Next Article in Journal
Xenoacremones D–H, Bioactive Tyrosine-decahydrofluorene Analogues from the Plant-Derived Fungus Xenoacremonium sinensis
Next Article in Special Issue
Taeanamides A and B, Nonribosomal Lipo-Decapeptides Isolated from an Intertidal-Mudflat-Derived Streptomyces sp.
Previous Article in Journal
The Development of the Bengamides as New Antibiotics against Drug-Resistant Bacteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genus Nocardiopsis: A Prolific Producer of Natural Products

1
College of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao 266590, China
2
State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200, China
*
Author to whom correspondence should be addressed.
Mar. Drugs 2022, 20(6), 374; https://doi.org/10.3390/md20060374
Submission received: 3 May 2022 / Revised: 26 May 2022 / Accepted: 30 May 2022 / Published: 31 May 2022
(This article belongs to the Special Issue Natural Products from Marine Bacteria)

Abstract

:
Actinomycetes are currently one of the major sources of bioactive secondary metabolites used for medicine development. Accumulating evidence has shown that Nocardiopsis, a key class of actinomycetes, has the ability to produce novel bioactive natural products. This review covers the sources, distribution, bioactivities, biosynthesis, and structural characteristics of compounds isolated from Nocardiopsis in the period between March 2018 and 2021. Our results reveal that 67% of Nocardiopsis-derived natural products are reported for the first time, and 73% of them are isolated from marine Nocardiopsis. The chemical structures of the Nocardiopsis-derived compounds have diverse skeletons, concentrating on the categories of polyketides, peptides, terphenyls, and alkaloids. Almost 50% of the natural products isolated from Nocardiopsis have been discovered to display various bioactivities. These results fully demonstrate the great potential of the genus Nocardiopsis to produce novel bioactive secondary metabolites that may serve as a structural foundation for the development of novel drugs.

Graphical Abstract

1. Introduction

Actinomycetes belong to Gram-positive bacteria and are one of the biggest bacterial phyla [1,2]. The high G+C DNA content of actinomycetes implied their enormous biosynthetic potential to produce various natural products with diverse structures and important commercial applications [1,2]. Two-thirds of all naturally derived antibiotics have been discovered from actinobacteria [3,4]. Approximately 70% of the pharmaceutically active natural products which are currently used in clinics are isolated from actinobacteria [5,6,7,8], including a series of anticancer, antifungal, antibacterial, antihelminthic, and immunosuppressive drugs [3].
Nocardiopsis is an important genus of actinobacterium for its extensive application in agriculture [9], industry [10], and environmental protection [11], especially for its potential ability to produce new natural products [5,12]. By mid-2021, 3% of marine actinomycetes-derived natural products were produced by Nocardiopsis, and Nocardiopsis are the third-largest actinomycetes in terms of producing marine compounds [13]. In addition, Nocardiopsis are a prolific source of bioactive natural products in both marine and terrestrial environments [1] and are widely distributed in multiple ecosystems [12], including deserts [14], deep ocean [15], coastal wetlands [16], and saline–alkali soil [17]. Nocardiopsis species have the ability to survive under different and hostile environmental conditions, mainly benefitting from their excretion of enzymes, multipurpose genetic constitution, and their ability to produce compuponible solutes and surfactants [12,18,19,20]. Besides these characteristics, the members of this genus have the ability to produce abundant bioactive compounds, which may allow them to prevail in different habitats [12]. Extracts from cultivated Nocardiopsis have exhibited cytotoxic [21,22], antimicrobial [23,24], antifibrotic [25], and anti-inflammatory [25] activities. Comprehensive analyses of Nocardiopsis metabolites by LC-HRES-MS, HRMS, or GC/MS have led to the identification of bioactive and structurally diverse compounds [21,26,27,28]. The bioactive secondary metabolites isolated from Nocardiopsis include antimicrobial agents [29,30,31], tumor promoters [32], cytotoxic compounds [33,34], kinase and P-glycoprotein inhibitors [35,36], immunoregulators [37], and natural products with other multiple bioactivities [38,39,40]. The secondary metabolites discovered from Nocardiopsis have shown a great diversity of structural frameworks, including polyketides [41,42,43], alkaloids [44,45], and terpenoids [46,47].
Bennur et al. reviewed the bioactive natural products derived from Nocardiopsis prior to 2015 [12], while Ibrahim et al. conducted a literature review regarding all of the secondary metabolites discovered from Nocardiopsis from 2016 to February 2018 [5]. In this review, the sources, distribution, bioactivities, biosynthesis, and structural characteristics of the compounds isolated from Nocardiopsis between March 2018 and 2021 are comprehensively summarized.

2. Polyketides

Three new angucyclines, nocardiopsistins A–C (13), were obtained from the deep-sea sponge-derived Nocardiopsis sp. strain HB-J378 (Figure 1) [48]. The antibacterial activities of compounds 13 were tested against MRSA (methicillin-resistant Staphylococcus aureus) and 13 exhibited antibacterial activity with MIC values of 3.12–12.5 μg/mL. Three core genes were identified through bioinformatic analysis of the sketch genome of the strain HB-J378 in a biosynthetic gene cluster encoding a typical aromatic or type II polyketide synthase (PKS) system, including acyl carrier protein (ACP), ketoacyl: ACP synthase α-subunit (KSα) and β-subunit (KSβ). The brief biosynthetic route for 13 was proposed according to the discovered oviedomycin pathway [49]. Compounds 13 were supposed to be biosynthesized by taking advantage of one molecule of isobutyral-CoA and nine molecules of malonyl-CoA, through complex enzymatic reactions to obtain the key angucycline biosynthetic intermediate UWM6, an analog of 13 [50], to acquire 13 (see Figure 1) [48].
The new strain Nocardiopsis sp. CG3 (DSM 106572), collected from the saltpan of Kenadsa (22 km west of Bechar, located in southwest Algeria), was discovered to have the ability to produce new bioactive natural products in a screening program [51]. Chemical investigation of the strain led to the isolation of five new polyene macrolactams, kenalactams A−E (48) (Figure 2) [51]. The biosynthetic pathway of polyketide kenalactam A (4) was studied by feeding experiments and was found to used L-alanine as the nitrogen-bearing starter unit. Compounds 68 exhibited cytotoxic activity against HeLa (cervical cancer cells KB3.1) and PC-3 (human prostate cancer) cell lines, with IC50 values ranging from 2.1 to 6.8 μM [51].
Eight new α-pyrone derivatives, nocahypyrones A–H (916), along with one known analog, germicidin G (17) (Figure 3) were isolated from the strains Nocardiopsis sp. HDN154-146 and HDN154-168, which were collected from soil samples derived from the Takla Makan desert area in Xinjiang Province, China. This was the first time it was reported that compounds 13 and 16 showed cytoprotective activity by inducing the expression of phase II detoxifying enzymes [52]. Aldo-keto reductase family1 member C1 (AKR1C1), human NAD(P)H: quinone oxidoreductase 1 (NQO1), superoxide dismutase 2 (SOD2), and heme oxygenase 1(HO-1), belonging to phase II detoxifying enzymes, have been illustrated to possess significant roles in defending mammalian cells against oxidative damage and excessive inflammatory reaction [53,54]. Compounds 13 and 16 displayed cytoprotective activity by inducing the expression level of SOD2 and HO-1 in HaCaT cells [52].
Chemical investigation of the actinobacterium Nocardiopsis sp. HDN 17-237 led to the isolation of one new β,γ-butenoate derivative, phenylbutenote (18), and one new α-pyrone, nocapyrone T (19) (Figure 3). The strain HDN 17-237 was collected from deep-sea water from the Mariana Trench (depth 4448 m, 10°21.100′ N, 142°17.574′ E, gathered in September 2016). Compounds 18 and 19 were evaluated for antibacterial and antioxidant activities, while neither of them exhibited obvious activity [55].
Comprehensive research of the secondary metabolites of an Antarctic marine animal sample-derived actinomycete, N. aegyptia HDN19-252, combined with a molecular networking approach in the Global Natural Products Social (GNPS) platform, obtained the isolation of four new anthraquinone derivatives, saliniquinones G−I (2022) and heraclemycin E (23) (Figure 3). Compounds 20 and 21 showed potent antibacterial activities against six evaluated bacterial strains—Bacillus subtilis, methicillin-resistant coagulase-negative staphylococci (MRCNS), B. cereus, Proteus sp., Mycobacterium phlei, and Escherichia coli—with MIC values of 3.1−12.5 µM [56].
Three new macrolides, borrelidins C−E, along with one known analog, borrelidin, which showed antibacterial and anticancer activities, were isolated from a saltern-derived halophilic Nocardiopsis sp. in 2017 [43]. These borrelidins were found to have the ability to relieve the amyloid-β induced toxicity in the HT22 cell line in 2021, indicating the potential of borrelidins to be developed as Alzheimer’s disease drugs [57].

