Next Article in Journal
Lipase-Catalyzed Production of Sorbitol Laurate in a “2-in-1” Deep Eutectic System: Factors Affecting the Synthesis and Scalability
Next Article in Special Issue
Nujiangexanthone A Inhibits Cervical Cancer Cell Proliferation by Promoting Mitophagy
Previous Article in Journal
Key Electronic, Linear and Nonlinear Optical Properties of Designed Disubstituted Quinoline with Carbazole Compounds
Previous Article in Special Issue
Olive-Derived Triterpenes Suppress SARS COV-2 Main Protease: A Promising Scaffold for Future Therapeutics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review: Halogenated Compounds from Marine Actinomycetes

1
Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi University for Nationalities, Nanning 530006, China
2
Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI 96720, USA
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(9), 2754; https://doi.org/10.3390/molecules26092754
Submission received: 3 April 2021 / Revised: 2 May 2021 / Accepted: 3 May 2021 / Published: 7 May 2021

Abstract

:
Marine actinomycetes, Streptomyces species, produce a variety of halogenated compounds with diverse structures and a range of biological activities owing to their unique metabolic pathways. These halogenated compounds could be classified as polyketides, alkaloids (nitrogen-containing compounds) and terpenoids. Halogenated compounds from marine actinomycetes possess important biological properties such as antibacterial and anticancer activities. This review reports the sources, chemical structures and biological activities of 127 new halogenated compounds originated mainly from Streptomyces reported from 1992 to 2020.

1. Introduction

Marine actinomycetes are a rich source of biologically active compounds, which have been widely studied worldwide. They can efficiently produce different secondary metabolites including simple benzene derivatives, polyketides and complex cyclic peptides. These secondary metabolites exhibit a wide range of biological activities including antibacterial, antifungal, anticancer and enzyme inhibition. Most of marine actinomyces were Streptomyces species, but rarer actinomycetes genera have been reported in the past twenty years. Consequently, more novel natural products including new halogenated compounds have been isolated in recent years. According to a review on marine microbial natural products from 2010 to 2013 [1], secondary metabolites from marine actinomycetes possess various structures, including terpenes, peptides, polyketides, alkaloids and halogenated molecules [2]. Due to the high concentration of chloride and bromine ions in seawater, marine actinomycetes usually produce more halogenated compounds than those of their terrestrial counterparts. The majority of the marine halogenated compounds showed certain kind of biological properties including antibacterial and anticancer activities [3]. This review focuses on the sources of marine actinomycetes, structures and biological activities of 127 new halogenated compounds derived from marine-derived actinomycetes from 1992 to 2020.

2. Halogenated Compounds from Streptomyces Species

2.1. Sponges-Associated Streptomyces sp.

Two indole-containing peptides JBIR-34 (1) (Figure 1) and JBIR-35 (2) were obtained from Streptomyces sp. Sp080513GE-23 strain collected from a marine sponge, Haliclona sp. [4]. The nonribosomal peptidesan has an unusual 4-methyloxazoline moiety. Experiments showed that the methyl group comes from alanine rather than methionine [5]. Ageloline A (3), a new chlorinated quinolone separated from a fermentation of Streptomyces sp. SBT345, showed antioxidant effect and reduced oxidative stress and genomic damage induced by an oxidative stress inducer 4-nitroquinoline-1-oxide [6]. Compound 3 also inhibited the growth of Chlamydia trachomatis with an IC50 value of 9.54 μM [7]. New 3-phenylpropanoic acids 3-(3,5-dichloro-4-hydroxyphenyl) propanoic acid (4), 3-(3,5-dichloro-4-hydroxyphenyl) propanoic acid methyl ester (5) and 3-(3-chloro-4-hydroxyphenyl) propanoic acid (6) were isolated from Streptomyces coelicolor LY001, which demonstrated a broad spectrum of antibacterial activities with MIC values ranging from 16 to 250 μg/mL [8].

2.2. Corals-Associated Streptomyces sp.

Strepchloritides A (7) and B (8) were separated from a culture of Streptomyces sp. OUCMDZ-1703 and were cytotoxic against MCF-7 with IC50 values of 9.9 and 20.2 μM, respectively [9].

2.3. Streptomyces sp. from Other Marine Animals

A new depsipeptide salinamide B (9) was isolated from Streptomyces hygroscopicus. Compound 9 exhibited inhibitory activity against Streptococcus pneumoniae and Staphylococcus pyrogenes, with MIC values of 4 and 2 μg/mL, respectively. Compound 9 also exhibited 83% inhibition of edema with a testing dose of 50 pg/ear [10]. Polyketide–cyclic peptide hybrid metabolites totopotensamides A (10) and B (11) were separated from Streptomyces sp. 1053U.I.1a.1b [11]. New napyradiomycin, MDN-0170 (12) was purified from Streptomyces sp. strain CA-271078 [12]. Streptomyces sp. strain CA-271078 yielded napyradiomycin analogs napyradiomycin B7a, napyradiomycin B7b and napyradiomycin D1 (1315), which were cytotoxic against HepG-2 tumor cell line with IC50 values of 41.7, 109.5 and 14.9 μM, respectively. Compounds 13 and 15 showed anti-bacterial activity against methicillin resistant Staphylococcus aureus (MRSA) and Mycobacterium tuberculosis with MIC values in the range of 12 to 48 μg/mL [13].

