1
|
Sinane M, Grunberger C, Gentile L, Moriou C, Chaker V, Coutrot P, Guenneguez A, Poullaouec MA, Connan S, Stiger-Pouvreau V, Zubia M, Fleury Y, Cérantola S, Kervarec N, Al-Mourabit A, Petek S, Voisset C. Potential of Marine Sponge Metabolites against Prions: Bromotyrosine Derivatives, a Family of Interest. Mar Drugs 2024; 22:456. [PMID: 39452864 PMCID: PMC11509309 DOI: 10.3390/md22100456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
The screening of 166 extracts from tropical marine organisms (invertebrates, macroalgae) and 3 cyclolipopeptides from microorganisms against yeast prions highlighted the potential of Verongiida sponges to prevent the propagation of prions. We isolated the known compounds purealidin Q (1), aplysamine-2 (2), pseudoceratinine A (3), aerophobin-2 (4), aplysamine-1 (5), and pseudoceratinine B (6) for the first time from the Wallisian sponge Suberea laboutei. We then tested compounds 1-6 and sixteen other bromotyrosine and bromophenol derivatives previously isolated from Verongiida sponges against yeast prions, demonstrating the potential of 1-3, 5, 6, aplyzanzine C (7), purealidin A (10), psammaplysenes D (11) and F (12), anomoian F (14), and N,N-dimethyldibromotyramine (15). Following biological tests on mammalian cells, we report here the identification of the hitherto unknown ability of the six bromotyrosine derivatives 1, 2, 5, 7, 11, and 14 of marine origin to reduce the spread of the PrPSc prion and the ability of compounds 1 and 2 to reduce endoplasmic reticulum stress. These two biological activities of these bromotyrosine derivatives are, to our knowledge, described here for the first time, offering a new therapeutic perspective for patients suffering from prion diseases that are presently untreatable and consequently fatal.
Collapse
Affiliation(s)
- Maha Sinane
- Univ Brest, Inserm, EFS, UMR 1078, GGB, School of Medicine, F-29200 Brest, France; (M.S.); (L.G.); (V.C.); (P.C.)
| | - Colin Grunberger
- IRD, CNRS, Ifremer, Univ Brest, LEMAR, IUEM, F-29280 Plouzane, France; (C.G.); (A.G.); (M.-A.P.); (S.C.); (V.S.-P.)
| | - Lucile Gentile
- Univ Brest, Inserm, EFS, UMR 1078, GGB, School of Medicine, F-29200 Brest, France; (M.S.); (L.G.); (V.C.); (P.C.)
| | - Céline Moriou
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, F-91190 Gif-sur-Yvette, France; (C.M.); (A.A.-M.)
| | - Victorien Chaker
- Univ Brest, Inserm, EFS, UMR 1078, GGB, School of Medicine, F-29200 Brest, France; (M.S.); (L.G.); (V.C.); (P.C.)
| | - Pierre Coutrot
- Univ Brest, Inserm, EFS, UMR 1078, GGB, School of Medicine, F-29200 Brest, France; (M.S.); (L.G.); (V.C.); (P.C.)
| | - Alain Guenneguez
- IRD, CNRS, Ifremer, Univ Brest, LEMAR, IUEM, F-29280 Plouzane, France; (C.G.); (A.G.); (M.-A.P.); (S.C.); (V.S.-P.)
| | - Marie-Aude Poullaouec
- IRD, CNRS, Ifremer, Univ Brest, LEMAR, IUEM, F-29280 Plouzane, France; (C.G.); (A.G.); (M.-A.P.); (S.C.); (V.S.-P.)
| | - Solène Connan
- IRD, CNRS, Ifremer, Univ Brest, LEMAR, IUEM, F-29280 Plouzane, France; (C.G.); (A.G.); (M.-A.P.); (S.C.); (V.S.-P.)
| | - Valérie Stiger-Pouvreau
- IRD, CNRS, Ifremer, Univ Brest, LEMAR, IUEM, F-29280 Plouzane, France; (C.G.); (A.G.); (M.-A.P.); (S.C.); (V.S.-P.)
| | - Mayalen Zubia
- UPF, Ifremer, ILM, IRD, UMR 241 SECOPOL, BP6570, 98702 Faa’a, Tahiti, French Polynesia;
| | - Yannick Fleury
- Univ Brest, Univ Bretagne Sud, CNRS, LBCM, EMR 6076, F-29000 Quimper, France;
| | | | - Nelly Kervarec
- Univ Brest, Plateforme Spectrométrie de Masse, F-29238 Brest, France;
| | - Ali Al-Mourabit
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, F-91190 Gif-sur-Yvette, France; (C.M.); (A.A.-M.)
| | - Sylvain Petek
- IRD, CNRS, Ifremer, Univ Brest, LEMAR, IUEM, F-29280 Plouzane, France; (C.G.); (A.G.); (M.-A.P.); (S.C.); (V.S.-P.)
| | - Cécile Voisset
- Univ Brest, Inserm, EFS, UMR 1078, GGB, School of Medicine, F-29200 Brest, France; (M.S.); (L.G.); (V.C.); (P.C.)
- Univ Brest, Inserm, UMR 1101, LaTIM, School of Medicine, F-29200 Brest, France
| |
Collapse
|
2
|
Ali ML, Noushin F, Azme E, Hasan MM, Hoque N, Metu AF. Marine natural compounds as potential CBP bromodomain inhibitors for treating cancer: an in-silico approach using molecular docking, ADMET, molecular dynamics simulations and MM-PBSA binding free energy calculations. In Silico Pharmacol 2024; 12:85. [PMID: 39310674 PMCID: PMC11411048 DOI: 10.1007/s40203-024-00258-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 09/08/2024] [Indexed: 09/25/2024] Open
Abstract
The cAMP-responsive element binding protein (CREB) binding protein (CBP), a bromodomain-containing protein, engages with multiple transcription factors and enhances the activation of many genes. CBP bromodomain acts as an epigenetic reader and plays an important role in the CBP-chromatin interaction which makes it an important drug target for treating many diseases. Though inhibiting CBP bromodomain was reported to have great potential in cancer therapeutics, approved CBP bromodomain inhibitor is yet to come. We utilized various in silico approaches like molecular docking, ADMET, molecular dynamics (MD) simulations, MM-PBSA calculations, and in silico PASS predictions to identify potential CBP bromodomain inhibitors from marine natural compounds as they have been identified as having distinctive chemical structures and greater anticancer activities. To develop a marine natural compound library for this investigation, Lipinski's rule of five was used. Sequential investigations utilizing molecular docking, ADMET studies, 100 ns MD simulations, and MM-PBSA calculations revealed that three marine compounds-ascididemin, neoamphimedine, and stelletin A-demonstrated superior binding affinity compared to the standard inhibitor, 69 A. These compounds also exhibited suitable drug-like properties, a favorable safety profile, and formed stable protein-ligand complexes. The in-silico PASS tool predicted that these compounds have significant potential for anticancer activity. Among them, ascididemin demonstrated the highest binding affinity in both molecular docking and MM-PBSA calculations, as well as a better stability profile in MD simulations. Hence, ascididemin can be a potential inhibitor of CBP bromodomain. However, in vitro and in vivo validation is required for further confirmation of these findings. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00258-5.
Collapse
Affiliation(s)
- Md. Liakot Ali
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Fabiha Noushin
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Eva Azme
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Md. Mahmudul Hasan
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Neamul Hoque
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| | - Afroz Fathema Metu
- Department of Pharmacy, Faculty of Biological Science, University of Chittagong, Chittagong, 4331 Bangladesh
| |
Collapse
|
3
|
Dukes DM, Atanassov VK, Smith JM. Enantioselective total synthesis of (+)-cylindricine B. Chem Sci 2024:d4sc04910a. [PMID: 39309100 PMCID: PMC11409986 DOI: 10.1039/d4sc04910a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
This article describes the first enantioselective synthesis of the Tasmanian marine alkaloid (+)-cylindricine B. The concise construction of the compound hinged on dearomative retrosynthetic logic combined with a tactical advance in the generation of congested, cyclic, alpha-tertiary amine centers. The scope of this key coupling reaction was explored in addition to providing a synthetic application for Cu-catalyzed enantioselective dearomatization of N-acyl-pyridiniums. The synthesis proceeds in five or six steps from commercially available starting materials.
Collapse
Affiliation(s)
- Dallas M Dukes
- Florida State University, Department of Chemistry and Biochemistry, Laboratories of Molecular Recognition 95 Chieftan Way Tallahassee FL 32306 USA
| | - Victor K Atanassov
- Florida State University, Department of Chemistry and Biochemistry, Laboratories of Molecular Recognition 95 Chieftan Way Tallahassee FL 32306 USA
| | - Joel M Smith
- Florida State University, Department of Chemistry and Biochemistry, Laboratories of Molecular Recognition 95 Chieftan Way Tallahassee FL 32306 USA
| |
Collapse
|
4
|
Surwase AJ, Thakur NL. Production of marine-derived bioactive peptide molecules for industrial applications: A reverse engineering approach. Biotechnol Adv 2024; 77:108449. [PMID: 39260778 DOI: 10.1016/j.biotechadv.2024.108449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 06/28/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
This review examines a wide range of marine microbial-derived bioactive peptide molecules, emphasizing the significance of reverse engineering in their production. The discussion encompasses the advancements in Marine Natural Products (MNPs) bio-manufacturing through the integration of omics-driven microbial engineering and bioinformatics. The distinctive features of non-ribosomally synthesised peptides (NRPs), and ribosomally synthesised precursor peptides (RiPP) biosynthesis is elucidated and presented. Additionally, the article delves into the origins of common peptide modifications. It highlights various genome mining approaches for the targeted identification of Biosynthetic Gene Clusters (BGCs) and novel RiPP and NRPs-derived peptides. The review aims to demonstrate the advancements, prospects, and obstacles in engineering both RiPP and NRP biosynthetic pathways.
Collapse
Affiliation(s)
- Akash J Surwase
- CSIR-National Institute of Oceanography, Dona Paula 403004, Goa, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Narsinh L Thakur
- CSIR-National Institute of Oceanography, Dona Paula 403004, Goa, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Zhang ZL, Xu HN, Gong CM, Li YZ, Li YM, Song XM, Wang R, Zhang DD. The Sources, Structures and Cytotoxicity of Animal-Derived Bisindole Compounds. Chem Biodivers 2024; 21:e202401165. [PMID: 38973453 DOI: 10.1002/cbdv.202401165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/09/2024]
Abstract
Bisindole compounds constitute a significant class of natural compounds distinguished by their characteristic bisindole structure and renowned for their anticancer properties. Over the past four decades, researchers have isolated 229 animal-derived bisindole compounds (ADBCs) from various animals. These compounds demonstrate a wide range of pharmacological properties, including cytotoxicity, antibacterial, antifungal, antiviral, and other activities. Notably, among these activities, cytotoxicity emerges as the most prominent characteristic of ADBCs. This review also summarizes the structureactivity relationship (SAR) studies associated with the cytotoxicity of these compounds and explores the druggability of these compounds. In summary, our objective is to provide an overview of the research progress concerning ADBCs, with the aim of fostering their continued development and utilization.
Collapse
Affiliation(s)
- Zi-Long Zhang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, P.R. China
| | - Hao-Nan Xu
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, P.R. China
| | - Chuan-Ming Gong
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, P.R. China
| | - Yu-Ze Li
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, P.R. China
| | - Yi-Ming Li
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Xiao-Mei Song
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, P.R. China
| | - Rui Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Dong-Dong Zhang
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, P.R. China
| |
Collapse
|
6
|
Smallwood TB, Krumpe LRH, Payne CD, Klein VG, O'Keefe BR, Clark RJ, Schroeder CI, Rosengren KJ. Picking the tyrosine-lock: chemical synthesis of the tyrosyl-DNA phosphodiesterase I inhibitor recifin A and analogues. Chem Sci 2024; 15:13227-13233. [PMID: 39183914 PMCID: PMC11339948 DOI: 10.1039/d4sc01976h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/11/2024] [Indexed: 08/27/2024] Open
Abstract
The peptide recifin A is the inaugural member of the structurally intriguing new fold referred to as a tyrosine-lock. Its central four stranded β-sheet is stabilized by a unique arrangement in which three disulfide bonds and their interconnecting backbone form a ring that wraps around one of the strands, resulting in a Tyr side chain being buried in the molecular core. Here we aimed to establish a synthetic route to this complex class of natural products. Full length recifin A was successfully generated through native chemical ligation chemistry joining two 21 amino acid residue fragments. Surprisingly, reduced linear recifin A readily adopts the correct, topologically-complex fold via random oxidation of the cysteines, suggesting it is highly energetically favored. Utilizing our synthetic strategy, we generated five recifin A analogues to investigate the structural role of the central Tyr residue and provide the first insights into the structure activity relationship of recifin A towards its cancer target tyrosyl-DNA phosphodiesterase I.
