1
|
Khairani AF, Shalannandia WA, Bashari MH, Atik N. Aaptamine Alters Vimentin Expression and Migration Capability of Triple-Negative Breast Cancer Cells. J Exp Pharmacol 2025; 17:239-247. [PMID: 40420842 PMCID: PMC12105642 DOI: 10.2147/jep.s512099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
Purpose This study aimed to explore the effect of Aaptamine, an alkaloid derived from marine sponges, on the vimentin expression in both mRNA and protein levels and the migration capacity of breast cancer cells. Methods The triple-negative breast cancer cell line MDA-MB-231 was used for in vitro experiments. Low-cytotoxicity concentrations of Aaptamine (12.5 to 50 μM) were given to MDA-MB-231 cells. The vimentin mRNA and protein expression were evaluated using RT-qPCR and immunofluorescence, respectively, 72 h after Aaptamine treatment. The migration scratch assay was conducted for 48 hours. Results Aaptamine treatment in three different doses did not affect the expression of vimentin at the mRNA level while significantly lowering vimentin protein expression at the concentration of 12.5 µM. In addition, Aaptamine significantly inhibited breast cancer cell migration in a dose-dependent manner. Conclusion Aaptamine inhibits vimentin protein expression and demonstrates anti-migration activity in the sub-cytotoxic dose.
Collapse
Affiliation(s)
- Astrid Feinisa Khairani
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Widad Aghnia Shalannandia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Muhammad Hasan Bashari
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| | - Nur Atik
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
- Graduate School of Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, Indonesia
| |
Collapse
|
2
|
Kavaliauskas P, Acevedo W, Mickevičiūtė E, Grigalevičiūtė R, Grybaitė B, Sapijanskaitė-Banevič B, Pranaitytė G, Petraitis V, Petraitienė R, Mickevičius V. 3,3'-((3-Hydroxyphenyl)azanediyl)dipropionic Acid Derivatives as a Promising Scaffold Against Drug-Resistant Pathogens and Chemotherapy-Resistant Cancer. Pathogens 2025; 14:484. [PMID: 40430804 PMCID: PMC12115217 DOI: 10.3390/pathogens14050484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
The synthesis and antimicrobial and anticancer activity of 3,3'-((3-hydroxyphenyl)azanediyl)dipropionic acid derivatives (2-25) against drug-resistant bacterial pathogens and FaDu head and neck cancer cells were investigated. The derivatives were synthesized through various methods, including esterification, hydrazinolysis, and condensation reactions. The compounds demonstrated structure-dependent antimicrobial activity, predominantly targeting Gram-positive pathogens. Compounds containing 4-nitrophenyl, 1-naphthyl, and 5-nitro-2-thienyl groups exhibited enhanced activity against S. aureus and E. faecalis. Additionally, compounds 5, 6, and 25 showed antiproliferative activity in cisplatin-resistant FaDu cells at low micromolar concentrations. The in silico modeling revealed that compound 25 interacts with the HER-2 and c-MET proteins. These compounds also induced significant oxidative stress in FaDu cells and demonstrated low cytotoxic activity in non-cancerous HEK293 cells. These results highlight the potential of N-aryl-substituted β-amino acid derivatives as promising scaffolds for the further development of novel amino acid-based antimicrobial and anticancer agents targeting drug-resistant pathogens and cancers.
Collapse
Affiliation(s)
- Povilas Kavaliauskas
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Street 18, LT-47181 Kaunas, Lithuania;
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birstono Street 38A, LT-59116 Prienai, Lithuania
| | - Waldo Acevedo
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile;
| | - Eglė Mickevičiūtė
- Department of Information Systems, Kaunas University of Technology, LT-51368 Kaunas, Lithuania;
| | - Ramunė Grigalevičiūtė
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Street 18, LT-47181 Kaunas, Lithuania;
- Department of Animal Nutrition, Lithuanian University of Health Sciences, Tilzes Street 18, LT-47181 Kaunas, Lithuania
| | - Birutė Grybaitė
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| | - Birutė Sapijanskaitė-Banevič
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| | - Guoda Pranaitytė
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| | - Vidmantas Petraitis
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (V.P.); (R.P.)
| | - Rūta Petraitienė
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (V.P.); (R.P.)
