Next Article in Journal
Antimicrobial Chlorinated 3-Phenylpropanoic Acid Derivatives from the Red Sea Marine Actinomycete Streptomyces coelicolor LY001
Next Article in Special Issue
Viridicatol Isolated from Deep-Sea Penicillium Griseofulvum Alleviates Anaphylaxis and Repairs the Intestinal Barrier in Mice by Suppressing Mast Cell Activation
Previous Article in Journal
Extraction, Enrichment, and LC-MSn-Based Characterization of Phlorotannins and Related Phenolics from the Brown Seaweed, Ascophyllum nodosum
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Structural Diversity of Marine Microbial Secondary Metabolites Based on Co-Culture Strategy: 2009–2019

1
College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
2
Phytochemistry and Plant Systematics Department, National Research Centre, 33 El Bohouth St., Dokki, Giza 12622, Egypt
*
Author to whom correspondence should be addressed.
Mar. Drugs 2020, 18(9), 449; https://doi.org/10.3390/md18090449
Submission received: 28 July 2020 / Revised: 25 August 2020 / Accepted: 25 August 2020 / Published: 27 August 2020
(This article belongs to the Special Issue Marine Microbial Diversity as Source of Bioactive Compounds)

Abstract

:
Marine microorganisms have drawn great attention as novel bioactive natural product sources, particularly in the drug discovery area. Using different strategies, marine microbes have the ability to produce a wide variety of molecules. One of these strategies is the co-culturing of marine microbes; if two or more microorganisms are aseptically cultured together in a solid or liquid medium in a certain environment, their competition or synergetic relationship can activate the silent biosynthetic genes to produce cryptic natural products which do not exist in monocultures of the partner microbes. In recent years, the co-cultivation strategy of marine microbes has made more novel natural products with various biological activities. This review focuses on the significant and excellent examples covering sources, types, structures and bioactivities of secondary metabolites based on co-cultures of marine-derived microorganisms from 2009 to 2019. A detailed discussion on future prospects and current challenges in the field of co-culture is also provided on behalf of the authors’ own views of development tendencies.

Graphical Abstract

1. Introduction

Although many industrial sectors have stopped their dependence on natural product (NP) drug discovery programs, NPs are still of great interest to many pharmaceutical communities and are important sources of bioactive compounds [1,2]. Marine microbes, as an important source of bioactive NPs, have elicited widespread attention [3,4,5]. However, the discovery of novel marine microbial NPs is becoming more difficult and the rate of rediscovery of known NPs is being gradually increased. On the other hand, recent genomic sequencing has revealed the presence of numerous biosynthetic gene clusters in some microbes that may be responsible for the biosynthesis of NPs which are not found under classical cultivation conditions [6,7]. Therefore, many alternative strategies have been explored to activate these silent and cryptic biosynthetic genes. The co-culturing of marine microbes involves the culturing of two or more marine microbes together on/in certain conditions; microorganisms can communicate with each other through direct or indirect contact, thereby stimulating the silent gene clusters to produce special NPs [2,8] (Figure 1). This strategy can promote the production of complex and novel skeletons with numerous stereocenters [9,10,11]. Hence, the co-culturing of marine microbes draws widespread attention in the scientific community as a potential source of unknown bioactive substances classified as alkaloids, polyketides, anthraquinone, flavonoids, cyclopeptides, etc. To exploit the NPs from the co-cultures of marine microbes and understand their medicinal significance, this review summarizes successful examples involved in NPs of marine microbes based on co-cultures from 2009 to 2019 (Table 1).

2. Compounds Derived from the Co-Cultures of Marine Microorganisms

Co-culturing or mixed fermentation is considered an important technique of inducing secondary metabolites hidden in the genomes of marine microbes by using appropriate physiological conditions, chemical communication and competition of microbes. Consequently, it is considered an easy, cheap and effective method [12,13]. This finding also explains the chemical communication and antagonism between different marine microorganisms, such as the interactions between marine fungi−fungi, fungi−bacteria and bacteria−bacteria, in which they act as signaling molecules, competitors or defense agents [14]. Herein, the metabolites based on co-cultures of marine microbes were classified according to their skeletons as alkaloids, anthraquinones, cyclopeptides, flavonoids, macrolides, phenylpropanoids, polyketides, steroids, terpenoids and others from 2009–2019. These excellent examples were found from SciFinder, Science Direct, PubMed, Springer and other databases. Among them, the interactions between marine fungi and bacteria were found to induce the most metabolites (Figure 2A), and the alkaloids played a significant role in co-cultures of marine microbes (Figure 2B), no matter whether the mixed cultivation was of marine fungi−fungi (Figure 2C), fungi−bacteria (Figure 2D) or bacteria−bacteria (Figure 2E).

2.1. Alkaloids

The nitrogenous alkaloids represented the most abundant class of compounds that were produced by the co-cultures of marine microorganisms with diverse skeletons and biological activities [15,16]. Eighty alkaloidal metabolites were isolated and identified from different microbial environments (Figure 2B), and the co-cultures of marine fungi–bacteria represented 51% of the total isolates (Figure 3).

2.1.1. Alkaloids Derived from the Co-Cultures of Different Marine Fungi

Several studies of co-cultures of fungal–fungal interactions from different marine sources were summarized as follows; 26 alkaloids were isolated and identified (Figure 2C and Figure 3). The mixed fermentation of marine-derived fungi Aspergillus sulphureus KMM 4640 from muddy sand of the eastern Sakhalin shelf (Sea of Okhotsk, 26 m depth) and Isaria felina KMM 4639 from sediments (South China Sea, Vietnam shores, 10 m depth), led to the production of five novel prenylated indole alkaloids, 17-hydroxynotoamide D (1), 17-O-ethylnotoamide M (2), 10-O-acetylsclerotiamide (3), 10-O-ethylsclerotiamide (4) and 10-O-ethylnotoamide R (5) together with known compounds (‒)-notoamide B (6), notoamide C (7), dehydronotoamide C (8), notoamide D (9), notoamide F (10), notoamide Q (11), 17-epi-notoamide Q (12), notoamide M (13) and sclerotiamide (14) (Figure 4) [17]. Among them, compounds 15 were only produced in the co-culturingprocess.
Compounds 2, 6, 8, 13 and 14 inhibited the proliferation of the human prostate cancer cells 22Rv1 at 100 μM. Notably, 2 and 13 drastically reduced the viability of 22Rv1 prostate cancer cells at 10 μM by 25% and 55%, respectively. 22Rv1 cancer cell lines were resistant to hormone therapy at conventional chemotherapy including two new 2nd generation drugs enzalutamide and abiraterone owing to the presence of the androgen receptor splice variant-7 (AR-V7). Therefore, the active NPs drugs in these cells might be further investigated in the treatment of different human drug-resistant prostate cancer. 6 and 7 displayed weak cytotoxicity against HeLa and L1210 cell lines with half maximal inhibitory concentration (IC50) in the range of 22–52 µg/mL [18]. Although 6 and 7 had the similar structure with 9, compound 9 did not display the similar cytotoxic activity against HeLa and L1210 cell lines. The significant difference in cytotoxicity might be attributed to the possible existence of pyrroloindole system in 9 rather than the dihydroxypyrano-2-oxindole ring system of 6 and 7 [18,19]. In addition, compounds 1, 2, 5, 9, 13 and 14 did not exhibit any cytotoxicity against human non-malignant (HEK 293 T and MRC-9) or malignant (PC-3, LNCaP, and 22Rv1) cell lines at concentrations up to 100 μM for 48 h [17].
The co-fermentation of marine mangrove epiphytic fungi Aspergillus sp. FSY-01 and FSW-02 collected from a rotten fruit of mangrove Avicennia marina in Zhanjiang, Guangdong Province, China, yielded a new alkaloid, aspergicin (15), together with two known secondary metabolites, neoaspergillic acid (16) and aspergicine (17) (Figure 5) [20,21]. Notably, compounds 17 and 15 are chemically isomers, and consequently aspergicine (17) may be the precursor of aspergicin (15) through a proton 1, 2-shift [22].
Compounds 15 and 16 showed potent inhibitory activities against three Gram-positive bacteria, Bacillus subtilis (MIC, minimum inhibitory concentration that inhibits the growth of microbes by 80%, 15.62 and 1.95 μg/mL), Staphylococcus epidermidis (MIC 31.25 and 0.49 μg/mL) and Staphylococcus aureus (MIC 62.50 and 0.98 μg/mL), and three Gram-negative bacteria, Escherichia coli (MIC 31.25 and 15.62 μg/mL), Bacillus proteus (MIC 62.50 and 7.80 μg/mL) and Bacillus dysenteriae (MIC 15.62 and 7.80 μg/mL), respectively [22].
Marine fungi Aspergillus sclerotiorum SCSGAF 0053 and Penicillium citrinum SCSGAF 0052 were isolated from the gorgonian corals Muricella flexuosa collected from South China Sea, Sanya (18°11′ N, 109°25′ E), Hainan Province, China [23]. Due to the mixed fermentation of marine fungi, a red pigment appeared in the mixed fermentation broth could not be observed in any strain cultured separately. This special phenomenon suggested that a novel biosynthesis route was activated. Four novel alkaloids were obtained, including one oxadiazin derivative sclerotiorumin C (18), a pyrrole derivative 1-(4-benzyl-1H-pyrrol-3-yl) ethanone (19), aluminumneohydroxyaspergillin (20) and ferrineohydroxyaspergillin (21), together with one known compound ferrineoaspergillin (22) (Figure 6) [23]. Compounds 1821 were only produced in the co-culture process.
Compound 20 exhibited potent toxicity towards brine shrimp with medium lethal concentration (LC50) value of 6.1 μM and high selective cytotoxicity towards histiocytic lymphoma U937 cell line with an IC50 value of 4.2 μM. 19, 21, and 22 showed moderate toxicity against brine shrimp with LC50 values of 46.2, 11.5 and 27.8 μM, respectively. 21 and 22 possessed mild cytotoxicity against U937 with IC50 values of 42.0 and 48.0 μM, respectively. These results suggested that the aluminum complex skeletons of compounds showed more potent toxicity and cytotoxicity than ferricomplex structures of compounds [23,24,25,26]. Moreover, aspergillic acid and 16 also showed more potent inhibitory activities than neohydroxyaspergillic acid and hydroxyaspergillic acid against B. subtilis, E. coli, S. aureus and Candida albicans [27].
The co-culture of mangrove fungi Phomopsis sp. K38 and Alternaria sp. E33 led to the identification of one new diimide derivative, (-)-byssochlamic acid bisdiimide (23) and a novel nonadride derivative, (-)-byssochlamic acid imide (24) (Figure 7) [28,29]. Ebada et al. (2014) investigated the mycelial extract of a co-cultivation of marine fungal strains Aspergillus. BM-05 and BM-05ML, and identified two alkaloids, protuboxepin A (25) and oxepinamide E (26) (Figure 7) [30]. 2324 were only found in the co-culture process.
Compound 23 exhibited moderate inhibitory activity against HepG2 and Hep-2 with IC50 values of 51 μg/mL and 45 μg/mL, respectively. 24 had moderate antifungal activities against Fusarium oxysporum and Fusarium graminearum with MIC values of 60 μg/mL and 50 μg/mL, respectively [28,29,31]. 25 possessed anti-proliferative activity against human breast cancer adenocarcinoma MDA-MB-231, human acute promyelocytic leukemia HL-60, hepatocellular carcinoma Hep3B, chronic myelogenous leukemia K562 and rat fibroblast 3Y1 cell lines with IC50 values of 130, 75, 150, 250 and 180 μM, respectively [32,33]. 26 showed transcriptional activation on liver X receptor α (LXRα) with a half maximal effective concentration (EC50) value of 12.8 µM. It was known that LXR was an important target in drug discovery; LXR agonists had been proven to exhibit remarkable therapeutic effects on diabetes, atherosclerosis, Alzheimer’s disease and anti-inflammation. Therefore, 26 was worthy of consideration as a potential lead compound for drug discovery [34].