Collapse
Affiliation(s)
- Taylor B Smallwood
- The University of Queensland, School of Biomedical Sciences Brisbane QLD 4072 Australia
| | - Lauren R H Krumpe
- Molecular Targets Program, Centre for Cancer Research, National Cancer Institute, National Institute of Health Frederick MD 21702 USA
| | - Colton D Payne
- The University of Queensland, School of Biomedical Sciences Brisbane QLD 4072 Australia
| | - Victoria G Klein
- Chemical Biology Laboratory, Centre for Cancer Research, National Cancer Institute, National Institute of Health Frederick MD 21702 USA
| | - Barry R O'Keefe
- Molecular Targets Program, Centre for Cancer Research, National Cancer Institute, National Institute of Health Frederick MD 21702 USA
- Natural Products Branch, Centre for Cancer Research, National Cancer Institute, National Institute of Health Frederick MD 21702 USA
| | - Richard J Clark
- The University of Queensland, School of Biomedical Sciences Brisbane QLD 4072 Australia
| | - Christina I Schroeder
- Chemical Biology Laboratory, Centre for Cancer Research, National Cancer Institute, National Institute of Health Frederick MD 21702 USA
- Peptide Therapeutics, Genentech Inc 1 DNA Way South San Francisco CA 94080 USA
| | - K Johan Rosengren
- The University of Queensland, School of Biomedical Sciences Brisbane QLD 4072 Australia
| |
Collapse
|
7
|
Shen SM, Yu DD, Ke LM, Yao LG, Su MZ, Guo YW. Polyoxygenated cembrane-type diterpenes from the Hainan soft coral Lobophytum crassum as a promising source of anticancer agents with ErbB3 and ROR1 inhibitory potential. Acta Pharmacol Sin 2024:10.1038/s41401-024-01347-z. [PMID: 39075227 DOI: 10.1038/s41401-024-01347-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
A detailed chemical investigation of the Hainan soft coral Lobophytum crassum led to the identification of a class of polyoxygenated cembrane-type macrocyclic diterpenes (1-28), including three new flexible cembranoids, lobophycrasins E-G (2-4), and twenty-five known analogues. Their structures were elucidated by combining extensive spectroscopic data analysis, quantum mechanical-nuclear magnetic resonance (QM-NMR) methods, the modified Mosher's method, X-ray diffraction analysis, and comparison with data reported in the literature. Bioassays revealed that sixteen cembranoids inhibited the proliferation of H1975, MDA-MB231, A549, and H1299 cells. Among them, Compounds 10, 17, and 20 exhibited significant antiproliferative activities with IC50 values of 1.92-8.82 μM, which are very similar to that of the positive control doxorubicin. Molecular mechanistic studies showed that the antitumour activity of Compound 10 was closely related to regulation of the ROR1 and ErbB3 signalling pathways. This study may provide insight into the discovery and utilization of marine macrocyclic cembranoids as lead compounds for anticancer drugs.
Collapse
Affiliation(s)
- Shou-Mao Shen
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China
- School of Pharmacy, Yancheng Teachers University, Yancheng, 224002, China
| | - Dan-Dan Yu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Lin-Mao Ke
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Li-Gong Yao
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Ming-Zhi Su
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Yue-Wei Guo
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
8
|
Im JW, Lim JH, Stonik VA, Kwak JY, Jin S, Son M, Bae HR. Stichoposide C and Rhizochalin as Potential Aquaglyceroporin Modulators. Mar Drugs 2024; 22:335. [PMID: 39195451 DOI: 10.3390/md22080335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Aquaporins (AQPs) are a family of integral membrane proteins that selectively transport water and glycerol across the cell membrane. Because AQPs are involved in a wide range of physiological functions and pathophysiological conditions, AQP-based therapeutics may have the broad potential for clinical utility, including for disorders of water and energy balance. However, AQP modulators have not yet been developed as suitable candidates for clinical applications. In this study, to identify potential modulators of AQPs, we screened 31 natural products by measuring the water and glycerol permeability of mouse erythrocyte membranes using a stopped-flow light scattering method. None of the tested natural compounds substantially affected the osmotic water permeability. However, several compounds considerably affected the glycerol permeability. Stichoposide C increased the glycerol permeability of mouse erythrocyte membranes, whereas rhizochalin decreased it at nanomolar concentrations. Immunohistochemistry revealed that AQP7 was the main aquaglyceroporin in mouse erythrocyte membranes. We further verified the effects of stichoposide C and rhizochalin on aquaglyceroporins using human AQP3-expressing keratinocyte cells. Stichoposide C, but not stichoposide D, increased AQP3-mediated transepithelial glycerol transport, whereas the peracetyl aglycon of rhizochalin was the most potent inhibitor of glycerol transport among the tested rhizochalin derivatives. Collectively, stichoposide C and the peracetyl aglycon of rhizochalin might function as modulators of AQP3 and AQP7, and suggests the possibility of these natural products as potential drug candidates for aquaglyceroporin modulators.
Collapse
Affiliation(s)
- Ji Woo Im
- Department of Physiology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Ju Hyun Lim
- Department of Physiology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Valentin A Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Jong-Young Kwak
- Department of Pharmacology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea
| | - Songwan Jin
- Department of Mechanical Engineering, Tech University of Korea, Siheung-si 15073, Gyeonggi-do, Republic of Korea
| | - Minkook Son
- Department of Physiology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Hae-Rahn Bae
- Department of Physiology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| |
Collapse
|
9
|
Buljan A, Stepanić V, Čikoš A, Babić Brčić S, Bojanić K, Roje M. Total Synthesis and Biological Profiling of Putative (±)-Marinoaziridine B and (±)- N-Methyl Marinoaziridine A. Mar Drugs 2024; 22:310. [PMID: 39057419 PMCID: PMC11278217 DOI: 10.3390/md22070310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The total synthesis of two new marine natural products, (±)-marinoaziridine B 7 and (±)-N-methyl marinoaziridine A 8, was accomplished. The (±)-marinoaziridine 7 was prepared in a six-step linear sequence with a 2% overall yield. The key steps in our strategy were the preparation of the chiral epoxide (±)-5 using the Johnson Corey Chaykovsky reaction, followed by the ring-opening reaction and the Staudinger reaction. The N,N-dimethylation of compound (±)-7 gives (±)-N-methyl marinoaziridine A 8. The NMR spectra of synthetized (±)-marinoaziridine B 7 and isolated natural product did not match. The compounds are biologically characterized using relevant in silico, in vitro and in vivo methods. In silico ADMET and bioactivity profiling predicted toxic and neuromodulatory effects. In vitro screening by MTT assay on three cell lines (MCF-7, H-460, HEK293T) showed that both compounds exhibited moderate to strong antiproliferative and cytotoxic effects. Antimicrobial tests on bacterial cultures of Escherichia coli and Staphylococcus aureus demonstrated the dose-dependent inhibition of the growth of both bacteria. In vivo toxicological tests were performed on zebrafish Danio rerio and showed a significant reduction of zebrafish mortality due to N-methylation in (±)-8.
Collapse
Affiliation(s)
- Anđela Buljan
- Laboratory for Chiral Technologies, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Višnja Stepanić
- Laboratory for Machine Learning and Knowledge Representation, Division of Electronics, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Ana Čikoš
- NMR Centre, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Sanja Babić Brčić
- Laboratory for Aquaculture Biotechnology, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (S.B.B.); (K.B.)
| | - Krunoslav Bojanić
- Laboratory for Aquaculture Biotechnology, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (S.B.B.); (K.B.)
| | - Marin Roje
- Laboratory for Chiral Technologies, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| |
Collapse
|
10
|
Lee J, Choi JH, Lee J, Cho E, Lee YJ, Lee HS, Oh KB. Halenaquinol Blocks Staphylococcal Protein A Anchoring on Cell Wall Surface by Inhibiting Sortase A in Staphylococcus aureus. Mar Drugs 2024; 22:266. [PMID: 38921577 PMCID: PMC11204543 DOI: 10.3390/md22060266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Sortase A (SrtA) is a cysteine transpeptidase that binds to the periplasmic membrane and plays a crucial role in attaching surface proteins, including staphylococcal protein A (SpA), to the peptidoglycan cell wall. Six pentacyclic polyketides (1-6) were isolated from the marine sponge Xestospongia sp., and their structures were elucidated using spectroscopic techniques and by comparing them to previously reported data. Among them, halenaquinol (2) was found to be the most potent SrtA inhibitor, with an IC50 of 13.94 μM (4.66 μg/mL). Semi-quantitative reverse transcription PCR data suggest that halenaquinol does not inhibit the transcription of srtA and spA, while Western blot analysis and immunofluorescence microscopy images suggest that it blocks the cell wall surface anchoring of SpA by inhibiting the activity of SrtA. The onset and magnitude of the inhibition of SpA anchoring on the cell wall surface in S. aureus that has been treated with halenaquinol at a value 8× that of the IC50 of SrtA are comparable to those for an srtA-deletion mutant. These findings contribute to the understanding of the mechanism by which marine-derived pentacyclic polyketides inhibit SrtA, highlighting their potential as anti-infective agents targeting S. aureus virulence.
Collapse
Affiliation(s)
- Jaepil Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Jae-Hyeong Choi
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jayho Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Eunji Cho
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Yeon-Ju Lee
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Hyi-Seung Lee
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Ki-Bong Oh
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| |
Collapse
|
11
|
Shama SM, Elissawy AM, Salem MA, Youssef FS, Elnaggar MS, El-Seedi HR, Khalifa SAM, Briki K, Hamdan DI, Singab ANB. Comparative metabolomics study on the secondary metabolites of the red alga, Corallina officinalis and its associated endosymbiotic fungi. RSC Adv 2024; 14:18553-18566. [PMID: 38903055 PMCID: PMC11187739 DOI: 10.1039/d4ra01055h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Marine endosymbionts have gained remarkable interest in the last three decades in terms of natural products (NPs) isolated thereof, emphasizing the chemical correlations with those isolated from the host marine organism. The current study aimed to conduct comparative metabolic profiling of the marine red algae Corallina officinalis, and three fungal endosymbionts isolated from its inner tissues namely, Aspergillus nidulans, A. flavipes and A. flavus. The ethyl acetate (EtOAc) extracts of the host organism as well as the isolated endosymbionts were analyzed using ultra-high performance liquid chromatography coupled to high resolution tandem mass spectrometry (UHPLC-MS/MS)in both positive and negative ion modes, applying both full scan (FS) and all ion fragmentation (AIF) modes. Extensive interpretation of the LC-MS/MS spectra had led to the identification of 76 metabolites belonging to different phytochemical classes including alkaloids, polyketides, sesquiterpenes, butyrolactones, peptides, fatty acids, isocoumarins, quinones, among others. Metabolites were tentatively identified by comparing the accurate mass and fragmentation pattern with metabolites previously reported in the literature, as well as bioinformatics analysis using GNPS. A relationship between the host C. officinalis and its endophytes (A. flavus, A. nidulans, and A. flavipes) was discovered. C. officinalis shares common metabolites with at least one of the three endosymbiotic fungi. Some metabolites have been identified in endophytes and do not exist in their host. Multivariate analysis (MVA) revealed discrimination of A. flavipes from Corallina officinalis and other associated endophytic Aspergillus fungi (A. flavus and A. nidulans).