| | - Vytautas Mickevičius
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| |
Collapse
|
3
|
Pang J, Yu Q, Wei H, Xia X, Lin Z, Du X, Wang C. Theoretical study on the structures and pharmacokinetic evaluation of verticillane-type diterpenes from soft coral Heteroxenia ghardaqensis. BMC Chem 2025; 19:122. [PMID: 40350455 PMCID: PMC12067919 DOI: 10.1186/s13065-025-01499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
The density functional theory (DFT) method ωB97XD/6-311++G(2d, p) was applied to calculate and analyze the geometric structures, spectral properties, frontier molecular orbitals, and molecular electrostatic potentials of 14 novel verticillane-type diterpenoids isolated from the soft coral Heteroxenia ghardaqensis. Additionally, reaction index analysis was conducted using conceptual density functional theory, and the drug-likeness of these compounds was evaluated using two different pharmacokinetic prediction platforms. The results showed that the hydroxyl hydrogen, secondary amine hydrogen, carbonyl oxygen, and hydroxyl oxygen in the molecules of these compounds have relatively high reactivity. Compounds 5, 8, and 9 exhibit significant anti-inflammatory activity and have similar electronic delocalization distribution characteristics, showing good stability and excellent biological activity, among which compound 5 demonstrates more significant drug potential. For compounds 2, 8, and 12 with hepatoprotective activity, through the analysis of comprehensive pharmacokinetic parameters and molecular docking data, compound 12 is considered more suitable as a potential hepatoprotective drug.
Collapse
Affiliation(s)
- Jiangmei Pang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Qinzhe Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Huining Wei
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiaoyun Xia
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zishan Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiandong Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Chaojie Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
4
|
Premarathna AD, Rjabovs V, Humayun S, Darko CNS, Robal M, Ahmed TAE, Hincke MT, Tuvikene R. Complex arabinose-containing polysaccharides from cyanobacterium Nostoc sp.: Extraction, structural characterization and antioxidant activity. Int J Biol Macromol 2025; 307:141793. [PMID: 40058433 DOI: 10.1016/j.ijbiomac.2025.141793] [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: 09/19/2024] [Revised: 02/08/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025]
Abstract
Freshwater Nostoc sp. polysaccharides (NSPs) have not been extensively studied, particularly regarding their structural and biological characteristics. This study assessed the extracted NSPs through a multi-faceted characterization approach using 1H and 13C NMR, FTIR spectroscopy, HP-SEC and HPAEC-PAD. The study presented novel insights into the extracted polysaccharide composition, extraction efficiency, and antioxidant activities. NSPs were extracted using two different ranges of heating regimes: cold (25 °C) and hot (95 °C), with hot extraction yielding up to 34.9 % of NSPs compared to 27.6 % from cold extraction. The NSPs were non-sulfated and mainly composed of four monosaccharide residues: (1 → 3)-β-d-mannopyranose-(1 → 4)-β-l-arabinopyranose-(1 → 4)-β-d-glucuronic acid-(1 → 4)-β-d-glucopyranose. Some glucose units were 6-O-methylated, and minor sidechain glycosylation by other monosaccharides was observed. β-d-glucuronic acid was present in higher amounts. The polysaccharides from the inner fluid (IF) fraction exhibited a higher molecular weight (up to 480 kDa) compared to outer layer (OL) fractions. The total phenolic content (TPC: 0.2 ± 0.0-2.8 ± 0.0 %), total sugar (9.1 ± 0.8-67.8 ± 0.4 %), and protein (1.2 ± 0.1-25.9 ± 0.6 %) content were determined. NSPs, especially from cold extraction (1B) exhibited the highest antioxidant activity, have potential applications in the skincare, food, pharmaceutical, and biotechnology industries due to their high yield, unique structural features.
Collapse
Affiliation(s)
- Amal D Premarathna
- School of Natural Sciences and Health, Tallinn University, Narva mnt 29, 10120 Tallinn, Estonia.
| | - Vitalijs Rjabovs
- National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia; Institute of Chemistry and Chemical Technology, Riga Technical University, Paula Valdena iela 3/7, LV-1048, Riga, Latvia
| | - Sanjida Humayun
- School of Natural Sciences and Health, Tallinn University, Narva mnt 29, 10120 Tallinn, Estonia
| | | | - Marju Robal
- School of Natural Sciences and Health, Tallinn University, Narva mnt 29, 10120 Tallinn, Estonia
| | - Tamer A E Ahmed
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ontario K1H 8M5, Canada; School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ontario K1H 8M5, Canada
| | - Maxwell T Hincke
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ontario K1H 8M5, Canada; Department of Innovation in Medical Education, Faculty of Medicine, University of Ottawa, Ontario K1H 8M5, Canada
| | - Rando Tuvikene
- School of Natural Sciences and Health, Tallinn University, Narva mnt 29, 10120 Tallinn, Estonia.
