Next Article in Journal
Anandins A and B, Two Rare Steroidal Alkaloids from a Marine Streptomyces anandii H41-59
Next Article in Special Issue
Crambescidin 800, Isolated from the Marine Sponge Monanchora viridis, Induces Cell Cycle Arrest and Apoptosis in Triple-Negative Breast Cancer Cells
Previous Article in Journal
Reclamation of Marine Chitinous Materials for the Production of α-Glucosidase Inhibitors via Microbial Conversion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioactive Secondary Metabolites from the Marine Sponge Genus Agelas

1
Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
2
Department of Chemistry & Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
*
Author to whom correspondence should be addressed.
Mar. Drugs 2017, 15(11), 351; https://doi.org/10.3390/md15110351
Submission received: 19 September 2017 / Revised: 25 October 2017 / Accepted: 3 November 2017 / Published: 8 November 2017
(This article belongs to the Special Issue Bioactive Compounds from Marine Sponges)

Abstract

:
The marine sponge genus Agelas comprises a rich reservoir of species and natural products with diverse chemical structures and biological properties with potential application in new drug development. This review for the first time summarized secondary metabolites from Agelas sponges discovered in the past 47 years together with their bioactive effects.

1. Introduction

The search for natural drug candidates from marine organisms is the eternal impetus to pharmaceutical scientists. For the past six decades, marine sponges have been a prolific and chemically diverse source of natural compounds with potential therapeutic application [1,2]. The marine sponge Agelas (Porifera, Demospongiae, Agelasida, Agelasidae) is widely distributed in the marine eco-system and includes at least 19 species (Figure 1): A. axifera, A. cerebrum, A. ceylonica, A. citrina, A. clathrodes, A. conifera, A. dendromorpha, A. dispar, A. gracilis, A. linnaei, A. longissima, A. mauritiana, A. nakamurai, A. nemoechinata, A. oroides, A. sceptrum, A. schmidtii, A. sventres, and A. wiedenmayeri. Since the beginning of the 1970s, many research groups around the world have carried out chemical investigation on Agelas spp., resulting in fruitful achievements. Their studies revealed that Agelas sponges harbor many bioactive secondary metabolites, including alkaloids (especially bromopyrrole derivatives), terpenoids, glycosphingolipids, carotenoids, fatty acids and meroterpenoids [3]. These natural products are an attractive resource for drug candidates due to their rich chemodiversity and interesting biological activities.

2. Natural Products from Agelas Genus

The chemical diversity of natural products is determined by the biological diversity of organisms. To date, 291 secondary metabolites (1291) have been isolated and characterized from the marine sponge Agelas spp. (Table 1). These chemicals were introduced and assorted as follows according to their biological sources.

2.1. Agelas axifera

Three new alkaloids, named axistatins 1 (1), 2 (2), and 3 (3) (Figure 2), were isolated and characterized from Agelas axifera collected in the Republic of Palau and found to exhibit inhibitory effects on cancer cell lines, including P388, BXPC-3, MCF-7, SF-268, NCI-H460, KM20L2 and DU-145. The exquisitely sensitive Gram-negative pathogen Neisseria gonorrheae and the opportunistic fungus Cryptococcus neoformans were inhibited by 13 with MIC values of 1–8, 2–4, and 8 μg/mL, and 1–4, 2, and 8–16 μg/mL, respectively. Furthermore, these compounds had antimicrobial effect on Gram-positive bacteria, including Staphylococcus aureus, Streptococcus pneumoniae, Enterococcus faecalis and Micrococcus luteus [4].

2.2. Agelas cerebrum

Marine sponge Agelas cerebrum was classified as a new species in 2001 [5]. Chemical investigation of Caribbean specimen A. cerebrum led to the isolation of three brominated compounds, 5-bromopyrrole-2-carboxylic acid (4), 4-bromopyrrole-2-carboxylic acid (5) and 3,4-bromopyrrole-2-carboxylic acid (6) (Figure 3) [6]. Biological tests indicated that these isolates had strong cytotoxic activities in vitro against human tumor cell lines at ≥1 mg/mL, including A549, HT29 and MDA-MB-231.

2.3. Agelas ceylonica

Only one case of chemical study on Agelas ceylonica has been reported [7]. The specimen of A. ceylonica collected from India Mandapam coast was found to produce one methyl ester hanishin (7) (Figure 4), which has been previously found in the marine sponge Homaxinella sp. [8].

2.4. Agelas citrina

The Caribbean specimen of Agelas citrina was firstly found to yield three new diterpene alkaloids, (−)-agelasidine E (8), (−)-agelasidine F (9) and agelasine N (10) [9]. Latter chemical investigation showed that this sponge also produces four new pyrrole-imidazole alkaloids, citrinamines A–D (1114), and one bromopyrrole alkaloid N-methylagelongine (15) (Figure 5) [10]. Compounds 1214 had antimicrobial activities whereas no inhibitory effect on cell proliferation of mouse fibroblasts was found for 1114.

2.5. Agelas clathrodes

Marine sponge Agelas clathrodes was the excellent producer of secondary metabolites, including glycosphingolipid derivatives (GSLs) and alkaloids. Clarhamnoside (16), containing an unusual l-rhamnose unit in the sugar head, was the first rhamnosylated α-galactosylceramide from A. clathrodes collected along the coast of Grand Bahamas Island (Sweetings Cay) [11]. The Caribbean sponge A. clathrodes could metabolize clathrosides A–C (1719) and isoclathrosides A–C (2022), which, respectively, belonged to two families of different glycolipids [12]. Compound 23 was also isolated from the Caribbean specimen (Figure 6) [13]. It was noted that all the GSLs from A. clathrodes were actually elucidated as mixtures of homologs, which play an important role in therapeutic immunomodulation.
Six alkaloids, (−)-agelasidine A (24), (−)-agelasidine C (25), (−)-agelasidine D (26), clathramide A (27), clathramide B (28) and clathrodin (29), were detected in the Caribbean sponge A. clathrodes (Figure 7). Bioassay results suggested that compound 24 possessed inhibitory effect on Staphilococcus aureus but no effect on fungi, while 25 and 26 were shown to have antimicrobial activities against S. aureus, Klebsiella pneumoniae and Proteus vulgaris [14]. In vitro cytotoxic test indicated that 25 and 26 significantly inhibited the growth of CHO-K1 cells with the ED50 values of 5.70 and 2.21 μg/mL, respectively. Compound 26 also possessed the inhibition against the growth of E. coli and Hafnia alvei [15], while 27 and 28 had a moderate antifungal activity against Aspergillus niger [16]. Interestingly, compound 29 contained a nonbrominated pyrrole and a guanidine moiety [17]. One specimen of A. clathrodes from the South China Sea was shown to produce an ionic compound (30), which had weak cytotoxicity against cancer cell lines A549 and SGC7901 with IC50 values of 26.5 and 22.7 μg/mL, respectively [18]. Four brominated compounds, dispacamides A–D (3134) (Figure 7), were detected not only in A. clathrodes, but also in A. conifera, A. dispar and A. longissima, and exhibited antihistamine activity [19,20].

2.6. Agelas conifera

Chemical study of two specimens of Agelas conifera from the Florida Keys and Belize led to the isolation of two new dimeric bromopyrrole alkaloids, bromosceptrin (35) and debromosceptrin (36), respectively [21,22]. Seven new bromopyrrole metabolites (3743) were firstly purified from the Caribbean sponge A. conifera [23], but the detailed structure elucidation of ageliferin (41), bromoageferin (42) and dibromoageliferin (43) were established by Kobayashi and his co-workers [24]. Bioassay results indicated that compounds 37, 41, 42 and 43 possessed biological activities against Bacillus subtilis at 10 μg/disk and 41 and 42 could inhibit the growth of E. coli at 10 μg/disk. Using new protein-guided methods by its affinity to proteins within tumor cell proteomes, one unique polyhydroxybutyrated β-GSL coniferoside (44), was detected in A. conifera derived from Puerto Rico as well as another GSL derivative (45) (Figure 8) [25,26].

2.7. Agelas dendromorpha

Natural product analysis of the marine sponge Agelas dendromorpha revealed three novel agelastatins (4648) with pyrrole-2-imidazole structure. Agelastatin A (46) was obtained from the Axinellid specimen grown in the Coral Sea and had strong cytotoxicity [27]. Agelastatins E (47) and F (48) (Figure 9) purified from the New Caledonian A. dendromorpha were shown to exhibit weak cytotoxicity against the KB cell line at 30 μM [28].

2.8. Agelas dispar

It is notable that Caribbean Agelas dispar harbors a distinct biogeographical bromination trend. Five compounds containing bromine, dispyrin (49), dibromoagelaspongin methyl ether (50), longamide B (51), clathramides C (52) and D (53), have been found in the Caribbean sponge A. dispar [29,30]. Only compound 51 had moderate anti-bacterial activities against B. subtilis and S. aureus with MIC values of about 50 μg/mL. The GSL derivative (54) and betaine alkaloids (5557) were detected in the Caribbean A. dispar [31,32]. Antibacterial tests indicated that compounds 55 and 56 had the inhibitory activities against B. subtilis and S. aureus with MIC values ranging from 2.5 to 8.0 μg/mL [32]. The first quaternary derivative of adenine in nature, agelasine (58) (Figure 10), was also found in A. dispar [33].

2.9. Agelas gracilis

Bioassay-guided fractionation of the crude extract of the deep-sea sponge Agelas gracilis collected in southern Japan afforded three novel antiprotozoan compounds, gracilioethers A–C (5961) (Figure 11) [34]. Antimalarial tests showed that these metabolites possessed inhibitory effects on Plasmodium falciparum with IC50 values of 0.5–10 μg/mL.

2.10. Agelas linnaei

Eleven novel brominated pyrrole derivatives (6272) (Figure 12) were purified from the Indonesian sponge Agelas linnaei and compounds 6669 had prominent activities against the murine L1578Y mouse lymphoma cell line with IC50 values of 9.55, 9.25, 16.76 and 13.06 μM, respectively [35].