3. Alkaloids

The approach of an optimized nitroso-based probe (dienophile probe), promoting the detection of compounds containing conjugated alkenes in crude broth extracts, was employed in the chemical investigation of a marine-derived Nocardiopsis sp. CNY-503 and gained the isolation of one new polyketide alkaloid, named nocarditriene (24), which contained an unprecedented epoxy-2,3,4,5-tetrahydropyridine structure (Figure 4) [58].
The structure of nocarbenzoxazole G (25), isolated from the marine-derived actinomycete N. lucentensis DSM 44048 in 2015 [60], was revised into the nocarbenzoxazole G (26) molecule by total synthesis in 2019. The benzoxazole skeleton was constructed with microwave assistance and continued by carbon–carbon bond formation with relevant aryl bromides [59]. Compound 26 was found to display moderate cytotoxicity against HepG2 and HeLa cell lines with IC50 values of 16 and 14 μM, respectively [60].
Chemical investigation of the strain N. flavescens NA01583, which was gained from marine sediment gathered at the coast near Hainan Island in 2016 through the genome mining of an indolocarbazole-type gene cluster, led to the isolation of three new indolocarbazole alkaloids, named loonamycins A−C (2729) (Figure 5) [61].
Compound 29 was produced successfully by the allogenetic expression of the complete loo gene cluster in a vicarious host, Streptomyces lividans K4−114. The indolocarbazole skeleton of 2729, belonging to the family of indolocarbazole alkaloids, is structurally similar to that of rebeccamycin and staurosporine with an additional rare modified tryptophan ring. The molecular bases of these modifications were investigated by carefully analyzing loo BGC for further genome mining and combinational biosynthetic research. The loo gene cluster sustains a ∼36 kb continuous DNA sequence, including 25 open reading frames which take charge of biosynthesis, regulation, and resistance (Figure 6A). A possible biosynthetic pathway for loonamycin was detected based on this bioinformatic analysis (Figure 6B). In particular, compound 27 showed potent cytotoxic activities toward eight cancer cell lines, including Sum1315 (breast cancer), SH-SY5Y (neuroblastoma), HCT116 (colorectal cancer), HT29 (colorectal cancer), HCC78 (lung cancer), HeLa (cervical cancer), SW620 (colorectal cancer), and SW872 (liposarcoma), with IC50 values of 41−283 nM [61].
The broth culture crude extracts of actinobacterium Nocardiopsis sp. SCA30, derived from marine sediments collected from Havelock Island, Andaman, and the Nicobar Islands, India (11.96° N, 93.00° E), displayed cytotoxic activities against a series of cell lines, including HT 29, HCT 15, MDA-MB 468, and MCF 7 at concentrations ranging from 62.5 to 1000 µg/mL. The strain extracts also showed antibacterial activities against MRSA ATCC NR-46171 and NR-46071 with MIC values of 7.81 and 15.62 µg/mL, respectively. Compound 1-acetyl-4-4(hydroxyphenyl)piperazine (30) (Figure 5) was isolated from the crude extracts of Nocardiopsis sp. SCA30 through LC-MS analysis and NMR chemical structural identification approved to be an antibacterial and cytotoxic compound [22].
Chemical investigation of actinomycete N. dassonvillei SCSIO 40065 derived from the marine sponge Petrosia sp., which was collected on the seabed near Yongxing Island in the South China Sea at a depth of 20 m, led to the isolation of two polycyclic thioalkaloides, dassonmycins A (31) and B (32) (Figure 5). The new isolated compounds 31 and 32 contained the skeleton of a 6/6/6/6-fused tetracyclic ring featuring a naphthoquinone [2,3-e] piperazine-[1,2-c] thiomorpholine. Both compounds exhibited antibacterial activities against Micrococcus luteus SCSIO ML01, B. subtilis 1064, MRSA shhsA1, and S. aureus ATCC 29213 with MICs of 8−64 μg/mL. Compound 31 was found to display weak inhibited growth of Vibrio alginolyticus ATCC 13214 and Enterococcus faecalis ATCC 29212, with MIC values of 32 μg/mL. Compounds 31 and 32 exhibited moderate cytotoxicity against four human cancer cell lines—HepG-2, SF-268, MCF-7, and A549—with IC50 values of 12−34 μM [62]. Biosynthetically, 31 and 32 are proposed to be biosynthesized by a non-ribosomal peptide synthetase (NRPS) route combined with a chorismate pathway (Figure 7) [62].
A marine sediment-derived actinobacterium N. dassonvillei JS106 showed potent antiquorum sensing activities against S. aureus and Pseudomonas aeruginosa [63]. The marine sediment sample was gathered from Lianyungang, China. Secondary metabolites research of the strain JS106 led to the isolation of one new compound, 2-hydroxyacetate-3-hydroxyacetamido-phenoxazine (HHP, 33), and one known analog questiomycin A (34) (Figure 5). Both of these two compounds (33 and 34) exhibited antibiofilm activity against Chromobacterium violaceum 12472 with IC50 values of 23.59 and 6.82 μg/mL, respectively [63].

4. Peptides

The secondary metabolites research of the broth culture of Nocardiopsis sp. UR67A, derived from the marine sponge Callyspongia sp. collected from the Red Sea in 2018, resulted in the purification of one new cyclic hexapeptide, nocardiotide A (35) (Figure 8) [64]. Compound 35 was synthesized by combining solid-and solution-phase synthesis methods in 2021 [65]. Compound 35 exhibited obvious cytotoxic activities against HeLa, CT26 (murine colon carcinoma), and MM.1S (human multiple myeloma) cell lines with IC50 values ranging from 8 to 12 µM [64].
One new diketopiperazine, 1-demethylnocazine A (36) (Figure 8), was separated from the broth culture of the actinomycete Nocardiopsis sp. TRM20105, which was derived from a local cotton field in Tarim Basin, through the approach of antifungal activity tracking purification against Candida albicans. Compound 36 displayed weak antifungal activity against C. albicans, with an MIC value of 3.16 mM [66].
One new cyclic tetrapeptide, androsamide (37), was isolated from the actinobacterium Nocardiopsis sp. CNT-189 was gathered from the surf zone sediment of the Bahamas shore (Figure 8) [67]. Androsamide (37) displayed moderate cytotoxicity against Caco2 (human colorectal adenocarcinoma), AGS (human gastric adenocarcinoma), and HCT116 (human colorectal carcinoma) cell lines with the IC50 values of 13 μM, 18 μM and 21 μM, respectively. Compound 37 exhibited significant inhibition to the motility of the Caco2 cell line caused by epithelial−mesenchymal transition (EMT) [67]. The expression of cell motility-related genes in Caco2 cells was also examined through Human Cell Motility RT2 Profiler PCR Array. Ras-related C3 botulinum toxin substrate 2 (RAC2) and calpain 1 (CAPN1) have been proved to be the human cell motility-related genes [69,70,71]. The RT2 Profiler PCR Array results showed that 37 with the significant inhibition of the expression of RAC2 in nontoxic concentration 0.65 μM (1/20 of IC50), and decreased the expression of CAPN1 by 1.3 μM (1/10 of IC50) in Caco2 cells [67].
Secondary metabolites of Nocardiopsis sp. HT88, which was separated from the fresh stems of Mallotus nudiflorus L. (plant), was comprehensively studied for its crude extracts displayed significant antibacterial activities. Eight proline (or hydroxyproline, Hyp)-containing cyclic dipeptides: cyclo(L-Pro-L-Leu) (38), cyclo(Pro-Leu) (39), cyclo(L-trans-Hyp-L-Leu) (40), cyclo(D-trans-Hyp-D-Leu) (41), cyclo(D-Pro-L-Phe) (42), cyclo(L-Pro-L-Phe) (43), and cyclo(D-cis-Hyp-L-Phe) (44), and cyclo(L-trans-Hyp-L-Phe) (45) (Figure 8) were isolated from the broth culture of HT88 guided by antibacterial bioassay. Unfortunately, none of the purified DKPs (3845) exhibited antibacterial activity against measured strains at 100 μg/disc [68].
Some of the peptides are hydrolyzed fragments of a precursor protein, called ribosomal posttranslational peptides (RiPPs) [72], and others are biosynthesized by non-ribosomal peptide synthetase (NRPS) assembly lines (Figure 9) [73,74].
There is no research on the biosynthesis of compounds 35 and 37. The cyclodipeptides’ (CDP) core structures are biosynthesized through NRPS or tRNA-dependent CDP synthase (CDPS) (Figure 10) pathways [75,76].
Compound 36 is belonging to the family of nocazine and is proposed to be synthesized through a CDPS-dependent pathway (Figure 11) [76]. Compounds 3845 are modified CDP and are also deduced to be synthesized by the CDPS assembly line.