2.4. Streptomyces sp. from Marine Sediments

Cultivation of Streptomyces sp. M045 afforded chinikomycins A-B (1617). Compound 16 inhibited tumor cell lines MAXF 401NL, MEXF 462NL and RXF 944L with IC50 values of 2.41, 4.15 and 4.02 µg/mL, respectively. Compound 17 inhibited MAXF 401NL with an IC50 value of 3.04 µg/mL [14]. An unusual meroterpenoid phthalazinone azamerone (18) was isolated from Streptomyces sp. CNQ766 [15]. In the synthesis of azamerone, Lewis-acid-induced cyclization, enantioselective synthesis of an epoxysilane and the formation of the pyridazine ring were three key steps [16]. A bohemamine-type pyrrolizidine alkaloid 5-chlorobohemamine C (19) was obtained from a culture of Streptomyces sp. CNQ-583 [17]. A pentacyclic C-glycoside marmycin B (20) was obtained from Streptomyces sp. CNH990, which displayed inhibitory activity against HCT-116 with an IC50 value of 1.09 μM [18]. Streptomyces sp. 04DH110 produced a new 3-substituted indole streptochlorin (21), which displayed cytotoxicity against human leukemia cells with an IC50 value of 1.05 μg/mL [19]. Total synthesis of streptochlorin started from indole, the synthetic product exhibited potential antifungal activity [20]. Three new hexadepsipeptides piperazimycins A–C (2224) were isolated from Streptomyces sp. CNQ-593, which showed cytotoxicity against HCT-116 with an equal IC50 value of 76 ng/mL [21]. The formation of dipeptide moiety and a macrocyclization by an SN2 reaction were the two key steps in the synthesis of piperazimycin A [22]. Three new chlorinated dihydroquinones (2527) were separated from culture of Actinomycete strain CNQ-525, and compounds 2527 were active against MRSA and vancomycin-resistant Enterococcus faecium with MIC values of 1.95 and 3.90, 15.6 and 15.6 and 1.95 and 1.95 µg/mL, respectively. Compounds 25 and 26 showed cytotoxicity against HCT-116 with the IC50 values of 2.40 and 0.97 μg/mL, respectively [23].
Heterologous expression of the CNQ-525-based nap biosynthetic cluster in Streptomyces albus produced 2-deschloro-2-hydroxy-A80915C (28), a new napyradiomycin [24]. Four napyradiomycin derivatives napyradiomycin CNQ525.510B (29), napyradiomycin CNQ525.538 (30), napyradiomycin CNQ525.554 (31) and napyradiomycin CNQ525.600 (32), were purified from the same strain, of which compounds 29, 30 and 32 displayed inhibitory activity against HCT-116 with IC50 values of 17, 6 and 49 μM, respectively [25]. New marinopyrroles dimeric marinopyrroles A (33) (Figure 2) and B (34) were obtained from Streptomyces sp. CNQ-418 and were cytotoxic toward HCT-116 with IC50 values of 8.8 and 9.0 μM, respectively. Compounds 33 and 34 were also active against methicillin-resistant S. aureus (MRSA) with MIC values of 0.61 and 1.1 μM, respectively [26]. Marinopyrroles C–F (3538) were obtained from the same strain, which displayed inhibitory activity against HCT-116 with IC50 values ranging from 1 to 5 μg/mL. Compound 35 was active against MRSA with an MIC value less than 1 μg/mL [27]. Ammosamides A (39) and B (40) were separated from Streptomyces sp. CNR-698, which showed cytotoxicity against HCT-116 cells with an equal IC50 value of 320 nM [28]. Ammosamides A and B were synthesized from 4-chloroisatin [29].
A cytotoxic compound, mansouramycin B (41) was isolated from the fermentation broth of Streptomyces sp. Mei37 [30]. Compound 41 was synthesized by using a new method through a catalytic acid-mediated cyclization of α-benzyl TosMIC derivatives [31]. Streptomyces malaysiensis CNQ-509 afforded nitropyrrolins C (42) and E (43), and 42 displayed cytotoxic activity against HCT-116 with an IC50 value of 31.0 μM [32]. Streptomyces sp. CNH-189 produced merochlorins A–D (4447) [33]. Spiroindimicins A–D (4851) [34], indimicins A–E (5256), lynamicin F (57) and lynamicin G (58) were separated from Streptomyces sp. SCSIO 03032, among which 4952 displayed cytotoxic activity against a panel of cancer cell lines with IC50 values ranging from 4 to 15 μM. Compound 53 also showed cytotoxicity toward MCF-7, with an IC50 value of 10.0 µM [35]. Merochlorins A and B were synthesized by heterologously produced enzymes and chemical synthesis [36]. (±)-Spiroindimicins B and C were synthesized, and central to the successful strategy was installing the spirocenter [37]. Chloroxiamycin (59), was isolated from Streptomyces sp. SCSIO 02999, which displayed antimicrobial activity against E. coli ATCC 25922, S. aureus ATCC29 213 and B. subtilis SCSIO BS01 with MIC values of 4, 8 and 64 μg/mL, respectively [38]. Streptomyces variabilis SNA-020 afforded an oxidatively ring opened ammosamide analog ammosamide D (60), which displayed cytotoxic activity against the MIA PaCa-2 with an IC50 value of 3.2 μM [39]. Cultivation of Streptomyces sp. CNT-179 strain afforded cyanosporasides C–E (6163) [40]. Chlorizidine A (64) was isolated from Streptomyces sp. CNH-287, which showed cytotoxic activity against the HCT-116 adenocarcinoma cell line with an IC50 value of 3.2–4.9 μM [41]. (±)-Chlorizidine A was synthesized by decarboxylative coupling and late-stage oxidation, Reformatsky reaction and Mitsunobu reactions [42]. Streptomyces sp. CNQ-329 yielded five new halogenated napyradiomycins A and C–E (6568) (Figure 3), of which compounds 65, 67 and 68 exhibited inhibitory activity towards HCT-116 with IC50 values of 4.2, 16.1 and 4.8 μg/mL, respectively. Compound 65 displayed antibacterial activity against MRSA with an MIC value of 16 µg/mL [43]. Napyradiomycin F (69) from Streptomyces sp. CNH-070 showed inhibitory activity against HCT-116, with an IC50 value of 9.42 μg/mL [43].
Streptomyces sp. SCSIO 10,428 afforded three new napyradiomycins 4a-dechloronapyradiomycin A1 (70), 3-dechloro-3-brominapyradiomycin A1 (71) and 3-chloro-6,8-dihydroxy-α-lapachone (72). Compound 70 demonstrated antibacterial activity against Staphylococcus aureus ATCC 29213, Bacillus subtilis SCSIO BS01 and Bacillus thuringensis SCSIO BT01 with MIC values of 4, 4 and 8 μg/mL; 71 exhibited antibacterial activity against Staphylococcus aureus ATCC 29213, Bacillus subtilis SCSIO BS01 and Bacillus thuringensis SCSIO BT01 with MIC values of 0.5, 1 and 1 μg/mL; and 72 showed antibacterial activity against Bacillus subtilis SCSIO BS01 and Bacillus thuringensis SCSIO BT01 with MIC values of 8 and 16 μg/mL, respectively. Compound 70 also displayed inhibitory activity against SF-268, MCF-7, NCI-H460 and HepG-2 with IC50 values of 22.8 ± 0.3, 20.6 ± 0.1, 22.4 ± 0.1 and 21.8 ± 0.5 μM, respectively; 71 showed inhibitory activity against SF-268, MCF-7, NCI-H460 and HepG-2 with IC50 values of 11.5 ± 1.2, 16.2 ± 0.7, 18.1 ± 0.3 and 17.1 ± 1.0 μM, respectively; and 72 exhibited inhibitory activity against SF-268, MCF-7, NCI-H460 and HepG-2 with IC50 values of 23.8 ± 2.2, 71.1 ± 0.4, 127.1 ± 0.9 and 59.4 ± 0.7 μM, respectively [44]. C-1027 chromophore-V (73) was obtained from a marine-derived Streptomyces sp. ART5, which showed inhibitory activity against Candida albicans isocitrate lyase with an IC50 value of 37.9 μM.
Compound 73 also inhibited MDA-MB231 and HCT-116 with IC50 values of 0.9 and 2.7 μM, respectively [45]. Chlorinated alkaloids inducamides A (74) and C (75) were separated from Streptomyces sp. SNC-109-M3, of which compound 75 showed cytotoxicity against NSCLC cell line HCC44 with an IC50 value of 10 μM [46]. Inducamide A (74) was synthesized from 6-hydroxy-3-chloro-2-methylbenzoic acid and L-6-chlorotryptophan [47]. Inducamide C (75) is unstable and easy to rearrange [48].
Hormaomycins B (76) and C (77) were obtained from a marine-derived Streptomyces sp. SNM55. Compounds 76 and 77 were active against S. aureus ATCC 25923, B. subtilis ATCC 6633, K. rhizophila NBRC 12708, S. pyogenes ATCC 19615, S. enterica ATCC 14,028 and P. hauseri NBRC 3851 with MIC values of 7/7, 14/56, 0.4/0.23, 14/8, 29/114 and 29/14 μM, respectively [49]. Two new phenazines marinocyanins A and B (78 and 79) were isolated from Streptomyces sp. CNS284, which inhibited the TNF-α-induced NF-κB activity with IC50 values of 4.1 and 24.2 μM and suppressed the PGE2 production with IC50 values of 7.15 and 0.89 μM, respectively [50]. Compound 78 inhibited LPS-induced nitric oxide production with an IC50 value of 15.1 μM [50]. Compounds 78 and 79 showed cytotoxicity against HCT-116 cell with IC50 values of 0.049 and 0.029 μM and inhibited S. aureus and C. albicans with MIC values of 2.37/33.92 and 0.95/5.79 μg/mL, respectively [51]. Marinocyanins A and B were synthesized through the establishment of the N-substituted phenazin-1-one skeleton [52]. Four phenazinone named marinocyanins C–F (8083) were purified from the marine actinomycete Streptomycetaceae CNS-284, which were active against S. aureus and C. albicans with MIC values ranging from 3.90 to 36.62 μg/mL. They also showed cytotoxicity against HCT-116 cell with IC50 values ranging from 0.078 to 17.14 μM [51]. A new tetrahydroanthracene alokicenone D (84) was isolated from the cultures of Streptomyces sp. HN-A101 [53]. New cyclizidine-type alkaloids cyclizidines D (85), H (86) and I (87) were purified from Streptomyces sp. HNA39. Compounds 86 and 87 exhibited inhibition against the ROCK2 protein kinase with IC50 values of 42 ± 3 and 39 ± 1 μM; 86 and 87 also showed cytotoxicity against PC-3 with IC50 values of 33 ± 1 and 17 ± 1 μM, respectively. Compound 87 demonstrated cytotoxicity against HCT-116 with an IC50 value of 40 ± 1 μM [54].
2,4-Dichlorophenyl 2,4-dichloro benzoate (88) (Figure 4) was obtained from Streptomyces sp. G212. Compound 88 was active against C. albicans with an MIC value of 64 μg/mL [55]. Streptomyces sp. CNH-189 afforded two new meroterpenoids merochlorins E (89) and F (90), which showed antibacterial activities against S. aureus, B. subtilis and K. rhizophila with MIC values ranging from 1 to 2 μg/mL [56]. Two new chlorinated bisindole alkaloids, dionemycin (91) and 6-OMe-7′,7″-dichorochromopyrrolic acid (92) were isolated from Streptomyces sp. SCSIO 11,791 [57]. Compound 91 displayed cytotoxic activity against MD1-MB-435, MDA-MB-231, NCI-H460, HCT-116, HepG2, and MCF10A with MIC values in the range of 3.1–11.2 μM. Compound 92 showed cytotoxic activity against human cancer cell lines MD1-MB-435, HCT-116, HepG2, and MCF10A with MIC values ranging from 2.9 to 19.4 μM.