2.1.2. Alkaloids Derived from the Co-Cultures of Marine Fungi and Bacteria

The alkaloids derived from the co-culture of marine fungi and bacteria were tallied to be 41 isolates (Figure 2D and Figure 3) and can be described as follows; prenylated 2,5-diketopiperazines (2,5-DKPs) were isolated from the co-culture of marine Penicillium sp. DT-F29 isolated from marine sediments of Dongtou country, China, and Bacillus sp. B31 collected from marine sediments of Changzhi Island, China [35], including ten novel metabolites, 12-β-hydroxy-13-butoxyethoxyfumitremorgin B (27), diprostatin A (28), 12-hydroxy-13α-ethoxyverruculogen TR-2 (29), hydrocycloprostatin A (30), 12-β-hydroxy-13α-butoxyethoxyverruculogen TR-2 (31), hydrocycloprostatin B (32), 26-α-hydroxyfumitremorgin A (33), 25-hydroxyfumitremorgin B (34), 12-β-hydroxy-13α-methoxyverruculogen (35), 25-hydroxyfumitremorgin A (36) and thirteen known isolates, verruculogen TR-2 (37), 12-α-hydroxy-13-α-prenylverruculogen TR-2 (38), 12-hydroxyverruculogen TR-2 (39), 13-prenyl fumitremorgin B (40), 12-β-hydroxy-13-α-methoxyverruculogen TR-2 (41), cycloprostatin C (42), cyclotryprostatin B (43), spirotryprostatin C (44), 12,13-dihydroxyfumitremorgin C (45), neofipiperzine C (46), prenylcycloprostatin B (47), fumitremorgin B (48) and fumitremorgin A (49) (Figure 8).
The secondary metabolites profile of the co-culture of Streptomyces sp. and Aspergillus flavipes, obtained from marine sediments of the Nanji Islands of the same habitat, showed an induced biosynthesis of a series of known cytochalasans, including rosellichalasin (50), and five aspochalasins (aspochalasin E 51, aspochalasin P 52, aspochalasin H 53, aspochalasin M 54 and 19,20-dihydro-aspochalasin D 55) (Figure 9) [36]. The chromatographic purification of the combination culture extract from marine-derived Aspergillus fumigatus MR2012 and Streptomyces leeuwenhoekii C34 led to the isolation of two novel compounds, luteoride D (56) and pseurotin G (57), along with the known isolates, nocardamine (58), terezine D (59), 11-O-methylpseurotin A (60) and lasso peptide chaxapeptin (61) [37]. In addition, seven known compounds, notoamide D (9), speramide B (62), notoamide E (63), stephacidin A (64), notoamide R (65), protuboxepin B (66) and 3,10-dehydrocyclopeptine (67) (Figure 9) were identified from the mixed-fermentation of the marine-derived fungus Aspergillus versicolor isolated from sponge Agelas oroides and B. subtilis [38].
Compounds 27, 28, 3840, 44 and 4649 displayed strong inhibitory effects on bromodomain-containing protein 4 (BRD4) at 20 µM. Notably, 39 and 48 exhibited the most inhibitory activity with 72.7% and 80.4%, compared with the positive control, BRD4 inhibitor (+)-JQ1 (85.7%) [35]. As reported in the previous study, BRD4 protein was a member of the bromodomain and extra-terminal domain (BET) family that carried two bromodomains and was associated with mitotic chromosomes. Bromodomains targeted genetic and epigenetic alterations and regulated chromatin remodeling, which were important therapeutic targets for major diseases, such as neurological disorders, obesity, cancer and inflammation [39,40]. Thus, these compounds further deserved development and research for the treatment of major diseases. Li et al. (2012) reported that 41 had potent inhibitory activities against Fusarium oxysporum f. sp. Niveum, Alternaria alternate, Fusarium oxysporum f. sp. vasinfectum and Fusarium solani with MIC values of 6.25–25 μg/mL and moderate brine shrimp toxicity (LC50 60.7 μg/mL) [41]. The occurrence of 41 could be involved in protecting microbes against invasion by other competing microbes. Therefore, 41 could be considered as a promising lead compound for developing new fungicides. Cui et al. reported 43 could completely inhibit the G2/M phase of tsFT210 cells at concentrations >29.4 µM [42]. Furthermore, Wang et al. (in 2008) showed that 44 had selective cytotoxicity against four cancer cell lines, MOLT-4, HL-60, A-549 and BEL-7402 [43].
Cytochalasans were fungal metabolites that were structurally identified by the presence of a reduced isoindone nucleus connected with a macrocyclic ring [44]. Six cytochalasans (5055) showed strong toxicity against Streptomyces sp. with 50–80% inhibition at 2–16 μg/mL, and most of them even exhibited 60% inhibition at 2 μg/mL, but they had no any effect on the fungus A. flavipes at the same concentration. This indicated that cytochalasans could help A. flavipes to compete with Streptomyces sp., which was an important support for their potential ecological role. All cytochalasans also exhibited obvious toxicity against human cell lines, as cytochalasans had the ability to inhibit, specifically, the actin filament elongation by blocking the polymerization sites [45,46,47]. Thus, all six compounds (5055) exhibited powerful toxicity against Streptomyces sp. at 2–16 μg/mL with inhibition rate of 50–80%. Notably, most of these compounds displayed strong inhibitory activity with inhibition rate of 60% even at 2 μg/mL, whereas none of them had antimicrobial activity against the marine-derived producer A. flavipes at the same concentration. These findings implied that the co-culture through microbial physical contact could stimulate the expression of silent gene cluster that was responsible for the production of cytochalasans.
The cyclic siderophore, nocardamine (58), had inhibitory effects on the proliferation of human tumor cell lines: SK-Mel-5 with an IC50 value of 18 μM, T-47D with an IC50 value of 6 μM, PRMI-7951 with an IC50 value of 14 μM and SK-Mel-28 with an IC50 value of 12 μM [48]. Compared with the pure cultures, some novel metabolites were observed in the mixed culture. Two fungal prenylated indole metabolites, 56 and 59, which were not traced before in A. fumigatus, were induced. Both of them had an oxazino [6,5-b]indole nucleus which was not previously found in nature. Additionally, the yield of compound 61 was obviously higher than that of the monoculture of Streptomyces leeuwenhoekii C58. It was the first time that a bi-lateral cross talk was proved, which resulted in dual induction of both fungal and bacterial metabolites in the same culture conditions. 64 displayed cytotoxic activities toward mouse lymphoma cell line L5178Y with an IC50 value of 16.7 μM and in vitro toward testosterone-dependent prostate LNCaP cells with an IC50 value of 2.1 μM [49].

2.1.3. Alkaloids Derived from the Co-Cultures of Different Marine Bacteria

Thirteen alkaloids were isolated from the co-culture of different marine bacteria (Figure 2E and Figure 3); the structures of these isolates were listed in Figure 10. The average yields of five known tryptamine derivatives, N-acetyltryptamine (68), N-propanoyltryptamine (69), bacillamide C (70), bacillamide B (71) and bacillamide A (72) using the co-fermentation of marine strain Streptomyces sp. CGMCC4.7185 and Bacillus mycoides isolated from marine sediments of the Nanji Island (China, 27°42′ N, 121°08′ E), were 14.9, 2.8, 9.6, 13.7 and 3.0 mg/L, respectively, which were all undetectable under simple culture conditions [50]. This was the first report of applying a microorganism co-culture system to enhance the yields of known compounds [50].
In 2018, El-Hawary et al. identified four indole alkaloids—a novel brominated oxindole alkaloid saccharomonosporine A (73), a novel convolutamydine F (74) and two known compounds, (S) 6-bromo-3-hydroxy-3-(1H-indol-3-yl) indolin-2-one (75) and vibrindole (76)—from the mixed fermentation culture of two sponge-associated actinomycetes, Saccharomonospora sp. UR22 and Dietzia sp. UR66 collected from the Red Sea sponge Callyspongia siphonella [51].
Two sponge-associated actinomycetes, Actinokineospora sp. EG49 isolated from the Red Sea sponge, Spheciospongia vagabunda, and Nocardiopsis sp. RV163 derived from the Mediterranean sponge, Dysidea avara, were co-cultivated together and yielded a novel 5a,6,11a,12-tetrahydro-5a,11a-dimethyl-1,4-benzoxazino[3,2-b][1,4]benzoxazine (77) and three known metabolites, N-(2-hydroxyphenyl)-acetamide (78), 1,6-dihydroxyphenazine (79) and 2,2′,3,3′-tetrahydro-2,2′-dimethyl-2,2′-bibenzoxazole (80) [52].
Pim-1 kinase is a well-established oncoprotein in several tumor entities, such as prostate cancer, pancreatic cancer, colorectal cancer and myeloid leukemia. Inhibition of Pim-1 kinase would prevent the growth of tumor cells. Compounds 73 and 75 exhibited potent Pim-1 kinase inhibitors with IC50 values of 0.3 μΜ and 0.946 μΜ, respectively. Docking studies showed the binding model of 73 and 75 in the ATP pocket of Pim-1 kinase. They also exhibited obvious antiproliferative activity against human promyelocytic leukemia HL-60 (IC50 2.8 and 4.9 µΜ) and human colon adenocarcinoma HT-29 (IC50 3.6 and 3.7 μΜ). This indicated that 73 and 75 could act as potential Pim-1 kinase inhibitors that mediated the inhibitory effects on the growth of tumor cells [51].
In addition, only compound 79 was documented against Trypanosoma brucei (IC50 19 μΜ), Bacillus sp. (11 mm inhibition zone diameter) and Actinokineospora sp. EG49 (15 mm inhibition zone diameter) [52]. The yield of 79 was very high in the co-culture process. However, it was not detected in the single microbial culture. Co-culture strategy not only enhanced the chemical diversity of the metabolites but also increased the production of metabolites undetected in the single microbial culture.

2.2. Anthraquinones

Thirteen different anthraquinone isolates were obtained from different marine microbial co-cultures; the co-cultures of marine fungi–bacteria represented the majority, 69% (9/13 isolates; Figure 2B and Figure 11).

2.2.1. Anthraquinones Derived from the Co-Cultures of Different Marine Fungi

In the recent study, the combination of cultures from two different developmental stages of marine alga-derived Aspergillus alliaceus (teleomorph: Petromyces alliaceus) drastically changed the metabolite profile and resulted in the production of allianthrone A (81) and two diastereomers, allianthrones B (82) and C (83) (Figure 12) [53]. 8183 exhibited cytotoxic activity against SK-Mel-5 melanoma cell lines with IC50 (11.0, 12.2, and 19.7 μM) and HCT-116 colon carcinoma cells with IC50 (9.0, 10.5 and 13.7 μM), respectively. This study presented the first example of elicitation of novel fungal chemical diversity by a co-existing strategy of two different developmental phenotypes of Aspergillus species. For several Aspergilli, e.g., A. alliaceus, asexual and sexual life developmental stages were known. However, rarely did they co-cultivate at the same time. Even more surprising was the presence of novel bianthrones when the sclerotial and asexual morphs of the same species co-existed. There were only a few examples that showed differences in secondary metabolites in fungi based on their distinct developmental stages or chemical profiles for the two mating types of heterothallic fungi. However, none of these compounds displayed any activity against P. aeruginosa, E. faecium, S. aureus, E. coli, C. albicans and B. subtilis. Furthermore, non-significant results were obtained against lung (A549), prostate (PC3) and breast (MCF-7) human cancer cells compared with the positive control, etoposide [53].