5. Terphenyls

Chemical investigation of the actinobacterium strain Nocardiopsis sp. OUCMDZ-4936, purified from a sample gathered from Dongzhaigang Mangrove Reserve, Hainan Province, China, led to the isolation of three new p-terphenyl derivatives, nocarterphenyls A−C (4648), together with three known analogs (4951) (Figure 12) [77]. Compound 46 exhibited significant cytotoxicity against the HL60 and HCC1954 cancer cell lines, with IC50 values of 0.38 ± 0.01 and 0.10 ± 0.01 μM, respectively. Compounds 49 and 51 showed potent cytotoxic activities against K562 and A549 cancer cell lines with IC50 values ranging from 0.10 to 0.77 μM. Compound 51 also exhibited cytotoxicity against MV4-11 and MDA-MB-468 cancer cell lines, with IC50 values of 0.77 ± 0.02 and 0.67 ± 0.01 μM, respectively [77].
Five new p-terphenyl derivatives—called nocarterphenyls D−H (5256) (Figure 12)—were gained and identified from the broth culture of the actinobacterium Nocardiopsis sp. HDN154086, separated from a marine sediment sample gathered from the South China Sea. It is the first time to find a 2,2′-bithiazole scaffold in natural products. p-Terphenylquinones 53 and 54 contain the unusual substitutions of thioether-linked fatty acid methyl ester. Compounds 52 and 53 demonstrated potent activities against seven kinds of bacteria—E. coli, Proteus sp., M. phlei, V. parahemolyticus, B. subtilis, B. cereus, and MRSA—with MIC values of 1.5−12 μM. Notably, compound 53 displayed significant antibacterial activity against MRSA, exceeding the positive control ciprofloxacin [78].

6. Others

Three known compounds, tryptophan (57), kynurenic acid (58), and 4-amino-3-methoxy benzoic acid (59) (Figure 13), were isolated from the actinobacterium Nocardiopsis sp. UR67, which was derived from the marine sponge Callyspongia sp. gained at a depth of 10 m from the Red Sea [64].
One new natural product phenolic acid derivative, 4-amino-6-methylsalicylic acid (60), along with two known compounds, 5-methylresorcinol (61) and linoleic acid (62), were isolated from the Nocardiopsis sp. AS23C strain, which was purified from the marine alga Sargassum arnaudianum derived from the Red Sea at the Hurghada coast, Egypt (Figure 13). The crude extracts of the strain AS23C displayed antibacterial activities against Gram-positive B. subtilis ATCC6051, S. viridochromogenes Tü 57, and S. aureus. Neither of the compounds 60 and 61 displayed cytotoxicity against KB-3-1 (human cervix carcinoma) cell line [79].
A rare actinomycete Nocardiopsis sp. SCA21, deriving from a marine sediment sample gathered from Havelock Island, Andaman, and the Nicobar Islands, India, exhibited the potential ability to produce bioactive secondary metabolites. Chemical investigation of the broth culture of the strain resulted in the purification of two known bioactive compounds—a bromophenol derivative, 4-bromophenol (63), and a phthalate ester, Bis (2-ethylhexyl) phthalate (64) (Figure 13). Compounds 63 and 64 displayed notable enzyme-inhibitory activities against α-glucosidase. However, there was no inhibited activity against α-amylase of compound 64. Both compounds 63 and 64 demonstrated potent free radical scavenging activities against DPPH and ABTS radicals. Additionally, 63 and 64 also revealed broad-spectrum inhibitory activities against MRSA ATCC-46071, MRSA ATCC NR-46171, S. aureus ATCC 12600, B. subtilis ATCC 6633, and Klebsiella pneumonia ATCC 13883 [80].
A novel sterol (65) with an unidentified substitution was isolated from marine actinomycete N. alba MCCB 110. Compound 65 was found to display antibacterial activity against the aquaculture pathogen V. harveyi, and did not exhibit toxicities to the VERO cell line and shrimp hemocytes, up to 1000 ppm [81].

7. Conclusions

From March 2018 to 2021, a total of 63 natural products have been isolated from the genus Nocardiopsis, and 67% of them are first-discovered compounds. These findings sufficiently demonstrate that actinomycetes Nocardiopsis have great potential to produce compounds with novel structures. The structures of the isolated compounds with diverse skeletons are mainly concentrated on the classes of polyketides, peptides, terphenyls, and alkaloids (Table 1 and Figure 14).
The sources of the genus Nocardiopsis are distributed throughout diverse ecological systems, including desert, marine, mangrove, and saltpan areas (Figure 15). More than two out of three natural products were isolated from marine-derived Nocardiopsis, demonstrating that the ocean is a vast treasure house with abundant microbial sources to produce various novel natural products. From March 2018 to 2021, the percentage of the marine-derived compounds increased by 10%, compared with the period 2015−February 2018 [5], which further demonstrates that more and more attention has been concentrated on the development of the marine resources. Among the marine-derived natural products, the sources of Nocardiopsis strains are mainly collected from marine sediment and sponges (Figure 15).
The genus Nocardiopsis has the potential ability to produce a great diversity of bioactive secondary metabolites, including antibacterial, cytotoxic, cytoprotective, enzyme inhibitory, free radical scavenging, antiquorum sensing, colorectal cancer motility inhibitory, and antifungal compounds (Figure 16). Almost 50% of the Nocardiopsis-derived natural products have been discovered to exhibit various bioactivities. The activities of the bioactive compounds are mainly focused on the categories of antibacterial and cytotoxic activities (Figure 16), which serves as a reminder that there is huge potential for new antibiotics and anticancer compounds to be developed from Nocardiopsis.
There were 13 antibacterial natural products isolated from Nocardiopsis in the period March 2018 to 2021 (Table 2). Among them, compounds 2, 30, and 53 exhibit strong antibacterial activity against MRSA, and compounds 20 and 21 display significant antibacterial activity against MRCNS (Table 2). These five compounds might be developed into new antibiotics to respond to the challenge of increasing antibacterial resistance. Compound 52 shows significant antibacterial activities against a series of strains, which has the potential to be exploited into novel antibiotics with a broad spectrum of antibacterial activity (Table 2). Compound 65 with antibacterial activity displays no toxic against VERO cell line and shrimp hemocytes up to 1000 ppm, could be developed as an environmentally friendly antibiotic (Table 2). All of the isolated antibacterial secondary metabolites provide the structural basis for novel antibiotic research.
There were 13 cytotoxic natural products isolated from Nocardiopsis in the period March 2018 to 2021 (Table 3). Compound 27 shows extremely significant and broad-spectrum cytotoxicity against a series of cell lines with the IC50 values at the nM level, and compounds 46 and 4951 display potent and broad-spectrum cytotoxicity (Table 3). All four compounds have the potential to be developed as new anticancer drugs. Compounds 8, 31, 32, 35, and 37 with moderate cytotoxicity provide structural inspiration for the research and development of new anticancer drugs (Table 3).
Eight secondary metabolites isolated from Nocardiopsis have various bioactivities, including cytoprotective, antiquorum sensing, colorectal cancer motility and enzyme inhibitory, and free radical scavenging activities (Table 3).
Although the Nocardiopsis-derived natural products exhibit excellent diverse bioactivities, further research on their bioactive mechanisms is deficient. Only three compounds, 13, 16, and 37, have been studied for their active mechanisms between March 2018 and 2021. To the best of our knowledge, all the activity tests were evaluated in vitro, and bioactive evaluations in vivo are also lacking between March 2018 and 2021. In terms of the current situation, it is still a long way from the drugs being approved for clinical use.
In this review, we summarized the secondary metabolites isolated from Nocardiopsis in the period between March 2018 and 2021. The literature survey comprehensively indicates that actinomycetes Nocardiopsis have great potential as producers to generate abundant and diverse novel bioactive secondary metabolites. Some potent antibacterial and cytotoxic compounds isolated from Nocardiopsis have the potential to be developed into new drugs. Additionally, all the natural products isolated from Nocardiopsis provide a structural foundation for drug design.