2.11. Agelas longissima

Five alkaloids (7377) (Figure 13) have been isolated from Agelas longissima specimens, all of which were collected from the Caribbean Sea. Agelongine (73) contained a pyridinium ring instead of the commonly found imidazole nucleus in Agelas alkaloids and was shown to be specific to inhibit the agonist 5-hydroxytryptamine (5-HT) [36]. Compound 75 was unique for its unusual pyrrolopiperazine nucleus [37]. Two new GSL analogs (76 and 77) were also found in the Caribbean A. longissima [38,39].

2.12. Agelas mauritiana

Agelas mauritiana is one of the most fruitful producers of secondary metabolites among all Agelas species. Thirty-five compounds (78112) have been isolated and identified from A. mauritiana, including terpenoids, pyrrole derivatives, GSLs, carotenoids and other alkaloids. Specimens of A. mauritiana collected from the South China Sea were found to metabolize eight terpenoids (7885) [40,41]. Compound 79 possessed inhibitory effects on S. aureus with MIC90 value of 1–8 μg/mL and activities against PC9, A549, HepG2, MCF-7, and U937 cell lines with IC50 values of 4.49–14.41 μM. Compound 84 possessed activities against C. neoformans, S. aureus, methicillin-resistant S. aureus and Leishmania donovani with IC50/MIC values of 4.96/10.00, 7.21/10.00, 9.20/20.00 and 28.55/33.19 μg/mL, respectively. Agelasimines A (86) and B (87) and an unusual purino-diterpene (88) were purified from Eniwetak A. mauritiana and 86 and 87 had inhibitory effect on L1210 leukemia with ED50 values of 2.1 and 3.9 nM, respectively. From Pohnpei-derived A. mauritiana, epi-agelasine C (89) was isolated and shown to have no activity against S. aureus, Vibrio costicola, E. coli and B. subtilis [42,43,44]. Chemical analysis of one specimen of A. auritiana collected from the Solomon Islands afforded agelasines J (90), K (91) and L (92) (Figure 14), which exhibited moderate activities against P. falciparum and low cytotoxicity on MCF-7 cells [45].
The same species of A. mauritiana grown in different places were found to produce different pyrrole derivatives, such as debromodispacamides B (93) and D (94) from Solomon Island specimen [46], 4-bromo-N-(butoxymethyl)-1H-pyrrole-2-carboxamide (95) from the South China Sea [41], 5-debromomidpacamide (96) from Enewetak Atoll [47], mauritamide A (97) from Fiji [48] and mauritiamine (98) from Hachijo-jima Island [49]. Compound 98 exhibited inhibitory effect on larval metamorphosis of the barnacle Balanus amphitrite with ED50 value of 15 μg/mL and moderate antibacterial activity against Flavobacterium marinotypicum with the inhibition zone of 10 mm at 10 µg/disk. Interestingly, the Pacific sponge A. mauritiana was found to metabolize other pyrroles, including debromokeramadine (99), benzosceptrin A (100), nagelamide S (101) and nagelamide T (102) (Figure 15) [50,51].
Agelasphins (103110) from the Okinawan A. mauritiana were the first example of galactosylceramides containing an α-galactosyl linkage [52,53]. These compounds exhibited high activity with the relative tumor proliferation rate (T/C) ranging from 160% to 190% and 200–400% relative 3H-TdR incorporation at <l µg/mL. But no activity was observed against B16 melanoma cells at 20 µg/mL. Two natural carotenoids, isotedanin (111) and isoclathriaxanthin (112) (Figure 16), were also detected in the specimen of A. mauritiana from Kagoshima [54].

2.13. Agelas nakamurai

Thirty-three chemicals have been characterized from Agelas nakamurai, including 16 terpenoids and 17 pyrrole alkaloids. The Okinawan A. nakamurai seems to occupy the majority of terpenoid compounds, including agelasidines B (113) and C (114) [55], nakamurols A–D (115118) [56], 2-oxoagelasiines A (119) and F (120), 10-hydro-9-hydroxyagelasine F (121) [57], agelasines E (122) and F (123) [58]. Compounds 113 and 114 were found to have inhibitory effects on the growth of S. aureus at 3.3 µg/mL and on contractile responses of smooth muscles. Compounds 119 and 120 showed markedly reduced activity against Mycobacterium smegmatis. The Indonesian A. nakamurai was found to yield two novel diterpenes, (−)-agelasine D (124) and (−)-ageloxime D (125). Antibacterial assay revealed 124 could inhibit the growth of Staphylococcus epidermidis with a MIC value < 0.0877 µM [35]. Isoagelasine C (126) and isoagelasidine B (127) were isolated from specimen of the South China Sea and possessed weak cytotoxicities against HL-60, K562 and HCT-116 cell lines with IC50 values ranging from 18.4 to 39.2 µM [59]. A new diterpene (128) (Figure 17) with a 9-methyladenum moiety produced by the Papua New Guinean A. nakamurai Hoshino was shown to be inactive against HIV-1 integrase, E. coli and Pseudomonas aeruginosa at 12.5 µg/mL [60].
Five non-brominated pyrrole derivatives, nakamurines A–E (129133), were purified from the South China Sea A. nakamurai [59,61]. Bioassay results showed that compound 130 had weak inhibition against Candida albicans with a MIC value of 60 µg/mL [61]. Bromopyrrole alkaloid was one of the most common secondary metabolites from marine sponges [62]. Two bromopyrrole alkaloids (134 and 135) were firstly isolated from the Papua New Guinean A. nakamurai in 1998 [60]. Ageladine A (136) containing 2-aminoimidazolopyridine was shown to have inhibitory effects on Matrix metalloproteinases-1, -2, -8, -9, -12 and -13 with IC50 values of 1.2, 2.0, 0.39, 0.79, 0.33, and 0.47 µg/mL, respectively [63]. Chemical investigation of the Indonesia A. nakamurai afforded longamide C (137) [35]. Nakamuric acid (138) and its methyl ester (139) were characterized from the Indopacific specimen and shown to be active against B. subtilis [64]. The Okinawan A. nakamurai was found to produce six brominated pyrrole derivatives, slagenins A–C (140142) and mukanadins A–C (143145) (Figure 18), of which 141 and 142 showed inhibitory effect on murine leukemia L1210 cells in vitro with IC50 values of 7.5 and 7.0 µg/mL, respectively [65,66].

2.14. Agelas nemoechinata

Nemoechines A–D (146149) and nemoechioxide A (150) were obtained from the sponge Agelas aff. nemoechinata collected from the South China Sea. Compounds 146148 had enantiomeric configurations and 146 had an unusual cyclopentene-fused imidazole ring system. Bioassay results suggested that only 149 had cytotoxicity against HL-60 cell lines with an IC50 value of 9.9 µM [67]. Two new nemoechine members, nemoechines F (151) and G (152) possessing N-methyladenine, were purified from the South China Sea-derived A. nemoechinata. Compound 152 had weak toxicity against Jurkat cell line with an IC50 value of 17.1 µM [68]. Oxysceptrin (153) (Figure 19) derived from the Okinawan A. nemoechinata was a potent actomyosin ATPase activator [69].

2.15. Agelas oroides

Thirty-six secondary metabolites (154189) (Figure 20) have been isolated from the marine sponge Agelas oroides, including pyrrole derivatives, sterols and fatty acids. Chemical investigation of A. oroides from the Great Barrier Reef afforded three fistularin-3 derivatives, agelorin A (154), agelorin B (155) and 11-epi-fistularin-3 (156). These metabolites exhibited antimicrobial activities against B. subtilis and M. luteus and 156 had moderate cytotoxicity against breast cancer cells [70]. Later on, two new naturally occurring pyrrole derivatives (157 and 158) and 2,4,6,6-tetramethyl-3(6H)-pyridone (159) were obtained from the same specimen [71,72]. Mediterranean A. oroides was shown to produce four novel compounds, cyclooroidin (160), taurodispacamide A (161), monobromoagelaspongin (162) and (−)-equinobetaine B (163), of which 161 displayed strong antihistaminic activity [73,74]. Five bromopyrrole alkaloids (164168) and fifteen sterols (169183) were detected in the sponge A. oroides collected in the Bay of Naples [75,76]. Interestingly, one imidazole compound (184), taurine (185) and some fatty acids (186189) were also found in the Northern Aegean Sea-derived specimen [77].

2.16. Agelas sceptrum

One novel C29 sterol containing the typical nucleus of ergosterol, 26-nor-25-isopropyl-ergosta-5,7,22E-trien-3β-ol (190), was purified from the Jamaican A. sceptrum [78]. Sceptrin (191) was obtained from A. sceptrum collected at Glover Reef and found to have a broad spectrum of antimicrobial activities against S. aureus, B. subtilis, C. albicans, Pseudomonas aeruginosa, Alternaria sp. and Cladosporium cucumerinum [79]. Chemical study of the sponge from Bahamas afforded two hybrid pyrrole-imidazole alkaloids: 15′-oxoadenosceptrin (192) and decarboxyagelamadin C (193) (Figure 21) [80].

2.17. Agelas schmidtii

Three monohydroxyl sterols (194196) were isolated from the Caribbean Agelas schmidtii [81]. Additionally, four carotenoids named α-carotene (197), isorenieratene (198), trikentriorhodin (199) and zeaxanthin (200) (Figure 22) were also derived from this sponge collected from West Indies [82].

2.18. Agelas sventres

Only one new bromopyrrole alkaloid, sventrin (201) (Figure 23), has been purified from the Caribbean sponge Agelas sventres. Biological assay showed that this chemical has feeding deterrent activity against omnivorous reef fish [83].