Collapse
Affiliation(s)
- Sherif M Shama
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University Shibin Elkom 32511 Egypt
| | - Ahmed M Elissawy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University Cairo 11566 Egypt
- Center of Drug Discovery Research and Development, Ain-Shams University Cairo 11566 Egypt
| | - Mohamed A Salem
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University Shibin Elkom 32511 Egypt
| | - Fadia S Youssef
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University Cairo 11566 Egypt
| | - Mohamed S Elnaggar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University Cairo 11566 Egypt
| | - Hesham R El-Seedi
- Chemistry Department, Faculty of Science, Islamic University of Madinah P. O. Box: 170 Madinah 42351 Saudi Arabia
| | - Shaden A M Khalifa
- International Research Center for Food Nutrition and Safety, Jiangsu University Zhenjiang 212013 China
- Psychiatry and Neurology Department, Capio Saint Göran's Hospital Sankt Göransplan 1 112 19 Stockholm Sweden
| | - Khaled Briki
- Laboratory of Organic Chemistry and Natural Substance, University Ziane Achour Djelfa Algeria
| | - Dalia Ibrahim Hamdan
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University Shibin Elkom 32511 Egypt
| | - Abdel Nasser B Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University Cairo 11566 Egypt
- Center of Drug Discovery Research and Development, Ain-Shams University Cairo 11566 Egypt
| |
Collapse
|
12
|
Wu X, Zhong S, Zhou N, Luo L. TRAF6 Inhibitors from Marine Compound Library: Pharmacophore, Virtual Screening, Fragment Replacement, ADMET, and Molecular Dynamics. Mar Drugs 2024; 22:260. [PMID: 38921571 PMCID: PMC11204769 DOI: 10.3390/md22060260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/23/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
TRAF6 is an E3 ubiquitin ligase that plays a crucial role in cell signaling. It is known that MMP is involved in tumor metastasis, and TRAF6 induces MMP-9 expression by binding to BSG. However, inhibiting TRAF6's ubiquitinase activity without disrupting the RING domain is a challenge that requires further research. To address this, we conducted computer-based drug screening to identify potential TRAF6 inhibitors. Using a ligand-receptor complex pharmacophore based on the inhibitor EGCG, known for its anti-tumor properties, we screened 52,765 marine compounds. After the molecular docking of 405 molecules with TRAF6, six compounds were selected for further analysis. By replacing fragments of non-binding compounds and conducting second docking, we identified two promising molecules, CMNPD9212-16 and CMNPD12791-8, with strong binding activity and favorable pharmacological properties. ADME and toxicity predictions confirmed their potential as TRAF6 inhibitors. Molecular dynamics simulations showed that CMNPD12791-8 maintained a stable structure with the target protein, comparable to EGCG. Therefore, CMNPD12791-8 holds promise as a potential inhibitor of TRAF6 for inhibiting tumor growth and metastasis.
Collapse
Affiliation(s)
- Xuexuan Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (X.W.); (N.Z.)
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China;
| | - Nan Zhou
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (X.W.); (N.Z.)
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
13
|
Qiu H, Qian S, Head SA, Sanchez PR, Liu JO, Jin Z. Insights into the structure-activity relationship of the anticancer compound ZJ-101: A role played by the amide moiety. Bioorg Med Chem Lett 2024; 105:129741. [PMID: 38599296 PMCID: PMC11060512 DOI: 10.1016/j.bmcl.2024.129741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/30/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
ZJ-101, a structurally simplified analog of marine natural product superstolide A, was previously designed and synthesized in our laboratory. In the present study four new analogs of ZJ-101 were designed and synthesized to investigate the structure-activity relationship of the acetamide moiety of the molecule. The biological evaluation showed that the amide moiety is important for the molecule's anticancer activity. Replacing the amide with other functional groups such as a sulfonamide group, a carbamate group, and a urea group resulted in the decrease in anticancer activity.
Collapse
Affiliation(s)
- Haibo Qiu
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA
| | - Shan Qian
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA
| | - Sarah A Head
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD 21205, USA
| | - Phillip R Sanchez
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD 21205, USA
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD 21205, USA
| | - Zhendong Jin
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
14
|
Peran JE, Salvador-Reyes LA. Modified oxylipins as inhibitors of biofilm formation in Staphylococcus epidermidis. Front Pharmacol 2024; 15:1379643. [PMID: 38846101 PMCID: PMC11153713 DOI: 10.3389/fphar.2024.1379643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/23/2024] [Indexed: 06/09/2024] Open
Abstract
New approaches to combating microbial drug resistance are being sought, with the discovery of biofilm inhibitors considered as alternative arsenal for treating infections. Natural products have been at the forefront of antimicrobial discovery and serve as inspiration for the design of new antibiotics. We probed the potency, selectivity, and mechanism of anti-biofilm activity of modified oxylipins inspired by the marine natural product turneroic acid. Structure-activity relationship (SAR) evaluation revealed the importance of the trans-epoxide moiety, regardless of the position, for inhibiting biofilm formation. trans-12,13-epoxyoctadecanoic acid (1) and trans-9,10 epoxyoctadecanoic acid (4) selectively target the early stage of biofilm formation, with no effect on planktonic cells. These compounds interrupt the formation of a protective polysaccharide barrier by significantly upregulating the ica operon's transcriptional repressor. This was corroborated by docking experiment with SarA and scanning electron micrographs showing reduced biofilm aggregates and the absence of thread-like structures of extrapolymeric substances. In silico evaluation revealed that 1 and 4 can interfere with the AgrA-mediated communication language in Staphylococci, typical to the diffusible signal factor (DSF) capacity of lipophilic chains.
Collapse
Affiliation(s)
| | - Lilibeth A. Salvador-Reyes
- Marine Science Institute, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| |
Collapse
|
15
|
Evidente A. Advances on anticancer fungal metabolites: sources, chemical and biological activities in the last decade (2012-2023). NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:31. [PMID: 38743184 PMCID: PMC11093966 DOI: 10.1007/s13659-024-00452-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
Among microorganisms, fungi are the ones that have the most imagination in producing secondary metabolites with the most varied structural differences, which are produced through different biosynthetic pathways. Therefore, they synthesize secondary metabolites classifiable into numerous families of natural compounds such as amino acids, alkaloids, anthraquinones, aromatic compounds, cyclohexene epoxides, furanones, macrolides, naphthoquinones, polyketides, pyrones, terpenes, etc. They also produced metabolites with very complex structures that can not be classified in the known families of natural compounds. Many fungal metabolites show different biological activities with potential applications in agriculture, food chemistry, cosmetics, pharmacology and medicine. This review is focused on the fungal secondary metabolites with anticancer activity isolated in the last ten years. For some metabolites, when described, their biosynthetic origin, the mode of action and the results of structure activity relationships studies are also reported.
Collapse
Affiliation(s)
- Antonio Evidente
- Institute Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, 80078 70125, Pozzuoli, NA, Italy.
| |
Collapse
|
16
|
Ngandjui YAT, Kereeditse TT, Kamika I, Madikizela LM, Msagati TAM. Nutraceutical and Medicinal Importance of Marine Molluscs. Mar Drugs 2024; 22:201. [PMID: 38786591 PMCID: PMC11123371 DOI: 10.3390/md22050201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Marine molluscs are of enormous scientific interest due to their astonishing diversity in terms of their size, shape, habitat, behaviour, and ecological roles. The phylum Mollusca is the second most common animal phylum, with 100,000 to 200,000 species, and marine molluscs are among the most notable class of marine organisms. This work aimed to show the importance of marine molluscs as a potential source of nutraceuticals as well as natural medicinal drugs. In this review, the main classes of marine molluscs, their chemical ecology, and the different techniques used for the extraction of bioactive compounds have been presented. We pointed out their nutraceutical importance such as their proteins, peptides, polysaccharides, lipids, polyphenolic compounds pigments, marine enzymes, minerals, and vitamins. Their pharmacological activities include antimicrobial, anticancer, antioxidant, anti-inflammatory, and analgesic activities. Moreover, certain molluscs like abalones and mussels contain unique compounds with potential medicinal applications, ranging from wound healing to anti-cancer effects. Understanding the nutritional and therapeutic value of marine molluscs highlights their significance in both pharmaceutical and dietary realms, paving the way for further research and utilization in human health.
Collapse
Affiliation(s)
- Yvan Anderson Tchangoue Ngandjui
- Institute for Nanotechnology and Water Sustainability, College of Engineering, Science and Technology, University of South Africa, Florida Science Campus, Johannesburg 1705, South Africa; (T.T.K.); (I.K.); (L.M.M.)
| | | | | | | | - Titus Alfred Makudali Msagati
- Institute for Nanotechnology and Water Sustainability, College of Engineering, Science and Technology, University of South Africa, Florida Science Campus, Johannesburg 1705, South Africa; (T.T.K.); (I.K.); (L.M.M.)
| |
Collapse
|
17
|
Lee J, Um S, Kim EH, Kim SH. Genomic and Metabolomic Analyses of Nocardiopsis maritima YSL2 as the Mycorrhizosphere Bacterium of Suaeda maritima (L.) Dumort. JOURNAL OF NATURAL PRODUCTS 2024; 87:733-742. [PMID: 38573876 DOI: 10.1021/acs.jnatprod.3c00843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Nine bacteria were isolated from the episphere of Suaeda maritima (L.) Dumort. Among them, the bacterial strain YSL2 displayed the highest antimicrobial activity on agar plates and exhibited significant novelty compared with other bacteria based on 16S rRNA analysis. Consequently, Nocardiopsis maritima YSL2T was subjected to phenotypic characterization and whole-genome sequencing. Phylogenetic analysis revealed its close association with Nocardiopsis aegyptia SNG49T. Furthermore, genomic analysis of strain YSL2T revealed the presence of various gene clusters, indicating its potential for producing antimicrobial secondary metabolites. Upon cultivation on a large scale, maritiamides A and B (1 and 2) were isolated and characterized as cyclic hexapeptides based on nuclear magnetic resonance, ultraviolet, infrared, and mass spectrometric data. The absolute configurations of the amino acid residues in the maritiamides were determined through chiral derivatization, utilizing FDAA and GITC. Maritiamides 1 and 2 exhibited promising antibacterial activities against Staphylococcus epidermidis and weakly inhibited the growth of Escherichia coli and Pseudomonas fluorescens.
Collapse
Affiliation(s)
- Jaeyoun Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea
| | - Soohyun Um
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea
| | - Eun-Hee Kim
- Ochang Center, Korea Basic Science Institute, 162 Yeongudanji-Ro, Ochang-Eup, Cheongju-Si, Chungcheongbuk-Do 28119, South Korea
| | - Seung Hyun Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea
| |
Collapse
|
18
|
Sun X, Zhang J, Han X, Li S, Zhang X, Bi X. Preparation of imidazole-modified paper membrane for selective extraction of gallic acid and its structural and functional analogues from Pomegranate Peel. RSC Adv 2024; 14:14202-14213. [PMID: 38690107 PMCID: PMC11058456 DOI: 10.1039/d3ra08576g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
In the search for pharmaceutically active compounds from natural products, it is crucial and challenging to develop separation methods that target not only structurally similar compounds but also a class of compounds with desired pharmaceutical functions. To achieve both structure-oriented and function-oriented selectivity, the choice of functional monomers with broad interactions or even biomimetic roles towards targeted compounds is essential. In this work, an imidazole (IM)-functionalized paper membrane was synthesized to realize selectivity. The IM was selected based on its capability to provide multiple interactions, participation in several bioprocesses, and experimental verification of adsorption performance. Using gallic acid as a representative component of Pomegranate Peel, the preparation conditions and extraction parameters were systematically investigated. The optimal membrane solid-phase extraction (MSPE) method allowed for enrichment of gallic acid from the complex matrix of Pomegranate Peel, enabling facile quantitative analysis with a limit of detection (LOD) of 0.1 ng mL-1. Furthermore, with the aid of cheminformatics, the extracted compounds were found to be similar in both their structures and pharmaceutical functions. This work offers a novel approach to preparing a readily synthesized extraction membrane capable of isolating compounds with similar structures and pharmaceutical effects, and provides an MSPE-based analytical method for natural products.
Collapse
Affiliation(s)
- Xiaoxue Sun
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan 250117 Shandong China
| | - Jingyu Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan 250117 Shandong China
| | - Xiaohui Han
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan 250117 Shandong China
| | - Shumin Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan 250117 Shandong China
| | - Xuerui Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan 250117 Shandong China
| | - Xiaodong Bi
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan 250117 Shandong China
- Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences) Jinan 250117 Shandong China
| |
Collapse
|
19
|
Chaudhary P, Janmeda P, Pareek A, Chuturgoon AA, Sharma R, Pareek A. Etiology of lung carcinoma and treatment through medicinal plants, marine plants and green synthesized nanoparticles: A comprehensive review. Biomed Pharmacother 2024; 173:116294. [PMID: 38401516 DOI: 10.1016/j.biopha.2024.116294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/29/2024] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
Lung cancer, a leading global cause of mortality, poses a significant public health challenge primarily linked to tobacco use. While tobacco contributes to over 90% of cases, factors like dietary choices and radiation exposure also play a role. Despite potential benefits from early detection, cancer patients face hurdles, including drug resistance, chemotherapy side effects, high treatment costs, and limited healthcare access. Traditional medicinal plant knowledge has recently unveiled diverse cancer chemopreventive agents from terrestrial and marine sources. These phytochemicals regulate intricate molecular processes, influencing the immune system, apoptosis, cell cycle, proliferation, carcinogen elimination, and antioxidant levels. In pursuing cutting-edge strategies to combat the diverse forms of cancer, technological advancements have spurred innovative approaches. Researchers have focused on the green synthesis of metallic nanoparticles using plant metabolites. This method offers distinct advantages over conventional physical and chemical synthesis techniques, such as cost-effectiveness, biocompatibility, and energy efficiency. Metallic nanoparticles, through various pathways such as the generation of reactive oxygen species, modulation of enzyme activity, DNA fragmentation, disruption of signaling pathways, perturbation of cell membranes, and interference with mitochondrial function resulting in DNA damage, cell cycle arrest, and apoptosis, exhibit significant potential for preventive applications. Thus, the amalgamation of phytocompounds and metallic nanoparticles holds promise as a novel approach to lung cancer therapy. However, further refinements and advancements are necessary to enhance the environmentally friendly process of metallic nanoparticle synthesis.