| |
Collapse
|
5
|
Qiu Y, Xu J, Liao W, Yang S, Wen Y, Farag MA, Zheng L, Zhao C. Ulvan derived from Ulva lactuca suppresses hepatocellular carcinoma cell proliferation through miR-542-3p-mediated downregulation of SLC35F6. Int J Biol Macromol 2025; 308:142252. [PMID: 40118430 DOI: 10.1016/j.ijbiomac.2025.142252] [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: 12/12/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Hepatocellular carcinoma (HCC) therapy still presents significant challenges, with a critical need for novel molecular targets and effective natural compound-based therapies. Despite its known oncogenic potential in other cancers, the role of SLC35F6 in HCC has not been previously reported, leaving a gap in our understanding of its function and therapeutic relevance. Here, we demonstrate that SLC35F6 is overexpressed in HCC and is associated with poor prognosis. Ulva lactuca polysaccharide (ULP), a natural extract with known antitumor properties, exerts its effects by upregulating miR-542-3p, which in turn inhibits SLC35F6 expression and significantly increases TP53 protein levels. Furthermore, TP53 is positively regulated by miR-542-3p, and our results indicate that SLC35F6 is a target gene of miR-542-3p. Knockdown of SLC35F6 in H22 and HepG2 cells markedly reduced cell growth while elevating TP53 expression, supporting SLC35F6 as a key regulatory factor in the miR-542-3p/TP53 axis. While this study did not confirm direct mutual regulation between SLC35F6 and TP53, our findings provide evidence that targeting SLC35F6 can suppress HCC progression. Collectively, these results identify SLC35F6 as a potential therapeutic target for HCC and provide mechanistic insights into its regulation through the miR-542-3p/SLC35F6/TP53 axis.
Collapse
Affiliation(s)
- Yinghui Qiu
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; School of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
| | - Jingxiang Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Wei Liao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Shuxin Yang
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yuxi Wen
- University of Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E32004 Ourense, Spain
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Lingjun Zheng
- School of Agriculture and Biology, Shanghai JiaoTong University, Shanghai 200240, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
6
|
Zhang S, Wang H, Sai C, Wang Y, Cheng Z, Zhang Z. The Cytotoxic Activity of Secondary Metabolites from Marine-Derived Penicillium spp.: A Review (2018-2024). Mar Drugs 2025; 23:197. [PMID: 40422787 DOI: 10.3390/md23050197] [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: 03/30/2025] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
Marine-derived Penicillium spp., including Penicillium citrinum, Penicillium chrysogenum, and Penicillium sclerotiorum, have emerged as prolific producers of structurally diverse secondary metabolites with cytotoxic activity. This review systematically categorizes 177 bioactive compounds isolated from marine Penicillium spp. between 2018 and 2024, derived from diverse marine environments such as sediments, animals, plants, and mangroves. These compounds, classified into polyketides, alkaloids, terpenoids, and steroids, exhibit a wide range of cytotoxic activities. Their potency is categorized as potent (<1 μM or <0.5 μg/mL), notable (1-10 μM or 0.5-5 μg/mL), moderate (10-30 μM or 5-15 μg/mL), mild (30-50 μM or 15-25 μg/mL), and negligible (>50 μM or >25 μg/mL). The current review highlights the promising role of marine Penicillium spp. as a rich repository for the discovery of anticancer agents and the advancement of marine-inspired drug development.
Collapse
Affiliation(s)
- Shuncun Zhang
- School of Pharmacy, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, China
- School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao 276800, China
| | - Huannan Wang
- School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao 276800, China
| | - Chunmei Sai
- School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao 276800, China
| | - Yan Wang
- School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao 276800, China
| | - Zhongbin Cheng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Zhen Zhang
- School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao 276800, China
| |
Collapse
|
7
|
Chang Y, Shen S, Zhang L, Zeng J, Sun J, Guo YW, Su MZ. 20-Acetylsinularolide B (ASB) From Lobophytum crassum Exhibits Anticancer Activity In Vitro Through IGF1R/PI3K/AKT/mTOR Pathway. Chem Biodivers 2025:e202500114. [PMID: 40246781 DOI: 10.1002/cbdv.202500114] [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: 01/10/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/19/2025]
Abstract
As lung cancer remains the leading cause of cancer-related deaths worldwide, the development of novel therapeutic drugs is essential. 20-Acetylsinularolide B (ASB) is a diterpene isolated from marine soft coral Lobophytum crassum. Our previous studies demonstrated that ASB exhibits growth-inhibitory effects on non-small cell lung cancer (NSCLC) cells. This study employed network pharmacology to predict ASB's potential targets in NSCLC treatment. The predicted target was validated using the cellular thermal shift assay (CETSA). In vitro anticancer activity was assessed through MTT and crystal violet assays for proliferation, along with Western blotting, cell cycle and apoptosis analysis, mitochondrial membrane potential, reactive oxygen species (ROS) levels, and nuclear morphology evaluation. Migration and invasion were evaluated using wound healing and Transwell assays. The results showed that ASB significantly arrests the cell cycle of H1299 cells at the G2/M phase by modulating the IGF1R/PI3K/AKT/mTOR signaling pathway, thereby inhibiting cell mitosis. Simultaneously, ASB promoted intracellular ROS production, reduced mitochondrial membrane potential, and ultimately induced cell apoptosis. In addition, ASB significantly inhibited the colony formation, migration, and invasion abilities of H1299 cells, which are closely associated with the function of the IGF1R target. These findings highlight the significant potential of ASB as a lead anticancer compound for NSCLC therapy.
