30,48,98]

Halichondria panaceaUnidentified bacteriumUnidentified compoundNeuroactive[100]

Halichondria panacea (Adriatic coast, Croatia)Microbacterium sp. (Actinobacteria)1-O-acyl-3-[R-glucopyranosyl-(1–3)-(6-O-acyl-R-manno-pyranosyl)]-glycerolAntitumor[101]

Halichondria japonica (Osaka Bay, Japan)Gymnascella dankaliensis OUPS-N134 (Ascomycota (fungus))Gymnostatin AAntileukemic, cytotoxic[78,102,103,105]
Gymnostatin BAntileukemic, cytotoxic[78,102,103,105]
Gymnostatin CAntileukemic, cytotoxic[78,102,103,105]
Gymnostatin FAntileukemic, cytotoxic[106]
Gymnostatin GAntileukemic, cytotoxic[106]
Gymnostatin QAntileukemic, anti cancer, cytotoxic[107]
Gymnostatin RAntileukemic, cytotoxic[107]
Gymnasterone ACytotoxic[108,109]
Gymnasterone BAntileukemic, cytotoxic[108,109]
Gymnasterone CAntileukemic, cytotoxic[108]
Gymnasterone DAntileukemic, cytotoxic[108]
Dankastatin AAntileukemic, cytotoxic[107]
Dankastatin BAntileukemic, cytotoxic[107]
Dankasterone AAntileukemic, anticancer, cytotoxic[104]

Halichondria Japonica (Japan)Phoma sp. Q60596 (Ascomycota (fungus))YM-202204Antifungal[110]

Halichondria JaponicaBacillus cereus QN03323 (Firmicutes)YM-266183Antibacterial[111113]
YM-266184Antibacterial[111113]

Acanthella acuta (Mediterranean Sea)Bacillus pumilus AAS3 (Firmicutes)GG11Antitumor[116]

Hymeniacidon perlevis (Nanji Island, China Sea)Pseudo-alteromonas piscicida NJ6-3-1 (γ-Proteobacteria)NorharmanAntimicrobial[43,117]

Hymeniacidon perlevis (Fujiazhuang coast, China)Fusarium oxysporum DLFP2008005 (Ascomycota (fungus))Unidentified compoundAntibacterial, antifungal[44]

Order
Haplosclerida
Family
Callyspongiidae
Callyspongia aerizusa (Indonesia)Cladosporium herbarum (Deuteromycota (fungus))Sumiki’s acidAntibacterial[118]
Acetyl Sumiki’s acidAntibacterial[118]

Callyspongia vaginalis (Caribbean Sea)Ulocladium botrylis 193A4 (Ascomycota (fungus))UlocladolAntimicrobial[119,120]
1-hydroxy-6-methyl-8-(hydroxylmethyl)-xanthoneAntifungal[119,120]

ChalinidaeHaliclona valliculata (Elba, Italy)Emericella variecolor (Ascomycota (fungus))EvariquinoneAnti-proliferative[122]

Haliclona simulans (Ireland)Pseudo-alteromonas sp. PA2 (γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Pseudo-alteromonas sp. PA4 (γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Pseudo-alteromonas sp. PA5 (γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Pseudo-alteromonas sp. PA5 (γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Halomonas sp. HM4 ( γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Psychrobacter sp. PB1 (γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Marinobacter sp. MB1 (γ-Proteobacteria)UnidentifiedAntimicrobial[123]
Pseudovibrio sp. PV1 (α-Proteobacteria)UnidentifiedAntimicrobial[123]
Pseudovibrio sp. PV2 ( α-Proteobacteria)UnidentifiedAntimicrobial[123]
Pseudovibrio sp. PV4 ( α-Proteobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM1 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM2 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM3 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM4 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM5 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM6 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM7 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM8 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM9 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM10 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM11 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM12 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM14 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM16 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM17 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM18 (Actinobacteria)UnidentifiedAntimicrobial[123]
Streptomyces sp. SM19 (Actinobacteria)UnidentifiedAntimicrobial[123]
Bacillus sp. BC1 (Firmicutes)UnidentifiedAntimicrobial[123]
Bacillus sp. BC2 (Firmicutes)UnidentifiedAntimicrobial[123]

Haliclona sp. (Tomini Bay, North Sulawesi, Indonesia)Unidentified fungusHirsutanol AAntibiotic[123]
ent-gloeosteretriolAntibiotic[123]

Haliclona sp.(North Java Sea, Indonesia)Unidentified bacterium 1UnidentifiedAntibacterial[2]
Unidentified bacterium 2UnidentifiedAntibacterial[2]
Unidentified bacterium 3UnidentifiedAntibacterial[2]
Unidentified bacterium 4UnidentifiedAntibacterial[2]
Unidentified bacterium 5UnidentifiedAntibacterial[2]