Collapse
Affiliation(s)
- Priya Chaudhary
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Pracheta Janmeda
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan 304022, India
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana (Ayurvedic Pharmaceutics), Banaras Hindu University, Varanasi 221005, India
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan 304022, India.
| |
Collapse
|
20
|
Rai T, Kaushik N, Malviya R, Sharma PK. A review on marine source as anticancer agents. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:415-451. [PMID: 37675579 DOI: 10.1080/10286020.2023.2249825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
This review investigates the potential of natural compounds obtained from marine sources for the treatment of cancer. The oceans are believed to contain physiologically active compounds, such as alkaloids, nucleosides, macrolides, and polyketides, which have shown promising effects in slowing human tumor cells both in vivo and in vitro. Various marine species, including algae, mollusks, actinomycetes, fungi, sponges, and soft corals, have been studied for their bioactive metabolites with diverse chemical structures. The review explores the therapeutic potential of various marine-derived substances and discusses their possible applications in cancer treatment.
Collapse
Affiliation(s)
- Tamanna Rai
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Gautam Budh Nagar, Greater Noida, Uttar Pradesh 201306, India
| | - Niranjan Kaushik
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Gautam Budh Nagar, Greater Noida, Uttar Pradesh 201306, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Gautam Budh Nagar, Greater Noida, Uttar Pradesh 201306, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Gautam Budh Nagar, Greater Noida, Uttar Pradesh 201306, India
| |
Collapse
|
21
|
Zhang H, Li X, Hui Z, Huang S, Cai M, Shi W, Lin Y, Shen J, Sui M, Lai Q, Shao Z, Dou J, Luo X, Ge Y, Tang X. A Semisynthesis Platform for the Efficient Production and Exploration of Didemnin-Based Drugs. Angew Chem Int Ed Engl 2024; 63:e202318784. [PMID: 38291557 DOI: 10.1002/anie.202318784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
Plitidepsin (or dehydrodidemnin B), an approved anticancer drug, belongs to the didemnin family of cyclic depsipeptides, which are found in limited quantities in marine tunicate extracts. Herein, we introduce a new approach that integrates microbial and chemical synthesis to generate plitidepsin and its analogues. We screened a Tistrella strain library to identify a potent didemnin B producer, and then introduced a second copy of the didemnin biosynthetic gene cluster into its genome, resulting in a didemnin B titer of approximately 75 mg/L. Next, we developed two straightforward chemical strategies to convert didemnin B into plitidepsin, one of which involved a one-step synthetic route giving over 90 % overall yield. Furthermore, we synthesized 13 new didemnin derivatives and three didemnin probes, enabling research into structure-activity relationships and interactions between didemnin and proteins. Our study highlights the synergistic potential of biosynthesis and chemical synthesis in overcoming the challenge of producing complex natural products sustainably and at scale.
Collapse
Affiliation(s)
- Haili Zhang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Xuyang Li
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Zhen Hui
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Shipeng Huang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
- Department of Chemistry and Shenzhen Grubbs Institute, Southern University of Science and Technology, 518000, Shenzhen, China
| | - Mingwei Cai
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Wenguang Shi
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Yang Lin
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| | - Jie Shen
- College of Life Science and Technology, China Pharmaceutical University, 211198, Nanjing, China
| | - Minghao Sui
- College of Life Science and Technology, China Pharmaceutical University, 211198, Nanjing, China
| | - Qiliang Lai
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, 361005, Xiamen, China
| | - Zongze Shao
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, 361005, Xiamen, China
| | - Jie Dou
- College of Life Science and Technology, China Pharmaceutical University, 211198, Nanjing, China
| | - Xiaozhou Luo
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Yun Ge
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Xiaoyu Tang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, 518132, Shenzhen, China
| |
Collapse
|
22
|
Sabotič J, Bayram E, Ezra D, Gaudêncio SP, Haznedaroğlu BZ, Janež N, Ktari L, Luganini A, Mandalakis M, Safarik I, Simes D, Strode E, Toruńska-Sitarz A, Varamogianni-Mamatsi D, Varese GC, Vasquez MI. A guide to the use of bioassays in exploration of natural resources. Biotechnol Adv 2024; 71:108307. [PMID: 38185432 DOI: 10.1016/j.biotechadv.2024.108307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/05/2023] [Accepted: 01/01/2024] [Indexed: 01/09/2024]
Abstract
Bioassays are the main tool to decipher bioactivities from natural resources thus their selection and quality are critical for optimal bioprospecting. They are used both in the early stages of compounds isolation/purification/identification, and in later stages to evaluate their safety and efficacy. In this review, we provide a comprehensive overview of the most common bioassays used in the discovery and development of new bioactive compounds with a focus on marine bioresources. We present a comprehensive list of practical considerations for selecting appropriate bioassays and discuss in detail the bioassays typically used to explore antimicrobial, antibiofilm, cytotoxic, antiviral, antioxidant, and anti-ageing potential. The concept of quality control and bioassay validation are introduced, followed by safety considerations, which are critical to advancing bioactive compounds to a higher stage of development. We conclude by providing an application-oriented view focused on the development of pharmaceuticals, food supplements, and cosmetics, the industrial pipelines where currently known marine natural products hold most potential. We highlight the importance of gaining reliable bioassay results, as these serve as a starting point for application-based development and further testing, as well as for consideration by regulatory authorities.
Collapse
Affiliation(s)
- Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia.
| | - Engin Bayram
- Institute of Environmental Sciences, Bogazici University, Bebek, Istanbul 34342, Turkey
| | - David Ezra
- Department of Plant Pathology and Weed Research, ARO, The Volcani Institute, P.O.Box 15159, Rishon LeZion 7528809, Israel
| | - Susana P Gaudêncio
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; UCIBIO - Applied Biomolecular Sciences Unit, Department of Chemistry, Blue Biotechnology & Biomedicine Lab, NOVA School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Berat Z Haznedaroğlu
- Institute of Environmental Sciences, Bogazici University, Bebek, Istanbul 34342, Turkey
| | - Nika Janež
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
| | - Leila Ktari
- B3Aqua Laboratory, National Institute of Marine Sciences and Technologies, Carthage University, Tunis, Tunisia
| | - Anna Luganini
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Manolis Mandalakis
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, 71500 Heraklion, Greece
| | - Ivo Safarik
- Department of Nanobiotechnology, Biology Centre, ISBB, CAS, Na Sadkach 7, 370 05 Ceske Budejovice, Czech Republic; Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Dina Simes
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, 8005-139 Faro, Portugal; 2GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Evita Strode
- Latvian Institute of Aquatic Ecology, Agency of Daugavpils University, Riga LV-1007, Latvia
| | - Anna Toruńska-Sitarz
- Department of Marine Biology and Biotechnology, Faculty of Oceanography and Geography, University of Gdańsk, 81-378 Gdynia, Poland
| | - Despoina Varamogianni-Mamatsi
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, 71500 Heraklion, Greece
| | | | - Marlen I Vasquez
- Department of Chemical Engineering, Cyprus University of Technology, 3036 Limassol, Cyprus
| |
Collapse
|
23
|
Chen HW, Liu FC, Kuo HM, Tang SH, Niu GH, Zhang MM, Tsou LK, Sung PJ, Wen ZH. Immunomodulatory and anti-angiogenesis effects of excavatolide B and its derivatives in alleviating atopic dermatitis. Biomed Pharmacother 2024; 172:116279. [PMID: 38368838 DOI: 10.1016/j.biopha.2024.116279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/03/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition primarily driven by T helper 2 (Th2) cytokines, resulting in skin barrier defects, angiogenesis, and inflammatory responses. The marine natural product excavatolide B (EXCB), isolated from the Formosan Gorgonian coral Briareum stechei, exhibits anti-inflammatory and analgesic properties. To enhance solubility, EXCB is chemically modified into the derivatives EXCB-61 salt and EXCB-79. The study aims to investigate the therapeutic effects of these compounds on dinitrochlorbenzene (DNCB)-induced skin damage and to elucidate the underlying anti-inflammatory and anti-angiogenesis mechanism. In vitro, using lipopolysaccharide (LPS)-induced RAW 264.7 cells, all compounds at 10 μM significantly inhibited expression of inflammatory proteins (inducible nitric oxide synthase and cyclooxygenase-2), vascular endothelial growth factor (VEGF), and cytokines (interleukin (IL)-1β, IL-6, and IL-17A). In vivo, topical application of these compounds on DNCB-induced AD mice alleviated skin symptoms, reduced serum levels of IgE, IL-4, IL-13, IL-17, and interferon-γ, and moderated histological phenomena such as hyperplasia, inflammatory cell infiltration, and angiogenesis. The three compounds restored the expression of skin barrier-related proteins (loricrin, filaggrin, and claudin-1) and reduced the expression of angiogenesis-related proteins (VEGF and platelet endothelial cell adhesion molecule-CD31) in the tissues. This is the first study to indicate that EXCB, EXCB-61 salt, and EXCB-79 can treat AD disease by reducing inflammation and angiogenesis. Hence, they may be considered potential candidates for the development of new drugs for AD.
Collapse
Affiliation(s)
- Hsiu-Wen Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Shih-Hsuan Tang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan.
| | - Ping-Jyun Sung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|
24
|
Li Q, Li S, Li S, Hao X, Wang A, Si S, Xu Y, Shu J, Gan M. Antimicrobial and Anti-inflammatory Cyclic Tetrapeptides from the Co-cultures of Two Marine-Derived Fungi. JOURNAL OF NATURAL PRODUCTS 2024; 87:365-370. [PMID: 38276888 DOI: 10.1021/acs.jnatprod.3c01115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Violaceotides B-E (1-4), four new cyclic tetrapeptides, along with seven known compounds, were identified from the sponge-associated Aspergillus insulicola IMB18-072 co-cultivated with the marine-derived Alternaria angustiovoidea IMB20-805. Their structures were elucidated by extensive analysis of spectroscopic data, including HRESIMS, 1D and 2D NMR, and MS/MS data. The absolute configurations were determined by the advanced Marfey's method. Compounds 2, 3, and violaceotide A (5) displayed selective antimicrobial activities against the aquatic pathogenic bacteria Edwardsiella tarda and E. ictaluri. In addition, compounds 1-5 showed inhibitory activities against the LPS-induced expression of the inflammatory mediator IL-6 in RAW264.7 cells at a concentration of 10 μM.