2.19. Agelas wiedenmayeri

Chemical investigation of Agelas wiedenmayeri from Florida Keys afforded one new pyrrole derivative, 4-bromopyrrole-2-carboxyhomoarginine (202) (Figure 24), which might be alternatively a biosynthetic precursor of hymenidin/oroidin-related alkaloids [84].

2.20. Other Agelas spp.

Eighty-nine secondary metabolites (203291) were isolated and chemically identified from unclassified Agelas species and assorted into two classes, ionic and non-ionic compounds as below.

2.20.1. Ionic Compounds

As described above, ionic compounds are the major secondary metabolites of Agelas sponge, which can be grouped in bromine-containing and non-bromine-containing compounds. It is eminent that all ionic brominated metabolites were produced by the Okinawan Agelas spp. besides dibromoagelaspongin hydrochloride (203) [85]. Agelamadins A (204) and B (205), possessing an agelastatin-like tetracyclic moiety and an oroidin-like linear moiety, were shown to have antimicrobial activity against B. subtilis, M. luteus and C. neoformans [86]. The same specimen was also found to metabolize agelamadins C–F (206209) and tauroacidin E (210) (Figure 25), of which 209 was the first occurrence bromopyrrole alkaloid for containing aminoimidazole and pyridinium moieties simultaneously [87,88].
Twenty-one nagelamides (211231) (Figure 26) have been characterized from the Okinawan Agelas spp. Nagelamides A–H (211218) and O (219) were shown to possess antimicrobial activities against M. luteus, B. subtilis and E. coli. Compounds 211, 217 and 218 were shown to inhibit the growth of protein phosphatase type 2A with IC50 values of 48, 13 and 46 µM, respectively [89,90]. Nagelamides K (220) and L (221) had inhibitory effect on M. luteus with a MIC value of 16.7 µg/mL [91]. Bioactivity test uncovered that nagelamides M (222) and N (223) exhibited inhibition against A. niger with the same MIC value of 33.3 µg/mL [92]. Nagelamides Q (224) and R (225), of which compound 225 possessed an oxazoline ring for the first time, showed antimicrobial activity against B. subtilis, Trichophyton mentagrophytes, C. neoformans, C. albicans and A. niger [93]. Nagelamides U (226) and V (227) were the first occurence for a bromopyrrole alkaloid containing a γ-lactam ring with an N-ethanesulfonic acid and guanidino moieties, while nagelamide W (228) was the first monomeric bromopyrrole alkaloid with two aminoimidazole moieties in the molecule. Compounds 226 and 228 could inhibit against C. albicans with the same IC50 value of 4 µg/mL [94]. Nagelamides X (229) and Y (230) were unique for their novel tricyclic skeleton consisting of spiro-bonded tetrahydrobenzaminoimidazole and aminoimidazolidine moieties. In addition, nagelamide Z (231) was the first example for dimeric bromopyrrole alkaloid involving the C-8 position in dimerization and displayed strong antimicrobial activity against C. albicans with an IC50 value of 0.25 µg/mL [95].
Eight new bromopyrrole alkaloids, 2-bromokeramadine (232), 2-bromo-9,10-dihydrokeramadine (233), tauroacidins C (234) and D (235), mukanadin G (236), 2-debromonagelamides U (237) and G (238), 2-debromonagelamide P (239), keramadine (240) and agelasine G (241) (Figure 27) were detected in the Okinawan Agelas spp. [96,97,98,99] Antimicrobial tests suggested that compound 236 exhibited inhibitory effects on C. albicans and C. neoformans with IC50 values of 16 and 8 µg/mL, respectively [96]. Compounds 237 and 239 could inhibit the growth of T. mentagrophytes with IC50 values of 16 and 32 µg/mL, respectively. Cytotoxicity assay revealed that 241 showed toxicity against murine lymphoma L1210 cells in vitro with an IC50 value of 3.1 µg/mL [97,99].
Nineteen non-bromine-containing ionic compounds have been isolated from unclassified Agelas spp., including eleven agalasines (242252) from Okinawa [100,101], two agelasines (253 and 254) from Yap Island [102], four higher unsaturated 9-N-methyladeninium bicyclic diterpenoids (255258) from Papua New Guinea [103] and two quarternary 9-methyladenine salts of diterpenes agelines (259 and 260) from Argulpelu Reef [104]. Compounds 242245 displayed strong inhibitory effects on Na, K-ATPase and antimicrobial activities [100]. Agelasine M (255) exhibited potent activity against Trypanosoma brucei [103], while agelines A (259) and B (260) (Figure 28) showed mild ichthyotoxins and antimicrobial activities [104].

2.20.2. Non-Ionic Compounds

Since 1983, 29 non-ionic brominated metabolites (261289) have been found in some unclassified Agelas spp. collected from the Okinawan Sea, the South China Sea, the Caribbean Sea, Papua New Guinea and the Indian Ocean. Agesamides A (261) and B (262) [105], benzosceptrin C (263) [106], nagelamide J (264) [107], nagelamide P (265), mukanadin E (266), mukanadin F (267) [92], nagelamide I (268) and 2,2’-didebromonagelamide B (269) [108] were obtained from the Okinawan specimen. Compound 264 had a cyclopentane ring fused to an amino imidazole ring and exhibited inhibitory effect on S. aureus and C. neoformans with MIC values of 8.35 and 16.7 µg/mL, respectively. Compounds 268 and 269 were inactive against murine lymphoma L1210 and human epidermoid carcinoma KB cells in vitro. Chemical study of an unidentified Agelas spp. from the South China Sea afforded ten new non-ionic bromopyrrole derivatives, longamides D–F (270272), 3-oxethyl-4-[1-(4,5-dibromopyrrole-2-yl)-formamido]-butanoic acid methyl ester (273), 2-oxethyl-3-[1-(4,5-dibromopyrrole-2-yl)-formamido]-methyl propionate (274), 9-oxethyl-mukanadin F (275) [109], hexazosceptrin (276), agelestes A (277) and B (278) and (9S, 10R, 9’S, 10’R)-nakamuric acid (279) [110]. Inspiringly, bioassay results revealed that (+)-270, (−)-271 and (+)-272 had significant antimicrobial activity against C. albicans with MIC values of 80, 20 and 140 µM, respectively. Monobromoisophakellin (280) was isolated from the Caribbean Agelas sp. and shown to possess antifeedant activity against Thalassoma bifasciatum [111]. Chemical investigation of Agelas sponges from Wewak and Indonesian sea respectively led to the isolation of two phakellin alkaloids (281,282) and 5-bromophakelline (283) [112,113]. In addition, 2,3-dibromopyrrole (284) and 2,3-dibromo-5-methoxymethylpyrrole (285) belonging to non-ionic bromopyrrole alkaloid were purified from the marine sponge Agelas sp. [114]. Apart from alkaloids, four new brominated phospholipid fatty acids (286289) (Figure 29) were also detected in the Caribbean Agelas spp. [115].
Only two non-ionic metabolites without bromine, agelasidine A (290) and agelagalastatin (291) (Figure 30), have been detected in two unclassified specimens of Agelas sp. Compound 290 was the first marine natural substance possessing sulfone and guanidine units purified from the Okinawan sample and showed antispasmodic activity [116]. It was notable that compound 24 from the Caribbean A. clathrodes is the optimal isomer of 290. Compound 291 was a new GSL derived from Agelas sp. collected in Papua New Guinea and found to exhibit significant in vitro activity against human cancer cell lines with lung NCI-H460 GI50 0.77 µg/mL to ovary OVCAR-3 GI50 2.8 µg/mL [117].

3. Conclusions

Many efforts have been devoted to implement chemical investigation of Agelas sponges during the past 47 years, from 1971 to 2017. Meanwhile, great achievements have been made on chemical diversity of their secondary metabolites. Agelas sponges are widely distributed in the ocean, especially in the Okinawa Sea, the Caribbean Sea and the South China Sea. Deep ocean technologies for specimen collecting should be used to search more unknown species of Agelas sponges, such as manned and remotely operated underwater vehicles. Advanced separation methodologies should be deployed to explore more bioactive secondary metabolites of these sponges, such as UPLC-MS, metabolomics approach [74]. Furthermore, special attention should be paid to symbiotic microorganisms of Agelas sponges owing to the fact that a great number of therapeutic agents of marine sponges are biosynthesized by their symbiotic microbes [118]. By a combination of gene engineering, pathway reconstructing, enzyme engineering and metabolic networks, these microbes can be modified to produce more novel chemicals containing enhanced structural features or a large quantity of known valuable compounds for pharmaceutical production.

Acknowledgments

Financial support from the National Natural Science Foundation of China (Nos. 41776139 and 81773628), the Zhejiang Provincial Natural Science Foundation of China (LY16H300007 and LY16H300008) and the US National Cancer Institute grants (R01 CA 047135) are gratefully acknowledged.