Author Contributions

T.S. collected the literature regarding natural products isolated from Nocardiopsis, and wrote the paper; Y.-F.W. and H.W. revised the manuscript; B.W. organized and guided the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (No. 82104029, 21868011); the Shandong Provincial Natural Science Foundation (No. ZR2020QD111), and supported by the Talent Support Program of Shandong University of Science and Technology.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jose, P.A.; Maharshi, A.; Jha, B. Actinobacteria in natural products research: Progress and prospects. Microbiol. Res. 2021, 246, 126708. [Google Scholar] [CrossRef] [PubMed]
  2. Barka, E.A.; Vatsa, P.; Sanchez, L.; Gaveau-Vaillant, N.; Jacquard, C.; Clement, C.; Meier-Kolthoff, J.P.; Klenk, H.-P.; Ouhdouch, Y.; van Wezel, G.P. Taxonomy, physiology, and natural products of actinobacteria. Microbiol. Mol. Biol. Rev. 2016, 80, 1–43. [Google Scholar] [CrossRef] [Green Version]
  3. Demain, A.L.; Sanchez, S. Microbial drug discovery: 80 years of progress. J. Antibiot. 2009, 62, 5–16. [Google Scholar] [CrossRef] [Green Version]
  4. Mahajan, G.B.; Balachandran, L. Antibacterial agents from actinomycetes—A review. Front. Biosci. Elite Ed. 2012, E4, 240–253. [Google Scholar] [CrossRef]
  5. Ibrahim, A.H.; Desoukey, S.Y.; Fouad, M.A.; Abdelmohsen, U.R.; Kamel, M.S.; Gulder, T.A.M. Natural product potential of the genus Nocardiopsis. Mar. Drugs 2018, 16, 147. [Google Scholar] [CrossRef] [PubMed]
  6. Li, J.W.H.; Vederas, J.C. Drug discovery and natural products: End of an era or an endless frontier? Science 2009, 325, 161–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Manivasagan, P.; Kang, K.-H.; Sivakumar, K.; Li-Chan, E.C.Y.; Oh, H.-M.; Kim, S.-K. Marine actinobacteria: An important source of bioactive natural products. Environ. Toxicol. Pharmacol. 2014, 38, 172–188. [Google Scholar] [CrossRef] [PubMed]
  8. Manivasagan, P.; Venkatesan, J.; Sivakumar, K.; Kim, S.-K. Pharmaceutically active secondary metabolites of marine actinobacteria. Microbiol. Res. 2014, 169, 262–278. [Google Scholar] [CrossRef]
  9. AbdElgawad, H.; Zinta, G.; Abuelsoud, W.; Hassan, Y.M.; Alkhalifah, D.H.M.; Hozzein, W.N.; Zrieq, R.; Beemster, G.T.; Schoenaers, S. An actinomycete strain of Nocardiopsis lucentensis reduces arsenic toxicity in barley and maize. J. Hazard. Mater. 2021, 417, 126055. [Google Scholar] [CrossRef] [PubMed]
  10. Adenan, N.H.; Lim, Y.Y.; Ting, A.S.Y. Nocardiopsis sp. for the removal of triphenylmethane dyes: Decolorization and optimization studies. Water Air Soil Pollut. 2021, 232, 414. [Google Scholar] [CrossRef]
  11. Patel, G.B.; Rakholiya, P.; Shindhal, T.; Varjani, S.; Tabhani, N.; Shah, K.R. Lipolytic Nocardiopsis for reduction of pollution load in textile industry effluent and SWISS model for structural study of lipase. Bioresour. Technol. 2021, 341, 125673. [Google Scholar] [CrossRef] [PubMed]
  12. Bennur, T.; Ravi Kumar, A.; Zinjarde, S.S.; Javdekar, V. Nocardiopsis species: A potential source of bioactive compounds. J. Appl. Microbiol. 2016, 120, 1–16. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, J.; Xu, L.; Zhou, Y.; Han, B. Natural products from actinomycetes associated with marine organisms. Mar. Drugs 2021, 19, 629. [Google Scholar] [CrossRef] [PubMed]
  14. Kumar, S.; Solanki, D.S.; Parihar, K.; Tak, A.; Gehlot, P.; Pathak, R.; Singh, S.K. Actinomycetes isolates of arid zone of Indian Thar Desert and efficacy of their bioactive compounds against human pathogenic bacteria. Biol. Futur. 2021, 72, 431–440. [Google Scholar] [CrossRef]
  15. Pinto-Almeida, A.; Bauermeister, A.; Luppino, L.; Grilo, I.R.; Oliveira, J.; Sousa, J.R.; Petras, D.; Rodrigues, C.F.; Prieto-Davo, A.; Tasdemir, D.; et al. The diversity, metabolomics profiling, and the pharmacological potential of actinomycetes isolated from the Estremadura Spur Pockmarks (Portugal). Mar. Drugs 2022, 20, 21. [Google Scholar] [CrossRef]
  16. Chen, L.; Wang, Z.; Du, S.; Wang, G. Antimicrobial activity and functional genes of actinobacteria from coastal wetland. Curr. Microbiol. 2021, 78, 3058–3067. [Google Scholar] [CrossRef]
  17. Tatar, D. Isolation, phylogenetic analysis and antimicrobial activity of halophilic actinomycetes from different saline environments located near Corum province. Biologia 2021, 76, 773–780. [Google Scholar] [CrossRef]
  18. Li, H.-W.; Zhi, X.-Y.; Yao, J.-C.; Zhou, Y.; Tang, S.-K.; Klenk, H.-P.; Zhao, J.; Li, W.-J. Comparative genomic analysis of the genus Nocardiopsis provides new insights into its genetic mechanisms of environmental adaptability. PLoS ONE 2013, 8, e61528. [Google Scholar]
  19. Bennur, T.; Kumar, A.R.; Zinjarde, S.; Javdekar, V. Nocardiopsis species as potential sources of diverse and novel extracellular enzymes. Appl. Microbiol. Biotechnol. 2014, 98, 9173–9185. [Google Scholar]
  20. Bennur, T.; Kumar, A.R.; Zinjarde, S.; Javdekar, V. Nocardiopsis species: Incidence, ecological roles and adaptations. Microbiol. Res. 2015, 174, 33–47. [Google Scholar]
  21. Shady, N.H.; Tawfike, A.F.; Yahia, R.; Fouad, M.A.; Brachmann, A.O.; Piel, J.; Abdelmohsen, U.R.; Kamel, M.S. Cytotoxic activity of actinomycetes Nocardia sp. and Nocardiopsis sp. associated with marine sponge Amphimedon sp. Nat. Prod. Res. 2021, 1–6. [Google Scholar] [CrossRef] [PubMed]
  22. Siddharth, S.; Aswathanarayan, J.B.; Kuruburu, M.G.; Madhunapantula, S.R.V.; Vittal, R.R. Diketopiperazine derivative from marine actinomycetes Nocardiopsis sp. SCA30 with antimicrobial activity against MRSA. Arch. Microbiol. 2021, 203, 6173–6181. [Google Scholar] [CrossRef]
  23. Trivedi, N.; Thumar, J. Chemical profiling of antimicrobial metabolites from halophilic actinomycete Nocardiopsis sp. Al-H10-1 (KF384482) isolated from Alang, Gulf of Khambhat, India. bioRxiv 2021. [Google Scholar] [CrossRef]
  24. Goel, N.; Fatima, S.W.; Kumar, S.; Sinha, R.; Khare, S.K. Antimicrobial resistance in biofilms: Exploring marine actinobacteria as a potential source of antibiotics and biofilm inhibitors. Biotechnol. Rep. 2021, 30, e00613. [Google Scholar] [CrossRef] [PubMed]
  25. Choi, G.; Kim, G.J.; Choi, H.; Choi, I.-W.; Lee, D.-S. Anti-inflammatory and anti-fibrotic activities of Nocardiopsis sp. 13G027 in lipopolysaccharides-induced RAW 264.7 macrophages and transforming growth factor beta-1-stimulated nasal polyp-derived fibroblasts. Microbiol. Biotechnol. Lett. 2021, 49, 543–551. [Google Scholar] [CrossRef]