Collapse
Affiliation(s)
- Qin Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Shasha Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shunwang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xiaomeng Hao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Anqi Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuyi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yanni Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Jicheng Shu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Maoluo Gan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
25
|
Zayed AOH, Altarabeen M, AlShamaileh E, Zain SM. The potential of some functional group compounds substituted 8-Manzamine A as RSK1 inhibitors: molecular docking and molecular dynamics simulations. J Biomol Struct Dyn 2024:1-10. [PMID: 38319051 DOI: 10.1080/07391102.2024.2310792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
Cancer, an incurable global disease, demands urgent anti-cancer drug development. Marine alkaloids like Manzamine and its derivatives show promise as RSK inhibitors against cancer cell invasion. Replacing the hydrogen at the 8-position of Manzamine A with a hydroxyl group has been shown to significantly enhance its biological activity. In this article, we designed various functional group compounds (A1-A21) substituted 8-Manzamine A by docking, MM-GBSA, molecular dynamics (MD) simulation, and well-tempered metadynamics (WT-MetaD) simulations to evaluate their potential as RSK1 inhibitors. Ligands A1-A21 were docked in the RSK1 N-terminal kinase domain (PDB ID: 2Z7Q) using the Glide module. The calculation of binding energy was performed using Prime MM-GB/SA, while MD simulations were conducted with the Desmond module of Schrodinger suite 2023. Compound A5 exhibits the highest G-score (-7.01) compared to 8-Hydroxymanzamine A (-6.08). Additionally, compounds A6, A10, A12, A17, A11, A4, and A13 demonstrate increased activity against RSK1 when compared to both 8-Hydroxymanzamine A and Manzamine A. Residues LEU68, VAL76, LEU141, PHE143, LEU144, PHE150, ASP148, GLU191, and LEU194 of RSK1 protein play a key role in binding with ligands. An MD simulation of Compound A5 was carried out to explore the dynamic interactions within the protein-ligand complex. Furthermore, WT-MetaD simulations validated the docking study results and identified the most energetically favored conformations for the A5/RSK1 complex. Ligands A5, A6, A10, A12, A17, A11, A4, and A13, featuring diverse functional groups and good Glide scores, may have the potential for significant RSK1 activity and merit further development.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ala' Omar Hasan Zayed
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Mousa Altarabeen
- Department of Basic Medical Sciences, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba, Jordan
| | - Ehab AlShamaileh
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Sharifuddin Md Zain
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
Zhang Z, Sun Y, Li Y, Song X, Wang R, Zhang D. The potential of marine-derived piperazine alkaloids: Sources, structures and bioactivities. Eur J Med Chem 2024; 265:116081. [PMID: 38181652 DOI: 10.1016/j.ejmech.2023.116081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/07/2024]
Abstract
Marine-derived piperazine alkaloids (MDPAs) constitute a significant group of natural compounds known for their diverse structures and biological activities. Over the past five decades, substantial efforts have been devoted to isolating these alkaloids from marine sources and characterizing their chemical and bioactive profiles. To date, a total of 922 marine-derived piperazine alkaloids have been reported from various marine organisms. These compounds demonstrate a wide range of pharmacological properties, including cytotoxicity, antibacterial, antifungal, antiviral, and various other activities. Notably, among these activities, cytotoxicity emerges as the most prominent characteristic of marine-derived piperazine alkaloids. This review also summarizes the structure-activity relationship (SAR) studies associated with the cytotoxicity of these compounds. In summary, our objective is to provide an overview of the research progress concerning marine-derived piperazine alkaloids, with the aim of fostering their continued development and utilization.
Collapse
Affiliation(s)
- Zilong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, PR China.
| | - Yu Sun
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, PR China.
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China.
| | - Xiaomei Song
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, PR China.
| | - Rui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China.
| | - Dongdong Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, PR China.
| |
Collapse
|
27
|
Jayaraman M, Gosu V, Kumar R, Jeyaraman J. Computational insights into potential marine natural products as selective inhibitors of Mycobacterium tuberculosis InhA: A structure-based virtual screening study. Comput Biol Chem 2024; 108:107991. [PMID: 38086160 DOI: 10.1016/j.compbiolchem.2023.107991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 01/22/2024]
Abstract
Several factors are associated with the emergence of drug resistance mechanisms, such as impermeable cell walls, gene mutations, and drug efflux systems. Consequently, bacteria acquire resistance, leading to a decrease in drug efficacy. A new and innovative strategy is required to combat drug resistance in tuberculosis (TB) effectively. Therefore, targeting the mycolic acid biosynthesis pathway, which is involved in synthesising mycolic acids (MAs), essential structural components responsible for mycobacterial pathogenicity, has garnered interest in TB research and the concept of drug resistance. In this context, InhA, which plays a crucial role in the fatty acid synthase-II (FAS-II) system of the MA biosynthetic pathway, was selected as a druggable target for screening investigation. To identify potential lead molecules against InhA, diverse marine natural products (MNPs) were collected from the comprehensive marine natural products database (CMNPD). Virtual screening studies aided in selecting potential lead molecules that best fit within the substrate-binding pocket (SBP) of InhA, forming crucial hydrogen bond interaction with the catalytic residue Tyr158. Three MNPs, CMNPD30814, CMNPD1702, and CMNPD27355, were chosen as prospective alternative molecules due to their favorable pharmacokinetic properties and lack of toxicity according to ProTox-II predictions. Additionally, improved reactivity of the MNPs was observed in the results of density functional theory (DFT) studies. Furthermore, comparative molecular dynamics simulation (MDS), principal component (PC)-based free energy landscape (FEL) analysis, and molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) were employed to show enhanced structural stability, increased H-bond potential, and high binding affinity toward the target InhA. Moreover, the hot spot residues that contributed to the high binding energy profile and anchored the stability of the complexes were revealed with their individual interaction energy. The computational insights from this study provide potential avenues to combat TB through the multifaceted mode of action of these marine lead molecules, which can be further explored in future experimental investigations.
Collapse
Affiliation(s)
- Manikandan Jayaraman
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu 630004, India
| | - Vijayakumar Gosu
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Rajalakshmi Kumar
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to be University), Pillayarkuppam, Puducherry 607402, India
| | - Jeyakanthan Jeyaraman
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu 630004, India; Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
28
|
Liu SJ, Zhong YN, Cheng ZQ, Meng N, Zhang J, Jiang CS. Discovery of Novel Marine-Derived Phidiandine/Lipoic Acid Hybrid as a Potential Anti-Atherosclerosis Agent: Design, Synthesis and in Vitro/in Vivo Evaluation. Chem Biodivers 2024; 21:e202301371. [PMID: 38069597 DOI: 10.1002/cbdv.202301371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
In the present study, a novel derivative, IOP-LA, was prepared by hybridizing antioxidant lipoic acid (LA) and our recently reported antioxidative marine phidianidine B-inspired indole/1,2,4-oxadiazole derivative. Our results demonstrated that IOP-LA could protect vascular endothelial cells (VECs) from oxidized low-density lipoprotein (oxLDL)-induced oxidative stress by activating the Nrf2 pathway, inhibit the production of atherosclerotic plaque, and promote the stability of atherosclerotic plaque in apoE-/- mice. Moreover, the protective effect of IOP-LA was superior to LA at the same concentration. Mechanistic studies revealed that IOP-LA significantly inhibited the increase of reactive oxygen species (ROS) levels and the translocation of nuclear factor kappa-B (NF-κB) nuclear induced by oxLDL through the nuclear factor erythroid2-related factor 2 (Nrf2) pathway. In summary, the data demonstrate that IOP-LA, as a new antioxidant, protects VECs from oxLDL-induced oxidative stress by activating the Nrf2 pathway. It is worth noting that this study provides a promising lead compound for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Shu-Jun Liu
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Ying-Nan Zhong
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Zhi-Qiang Cheng
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| |
Collapse
|
29
|
Yamazaki K, Okuda Y, Takaya A, Nemoto T. Total Synthesis of Dragmacidins G and H. Org Lett 2024; 26:670-675. [PMID: 38206835 DOI: 10.1021/acs.orglett.3c04039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
The total synthesis of dragmacidins G and H was achieved for the first time by employing nucleophilic aromatic substitution and site-selective cross-coupling reactions using appropriately functionalized pyrazines as substrates. The evaluation of antibacterial activities of dragmacidin G, dragmacidin H, and synthetic analogues against Staphylococcus aureus and the efflux pump-deficient Salmonella Typhimurium revealed that the presence of a Br group on the indole ring adjacent to the sulfide unit was important for increasing antibacterial activities.
Collapse
Affiliation(s)
- Keita Yamazaki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Yuma Okuda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Akiko Takaya
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Tetsuhiro Nemoto
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
30
|
Lu CJ, Liang LF, Zhang GS, Li HY, Fu CQ, Yu Q, Zhou DM, Su ZW, Liu K, Gao CH, Xu XY, Liu YH. Carneusones A-F, Benzophenone Derivatives from Sponge-Derived Fungus Aspergillus carneus GXIMD00543. Mar Drugs 2024; 22:63. [PMID: 38393034 PMCID: PMC10890008 DOI: 10.3390/md22020063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Six benzophenone derivatives, carneusones A-F (1-6), along with seven known compounds (7-13) were isolated from a strain of sponge-derived marine fungus Aspergillus carneus GXIMD00543. Their chemical structures were elucidated by detailed spectroscopic data and quantum chemical calculations. Compounds 5, 6, and 8 exhibited moderate anti-inflammatory activity on NO secretion using lipopolysaccharide (LPS)-induced RAW 264.7 cells with EC50 values of 34.6 ± 0.9, 20.2 ± 1.8, and 26.8 ± 1.7 μM, while 11 showed potent effect with an EC50 value of 2.9 ± 0.1 μM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xin-Ya Xu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.-J.L.); (L.-F.L.); (G.-S.Z.); (H.-Y.L.); (C.-Q.F.); (Q.Y.); (D.-M.Z.); (Z.-W.S.); (K.L.); (C.-H.G.)
| | - Yong-Hong Liu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.-J.L.); (L.-F.L.); (G.-S.Z.); (H.-Y.L.); (C.-Q.F.); (Q.Y.); (D.-M.Z.); (Z.-W.S.); (K.L.); (C.-H.G.)
| |
Collapse
|
31
|
Barzkar N, Sukhikh S, Babich O. Study of marine microorganism metabolites: new resources for bioactive natural products. Front Microbiol 2024; 14:1285902. [PMID: 38260902 PMCID: PMC10800913 DOI: 10.3389/fmicb.2023.1285902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024] Open
Abstract
The marine environment has remained a source of novel biological molecules with diversified applications. The ecological and biological diversity, along with a unique physical environment, have provided the evolutionary advantage to the plant, animals and microbial species thriving in the marine ecosystem. In light of the fact that marine microorganisms frequently interact symbiotically or mutualistically with higher species including corals, fish, sponges, and algae, this paper intends to examine the potential of marine microorganisms as a niche for marine bacteria. This review aims to analyze and summarize modern literature data on the biotechnological potential of marine fungi and bacteria as producers of a wide range of practically valuable products (surfactants, glyco-and lipopeptides, exopolysaccharides, enzymes, and metabolites with different biological activities: antimicrobial, antitumor, and cytotoxic). Hence, the study on bioactive secondary metabolites from marine microorganisms is the need of the hour. The scientific novelty of the study lies in the fact that for the first time, the data on new resources for obtaining biologically active natural products - metabolites of marine bacteria and fungi - were generalized. The review investigates the various kinds of natural products derived from marine microorganisms, specifically focusing on marine bacteria and fungi as a valuable source for new natural products. It provides a summary of the data regarding the antibacterial, antimalarial, anticarcinogenic, antibiofilm, and anti-inflammatory effects demonstrated by marine microorganisms. There is currently a great need for scientific and applied research on bioactive secondary metabolites of marine microorganisms from the standpoint of human and animal health.
Collapse
Affiliation(s)
- Noora Barzkar
- Department of Agro-Industrial Technology, Faculty of Applied Science, Food and Agro-Industrial Research Center, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| | - Stanislav Sukhikh
- Research and Education Center “Industrial Biotechnologies”, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Olga Babich
- Research and Education Center “Industrial Biotechnologies”, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| |
Collapse
|
32
|
Ye W, Lui ST, Zhao Q, Wong YM, Cheng A, Sung HHY, Williams ID, Qian PY, Huang P. Novel marine natural products as effective TRPV1 channel blockers. Int J Biol Macromol 2023; 253:127136. [PMID: 37776932 DOI: 10.1016/j.ijbiomac.2023.127136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Chronic pain management poses a formidable challenge to healthcare, exacerbated by current analgesic options' limitations and adverse effects. Transient receptor potential vanilloid 1 (TRPV1), a non-selective cation channel, has emerged as a promising target for novel analgesics. However, safety and tolerability concerns have constrained the development of TRPV1 modulators. In this study, we explored marine-derived natural products as a source of potential TRPV1 modulators using high-throughput dye-uptake assays. We identified chrexanthomycins, a family of hexacyclic xanthones, exhibited potent TRPV1 inhibitory effects, with compounds cC and cF demonstrating the most significant activity. High-resolution patch-clamp assays confirmed the direct action of these compounds on the TRPV1 channel. Furthermore, in vivo assays revealed that cC and cF effectively suppressed capsaicin-induced pain sensation in mice, comparable to the known TRPV1 inhibitor, capsazepine. Structural-activity relationship analysis highlighted the importance of specific functional groups in modulating TRPV1 activity. Our findings underscore the therapeutic potential of chrexanthomycins and pave the way for further investigations into marine-derived TRPV1 modulators for pain management.