2.5. Streptomyces sp. from Other Marine Sources

New dibenzoxazepinones mycemycins C–E (9395) were separated from Streptomyces olivaceus FXJ8.012Δ1741 [58]. A sulfonate-containing analog totopotensamide C (96) was isolated from Streptomyces pactum SCSIO 02,999 [59]. One new polycyclictetramate macrolactam pactamide F (97) was also purified from Streptomyces pactum SCSIO 02,999 [60]. Cultivation of Streptomyces sp. ZZ502 afforded a new cyclohexene 3-amino-2-carboxamine-6(R)-chloro-4(R)-5(S)-dihydroxy-cyclohex-2-en-1-one (98) [61].

3. Halogenated Compounds from Other Marine Actinomycetes

3.1. Other Marine Sediments-Associated Actinomycetes

Actinomycete CNB-632 (sediment, the Tot-my Pines Estuary) yielded a sesquiterpenoid naphthoquinone marinone (99) that was active against Bacillus subtilis with an MIC value of 1 μg/mL [62]. An actinomycete strain (# CNH-099) produced isomarinone (100). Compound 100 displayed cytotoxicity against a colon carcinoma cell line HCT-116 with an MIC value of 8 μg/mL [63]. Salinosporamide A (101) with a γ-lactam-β-lactone bicyclic ring was isolated from Salinospora strain CNB-392 (later assigned as Salinispora tropica), which showed cytotoxicity against a panel of cancer cell lines with IC50 values less than 10 nM and exhibited prominent inhibition of the 20S proteasome [64]. Compound 101 entered phase I human clinical trials for the treatment of multiple myeloma three years after its discovery in 2003 [64]. Salinosporamide A was synthesized from (R)-pyroglutamic acid [65]. Salinispora tropica CNB-392 produced sporolides A (102) and B (103) [66]. Compound 103 exhibited inhibitory activity against HIV-1 reverse transcriptase [67]. Compound 103 was synthesized by ruthenium-catalyzed [2+2+2] cycloaddition and Diels–Alder-type reaction [68]. Salinispora tropica CNB-392 yielded salinosporamide F (104), salinosporamide I (105), salinosporamide J (106) and bromosalinosporamide (107). Compound 106 displayed RPMI 8226 and chymotrypsin-like activity with IC50 values of 52 and 250 nM, respectively [69]. Fermentation of Salinispora pacifica (designated CNS103) derived from sediments led to the identification of cyclopenta[a]indene glycosides cyanosporasides A and B (108 and 109). Compound 108 was cytotoxic against HCT-116 with an IC50 value of 30 µg/mL [70]. Lynamicins A–E (110114) were afforded by Marinispora sp. NPS12745, which showed antibacterial activity against MRSA and vancomycin-resistant E. faecium with IC50 values in the range of 1.8–57.0 μg/mL [71].
Lodopyridone (115) (Figure 5) from Saccharomonospora sp. (marine sediment, the La Jolla Submarine Canyon) showed cytotoxicity against HCT-116 cell line with an IC50 value of 3.6 μM [72]. Saccharomonospora sp. CNQ-490 afforded taromycin A (116), which exhibited antibacterial activity against MRSA and Enterococcus faecalis 613D with MIC values ranging from 6 to 100 μM [73]. Fijiolides A and B (117 and 118) were isolated from the cultures of bacterium of the genus Nocardiopsis CNS-653 (sediment sample, Fiji). Compound 117 showed QR1 activity and was active against TNF-R-induced NF-κB with an IC50 value of 0.57 μM. Compound 118 showed activity against TNF-R-induced NFκB [74].
Phocoenamicins B (119) and C (120) were isolated from Micromonospora sp. CA-214671, and both compounds showed a broad spectrum of antibacterial activities with MIC values ranging from 2 to 64 μg/mL [75].

3.2. Other Ascidian-Associated Actinomycetes

Halomadurones A–D (121124) were obtained from Actinomadura sp. WMMB499 (ascidian Ecteinascidia turbinata), among which 123 and 124 showed activity against Nrf2-ARE [76]. A new halimane-type diterpenoid micromonohalimane B (125) was isolated from a culture of Micromonospora sp. WMMC-218, which inhibited MRSA with an MIC value of 40 μg/mL [77].

3.3. Other Marine Source-Associated Actinomycetes

Saccharochlorines A (126) and B (127) were isolated from Saccharomonospora sp. KCTC-19160, and both compounds showed BACE1 inhibition of 41.4 ± 3.6% and 32.0 ± 9.7%, respectively. at the same concentration of 50 μM (a positive control, isoliquiritigenin, 56.7% inhibition at 50 μM) [78].

4. Summary

According to the summary of halogenated compounds from marine-derived actinomycetes (Figure 6 and Table 1), the study of halogenated compounds from marine-derived actinomycetes could be traced back to 1992 when marinone (99) was purified from an actinomycete strain CNB-632 isolated from a sediment sample (Table 2) [62]. Since 2005, more new halogenated compounds from marine-derived actinomycetes have been isolated annually than ever before except for 2016. From 2010 to 2014 and in 2020, 10 or more new halogenated compounds were reported annually. By the end of 2020, 127 new halogenated compounds from marine-derived actinomycetes have been reported.
Sediments were the richest source of marine-derived actinomycetes, which produced about 78% of new halogenated compounds (Figure 7). It was reported that sediments are rich in nutrients, which can harbor an enormous quantity of microorganisms, including actinomycetes. It is worth mentioning that, the deeper and older the sediment is, the less abundant the microbes. Nevertheless, marine actinobacteria in sediments will keep providing opportunities for natural product research and natural product drug discovery.
Marine Streptomyces spp. had the highest occurrence of halogenated compounds (98/127 = 77%) (Figure 8), which might be due to their unique and diverse biosynthetic machinery, high halogenase activity or simply Streptomyces being the largest genus of Actinobacteria. Overall, 70.1% of halogenated compounds from marine actinomycetes is biologically active, and 37.3% and 24.6% of the halogenated compounds showed anticancer and antimicrobial activity, respectively (Figure 9).
The structure types of the new halogenated compounds were diverse, which could be classified as nitrogen-containing compounds, polyketides and terpenoids. Nitrogen-containing compounds and polyketides were two main classes of compounds produced by marine actinomycetes (Figure 10). The number of chlorinated compounds generated by marine actinomycetes is 10 times more than that of brominated compounds (Figure 11), which may be related to the concentrations of chloride and bromide ions in the ocean. Fluorinated natural products were reported before, but no new fluorinated compounds were discovered from marine actinomycetes recently.
In short, marine actinomycetes have unique and diverse biogenetic machinery, which can produce different halogenated compounds with novel structure skeletons and various biological activities, and Streptomyces spp. from sediments are the main producers. Some halogen-containing drugs such as chloramphenicol, vancomycin, chlortetracycline, calicheamicin, rebeccamycin and complestatin have been developed from secondary metabolites isolated from terrestrial actinomycetes [3]. Marine natural products have higher success rate (1 in 3500) in drug discovery, compared with the industry average of 1 in 5000–10,000 compounds [79]. Therefore, halogenated compounds from marine actinomycetes are expected to be a promising source of lead compounds for natural product drug discovery.