Collapse
Affiliation(s)
- Yuanmin Chang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| | - Shoumao Shen
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
| | - Liting Zhang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| | - Jianang Zeng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| | - Jingyong Sun
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yue-Wei Guo
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Ming-Zhi Su
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| |
Collapse
|
8
|
Niu S, Huang S, Shi M, Luo Z, Shao Z, Hong B, Tian W. Discovery of new aphidicolin diterpenoids from the deep-sea-derived fungus Botryotinia fuckeliana with cytotoxic activity against human bladder cancer cells. Bioorg Chem 2025; 157:108311. [PMID: 40022848 DOI: 10.1016/j.bioorg.2025.108311] [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: 12/24/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Aphidicolin, characterized by a highly fused 6/6/5/6 tetracyclic diterpenoid skeleton, had been explored as a potential anticancer drug in clinical trials. However, its development has been constrained by poor solubility. The discovery of new aphidicolin derivatives offers promising prospects for anticancer drug development. In the present study, 37 new aphidicolin derivatives, designated as aphidicolins B1-B37 (1-37), together with 34 known analogues (38-71), were discovered from the ethyl acetate (EtOAc) extract of the deep-sea-derived fungus Botryotinia fuckeliana. Their structures, including absolute configurations, were determined by extensive analyses of spectroscopic data, the phenylglycine methyl ester (PGME) method, modified Mosher's method, and comparison of experimental and calculated electronic circular dichroism (ECD) data. Notably, aphidicolin B12 (12) features a 6/6/5/6/6 pentacyclic framework with an unprecedented γ-lactone ring E, while compound 31 contains a novel 6/6/5/6/5/5 hexacyclic system bearing an unprecedented tetrahydrofuran ring formed by an ether bridge between C-3 and C-6. All isolated aphidicolins were evaluated for their cytotoxic effects against T24 human bladder cancer cells. Among them, 11 diterpenoids showed stronger inhibitory activity than aphidicolin (60), especially compound 32, which exhibited an IC50 value of 1.9 μM, significantly more potent than 60 (IC50 = 27.6 μM). The structure-bioactivity relationships were also discussed. Further mechanistic studies revealed that 32 inhibits T24 cells proliferation by inducing cell cycle arrest in the G0/G1 phase, suggesting its potential as a therapeutic agent for bladder cancer treatment.
Collapse
Affiliation(s)
- Siwen Niu
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China.
| | - Shuhuan Huang
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Menglei Shi
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Zhuhua Luo
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Zongze Shao
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China
| | - Bihong Hong
- Technology Innovation Center for Exploitation of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen 361005, PR China.
| | - Wenjing Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China.
| |
Collapse
|
9
|
Selvaraj C, Santhosh R, Alothaim AS, Vijayakumar R, Desai D, Safi SZ, Singh SK. Advances in cancer therapy: unveil the immunomodulatory protein involved in signaling pathways as molecular targets. CHEMICAL PAPERS 2025. [DOI: 10.1007/s11696-025-04007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
|
10
|
Osorio-Ramírez MDC, Hernández-Melgar AG, Cembella AD, Maskrey BH, Díaz-Rubio LJ, Córdova-Guerrero I, Bernáldez-Sarabia J, González-Maya L, Esquivel-Rodríguez B, Bustos-Brito C, Licea-Navarro AF, Durán-Riveroll LM. Untargeted Metabolomic Analysis and Cytotoxicity of Extracts of the Marine Dinoflagellate Amphidinium eilatiense Against Human Cancer Cell Lines. Toxins (Basel) 2025; 17:150. [PMID: 40278648 PMCID: PMC12030893 DOI: 10.3390/toxins17040150] [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: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Members of the benthic marine dinoflagellate genus Amphidinium produce a variety of bioactive compounds, exhibiting potent cytotoxicity in cell assays. Crude methanolic extracts from three genetically distinct cultured strains of A. eilatiense J.J. Lee were screened for cytotoxicity against three human breast and four lung cancer cell lines to evaluate potential applications in anticancer therapy. A standard tetrazolium cell viability assay demonstrated that the methanolic crude extract (100 µg mL-1) from strain AeSQ181 reduced cell viability by 20-35% in five cancer cell lines. Further bioassay-guided fractionation of these crude extracts yielded non-polar fractions (FNP-5 and FNP-6) with particularly high cytotoxic activity against lung (H1563) and breast (MDA-MB-231) adenocarcinoma cell lines. Untargeted metabolomic analysis of cytotoxic fractions by liquid chromatography coupled with high-resolution mass spectrometry (LC-HRMS) revealed a much richer chemical diversity profile than previous toxigenicity studies on Amphidinium that exclusively focused on linear and cyclic polyethers and their macrolide analogs as putative cytotoxins. This untargeted metabolomic study showed substantial differences in chemical composition between the biologically active and non-active fractions. Preliminary biological and chemical characterization of these A. eilatiense fractions confirms that this species is a rich source of bioactive natural products with potential applications such as anticancer therapeutics.