Author Contributions

H.Z. conceived and wrote the paper; M.D. and J.C. searched and collected all references; and H.W., K.T. and P.C. made suggestive revision and provided eight photos of Agelas sponges.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rane, R.; Sahu, N.; Shah, C.; Karpoormath, R. Marine bromopyrrole alkaloids: Synthesis and diverse medicinal applications. Curr. Top. Med. Chem. 2014, 14, 253–273. [Google Scholar] [CrossRef] [PubMed]
  2. Ancheeva, E.; El-Neketi, M.; Song, W.; Lin, W.; Daletos, G.; Ebrahim, W.; Proksch, P. Structurally unprecedented metabolites from marine sponges. Curr. Org. Chem. 2017, 21, 426–449. [Google Scholar] [CrossRef]
  3. Bickmeyer, U.; Drechsler, C.; Köck, M.; Assmann, M. Brominated pyrrole alkaloids from marine Agelas sponges reduce depolarization-induced cellular calcium elevation. Toxicon 2004, 44, 45–51. [Google Scholar] [CrossRef] [PubMed]
  4. Pettit, G.R.; Tang, Y.; Zhang, Q.; Bourne, G.T.; Arm, C.A.; Leet, J.E.; Knight, J.C.; Pettit, R.K.; Chapuis, J.C.; Doubek, D.L.; et al. Isolation and structures of axistatins 1-3 from the Republic of Palau marine sponge Agelas axifera Hentschel. J. Nat. Prod. 2013, 76, 420–424. [Google Scholar] [CrossRef] [PubMed]
  5. Assmann, M.; Soest, R.W.M.; Köck, M. Description of Agelas cerebrum, a new species and redescription of A. dilata. Proc. Biol. Soc. Wash. 2001, 114, 359–366. [Google Scholar]
  6. Regalado, E.L.; Laguna, A.; Mendiola, J.; Thomas, O.P.; Nogueiras, C. Bromopyrrole alkaloids from the Caribbean sponge Agelas cerebrum. Quim. Nova 2011, 34, 289–291. [Google Scholar] [CrossRef]
  7. Reddy, N.S.; Venkateswarlu, Y. S-(+)-methyl ester of hanishin from the marine sponge Agelas ceylonica. Biochem. Syst. Ecol. 2000, 28, 1035–1037. [Google Scholar] [CrossRef]
  8. Umeyama, A.; Ito, S.; Yuasa, E.; Arihara, S.; Yamada, T. A new bromopyrrole alkaloid and the optical resolution of the racemate from the marine sponge Homaxinella sp. J. Nat. Prod. 1998, 61, 1433–1434. [Google Scholar] [CrossRef] [PubMed]
  9. Stout, E.P.; Yu, L.C.; Molinski, T.F. Antifungal diterpene alkaloids from the Caribbean sponge Agelas citrina: Unified configurational assignments of agelasidines and agelasines. Eur. J. Org. Chem. 2012, 5131–5135. [Google Scholar] [CrossRef] [PubMed]
  10. Cychon, C.; Lichte, E.; Köck, M. The marine sponge Agelas citrina as a source of the new pyrrole-imidazole alkaloids citrinamines A–D and N-methylagelongine. Beilstein J. Org. Chem. 2015, 11, 2029–2037. [Google Scholar] [CrossRef] [PubMed]
  11. Costantino, V.; Fattorusso, E.; Imperatore, C.; Mangoni, A. Clarhamnoside, the first rhamnosylated α-galactosylceramide from Agelas clathrodes. Improving spectral strategies for glycoconjugate structure determination. J. Org. Chem. 2004, 69, 1174–1179. [Google Scholar] [CrossRef] [PubMed]
  12. Costantino, V.; Fattorusso, E.; Imperatore, C.; Mangoni, A. Clathrosides and isoclathrosides, unique glycolipids from the Caribbean sponge Agelas clathrode. J. Nat. Prod. 2006, 69, 73–78. [Google Scholar] [CrossRef] [PubMed]
  13. Costantino, V.; Fattorusso, E.; Mangoni, A. Glycosyl ceramide composition of the marine sponge Agelas clathrodes. Liebigs Ann. 1995, 1995, 1471–1475. [Google Scholar] [CrossRef]
  14. Medeiros, A.M.; Lourenço, A.; Tavares, M.R.; Curto, M.J.M.; Feio, S.S.; Roseiro, J.C. (−)-Agelasidine A from Agelas clathrodes. Z. Naturforsch. 2006, 61, 472–476. [Google Scholar] [CrossRef]
  15. Morales, J.J.; Rodríguez, A.D. (−)-Agelasidine C and (−)-agelasidine D, two new hypotaurocyamine diterpenoids from the Caribbean Sea sponge Agelas clathrodes. J. Nat. Prod. 1992, 55, 389–394. [Google Scholar] [CrossRef] [PubMed]
  16. Cafieri, F.; Fattorusso, E.; Mangoni, A.; Taglialatela-Scafati, O. Clathramides, unique bromopyrrole alkaloids from the Caribbean sponge Agelas clathrodes. Tetrahedron 1996, 52, 13713–13720. [Google Scholar] [CrossRef]
  17. Morales, J.J.; Rodríguez, A.D. The structure of clathrodin, a novel alkaloid isolated from the Caribbean Sea sponge Agelas clathrodes. J. Nat. Prod. 1991, 54, 629–631. [Google Scholar] [CrossRef]
  18. Yang, F.; Ji, R.H.; Li, J.; Gan, J.H. N-containing metabolites from the marine sponge Agelas clathrodes. Nat. Prod. Commun. 2013, 8, 1713–1714. [Google Scholar] [PubMed]
  19. Cafieri, F.; Carnuccio, R.; Fattorusso, E.; Taglialatela-Scafati, O.; Vallefuoco, T. Anti-histaminic activity of bromopyrrole alkaloids isolated from Caribbean Agelas sponges. Bioorg. Med. Chem. Lett. 1997, 7, 2283–2288. [Google Scholar] [CrossRef]
  20. Cafieri, F.; Fattorusso, E.; Mangoni, A.; Taglialatela-Scafati, O. Dispacamides, anti-histamine alkaloids from Caribbean Agelas sponges. Tetrahedron Lett. 1996, 37, 3587–3590. [Google Scholar] [CrossRef]
  21. Assmann, M.; Köck, M. Bromosceptrin, an alkaloid from the marine sponge Agelas conifera. Z. Naturforsch. C 2002, 57, 157–160. [Google Scholar] [CrossRef] [PubMed]
  22. Shen, X.; Perry, T.L.; Dunbar, C.D.; Kelly-Borges, M.; Hamann, M.T. Debromosceptrin, an alkaloid from the Caribbean sponge Agelas conifera. J. Nat. Prod. 1998, 61, 1302–1303. [Google Scholar] [CrossRef] [PubMed]
  23. Keifer, P.A.; Schwartz, R.E.; Koker, M.E.S.; Hughes, R.G.; Rittschof, D.; Rinehart, K. Bioactive bromopyrrole metabolites from the Caribbean sponge Agelas conifera. J. Org. Chem. 1991, 56, 2965–2975. [Google Scholar] [CrossRef]
  24. Kobayashi, J.; Tsuda, M. Ageliferins, potent actomyosin ATPase activators from the Okinawan marine sponge Agelas sp. Tetrahedron 1990, 46, 5579–5586. [Google Scholar] [CrossRef]
  25. La Clair, J.J.; Rodríguez, A.D. Isolation of the β-galactosphingolipid coniferoside using a tumor cell proteome reverse affinity protocol. Bioorg. Med. Chem. 2011, 19, 6645–6653. [Google Scholar] [CrossRef] [PubMed]
  26. Costantino, V.; Fattorusso, E.; Mangoni, A. Glycosyl ceramides from the marine sponge Agelas conifera. Liebigs Ann. 1995, 2, 2133–2136. [Google Scholar] [CrossRef]
  27. D’Ambrosio, M.; Guerriero, A.; Debitus, C.; Ribes, O.; Pusset, J.; Leroy, S.; Pietra, F. Agelastatin A, a new skeleton cytotoxic alkaloid of the oroidin family. Isolation from the Axinellid sponge Agelas dendromorpha of the Coral Sea. J. Chem. Soc. Chem. Commun. 1993, 16, 1305–1306. [Google Scholar] [CrossRef]
  28. Tilvi, S.; Moriou, C.; Martin, M.T.; Gallard, J.F.; Sorres, J.; Patel, K.; Petek, S.; Debitus, C.; Ermolenko, L.; Al-Mourabit, A. Agelastatin E, agelastatin F, and benzosceptrin C from the marine sponge Agelas dendromorpha. J. Nat. Prod. 2010, 73, 720–723. [Google Scholar] [CrossRef] [PubMed]
  29. Piňa, I.C.; White, K.N.; Cabrera, G.; Rivero, E.; Crews, P. Bromopyrrole carboxamide biosynthetic products from the Caribbean sponge Agelas dispar. J. Nat. Prod. 2007, 70, 613–617. [Google Scholar] [CrossRef] [PubMed]
  30. Cafieri, F.; Fattorusso, E.; Taglialatela-scafati, O. Novel bromopyrrole alkaloids from the sponge Agelas dispar. J. Nat. Prod. 1998, 61, 122–125. [Google Scholar] [CrossRef] [PubMed]
  31. Costantino, V.; Fattorusso, E.; Mangoni, A.; DiRosa, M.; Ianaro, A.; Maffia, P. Immunomodulating glycosyl ceramides from the marine sponge Agehs dispar. Tetrahedron 1996, 52, 1573–1578. [Google Scholar] [CrossRef]
  32. Cafieri, F.; Fattorusso, E.; Taglialatela-scafati, O. Novel betaines from the marine sponge Agelas dispar. J. Nat. Prod. 1998, 61, 1171–1173. [Google Scholar] [CrossRef] [PubMed]
  33. Cullen, E.; Devlin, J.P. Agelasine: A novel quaternary 9-methyladenine from the sponge Agelas dispar. Can. J. Chem. 1975, 53, 1690–1691. [Google Scholar] [CrossRef]