Author Contributions

S.C. and C.W. conceived and designed the format of the paper; C.W. and W.D. edited the article; J.L. analyzed the data; H.L. and K.L. drew the structures of the compounds; and S.C. revised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Specific research project of Guangxi for research bases and talents (AD20297036), Guangxi Natural Science Foundation under Grant (2019GXNSFBA185002), Specific research project of Guangxi for research bases and talents (AD18281066, AD18126005), Seed Grants from University of Hawaii at Hilo (UHH), start-up funding from University of Hawaii Cancer Center (UHCC) and Daniel K. Inouye College of Pharmacy (DKICP), and Hawaii Community Foundation (15ADVC-74420, 17CON-86295, and 20CON-102163) (to SC). Funding for this work was also supported by Hawaii IDeA Network for Biomedical Research Excellence III and IV (INBRE-III and INBRE-IV) project: NIGMS Grant 5P20GM103466.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This research was supported by Specific research project of Guangxi for research bases and talents (AD20297036), Guangxi Natural Science Foundation under Grant (2019GXNSFBA185002), Specific research project of Guangxi for research bases and talents (AD18281066, AD18126005), Seed Grants from University of Hawaii at Hilo (UHH), start-up funding from University of Hawaii Cancer Center (UHCC) and Daniel K. Inouye College of Pharmacy (DKICP), and Hawaii Community Foundation (15ADVC-74420, 17CON-86295, and 20CON-102163) (to SC). Funding for this work was also supported by Hawaii IDeA Network for Biomedical Research Excellence III and IV (INBRE-III and INBRE-IV) project: NIGMS Grant 5P20GM103466.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhao, C.; Zhu, T.; Zhu, W. New marine natural products of microbial origin from 2010 to 2013. Chin. J. Org. Chem. 2013, 33, 1195–1234. [Google Scholar] [CrossRef] [Green Version]
  2. Wang, C.; Mei, X.G.; Zhu, W.M. New natural products from the marine-derived Streptomyces actinobacteria. Stud. Mar. Sin. 2016, 51, 86–124. [Google Scholar]
  3. Kasanah, N.; Triyanto, T. Bioactivities of halometabolites from marine actinobacteria. Biomolecules 2019, 9, 225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Motohashi, K.; Takagi, M.; Shin-Ya, K. Tetrapeptides possessing a unique skeleton, JBIR-34 and JBIR-35, isolated from a sponge-derived actinomycete, Streptomyces sp. Sp080513GE-23. J. Nat. Prod. 2010, 73, 226–228. [Google Scholar] [CrossRef]
  5. Muliandi, A.; Katsuyama, Y.; Sone, K.; Izumikawa, M.; Moriya, T.; Hashimoto, J.; Kozone, I.; Takagi, M.; Shin-ya, K.; Ohnishi, Y. Biosynthesis of the 4-methyloxazoline-containing nonribosomal peptides, JBIR-34 and-35, in Streptomyces sp. Sp080513GE-23. Chem. Biol. 2014, 21, 923–934. [Google Scholar] [CrossRef] [Green Version]
  6. Jakubiec-Krzesniak, K.; Rajnisz-Mateusiak, A.; Guspiel, A.; Ziemska, J.; Solecka, J. Secondary metabolites of actinomycetes and their antibacterial, antifungal and antiviral properties. Pol. J. Microbiol. 2018, 67, 259. [Google Scholar] [CrossRef] [Green Version]
  7. Cheng, C.; Othman, E.M.; Reimer, A.; Grüne, M.; Kozjak-Pavlovic, V.; Stopper, H.; Hentschel, U.; Abdelmohsen, U.R. Ageloline A, new antioxidant and antichlamydial quinolone from the marine sponge-derived bacterium Streptomyces sp. SBT345. Tetrahedron Lett. 2016, 57, 2786–2789. [Google Scholar] [CrossRef]
  8. Shaala, L.A.; Youssef, D.T.; Alzughaibi, T.A.; Elhady, S.S. Antimicrobial chlorinated 3–phenylpropanoic acid derivatives from the Red Sea marine actinomycete Streptomyces coelicolor LY001. Mar. Drugs 2020, 18, 450. [Google Scholar] [CrossRef]
  9. Fu, P.; Kong, F.; Wang, Y.; Wang, Y.; Liu, P.; Zuo, G.; Zhu, W. Antibiotic metabolites from the coral–associated actinomycete Streptomyces sp. OUCMDZ–1703. Chin. J. Chem. 2013, 31, 100–104. [Google Scholar] [CrossRef]
  10. Trischman, J.A.; Tapiolas, D.M.; Jensen, P.R.; Dwight, R.; Fenical, W.; McKee, T.C.; Ireland, C.M.; Stout, T.J.; Clardy, J. Salinamides A and B anti-inflammatory depsipeptides from a marine Streptomycete. J. Am. Chem. Soc. 1994, 116, 757–758. [Google Scholar] [CrossRef]
  11. Lin, Z.; Flores, M.; Forteza, I.; Henriksen, N.M.; Concepcion, G.P.; Rosenberg, G.; Haygood, M.G.; Olivera, B.M.; Light, A.R.; Cheatham, T.E., III; et al. Totopotensamides, polyketide–cyclic peptide hybrids from a mollusk-associated bacterium Streptomyces sp. J. Nat. Prod. 2012, 75, 644–649. [Google Scholar] [CrossRef] [Green Version]
  12. Lacret, R.; Pérez-Victoria, I.; Oves-Costales, D.; De la Cruz, M.; Domingo, E.; Martín, J.; Díaz, C.; Vicente, F.; Genilloud, O.; Reyes, F. MDN-0170, a new napyradiomycin from Streptomyces sp. strain CA-271078. Mar. Drugs 2016, 14, 188. [Google Scholar] [CrossRef] [Green Version]
  13. Carretero-Molina, D.; Ortiz-López, F.J.; Martín, J.; Oves-Costales, D.; Díaz, C.; de la Cruz, M.; Cautain, B.; Vicente, F.; Genilloud, O.; Reyes, F. New napyradiomycin analogues from Streptomyces sp. strain CA-271078. Mar. Drugs 2020, 18, 22. [Google Scholar] [CrossRef] [Green Version]
  14. Li, F.; Maskey, R.P.; Qin, S.; Sattler, I.; Fiebig, H.H.; Maier, A.; Zeeck, A.; Laatsch, H. Chinikomycins A and B: Isolation, structure elucidation, and biological activity of novel antibiotics from a marine Streptomyces sp. isolate M045#. J. Nat. Prod. 2005, 68, 349–353. [Google Scholar]
  15. Cho, J.Y.; Kwon, H.C.; Williams, P.G.; Jensen, P.R.; Fenical, W. Azamerone, a terpenoid phthalazinone from a marine-derived bacterium related to the genus Streptomyces (Actinomycetales). Org. Lett. 2006, 8, 2471–2474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Schnell, S.D.; Linden, A.; Gademann, K. Synthesis of two key fragments of the complex polyhalogenated marine meroterpenoid azamerone. Org. Lett. 2019, 21, 1144–1147. [Google Scholar] [CrossRef] [Green Version]