2.2.2. Anthraquinones Derived from the Co-Cultures of Marine Fungi and Bacteria

Two novel anthraquinones, (z)(11S,12R)-versicolorin B (84) and 6,8-O-dimethylbipolarin (85), along with seven known substances bipolarin (86), versiconol (87), versiconol acetate (88), versicolorin B (89), 8-O-Methylversicolorin B (90), averufin (91) and endocrocin (92) (Figure 13) were isolated and identified from the mixed fermentation broth of the marine fungus A. versicolor and B. subtilis [38].
Versiconol (87) was characterized as an inhibitor of protein tyrosine kinases against EGF-R and v-abl protein tyrosine kinases that were responsible for catalyzing phosphorylation of tyrosine residues of protein substrates, and suppression of MK-cells [54]. 89 displayed inhibitory activity against the Gram-positive S. aureus with MIC value of 50 μM and antifungal activity against Fusarium solani with MIC values of 16–32 µg/mL [38,55]. The cytotoxic bioassay of 90 was recorded against mouse lymphoma cell line L5178Y with an IC50 value of 21.2 μM. Moreover, 91 displayed antibacterial activity against B. subtilis (MIC = 8–16 μg/mL) and the Gram-positive S. aureus (MIC = 25 μM) and four Gram-positive microbes, including two E. faecalis and two E. faecium (MIC = 12.5–25 μM) [38,55]. Neither 89 nor 91 had cytotoxicity against L5178Y cell line, which implied that their antimicrobial activities were not associated with their respective general toxicities. Besides, 90 also displayed mild cytotoxic activity against human lung cancer cells H460 and the human prostate cancer cells PC-3 with IC50 values of 27.2 and 19.5 μM, respectively [56]. Other compounds did not exhibit distinct cytotoxic activity against L5178Y cell line and antibacterial activity against five Gram-positive microbes, including one S. aureus, two E. faecalis and two E. faecium.

2.2.3. Anthraquinones Derived from the Co-Cultures of Different Marine Bacteria

A new antibiotic, keyicin (93) (Figure 14), was purified and identified from a co-culture of two marine invertebrate-associated bacteria Micromonospora sp. WMMB-235 and Rhodococcus sp. WMMA-185 [57]. It showed selective inhibitory activity against Gram-positive bacteria and could inhibit the growth of B. subtilis and Methicillin Sensitive Staphylococcus aureus (MSSA) with MIC values of 9.9 μM and 2.5 μM, respectively. In contrast to many anthracyclines, 93 might modulate fatty acid metabolism and exhibit antibacterial activity without nucleic acid damage that is explained by keyicin’s mechanism of action (MOA) based on E. coli chemical genomics studies [57].

2.3. Cyclopeptides

Cyclopeptides are cyclic compounds mainly formed by the amide bonds of proteinogenic or non-proteinogenic amino acids bound together. Several fungal cyclic peptides have been developed as pharmaceuticals, such as the echinocandins, pneumocandins and cyclosporin A [58]. Six cyclopeptides were produced by the co-cultures of marine fungi–fungi (four isolates, 67%) and fungi–bacteria (two isolates, 33%) from different marine sources. However, marine bacteria–bacteria did not yield these structures in this period of investigation.

2.3.1. Cyclopeptides Derived from the Co-Cultures of Different Marine Fungi

Three new cyclic tetrapeptides, named cyclo-(L-leucyl-trans-4-hydroxy-L-prolyl-D-leucyl-trans-4-hydroxy-L-proline) (94) [59] cyclo (D-Pro-L-Tyr-L-Pro-L-Tyr) (95) and cyclo (Gly-L-Phe-L-Pro-L-Tyr) (96) (Figure 15) [60] were identified from the co-culture of two mangrove fungi Phomopsis sp. K38 and Alternaria sp. E33 isolated from the South China Sea. Meanwhile, the co-cultivation of two marine alga-derived fungi Aspergillus sp. BM-05 and BM-05ML isolated from a brown algal species collected off Helgoland, North Sea, Germany, yielded a new cyclotripeptide, psychrophilin E (97) (Figure 15) [30].
Compound 94 exhibited in vitro moderate to high inhibitory activity towards four crop-threatening fungi, Helminthosporium sativum, Gaeumannomyces graminis, F. graminearum and Rhizoctonia cereals with MIC values of 130, 220, 250 and 160 μg/mL, respectively [59]. 95 and 96 showed high in vitro antifungal activity against human fungus (Candida albicans) with MIC values of 35 μg/mL and 25 μg/mL, respectively [60]. 97 exhibited anti-proliferative activities against four human cancer cells, human cisplatin-resistant ovarian cancer A2780CisR, colon carcinoma HCT116, ovarian cancer A2780 and chronic myelogenous leukemia K562 with IC50 values of 49.4, 28.5, 27.3 and 67.8 µM, respectively. The inhibition of HCT116 cells by 97 was more potent than that of the positive control, cisplatin (IC50 33.4 µM) [30].

2.3.2. Cyclopeptides Derived from the Co-Cultures of Marine Fungi and Bacteria

Recently, the chemical investigation of the mixed-fermentation of a marine fungus Aspergillus versicolor isolated from the sponge Agelas oroides and B. subtilis yielded two cyclic pentapeptides, one new cotteslosin C (98) and a known cotteslosin A (99) (Figure 16) [38]. Both of them did not show significant cytotoxic activity towards mouse lymphoma cell line L5178Y, or even antibacterial activity against five Gram-positive microbes, including one S. aureus, two E. faecalis and two E. faecium [38]. 99 displayed weak cytotoxicity against another three human cancer cell lines, prostate DU145, melanoma MM418c5 and breast T47D, with EC50 values of 90, 66 and 94 µg/mL, respectively [61].

2.4. Macrolide

There were no reported macrolides from the co-cultures of marine fungi–fungi and fungi–bacteria. Only one isolate was identified from a co-culture of marine bacteria–bacteria.

Macrolides Derived from the Co-Cultures of Different Marine Bacteria

A known compound, nonactin (100) (Figure 17) was isolated from the co-culture of two marine bacteria, Saccharomonospora sp. UR22 and Dietzia sp. UR66 [51]. It possessed a macrotetrolide structure integrated from nonactic acid, and exhibited antitumor and antibacterial activity, especially its inhibitory effects on the P170 glycoprotein-mediated efflux of chemotherapeutic agents in multiple-drug-resistant cancer cells [62,63,64,65].

2.5. Phenylpropanoids

Phenylpropanoids are a big and structurally diverse group of secondary metabolites, which bear a C6–C3 phenolic scaffold that play crucial roles in a wide spectrum of biological and pharmacological activities [66]. Twenty-three phenylpropanoids were isolated from co-culture of marine fungi–fungi (12 isolates, 52%) and fungi–bacteria (11 isolates, 48%), while there are no reported phenylpropanoids from the co-culture of different marine bacteria.

2.5.1. Phenylpropanoids Derived from the Co-Cultures of Different Marine Fungi

A xanthone derivative known as 8-hydroxy-3-methyl-9-oxo-9H-xanthene-1-carboxylic acid methyl ether (101) (Figure 18) was discovered from the mixed culture of two mangrove fungi, Phomopsis sp. K38 and Alternaria sp. E33 [67] from the South China Sea coast. It showed a broad spectrum of antifungal activities against plant pathogens, Blumeria graminearum, Gloeasporium musae, F. oxysporum, Colletotrichum glocosporioides and Peronophthora cichoralearum.
Ten citrinin analogues were isolated and identified from the co-culture of two marine algal-derived endophytic fungal strains, Aspergillus sydowii EN-534 and Penicillium citrinum EN-535 collected from marine red alga Laurencia okamurai, including two novel compounds, citrinin dimer seco-penicitrinol A (102) and citrinin monomer penicitrinol L(103), and the known penicitrinone A (104), penicitrinone F (105), penicitrinol A (106), citrinin (107), dihydrocitrinone (108), decarboxydihydrocitrinone (109) phenol A acid (110) and phenol A (111) (Figure 19) [68]. In addition, one novel coumarin named 7-(γ,γ-dimethylallyloxy)-6-hydroxy-4-methylcoumarin (112) (Figure 19) was detected and characterized from the co-culture of the two mangrove fungi, Phomopsis sp. K38 and Alternaria sp. E33 [69].
Compounds 104, 106 and 107 exhibited inhibitory activities against two human pathogens Micrococcus luteus and E. coli, and three aquatic bacteria Vibrio parahaemolyticus, Vibrio alginolyticus and Edwardsiella ictaluri with MIC values of 4–64 μg/mL. 102, 103 and 105 inhibited V. alginolyticus and E. ictaluri with MIC values of 32–64 μg/mL. 103 and 105 inhibited V. parahaemolyticus and E. coli with MIC values of 32 and 64 μg/mL, respectively. Moreover, 102107 were further evaluated for anti-influenza neuraminidase (homologous protein of H5N1) activity. 104 and 105 exhibited significant inhibitory activities with IC50 values of 12.9 and 18.5 nM, respectively [68]. Thus, these bioactive substances could be further optimized for the development of antibacterial and anti-influenza agents. In addition to the anti-influenza activity, the activated metabolite penicitrinone A (104) also exerted an inhibitory effect on four human cancer cell lines, HL-60, K562, BGC-823 and HeLa cells with IC50 values of 43.2, 50.8, 54.2 and 65.6 μM, respectively [70].

2.5.2. Phenylpropanoids Derived from the Co-Cultures of Marine Fungi and Bacteria

The chemical investigation of the mixed culture of the marine fungus A. versicolor and B. subtilis resulted in the isolation of one novel aflaquinolone, 22-epi-aflaquinolone B (113); and ten known metabolites, aflaquinolone A, F and G (114116), 3-O-methylviridicatin (117), 9-hydroxy-3-methoxyviridicatin (118), O-demethylsterigmatocystin (119), sterigmatocystin (120), sterigmatin (121), AGI-B4 (122) and sydowinin B (123) (Figure 20) [38].
The metabolite 3-O-methylviridicatin (117) was reported to possess inhibitory activity against human immunodeficiency virus (HIV) (Heguy et al., 1998). It could prevent cytokine tumor necrosis factor α (TNF-α), induce the HIV expression with long terminal repeat in HeLa cells (IC50, 5μM) and block the viral replication in the model of chronic infection in OM-10.1 cell lines which directed at the induction of TNF-α [71]. 119 exhibited cytotoxic activities towards mouse lymphoma cell line L5178Y with an IC50 value of 5.8 μM. Three xanthone derivatives (120122) showed potent cytotoxic activities towards the mouse lymphoma cell lines with IC50 values of 2.3, 2.2 and 2.0 μM, respectively, compared with a positive control, kahalalide F (IC50 = 4.3 μM). Sterigmatocystin (120) also exhibited strong cytotoxicity towards human hepatoma cells (HepG2) at 3 μM [72]. Its mechanism suggested that it could stimulate a biotransformation process, increase the population of reactive oxygen species and promote the imbalance in the antioxidant defense system caused by the process of lipid peroxidation [73]. Recently, Zingales et al. (2020) displayed the significant role of mitochondria in sterigmatocystin-induced toxicity in SH-SY5Y cells [74]. The reduced viability of SH-SY5Y cells displayed time- and dose-dependence with mitochondrial dysfunction when exposed to 120 in response to the forced dependency of the cells on mitochondrial oxidative phosphorylation [74]. Thus, these findings provided us a valuable direction for the application of neuroprotective mitochondria-target functional peptides. Moreover, compound 122 inhibited human umbilical vein endothelial cells (VEGF-induced proliferation of HUVECs) with an IC50 value of 1.4 μM [75]. It is considered as a novel inhibitor of vascular endothelial cell growth factor, which is one of the main stimulants of angiogenesis.