NiphatidaeNiphates olemda (Indonesia)Curvularia lunata (Ascomycota (fungus))LunatinAntibacterial[124,125]
Cytoskyrin AAntibacterial[124,125]

PetrosiidaePetrosia ficiformis (Capo S. Andrea, Elba, Italy)Penicillium brevicompactum (Ascomycota (fungus))Mycophenolic acidImmuno-suppressant[128]
Aspergillus insuetus (Ascomycota (fungus))Terretonins EInhibit mammalian mitochondrial respiratory chain[132]
Terretonins FInhibit mammalian mitochondrial respiratory chain[132]

Petrosia sp. (Jeju Island, Korea)Aspergillus versicolor (Ascomycota (fungus))Decumbenone AMelanin inhibitor[139]
Fellutamide CCytotoxic[140]

Xestospongia sp. (Off Noumea (New Caledonia, southwest Pacific))Micrococcus luteus R-1588-10 (Actinobacteria)2,4,4′-trichloro-2′-hydroxy-diphenylether (Triclosan)Antimicrobial[133]
Acyl-1-(acyl-6′-mannobiosyl)-3-glycerol (Lutoside)Antimicrobial[133]

Xestospongia exigua (Bali Sea, Indonesia)Penicillium cf. montanense (Ascomycota (fungus))Xestodecalactone BAntifungal[79,134]

Xestospongia exigua (Indonesia)Aspergillus versicolor (Ascomycota (fungus))AspergillitineAntibacterial[124,135]

Acantho-strongylophora sp. (Indonesia)Micromonospora sp. (Actinobacteria)Manzamine AAntitumor, antimalarial[82,137, 138]

Order
Lithistida
Family
Neopeltidae
Homophymia sp. (Off Touho, New Caledonia)Pseudomonas sp. 1537-E7 (γ-Proteobacteria)2-undecyl-4-quinoloneAntimalarial Anti HIV[142]
2-undecen-1′-yl-4-quinoloneCytotoxic[142]
2-nonyl-4-hydroxy-quinoline N-oxideAntibacterial, cytotoxic[142]

TheonellidaeTheonella swinhoei (Palau)Unidentified bacteriumSwinholide ACytotoxic[144,145]
Candidatus Entotheonella palauensis (δ-Proteobacteria)TheopalauamideAntifungal[26,144,146]

Theonella swinhoei (Philippines)Entotheonella palauenis (δ-Proteobacteria)TheonegramideAntifungal[145,147]

Theonella swinhoei (Hachijojima Island, Japan)Uncultured bacteriumOnnamide AAntitumor[149,150]

Order
Poecilosclerida
Family
Acarnidae
Zyzzya sp. (Fiji)Penicillium brocae (Ascomycota (fungus))Brocaenol ACytotoxic[151,152]
Brocaenol BCytotoxic[151,152]
Brocaenol CCytotoxic[151,152]

IsodictyidaeIsodictya setifera (Hut Point and Danger Slopes, Ross Island, Antarctica)Pseudomonas aeruginosa (γ-Proteobacteria)Cyclo-(L-proline-L-methionine)Antibacterial[153]

RaspailiidaeEctyoplasia ferox (Dominica, Carribean Island)Coniothyrium sp. 193477 (Deuteromycota (fungus))(3S)-(3′,5′-dihydroxyphenyl) butan-2-oneAntimicrobial[154]
2-(1′(E)-propenyl)-octa-4(E),6(Z)-diene-1,2-DiolAntimicrobial[154]
(3R) 6-methoxymelleinAntimicrobial[154]
(3R)-6-methoxy-7-chloromelleinAntimicrobial[154]
Crypto-sporiopsinolAntimicrobial[154]
Phoma sp. (Ascomycota (fungus))Epoxyphomalin AAntitumor[155]
Spicellum roseum 193H15 (Deuteromycota (fungus))TrichodermolAnticancer[156,157]
8-deoxytrichothecinAnticancer[156,157]

MycalidaeMycale plumose (Qingdao coast, China)Saccharopolyspora sp. nov. (Actinobacteria)Metacyclo-prodigiosinAnticancer[159]
Undecyl-prodigiosinAnticancer[159]
Penicillium auratiogriseum (Ascomycota (fungus))(S)-2,4-dihydroxy-1-butyl(4-hydroxy)-benzoateAntitumor[160]
Fructigenin AAntitumor[160]
Aurantiomide BCytotoxic[161]
Aurantiomide CCytotoxic[161]

Mycale adhaerensExophiala pisciphila N110102 (Ascomycota (fungus))Exophilin AAntibacterial[48,162]