  17. Bugni, T.S.; Woolery, M.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Bohemamines from a marine-derived Streptomyces sp. J. Nat. Prod. 2006, 69, 1626–1628. [Google Scholar] [CrossRef] [PubMed]
  18. Martin, G.D.; Tan, L.T.; Jensen, P.R.; Dimayuga, R.E.; Fairchild, C.R.; Raventos-Suarez, C.; Fenical, W. Marmycins A and B, cytotoxic pentacyclic C-glycosides from a marine sediment-derived actinomycete related to the genus Streptomyces. J. Nat. Prod. 2007, 70, 1406–1409. [Google Scholar] [CrossRef] [PubMed]
  19. Shin, H.J.; Jeong, H.S.; Lee, H.S.; Park, S.K.; Kim, H.M.; Kwon, H.J. Isolation and structure determination of streptochlorin, an antiproliferative agent from a marine-derived Streptomyces sp. 04DH110. J. Microbiol. Biotechnol. 2007, 17, 1403–1406. [Google Scholar]
  20. Jia, C.Y.; Xu, L.Y.; Yu, X.; Ding, Y.B.; Jin, B.; Zhang, M.Z.; Zhang, W.H.; Yang, G.F. An efficient synthesis and antifungal evaluation of natural product streptochlorin and its analogues. Fitoterapia 2018, 125, 106–110. [Google Scholar] [CrossRef] [PubMed]
  21. Miller, E.D.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Piperazimycins: Cytotoxic hexadepsipeptides from a marine–derived bacterium of the genus Streptomyces. J. Org. Chem. 2007, 72, 323–330. [Google Scholar] [CrossRef]
  22. Li, W.; Gan, J.; Ma, D. Total synthesis of piperazimycin A: A cytotoxic cyclic hexadepsipeptide. Angew. Chem. Int. Ed. 2009, 121, 9053–9057. [Google Scholar] [CrossRef]
  23. Soria–Mercado, I.E.; Prieto–Davo, A.; Jensen, P.R.; Fenical, W. Antibiotic terpenoid chloro–dihydroquinones from a new marine actinomycete. J. Nat. Prod. 2005, 68, 904–910. [Google Scholar] [CrossRef] [PubMed]
  24. Winter, J.M.; Moffitt, M.C.; Zazopoulos, E.; McAlpine, J.B.; Dorrestein, P.C.; Moore, B.S. Molecular basis for chloronium–mediated meroterpene cyclization-cloning, sequencing, and heterologous expression of the napyradiomycin biosynthetic gene cluster. J. Biol. Chem. 2007, 282, 16362–16368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Farnaes, L.; Coufal, N.G.; Kauffman, C.A.; Rheingold, A.L.; DiPasquale, A.G.; Jensen, P.R.; Fenical, W. Napyradiomycin derivatives, produced by a marine–derived actinomycete, illustrate cytotoxicity by induction of apoptosis. J. Nat. Prod. 2014, 77, 15–21. [Google Scholar] [CrossRef] [Green Version]
  26. Hughes, C.C.; Prieto-Davo, A.; Jensen, P.R.; Fenical, W. The marinopyrroles, antibiotics of an unprecedented structure class from a marine Streptomyces sp. Org. Lett. 2008, 10, 629–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Hughes, C.C.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Structures, reactivities, and antibiotic properties of the marinopyrroles A–F. J. Org. Chem. 2010, 75, 3240–3250. [Google Scholar] [CrossRef] [Green Version]
  28. Hughes, C.C.; MacMillan, J.B.; Gaudêncio, S.P.; Jensen, P.R.; Fenical, W. The ammosamides: Structures of cell cycle modulators from a marine–derived Streptomyces species. Angew. Chem. Int. Ed. Engl. 2009, 48, 725–727. [Google Scholar] [CrossRef] [Green Version]
  29. Hughes, C.C.; Fenical, W. Total synthesis of the ammosamides. J. Am. Chem. Soc. 2010, 132, 2528–2529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Hawas, U.W.; Shaaban, M.; Shaaban, K.A.; Speitling, M.; Maier, A.; Kelter, G.; Fiebig, H.H.; Meiners, M.; Helmke, E.; Laatsch, H. Mansouramycins A–D, cytotoxic isoquinolinequinones from a marine Streptomycete. J. Nat. Prod. 2009, 72, 2120–2124. [Google Scholar] [CrossRef]
  31. Coppola, A.; Sucunza, D.; Burgos, C.; Vaquero, J.J. Isoquinoline synthesis by heterocyclization of tosylmethyl isocyanide derivatives: Total synthesis of mansouramycin B. Org. Lett. 2015, 17, 78–81. [Google Scholar] [CrossRef] [PubMed]
  32. Kwon, H.C.; Espindola, A.P.D.; Park, J.S.; Prieto-Davó, A.; Rose, M.; Jensen, P.R.; Fenical, W. Nitropyrrolins A–E, cytotoxic farnesyl–α–nitropyrroles from a marine–derived bacterium within the actinomycete family Streptomycetaceae. J. Nat. Prod. 2010, 73, 2047–2052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kaysser, L.; Bernhardt, P.; Nam, S.J.; Loesgen, S.; Ruby, J.G.; Skewes-Cox, P.; Jensen, P.R.; Fenical, W.; Moore, B.S. Merochlorins A–D, cyclic meroterpenoid antibiotics biosynthesized in divergent pathways with vanadium–dependent chloroperoxidases. J. Am. Chem. Soc. 2012, 134, 11988–11991. [Google Scholar] [CrossRef] [Green Version]
  34. Zhang, W.; Liu, Z.; Li, S.; Yang, T.; Zhang, Q.; Ma, L.; Tian, X.; Zhang, H.; Huang, C.; Zhang, S.; et al. Spiroindimicins A–D: New bisindole alkaloids from a deep–sea–derived actinomycete. Org. Lett. 2012, 14, 3364–3367. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, W.; Ma, L.; Li, S.; Liu, Z.; Chen, Y.; Zhang, H.; Zhang, G.; Zhang, Q.; Tian, X.; Yuan, C.; et al. Indimicins A–E, bisindole alkaloids from the deep–sea–derived Streptomyces sp. SCSIO 03032. J. Nat. Prod. 2014, 77, 1887–1892. [Google Scholar] [CrossRef]
  36. Teufel, R.; Kaysser, L.; Villaume, M.T.; Diethelm, S.; Carbullido, M.K.; Baran, P.S.; Moore, B.S. One-pot enzymatic synthesis of merochlorin A and B. Angew. Chem. Int. Ed. 2014, 53, 11019–11022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Blair, L.M.; Sperry, J. Total syntheses of (±)-spiroindimicins B and C enabled by a late-stage Schöllkopf–Magnus–Barton–Zard (SMBZ) reaction. Chem. Commun. 2016, 52, 800–802. [Google Scholar] [CrossRef]
  38. Zhang, Q.; Mándi, A.; Li, S.; Chen, Y.; Zhang, W.; Tian, X.; Zhang, H.; Li, H.; Zhang, W.; Zhang, S.; et al. N–N–Coupled indolo–sesquiterpene atropo–diastereomers from a marine–derived actinomycete. Eur. J. Org. Chem. 2012, 16, 2752–2755. [Google Scholar] [CrossRef]
  39. Pan, E.; Jamison, M.; Yousufuddin, M.; MacMillan, J.B. Ammosamide D, an oxidatively ring opened ammosamide analog from a marine–derived Streptomyces variabilis. Org. Lett. 2012, 14, 2390–2393. [Google Scholar] [CrossRef] [Green Version]