  26. Sarmiento-Vizcaíno, A.; Martín, J.; Reyes, F.; García, L.A.; Blanco, G. Bioactive natural products in actinobacteria isolated in rainwater from storm clouds transported by western winds in Spain. Front. Microbiol. 2021, 12, 773095. [Google Scholar] [CrossRef]
  27. Gamaleldin, N.M.; Bakeer, W.; El-Gendy, A.O.; Sayed, A.M.; Shamikh, Y.I.; Hassan, H.M.; Horn, H.; Abdelmohsen, U.R.; Hozzein, W.N. Exploration of chemical diversity and antitrypanosomal activity of some Red Sea-derived actinomycetes using the OSMAC approach supported by LC-MS-based metabolomics and molecular modelling. Antibiotics 2020, 9, 629. [Google Scholar] [CrossRef]
  28. Widada, J.; Damayanti, E.; Alhakim, M.R.; Yuwono, T.; Mustofa, M. Two strains of airborne Nocardiopsis alba producing different volatile organic compounds (VOCs) as biofungicide for Ganoderma boninense. FEMS Microbiol. Lett. 2021, 368, fnab138. [Google Scholar] [CrossRef]
  29. Wang, Z.; Fu, P.; Liu, P.; Wang, P.; Hou, J.; Li, W.; Zhu, W. New pyran-2-ones from alkalophilic actinomycete, Nocardiopsis alkaliphila sp. nov. YIM-80379. Chem. Biodivers. 2013, 10, 281–287. [Google Scholar] [CrossRef]
  30. Lu, C.; Li, Y.; Wang, H.; Wang, B.; Shen, Y. A new phenoxazine derivative isolated from marine sediment actinomycetes, Nocardiopsis sp. 236. Drug Discov. Ther. 2013, 7, 101–104. [Google Scholar]
  31. Tian, S.; Yang, Y.; Liu, K.; Xiong, Z.; Xu, L.; Zhao, L. Antimicrobial metabolites from a novel halophilic actinomycete Nocardiopsis terrae YIM 90022. Nat. Prod. Res. 2014, 28, 344–346. [Google Scholar] [CrossRef] [PubMed]
  32. Yamashita, T.; Imoto, M.; Isshiki, K.; Sawa, T.; Naganawa, H.; Kurasawa, S.; Zhu, B.; Umezawa, K. Isolation of a new indole alkaloid, pendolmycin, from Nocardiopsis. J. Nat. Prod. 1988, 51, 1184–1187. [Google Scholar] [CrossRef]
  33. Lin, Z.; Torres, J.P.; Ammon, M.A.; Marett, L.; Teichert, R.W.; Reilly, C.A.; Kwan, J.C.; Hughen, R.W.; Flores, M.; Tianero, M.D.; et al. A Bacterial source for mollusk pyrone polyketides. Chem. Biol. 2013, 20, 73–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gao, X.; Lu, Y.; Xing, Y.; Ma, Y.; Lu, J.; Bao, W.; Wang, Y.; Xi, T. A novel anticancer and antifungus phenazine derivative from a marine actinomycete BM-17. Microbiol. Res. 2012, 167, 616–622. [Google Scholar] [CrossRef]
  35. Kase, H.; Iwahashi, K.; Matsuda, Y. K-252a, a potent inhibitor of protein kinase C from microbial origin. J. Antibiot. 1986, 39, 1059–1065. [Google Scholar] [CrossRef] [Green Version]
  36. Raju, R.; Piggott, A.M.; Huang, X.-C.; Capon, R.J. Nocardioazines: A novel bridged diketopiperazine scaffold from a marine-derived bacterium inhibits P-glycoprotein. Org. Lett. 2011, 13, 2770–2773. [Google Scholar] [CrossRef]
  37. Kim, M.C.; Kwon, O.-W.; Park, J.-S.; Kim, S.Y.; Kwon, H.C. Nocapyrones H–J, 3,6-disubstituted α-pyrones from the marine actinomycete Nocardiopsis sp. KMF-001. Chem. Pharm. Bull. 2013, 61, 511–515. [Google Scholar] [CrossRef] [Green Version]
  38. Wu, Z.; Xie, L.; Xia, G.; Zhang, J.; Nie, Y.; Hu, J.; Wang, S.; Zhang, R. A new tetrodotoxin-producing actinomycete, Nocardiopsis dassonvillei, isolated from the ovaries of puffer fish Fugu rubripes. Toxicon 2005, 45, 851–859. [Google Scholar] [CrossRef]
  39. Peltola, J.S.P.; Andersson, M.A.; Kampfer, P.; Auling, G.; Kroppenstedt, R.M.; Busse, H.-J.; Salkinoja-Salonen, M.S.; Rainey, F.A. Isolation of toxigenic Nocardiopsis strains from indoor environments and description of two new Nocardiopsis species, N. exhalans sp. nov. and N. umidischolae sp. nov. Appl. Environ. Microbiol. 2001, 67, 4293–4304. [Google Scholar] [CrossRef] [Green Version]
  40. Kim, Y.; Ogura, H.; Akasaka, K.; Oikawa, T.; Matsuura, N.; Imada, C.; Yasuda, H.; Igarashi, Y. Nocapyrones: α- and γ-pyrones from a marine-derived Nocardiopsis sp. Mar. Drugs 2014, 12, 4110–4125. [Google Scholar] [CrossRef] [Green Version]
  41. Sun, M.-W.; Guo, Z.-X.; Lu, C.-H. Two new polyketides from Nocardiopsis lucentensis DSM 44048. Nat. Prod. Res. 2016, 30, 1036–1041. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, H.; Saurav, K.; Yu, Z.; Mandi, A.; Kurtan, T.; Li, J.; Tian, X.; Zhang, Q.; Zhang, W.; Zhang, C. α-Pyrones with diverse hydroxy substitutions from three marine-derived Nocardiopsis strains. J. Nat. Prod. 2016, 79, 1610–1618. [Google Scholar] [CrossRef] [PubMed]
  43. Kim, J.; Shin, D.; Kim, S.-H.; Park, W.; Shin, Y.; Kim, W.K.; Lee, S.K.; Oh, K.-B.; Shin, J.; Oh, D.-C. Borrelidins C.–E: New antibacterial macrolides from a saltern-derived halophilic Nocardiopsis sp. Mar. Drugs 2017, 15, 166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhang, X.; He, H.; Ma, R.; Ji, Z.; Wei, Q.; Dai, H.; Zhang, L.; Song, F. Madurastatin B3, a rare aziridine derivative from actinomycete Nocardiopsis sp. LS150010 with potent anti-tuberculosis activity. J. Ind. Microbiol. Biotechnol. 2017, 44, 589–594. [Google Scholar] [CrossRef]
  45. Eliwa, E.M.; Abdel-Razek, A.S.; Frese, M.; Wibberg, D.; Halawa, A.H.; El-Agrody, A.M.; Bedair, A.H.; Kalinowski, J.; Sewald, N.; Shaaban, M. New bioactive compounds from the marine-derived actinomycete Nocardiopsis lucentensis sp. ASMR2. Z. Naturforsch. B J. Chem. Sci. 2017, 72, 351–360. [Google Scholar] [CrossRef]
  46. Sun, M.-W.; Zhang, X.-M.; Bi, H.-L.; Li, W.-J.; Lu, C.-H. Two new sesquiterpenoids produced by halophilic Nocardiopsis chromatogenes YIM 90109. Nat. Prod. Res. 2017, 31, 77–83. [Google Scholar] [CrossRef] [PubMed]
  47. Hamed, A.; Abdel-Razek, A.S.; Frese, M.; Stammler, H.G.; El-Haddad, A.F.; Ibrahim, T.M.A.; Sewald, N.; Shaaban, M. Terretonin N: A new meroterpenoid from Nocardiopsis sp. Molecules 2018, 23, 299. [Google Scholar] [CrossRef] [Green Version]
  48. Xu, D.; Nepal, K.K.; Harmody, D.; McCarthy, P.J.; Wright, A.E.; Wang, G.; Chen, J.; Zhu, H. Nocardiopsistins A–C: New angucyclines with anti-MRSA activity isolated from a marine sponge-derived Nocardiopsis sp. HB-J378. Synth. Syst. Biotechnol. 2018, 3, 246–251. [Google Scholar] [CrossRef]
  49. Lombo, F.; Brana, A.F.; Salas, J.A.; Mendez, C. Genetic organization of the biosynthetic gene cluster for the antitumor angucycline oviedomycin in Streptomyces antibioticus ATCC 11891. ChemBioChem 2004, 5, 1181–1187. [Google Scholar] [CrossRef] [PubMed]