2.6. Polyketides

Twelve polyketides were isolated and characterized from the marine microbial co-cultures in recent years (Figure 2B and Figure 21).

2.6.1. Polyketides Derived from the Co-Cultures of Different Marine Fungi

In 2014, Ebada et al. identified three previously reported polyketide derivatives, sterigmatocystin (124), 5-methoxysterigmatocystin (125) and aversin (126) (Figure 22) from the ethyl acetate extract of two marine alga-derived fungi, Aspergillus sp. BM-05 and BM-05ML [30]. Kossuga et al. isolated two new and unusual polyketides: (Z)-2-ethylhex-2-enedioic acid (127) and (E)-4-oxo-2-propylideneoct-7-enoic acid (128) (Figure 22) from the marine-derived fungi Penicillium sp. Ma(M3)V isolated from the marine sponge Mycale angulosa co-cultivated with Trichoderma sp. Gc(M2)1 isolated from the marine sponge Geodia corticostylifera [76]. Two unprecedented polyketides (127128) had a common feature—a conjugated carboxylic acid group that could be biogenetically generated from the methyl group of an acetate rather than a methionine precursor in 127, and the same group could be derived from C-1 position of an acetate or C-2 position of a propionate in 128 based on the precursor of the ethyl group connected to a double bond. It was an excellent case of a truly novel carbon skeleton induced by the powerful and underexplored method, marine microbial co-cultivation.
Compound 124 showed in vitro anti-proliferative activities towards human cisplatin-resistant ovarian cancer A2780CisR, ovarian cancer A2780 and chronic myelogenous leukemia K562 cell lines with IC50 values of 95.5, 30.6 and 57.0 µM, respectively. Moreover, it also displayed more significant anti-proliferative activities against human colon carcinoma HCT116 cells with an IC50 value of 10.3 µM (cf. to cisplatin’s IC50 33.4 µM). Compound 125 exhibited potent in vitro anti-proliferative activities towards three human cancer cell lines, HCT116, A2780 and human chronic myelogenous leukemia (K562) with IC50 values of 4.4, 51.0 and 13.4 µM, respectively [30].

2.6.2. Polyketides Derived from the Co-Cultures of Marine Fungi and Bacteria

A pair of enantiomers (9R,14S)-epoxy-11-deoxyfunicone (129) and (9S,14R)-epoxy-11-deoxyfunicone (130), along with deoxyfunicone (131), alternariol (132) and vermistatin (133) (Figure 23) were isolated from the co-culture of Penicillium sp. WC-29-5 isolated from the mangrove soil around the roots of Aegiceras corniculatum and Streptomyces fradiae 007 isolated from a sediment sample in the Jiaozhou Bay, Shandong Province, China [77].
Both 129 and 130 exhibited moderate inhibitory activity against H1975 tumor cell lines with IC50 values of 3.97 and 5.73 μM, respectively. Deoxyfunicone (131) was found to exert anti-inflammatory activity, exhibiting the inhibition effect on overproduction of nitric oxide (NO) and the prostaglandin E2 in both lipopolysaccharide-provoked BV2 microglial and lipopolysaccharide-stimulated RAW264.7 macrophage cells (IC50 = 10.6 and 40.1 μM, respectively) [78]. 132 was known as a cytotoxic, genotoxic, mutagenic and fetotoxic mycotoxin [79,80]. However, in the IL-1β-stimulated Caco-2 cells, the metabolite 132 increased the transcription of TNF-α; inversely reduced the transcription of IL-1β and IL-6; and decreased the transcription and secretion of IL-8, suggesting that 132 possessed immunomodulatory activities on both lipopolysaccharide- and IL-1 β-related pathways in non-immune intestinal epithelial cells [79].

2.6.3. Polyketides Derived from the Co-Cultures of Different Marine Bacteria

Recently, two unusual polyketides, janthinopolyenemycins A (134) and B (135) (Figure 24) were purified and identified from the co-cultivation broth of two marine bacteria Janthino bacterium spp. ZZ145 and ZZ148 isolated from marine soil sample [81]. Both 134 and 135 displayed the same antifungal activity against C. albicans with a minimum bactericidal concentration (MBC) value of 31.25 μg/mL and an MIC value of 15.6 μg/mL. However, none of them could suppress the growth of methicillin-resistant S. aureus or E. coli (MIC > 100 μg/mL) [81].

2.7. Steroids

Steroids contain a characteristic arrangement of four cycloalkane rings that are joined together. They represent a large family of compounds that play important roles as chemical messengers, and the scaffold is present in many FDA-approved drugs [82,83,84]. A total of five steroidal metabolites were reported; four of them were isolated from the co-culture of marine fungi–bacteria (80%); only one isolate was identified from the co-culture of marine fungi–fungi (20%). No isolates were obtained from the co-culture of marine bacteria–bacteria.

2.7.1. Steroids Derived from the Co-Cultures of Different Marine Fungi

To the best of our knowledge, the only one steroid, ergosterol (136), was found from the co-culture broth of two marine mangrove epiphytic fungi, Aspergillus sp. FSY-01 and FSW-02 (Figure 25) [21,85]. It was an essential component of fungal cell membrane with strong specificity and stable structure. Therefore, 136 was widely applied to detecting fungal containment as an indicator of fungal biomass [86].

2.7.2. Steroids Derived from the Co-Cultures of Marine Fungi and Bacteria

An unprecedented steroid, 7β-hydroxycholesterol-1β-carboxylic acid (137), together with three known steroidal metabolites, 7β-hydroxycholesterol (138), 7α-hydroxycholesterol (139) and ergosterol-5α,8α-peroxide (140) (Figure 26), have been confirmed from the co-culture of two marine alga-derived microbes, Aspergillus sp. BM05, and an unidentified bacterium (BM05BL), isolated from the brown alga of the genus Sargassum collected off Helgoland, North Sea, Germany [87].
Compounds 137140 showed moderate activities against four human tumor cell lines, A2780, HCT116, K562 and A2780 CisR with the IC50 values of 10.0–100.0 µM. At the same time, the total extract of co-culture of Aspergillus sp. BM05 and BM05BL showed obvious antiproliferative activity compared with its single steroidal compounds. This implied a synergistic role of these steroidal metabolites in the extract. Furthermore, 140 was reported as a promising new candidate that could overcome the drug-resistant property of malignant cancer cells through abolishing miR-378, a microRNA involved in new tumor initiation, unlimited self-renewal and recurrence of tumor cells after chemotherapy [88].

2.8. Terpenoids

Terpenoids known as isoprenoids are structurally diverse metabolites found in many natural sources. This class of compounds displays a wide sector of important pharmacological entities that confirmed by several preclinical and clinical studies [89,90]. Only two terpenoidals were isolated from the co-cultures of marine fungi–bacteria (one compound, 50%) and bacteria–bacteria (one compound, 50%).

2.8.1. Terpenoids Derived from the Co-Cultures of Marine Fungi and Bacteria

The production of the bacterial sesquiterpene pentalenic acid (141) (Figure 27) might be attributed to the competition relationship between marine fungus A. fumigatus MR2012 isolated from a Red Sea sediment in Hurghada, Egypt and terrestrial bacterium S. leeuwenhoekii C58 collected from the hyper-arid soil of Laguna de Chaxa Salar de Atacama, Chile, in which S. leeuwenhoekii C58 suppressed the production of A. fumigatus MR2012 and enhanced the production of 141 [37]. This suggested that S. leeuwenhoekii C58 appeared to activate the cryptic biosynthetic gene clusters to construct a defense mechanism based on the chemical signals generated by the competitive fungus, A. fumigatus MR2012. Thus, the bacterial strain was capable of suppressing the biosynthesis of the fungus metabolites that were present in the axenic cultures.

2.8.2. Terpenoids Derived from the Co-Cultures of Different Marine Bacteria

A diterpene lobocompactol (142) (Figure 28) was isolated from the co-culture of marine actinomycete Streptomyces cinnabarinus PK209 collected from the seaweed rhizosphere, obtained at a depth of 10 m along the coast of Korea and its competitor Alteromonas sp. KNS-16. Its productivity was increased 10.4-fold higher than that of the pure culture of PK209 [91]. Moreover, its antifouling activities were recently confirmed against primary fouling organisms, including diatoms, bacteria, and macroalgae zoospores. In order to further determine whether 142 was a non-toxic antifoulant, the therapeutic rate (LC50/EC50) was used to evaluate its toxicity, the LC50/EC50 of 142 was more than that of 15, indicating that the metabolite 142 was a non-toxic antifoulant. Thus, this compound could be valuable as an antifouling agent in both antifouling coating industry and marine ecology.

2.9. Others

Twelve compounds with other structures were obtained by co-culture of marine fungi–bacteria (4 compounds, 33%) and fungi–fungi (8 compounds, 67%).

2.9.1. Other Compounds Derived from the Co-Cultures of Different Marine Fungi

A novel polysubstituted benzaldehyde derivative, ethyl-5-ethoxy-2-formyl-3-hydroxy-4- methylbenzoate (143) (Figure 29) was identified from the mixed fermentation of the two mangrove fungi, Phomopsis sp. K38 and Alternaria sp. E33 that were collected from the South China Sea [92]. Another two novel furanone derivatives were identified as sclerotiorumins A and B (144, 145) (Figure 29) from the co-culture of the two marine fungi, sclerotiorum SCSGAF 0053 and P. citrinum SCSGAF 0052 isolated from gorgonian Muricella flexuosa collected from the South China Sea, Sanya (18°11′ N, 109°25′ E), Hainan Province, China [23]. Five diorcinols, including one novel diorcinol J (146) and four known diorcinols B-E (147150) (Figure 29), were characterized during the co-culturing of two marine-derived fungi, A. sulphureus KMM 4640 and I. felina KMM 4639 [93].
Compound 143 showed in vitro inhibitory activity against G. musae, F. graminearum, P. sojae (Kaufmann and Gerdemann) and Rhizoctonia solani Kuhn at 0.25 mM with inhibition zone diameters of 11.57, 12.06, 8.5 and 10.21 mm, respectively. This suggested that 143 had broad inhibitory activity against these microbes [92]. 144 and 145 exhibited weak toxicity against brine shrimp (LC50 > 100 μM) and none of them displayed cytotoxicity against the liver hepatocellular carcinoma Huh7 and HepG2 (LC50 > 100 μM) and obvious inhibitory activities towards three marine-derived bacteria, Bacillus stearothermophilus, Pseudoalteromonas nigrifaciens and Bacillus amyloliquefaciens, and two common pathogens, P. aeruginosa and S. aureus [23].
Among the five diorcinols, only 146 showed apparent cytotoxicity against murine Ehrlich carcinoma cells and hemolytic activity against mouse erythrocytes. The significant hemolytic activity of 146 suggested that its cytotoxic activity against murine Ehrlich carcinoma cells was due to a membranolytic mechanism. It is well known that the heat shock protein 70 (HSP70) was frequently overexpressed in tumor cell lines as an ATP-dependent molecular chaperone and played a significant role in refolding misfolded proteins and promoting cell survival under stress [94]. Thus, compounds that could inhibit HSP70 had great potential in tumor therapy. 147 could decrease the expression of HSP70 in the Ehrlich carcinoma cells, which made it possible to develop as a new antitumor drug/lead. Diorcinol D (149) was studied for its combined therapy against planktonic Candida albicans with a broad-spectrum antifungal agent fluconazole [95]. The combined therapy exhibited considerable antifungal activity against ten clinical isolates of C. albicans containing five fluconazole-resistant isolates and five fluconazole-sensitive isolates, whereas fluconazole alone did not display antifungal activity. This suggested that diorcinol D (149) restored the susceptibility of fluconazole to C. albicans.
Moreover, the efficiencies of fluconazole inhibiting mature biofilms were also drastically boosted by the addition of 149 [95]. The fractional inhibitory concentration index (FICI) model and ΔE model unclosed that the synergistic actions indeed existed in combination of diorcinol D (149) and fluconazole [95]. Two resistance mechanisms of azoles were overexpression of efflux pumps genes and alterations of genes (point mutations). 149 mainly suppressed the activity of efflux pump in cells partly by decreasing the expression of Cdr1 (one mediator of azole efflux pumps) in Candida albicans CASA1. On the other hand, 149 also inhibited ergosterol synthesis and CYP51 (the target of fluconazole) expression [95]. Thus, the significant synergistic interaction and drug-resistant reversion of fluconazole combined with diorcinol D (149) were caused by the two latent mechanisms, the block of efflux pump and ergosterol biosynthesis. Notably, 149 was still needed to further in vivo study in the combination therapy field to settle rock-ribbed clinical fungal infection in response to the azole resistance.