  34. Ueoka, R.; Nakao, Y.; Kawatsu, S.; Yaegashi, J.; Matsumoto, Y.; Matsunaga, S.; Furihata, K.; Soest, R.W.M.; Fusetani, N. Gracilioethers A-C, antimalarial metabolites from the marine sponge Agelas gracilis. J. Org. Chem. 2009, 74, 4203–4207. [Google Scholar] [CrossRef] [PubMed]
  35. Hertiani, T.; Edrada-Ebel, R.; Ortlepp, S.; Soest, R.W.M.; Voogd, N.J.; Wray, V.; Hentschel, U.; Kozytska, S.; Müller, W.E.G.; Proksch, P. From anti-fouling to biofilm inhibition: New cytotoxic secondary metabolites from two Indonesian Agelas sponges. Bioorg. Med. Chem. 2010, 18, 1297–1311. [Google Scholar] [CrossRef] [PubMed]
  36. Cafieri, F.; Fattorusso, E.; Mangoni, A.; Taglialatela-scafati, O. A novel bromopyrrole alkaloid from the sponge Agelas longissima with antiserotonergic activity. Bioorg. Med. Chem. Lett. 1995, 5, 799–804. [Google Scholar] [CrossRef]
  37. Cafieri, F.; Fattorusso, E.; Mangoni, A.; Taglialatela-scafati, O. Longamide and 3,7-dimethylisoguanine, two novel alkaloids from the marine sponge Agelas longissima. Tetrahedron Lett. 1995, 36, 7893–7896. [Google Scholar] [CrossRef]
  38. Cafieri, F.; Fattorusso, E.; Mahajnah, Y.; Mangoni, A. Longiside, a novel digalactosylceramide from the Caribbean sponge Agelas longissima. Liebigs Ann. Chem. 1994, 2, 1187–1189. [Google Scholar] [CrossRef]
  39. Cafieri, F.; Fattorusso, E.; Mangoni, A.; Taglialatela-scafati, O. Glycosyl ceramide composition of the marine sponge Agelas longissima. Liebigs Ann. 1995, 1995, 1477–1481. [Google Scholar] [CrossRef]
  40. Hong, L.L.; Sun, J.B.; Yang, F.; Liu, M.; Tang, J.; Sun, F.; Jiao, W.H.; Wang, S.P.; Zhang, W.; Lin, H.W. New diterpene alkaloids from the marine sponge Agelas mauritiana. RSC Adv. 2017, 7, 23970–23976. [Google Scholar] [CrossRef]
  41. Yang, F.; Hamann, M.T.; Zou, Y.; Zhang, M.Y.; Gong, X.B.; Xiao, J.R.; Chen, W.S.; Lin, H.W. Antimicrobial metabolites from the Paracel Islands sponge Agelas mauritiana. J. Nat. Prod. 2012, 75, 774–778. [Google Scholar] [CrossRef] [PubMed]
  42. Fathi-Afshar, R.; Allen, T.M.; Krueger, C.A.; Cook, D.K.; Clanachan, A.S.; Vriend, R.; Baer, H.P. Some pharmacological activities of novel adenine-related compounds isolated from a marine sponge Agelas mauritiana. Can. J. Physiol. Pharmacol. 1989, 67, 276–281. [Google Scholar] [CrossRef] [PubMed]
  43. Nakatsu, T.; Faulkner, D.J.; Matsumoto, G.K.; Clardy, J. Structure of the diterpene portion of a novel base from the sponge Agelas mauritiana. Tetrahedron Lett. 1984, 25, 935–938. [Google Scholar] [CrossRef]
  44. Hattori, T.; Adachi, K.; Shizuri, Y. New agelasine compound from the marine sponge Agelas mauritiana as an antifouling substance against macroalgae. J. Nat. Prod. 1997, 60, 411–413. [Google Scholar] [CrossRef]
  45. Appenzeller, J.; Mihci, G.; Martin, M.T.; Gallard, J.F.; Menou, J.L.; Boury-Esnault, N.; Hooper, J.; Petek, S.; Chevalley, S.; Valentin, A.; et al. Agelasines J, K, and L from the Solomon Islands marine sponge Agelas cf. mauritiana. J. Nat. Prod. 2008, 71, 1451–1454. [Google Scholar] [CrossRef] [PubMed]
  46. Vergne, C.; Appenzeller, J.; Ratinaud, C.; Martin, M.T.; Debitus, C.; Zaparucha, A.; Al-Mourabit, A. Debromodispacamides B and D: Isolation from the marine sponge Agelas mauritiana and stereoselective synthesis using a biomimetic proline route. Org. Lett. 2008, 10, 493–496. [Google Scholar] [CrossRef] [PubMed]
  47. Fathi-Afshar, R.; Allen, T.M. Biologically active metabolites from Agelas mauritiana. Can. J. Chem. 1988, 66, 45–50. [Google Scholar] [CrossRef]
  48. Jiménez, C.; Crews, P. Mauritamide A and accompanying oroidin alkaloids from the sponge Agelas mauritiana. Tetrahedron Lett. 1994, 35, 1375–1378. [Google Scholar] [CrossRef]
  49. Tsukamoto, S.; Kato, H.; Hirota, H.; Fusetani, N. Mauritiamine, a new antifouling oroidin dimer from the marine sponge Agelas mauritiana. J. Nat. Prod. 1996, 59, 501–503. [Google Scholar] [CrossRef]
  50. Schroif-Grégoire, C.; Appenzeller, J.; Debitus, C.; Zaparucha, A.; Al-Mourabit, A. Debromokeramadine from the marine sponge Agelas cf. mauritiana: Isolation and short regioselective and flexible synthesis. Tetrahedron 2015, 71, 3609–3613. [Google Scholar]
  51. Appenzeller, J.; Tilvi, S.; Martin, M.T.; Gallard, J.F.; El-bitar, H.; Dau, E.T.H.; Debitus, C.; Laurent, D.; Moriou, C.; Al-Mourabit, A. Benzosceptrins A and B with a unique benzocyclobutane skeleton and nagelamide S and T from Pacific sponges. Org. Lett. 2009, 11, 4874–4877. [Google Scholar] [CrossRef] [PubMed]
  52. Natori, T.; Koezuka, Y.; Higa, T. Agelasphins, novel α-galactosylceramides from the marine sponge Agelas mauritianus. Tetrahedron Lett. 1993, 34, 5591–5592. [Google Scholar] [CrossRef]
  53. Natori, T.; Morita, M.; Akimoto, K.; Koezuka, Y. Agelasphins, novel antitumor and immunostimulatory cerebrosides from the marine sponge Agelas mauritianus. Tetrahedron 1994, 50, 2771–2784. [Google Scholar] [CrossRef]
  54. Tanaka, Y.; Katayama, T. The structures of isotedanin and isoclathriaxanthin in sea sponge Agelas mauritiana. Bull. Jpn. Soc. Sci. Fish. 1982, 48, 531–533. [Google Scholar] [CrossRef]
  55. Nakamura, H.; Wu, H.; Kobayashi, J.; Kobayashi, M.; Ohizumi, Y.; Hirata, Y. Agelasidines. Novel hypotaurocyamine derivatives from the Okinawan Sea sponge Agelas nakamurai Hoshino. J. Org. Chem. 1985, 50, 2494–2497. [Google Scholar] [CrossRef]
  56. Shoji, N.; Umeyama, A.; Teranaka, M.; Arihara, S. Four novel diterpenoids, including nakamurol A with a unique thelepogane skeleton, from the marine sponge Agelas nakamurai. J. Nat. Prod. 1996, 59, 448–450. [Google Scholar] [CrossRef]
  57. Abdjul, D.B.; Yamazaki, H.; Kanno, S.; Takahashi, O.; Kirikoshi, R.; Ukai, K.; Namikoshi, M. Structures and Biological evaluations of agelasines isolated from the Okinawan marine sponge Agelas nakamurai. J. Nat. Prod. 2015, 78, 1428–1433. [Google Scholar] [CrossRef] [PubMed]
  58. Wu, H.; Nakamura, H.; Kobayashi, J.; Ohizumi, Y. Agelasine-E and -F, novel monocyclic diterpenoids with 9-methyladeninium unit possessing inhibitory effects on Na, K-ATPase isolated from the Okinawan Sea sponge Agelas nakamurai Hoshino. Tetrahedron Lett. 1984, 25, 3719–3722. [Google Scholar] [CrossRef]
  59. Chu, M.J.; Tang, X.L.; Qin, G.F.; Sun, Y.T.; Li, L.; Voogd, N.J.; Li, P.L.; Li, G.Q. Pyrrole derivatives and diterpene alkaloids from the South China Sea sponge Agelas nakamurai. Chem. Biodivers. 2017, 14, e1600446. [Google Scholar] [CrossRef] [PubMed]
  60. Iwagawa, T.; Kaneko, M.; Okamura, H.; Nakatani, M.; Soest, R.W.M. New alkaloids from the Papua New Guinean sponge Agelas nakamurai. J. Nat. Prod. 1998, 61, 1310–1312. [Google Scholar] [CrossRef] [PubMed]
  61. Chu, M.J.; Tang, X.L.; Qin, G.F.; Voogd, N.J.; Li, P.L.; Li, G.Q. Three new non-brominated pyrrole alkaloids from the South China Sea sponge Agelas nakamurai. Chin. Chem. Lett. 2017, 28, 1210–1213. [Google Scholar] [CrossRef]
  62. Tanaka, N.; Kusama, T.; Kashiwada, Y.; Kobayashi, J. Bromopyrrole alkaloids from Okinawan marine sponges Agelas spp. Chem. Pharm. Bull. 2016, 64, 691–694. [Google Scholar] [CrossRef] [PubMed]
  63. Fujita, M.; Nakao, Y.; Matsunaga, S.; Seiki, M.; Itoh, Y.; Yamashita, J.; Soest, R.W.M.; Fusetani, N. Ageladine A: An antiangiogenic matrixmetalloproteinase inhibitor from the marine sponge Agelas nakamurai. J. Am. Chem. Soc. 2003, 125, 15700–15701. [Google Scholar] [CrossRef] [PubMed]
  64. Eder, C.; Proksch, P.; Wray, V.; Soest, R.W.M.; Ferdinandus, E.; Pattisina, L.A.; Sudarsono. New bromopyrrole alkaloids from the Indopacific sponge Agelas nakamurai. J. Nat. Prod. 1999, 62, 1295–1297. [Google Scholar] [CrossRef] [PubMed]