Collapse
Affiliation(s)
- Wenkang Ye
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China; SZU-HKUST Joint Ph.D. Program in Marine Environmental Science, Shenzhen University, Shenzhen 518060, China
| | - Sin Tung Lui
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Qirui Zhao
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuk Ming Wong
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Aifang Cheng
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China
| | - Herman H-Y Sung
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ian D Williams
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Pei-Yuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Pingbo Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
33
|
Cahyani NKD, Kasanah N, Kurnia DS, Hamann MT. Profiling Prokaryotic Communities and Aaptamines of Sponge Aaptos suberitoides from Tulamben, Bali. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1158-1175. [PMID: 38008858 PMCID: PMC11329227 DOI: 10.1007/s10126-023-10268-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/04/2023] [Indexed: 11/28/2023]
Abstract
Sponges (Porifera) harbor a diversity of microorganisms that contribute largely to the production a vast array of bioactive compounds. The microorganisms associated with sponge have an important impact on the chemical diversity of the natural products. Herein, our study focuses on an Aaptos suberitoides commonly found in Indonesia. The objective of this study was to investigate the profile of prokaryotic community and the presence of aaptamine metabolites in sponge Aaptos suberitoides. Sponges were collected from two site locations (Liberty Wreck and Drop Off) in Tulamben, Bali. The sponges were identified by barcoding DNA cytochrome oxidase subunit I (COI) gene. The profile of prokaryotic composition was investigated by amplifying the 16S rRNA gene using primers 515f and 806r to target the V4 region. The metabolites were analyzed using LC-MS, and dereplication was done to identify the aaptamines and its derivates. The barcoding DNA of the sponges confirmed the identity of samples as Aaptos suberitoides. The prokaryotic communities of samples A. suberitoides were enriched and dominated by taxa Proteobacteria, Chloroflexi, Actinobacteria, and Acidobacteria. The chemical analysis showed that all sponges produce aaptamine and isoaaptamine except A. suberitoides S2421 produce analog of aaptamines. This is the first report on the profile of prokaryotic community and the aaptamine of tropical marine sponges, A. suberitoides, from Tulamben, Bali.
Collapse
Affiliation(s)
- Ni Kadek Dita Cahyani
- Biology Department, Faculty of Science and Mathematics, Diponegoro University, Semarang, Central Java, Indonesia
| | - Noer Kasanah
- Department of Fisheries, Faculty of Agriculture, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Dewi Sri Kurnia
- Department of Biotechnology, Graduate School, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Mark T Hamann
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
34
|
Matsumoto-Elliott O, Sanchez LM. I-SMEL a Big Catch! ACS CENTRAL SCIENCE 2023; 9:2006-2008. [PMID: 38033790 PMCID: PMC10683497 DOI: 10.1021/acscentsci.3c01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Affiliation(s)
- Olivia Matsumoto-Elliott
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High St, Santa Cruz, California 95064, United States
| | - Laura M Sanchez
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High St, Santa Cruz, California 95064, United States
| |
Collapse
|
35
|
Orefice I, Balzano S, Romano G, Sardo A. Amphidinium spp. as a Source of Antimicrobial, Antifungal, and Anticancer Compounds. Life (Basel) 2023; 13:2164. [PMID: 38004303 PMCID: PMC10671881 DOI: 10.3390/life13112164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Dinoflagellates make up the second largest marine group of marine unicellular eukaryotes in the world ocean and comprise both heterotrophic and autotrophic species, encompassing a wide genetic and chemical diversity. They produce a plethora of secondary metabolites that can be toxic to other species and are mainly used against predators and competing species. Dinoflagellates are indeed often responsible for harmful algal bloom, where their toxic secondary metabolites can accumulate along the food chain, leading to significant damages to the ecosystem and human health. Secondary metabolites from dinoflagellates have been widely investigated for potential biomedical applications and have revealed multiple antimicrobial, antifungal, and anticancer properties. Species from the genus Amphidinium seem to be particularly interesting for the production of medically relevant compounds. The present review aims at summarising current knowledge on the diversity and the pharmaceutical properties of secondary metabolites from the genus Amphidinium. Specifically, Amphidinium spp. produce a range of polyketides possessing cytotoxic activities such as amphidinolides, caribenolides, amphidinins, and amphidinols. Potent antimicrobial properties against antibiotic-resistant bacterial strains have been observed for several amphidinins. Amphidinols revealed instead strong activities against infectious fungi such as Candida albicans and Aspergillus fumigatus. Finally, compounds such as amphidinolides, isocaribenolide-I, and chlorohydrin 2 revealed potent cytotoxic activities against different cancer cell lines. Overall, the wide variety of antimicrobial, antifungal, and anticancer properties of secondary metabolites from Amphidinium spp. make this genus a highly suitable candidate for future medical applications, spanning from cancer drugs to antimicrobial products that are alternatives to currently available antibiotic and antimycotic products.
Collapse
Affiliation(s)
| | | | | | - Angela Sardo
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80131 Naples, Italy; (I.O.); (S.B.); (G.R.)
| |
Collapse
|
36
|
Jahajeeah D, Ranghoo-Sanmukhiya M, Schäfer G. Metabolic Profiling, Antiviral Activity and the Microbiome of Some Mauritian Soft Corals. Mar Drugs 2023; 21:574. [PMID: 37999398 PMCID: PMC10672535 DOI: 10.3390/md21110574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
Soft corals, recognized as sessile marine invertebrates, rely mainly on chemical, rather than physical defense, by secreting intricate secondary metabolites with plausible pharmaceutical implication. Their ecological niche encompasses a diverse community of symbiotic microorganisms which potentially contribute to the biosynthesis of these bioactive metabolites. The emergence of new viruses and heightened viral resistance underscores the urgency to explore novel pharmacological reservoirs. Thus, marine organisms, notably soft corals and their symbionts, have drawn substantial attention. In this study, the chemical composition of four Mauritian soft corals: Sinularia polydactya, Cespitularia simplex, Lobophytum patulum, and Lobophytum crassum was investigated using LC-MS techniques. Concurrently, Illumina 16S metagenomic sequencing was used to identify the associated bacterial communities in the named soft corals. The presence of unique biologically important compounds and vast microbial communities found therein was further followed up to assess their antiviral effects against SARS-CoV-2 and HPV pseudovirus infection. Strikingly, among the studied soft corals, L. patulum displayed an expansive repertoire of unique metabolites alongside a heightened bacterial consort. Moreover, L. patulum extracts exerted some promising antiviral activity against SARS-CoV-2 and HPV pseudovirus infection, and our findings suggest that L. patulum may have the potential to serve as a therapeutic agent in the prevention of infectious diseases, thereby warranting further investigation.
Collapse
Affiliation(s)
- Deeya Jahajeeah
- Department of Agricultural & Food Science, Faculty of Agriculture, University of Mauritius, Reduit 80837, Mauritius;
- International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa;
| | - Mala Ranghoo-Sanmukhiya
- Department of Agricultural & Food Science, Faculty of Agriculture, University of Mauritius, Reduit 80837, Mauritius;
| | - Georgia Schäfer
- International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa;
| |
Collapse
|
37
|
Holland DC, Carroll AR. Marine indole alkaloid diversity and bioactivity. What do we know and what are we missing? Nat Prod Rep 2023; 40:1595-1607. [PMID: 36790012 DOI: 10.1039/d2np00085g] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Covering: marine indole alkaloids (n = 2048) and their reported bioactivities up to the end of 2021Despite increasing numbers of marine natural products (MNPs) reported each year, most have only been examined for cytotoxic, antibacterial, and/or antifungal biological activities with the majority found to be inactive in these assays. In this context, why are natural products continuing to be examined in assays they are unlikely to show significant activity in, and what targets might be more useful for expanding knowledge of their biologically relevant chemical space? We have undertaken a meta-analysis of the biological activities for 2048 marine indole alkaloids (MIAs), a diverse sub-class of MNPs reported up to the end of 2021, and this has highlighted that the bioactivity potentials for up to 86% of published MIAs remains underexplored and/or undefined. Although most published MIAs are not cytotoxic or antimicrobial, there is a continued focus on using these assays to evaluate new structurally related analogues. Using cheminformatics analyses, the chemical diversity of the 2048 MIAs were clustered using fragment based fingerprints and their reported bioactivity potency towards specific disease targets was assessed for structure activity trends. These analyses showed that there are groups of MIAs that possess potent and diverse activities and that many analogues, previously tested only in cellular toxicity assays, could be better exploited to generate structure activity relationships associated with leads to treat emerging diseases. A collection of indole drug and drug-lead structures from non-natural sources were also incorporated into the dataset providing complementary bioactivity profiles that were further used to predict underexplored areas of potential new activity and to better direct future testing of MIAs. Our findings clearly suggest the biological evaluation of MIAs continues to be conducted on a narrow range of bioassays and disease targets, and that shifting the focus to non-toxic disease targets should provide expanded knowledge of biologically relevant chemical space aimed at maximising the potential of MIAs for drug discovery.
Collapse
Affiliation(s)
- Darren C Holland
- School of Environment and Science, Griffith University, Gold Coast, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia.
| | - Anthony R Carroll
- School of Environment and Science, Griffith University, Gold Coast, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia.
| |
Collapse
|
38
|
Nugraha AS, Firli LN, Rani DM, Hidayatiningsih A, Lestari ND, Wongso H, Tarman K, Rahaweman AC, Manurung J, Ariantari NP, Papu A, Putra MY, Pratama ANW, Wessjohann LA, Keller PA. Indonesian marine and its medicinal contribution. NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:38. [PMID: 37843645 PMCID: PMC10579215 DOI: 10.1007/s13659-023-00403-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023]
Abstract
The archipelagic country of Indonesia is populated by the densest marine biodiversity in the world which has created strong global interest and is valued by both Indigenous and European settlements for different purposes. Nearly 1000 chemicals have been extracted and identified. In this review, a systematic data curation was employed to collate bioprospecting related manuscripts providing a comprehensive directory based on publications from 1988 to 2022. Findings with significant pharmacological activities are further discussed through a scoping data collection. This review discusses macroorganisms (Sponges, Ascidian, Gorgonians, Algae, Mangrove) and microorganism (Bacteria and Fungi) and highlights significant discoveries, including a potent microtubule stabilizer laulimalide from Hyattella sp., a prospective doxorubicin complement papuamine alkaloid from Neopetrosia cf exigua, potent antiplasmodial manzamine A from Acanthostrongylophora ingens, the highly potent anti trypanosomal manadoperoxide B from Plakortis cfr. Simplex, mRNA translation disrupter hippuristanol from Briareum sp, and the anti-HIV-1 (+)-8-hydroxymanzamine A isolated from Acanthostrongylophora sp. Further, some potent antibacterial extracts were also found from a limited biomass of bacteria cultures. Although there are currently no examples of commercial drugs from the Indonesian marine environment, this review shows the molecular diversity present and with the known understudied biodiversity, reveals great promise for future studies and outcomes.
Collapse
Affiliation(s)
- Ari Satia Nugraha
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember, 68121, Indonesia.
- Leibniz Institute Für Pflanzenbiochemie, Weinberg 3, 06120, Halle (Saale), Germany.