2.9.2. Other Compounds Derived from the Co-Cultures of Marine Fungi and Bacteria

Five known metabolites, diorcinol D (149), penicillanone (151), diorcinol G (152), diorcinol I (153) and radiclonic acid (154) (Figure 30) were obtained from the co-culture of the sponge-derived fungi A. versicolor and B. subtilis [38].
Compounds 149, 152 and 153 displayed antibacterial activities against five Gram-positive microbes, including one S. aureus, two E. faecalis and two E. faecium with the MIC values of 12.5–50 μM. In addition, 152 displayed potent inhibitory activities against all tested bacteria with an MIC value of 12.5 μM. 149 displayed inhibitory activity against E. coli with an MIC value of 8 µg/mL; and 153 showed significant antibacterial activity against S. aureus with an MIC value of 6.25 μg/mL [96,97]. In contrast, 149, 152 and 153 did not display any obvious activity against L5178Y cell lines, which suggested that the antimicrobial activities of these products were not associated with their respective general toxicities [38].

3. Conclusions

Marine microorganisms have attracted more attention as natural producers of lead compounds. Marine microbes especially are considered as a renewable and reproducible source that can be easily cultured [98,99]. However, the speed of new lead compound discovery is slowing down. Thus, marine microbial co-culturing represents a powerful strategy for the production of novel bio-substances. The strategy can induce the biosynthesis of novel compounds and various NPs coded by corresponding genomes through the activation of the silent gene clusters or previously unexpressed biosynthetic routes.
In the last ten years, the overall statistical studies showed that 156 metabolites were discovered from the co-culture of different marine microbes. Figure 2 and Table 1 illustrated that 59 compounds were isolated from the co-culturing of different marine fungi; 79 compounds were isolated from marine fungi and bacteria; and only 18 compounds were disclosed from co-culturing different marine bacteria. The metabolites by co-culture of marine fungi and bacteria accounted for the largest proportion (51% of all metabolites of marine microbial co-culture). Alkaloids were the largest group with ≥51.9%, whereas macrolides were the lowest group with <0.65%. Just only one macrolide was identified from the co-cultures of different marine bacteria. Furthermore, co-cultures of different marine bacteria did not produce cyclopeptides, phenylpropanoids and steroids, and co-cultures of different marine fungi did not induce the biosynthesis of terpenoids.
Several studies suggest that Aspergillus spp. are the most common fungi that co-fermented with other microbes and produce numerous novel skeletons. The majority of these NPs have antimicrobial or/and antitumor activities. However, some significant restrictions obstruct the development of the co-culture technology; e.g., cryptic and undefined biosynthesis routes and the producers of NPs from the co-cultivation of two or more microorganisms, the particularities of strains and environmental and nutritional requirements, the instability of the ecological relationship, the uncertainty of the interaction relationship and the high contamination probability. Therefore, new technology and equipment need to be created, such as metabolomics analysis and molecular network technology. The new mechanisms of chemical communication of microbes (through direct/mediate contact) also need to be further investigated. In conclusion, co-culture is still shrouded in mystery as a prospective experimental tool for novel bioactive NPs. This article embodies the value and diversity of NPs from the co-cultivation of marine-derived microorganisms and it is considered as a guided reference for studying NPs.