  50. Kharel, M.K.; Pahari, P.; Shepherd, M.D.; Tibrewal, N.; Nybo, S.E.; Shaaban, K.A.; Rohr, J. Angucyclines: Biosynthesis, mode-of-action, new natural products, and synthesis. Nat. Prod. Rep. 2012, 29, 264–325. [Google Scholar] [CrossRef]
  51. Messaoudi, O.; Sudarman, E.; Bendahou, M.; Jansen, R.; Stadler, M.; Wink, J. Kenalactams A–E, polyene macrolactams isolated from Nocardiopsis CG3. J. Nat. Prod. 2019, 82, 1081–1088. [Google Scholar] [CrossRef]
  52. Zhao, T.; Chang, Y.; Zhu, T.; Li, J.; Gu, Q.; Li, D.; Che, Q.; Zhang, G. α-Pyrone derivatives with cyto-protective activity from two Takla Makan desert soil derived actinomycete Nocardiopsis strains recovered in seawater based medium. Nat. Prod. Res. 2019, 33, 2498–2506. [Google Scholar] [CrossRef] [PubMed]
  53. Bolling, B.W.; Parkin, K.L. Limited contribution of isoflavones to hepatocellular phase II enzyme-inducing activity of soybean (Glycine max) extracts. Food Chem. 2009, 113, 1069–1075. [Google Scholar] [CrossRef]
  54. Park, J.S.; Jung, J.S.; Jeong, Y.H.; Hyun, J.W.; Le, T.K.V.; Kim, D.H.; Choi, E.C.; Kim, H.S. Antioxidant mechanism of isoflavone metabolites in hydrogen peroxide-stimulated rat primary astrocytes: Critical role of hemeoxygenase-1 and NQO1 expression. J. Neurochem. 2011, 119, 909–919. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, J.-X.; Sun, C.-X.; Shah, M.; Zhang, G.-J.; Gu, Q.-Q.; Zhu, T.-J.; Che, Q.; Li, D.-H. New metabolites from a Mariana Trench-derived actinomycete Nocardiopsis sp. HDN 17-237. J. Asian Nat. Prod. Res. 2020, 22, 1031–1036. [Google Scholar] [CrossRef] [PubMed]
  56. Zhou, L.; Chen, X.; Sun, C.; Chang, Y.; Huang, X.; Zhu, T.; Zhang, G.; Che, Q.; Li, D. Saliniquinone derivatives, saliniquinones G−I and heraclemycin E, from the marine animal-derived Nocardiopsis aegyptia HDN19-252. Mar. Drugs 2021, 19, 575. [Google Scholar] [CrossRef] [PubMed]
  57. Shin, J.; Yang, S.-H.; Du, Y.E.; Park, K.; Kim, D.; Shin, D.; Kim, J.; Kim, S.-H.; Kim, Y.K.; Shin, J. Borrelidin from saltern-derived halophilic Nocardiopsis sp. dissociates amyloid-β and tau fibrils. J. Alzheimer’s Dis. Rep. 2021, 5, 7–13. [Google Scholar] [CrossRef]
  58. Castro-Falcon, G.; Millan-Aguinaga, N.; Roullier, C.; Jensen, P.R.; Hughes, C.C. Nitrosopyridine probe to detect polyketide natural products with conjugated alkenes: Discovery of novodaryamide and nocarditriene. ACS Chem. Biol. 2018, 13, 3097–3106. [Google Scholar] [CrossRef]
  59. Kim, T.; Lee, S.-A.; Noh, T.; Choi, P.; Choi, S.-J.; Song, B.G.; Kim, Y.; Park, Y.-T.; Huh, G.; Kim, Y.-J.; et al. Synthesis, structure revision, and cytotoxicity of nocarbenzoxazole G. J. Nat. Prod. 2019, 82, 1325–1330. [Google Scholar] [CrossRef] [PubMed]
  60. Sun, M.; Zhang, X.; Hao, H.; Li, W.; Lu, C. Nocarbenzoxazoles A–G, benzoxazoles produced by halophilic Nocardiopsis lucentensis DSM 44048. J. Nat. Prod. 2015, 78, 2123–2127. [Google Scholar] [CrossRef]
  61. Yang, C.L.; Zhang, B.; Xue, W.W.; Li, W.; Xu, Z.F.; Shi, J.; Shen, Y.; Jiao, R.H.; Tan, R.X.; Ge, H.M. Discovery, biosynthesis, and heterologous production of loonamycin, a potent anticancer indolocarbazole alkaloid. Org. Lett. 2020, 22, 4665–4669. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, X.; Chen, S.; Zhang, L.; Zhang, Q.; Zhang, W.; Chen, Y.; Zhang, W.; Zhang, H.; Zhang, C. Dassonmycins A and B, polycyclic thioalkaloids from a marine sponge-derived Nocardiopsis dassonvillei SCSIO 40065. Org. Lett. 2021, 23, 2858–2862. [Google Scholar] [CrossRef]
  63. Miao, L.; Qian, S.; Qi, S.; Jiang, W.; Dong, K. Culture medium optimization and active compounds investigation of an anti-quorum sensing marine actinobacterium Nocardiopsis dassonvillei JS106. Microbiology 2021, 90, 112–123. [Google Scholar] [CrossRef]
  64. Ibrahim, A.H.; Attia, E.Z.; Desoukey, S.Y.; Fouad, M.A.; Abdelmohsen, U.R.; Hajjar, D.; Anany, M.A.; Wajant, H.; Kamel, M.S.; Gulder, T.A.M. New cytotoxic cyclic peptide from the marine sponge-associated Nocardiopsis sp. UR67. Mar. Drugs 2018, 16, 290. [Google Scholar] [CrossRef] [Green Version]
  65. Muhajir, M.I.; Hardianto, A.; Al-Anshori, J.; Sumiarsa, D.; Mayanti, T.; Harneti, D.; Hidayat, A.T.; Supratman, U.; Maharani, R. Total synthesis of nocardiotide A by using a combination of solid-and solution-phase methods. ChemistrySelect 2021, 6, 12941–12946. [Google Scholar] [CrossRef]
  66. Chen, H.; Wan, C.; Zhang, L. A new diketopiperazine isolated from a Nocardiopsis strain TRM20105 guided by bioassay against Candida albicans. Nat. Prod. Res. 2019, 33, 3421–3425. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, J.; Gamage, C.D.B.; Kim, G.J.; Hillman, P.F.; Lee, C.; Lee, E.Y.; Choi, H.; Kim, H.; Nam, S.-J.; Fenical, W. Androsamide, a cyclic tetrapeptide from a marine Nocardiopsis sp. suppresses motility of colorectal cancer cells. J. Nat. Prod. 2020, 83, 3166–3172. [Google Scholar] [CrossRef] [PubMed]
  68. Xiang, W.-X.; Liu, Q.; Li, X.-M.; Lu, C.-H.; Shen, Y.-M. Four pairs of proline-containing cyclic dipeptides from Nocardiopsis sp. HT88, an endophytic bacterium of Mallotus nudiflorus L. Nat. Prod. Res. 2020, 34, 2219–2224. [Google Scholar] [CrossRef]
  69. Storr, S.J.; Carragher, N.O.; Frame, M.C.; Parr, T.; Martin, S.G. The calpain system and cancer. Nat. Rev. Cancer 2011, 11, 364–374. [Google Scholar] [CrossRef]
  70. Joshi, S.; Singh, A.R.; Zulcic, M.; Bao, L.; Messer, K.; Ideker, T.; Dutkowski, J.; Durden, D.L. Rac2 controls tumor growth, metastasis and M1-M2 macrophage differentiation in vivo. PLoS ONE 2014, 9, e95893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Wertheimer, E.; Gutierrez-Uzquiza, A.; Rosemblit, C.; Lopez-Haber, C.; Sosa, M.S.; Kazanietz, M.G. Rac signaling in breast cancer: A tale of GEFs and GAPs. Cell. Signal. 2012, 24, 353–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Arnison, P.G.; Bibb, M.J.; Bierbaum, G.; Bowers, A.A.; Bugni, T.S.; Bulaj, G.; Camarero, J.A.; Campopiano, D.J.; Challis, G.L.; Clardy, J. Ribosomally synthesized and post-translationally modified peptide natural products: Overview and recommendations for a universal nomenclature. Nat. Prod. Rep. 2013, 30, 108–160. [Google Scholar] [CrossRef] [PubMed]