Collapse
Affiliation(s)
- María del Carmen Osorio-Ramírez
- Departamento de Biotecnología Marina, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada 22860, Baja California, Mexico; (M.d.C.O.-R.); (A.D.C.)
| | - Alan Gerardo Hernández-Melgar
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada 22860, Baja California, Mexico; (A.G.H.-M.); (J.B.-S.); (A.F.L.-N.)
| | - Allan D. Cembella
- Departamento de Biotecnología Marina, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada 22860, Baja California, Mexico; (M.d.C.O.-R.); (A.D.C.)
- Department of Ecological Chemistry, Alfred-Wegener-Institut, Helmholtz-Zentrum für Polar-und Meeresforschung, 27570 Bremerhaven, Germany
| | - Benjamin H. Maskrey
- Centre for Environment, Fisheries and Aquaculture Science (CEFAS), Barrack Road, The Nothe, Weymouth DT4 8UB, UK;
| | - Laura Janeth Díaz-Rubio
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana 22424, Baja California, Mexico; (L.J.D.-R.); (I.C.-G.)
| | - Iván Córdova-Guerrero
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana 22424, Baja California, Mexico; (L.J.D.-R.); (I.C.-G.)
| | - Johanna Bernáldez-Sarabia
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada 22860, Baja California, Mexico; (A.G.H.-M.); (J.B.-S.); (A.F.L.-N.)
| | - Leticia González-Maya
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico;
| | - Baldomero Esquivel-Rodríguez
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico; (B.E.-R.); (C.B.-B.)
| | - Celia Bustos-Brito
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico; (B.E.-R.); (C.B.-B.)
| | - Alexei F. Licea-Navarro
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada 22860, Baja California, Mexico; (A.G.H.-M.); (J.B.-S.); (A.F.L.-N.)
| | - Lorena M. Durán-Riveroll
- SECIHTI-Departamento de Biotecnología Marina, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada 22860, Baja California, Mexico
| |
Collapse
|
11
|
Liu C, Gu F, Zou Z, Wang F, Li D, Song J, Hong Y, Wu X, Yang X, Liu WH, Liu G, Zhou Y, Liu Q. Marine-Derived Alternariol Suppresses Inflammation by Regulating T Cell Activation and Migration. Mar Drugs 2025; 23:133. [PMID: 40137319 PMCID: PMC11944012 DOI: 10.3390/md23030133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
T cells play pivotal roles in inflammation's initiation and progression. Exploring natural compounds that regulate T cell function is crucial for preventing and treating inflammation. Herein, we report that Alternariol (AOH), a marine-derived secondary metabolite, exerts an anti-inflammatory activity by targeting T cell function. Using an ovalbumin (OVA)-induced OT-II CD4+ T cell activation model, we demonstrated that AOH potently suppresses T cell proliferation and cytokine secretion, mildly promotes T cell apoptosis, and spares antigen presentation processes. Mechanistically, AOH controlled early T cell activation by inhibiting the expression of activation markers (CD69, CD25, CD44) and transcription factors (T-bet, Eomes), leading to impaired Th1 cytokine production. In vivo experiments revealed that AOH attenuated OVA-induced lung injury in mice by reducing immune cell infiltration in pulmonary tissues and draining lymph nodes. Notably, AOH dramatically suppressed OVA-specific T cells migrating to the inflammatory lung while impairing T-cell-mediated other immune cell infiltration. Collectively, AOH exhibited potent anti-inflammatory effects by modulating T cell proliferation, function, and migration, offering a promising therapeutic strategy for T-cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chenfeng Liu
- Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei 230031, China (F.W.); (D.L.)
| | - Fudie Gu
- Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (F.G.)
| | - Zhengbiao Zou
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou 571199, China (X.Y.)
| | - Fengli Wang
- Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei 230031, China (F.W.); (D.L.)
| | - Dashuai Li
- Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei 230031, China (F.W.); (D.L.)
| | - Jing Song
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xuhui Wu
- School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xianwen Yang
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou 571199, China (X.Y.)
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Guangming Liu
- Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (F.G.)