Author Contributions

J.C. and P.Z. conceived and wrote the review; X.Y. and B.W. offered important advice to improve the review; M.E., H.Z. and H.W. revised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research work was supported by National Natural Science Foundation of China (number 81773628 and 41776139), and the 111-National Overseas Expertise Introduction Center for Green Pharmaceutical Discipline Innovation (number D17012).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [Green Version]
  2. Bertrand, S.; Bohni, N.; Schnee, S.; Schumpp, O.; Gindro, K.; Wolfender, J.L. Metabolite induction via microorganism co-culture: A potential way to enhance chemical diversity for drug discovery. Biotechnol. Adv. 2014, 32, 1180–1204. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, J.W.; Wu, Q.H.; Rowley, D.C.; Al-Kareef, A.M.; Wang, H. Anticancer agent-based marine natural products and related compounds. J. Aisan. Nat. Prod. Res. 2015, 17, 199–216. [Google Scholar] [CrossRef]
  4. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2019, 36, 122–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Chen, J.W.; Wang, B.X.; Lu, Y.J.; Guo, Y.Q.; Sun, J.D.; Wei, B.; Zhang, H.W.; Wang, H. Quorum sensing inhibitors from marine microorganisms and their synthetic derivatives. Mar. Drugs 2019, 17, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Brakhage, A.A.; Schroeckh, V. Fungal secondary metabolites–Strategies to activate silent gene clusters. Fungal Genet. Biol. 2011, 48, 15–22. [Google Scholar] [CrossRef] [PubMed]
  7. Mao, D.; Okada, B.K.; Wu, Y.; Xu, F.; Seyedsayamdost, M.R. Recent advances in activating silent biosynthetic gene clusters in bacteria. Curr. Opin. Microbiol. 2018, 45, 156–163. [Google Scholar] [CrossRef]
  8. Romano, S.; Jackson, S.A.; Patry, S.; Dobson, A.D.W. Extending the “one strain many compounds” (OSMAC) principle to marine microorganisms. Mar. Drugs 2018, 16, 224. [Google Scholar] [CrossRef] [Green Version]
  9. Dalisay, D.S.; Rogers, E.W.; Edison, A.S.; Molinski, T.F. Structure elucidation at the nanomole scale. 1. Trisoxazole macrolides and thiazole-containing cyclic peptides from the nudibranch Hexabranchus sanguineus. J. Nat. Prod. 2009, 72, 732–738. [Google Scholar] [CrossRef] [Green Version]
  10. Wolfender, J.L.; Queiroz, E.F. Chemical diversity of natural resources and the bioactivity of their constituents. CHIMIA Int. J. Chem. 2012, 66, 324–329. [Google Scholar] [CrossRef]
  11. Bohni, N.; Cordero-Maldonado, M.L.; Maes, J.; Siverio-Mota, D.; Marcourt, L.; Munck, S.; Kamuhabwa, A.R.; Moshi, M.J.; Esguerra, C.V.; de Witte, P.A.M.; et al. Integration of microfractionation, qNMR and zebrafish screening for the in vivo bioassay-guided isolation and quantitative bioactivity analysis of natural products. PLoS ONE 2013, 8, e64006. [Google Scholar] [CrossRef] [Green Version]
  12. Scherlach, K.; Hertweck, C. Triggering cryptic natural product biosynthesis in microorganisms. Org. Biomol. Chem. 2009, 7, 1753–1760. [Google Scholar] [CrossRef] [PubMed]
  13. Abdalla, M.; Sulieman, S.; McGaw, L. Microbial communication: A significant approach for new leads. S. Afr. J. Bot. 2017, 113, 461–470. [Google Scholar] [CrossRef]
  14. Ola, A.R.B.; Thomy, D.; Lai, D.; Brötz-Oesterhelt, H.; Proksch, P. Inducing secondary metabolite production by the endophytic fungus Fusarium tricinctum through coculture with Bacillus subtilis. J. Nat. Prod. 2013, 76, 2094–2099. [Google Scholar] [CrossRef] [PubMed]
  15. Kochanowska-Karamyan, A.J.; Hamann, M.T. Marine indole alkaloids: Potential new drug leads for the control of depression and anxiety. Chem. Rev. 2010, 110, 4489–4497. [Google Scholar] [CrossRef] [Green Version]
  16. Singh, K.S.; Majik, M.S. Bioactive alkaloids from marine sponges. In Marine Sponges: Chemicobiological and Biomedical Applications; Ehrlich, R.P.H., Ed.; Springer: New Delhi, India, 2016; pp. 257–286. [Google Scholar]
  17. Afiyatullov, S.S.; Zhuravleva, O.I.; Antonov, A.S.; Berdyshev, D.V.; Pivkin, M.V.; Denisenko, V.A.; Popov, R.S.; Gerasimenko, A.V.; von Amsberg, G.; Dyshlovoy, S.A.; et al. Prenylated indole alkaloids from co-culture of marine-derived fungi Aspergillus sulphureus and Isaria felina. J. Antibiot. 2018, 71, 846–853. [Google Scholar] [CrossRef] [PubMed]
  18. Kato, H.; Yoshida, T.; Tokue, T.; Nojiri, Y.; Hirota, H.; Ohta, T.; Williams, R.M.; Tsukamoto, S. Notoamides A-D: Prenylated indole alkaloids isolated from a marine-derived fungus, Aspergillus sp. Angew. Chem. Int. Ed. Engl. 2007, 46, 2254–2256. [Google Scholar] [CrossRef] [PubMed]
  19. Tsukamoto, S.; Kato, H.; Samizo, M.; Nojiri, Y.; Onuki, H.; Hirota, H.; Ohta, T. Notoamides F-K, prenylated indole alkaloids isolated from a marine-derived Aspergillus sp. J. Nat. Prod. 2008, 71, 2064–2067. [Google Scholar] [CrossRef]
  20. Assante, G.; Camarda, L.; Locci, R.; Merlini, L.; Nasini, G.; Papadopoulos, E. Isolation and structure of red pigments from Aspergillus flavus and related species, grown on a differential medium. J. Agric. Food Chem. 1981, 29, 785–787. [Google Scholar] [CrossRef]
  21. Zhu, F.; Chen, G.Y.; Chen, X.; Huang, M.Z.; Wan, X. Aspergicin, a new antibacterial alkaloid produced by mixed fermentation of two marine-derived mangrove epiphytic fungi. Chem. Nat. Compd. 2011, 47, 767–769. [Google Scholar] [CrossRef]
  22. Zhu, F.; Li, J.S.; Xie, W.C.; Shi, J.J.; Xu, F.; Song, Z.F.; Liu, Y.L. Structure revision of aspergicin by the crystal structure of aspergicine, a co-occurring isomer produced by co-culture of two mangrove epiphytic fungi. Nat. Prod. Res. 2017, 31, 2268–2272. [Google Scholar] [CrossRef] [PubMed]
  23. Bao, J.; Wang, J.; Zhang, X.Y.; Nong, X.H.; Qi, S.H. New furanone derivatives and alkaloids from the co-culture of marine-derived fungi Aspergillus sclerotiorum and Penicillium citrinum. Chem. Biodivers. 2017, 14, e1600327. [Google Scholar] [CrossRef] [PubMed]
  24. Weiss, U.; Strelitz, F.; Flon, H.; Asheshovl, I.N. Antibiotic compounds with action against bacterial viruses: Neohydroxyaspergillic acid. Arch. Biochem. Biophys. 1958, 74, 150–157. [Google Scholar] [CrossRef]
  25. Wan, X.Q.; Zhu, F.; Chen, G.Y.; Li, H.M.; Tan, S.Y.; Pan, Y.Q.; Hong, Y. Biological evaluation of neoaspergillic acid, a pyrazine hydroxamic acid produced by mixed cultures of two marine-derived mangrove epiphytic fungi. J. Biomed. Inform. 2010, 5, 1932–1935. [Google Scholar]
  26. Xu, X.; He, F.; Zhang, X.; Bao, J.; Qi, S. New mycotoxins from marine-derived fungus Aspergillus sp. SCSGAF0093. Food Chem. Toxicol. 2013, 53, 46–51. [Google Scholar] [CrossRef] [PubMed]
  27. MacDonald, J.C.; Micetich, R.G.; Haskins, R.H. Antibiotic activity of neoaspergillic acid. Can. J. Microbiol. 1964, 10, 90–92. [Google Scholar] [CrossRef]
  28. Li, C.Y.; Ding, W.J.; Shao, C.L.; She, Z.G.; Lin, Y.C. A new diimide derivative from the co-culture broth of two mangrove fungi (strain no. E33 and K38). J. Asian Nat. Prod. Res. 2010, 12, 809–813. [Google Scholar] [CrossRef]
  29. Ding, W.J.; Lu, Y.C.; Feng, Z.H.; Luo, S.H.; Li, C.Y. A new nonadride derivative from the co-culture broth of two mangrove fungi. Chem. Nat. Compd. 2017, 53, 691–693. [Google Scholar] [CrossRef]
  30. Ebada, S.S.; Fischer, T.; Hamacher, A.; Du, F.Y.; Roth, Y.O.; Kassack, M.U.; Wang, B.G.; Roth, E.H. Psychrophilin E, a new cyclotripeptide, from co-fermentation of two marine alga-derived fungi of the genus Aspergillus. Nat. Prod. Res. 2014, 28, 776–781. [Google Scholar] [CrossRef]
  31. Wang, J.H.; Ding, W.J.; Wang, R.M.; Du, Y.P.; Liu, H.L.; Kong, X.H.; Li, C.Y. Identification and bioactivity of compounds from the mangrove endophytic fungus Alternaria sp. Mar. Drugs 2015, 13, 4492–4504. [Google Scholar] [CrossRef] [Green Version]
  32. Lee, S.U.; Asami, Y.; Lee, D.; Jang, J.-H.; Ahn, J.S.; Oh, H. Protuboxepins A and B and protubonines A and B from the marine-derived fungus Aspergillus sp. SF-5044. J. Nat. Prod. 2011, 74, 1284–1287. [Google Scholar] [CrossRef] [PubMed]
  33. Asami, Y.; Jang, J.H.; Soung, N.K.; He, L.; Moon, D.O.; Kim, J.W.; Oh, H.; Muroi, M.; Osada, H.; Kim, B.Y.; et al. Protuboxepin A, a marine fungal metabolite, inducing metaphase arrest and chromosomal misalignment in tumor cells. Bioorg. Med. Chem. 2012, 20, 3799–3806. [Google Scholar] [CrossRef] [PubMed]
  34. Lu, X.H.; Shi, Q.W.; Zheng, Z.H.; Ke, A.B.; Zhang, H.; Huo, C.H.; Ma, Y.; Ren, X.; Li, Y.Y.; Lin, J.; et al. Oxepinamides: Novel liver X receptor agonists from Aspergillus puniceus. Eur. J. Org. Chem. 2011, 2011, 802–807. [Google Scholar] [CrossRef]
  35. Yu, L.Y.; Ding, W.J.; Wang, Q.Q.; Ma, Z.J.; Xu, X.W.; Zhao, X.F.; Chen, Z. Induction of cryptic bioactive 2,5-diketopiperazines in fungus Penicillium sp. DT-F29 by microbial co-culture. Tetrahedron 2017, 73, 907–914. [Google Scholar] [CrossRef]
  36. Yu, L.Y.; Ding, W.J.; Ma, Z.J. Induced production of cytochalasans in co-culture of marine fungus Aspergillus flavipes and actinomycete Streptomyces sp. Nat. Prod. Res. 2016, 30, 1718–1723. [Google Scholar] [CrossRef] [PubMed]
  37. Wakefield, J.; Hassan, H.M.; Jaspars, M.; Ebel, R.; Rateb, M.E. Dual induction of new microbial secondary metabolites by fungal bacterial co-cultivation. Front. Microbiol. 2017, 8, 1284. [Google Scholar] [CrossRef] [Green Version]
  38. Abdel-Wahab, N.M.; Scharf, S.; Özkaya, F.C.; Kurtán, T.; Mándi, A.; Fouad, M.A.; Kamel, M.S.; Müller, W.E.G.; Kalscheuer, R.; Lin, W.H.; et al. Induction of secondary metabolites from the marine-derived fungus Aspergillus versicolor through co-cultivation with Bacillus subtilis. Planta Med. 2019, 85, 503–512. [Google Scholar] [CrossRef] [Green Version]
  39. Dey, A.; Chitsaz, F.; Abbasi, A.; Misteli, T.; Ozato, K. The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis. Proc. Natl. Acad. Sci. USA 2003, 100, 8758–8763. [Google Scholar] [CrossRef] [Green Version]
  40. Yang, Z.Y.; He, N.H.; Zhou, Q. Brd4 recruits P-TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol. Cell Biol. 2008, 28, 967–976. [Google Scholar] [CrossRef] [Green Version]
  41. Li, X.J.; Zhang, Q.; Zhang, A.L.; Gao, J.M. Metabolites from Aspergillus fumigatus, an endophytic fungus associated with Melia azedarach, and their antifungal, antifeedant, and toxic activities. J. Agric. Food Chem. 2012, 60, 3424–3431. [Google Scholar] [CrossRef]
  42. Cui, C.B.; Kakeya, H.; Osada, H. Novel mammalian cell cycle inhibitors, cyclotryprostatins A-D, produced by Aspergillus fumigatus, which inhibit mammalian cell cycle at G2/M phase. Terrahedron 1997, 53, 59–72. [Google Scholar] [CrossRef]
  43. Wang, F.Z.; Fang, Y.C.; Zhu, T.J.; Zhang, M.; Lin, A.Q.; Gu, Q.Q.; Zhu, W.M. Seven new prenylated indole diketopiperazine alkaloids from holothurian-derived fungus Aspergillus fumigatus. Tetrahedron 2008, 64, 7986–7991. [Google Scholar] [CrossRef]
  44. Thomas, E.J. Cytochalasan synthesis: Macrocycle formation via intramolecular Diels-Alder reactions. Acc. Chem. Res. 1991, 24, 229–235. [Google Scholar] [CrossRef]