  73. Walsh, C.T.; O’Brien, R.V.; Khosla, C. Nonproteinogenic amino acid building blocks for nonribosomal peptide and hybrid polyketide scaffolds. Angew. Chem. Int. Ed. 2013, 52, 7098–7124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Walsh, C.T.; Tang, Y. Natural Product Biosynthesis; The Royal Society of Chemistry: London, UK, 2017. [Google Scholar]
  75. Mishra, A.K.; Choi, J.; Choi, S.-J.; Baek, K.-H. Cyclodipeptides: An overview of their biosynthesis and biological activity. Molecules 2017, 22, 1796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Giessen, T.W.; Marahiel, M.A. The tRNA-dependent biosynthesis of modified cyclic dipeptides. Int. J. Mol. Sci. 2014, 15, 14610–14631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, D.; Wang, Y.; Ouyang, Y.; Fu, P.; Zhu, W. Cytotoxic p-terphenyls from a marine-derived Nocardiopsis species. J. Nat. Prod. 2019, 82, 3504–3508. [Google Scholar] [CrossRef] [PubMed]
  78. Chang, Y.; Che, Q.; Xing, L.; Ma, C.; Han, Y.; Zhu, T.; Pfeifer, B.A.; Peng, J.; Zhang, G.; Li, D. Antibacterial p-terphenyl with a rare 2,2′-bithiazole substructure and related compounds isolated from the marine-derived actinomycete Nocardiopsis sp. HDN154086. J. Nat. Prod. 2021, 84, 1226–1231. [Google Scholar] [CrossRef]
  79. Eliwa, E.M.; Abdel-Razek, A.S.; Frese, M.; Halawa, A.H.; El-Agrody, A.M.; Bedair, A.H.; Sewald, N.; Shaaban, M. New naturally occurring phenolic derivatives from marine Nocardiopsis sp. AS23C: Structural elucidation and in silico computational studies. Vietnam J. Chem. 2019, 57, 164–174. [Google Scholar] [CrossRef] [Green Version]
  80. Siddharth, S.; Rai, V.R. Isolation and characterization of bioactive compounds with antibacterial, antioxidant and enzyme inhibitory activities from marine-derived rare actinobacteria, Nocardiopsis sp. SCA21. Microb. Pathog. 2019, 137, 103775. [Google Scholar] [CrossRef]
  81. Sunish, K.S.; Sreedharan, P.; Daniel, S.; Biji, M.; Rosamma, P.; Sukumaran, V.; Mohandas, A.; Singh, I.S.B. A novel substituted derivative of sterol from marine actinomycetes Nocardiopsis alba MCCB 110 antagonistic to the aquaculture pathogen Vibrio harveyi. Microb. Pathog. 2021, 157, 104967. [Google Scholar] [CrossRef]
Figure 1. Structural formulas and proposed biosynthesis mechanism for compounds 13 [48].
Figure 1. Structural formulas and proposed biosynthesis mechanism for compounds 13 [48].
Marinedrugs 20 00374 g001
Figure 2. Structural formulas of compounds 48 [51]. (Reprinted with permission from Ref. [51], Copyright 2019, Journal of Natural Products, published by American Chemical Society).
Figure 2. Structural formulas of compounds 48 [51]. (Reprinted with permission from Ref. [51], Copyright 2019, Journal of Natural Products, published by American Chemical Society).
Marinedrugs 20 00374 g002
Figure 3. Structural formulas of compounds 923 [52,55,56].
Figure 3. Structural formulas of compounds 923 [52,55,56].
Marinedrugs 20 00374 g003
Figure 4. Structural formulas of compounds 2426 [58,59].
Figure 4. Structural formulas of compounds 2426 [58,59].
Marinedrugs 20 00374 g004
Figure 5. Structural formulas of compounds 2734 [22,61,62,63].
Figure 5. Structural formulas of compounds 2734 [22,61,62,63].
Marinedrugs 20 00374 g005
Figure 6. Biosynthesis of loonamycins: (A) loo, atm, and reb gene clusters. (B) Detected biosynthetic route for compounds 27 and 28 [61]. (Reprinted with permission from Ref. [61], Copyright 2020, Organic Letters, published by American Chemical Society).
Figure 6. Biosynthesis of loonamycins: (A) loo, atm, and reb gene clusters. (B) Detected biosynthetic route for compounds 27 and 28 [61]. (Reprinted with permission from Ref. [61], Copyright 2020, Organic Letters, published by American Chemical Society).
Marinedrugs 20 00374 g006
Figure 7. Proposed biosynthesis of compounds 31 and 32 [62]. (Reprinted with permission from Ref. [62], Copyright 2021, Organic Letters, published by American Chemical Society).
Figure 7. Proposed biosynthesis of compounds 31 and 32 [62]. (Reprinted with permission from Ref. [62], Copyright 2021, Organic Letters, published by American Chemical Society).
Marinedrugs 20 00374 g007
Figure 8. Structural formulas of compounds 3545 [64,66,67,68].
Figure 8. Structural formulas of compounds 3545 [64,66,67,68].
Marinedrugs 20 00374 g008
Figure 9. Biosynthesis of ribosomal and non-ribosomal peptides [73].
Figure 9. Biosynthesis of ribosomal and non-ribosomal peptides [73].
Marinedrugs 20 00374 g009
Figure 10. Biosynthesis of CDP through CDPS pathway [76].
Figure 10. Biosynthesis of CDP through CDPS pathway [76].
Marinedrugs 20 00374 g010
Figure 11. Proposed biosynthesis of compound 36 [76].
Figure 11. Proposed biosynthesis of compound 36 [76].
Marinedrugs 20 00374 g011
Figure 12. Structural formulas of compounds 4656 [77,78].
Figure 12. Structural formulas of compounds 4656 [77,78].
Marinedrugs 20 00374 g012
Figure 13. Structural formulas of compounds 5764 [64,79,80].
Figure 13. Structural formulas of compounds 5764 [64,79,80].
Marinedrugs 20 00374 g013
Figure 14. Structural types of compounds isolated from Nocardiopsis from March 2018 to 2021.
Figure 14. Structural types of compounds isolated from Nocardiopsis from March 2018 to 2021.
Marinedrugs 20 00374 g014
Figure 15. Sources of natural products from Nocardiopsis from March 2018 to 2021.
Figure 15. Sources of natural products from Nocardiopsis from March 2018 to 2021.
Marinedrugs 20 00374 g015
Figure 16. Bioactivities of natural products from Nocardiopsis discovered from March 2018 to 2021.
Figure 16. Bioactivities of natural products from Nocardiopsis discovered from March 2018 to 2021.
Marinedrugs 20 00374 g016
Table 1. Compounds isolated from Nocardiopsis during March 2018–2021.
Table 1. Compounds isolated from Nocardiopsis during March 2018–2021.
TypesComps.SourcesDistributionBioactivitiesYearsRefs
Polyketides1–3Sponge Theonella sp. derived Nocardiopsis sp. HB-J378 (GenBank No. MH779065) Antibacterial activity2018[48]
4, 5Saltpan-derived Nocardiopsis sp. CG3 (GenBank No. MG972881)Kenadsa, Algeria 2019[51]
6−8Saltpan-derived Nocardiopsis sp. CG3 (GenBank No. MG972881)Kenadsa, AlgeriaCytotoxicity2019[51]
9−12, 14, 15, 17Desert soil-derived Nocardiopsis spp. HDN154-146 (Genbank no. KY794927) and HDN154-168 (Genbank No. MF952649)Xinjiang, China 2019[52]
13, 16Desert soil-derived Nocardiopsis spp. HDN154-146 (Genbank no. KY794927) and HDN154-168 (Genbank No. MF952649)Xinjiang, ChinaCytoprotective activity2019[52]