- Faculty of Marine Biology, Xiamen Ocean Vocational College, Xiamen 361102, China
| | - Yu Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qingmei Liu
- Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (F.G.)
| |
Collapse
|
12
|
Ferdous UT, Nurdin A, Ismail S, Shaari K, Norhana Balia Yusof Z. A comparative study on antioxidant properties, total phenolics, total flavonoid contents, and cytotoxic properties of marine green microalgae and diatoms. J Genet Eng Biotechnol 2025; 23:100456. [PMID: 40074430 PMCID: PMC11795137 DOI: 10.1016/j.jgeb.2024.100456] [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: 10/09/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 03/14/2025]
Abstract
Despite having valuable and novel metabolites, the marine microalgae species are still not thoroughly investigated for their pharmaceutical and nutraceutical importance. Therefore, this study was focused on investigating the crude extracts of marine green microalgae species, Tetraselmis sp., Nannochloropsis sp., and diatoms Chaetoceros sp., and Thalassiosira sp., isolated from the Malaysian coastal region in terms of their antioxidant activity, total phenolics, total flavonoid contents and cytotoxicity against human breast cancer cells, MCF-7. Among twenty-eight crude extracts, Tetraselmis ethanol and ethyl acetate extract showed the highest amount of total phenolic (19.87 mg GAE/g), and total flavonoid content (38.58 mg QE/g of extract), respectively. From the antioxidant assays, methanol and ethyl acetate extract of Tetraselmis sp. exhibited significantly higher (p < 0.05) antioxidant activities, revealed through DPPH (54.41 ± 1.18 mg Trolox Equivalent Antioxidant Capacity or TEAC/g extract) and ABTS (41.57 ± 0.83 mg TEAC/g extract) radical scavenging activities, respectively than the rest. Ethyl acetate extract of Tetraselmis sp. also showed high ferric reducing power (113.46 ± 4.83 mg TEAC/g extract). On the contrary, methanol and ethyl acetate extract of Chaetoceros sp. showed the highest cytotoxicity towards MCF-7 and reduced the cell viability to 21.26 % and 21.56 %, respectively. The data suggest that marine diatom Chaetoceros sp. has a good cytotoxic effect on MCF-7, while marine green microalga Tetraselmis sp. has good radical scavenging and ferric reduction capabilities, warranting further investigation along with their metabolic profiling, cancer cell killing mechanism and extensive in vivo study.
Collapse
Affiliation(s)
- Umme Tamanna Ferdous
- Center for Biosystems and Machines (IRC-BSM), King Fahd University of Petroleum & Minerals, Dhahran, Saudi Arabia
| | - Armania Nurdin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Laboratory of UPM-MAKNA Cancer Research (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Saila Ismail
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Khozirah Shaari
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Zetty Norhana Balia Yusof
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Aquatic Animal Health and Therapeutics Laboratory (AquaHealth), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Bioprocessing and Biomanufacturing Research Complex, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| |
Collapse
|
13
|
Pradhan B, Ki JS. Seaweed-derived laminarin and alginate as potential chemotherapeutical agents: An updated comprehensive review considering cancer treatment. Int J Biol Macromol 2025; 293:136593. [PMID: 39426775 DOI: 10.1016/j.ijbiomac.2024.136593] [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: 03/23/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Seaweed-derived bioactive substances such as polysaccharides have proven to be effective chemotherapeutic and chemopreventive agents. Laminarin and alginate antioxidant properties aid in the prevention of cancer through dynamic modulation of critical intracellular signaling pathways via apoptosis which produce low cytotoxicity and potential chemotherapeutic effects. Understanding the effects of laminarin and alginate on human cancer cells and their molecular roles in cell death pathways can help to develop a novel chemoprevention strategy. This review emphasizes the importance of apoptosis-modulating laminarin and alginate in a range of malignancies as well as their extraction, molecular structure, and weight. In addition, future nano-formulation enhancements for greater clinical efficacy are discussed. Laminarin and alginate are perfect ingredients because of their distinct physicochemical and biological characteristics and their use-based delivery systems in cancer. The effectiveness of laminarin and alginate against cancer and more preclinical and clinical trials will open up as new chemotherapeutic natural drugs which lead to established as potential cancer drugs.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea; Department of Botany, Model Degree College, Rayagada 765017, Odisha, India
| | - Jang-Seu Ki
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea.