MyxillidaeMyxilla incrustance (Helgoland, Germany)Microsphaeropsis sp. H5-50 (Anamorphic fungus)MicrosphaeropsisinAntifungal[79,154]
(R)-melleinAntimicrobial[154]
(3R,4S)-hydroxymelleinAntimicrobial[154]
(3R,4R)-hydroxymelleinAntimicrobial[154]
4,8-dihydroxy-3,4-dihydro-2H-naphthalen-1-oneAntimicrobial[154]

Order
Spirophorida
Family
Tetillidae
Craniella australiensis (South China Sea)Streptomyces sp. DA11 (Actinobacteria)ChitinaseAntifungal[163]

Order
Verongida
Family
Aplysinellidae
Suberea clavata (Great Barrier Reef, Australia)Salinospora sp. (Actinobacteria)Rifamycin BAntibiotic[164,166]
Rifamycin SVAntibiotic[164,166]

AplysinidaeAplysina aerophoba (Mediterranean coast, France)Bacillus subtilis A184 (Firmicutes)Surfactin, iturin and fengycinAntifungal, antibacterial, hemolytic[170]
Bacillus subtilis A190 (Firmicutes)SurfactinAntifungal, hemolytic[170]
Bacillus subtilis A202 (Firmicutes)IturinAntifungal, hemolytic[170]
Bacillus pumilus A586 (Firmicutes)Pumilacidin containing β-hydroxy fatty-acidAntibacterial[170]

Aplysina aerophoba (Mediterranean Sea)Microsphaeropsis sp. (Anamorphic fungus)10-Hydroxy-18-methoxyl-betaenoneProtein Kinase C ɛ inhibitor[174]

Aplysina aerophoba (Banyuls sur Mer)Bacillus sp. SB8 (Firmicutes)Unidentified compoundAntibacterial[41]

Bacillus sp. SB17 (Firmicutes)Unidentified compoundAntibacterial[41]
Micrococcus sp. SB58 (Actinobacteria)Unidentified compoundAntibacterial[41]
Enterococcus sp. SB91 (Firmicutes)Unidentified compoundAntibacterial[41]
Arthrobacter sp. SB95 (Actinobacteria)Unidentified compoundAntibacterial[41]
Unidentified bacteria SB122Unidentified compoundAntibacterial[41]
Unidentified bacteria SB144Unidentified compoundAntibacterial[41]
α-Proteobacteria SB6Unidentified compoundAntibacterial[41]
α-Proteobacteria SB55Unidentified compoundAntibacterial[41]
α-Proteobacteria SB63Unidentified compoundAntibacterial[41]
α-Proteobacteria SB89Unidentified compoundAntibacterial[41]
α-Proteobacteria SB156Unidentified compoundAntibacterial[41]
α-Proteobacteria SB197Unidentified compoundAntibacterial[41]
α-Proteobacteria SB202Unidentified compoundAntibacterial[41]
α-Proteobacteria SB207Unidentified compoundAntibacterial[41]
α-Proteobacteria SB214Unidentified compoundAntibacterial[41]
Vibrio halioticoli SB177 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB181 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB182 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB183 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB185 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseud-oalteromonas sp. SB186 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB192 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB194 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB200 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB208 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]
Pseudo-alteromonas sp. SB213 (γ-Proteobacteria)Unidentified compoundAntibacterial[41]

Pseudo-ceratinidaePseudoceratina purpurea (Fiji)Metarrhizium sp. 001103 (Ascomycota (fungus))Destruxin AAntitumor[90]
Destruxin B2Antitumor[90]
Desmethyl BAntitumor[90]
E chlorohydrinAntitumor[90]
E2 chlorohydrinAntitumor[90]

Class: Calcarea
Order
Clathrinida
Family
Leucettidae
Leucetta microraphis (Great Barrier Reef, Australia)Unidentified cyanobacteriaLeucamide AAntitumor[120]

Unidentified sponges
UnidentifiedUnidentified (Venezuela)Emericella variecolor (Ascomycota (fungus))VaritriolAnticancer[176]
VarixanthoneAntimicrobial[176]

UnidentifiedStreptomyces sp. Ni-80 (Actinobacteria)Urauchimycin AAntibiotic[177]
Urauchimycin BAntibiotic[177]

Unidentified (Harrington Point, Otago Harbor, New Zealand)Microascus longirostris SF-73 (Ascomycota (fungus))Cathestatin ACysteine protease inhibitor[178]
Cathestatin BCysteine protease inhibitor[178]
Cathestatin CCysteine protease inhibitor[178]