  45. Lin, Z.J.; Zhu, T.J.; Wei, H.J.; Zhang, G.J.; Wang, H.; Gu, Q.Q. Spicochalasin A and new aspochalasins from the marine-derived fungus Spicaria elegans. Eur. J. Org. Chem. 2009, 2009, 3045–3051. [Google Scholar] [CrossRef]
  46. Scherlach, K.; Boettger, D.; Remme, N.; Hertweck, C. The chemistry and biology of cytochalasans. Nat. Prod. Rep. 2010, 27, 869–886. [Google Scholar] [CrossRef] [PubMed]
  47. Xiao, L.; Liu, H.Z.; Wu, N.; Liu, M.; Wei, J.T.; Zhang, Y.Y.; Lin, X.K. Characterization of the high cytochalasin E and rosellichalasin producing-Aspergillus sp. nov. F1 isolated from marine solar saltern in China. World J. Microbiol. Biotechnol. 2013, 29, 11–17. [Google Scholar] [CrossRef]
  48. Kalinovskaya, N.I.; Romanenko, L.A.; Irisawa, T.; Ermakova, S.P.; Kalinovsky, A.I. Marine isolate Citricoccus sp. KMM 3890 as a source of a cyclic siderophore nocardamine with antitumor activity. Microbiol. Res. 2011, 166, 654–661. [Google Scholar] [CrossRef]
  49. Qian-Cutrone, J.F.; Huang, S.; Shu, Y.Z.; Vyas, D.; Fairchild, C.; Menendez, A.; Krampitz, K.; Dalterio, R.; Klohr, S.E.; Gao, Q. Stephacidin A and B: Two structurally novel, selective inhibitors of the testosterone-dependent prostate LNCaP cells. J. Am. Chem. Soc. 2002, 124, 14556–14557. [Google Scholar] [CrossRef]
  50. Yu, L.Y.; Hu, Z.F.; Ma, Z.J. Production of bioactive tryptamine derivatives by co-culture of marine Streptomyces with Bacillus mycoides. Nat. Prod. Res. 2015, 29, 2087–2091. [Google Scholar] [CrossRef]
  51. El-Hawary, S.S.; Sayed, A.M.; Mohammed, R.; Khanfar, M.A.; Rateb, M.E.; Mohammed, T.A.; Hajjar, D.; Hassan, H.M.; Gulder, T.A.M.; Abdelmohsen, U.R. New Pim-1 kinase inhibitor from the co-culture of two sponge-associated actinomycetes. Front. Chem. 2018, 6, 538. [Google Scholar] [CrossRef] [Green Version]
  52. Dashti, Y.; Grkovic, T.; Abdelmohsen, U.R.; Hentschel, U.; Quinn, R.J. Production of induced secondary metabolites by a co-culture of sponge-associated actinomycetes, Actinokineospora sp. EG49 and Nocardiopsis sp. RV163. Mar. Drugs 2014, 12, 3046–3059. [Google Scholar] [CrossRef] [PubMed]
  53. Mandelare, P.E.; Adpressa, D.A.; Kaweesa, E.N.; Zakharov, L.N.; Loesgen, S. Coculture of two developmental stages of a marine-derived Aspergillus alliaceus results in the production of the cytotoxic bianthrone allianthrone A. J. Nat. Prod. 2018, 81, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
  54. Petersen, F.; Fredenhagen, A.; Mett, H.; Lydon, N.B.; Delmendo, R.; Jenny, H.B.; Peter, H.H. Paeciloquinones A, B, C, D, E and F: New potent inhibitors of protein tyrosine kinases produced by Paecilomyces carneus. I. Taxonomy, fermentation, isolation and biological activity. J. Antibiot. 1995, 48, 191–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Liu, K.; Zheng, Y.K.; Miao, C.P.; Xiong, Z.J.; Xu, L.H.; Guan, H.L.; Yang, Y.B.; Zhao, L.X. The antifungal metabolites obtained from the rhizospheric Aspergillus sp. YIM PH30001 against pathogenic fungi of Panax notoginseng. Nat. Prod. Res. 2014, 28, 2334–2337. [Google Scholar] [CrossRef] [PubMed]
  56. Dou, Y.L.; Wang, X.L.; Jiang, D.F.; Wang, H.Y.; Jiao, Y.; Lou, H.X.; Wang, X.N. Metabolites from Aspergillus versicolor, an endolichenic fungus from the lichen Lobaria retigera. Drug. Discov. Ther. 2014, 8, 84–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Adnani, N.; Chevrette, M.G.; Adibhatla, S.N.; Zhang, F.; Yu, Q.; Braun, D.R.; Nelson, J.; Simpkins, S.W.; McDonald, B.R.; Myers, C.L.; et al. Coculture of marine invertebrate-associated bacteria and interdisciplinary technologies enable biosynthesis and discovery of a new antibiotic, keyicin. ACS. Chem. Biol. 2017, 12, 3093–3102. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, X.H.; Lin, M.Y.; Xu, D.; Lai, D.W.; Zhou, L.G. Structural diversity and biological activities of fungal cyclic peptides, excluding cyclodipeptides. Molecules 2017, 22, 2069. [Google Scholar] [CrossRef] [Green Version]
  59. Li, C.Y.; Wang, J.H.; Luo, C.P.; Ding, W.J.; Cox, D.G. A new cyclopeptide with antifungal activity from the co-culture broth of two marine mangrove fungi. Nat. Prod. Res. 2014, 28, 616–621. [Google Scholar] [CrossRef]
  60. Huang, S.; Ding, W.J.; Li, C.Y.; Cox, D.G. Two new cyclopeptides from the co-culture broth of two marine mangrove fungi and their antifungal activity. Pharmacogn. Mag. 2014, 10, 410–414. [Google Scholar]
  61. Fremlin, L.J.; Piggott, A.M.; Lacey, E.; Capon, R.J. Cottoquinazoline A and cotteslosins A and B, metabolites from an Australian marine-derived strain of Aspergillus versicolor. J. Nat. Prod. 2009, 72, 666–670. [Google Scholar] [CrossRef] [PubMed]
  62. Meyers, E.; Pansy, F.E.; Perlman, D.; Smith, D.A.; Weisenborn, F.L. The in vitro activity of nonactin and its homologs: Monactin, dinactin and trinactin. J. Antibiot. 1965, 18, 128–129. [Google Scholar] [PubMed]
  63. Borrel, M.N.; Pereira, E.; Fiallo, M.; Garnier-Suillerot, A. Mobile ionophores are a novel class of P-glycoprotein inhibitors: The effects of ionophores on 4′-O-tetrahydropyranyl-adriamycin incorporation in K562 drug-resistant cells. Eur. J. Biochem. 1994, 223, 125–133. [Google Scholar] [CrossRef] [PubMed]
  64. Woo, A.J.; Strohl, W.R.; Priestley, N.D. Nonactin biosynthesis: The product of nonS catalyzes the formation of the furan ring of nonactic acid. Antimicrob. Agents Chemother. 1999, 43, 1662–1668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kusche, B.R.; Phillips, J.B.; Priestley, N.D. Nonactin biosynthesis: Setting limits on what can be achieved with precursor-directed biosynthesis. Bioorg. Med. Chem. Lett. 2009, 19, 1233–1235. [Google Scholar] [CrossRef] [Green Version]
  66. Yang, C.H.; Ma, L.; Wei, Z.P.; Han, F.; Gao, J. Advances in isolation and synthesis of xanthone derivatives. Chin. Herb. Med. 2012, 4, 87–102. [Google Scholar]
  67. Li, C.Y.; Zhang, J.; Shao, C.L.; Ding, W.J.; She, Z.G.; Lin, Y.C. A new xanthone derivative from the co-culture broth of two marine fungi (strain No. E33 and K38). Chem. Nat. Compd. 2011, 47, 382–384. [Google Scholar] [CrossRef]
  68. Yang, S.Q.; Li, X.M.; Li, X.; Li, H.L.; Meng, L.H.; Wang, B.G. New citrinin analogues produced by coculture of the marine algal-derived endophytic fungal strains Aspergillus sydowii EN-534 and Penicillium citrinum EN-535. Phytochem. Lett. 2018, 25, 191–195. [Google Scholar] [CrossRef]
  69. Wang, J.H.; Huang, S.; Li, C.Y.; Ding, W.J.; She, Z.G.; Li, C.L. A new coumarin produced by mixed fermentation of two marine fungi. Chem. Nat. Compd. 2015, 51, 239–241. [Google Scholar] [CrossRef]
  70. Wu, C.J.; Yi, L.; Cui, C.B.; Li, C.W.; Wang, N.; Han, X. Activation of the silent secondary metabolite production by introducing neomycin-resistance in a marine-derived Penicillium purpurogenum G59. Mar. Drugs 2015, 13, 2465–2487. [Google Scholar] [CrossRef] [Green Version]
  71. Heguy, A.; Cai, P.; Meyn, P.; Houck, D.; Russo, S.; Michitsch, R.; Pearce, C.; Katz, B.; Bringmann, G.; Feineis, D.; et al. Isolation and characterization of the fungal metabolite 3-O-methylviridicatin as an inhibitor of tumour necrosis factor α-induced human immunodeficiency virus replication. Antivir. Chem. Chemother. 1998, 9, 149–155. [Google Scholar] [CrossRef] [Green Version]
  72. Gao, W.; Jiang, L.P.; Ge, L.; Chen, M.; Geng, C.Y.; Yang, G.; Li, Q.J.; Ji, F.; Yan, Q.; Zou, Y.; et al. Sterigmatocystin-induced oxidative DNA damage in human liver-derived cell line through lysosomal damage. Toxicol. In Vitro 2015, 29, 1–7. [Google Scholar] [CrossRef] [PubMed]
  73. Sivakumar, V.; Thanislass, J.; Niranjali, S.; Devaraj, H. Lipid peroxidation as a possible secondary mechanism of sterigmatocystin toxicity. Hum. Exp. Toxicol. 2001, 20, 398–403. [Google Scholar] [CrossRef] [PubMed]
  74. Zingales, V.; Fernandez-Franzon, M.; Ruiz, M.J. The role of mitochondria in sterigmatocystin-induced apoptosis on SH-SY5Y cells. Food Chem. Toxicol. 2020, 142, 111493. [Google Scholar] [CrossRef] [PubMed]
  75. Kim, H.S.; Park, I.Y.; Park, Y.J.; Lee, J.H.; Hong, Y.S.; Lee, J.J. A novel dihydroxanthenone, AGI-B4 with inhibition of VEGF-induced endothelial cell growth. J. Antibiot. 2002, 55, 669–672. [Google Scholar] [CrossRef] [Green Version]
  76. Kossuga, M.H.; Ferreira, A.G.; Sette, L.D.; Berlinck, R.G.S. Two polyketides from a co-culture of two marine-derived fungal strains. Nat. Prod. Commun. 2013, 8, 721–724. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, Y.; Wang, L.P.; Zhuang, Y.B.; Kong, F.D.; Zhang, C.X.; Zhu, W.M. Phenolic polyketides from the co-cultivation of marine-derived Penicillium sp. WC-29-5 and Streptomyces fradiae 007. Mar. Drugs 2014, 12, 2079–2088. [Google Scholar] [CrossRef]
  78. Ha, T.M.; Kim, D.C.; Sohn, J.H.; Yim, J.H.; Oh, H. Anti-inflammatory and protein tyrosine phosphatase 1B inhibitory metabolites from the antarctic marine-derived fungal strain Penicillium glabrum SF-7123. Mar. Drugs. 2020, 18, 247. [Google Scholar] [CrossRef]
  79. Schmutz, C.; Cenk, E.; Marko, D. The alternaria mycotoxin alternariol triggers the immune response of IL-1β-stimulated, differentiated Caco-2 cells. Mol. Nutr. Food Res. 2019, 63, 1900341. [Google Scholar] [CrossRef] [Green Version]
  80. Fliszár-Nyúl, E.; Lemli, B.; Kunsági-Máté, S.; Dellafiora, L.; Dall’Asta, C.; Cruciani, G.; Pethő, G.; Poór, M. Interaction of mycotoxin alternariol with serum albumin. Int. J. Mol. Sci. 2019, 20, 2352. [Google Scholar] [CrossRef] [Green Version]
  81. Anjum, K.; Sadiq, I.; Chen, L.; Kaleem, S.; Li, X.C.; Zhang, Z.Z.; Lian, X.Y. Novel antifungal janthinopolyenemycins A and B from a co-culture of marine-associated Janthinobacterium spp. ZZ145 and ZZ148. Tetrahedron. Lett. 2018, 59, 3490–3494. [Google Scholar] [CrossRef]
  82. Konoshima, T.; Takasaki, M. Anti-tumor-promoting activities (cancer chemopreventive activities) of natural products. Stud. Nat. Prod. Chem. 2000, 24, 215–267. [Google Scholar]
  83. Ouellette, R.J.; Rawn, J.D. Organic Chemistry: Structure, Mechanism, and Synthesis; Elsevier: Boston, MA, USA, 2014. [Google Scholar]
  84. Roos, G.; Roos, C. Organic Chemistry Concepts: An EFL Approach; Academic Press: London, UK, 2014. [Google Scholar]
  85. Dias, D.A.; Urban, S. HPLC and NMR studies of phenoxazone alkaloids from Pycnoporus cinnabarinus. Nat. Prod. Commun. 2009, 4, 489–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ng, H.E.; Raj, S.S.; Wong, S.H.; Tey, D.; Tan, H.M. Estimation of fungal growth using the ergosterol assay: A rapid tool in assessing the microbiological status of grains and feeds. Lett. Appl. Microbiol. 2008, 46, 113–118. [Google Scholar] [CrossRef] [PubMed]
  87. Ebada, S.S.; Fischer, T.; Klassen, S.; Hamacher, A.; Roth, Y.O.; Kassack, M.U.; Roth, E.H. A new cytotoxic steroid from co-fermentation of two marine alga-derived micro-organisms. Nat. Prod. Res. 2014, 28, 1241–1245. [Google Scholar] [CrossRef]
  88. Wu, Q.P.; Xie, Y.Z.; Deng, Z.; Li, X.M.; Yang, W.; Jiao, C.W.; Fang, L.; Li, S.Z.; Pan, H.H.; Yee, A.J.; et al. Ergosterol peroxide isolated from Ganoderma lucidum abolishes microRNA miR-378-mediated tumor cells on chemoresistance. PLoS ONE 2012, 7, e44579. [Google Scholar] [CrossRef]