  40. Lane, A.L.; Nam, S.J.; Fukuda, T.; Yamanaka, K.; Kauffman, C.A.; Jensen, P.R.; Fenical, W.; Moore, B.S. Structures and comparative characterization of biosynthetic gene clusters for cyanosporasides, enediyne–derived natural products from marine actinomycetes. J. Am. Chem. Soc. 2013, 135, 4171–4174. [Google Scholar] [CrossRef] [Green Version]
  41. Alvarez-Mico, X.; Jensen, P.R.; Fenical, W.; Hughes, C.C. Chlorizidine, a cytotoxic 5H–pyrrolo [2,1–a]isoindol–5–one–containing alkaloid from a marine Streptomyces sp. Org. Lett. 2013, 15, 988–991. [Google Scholar] [CrossRef] [Green Version]
  42. Mahajan, J.P.; Mhaske, S.B. Synthesis of methyl-protected (±)-chlorizidine A. Org. Lett. 2017, 19, 2774–2776. [Google Scholar] [CrossRef]
  43. Cheng, Y.B.; Jensen, P.R.; Fenical, W. Cytotoxic and antimicrobial napyradiomycins from two marine–derived MAR 4 Streptomyces strains. Eur. J. Org. Chem. 2013, 18, 3751–3757. [Google Scholar] [CrossRef] [Green Version]
  44. Wu, Z.; Li, S.; Li, J.; Chen, Y.; Saurav, K.; Zhang, Q.; Zhang, H.; Zhang, W.; Zhang, W.; Zhang, S.; et al. Antibacterial and cytotoxic new napyradiomycins from the marine–derived Streptomyces sp. SCSIO 10428. Mar. Drugs 2013, 11, 2113–2125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Moon, K.; Ahn, C.H.; Shin, Y.; Won, T.H.; Ko, K.; Lee, S.K.; Oh, K.B.; Shin, J.; Nam, S.I.; Oh, D.C. New benzoxazine secondary metabolites from an arctic actinomycete. Mar. Drugs 2014, 12, 2526–2538. [Google Scholar] [CrossRef] [Green Version]
  46. Fu, P.; Jamison, M.; La, S.; MacMillan, J.B. Inducamides A–C, chlorinated alkaloids from an RNA polymerase mutant strain of Streptomyces sp. Org. Lett. 2014, 16, 5656–5659. [Google Scholar] [CrossRef] [Green Version]
  47. Scott, L.M.; Sperry, J. Synthesis of inducamides A and B. J. Nat. Prod. 2016, 79, 519–522. [Google Scholar] [CrossRef] [PubMed]
  48. Nabi, A.A.; Scott, L.M.; Furkert, D.P.; Sperry, J. Synthetic studies toward inducamide C. Org. Biomol. Chem. 2021, 19, 416–420. [Google Scholar] [CrossRef] [PubMed]
  49. Bae, M.; Chung, B.; Oh, K.B.; Shin, J.; Oh, D.-C. Hormaomycins B and C: New antibiotic cyclic depsipeptides from a marine mudflat–derived Streptomyces sp. Mar. Drugs 2015, 13, 5187–5200. [Google Scholar] [CrossRef] [Green Version]
  50. Kondratyuk, T.P.; Park, E.J.; Yu, R.; Van Breemen, R.B.; Asolkar, R.N.; Murphy, B.T.; Fenical, W.; Pezzuto, J.M. Novel marine phenazines as potential cancer chemopreventive and anti–inflammatory agents. Mar. Drugs 2012, 10, 451–464. [Google Scholar] [CrossRef] [Green Version]
  51. Asolkar, R.N.; Singh, A.; Jensen, P.R.; Aalbersberg, W.; Carté, B.K.; Feussner, K.D.; Subramani, R.; DiPasquale, A.; Rheingold, A.L.; Fenical, W. Marinocyanins, cytotoxic bromo–phenazinone meroterpenoids from a marine bacterium from the Streptomycete clade MAR4. Tetrahedron 2017, 73, 2234–2241. [Google Scholar] [CrossRef] [Green Version]
  52. Kohatsu, H.; Kamo, S.; Tomoshige, S.; Kuramochi, K. Total syntheses of pyocyanin, lavanducyanin, and marinocyanins A and B. Org. Lett. 2019, 21, 7311–7314. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, D.; Jiang, Y.; Li, J.; Zhang, H.; Ding, W.; Ma, Z. Alokicenones A-H, eight tetrahydroanthracenes from the mangrove–derived Streptomyces sp. HN–A101. Tetrahedron 2018, 74, 6667–6672. [Google Scholar] [CrossRef]
  54. Jiang, Y.J.; Li, J.Q.; Zhang, H.J.; Ding, W.J.; Ma, Z.J. Cyclizidine–type alkaloids from Streptomyces sp. HNA39. J. Nat. Prod. 2018, 81, 394–399. [Google Scholar] [CrossRef]
  55. Cao, D.T.; Nguyen, T.L.; Tran, V.H.; Doan-Thi-Mai, H.; Vu-Thi, Q.; Nguyen, M.A.; Le-Thi, H.M.; Chau, V.M.; Pham, V.C. Synthesis, structure and antimicrobial activity of novel metabolites from a marine actinomycete in Vietnam’s East Sea. Nat. Prod. Commun. 2019, 14, 121–124. [Google Scholar] [CrossRef]
  56. Ryu, M.J.; Hwang, S.; Kim, S.; Yang, I.; Oh, D.C.; Nam, S.J.; Fenical, W. Meroindenon and merochlorins E and F, antibacterial meroterpenoids from a marine–derived sediment bacterium of the genus Streptomyces. Org. Lett. 2019, 21, 5779–5783. [Google Scholar] [CrossRef] [PubMed]
  57. Song, Y.; Yang, J.; Yu, J.; Li, J.; Yuan, J.; Wong, N.K.; Ju, J. Chlorinated bis–indole alkaloids from deep–sea derived Streptomyces sp. SCSIO 11791 with antibacterial and cytotoxic activities. J. Antibiot. 2020, 73, 542–547. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, N.; Song, F.; Shang, F.; Huang, Y. Mycemycins A–E, new dibenzoxazepinones isolated from two different Streptomycetes. Mar. Drugs 2015, 13, 6247–6258. [Google Scholar] [CrossRef] [Green Version]
  59. Chen, R.; Zhang, Q.; Tan, B.; Zheng, L.; Li, H.; Zhu, Y.; Zhang, C. Genome mining and activation of a silent PKS/NRPS gene cluster direct the production of totopotensamides. Org. Lett. 2017, 19, 5697–5700. [Google Scholar] [CrossRef]
  60. Saha, S.; Zhang, W.; Zhang, G.; Zhu, Y.; Chen, Y.; Liu, W.; Yuan, C.; Zhang, Q.; Zhang, H.; Zhang, L.; et al. Activation and characterization of a cryptic gene cluster reveals a cyclization cascade for polycyclic tetramate macrolactams. Chem. Sci. 2017, 8, 1607–1612. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, X.; Shu, C.; Li, Q.; Lian, X.Y.; Zhang, Z. Novel cyclohexene and benzamide derivatives from marine–associated Streptomyces sp. ZZ502. Nat. Prod. Res. 2019, 33, 2151–2159. [Google Scholar] [CrossRef]
  62. Pathirana, C.; Jensen, P.R.; Fenical, W. Marinone and debromomarinone: Antibiotic sesquiterpenoid naphthoquinones of a new structure class from a marine bacterium. Tetrahedron Lett. 1992, 33, 7663–7666. [Google Scholar] [CrossRef]
  63. Hardt, I.H.; Jensen, P.R.; Fenical, W. Neomarinone, and new cytotoxic marinone derivatives, produced by a marine filamentous bacterium (actinomycetales). Tetrahedron Lett. 2000, 41, 2073–2076. [Google Scholar] [CrossRef]
  64. Feling, R.H.; Buchanan, G.O.; Mincer, T.J.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Salinosporamide A: A highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora. Angew. Chem. Int. Ed. Engl. 2003, 42, 355–357. [Google Scholar] [CrossRef]