- School of Chemistry and Molecular Biosciences, Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia.
| | - Lilla Nur Firli
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember, 68121, Indonesia
| | - Dinar Mutia Rani
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember, 68121, Indonesia
| | - Ayunda Hidayatiningsih
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember, 68121, Indonesia
| | - Nadya Dini Lestari
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember, 68121, Indonesia
| | - Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, J1. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Kustiariyah Tarman
- Department of Aquatic Product Technology, Faculty of Fisheries and Marine Sciences; and Division of Marine Biotechnology, Centre for Coastal and Marine Resources Studies (CCMRS), IPB University, Bogor, 16680, Indonesia
| | | | - Jeprianto Manurung
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Puschstrasse 4, 04103, Leipzig, Germany
| | - Ni Putu Ariantari
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Udayana University, Badung, Bali, 80361, Indonesia
| | - Adelfia Papu
- Biology Department, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, 95115, Indonesia
| | - Masteria Yunovilsa Putra
- Vaccine and Drug Research Center, National Research and Innovation Agency, Cibinong, Jawa Barat, 16911, Indonesia
| | | | - Ludger A Wessjohann
- Leibniz Institute Für Pflanzenbiochemie, Weinberg 3, 06120, Halle (Saale), Germany
| | - Paul A Keller
- School of Chemistry and Molecular Biosciences, Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia
| |
Collapse
|
39
|
Zhang J, Cai YS, Ji HL, Ma M, Zhang JH, Cheng ZQ, Wang KM, Jiang CS, Zhuang C, Hu Y, Meng N. Discovery of marine phidianidine-based Nrf2 activators and their potential against oxLDL- and HG-induced injury in HUVECs. Bioorg Med Chem Lett 2023; 95:129468. [PMID: 37689216 DOI: 10.1016/j.bmcl.2023.129468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
One effective strategy for treating atherosclerosis is to inhibit the injury of vascular endothelial cells (VECs) induced by oxidized low-density lipoprotein (oxLDL) and high glucose (HG). This study synthesized and evaluated a series of novel Nrf2 activators derived from the marine natural product phidianidine for their ability to protect human umbilical VECs against oxLDL- and HG-induced injury. The results of in vitro bioassays demonstrated that compound D-36 was the most promising Nrf2 activator, effectively inhibiting the apoptosis of HUVECs induced by oxLDL and HG. Furthermore, Nrf2 knockdown experiments confirmed that compound D-36 protected against oxLDL- and HG-induced apoptosis in HUVECs by activating the Nrf2 pathway. These findings provide important insights into a new chemotype of marine-derived Nrf2 activators that could potentially be optimized to develop effective anti-atherosclerosis agents.
Collapse
Affiliation(s)
- Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yong-Si Cai
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Hua-Long Ji
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Mengqi Ma
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Jin-He Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Zhi-Qiang Cheng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kai-Ming Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Chunlin Zhuang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Yang Hu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China.
| |
Collapse
|
40
|
Alzain AA, Elbadwi FA, Mohamed SGA, Kushk KSA, Bafarhan RI, Alswiri SA, Khushaim SN, Hussein HGA, Abuhajras MYA, Mohamed GA, Ibrahim SRM. Exploring marine-derived compounds for MET signalling pathway inhibition in cancer: integrating virtual screening, ADME profiling and molecular dynamics investigations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:1003-1021. [PMID: 38014514 DOI: 10.1080/1062936x.2023.2284917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
The MET signalling pathway regulates fundamental cellular processes such as growth, division, and survival. While essential for normal cell function, dysregulation of this pathway can contribute to cancer by triggering uncontrolled proliferation and metastasis. Targeting MET activity holds promise as an effective strategy for cancer therapy. Among potential sources of anti-cancer agents, marine organisms have gained attention. In this study, we screened 47,450 natural compounds derived from marine sources within the CMNPD database against the Met crystal structure. By employing HTVS, SP, and XP docking modes, we identified three compounds (CMNPD17595, CMNPD14026, and CMNPD19696) that outperformed a reference molecule in binding affinity to the Met structure. These compounds demonstrated desirable ADME properties. Molecular Dynamics (MD) simulations for 200 ns confirmed the stability of their interactions with Met. Our findings highlight CMNPD17595, CMNPD14026, and CMNPD19696 as potential inhibitors against Met-dependent cancers. Additionally, these compounds offer new avenues for drug development, leveraging their inhibitory effects on Met to combat carcinogenesis.
Collapse
Affiliation(s)
- A A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - F A Elbadwi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S G A Mohamed
- Faculty of Dentistry, British University, El Sherouk City, Egypt
| | - K S A Kushk
- Operations Sales Department, United Pharmaceuticals & Medical Supply Co. Ltd, Al Madinah Al-Munawwarah, Saudi Arabia
| | - R I Bafarhan
- Pharmaceutical Care Services, Medical Department, Private Sector, Tabuk, Saudi Arabia
| | - S A Alswiri
- Pharmaceutical Company, Medical Department, Private Sector, Al Madinah Al-Munawwarah, Saudi Arabia
| | - S N Khushaim
- College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - H G A Hussein
- Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - M Y A Abuhajras
- Medical Claims Department, Bupa Arabia, Prince Saud AlFaisal, Jeddah, Saudi Arabia
| | - G A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - S R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
41
|
Lin SH, Yan QX, Zhang Y, Wu TZ, Zou ZB, Liu QM, Jiang JY, Xie MM, Xu L, Hao YJ, Liu Z, Liu GM, Yang XW. Citriquinolinones A and B: Rare Isoquinolinone-Embedded Citrinin Analogues and Related Metabolites from the Deep-Sea-Derived Aspergillus versicolor 170217. Mar Drugs 2023; 21:504. [PMID: 37888439 PMCID: PMC10608187 DOI: 10.3390/md21100504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
A systematic chemical investigation of the deep-sea-derived fungus Aspergillus versicolor 170217 resulted in the isolation of six new (1-6) and 45 known (7-51) compounds. The structures of the new compounds were established on the basis of exhaustive analysis of their spectroscopic data and theoretical-statistical approaches including GIAO-NMR, TDDFT-ECD/ORD calculations, DP4+ probability analysis, and biogenetic consideration. Citriquinolinones A (1) and B (2) feature a unique isoquinolinone-embedded citrinin scaffold, representing the first exemplars of a citrinin-isoquinolinone hybrid. Dicitrinones K-L (3-4) are two new dimeric citrinin analogues with a rare CH-CH3 bridge. Biologically, frangula-emodin (32) and diorcinol (17) displayed remarkable anti-food allergic activity with IC50 values of 7.9 ± 3.0 μM and 13.4 ± 1.2 μM, respectively, while diorcinol (17) and penicitrinol A (20) exhibited weak inhibitory activity against Vibrio parahemolyticus, with MIC values ranging from 128 to 256 μM.
Collapse
Affiliation(s)
- Shui-Hua Lin
- Department of Pharmacy, Quanzhou Medical College, 2 Anji Road, Quanzhou 362000, China;
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Qing-Xiang Yan
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Yong Zhang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Tai-Zong Wu
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Zheng-Biao Zou
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Qing-Mei Liu
- College of Food and Biological Engineering, Jimei University, 43 Yindou Road, Xiamen 361021, China; (Q.-M.L.); (G.-M.L.)
| | - Jia-Yang Jiang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
- College of Life Sciences, Hainan University, 58 People’s Avenue, Haikou 570228, China;
| | - Ming-Min Xie
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Lin Xu
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - You-Jia Hao
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| | - Zhu Liu
- College of Life Sciences, Hainan University, 58 People’s Avenue, Haikou 570228, China;
| | - Guang-Ming Liu
- College of Food and Biological Engineering, Jimei University, 43 Yindou Road, Xiamen 361021, China; (Q.-M.L.); (G.-M.L.)
| | - Xian-Wen Yang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Q.-X.Y.); (Y.Z.); (T.-Z.W.); (Z.-B.Z.); (J.-Y.J.); (M.-M.X.); (L.X.); (Y.-J.H.)
| |
Collapse
|
42
|
Huynh TH, Bae ES, Heo BE, Lee J, An JS, Kwon Y, Nam SJ, Oh KB, Jang J, Lee SK, Oh DC. Tandocyclinones A and B, Ether Bridged C-Glycosyl Benz[ a]anthracenes from an Intertidal Zone Streptomyces sp. Mar Drugs 2023; 21:500. [PMID: 37755113 PMCID: PMC10533038 DOI: 10.3390/md21090500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/28/2023] Open
Abstract
Two new proton-deficient metabolites, tandocyclinones A and B (1 and 2), were discovered via the chemical profiling of the Streptomyces sp. strain TDH03, which was isolated from a marine sediment sample collected from the intertidal mudflat in Tando Port, the Republic of Korea. The structures of 1 and 2 were elucidated as new ether-bridged C-glycosyl benz[a]anthracenes by using a combination of spectroscopic analyses of ultraviolet (UV) and mass spectrometry (MS) data, along with nuclear magnetic resonance (NMR) spectra, which were acquired in tetrahydrofuran (THF)-d8 selected after an extensive search for a solvent, resulting in mostly observable exchangeable protons in the 1H NMR spectrum. Their configurations were successfully assigned by applying a J-based configuration analysis, rotating-frame Overhauser enhancement spectroscopy (ROESY) NMR correlations, chemical derivatization methods based on NMR (a modified version of Mosher's method) and circular dichroism (CD) (Snatzke's method using Mo2(OAc)4-induced CD), as well as quantum-mechanics-based computational methods, to calculate the electronic circular dichroism (ECD). Tandocyclinones A and B (1 and 2) were found to have weak antifungal activity against Trichophyton mentagrophytes IFM40996 with an MIC value of 128 μg/mL (244 and 265 μM for 1 and 2, respectively). A further biological evaluation revealed that tandocyclinone A (1) displayed inhibitory activity against Mycobacterium avium (MIC50 = 40.8 μM) and antiproliferative activity against SNU638 and HCT116 cancer cells, with IC50 values of 31.9 µM and 49.4 µM, respectively.
Collapse
Affiliation(s)
- Thanh-Hau Huynh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (T.-H.H.); (E.S.B.); (J.S.A.); (S.K.L.)
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (T.-H.H.); (E.S.B.); (J.S.A.); (S.K.L.)
| | - Bo Eun Heo
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (B.E.H.); (J.J.)
| | - Jayho Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (K.-B.O.)
| | - Joon Soo An
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (T.-H.H.); (E.S.B.); (J.S.A.); (S.K.L.)
| | - Yun Kwon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Ki-Bong Oh
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (K.-B.O.)
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (B.E.H.); (J.J.)
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (T.-H.H.); (E.S.B.); (J.S.A.); (S.K.L.)
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (T.-H.H.); (E.S.B.); (J.S.A.); (S.K.L.)
| |
Collapse
|
43
|
Hermans C, De Mol ML, Mispelaere M, De Rop AS, Rombaut J, Nusayr T, Creamer R, De Maeseneire SL, Soetaert WK, Hulpiau P. MariClus: Your One-Stop Platform for Information on Marine Natural Products, Their Gene Clusters and Producing Organisms. Mar Drugs 2023; 21:449. [PMID: 37623730 PMCID: PMC10455768 DOI: 10.3390/md21080449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The marine environment hosts the vast majority of living species and marine microbes that produce natural products with great potential in providing lead compounds for drug development. With over 70% of Earth's surface covered in water and the high interaction rate associated with liquid environments, this has resulted in many marine natural product discoveries. Our improved understanding of the biosynthesis of these molecules, encoded by gene clusters, along with increased genomic information will aid us in uncovering even more novel compounds. RESULTS We introduce MariClus (https://www.mariclus.com), an online user-friendly platform for mining and visualizing marine gene clusters. The first version contains information on clusters and the predicted molecules for over 500 marine-related prokaryotes. The user-friendly interface allows scientists to easily search by species, cluster type or molecule and visualize the information in table format or graphical representation. CONCLUSIONS This new online portal simplifies the exploration and comparison of gene clusters in marine species for scientists and assists in characterizing the bioactive molecules they produce. MariClus integrates data from public sources, like GenBank, MIBiG and PubChem, with genome mining results from antiSMASH. This allows users to access and analyze various aspects of marine natural product biosynthesis and diversity.