  65. Tsuda, M.; Uemoto, H.; Kobayashi, J. Slagenins A-C, novel bromopyrrole alkaloids from marine sponge Agelas nakamurai. Tetrahedron Lett. 1999, 40, 5709–5712. [Google Scholar] [CrossRef]
  66. Uemoto, H.; Tsuda, M.; Kobayashi, J. Mukanadins A−C, new bromopyrrole alkaloids from marine sponge Agelas nakamurai. J. Nat. Prod. 1999, 62, 1581–1583. [Google Scholar] [CrossRef] [PubMed]
  67. An, L.; Song, W.; Tang, X.; Voogd, N.J.; Wang, Q.; Chu, M.; Li, P.; Li, G. Alkaloids and polyketides from the South China Sea sponge Agelas aff. nemoechinata. RSC Adv. 2017, 7, 14323–14329. [Google Scholar] [CrossRef]
  68. Li, T.; Wang, B.; Voogd, N.J.; Tang, X.L.; Wang, Q.; Chu, M.J.; Li, P.L.; Li, G.Q. Two new diterpene alkaloids from the South China Sea sponge Agelas aff. nemoechinata. Chin. Chem. Lett. 2016, 27, 1048–1051. [Google Scholar] [CrossRef]
  69. Kobayashi, J.; Tsuda, M.; Ohizumi, Y. A potent actomyosin ATPase activator from the Okinawan marine sponge Agelas cf. nemoechinata. Experientia 1991, 47, 301–304. [Google Scholar] [CrossRef] [PubMed]
  70. König, G.M.; Wright, A.D. Agelorins A and B and 11-Epi-fistularin-3, three new antibacterial fistularin-3 derivatives from the tropical marine sponge Agelas oroides. Heterocycles 1993, 36, 1351–1358. [Google Scholar]
  71. König, G.M.; Wright, A.D. Two new naturally occurring pyrrole derivatives from the tropical marine sponge Agelas oroides. Nat. Prod. Lett. 1994, 5, 141–146. [Google Scholar] [CrossRef]
  72. König, G.M.; Wright, A.D. An unprecedented compound from the dichloromethane solubles of the tropical marine sponge Agelas oroides. Planta Med. 1998, 64, 88–89. [Google Scholar] [CrossRef] [PubMed]
  73. Fattorusso, E.; Taglialatela-Scafati, O. Two novel pyrrole-imidazole alkaloids from the Mediterranean sponge Agelas oroides. Tetrahedron Lett. 2000, 41, 9917–9922. [Google Scholar] [CrossRef]
  74. Sauleau, P.; Moriou, C.; Al Mourabit, A. Metabolomics approach to chemical diversity of the Mediterranean marine sponge Agelas oroides. Nat. Prod. Res. 2017, 31, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  75. Forenza, S.; Minale, L.; Riccio, R. New bromo-pyrrole derivatives from the sponge Agelas oroides. Chem. Commun. 1971, 18, 1129–1130. [Google Scholar] [CrossRef]
  76. Giacomo, G.D.; Dini, A.; Falco, B.; Marino, A.; Sica, D. Sterols from the sponge Agelas oroides. Comp. Biochem. Physiol. 1983, 74B, 499–501. [Google Scholar]
  77. Tasdemir, D.; Topaloglu, B.; Perozzo, R.; Brun, R.; O’Neill, R.; Carballeira, N.M.; Zhang, X.; Tonge, P.J.; Linden, A.; Rüedi, P. Marine natural products from the Turkish sponge Agelas oroides that inhibit the enoyl reductases from Plasmodium falciparum, Mycobacterium tuberculosis and Escherichia coli. Bioorg. Med. Chem. 2007, 15, 6834–6845. [Google Scholar] [CrossRef] [PubMed]
  78. Hu, J.F.; Kelly, M.; Hamann, M.T. 26-Nor-25-isopropyl-ergosta-5,7,22E-trien-3β-ol: A new C29 sterol from the sponge Agelas sceptrum from Jamaica. Steroids 2002, 67, 743–747. [Google Scholar] [CrossRef]
  79. Walker, R.P.; Faulkner, D.J. Sceptrin, an antimicrobial agent from the sponge Agelas sceptrum. J. Am. Chem. Soc. 1981, 103, 6772–6773. [Google Scholar] [CrossRef]
  80. Muûoz, J.; Köck, M. Hybrid pyrrole-imidazole alkaloids from the sponge Agelas sceptrum. J. Nat. Prod. 2016, 79, 434–437. [Google Scholar]
  81. Duque, C.; Castillo, G.; Buitrago, S.; Zea, O.O.S. Three monohydroxyl sterols from Agelas schmidti (Wilson, 1902). Rev. Colomb. Quim. 1994, 23, 63–72. [Google Scholar]
  82. Buchecker, R.; Eugster, C.H.; Litchfield, C. Carotenoids from marine sponges (Porifera): Isolation and structure of the seven main carotenoids from Agelas schmidtii. Helv. Chim. Acta 1977, 60, 2780–2788. [Google Scholar] [CrossRef]
  83. Assmann, M.; Zea, S.; Köck, M. Sventrin, a new bromopyrrole alkaloid from the Caribbean sponge Agelas sventres. J. Nat. Prod. 2001, 64, 1593–1595. [Google Scholar] [CrossRef] [PubMed]
  84. Assmann, M.; Lichte, E.; Soest, R.W.M.; Köck, M. New bromopyrrole alkaloid from the marine sponge Agelas wiedenmayeri. Org. Lett. 1999, 1, 455–457. [Google Scholar] [CrossRef]
  85. Fedoreev, S.A.; Ilyin, S.G.; Utkina, N.K.; Maxtmov, O.B.; Reshetnyak, M.V. The structure of dibromoagelaspongin—A novel bromine-containing guanidine derivative from the marine sponge Agelas sp. Tetrahedron 1989, 45, 3487–3492. [Google Scholar] [CrossRef]
  86. Kusama, T.; Tanaka, N.; Sakai, K.; Gonoi, T.; Fromont, J.; Kashiwada, Y.; Kobayashi, J. Agelamadins A and B, dimeric bromopyrrole alkaloids from a marine sponge Agelas sp. Org. Lett. 2014, 16, 3916–3918. [Google Scholar] [CrossRef] [PubMed]
  87. Kusama, T.; Tanaka, N.; Sakai, K.; Gonoi, T.; Fromont, J.; Kashiwada, Y.; Kobayashi, J. Agelamadins C–E, bromopyrrole alkaloids comprising oroidin and 3-hydroxykynurenine from a marine sponge Agelas sp. Org. Lett. 2014, 16, 5176–5179. [Google Scholar] [CrossRef] [PubMed]
  88. Kusama, T.; Tanaka, N.; Kashiwada, Y.; Kobayashi, J. Agelamadin F and tauroacidin E, bromopyrrole alkaloids from an Okinawan marine sponge Agelas sp. Tetrahedron Lett. 2015, 56, 4502–4504. [Google Scholar] [CrossRef]
  89. Endo, T.; Tsuda, M.; Okada, T.; Mitsuhashi, S.; Shima, H.; Kikuchi, K.; Mikami, Y.; Fromont, J.; Kobayashi, J. Nagelamides A–H, new dimeric bromopyrrole alkaloids from marine sponge Agelas Species. J. Nat. Prod. 2004, 67, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
  90. Yasuda, T.; Araki, A.; Kubota, T.; Ito, J.; Mikami, Y.; Fromont, J.; Kobayashi, J. Bromopyrrole alkaloids from marine sponges of the genus Agelas. J. Nat. Prod. 2009, 72, 488–491. [Google Scholar] [CrossRef] [PubMed]
  91. Araki, A.; Kubota, T.; Tsuda, M.; Mikami, Y.; Fromont, J.; Kobayashi, J. Nagelamides K and L, dimeric bromopyrrole alkaloids from sponge Agelas Species. Org. Lett. 2008, 10, 2099–2102. [Google Scholar] [CrossRef] [PubMed]
  92. Kubota, T.; Araki, A.; Ito, J.; Mikami, Y.; Fromont, J.; Kobayashi, J. Nagelamides M and N, new bromopyrrole alkaloids from sponge Agelas species. Tetrahedron 2008, 64, 10810–10813. [Google Scholar] [CrossRef]
  93. Araki, A.; Kubota, T.; Aoyama, K.; Mikami, Y.; Fromont, J.; Kobayashi, J. Nagelamides Q and R, novel dimeric bromopyrrole alkaloids from sponges Agelas sp. Org. Lett. 2009, 11, 1785–1788. [Google Scholar] [CrossRef] [PubMed]
  94. Tanaka, N.; Kusama, T.; Takahashi-Nakaguchi, A.; Gonoi, T.; Fromont, J.; Kobayashi, J. Nagelamides U–W, bromopyrrole alkaloids from a marine sponge Agelas sp. Tetrahedron Lett. 2013, 54, 3794–3796. [Google Scholar] [CrossRef]
  95. Tanaka, N.; Kusama, T.; Takahashi-Nakaguchi, A.; Gonoi, T.; Fromont, J.; Kobayashi, J. Nagelamides X–Z, dimeric bromopyrrole alkaloids from a marine sponge Agelas sp. Org. Lett. 2013, 15, 3262–3265. [Google Scholar] [CrossRef] [PubMed]
  96. Kusama, T.; Tanaka, N.; Takahashi-Nakaguchi, A.; Gonoi, T.; Fromont, J.; Kobayashi, J. Bromopyrrole alkaloids from a marine sponge Agelas sp. Chem. Pharm. Bull. 2014, 62, 499–503. [Google Scholar] [CrossRef] [PubMed]
  97. Nakamura, K.; Kusama, T.; Tanaka, N.; Sakai, K.; Gonoi, T.; Fromont, J.; Kobayashi, J. 2-Debromonagelamide U, 2-debromomukanadin G, and 2-debromonagelamide P from marine sponge Agelas sp. Heterocycles 2015, 90, 425–431. [Google Scholar]
  98. Nakamura, H.; Ohizumi, Y.; Kobayashi, J. Keramadine, a novel antagonist of serotonergic receptors isolated from the Okinawan Sea sponge Agelas sp. Tetrahedron Lett. 1984, 25, 2475–2478. [Google Scholar] [CrossRef]
  99. Ishida, K.; Ishibashi, M.; Shigemori, H.; Sasaki, T.; Kobayashi, J. Agelasine G, a new antileukemic alkaloid from the Okinawan marine sponge Agelas sp. Chem. Pharm. Bull. 1992, 40, 766–767. [Google Scholar] [CrossRef] [PubMed]