Unidentified (Pohnpei, The federated state of Micronesia)Aspergillus ostianus 01F313 (Ascomycota (fungus))8-chloro-9-hydroxy-8,9-deoxyasperlactoneAntibacterial[179]
9-chloro-8-hydroxy-8,9-deoxyasperlactoneAntibacterial[179]
9-chloro-8-hydroxy-8,9-deoxyaspyroneAntibacterial[179]
AspinoneneAntileukemic[48,180]
DihydroaspyroneAntileukemic[48,180]
Aspergillide AAntileukemic[48,181]
Aspergillide BAntileukemic[48,181]
Aspergillide CAntileukemic[48,181]
Cryptosphaeria eunomia (Ascomycota (fungus))Diaporthein AAntibacterial[48,182]
Diaporthein BAntibacterial[48,182]
Table 3. Chemical diversity of therapeutics produced by sponge-microbe associations.
Table 3. Chemical diversity of therapeutics produced by sponge-microbe associations.
CategoryChemical diversity

AntiandrogenicFattyacid esters, fatty acids
AntiangiogenicQuinone
AnticancerQuinone, steroid, fatty acid esters, fatty acids, diketopiperazine, alkaloid, terpenes, terpenoids, trichoverroids, prodigiosin derivative
AntiHIVQuinolone derivative
Anti-inflammatoryFatty acid esters, fatty acid
AntimalarialAlkaloid, quinolone derivative
AntimicrobialPolyketide, glycopeptides, α-pyrone derivative, peptide, protein, antimycin, lipopeptides, polybrominated biphenyl ether, cyclic depsipeptide, terpenes, pentaketides, furan carboxylic acid, alkaloid, diketopiperazine, anthraquinone, chromones, steroid, lactone, quinolone derivative, trisindole derivative, macrolactam, ethers, phenol derivative
AntiinfectivePolyketides
AntioxidantFatty acid esters, fatty acid, carotenoic acid
Anti-respiratoryTerpenoids
AntitumorDiglucosyl-glycerol, polyketides, alkaloids, cyclopeptides, glycoglycerolipid, benzoic acid derivative, terpenoids, terpenes, trichoverroids
HemolyticFatty acid ester, fatty acids
HypocholesterolemicFatty acid ester, fatty acids
ImmunosupressantMycophenolic acid
Melanin inhibitorPolyketide
NematicideFatty acid ester, fatty acids
NeuroactiveUnknown
NeuroprotectiveDihydropyridine
Table 4. Microbial groups in various orders of sponges producing functionally diverse therapeutics.
Table 4. Microbial groups in various orders of sponges producing functionally diverse therapeutics.
SymbiontSponge orderCompound fuction
BacteriaDendroceratidaAntiandrogenic
BacteriaDictyoceratida, HadromeridaAntiangiogenic
BacteriaHalichondrida, Dendroceratida, PoeciloscleridaAnticancer
FungiDictyoceratida, Halichondrida, Haplosclerida, Poecilosclerida
BacteriaLithistidaAntiHIV
FungiDictyoceratida
FungiChondrosidaAnti-infective
BacteriaDendroceratidaAnt-inflammatory
BacteriaLithistida, HaploscleridaAntimalarial
BacteriaAstrophorida, Dendroceratida Dictyoceratida, Hadromerida, Haplosclerida, Halichondrida, Lithistida, Poecilosclerida, Spirophorida, VerongidaAntimicrobial
FungiHadromerida, Halichondrida, Haplosclerida, Poecilosclerida
BacteriaDendroceratida, HalichondridaAntioxidant
FungiHaploscleridaAnti-respiratory
BacteriaClathrinida, Halichondrida, Haplosclerida, LithistidaAntitumor
FungiAstrophorida, Dictyoceratida, Poecilosclerida, Verongida
BacteriaHadromerida, DendroceratidaHemolytic
BacteriaDendroceratidaHypocholesterolemic
FungiHaploscleridaImmunosuppressant
FungiHaploscleridaMelanin inhibitor
BacteriaDendroceratidaNematicide
BacteriaHalichondridaNeuroactive
FungiHalichondridaNeuroprotective

Share and Cite

MDPI and ACS Style

Thomas, T.R.A.; Kavlekar, D.P.; LokaBharathi, P.A. Marine Drugs from Sponge-Microbe Association—A Review. Mar. Drugs 2010, 8, 1417-1468. https://doi.org/10.3390/md8041417

AMA Style

Thomas TRA, Kavlekar DP, LokaBharathi PA. Marine Drugs from Sponge-Microbe Association—A Review. Marine Drugs. 2010; 8(4):1417-1468. https://doi.org/10.3390/md8041417

Chicago/Turabian Style

Thomas, Tresa Remya A., Devanand P. Kavlekar, and Ponnapakkam A. LokaBharathi. 2010. "Marine Drugs from Sponge-Microbe Association—A Review" Marine Drugs 8, no. 4: 1417-1468. https://doi.org/10.3390/md8041417

Article Metrics

Back to TopTop