Collapse
Affiliation(s)
- Cedric Hermans
- Bioinformatics Knowledge Center (BiKC), Campus Brugge Station, Howest University of Applied Sciences, Rijselstraat 5, 8200 Bruges, Belgium; (C.H.)
| | - Maarten Lieven De Mol
- Centre for Industrial Biotechnology and Biocatalysis (InBio.be), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Marieke Mispelaere
- Bioinformatics Knowledge Center (BiKC), Campus Brugge Station, Howest University of Applied Sciences, Rijselstraat 5, 8200 Bruges, Belgium; (C.H.)
| | - Anne-Sofie De Rop
- Centre for Industrial Biotechnology and Biocatalysis (InBio.be), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Jeltien Rombaut
- Centre for Industrial Biotechnology and Biocatalysis (InBio.be), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Tesneem Nusayr
- Life Sciences, Texas A&M-Corpus Christi, Corpus Christi, TX 78412, USA
| | - Rebecca Creamer
- Entomology, Plant Pathology, and Weed Science, New Mexico State University, Las Cruces, NM 88003, USA
| | - Sofie L. De Maeseneire
- Centre for Industrial Biotechnology and Biocatalysis (InBio.be), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Wim K. Soetaert
- Centre for Industrial Biotechnology and Biocatalysis (InBio.be), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Paco Hulpiau
- Bioinformatics Knowledge Center (BiKC), Campus Brugge Station, Howest University of Applied Sciences, Rijselstraat 5, 8200 Bruges, Belgium; (C.H.)
| |
Collapse
|
44
|
Abdelghany FM, Abdel-Warith AWA, Younis EM, Davies SJ, Elnakeeb MA, El-Nawasany MM, El-Bahlol AA, Bauomi MA, Abu-Almaaty AH, Omer MY. Genetic Differentiation of Five Sea Cucumber Species from the Red Sea, Hurghada, Egypt. BRAZ J BIOL 2023; 83:e271983. [PMID: 37283337 DOI: 10.1590/1519-6984.271983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/16/2023] [Indexed: 06/08/2023] Open
Abstract
This study aimed to assess the genetic differentiation and relationship among five sea cucumber species from the Red Sea in Egypt, namely Holothuria atra, H. impatiens, H. leucospilota, Actinopyga crassa and A. mauritiana, using Inter Simple Sequence Repeated (ISSR) and Start Codon Targeted (SCoT) markers. A collection of 100 specimens, with 20 individuals per species, was gathered for the analysis. With ten ISSR primers, 135 amplified bands were detected, including 11 distinct species-specific bands, indicating high-level polymorphism among species. Using ten SCoT primers, 151 amplicons were generated, including 30 species-specific bands, with 52% polymorphic bands indicating high-level polymorphism among species. The degree of genetic similarity (GS) among the different genotypes of species was calculated based on ISSR bands analysis, which ranged from 93% between H. atra and H. impatiens to 86% between H. atra and A. crassa. The highest genetic similarity was observed between H. atra and H. impatiens (90%), while the lowest was identified between A. crassa and A. mauritiana (75%) using SCoT bands. Notably, the ISSR and SCoT-based DNA analysis revealed similar genetic relationships between H. atra and H. impatiens compared to other sea cucumber species studied. This study provides new insights into the genetic diversity and relationship among sea cucumber species in the Red Sea, which could have implications for their conservation and management.
Collapse
Affiliation(s)
- F M Abdelghany
- Al-Azhar University, Faculty of Agriculture, Fish Production Department, Nasr City, Cairo, Egypt
| | - A W A Abdel-Warith
- King Saud University, College of Science, Department of Zoology, Riyadh, Saudi Arabia
| | - E M Younis
- King Saud University, College of Science, Department of Zoology, Riyadh, Saudi Arabia
| | - S J Davies
- National University of Ireland Galway, School of Science and Engineering, Galway, Ireland
| | - M A Elnakeeb
- Al-Azhar University, Faculty of Agriculture, Fish Production Department, Nasr City, Cairo, Egypt
| | - M M El-Nawasany
- Al-Azhar University, Faculty of Agriculture, Fish Production Department, Nasr City, Cairo, Egypt
| | - A A El-Bahlol
- Al-Azhar University, Faculty of Agriculture, Fish Production Department, Nasr City, Cairo, Egypt
| | - M A Bauomi
- Al-Azhar University, Faculty of Agriculture, Fish Production Department, Nasr City, Cairo, Egypt
| | - A H Abu-Almaaty
- Port Said University, Faculty of Science, Zoology Department, Port Said, Egypt
| | - M Y Omer
- National Institute of Oceanography and Fisheries, Hurghada, Egypt
| |
Collapse
|
45
|
Piwko AT, Miller BG, Smith JM. Revisiting the manzamine biosynthetic hypothesis. Nat Prod Rep 2023; 40:964-971. [PMID: 36648485 PMCID: PMC10773000 DOI: 10.1039/d2np00082b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Covering: up to 2023The marine environment represents a rich yet challenging source of novel therapeutics. These challenges are best exemplified by the manzamine class of alkaloids, featuring potent bioactivities, difficult procurement, and a biosynthetic pathway that has eluded characterization for over three decades. This review highlights postulated biogenic pathways toward the manzamines, evaluated in terms of current biosynthetic knowledge and metabolic precedent.
Collapse
Affiliation(s)
- Alexander T Piwko
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32308, USA.
| | - Brian G Miller
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32308, USA.
| | - Joel M Smith
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32308, USA.
| |
Collapse
|
46
|
Lauritano C, Galasso C. Microbial Interactions between Marine Microalgae and Fungi: From Chemical Ecology to Biotechnological Possible Applications. Mar Drugs 2023; 21:md21050310. [PMID: 37233504 DOI: 10.3390/md21050310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
Chemical interactions have been shown to regulate several marine life processes, including selection of food sources, defense, behavior, predation, and mate recognition. These chemical communication signals have effects not only at the individual scale, but also at population and community levels. This review focuses on chemical interactions between marine fungi and microalgae, summarizing studies on compounds synthetized when they are cultured together. In the current study, we also highlight possible biotechnological outcomes of the synthetized metabolites, mainly for human health applications. In addition, we discuss applications for bio-flocculation and bioremediation. Finally, we point out the necessity of further investigating microalgae-fungi chemical interactions because it is a field still less explored compared to microalga-bacteria communication and, considering the promising results obtained until now, it is worthy of further research for scientific advancement in both ecology and biotechnology fields.
Collapse
Affiliation(s)
- Chiara Lauritano
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Acton n. 55, 80133 Naples, Italy
| | - Christian Galasso
- Department of Ecosustainable Marine Biotechnology, Calabria Marine Centre, Stazione Zoologica Anton Dohrn, C. da Torre Spaccata, 87071 Amendolara, Italy
| |
Collapse
|
47
|
Sharma A, Kaur I, Dheer D, Nagpal M, Kumar P, Venkatesh DN, Puri V, Singh I. A propitious role of marine sourced polysaccharides: Drug delivery and biomedical applications. Carbohydr Polym 2023; 308:120448. [PMID: 36813329 DOI: 10.1016/j.carbpol.2022.120448] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Numerous compounds, with extensive applications in biomedical and biotechnological fields, are present in the oceans, which serve as a prime renewable source of natural substances, further promoting the development of novel medical systems and devices. Polysaccharides are present in the marine ecosystem in abundance, promoting minimal extraction costs, in addition to their solubility in extraction media, and an aqueous solvent, along with their interactions with biological compounds. Certain algae-derived polysaccharides include fucoidan, alginate, and carrageenan, while animal-derived polysaccharides comprise hyaluronan, chitosan and many others. Furthermore, these compounds can be modified to facilitate their processing into multiple shapes and sizes, as well as exhibit response dependence to external conditions like temperature and pH. All these properties have promoted the use of these biomaterials as raw materials for the development of drug delivery carrier systems (hydrogels, particles, capsules). The present review enlightens marine polysaccharides providing its sources, structures, biological properties, and its biomedical applications. In addition to this, their role as nanomaterials is also portrayed by the authors, along with the methods employed to develop them and associated biological and physicochemical properties designed to develop suitable drug delivery systems.
Collapse
Affiliation(s)
- Ameya Sharma
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; University of Glasgow, College of Medical, Veterinary and Life Sciences, Glasgow, United Kingdom, G12 8QQ
| | - Divya Dheer
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - D Nagasamy Venkatesh
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Vivek Puri
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India.
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
48
|
Lu S, Zhang Z, Sharma AR, Nakajima-Shimada J, Harunari E, Oku N, Trianto A, Igarashi Y. Bulbiferamide, an Antitrypanosomal Hexapeptide Cyclized via an N-Acylindole Linkage from a Marine Obligate Microbulbifer. JOURNAL OF NATURAL PRODUCTS 2023; 86:1081-1086. [PMID: 36843290 DOI: 10.1021/acs.jnatprod.2c01083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
UV absorption spectroscopy-guided fractionation of the culture extract of a marine obligate bacterium of the genus Microbulbifer yielded a novel cyclic hexapeptide, bulbiferamide (1). NMR spectroscopic and mass spectrometric analyses revealed the structure of 1 to be a cyclic tetrapeptide appending a ureido-bridged two amino acid unit. Notably, Trp is a junction residue, forming on one hand a very rare N-aminoacylated indole linkage for cyclization and on the other hand connecting the ureido-containing tail structure, which is an unprecedented way of configuring peptides. The component amino acids were determined to be l by the advanced Marfey's method. Compound 1 displayed growth inhibitory activity against Trypanosoma cruzi epimastigotes with an IC50 value of 4.1 μM, comparable to the currently approved drug benznidazole, while it was not cytotoxic to P388 murine leukemia cells at 100 μM.
Collapse
Affiliation(s)
- Shiyang Lu
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Zhiwei Zhang
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Amit Raj Sharma
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Junko Nakajima-Shimada
- Department of Molecular and Cellular Parasitology, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Enjuro Harunari
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Naoya Oku
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Agus Trianto
- Department of Marine Science, Faculty of Fisheries and Marine Science, Diponegoro University, Tembalang Campus, St. Prof. Soedarto SH, Semarang, 50275 Central Java, Indonesia
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| |
Collapse
|
49
|
Buskes M, Coffin A, Troast DM, Stein R, Blanco MJ. Accelerating Drug Discovery: Synthesis of Complex Chemotypes via Multicomponent Reactions. ACS Med Chem Lett 2023; 14:376-385. [PMID: 37077380 PMCID: PMC10107905 DOI: 10.1021/acsmedchemlett.3c00012] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/20/2023] [Indexed: 04/21/2023] Open
Abstract
The generation of multiple bonds in one reaction step has attracted massive interest in drug discovery and development. Multicomponent reactions (MCRs) offer the advantage of combining three or more reagents in a one-pot fashion to effectively yield a synthetic product. This approach significantly accelerates the synthesis of relevant compounds for biological testing. However, there is a perception that this methodology will only produce simple chemical scaffolds with limited use in medicinal chemistry. In this Microperspective, we want to highlight the value of MCRs toward the synthesis of complex molecules characterized by the presence of quaternary and chiral centers. This paper will cover specific examples showing the impact of this technology toward the discovery of clinical compounds and recent breakthroughs to expand the scope of the reactions toward topologically rich molecular chemotypes.
Collapse
Affiliation(s)
- Melissa
J. Buskes
- Atavistik Bio 75 Sidney Street, Cambridge, Massachusetts 02139, United States
| | - Aaron Coffin
- Atavistik Bio 75 Sidney Street, Cambridge, Massachusetts 02139, United States
| | - Dawn M. Troast
- Atavistik Bio 75 Sidney Street, Cambridge, Massachusetts 02139, United States
| | - Rachel Stein
- Atavistik Bio 75 Sidney Street, Cambridge, Massachusetts 02139, United States
| | - Maria-Jesus Blanco
- Atavistik Bio 75 Sidney Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
50
|
Tsai KC, Chen CS, Su JH, Lee YC, Tseng YH, Wei WC. The Blockade of Mitogen-Activated Protein Kinase 14 Activation by Marine Natural Product Crassolide Triggers ICD in Tumor Cells and Stimulates Anti-Tumor Immunity. Mar Drugs 2023; 21:md21040225. [PMID: 37103364 PMCID: PMC10141386 DOI: 10.3390/md21040225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Immunogenic cell death (ICD) refers to a type of cell death that stimulates immune responses. It is characterized by the surface exposure of damage-associated molecular patterns (DAMPs), which can facilitate the uptake of antigens by dendritic cells (DCs) and stimulate DC activation, resulting in T cell immunity. The activation of immune responses through ICD has been proposed as a promising approach for cancer immunotherapy. The marine natural product crassolide, a cembranolide isolated from the Formosan soft coral Lobophytum michaelae, has been shown to have cytotoxic effects on cancer cells. In this study, we investigated the effects of crassolide on the induction of ICD, the expression of immune checkpoint molecules and cell adhesion molecules, as well as tumor growth in a murine 4T1 mammary carcinoma model. Immunofluorescence staining for DAMP ectolocalization, Western blotting for protein expression and Z′-LYTE kinase assay for kinase activity were performed. The results showed that crassolide significantly increased ICD and slightly decreased the expression level of CD24 on the surface of murine mammary carcinoma cells. An orthotopic tumor engraftment of 4T1 carcinoma cells indicated that crassolide-treated tumor cell lysates stimulate anti-tumor immunity against tumor growth. Crassolide was also found to be a blocker of mitogen-activated protein kinase 14 activation. This study highlights the immunotherapeutic effects of crassolide on the activation of anticancer immune responses and suggests the potential clinical use of crassolide as a novel treatment for breast cancer.
Collapse
Affiliation(s)
- Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112026, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Chia-Sheng Chen
- Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Jui-Hsin Su
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Yu-Ching Lee
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yu-Hwei Tseng
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112026, Taiwan
| | - Wen-Chi Wei
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112026, Taiwan
- Correspondence: ; Tel.: +886-2-28201999 (ext. 3561)
| |
Collapse
|