  89. Ludwiczuk, A.; Skalicka-Woźniak, K.; Georgiev, M.I. Chapter 11–Terpenoids. In Pharmacognosy Fundamentals, Applications and Strategies; Badal, S., Delgoda, R., Eds.; Academic Press: London, UK, 2017; pp. 233–266. [Google Scholar]
  90. Teufel, R. Unusual “head-to-torso” coupling of terpene precursors as a new strategy for the structural diversification of natural products. Methods Enzymol. 2018, 604, 425–439. [Google Scholar]
  91. Cho, J.Y.; Kim, M.S. Induction of antifouling diterpene production by Streptomyces cinnabarinus PK209 in co-culture with marine-derived Alteromonas sp. KNS-16. Biosci. Biotechnol. Biochem. 2012, 76, 1849–1854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Wang, J.H.; Ding, W.J.; Li, C.Y.; Huang, S.P.; She, Z.G.; Lin, Y.C. New polysubstituted benzaldehyde from the co-culture broth of two marine fungi (strains Nos. E33 and K38). Chem. Nat. Compd. 2013, 49, 799–802. [Google Scholar] [CrossRef]
  93. Zhuravleva, O.I.; Kirichuk, N.N.; Denisenko, V.A.; Dmitrenok, P.S.; Yurchenko, E.A.; Min′ko, E.M.; Ivanets, E.V.; Afiyatullov, S.S. New diorcinol J produced by co-cultivation of marine fungi Aspergillus sulphureus and Isaria felina. Chem. Nat. Compd. 2016, 52, 227–230. [Google Scholar] [CrossRef]
  94. Balaburski, G.M.; Leu, J.I.; Beeharry, N.; Hayik, S.; Andrake, M.D.; Zhang, G.; Herlyn, M.; Villanueva, J.; Dunbrack, R.L., Jr.; Yen, T.; et al. A modified HSP70 inhibitor shows broad activity as an anticancer agent. Mol. Cancer Res. 2013, 11, 219–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Li, Y.; Chang, W.; Zhang, M.; Li, X.; Jiao, Y.; Lou, H. Synergistic and drug-resistant reversing effects of diorcinol D combined with fluconazole against Candida albicans. FEMS. Yeast Res. 2015, 15, fov001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Li, Z.X.; Wang, X.F.; Ren, G.W.; Yuan, X.L.; Deng, N.; Ji, G.X.; Li, W.; Zhang, P. Prenylated diphenyl ethers from the marine algal-derived endophytic fungus Aspergillus tennesseensis. Molecules 2018, 23, 2368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Xu, X.L.; Yang, H.J.; Xu, H.T.; Yin, L.Y.; Chen, Z.K.; Shen, H.H. Diphenyl ethers from a marine-derived isolate of Aspergillus sp. CUGB-F046. Nat. Prod. Res. 2018, 32, 821–825. [Google Scholar] [CrossRef]
  98. Netzker, T.; Fischer, J.; Weber, J.; Mattern, D.J.; Konig, C.C.; Valiante, V.; Schroeckh, V.; Brakhage, A.A. Microbial communication leading to the activation of silent fungal secondary metabolite gene clusters. Front. Microbiol. 2015, 6, 299. [Google Scholar] [CrossRef]
  99. Chen, J.W.; Guo, Y.Q.; Lu, Y.J.; Wang, B.X.; Sun, J.D.; Zhang, H.W.; Wang, H. Chemistry and biology of siderophores from marine microbes. Mar. Drugs 2019, 17, 562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The schematic diagram of novel and bioactive natural products (NPs) using co-cultures of marine fungi−fungi, fungi−bacteria and bacteria−bacteria in direct or indirect contact.
Figure 1. The schematic diagram of novel and bioactive natural products (NPs) using co-cultures of marine fungi−fungi, fungi−bacteria and bacteria−bacteria in direct or indirect contact.
Marinedrugs 18 00449 g001
Figure 2. Numbers and the percentage of (A) isolates from the co-cultures of different marine microbes; (B) different classes of NPs from the co-cultures of marine microbes. The classes, numbers and proportions of NPs isolated from the co-cultures of marine (C) fungi and fungi, (D) fungi and bacteria, (E) bacteria and bacteria.
Figure 2. Numbers and the percentage of (A) isolates from the co-cultures of different marine microbes; (B) different classes of NPs from the co-cultures of marine microbes. The classes, numbers and proportions of NPs isolated from the co-cultures of marine (C) fungi and fungi, (D) fungi and bacteria, (E) bacteria and bacteria.
Marinedrugs 18 00449 g002
Figure 3. Alkaloids isolated from the co-cultures of marine fungi–fungi, fungi–bacteria and bacteria–bacteria.
Figure 3. Alkaloids isolated from the co-cultures of marine fungi–fungi, fungi–bacteria and bacteria–bacteria.
Marinedrugs 18 00449 g003
Figure 4. Chemical structures of 114.
Figure 4. Chemical structures of 114.
Marinedrugs 18 00449 g004
Figure 5. Chemical structures of 1517.
Figure 5. Chemical structures of 1517.
Marinedrugs 18 00449 g005
Figure 6. Chemical structures of 1822.
Figure 6. Chemical structures of 1822.
Marinedrugs 18 00449 g006
Figure 7. Chemical structures of 2326.
Figure 7. Chemical structures of 2326.
Marinedrugs 18 00449 g007
Figure 8. Chemical structures of 2749.
Figure 8. Chemical structures of 2749.
Marinedrugs 18 00449 g008
Figure 9. Chemical structures of 5067.
Figure 9. Chemical structures of 5067.
Marinedrugs 18 00449 g009
Figure 10. Chemical structures of 6880.
Figure 10. Chemical structures of 6880.
Marinedrugs 18 00449 g010
Figure 11. Anthraquinones isolated from the co-cultures of marine fungi–fungi, fungi–bacteria and bacteria–bacteria.
Figure 11. Anthraquinones isolated from the co-cultures of marine fungi–fungi, fungi–bacteria and bacteria–bacteria.
Marinedrugs 18 00449 g011
Figure 12. Chemical structures of 8183.
Figure 12. Chemical structures of 8183.
Marinedrugs 18 00449 g012
Figure 13. Chemical structures of 8492.
Figure 13. Chemical structures of 8492.
Marinedrugs 18 00449 g013
Figure 14. Chemical structures of 93.
Figure 14. Chemical structures of 93.
Marinedrugs 18 00449 g014
Figure 15. Chemical structures of 9497.
Figure 15. Chemical structures of 9497.
Marinedrugs 18 00449 g015
Figure 16. Chemical structures of 9899.
Figure 16. Chemical structures of 9899.
Marinedrugs 18 00449 g016
Figure 17. Chemical structures of 100.
Figure 17. Chemical structures of 100.
Marinedrugs 18 00449 g017
Figure 18. Chemical structures of 101.
Figure 18. Chemical structures of 101.
Marinedrugs 18 00449 g018
Figure 19. Chemical structures of 102112.
Figure 19. Chemical structures of 102112.
Marinedrugs 18 00449 g019
Figure 20. Chemical structures of 113123.
Figure 20. Chemical structures of 113123.
Marinedrugs 18 00449 g020
Figure 21. Polyketides isolated from the co-culture of marine fungi–fungi, fungi–bacteria and bacteria–bacteria.
Figure 21. Polyketides isolated from the co-culture of marine fungi–fungi, fungi–bacteria and bacteria–bacteria.
Marinedrugs 18 00449 g021
Figure 22. Chemical structures of 124128.
Figure 22. Chemical structures of 124128.
Marinedrugs 18 00449 g022
Figure 23. Chemical structures of 129133.
Figure 23. Chemical structures of 129133.
Marinedrugs 18 00449 g023
Figure 24. Chemical structures of 134135.
Figure 24. Chemical structures of 134135.
Marinedrugs 18 00449 g024
Figure 25. Chemical structures of 136.
Figure 25. Chemical structures of 136.
Marinedrugs 18 00449 g025
Figure 26. Chemical structures of 137140.
Figure 26. Chemical structures of 137140.
Marinedrugs 18 00449 g026
Figure 27. Chemical structures of 141.
Figure 27. Chemical structures of 141.
Marinedrugs 18 00449 g027
Figure 28. Chemical structures of 142.
Figure 28. Chemical structures of 142.
Marinedrugs 18 00449 g028
Figure 29. Chemical structures of 143150.
Figure 29. Chemical structures of 143150.
Marinedrugs 18 00449 g029
Figure 30. Chemical structures of 151154.
Figure 30. Chemical structures of 151154.
Marinedrugs 18 00449 g030
Table 1. Summarized NPs identified from the co-culture of marine microbes: 2009–2019.
Table 1. Summarized NPs identified from the co-culture of marine microbes: 2009–2019.
ClassesThe Number of NPsIdentified DateBioactivitiesCo-Culture of Marine Microorganisms
Alkaloids80 isolates
(180)
2010 and 2014–2019Cytotoxicity, enzyme Inhibitors, antimicrobial activitiesFungi and fungi
A. sulphureus KMM 4640 and I. felina KMM 4639
Aspergillus. sp. FSY-01 and FSW-02
P. citrinum SCSGAF 0052 and A. sclerotiorum SCSGAF 0053
Phomopsis sp. K38 and Alternaria sp. E33
Fungi and bacteria
Penicillium sp. DT-F29 and Bacillus sp. B31
A. flavipes fungus and S. sp. CGMCC4.7185
A. fumigatus MR2012 and S. leeuwenhoekii C34
A. versicolor and B. subtilis,
Bacteria and bacteria
Streptomyces sp. CGMCC4.7185 and B. mycoides
Saccharomonospora sp. UR22 and Dietzia sp. UR66
Anthraquinones13 isolates
(8193)
2017–2019 Cytotoxicity and antimicrobial activitiesFungi and fungi
Asexual morph and sclerotial morph of A. alliaceus
Fungi and bacteria
A. versicolor and B. subtilis
Bacteria and bacteria
Micromonospora sp. WMMB-235 and Rhodococcus sp. WMMA-185
Cyclopeptides6 isolates
(9499)
2014 and 2019Antifungal and anti-proliferative activitiesFungi and fungi
Phomopsis sp. K38 and Alternaria sp. E33
Aspergillus sp. BM and 05-BM-05ML
Fungi and bacteria
A. versicolor and B. subtilis
Macrolides1 isolate
(100)
2018Antitumor and antibacterial activityBacteria and bacteria
Saccharomonospora sp. UR22 and Dietzia sp. UR66
Phenylpropanoids23 isolates
(101123)
2011, 2015 and 2019Cytotoxic, antifungal, antibacterial and anti-influenza activitiesFungi and fungi
Phomopsis sp. K38 and Alternaria sp. E33
A. sydowii EN-534 and P. citrinum EN-535
Fungi and bacteria
A. versicolor and B. subtilis
Polyketides12 isolates
(124135)
2013, 2014 and 2018Anti-proliferative, cytotoxicity and antifungal activitiesFungi and fungi
Aspergillus sp. BM and 05 and BM-05ML
Penicillium sp. Ma(M3)V and Trichoderma sp. Gc(M2)1
Fungi and bacteria
Penicillium sp. WC-29-5 and S. fradiae 007
Bacteria and bacteria
Janthinobacterium spp. ZZ145 and ZZ148
Steroids5 isolates
(136140)
2009, 2010 and 2014Antiproliferative activityFungi and fungi
Aspergillus sp. FSY-01 and FSW-02
Fungi and bacteria
Aspergillus sp. BM05 and an unknown bacteria (BM05BL)
Terpenoids2 isolates
(141142)
2012 and 2017Inhibition of diatom N. annexa and macroalga U. pertusa Fungi and bacteria
A. fumigatus MR2012 and S. leeuwenhoekii C58
Bacteria and bacteria
S. cinnabarinus PK209 and Alteromonas sp. KNS-16
Others 12 isolates
(143154)
2013, 2016, 2017 and 2019Antimicrobial, toxicity, cytotoxicity, Hemolytic activitiesFungi and fungi
Phomopsis sp. K38 and Alternaria sp. E33
P. citrinum SCSGAF 0052 and A. sclerotiorum SCSGAF 0053
A. sulphureus KMM 4640 and I. felina KMM 4639
Fungi and bacteria
A. versicolor and B. subtilis

Share and Cite

MDPI and ACS Style

Chen, J.; Zhang, P.; Ye, X.; Wei, B.; Emam, M.; Zhang, H.; Wang, H. The Structural Diversity of Marine Microbial Secondary Metabolites Based on Co-Culture Strategy: 2009–2019. Mar. Drugs 2020, 18, 449. https://doi.org/10.3390/md18090449

AMA Style

Chen J, Zhang P, Ye X, Wei B, Emam M, Zhang H, Wang H. The Structural Diversity of Marine Microbial Secondary Metabolites Based on Co-Culture Strategy: 2009–2019. Marine Drugs. 2020; 18(9):449. https://doi.org/10.3390/md18090449

Chicago/Turabian Style

Chen, Jianwei, Panqiao Zhang, Xinyi Ye, Bin Wei, Mahmoud Emam, Huawei Zhang, and Hong Wang. 2020. "The Structural Diversity of Marine Microbial Secondary Metabolites Based on Co-Culture Strategy: 2009–2019" Marine Drugs 18, no. 9: 449. https://doi.org/10.3390/md18090449

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

Article Metrics

Back to TopTop