  65. Endo, A.; Danishefsky, S.J. Total synthesis of salinosporamide A. J. Am. Chem. Soc. 2005, 127, 8298–8299. [Google Scholar] [CrossRef]
  66. Buchanan, G.O.; Williams, P.G.; Feling, R.H.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Sporolides A and B: Structurally unprecedented halogenated macrolides from the marine actinomycete Salinispora tropica. Org. Lett. 2005, 7, 2731–2734. [Google Scholar] [CrossRef] [PubMed]
  67. Dineshkumar, K.; Aparna, V.; Madhuri, K.Z.; Hopper, W. Biological activity of sporolides A and B from Salinispora tropica: In silico target prediction using ligand–based pharmacophore mapping and in vitro activity validation on HIV–1 reverse transcriptase. Chem. Biol. Drug Des. 2014, 83, 350–361. [Google Scholar] [CrossRef] [PubMed]
  68. Nicolaou, K.E.; Tang, Y.; Wang, J. Total synthesis of sporolide B. Angew. Chem. Int. Ed. 2009, 48, 3449–3453. [Google Scholar] [CrossRef] [Green Version]
  69. Reed, K.A.; Manam, R.R.; Mitchell, S.S.; Xu, J.; Teisan, S.; Chao, T.H.; Deyanat–Yazdi, G.; Neuteboom, S.T.; Lam, K.S.; Potts, B.C. Salinosporamides D–J from the marine actinomycete Salinispora tropica, bromosalinosporamide, and thioester derivatives are potent inhibitors of the 20S proteasome. J. Nat. Prod. 2007, 70, 269–276. [Google Scholar] [CrossRef] [PubMed]
  70. Oh, D.C.; Williams, P.G.; Kauffman, C.A.; Jensen, P.R.; Fenical, W. Cyanosporasides A and B, chloro– and cyano–cyclopenta [a] indene glycosides from the marine actinomycete “Salinispora pacifica”. Org. Lett. 2006, 8, 1021–1024. [Google Scholar] [CrossRef] [PubMed]
  71. McArthur, K.A.; Mitchell, S.S.; Tsueng, G.; Rheingold, A.; White, D.J.; Grodberg, J.; Lam, K.S.; Potts, B.C. Lynamicins A-E, chlorinated bisindole pyrrole antibiotics from a novel marine actinomycete. J. Nat. Prod. 2008, 71, 1732–1737. [Google Scholar] [CrossRef] [PubMed]
  72. Maloney, K.N.; MacMillan, J.B.; Kauffman, C.A.; Jensen, P.R.; DiPasquale, A.G.; Rheingold, A.L.; Fenical, W. Lodopyridone, a structurally unprecedented alkaloid from a marine actinomycete. Org. Lett. 2009, 11, 5422–5424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yamanaka, K.; Reynolds, K.A.; Kersten, R.D.; Ryan, K.S.; Gonzalez, D.J.; Nizet, V.; Dorrestein, P.C.; Moore, B.S. Direct cloning and refactoring of a silent lipopeptide biosynthetic gene cluster yields the antibiotic taromycin A. Proc. Natl. Acad. Sci. USA 2014, 111, 1957–1962. [Google Scholar] [CrossRef] [Green Version]
  74. Nam, S.J.; Gaudêncio, S.P.; Kauffman, C.A.; Jensen, P.R.; Kondratyuk, T.P.; Marler, L.E.; Pezzuto, J.M.; Fenical, W. Fijiolides A and B, inhibitors of TNF–alpha–Induced NF kappa B activation, from a marine–derived sediment bacterium of the genus Nocardiopsis. J. Nat. Prod. 2010, 73, 1080–1086. [Google Scholar] [CrossRef] [Green Version]
  75. Pérez–Bonilla, M.; Oves–Costales, D.; De la Cruz, M.; Kokkini, M.; Martín, J.; Vicente, F.; Genilloud, O.; Reyes, F. Phocoenamicins B and C, new antibacterial spirotetronates isolated from a marine Micromonospora sp. Mar. Drugs 2018, 16, 95. [Google Scholar] [CrossRef] [Green Version]
  76. Wyche, T.P.; Standiford, M.; Hou, Y.; Braun, D.; Johnson, D.A.; Johnson, J.A.; Bugni, T.S. Activation of the nuclear factor E2–related factor 2 pathway by novel natural products halomadurones A–D and a synthetic analogue. Mar. Drugs 2013, 11, 5089–5099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Zhang, Y.; Adnani, N.; Braun, D.R.; Ellis, G.A.; Barns, K.J.; Parker–Nance, S.; Guzei, I.A.; Bugni, T.S. Micromonohalimanes A and B: Antibacterial halimane–type diterpenoids from a marine Micromonospora species. J. Nat. Prod. 2016, 79, 2968–2972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Le, T.C.; Katila, N.; Park, S.; Lee, J.; Yang, I.; Choi, H.; Choi, D.Y.; Nam, S.J. Two new secondary metabolites, saccharochlorines A and B, from a marine bacterium Saccharomonospora sp. KCTC–19160. Bioorg. Med. Chem. Lett. 2020, 30, 127–145. [Google Scholar] [CrossRef]
  79. Pereira, F. Have marine natural product drug discovery efforts been productive and how can we improve their efficiency? Expert Opin. Drug Discov. 2019, 14, 717–722. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Structures of compounds 132.
Figure 1. Structures of compounds 132.
Molecules 26 02754 g001
Figure 2. Structures of compounds 3365.
Figure 2. Structures of compounds 3365.
Molecules 26 02754 g002
Figure 3. Structures of compounds 6687.
Figure 3. Structures of compounds 6687.
Molecules 26 02754 g003
Figure 4. Structures of compounds 88114.
Figure 4. Structures of compounds 88114.
Molecules 26 02754 g004
Figure 5. Structures of compounds 115127.
Figure 5. Structures of compounds 115127.
Molecules 26 02754 g005
Figure 6. Numbers of new halogenated compounds from actinomycetes reported annually from 1992 to 2020.
Figure 6. Numbers of new halogenated compounds from actinomycetes reported annually from 1992 to 2020.
Molecules 26 02754 g006
Figure 7. Percentages of new halogenated compounds from different sources of marine origins (1992–2020).
Figure 7. Percentages of new halogenated compounds from different sources of marine origins (1992–2020).
Molecules 26 02754 g007
Figure 8. Numbers of new halogenated compounds from different marine actinomycetes (1994–2019).
Figure 8. Numbers of new halogenated compounds from different marine actinomycetes (1994–2019).
Molecules 26 02754 g008
Figure 9. Activity of new halogenated compounds from marine actinomycetes (1992–2020).
Figure 9. Activity of new halogenated compounds from marine actinomycetes (1992–2020).
Molecules 26 02754 g009
Figure 10. Structural classes of new halogenated compounds from actinomycetes (1992–2020).
Figure 10. Structural classes of new halogenated compounds from actinomycetes (1992–2020).
Molecules 26 02754 g010
Figure 11. Proportion of new halogenated compounds from actinomycetes (1992–2020).
Figure 11. Proportion of new halogenated compounds from actinomycetes (1992–2020).