  100. Nakamura, H.; Wu, H.; Ohizumi, Y.; Hirata, Y. Agelasine-A, -B, -C and -D, novel bicyclic diterpenoids with a 9-methyladeninium unit possessing inhibitory effects on Na, K-ATPase from the Okinawa Sea sponge Agelas sp. Tetrahedron Lett. 1984, 25, 2989–2992. [Google Scholar] [CrossRef]
  101. Kubota, T.; Iwai, T.; Takahashi-Nakaguchi, A.; Fromont, J.; Gonoi, T.; Kobayashi, J. Agelasines O–U, new diterpene alkaloids with a 9-N-methyladenine unit from a marine sponge Agelas sp. Tetrahedron 2012, 68, 9738–9744. [Google Scholar] [CrossRef]
  102. Fu, X.; Schmiz, F.J.; Tanner, R.S.; Kelly-Borges, M. Agelasines H and I, 9-methyladenine-containing diterpenoids from an Agelas sponge. J. Nat. Prod. 1998, 61, 548–550. [Google Scholar] [CrossRef] [PubMed]
  103. Calcul, L.; Tenney, K.; Ratnam, J.; Mckerrow, J.H.; Crews, P. Structural variations to the 9-N-methyladeninium diterpenoid hybrid commonly isolated from Agelas sponges. Aust. J. Chem. 2010, 63, 915–921. [Google Scholar] [CrossRef]
  104. Capon, R.J.; Faulkner, D.J. Antimicrobial metabolites from a Pacific sponge, Agelas sp. J. Am. Chem. Soc. 1984, 106, 1819–1822. [Google Scholar] [CrossRef]
  105. Tsuda, M.; Yasuda, T.; Fukushi, E.; Kawabata, J.; Sekiguchi, M.; Fromont, J.; Kobayashi, J. Agesamides A and B, bromopyrrole alkaloids from sponge Agelas Species: Application of DOSY for chemical screening of new metabolites. Org. Lett. 2006, 8, 4235–4238. [Google Scholar] [CrossRef] [PubMed]
  106. Kubota, T.; Araki, A.; Yasuda, T.; Tsuda, M.; Fromont, J.; Aoyama, K.; Mikami, Y.; Wälchli, M.R.; Kobayashi, J. Benzosceptrin C, a new dimeric bromopyrrole alkaloid from sponge Agelas sp. Tetrahedron Lett. 2009, 50, 7268–7270. [Google Scholar] [CrossRef]
  107. Araki, A.; Tsuda, M.; Kubota, T.; Mikami, Y.; Fromont, J.; Kobayashi, J. Nagelamide J, a novel dimeric bromopyrrole alkaloid from a sponge Agelas species. Org. Lett. 2007, 9, 2369–2371. [Google Scholar] [CrossRef] [PubMed]
  108. Iwai, T.; Kubota, T.; Fromont, J.; Kobayashi, J. Nagelamide I and 2,2′-didebromonagelamide B, new dimeric bromopyrrole-imidazole alkaloids from a marine sponge Agelas sp. Chem. Pharm. Bull. 2014, 62, 213–216. [Google Scholar] [CrossRef] [PubMed]
  109. Zhu, Y.; Wang, Y.; Gu, B.B.; Yang, F.; Jiao, W.H.; Hu, G.H.; Yu, H.B.; Han, B.N.; Zhang, W.; Shen, Y.; Lin, H.W. Antifungal bromopyrrole alkaloids from the South China Sea sponge Agelas sp. Tetrahedron 2016, 72, 2964–2971. [Google Scholar] [CrossRef]
  110. Sun, Y.T.; Lin, B.; Li, S.G.; Liu, M.; Zhou, Y.J.; Xu, Y.; Hua, H.M.; Lin, H.W. New bromopyrrole alkaloids from the marine sponge Agelas sp. Tetrahedron 2017, 73, 2786–2792. [Google Scholar] [CrossRef]
  111. Assmann, M.; Köck, M. Monobromoisophakellin, a new bromopyrrole alkaloid from the Caribbean sponge Agelas sp. Z. Naturforsch. 2002, 57, 153–156. [Google Scholar] [CrossRef]
  112. Gautschi, J.T.; Whitman, S.; Holman, T.R.; Crews, P. An analysis of phakellin and oroidin structures stimulated by further study of an Agelas sponge. J. Nat. Prod. 2004, 67, 1256–1261. [Google Scholar] [CrossRef] [PubMed]
  113. Abdjul, D.B.; Yamazaki, H.; Kanno, S.I.; Tomizawa, A.; Rotinsulu, H.; Wewengkang, D.S.; Sumilat, D.A.; Ukai, K.; Kapojos, M.M.; Namikoshi, M. An anti-mycobacterial bisfunctionalized sphingolipid and new bromopyrrole alkaloid from the Indonesian marine sponge Agelas sp. J. Nat. Med. 2017, 71, 531–536. [Google Scholar] [CrossRef] [PubMed]
  114. Tada, H.; Tozyo, T. Two bromopyrroles from a marine sponge Agelas sp. Chem. Lett. 1988, 117, 803–804. [Google Scholar] [CrossRef]
  115. Carballeira, N.M.; Emiliano, A. Novel brominated phospholipid fatty acids from the Caribbean sponge Agelas sp. Lipids 1993, 28, 763–766. [Google Scholar] [CrossRef] [PubMed]
  116. Nakamura, H.; Wu, H.; Kobayashi, J.; Ohizumi, Y. Agelasidine-A, a novel sesquiterpene possessing antispasmodic activity from the Okinawa Sea sponge Agelas sp. Tetrahedron Lett. 1983, 24, 4105–4108. [Google Scholar] [CrossRef]
  117. Pettit, G.R.; Xu, J.P.; Gingrich, D.E.; Williams, M.D.; Doubek, D.L.; Chapuis, J.C.; Schmidt, J.M. Isolation and structure of agelagalastatin from the Papua New Guinea marine sponge Agelas sp. Chem. Commun. 1999, 30, 915–916. [Google Scholar] [CrossRef]
  118. Zhang, H.W.; Zhao, Z.P.; Wang, H. Cytotoxic natural products from marine sponge-derived microorganisms. Mar. Drugs 2017, 15, 68. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Photos of Agelas sponges provided by professor Crews.
Figure 1. Photos of Agelas sponges provided by professor Crews.
Marinedrugs 15 00351 g001
Figure 2. Chemical structures of compounds 13.
Figure 2. Chemical structures of compounds 13.
Marinedrugs 15 00351 g002
Figure 3. Chemical structures of compounds 46.
Figure 3. Chemical structures of compounds 46.
Marinedrugs 15 00351 g003
Figure 4. Chemical structures of compounds 7.
Figure 4. Chemical structures of compounds 7.
Marinedrugs 15 00351 g004
Figure 5. Chemical structures of compounds 815.
Figure 5. Chemical structures of compounds 815.
Marinedrugs 15 00351 g005
Figure 6. Chemical structures of compounds 1623.
Figure 6. Chemical structures of compounds 1623.
Marinedrugs 15 00351 g006aMarinedrugs 15 00351 g006b
Figure 7. Chemical structures of compounds 2434.
Figure 7. Chemical structures of compounds 2434.
Marinedrugs 15 00351 g007
Figure 8. Chemical structures of compounds 3545.
Figure 8. Chemical structures of compounds 3545.
Marinedrugs 15 00351 g008
Figure 9. Chemical structures of compounds 4648.
Figure 9. Chemical structures of compounds 4648.
Marinedrugs 15 00351 g009
Figure 10. Chemical structures of compounds 4958.
Figure 10. Chemical structures of compounds 4958.
Marinedrugs 15 00351 g010
Figure 11. Chemical structures of compounds 5961.
Figure 11. Chemical structures of compounds 5961.
Marinedrugs 15 00351 g011
Figure 12. Chemical structures of compounds 6272.
Figure 12. Chemical structures of compounds 6272.
Marinedrugs 15 00351 g012
Figure 13. Chemical structures of compounds 7377.
Figure 13. Chemical structures of compounds 7377.
Marinedrugs 15 00351 g013
Figure 14. Chemical structures of compounds 7892.
Figure 14. Chemical structures of compounds 7892.
Marinedrugs 15 00351 g014aMarinedrugs 15 00351 g014b
Figure 15. Chemical structures of compounds 93102.
Figure 15. Chemical structures of compounds 93102.
Marinedrugs 15 00351 g015
Figure 16. Chemical structures of compounds 103112.
Figure 16. Chemical structures of compounds 103112.
Marinedrugs 15 00351 g016
Figure 17. Chemical structures of compounds 113128.
Figure 17. Chemical structures of compounds 113128.
Marinedrugs 15 00351 g017aMarinedrugs 15 00351 g017b
Figure 18. Chemical structures of compounds 129145.
Figure 18. Chemical structures of compounds 129145.
Marinedrugs 15 00351 g018
Figure 19. Chemical structures of compounds 146153.
Figure 19. Chemical structures of compounds 146153.
Marinedrugs 15 00351 g019
Figure 20. Chemical structures of compounds 154189.
Figure 20. Chemical structures of compounds 154189.
Marinedrugs 15 00351 g020
Figure 21. Chemical structures of compounds 190193.
Figure 21. Chemical structures of compounds 190193.
Marinedrugs 15 00351 g021
Figure 22. Chemical structures of compounds 194200.
Figure 22. Chemical structures of compounds 194200.
Marinedrugs 15 00351 g022
Figure 23. Chemical structure of compounds 201.
Figure 23. Chemical structure of compounds 201.
Marinedrugs 15 00351 g023
Figure 24. Chemical structure of compounds 202.