18, 19Deep-sea water-derived N. dassonvillei subsp. albirubida HDN 17-237 (Genbank No. MN416280)Mariana Trench 2020[55]
20, 21Marine animal-derived N. aegyptia HDN19-252 (GenBank No. MN822699)AntarcticAntibacterial activity2021[56]
22, 23Marine animal derived strain N. aegyptia HDN19-252 (GenBank No. MN822699)Antarctic 2021[56]
Alkaloids24Marine-derived Nocardiopsis sp. CNY-503 2018[58]
27Marine sediment-derived N. flavescens NA01583 (GenBank No. MT371575)Hainan ChinaCytotoxicity2020[61]
28, 29Marine sediment-derived N. flavescens NA01583 (GenBank No. MT371575)Hainan China 2020[61]
30Marine sediment-derived Nocardiopsis sp. SCA30 (GenBank No. MT573349)Havelock IslandAntibacterial and anticancer activities2021[22]
31, 32Sponge Petrosia sp.-derived N. dassonvillei SCSIO 40065 (GenBank No. MW492395)South China SeaAntibacterial and cytotoxic activities2021[62]
33, 34Marine sediment-derived N. dassonvillei JS106 (GenBank No. MN416229)Lianyungang, China,Antiquorum sensing activity2021[63]
Peptides35Sponge Callyspongia sp.-derived Nocardiopsis sp. UR67Red SeaCytotoxicity2018[64]
36Cotton field-derived Nocardiopsis sp. TRM20105Tarim BasinAntifungal activity2019[66]
37Shore sediment-derived Nocardiopsis sp. CNT-189 (GenBank No. KY111725.1)BahamasCytotoxicity and colorectal cancer motility inhibitor2020[67]
38−45Stems of Mallotus nudiflorus L-derived Nocardiopsis sp. HT88 (Genbank No. MH817156) 2020[68]
Terphenyls46, 49−51Mangrove-derived Nocardiopsis sp. OUCMDZ-4936 (Genbank No. MK129184)Hainan ChinaCytotoxicity2019[77]
47, 48Mangrove-derived Nocardiopsis sp. OUCMDZ-4936 (Genbank No. MK129184)Hainan China 2019[77]
52, 53Marine sediment-derived Nocardiopsis sp. HDN154086 (GenBank No. MK129184)South China SeaAntibacterial activity2021[78]
54−56Marine sediment-derived Nocardiopsis sp. HDN154086 (GenBank No. MK129184)South China Sea 2021[78]
Others57−59Sponge Callyspongia sp.-derived Nocardiopsis sp. UR67Red Sea 2018[64]
60−62Alga Sargassum arnaudianum-derived Nocardiopsis sp. AS23C (GenBank No. MH144210)Red Sea 2019[79]
63, 64Marine sediment-derived Nocardiopsis sp. SCA21 (GenBank No. MH105056)Havelock IslandEnzyme inhibitory, antibacterial, and free radical scavenging activities2019[80]
65Marine-derived N. alba MCCB110 (GenBank No. EU008081) Antibacterial activity2021[81]
Table 2. Antibacterial activities of compounds isolated from Nocardiopsis from March 2018 to 2021.
Table 2. Antibacterial activities of compounds isolated from Nocardiopsis from March 2018 to 2021.
StrainsComps.Values (MIC)ProsCons
MRSA1, 312.5 μg/mLSpecific inhibition of MRSAModerate activity [48]
23.12 μg/mLStrong activity; Specific inhibition of MRSA[48]
MRCNS20/21 (μM)6.2/6.2Broad-spectrum antibacterial activity; Strong activity against MRCNS compared with positive controlModerate activity against B. subtilis and Proteus sp. compared with positive control [56]
B. subtilis6.2/6.2
Proteus sp.12.5/6.2
B. cereus6.2/6.2
E. coli6.2/6.2
M. Phlei6.2/3.1
MRSA ATCC NR-4607130 (μg/mL)15.6Strong activity[22]
MRSA ATCC NR-461717.8
B. subtilis 106431/32 (μg/mL)8/16Broad-spectrum antibacterial activityWeak activity [62]
M. Luteus SCSIO ML0116/32
S. aureus ATCC 2921364/64
MRSA shhsA132/32
E. faecalis ATCC 2921232/-
V. alginolyticus 1321432/64
Proteus sp.52/53 (μM)3.1/1252 showed strong and broad-spectrum activity, and 53 displayed strong activity of MRSA53 displayed moderate activity against Proteus sp. and B. subtilis [78]
B. cereus1.5/-
M. phlei6.2/-
B. subtilis3.1/12
MRSA-/6.2
V. parahemolyticus6.2/-
E. coli3.1/-
K. pneumoniae ATCC 1388363/64 (μg/mL)125/250Broad-spectrum antibacterial activityWeak activity [80]
Listeria cytogens ATCC 1393262.5/-
S. aureus ATCC 1260062.5/125
B. subtilis ATCC 66337.81/7.81
MRSA ATCC NR-4617115.62/7.81
MRSA ATCC NR-46071125/15.62
V. harveyi MCCB 1116520 mm (zone of inhibition)Not toxic against VERO cell line and shrimp hemocytes up to 1000 ppm[81]
Table 3. Cytotoxicity and other activities of compounds isolated from Nocardiopsis from March 2018 to 2021.
Table 3. Cytotoxicity and other activities of compounds isolated from Nocardiopsis from March 2018 to 2021.
BioactivitiesCells/Stains/EnzymeComps.ValuesProsCons
Cytotoxicity (IC50)Hela cells KB3.16/7/8 (μM)6.8/5.4/2.4Compound 8 with broad-spectrum cytotoxicityModerate activity [51]
PC-36.3/5.0/2.1
A549-/-/6.5
SKOV-3-/10.0/5.5
SH-SY5Y27 (nM)283.6Extremely potent and broad-spectrum cytotoxicity[61]
Sum1315121.3
HT2981.3
SW62090.5
HCT11631.4
HeLa100.1
SW87292.3
HCC7841.5
30 Broad-spectrum cytotoxicityIC50 untested [22]
SF-26831/32 (μM)17.0/11.9Broad-spectrum cytotoxicityModerate activity [62]
MCF-725.7/20.7
HepG231.2/12.0
A54934.4/13.5
MM. 1S35 (μM)8Broad-spectrum cytotoxicityModerate activity [64]
HeLa11
CT2612
AGS37 (μM)13Broad-spectrum cytotoxicityModerate activity [67]
Caco218
HCT11621
L-0249 (μM)17Strong and broad-spectrum cytotoxicity[77]
A5495.1
K5620.77
MCF-76.0
P6C9.4
N8746/50/51 (μM)-/1.0/-
A673-/0.76/8.9
MV4-114.0/0.16/0.77
K5629.0/4.8/8.9
A5497.8/0.48/9.7
BT4746.0/3.6/-
H1229-/0.72/-
HUCCT1-/0.20/-
B16F10-/0.76/-
MDA-MB-4682.8/1.1/0.67
H1975-/3.1/4.4
HL600.38/0.17/5.0
A4314.6/-/-
U251-/4.7/-
HCC19540.10/0.48/2.0
MCF-718/-/17
MKN-45-/0.49/12
DU-145-/0.52/1.0
SPC-A1-/2.0/9.8
HCT116-/1.9/-
143B5.5/5.0/7.7
H22281.7/0.94/5.0
MDA-MB-231-/2.0/2.0
Cytoprotective activityHaCaT cells13, 16 [52]
Antiquorum sensing activity (IC50)C. violaceum 1247233/34 (μg/mL)23.59/6.82 [63]
Antifungal activity (MIC)C. albicans363.16 mM Weak activity [66]
Colorectal cancer motility inhibitionCaco237 Strong activity[67]
Enzyme-inhibitory activity (IC50)α-glucosidase63/6494.61/202.33strong activity against α-glucosidase[80]
α-amylase103.23/-
Free radical scavenging activity 63/64 Strong activity[80]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shi, T.; Wang, Y.-F.; Wang, H.; Wang, B. Genus Nocardiopsis: A Prolific Producer of Natural Products. Mar. Drugs 2022, 20, 374. https://doi.org/10.3390/md20060374

AMA Style

Shi T, Wang Y-F, Wang H, Wang B. Genus Nocardiopsis: A Prolific Producer of Natural Products. Marine Drugs. 2022; 20(6):374. https://doi.org/10.3390/md20060374

Chicago/Turabian Style

Shi, Ting, Yi-Fei Wang, Han Wang, and Bo Wang. 2022. "Genus Nocardiopsis: A Prolific Producer of Natural Products" Marine Drugs 20, no. 6: 374. https://doi.org/10.3390/md20060374

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