Molecules 26 02754 g011
Table 1. The initial research on halogenated compounds from marine-derived actinomycetes.
Table 1. The initial research on halogenated compounds from marine-derived actinomycetes.
First Producing StrainEnvironment SourceCompound.Time
Streptomyces sp. 1053U.I.1a.1bLienardia totopotens, Mactan Island, Cebu, Philippinestotopotensamides A (10) and B (11)1994
Actinomycete CNB-632 (other marine actinomycetes)Sediment sample, Tot-my Pines Estuary, La Jolla, CAmarinone (99)1992
Table 2. Halogenated compounds isolated from marine-derived actinomycetes.
Table 2. Halogenated compounds isolated from marine-derived actinomycetes.
Compound Producing Strain Environment Source BioactivityRef.
1–2Streptomyces sp. Sp080513GE-23Haliclona sp. Sponge, Chiba, Japan/[4,5]
3Streptomyces sp. SBT345Agelas oroides sponge, Mediterranean SeaAntioxidant and antichlamydial effects[6,7]
4–6Streptomyces coelicolor LY001sponge Callyspongia siphonella, the Saudi Red SeaAntibacterial activity[8]
7–8Streptomyces sp. OUCMDZ-1703Unidentified soft coral, Weizhou Island, Guangxi, ChinaCytotoxicity[9]
9Streptomyces hygroscopicusJellyfish Cassiopeia xamachana, Florida KeysAntibacterial, Anti- inflammatory activity[10]
10–11Streptomyces sp. 1053U.I.1a.1bLienardia totopotens, Mactan Island, Cebu, Philippines/[11]
12–15Streptomyces sp. Strain CA-271078Ascidian, the sea shore in Baía Ana Chaves, Sao Tome13–15: Cytotoxicity
13, 15: Antibacterial activity
[12,13]
16–17Streptomyces sp. M045Sediment, Jiaozhou Bay, ChinaCytotoxicity[14]
18Streptomyces sp. CNQ766Sediment, Island of Guam/[15,16]
19Streptomyces sp. CNQ-583Sediment, Island of Guam/[17]
20Streptomyces sp. CNH990Sediment, Cabo San Lucas, Mexico.Cytotoxicity[18]
21Streptomyces sp. 04DH110Sediments, Ayajin Bay, East Sea of KoreaCytotoxicity[19,20]
22–24Streptomyces sp. CNQ-593Sediment, Island of GuamCytotoxicity[21,22]
25–32Streptomcyces sp. CNQ525Sediment, La Jolla, CA26–27, 29–30, 32: Cytotoxicity
25–27: Antibacterial activity
[23,24,25]
33–38Streptomyces sp CNQ-418Sediment, La Jolla, CACytotoxicity[26,27]
39–40Streptomyces sp. CNR-698Sediment, Bahamas IslandsCytotoxicity[28,29]
41Streptomyces sp. Mei37Sediment, Jade Bay, GermanCytotoxicity[30,31]
42–43S. malaysiensis CNQ-509Sediment, California42: Cytotoxicity[32]
44–47Streptomyces sp. CNH-189Sediment, Oceanside, California/[33]
48–58Streptomyces sp. SCSIO 03032Sediment, Bay of Bengal49–51, 53: Cytotoxicity[34,35,36,37]
59Streptomyces sp. SCSIO 02999Sediment, South China SeaAntibacterial activity[38]
60Streptomyces variabilis SNA-020Sediment, BahamasCytotoxicity[39]
61–63Streptomyces sp. CNT-179Sediment, Bahamas/[40]
64Streptomyces sp. CNH-287Sediment, San Diego, CA.Cytotoxicity[41,42]
65–68Streptomyces sp. CNQ-329Sediment, San Diego, CA.65, 67–68: Cytotoxicity
65: Antibacterial
[43]
69Streptomyces sp. CNH-070Sediment, Encinitas, CaliforniaCytotoxicity[43]
70–72Streptomyces sp. SCSIO 10428Sediment, Beihai, Guangxi, ChinaAntibacterial activity [44]
73Streptomyces sp. ART5Sediment, East Siberian, Arctic OceanCytotoxicity[45]
74–75Streptomyces sp. SNC-109-M3 Sediment, Vava’u, Tonga74: Cytotoxicity[46,47,48]
76–77Streptomyces sp. SNM55Sediment, Buan, KoreaAntibacterial activity[49]
78–83Streptomycetaceae CNS-284 Marine sediments, the Solomon Islands and in Palau78–79: TNF-α-induced NFκB activity and antibacterial activity; 8083: Antibacterial activity and cytotoxicity[50,51,52]
84Streptomyces sp. HN-A101Mangrove soil, Hainan, China/[53]
85–87Streptomyces sp. HNA39 Marine sediment, Hainan, China86–87: Cytotoxicity[54]
88Streptomyces sp. G212Marine sediment, Quang Binh-VietnamAntifungl activity[55]
89–90Streptomyces sp. CNH-189Sediment, near Oceanside, California.Antibacterial activity[56]
91–92Streptomyces sp. SCSIO 11791Sediment, South China Seacytotoxicity[57]
93–95Streptomyces olivaceus FXJ8.012Δ1741a gntR gene-disrupted deep-sea strain/[58]
96–97Streptomyces pactum SCSIO 02999Sediment, South China Sea/[59,60]
98Streptomyces sp. ZZ502Seaweed Ulva conglobatea (Family Ulvaceae)./[61]
99Actinomycete CNB-632Sediment sample, Tot-my Pines Estuary, La Jolla, CA Antibacterial activity[62]
100Actinomycete (strain # CNH-099)Sediment, Batiquitos Lagoon, North of San Diego, CACytotoxicity[63]
101–107Salinospora strain CNB-392(later assigned as Salinispora tropica)Sediment, Chub Cay, Bahamas101, 106: Cytotoxicity
103: inhibitory activity against HIV-1 reverse transcriptase
106: chymotrypsin-like activity
[64,65,66,67,68,69]
108–109Salinispora pacifica (designated CNS103)Sediment, Palau108: Cytotoxicity[70]
110–114Marinispora sp. NPS12745Sediment, the coast of San Diego, CaliforniaAntibacterial activity[71]
115–116Saccharomonospora sp. CNQ490Marine sediment, the La Jolla Submarine CanyonCytotoxicity
116: Antibacterial activity
[72,73]
117–118Nocardiopsis CNS-653Sediment sample, FijiTNF-R-induced NFκB[74]
119–120Micromonospora sp. CA-214671Marine sediments, the Canary IslandsAntibacterial activity[75]
121–124Actinomadura sp. WMMB499Ascidian Ecteinascidia turbinata123–124: Nrf2-ARE activity[76]
125Micromonospora sp. WMMC-218Ascidian Symplegma brakenhielmi,
Florida, Stanblum State Park
Antibacterial activity[77]
126–127Saccharomonospora sp. KCTC-19160Korean Collection for Type CulturesBACE1 activity[78]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, C.; Du, W.; Lu, H.; Lan, J.; Liang, K.; Cao, S. A Review: Halogenated Compounds from Marine Actinomycetes. Molecules 2021, 26, 2754. https://doi.org/10.3390/molecules26092754

AMA Style

Wang C, Du W, Lu H, Lan J, Liang K, Cao S. A Review: Halogenated Compounds from Marine Actinomycetes. Molecules. 2021; 26(9):2754. https://doi.org/10.3390/molecules26092754

Chicago/Turabian Style

Wang, Cong, Weisheng Du, Huanyun Lu, Jianzhou Lan, Kailin Liang, and Shugeng Cao. 2021. "A Review: Halogenated Compounds from Marine Actinomycetes" Molecules 26, no. 9: 2754. https://doi.org/10.3390/molecules26092754

Article Metrics

Back to TopTop