| |
Collapse
|
14
|
Yan L, Liu Y, Huang Y, Sun X, Jiang H, Gu J, Xia J, Sun X, Sui X. Erianin inhibits the proliferation of lung cancer cells by suppressing mTOR activation and disrupting pyrimidine metabolism. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0385. [PMID: 39995202 PMCID: PMC11899589 DOI: 10.20892/j.issn.2095-3941.2024.0385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/03/2025] [Indexed: 02/26/2025] Open
Abstract
OBJECTIVE Erianin has potential anticancer activities, especially against lung cancer. The specific mechanisms underlying the anti-cancer effects, including the molecular targets and signaling pathways in lung cancer, remain poorly understood and necessitate further investigation. METHODS Lung cancer cell viability was evaluated using the CCK-8 assay. Flow cytometry was used to examine the effects of erianin on apoptosis and cell cycle progression. mRNA sequencing and metabolomics analysis were utilized to explore erianin-induced biological changes. Potential targets were identified and validated through molecular docking and Western blot analysis. The roles of mammalian target of rapamycin (mTOR) and carbamoyl-phosphate synthetase/aspartate transcarbamylase/dihydroorotase (CAD) in erianin-induced growth inhibition were studied using gene overexpression/knockdown techniques with uridine and aspartate supplementation confirming pyrimidine metabolism involvement. Additionally, lung cancer-bearing nude mouse models were established to evaluate the anti-lung cancer effects of erianin in vivo. RESULTS Erianin significantly inhibits the proliferation of lung cancer cells, induces apoptosis, and causes G2/M phase cell cycle arrest. Integrative analysis of mRNA sequencing and metabolomics data demonstrated that erianin disrupts pyrimidine metabolism in lung cancer cells. Notably, uridine supplementation mitigated the inhibitory effects of erianin, establishing a connection between pyrimidine metabolism and anticancer activity. Network pharmacology analyses identified mTOR as a key target of erianin. Erianin inhibited mTOR phosphorylation, thereby blocking downstream effectors (S6K and CAD), which are essential regulators of pyrimidine metabolism. CONCLUSIONS Erianin is a promising therapeutic candidate for lung cancer. Erianin likely inhibits lung cancer cell growth by disrupting pyrimidine metabolism by suppressing mTOR activation.
Collapse
Affiliation(s)
- Lili Yan
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Yanfen Liu
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Yufei Huang
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Xiaoyu Sun
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
- Department of Gastrointestinal & Pancreatic Surgery, Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Haiyang Jiang
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Jie Gu
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Jing Xia
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Xueni Sun
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
- Department of Gastrointestinal & Pancreatic Surgery, Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Xinbing Sui
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
- Department of Gastrointestinal & Pancreatic Surgery, Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
15
|
Li N, Xu J, Li Y, Elango J, Wu W. Polyethylene Glycolylation of the Purified Basic Protein (Protamine) of Squid ( Symplectoteuthis oualaniensis): Structural Changes and Evaluation of Proliferative Effects on Fibroblast. Int J Mol Sci 2025; 26:1869. [PMID: 40076495 PMCID: PMC11899872 DOI: 10.3390/ijms26051869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
In recent years, arginine-rich basic proteins have garnered significant attention due to their essential roles in various biological processes. However, the potential of marine-derived proteins in this domain remains largely unexplored. This study presents, for the first time, the isolation and purification of a 14.3 kDa protamine (SOP) from the mature spermatogonial tissues of Symplectoteuthis oualaniensis. Additionally, we obtained an 18.5 kDa PEGylated derivative, SOP-PEG. The physicochemical properties of both SOP and SOP-PEG were comprehensively characterized using SEM, FTIR, CD, and TGA. PEGylation markedly altered the surface morphology, secondary structure, and thermal stability of SOP. In vitro studies demonstrated that PEGylation significantly enhanced the biocompatibility of SOP, leading to improved proliferation of L-929 fibroblasts. Furthermore, both SOP and its PEGylated derivative (SOP-PEG) regulated the cell cycle, activated the PI3K-Akt signaling pathway, and modulated anti-apoptotic mechanisms, suggesting their potential to support cell survival and facilitate tissue regeneration. Notably, SOP-PEG exhibited superior bioactivity, likely attributable to its enhanced delivery efficiency conferred by PEGylation. Collectively, these findings underscore the promising applications of SOP and SOP-PEG in regenerative medicine and highlight the pivotal role of PEGylation in augmenting the bioactivity of SOP.
Collapse
Affiliation(s)
- Na Li
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
| | - Jiren Xu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
| | - Yu Li
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
| | - Jeevithan Elango
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
- Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Wenhui Wu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (J.X.); (Y.L.)