Figure 24. Chemical structure of compounds 202.
Marinedrugs 15 00351 g024
Figure 25. Chemical structures of compounds 203210.
Figure 25. Chemical structures of compounds 203210.
Marinedrugs 15 00351 g025aMarinedrugs 15 00351 g025b
Figure 26. Chemical structures of compounds 211231.
Figure 26. Chemical structures of compounds 211231.
Marinedrugs 15 00351 g026aMarinedrugs 15 00351 g026b
Figure 27. Chemical structures of compounds 232241.
Figure 27. Chemical structures of compounds 232241.
Marinedrugs 15 00351 g027aMarinedrugs 15 00351 g027b
Figure 28. Chemical structures of compounds 242260.
Figure 28. Chemical structures of compounds 242260.
Marinedrugs 15 00351 g028aMarinedrugs 15 00351 g028b
Figure 29. Chemical structures of compounds 261289.
Figure 29. Chemical structures of compounds 261289.
Marinedrugs 15 00351 g029aMarinedrugs 15 00351 g029b
Figure 30. Chemical structures of compounds 290 and 291.
Figure 30. Chemical structures of compounds 290 and 291.
Marinedrugs 15 00351 g030
Table 1. Agelas-derived secondary metabolites.
Table 1. Agelas-derived secondary metabolites.
OrganismLocalitySecondary MetaboliteReferences
Agelas axiferathe Republic of Palauaxistatins 1 (1), 2 (2), 3 (3)[4]
A. cerebrumCaribbean5-bromopyrrole-2-carboxylic acid (4), 4-bromopyrrole-2-carboxylic acid (5), 3,4-bromopyrrole-2-carboxylic acid (6)[6]
A. ceylonicathe Mandapam coasthanishin (7)[7]
A. citrinaCaribbean(−)-agelasidine E (8), (−)-agelasidine F (9), agelasine N (10), citrinamines A–D (1114), N-methylagelongine (15)[9,10]
A. clathrodesGrand Bahamas Islandclarhamnoside (16)[11]
Caribbeanclathrosides A–C (1719), isoclathrosides A–C (2022), glycosphingolipid (23), (−)-agelasidine A (24), (−)-agelasidine C (25), (−)-agelasidine D (26), clathramides A (27) and B (28), clathrodin (29), dispacamides A–D (3134)[12,13,14,15,16,17,19,20]
South China Sea3,3-bis(uracil-l-yl)-propan-1-aminium (30)[18]
A. coniferaFlorida Keysbromosceptrin (35)[21]
Belizedebromosceptrin (36)[22]
Caribbeanbromopyrroles (3743), glycosphingolipid (45)[23,24,26]
Puerto Ricoconiferoside (44)[25]
A. dendromorphathe Coral Seaagelastatin A (46)[27]
the New Caledoniaagelastatins E (47) and F (48)[28]
A. disparCaribbeandispyrin (49), dibromoagelaspongin methyl ether (50), longamide B (51), clathramides C (52) and D (53), aminozooanemonin (55), pyridinebetaines A (56) and B (57)[29,30,32]
San Salvador Islandtriglycosylceramide (54)[31]
agelasine (58)[33]
A. gracilisSouth Japangracilioethers A–C (5961)[34]
A. linnaeiIndonesiabrominated pyrrole derivatives (6272)[35]
A. longissimaCaribbeanagelongine (73), 3,7-dimethylisoguanine (74), longamide (75), glycosphingolipids (76 and 77)[36,37,38,39]
A. mauritianaSouth China Sea(−)-80-oxo-agelasine B (78), (+)-agelasine B (79), (+)-8’-oxo-agelasine C (80), agelasine V (81), (+)-8’-oxo-agelasine E (82), 8’-oxo-agelasine D (83), ageloxime B (84), (+)-2-oxo-agelasidine C (85), 4-bromo-N-(butoxymethyl)-1H-pyrrole-2-carboxamide (95)[40,41]
Enewetakagelasimine A (86), agelasimine B (87), purino-diterpene (88), 5-debromomidpacamide (96)[42,43,47]
Pohnpeiepi-agelasine C (89)[44]
Solomon Islandsagelasines J (90), K (91) and L (92), debromodispacamides B (93) and D (94)[45,46]
Fijimauritamide A (97)[48]
Hachijo-jima Islandmauritiamine (98)[49]
the Pacific seaebromokeramadine (99), benzosceptrin A (100), nagelamides S (101) and T (102)[50,51]
Okinawaagelasphins (103110)[52,53]
Kagoshimaisotedanin (111), isoclathriaxanthin (112)[54]
A. nakamuraiOkinawaagelasidines B (113) and C (114), nakamurols A–D (115118), 2-oxoagelasiines A (119) and F (120), 10-hydro-9-hydroxyagelasine F (121), agelasines E (122) and F (123), slagenins A–C (140142), mukanadins A–C (143145)[55,56,57,58,65,66]
Indonesia(−)-agelasine D (124), (−)-ageloxime D (125)[35]
South China Seaisoagelasine C (126), isoagelasidine B (127)[59]
Papua New Guineaditerpene (128), bromopyrrole alkaloids (134 and 135)[60]
South China Seanakamurines A–E (129133)[59,61]
Japanageladine A (136)[63]
Indonesialongamide C (137)[35]
Indopacificnakamuric acid (138) and its methyl ester (139)[64]
A. nemoechinataSouth China Seanemoechines A–D (146149), nemoechioxide A (150), nemoechines F (151) and G (152)[67,68]
Okinawaoxysceptrin (153)[69]
A. oroidesthe Great Barrier Reefagelorin A (154), agelorin B (155), 11-epi-fistularin-3 (156), pyrrole-2-carboxamide (157), N-formyl-pymole-2-carboxamid (158), 2,4,6,6-tetramethyl-3(6H)-pyridone (159)[70,71,72]
Mediterranea Seacyclooroidin (160) and taurodispacamide A (161), monobromoagelaspongin (162), (−)-equinobetaine B (163)[73,74]
Naplesbromopyrroles (164168), sterols (169183)[75,76]
the Northern Aegean Sea3-amino-1-(2-aminoimidazoyl)-prop-1-ene (184), taurine (185), fatty acid mixtures (186189)[77]
A. sceptrumJamaica26-nor-25-isopropyl-ergosta-5,7,22 E-trien-3β-ol (190)[78]
Belizesceptrin (191)[79]
Bahamas15′-oxoadenosceptrin (192), decarboxyagelamadin C (193)[80]
A. schmidtiiCaribbeanmonohydroxyl sterols (194196)[81]
West Indiesα-carotene (197), isorenieratene (198), trikentriorhodin (199) and zeaxanthin (200)[82]
A. sventresCaribbeansventrin (201)[83]
A. wiedenmayeriFlorida Keys4-bromopyrrole-2-carboxyhomoarginine (202)[84]
Unclassified Agelas sp.No recorddibromoagelaspongin hydrochloride (203)[85]
Okinawaagelamadins A (204) and B (205), agelamadins C–F (206209), tauroacidin E (210), nagelamides A–H (211218), nagelamides K–O (219223), nagelamides Q (224) and R (225), nagelamides U–Z (226231), 2-bromokeramadine (232), 2-bromo-9,10-dihydrokeramadine (233), tauroacidins C (234) and D (235), mukanadin G (236), 2-debromonagelamides U (237) and G (238), 2-debromonagelamide P (239), keramadine (240), agelasine G (241), agelasines A–D (242245), agelasines O–U (246252), agesamides A (261) and B (262), benzosceptrin C (263), nagelamides J (264) and P (265), mukanadins E (266) and F (267), nagelamide I (268), 2,2’-didebromonagelamide B (269), agelasidine A (290)[86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,105,106,107,108,116]
Yap Islandagelasines H (253) and I (254)[102]
Papua New Guineaagelasine M (255), 2-oxo-agelasine B (256), gelasines A (257) and B (258), (−)-7-N-methyldibromophakellin (281), (−)-7-N-methylmonobromophakellin (282), agelagalastatin (291)[103,112,117]
Palau Islandagelines A (259) and B (260)[104]
South China Sealongamides D–F (270272), 3-oxethyl-4-[1-(4,5-dibromopyrrole-2-yl)-formamido]-butanoic acid methyl ester (273), 2-oxethyl-3-[1-(4,5-dibromopyrrole-2-yl)-formamido]-methyl propionate (274), 9-oxethyl-mukanadin F (275), hexazosceptrin (276), agelestes A (277) and B (278) and (9S,10R,9’S,10’R)-nakamuric acid (279)[109,110]
Caribbean Seamonobromoisophakellin (280), brominated phospholipid fatty acids (286289)[111,115]
Indonesia5-bromophakelline (283)[113]
No record2,3-dibromopyrrole (284) and 2,3-dibromo-5-methoxymethylpyrrole (285)[114]

Share and Cite

MDPI and ACS Style

Zhang, H.; Dong, M.; Chen, J.; Wang, H.; Tenney, K.; Crews, P. Bioactive Secondary Metabolites from the Marine Sponge Genus Agelas. Mar. Drugs 2017, 15, 351. https://doi.org/10.3390/md15110351

AMA Style

Zhang H, Dong M, Chen J, Wang H, Tenney K, Crews P. Bioactive Secondary Metabolites from the Marine Sponge Genus Agelas. Marine Drugs. 2017; 15(11):351. https://doi.org/10.3390/md15110351

Chicago/Turabian Style

Zhang, Huawei, Menglian Dong, Jianwei Chen, Hong Wang, Karen Tenney, and Phillip Crews. 2017. "Bioactive Secondary Metabolites from the Marine Sponge Genus Agelas" Marine Drugs 15, no. 11: 351. https://doi.org/10.3390/md15110351

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