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
- Putuo Branch of International Combined Research Center for Marine Biological Sciences, Zhoushan 316104, China
| |
Collapse
|
16
|
Yassein AS, Elamary RB, Alwaleed EA. Biogenesis, characterization, and applications of Spirulina selenium nanoparticles. Microb Cell Fact 2025; 24:39. [PMID: 39915798 PMCID: PMC11804068 DOI: 10.1186/s12934-025-02656-6] [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: 11/14/2024] [Accepted: 01/16/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Nowadays, researchers are attracted to the phyco-synthesis of selenium nanoparticles (SeNPs) for biotechnological and medical applications as they possess many advantages such as safety, nutritional value, and easy biodegradation than gold, copper, and silver nanoparticles. Spirulina platensis is the preferred microalgae for SeNPs synthesis because it contains many compounds that increase their stability making them fit for biomedical treatments. RESULTS The biosynthesized Spirulina platensis selenium nanoparticles (SP-SeNPs) were spherical and crystalline, with a diameter of 65 nm and a net charge of -16.7 mV. Furthermore, they were surrounded by active groups responsible for stability. The DPPH radical scavenging test assessed the antioxidant efficacy of SP-SeNPs and exposed scavenging inhibition of 79.234% at a 100 µM dosage. ABTS and H2O2 radical scavenging assay is dose-dependent recording IC50 of 50.69 and 116.18 µg/ml, respectively. The antibacterial efficacy was investigated against 13 G-negative & G-positive bacteria. The study demonstrated that SP-SeNPs had antibacterial and antibiofilm efficiencies against the tested strains with MBC of 286-333 µg/ml. The highest percentages of biofilm inhibition were recorded for Bacillus subtilis and Klebsiella pneumoniae, with ratios of 78.8 and 69.9%, respectively. The prepared SP-SeNPS efficiently suppressed the tested fungi growth with MIC (350 µg/ml) and MFCs (480-950 µg/ml). Most notably, biogenic SeNPs effectively extended the clot formation period recording 170.4 S for prothrombin time (PT) and 195.6 S for the activated partial thromboplastin time (aPTT). SP-SeNPs reduced the cell viability of breast adenocarcinoma (MCF-7) and ovarian cancer (SKOV-3) cell lines with a percentage of 17.6009% and 14.9484% at a concentration of 100 ug/ml, respectively. Moreover, SP-SeNPs could effectively alleviate the inflammation in RAW 264.7 macrophages with a reduction percentage of 8.82% in Nitric oxide concentration. CONCLUSION The investigation findings reveal that SP-SeNPs are a hopeful antimicrobial, anti-tumor, anticoagulant, antioxidant, and anti-inflammatory factor that can be applied in medical cures.
Collapse
Affiliation(s)
- Asmaa S Yassein
- Faculty of Science, Botany and Microbiology Department, South Valley University, Qena, 83523, Egypt.
| | - Rokaia B Elamary
- Faculty of Science, Botany and Microbiology Department, Luxor University, Luxor, Egypt
| | - Eman A Alwaleed
- Faculty of Science, Botany and Microbiology Department, South Valley University, Qena, 83523, Egypt
| |
Collapse
|
17
|
Martínez H, Santos M, Pedraza L, Testera AM. Advanced Technologies for Large Scale Supply of Marine Drugs. Mar Drugs 2025; 23:69. [PMID: 39997193 PMCID: PMC11857447 DOI: 10.3390/md23020069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Marine organisms represent a source of unique chemical entities with valuable biomedical potentialities, broad diversity, and complexity. It is essential to ensure a reliable and sustainable supply of marine natural products (MNPs) for their translation into commercial drugs and other valuable products. From a structural point of view and with few exceptions, MNPs of pharmaceutical importance derive from the so-called secondary metabolism of marine organisms. When production strategies rely on marine macroorganisms, harvesting or culturing coupled with extraction procedures frequently remain the only alternative to producing these compounds on an industrial scale. Their supply can often be implemented with laboratory scale cultures for bacterial, fungal, or microalgal sources. However, a diverse approach, combining traditional methods with modern synthetic biology and biosynthesis strategies, must be considered for invertebrate MNPs, as they are usually naturally accumulated in only very small quantities. This review offers a comprehensive examination of various production strategies for MNPs, addressing the challenges related to supply, synthesis, and scalability. It also underscores recent biotechnological advancements that are likely to transform the current industrial-scale manufacturing methods for pharmaceuticals derived from marine sources.
Collapse
Affiliation(s)
- Henar Martínez
- Department of Organic Chemistry, School of Engineering (EII), University of Valladolid (UVa), Dr. Mergelina, 47002 Valladolid, Spain; (H.M.); (M.S.)
- G.I.R. Computational Chemistry Group, Department of Physical Chemistry and Inorganic Chemistry, Science Faculty, University of Valladolid (UVa), Paseo de Belén 7, 47011 Valladolid, Spain
| | - Mercedes Santos
- Department of Organic Chemistry, School of Engineering (EII), University of Valladolid (UVa), Dr. Mergelina, 47002 Valladolid, Spain; (H.M.); (M.S.)
- G.I.R. Bioforge, University of Valladolid (UVa), CIBER-BBN, Paseo de Belén 19, 47011 Valladolid, Spain
| | - Lucía Pedraza
- Department of Organic Chemistry, Science Faculty, University of Valladolid (UVa), Paseo de Belén 7, 47011 Valladolid, Spain;
| | - Ana M. Testera
- Department of Organic Chemistry, School of Engineering (EII), University of Valladolid (UVa), Dr. Mergelina, 47002 Valladolid, Spain; (H.M.); (M.S.)
- G.I.R. Bioforge, University of Valladolid (UVa), CIBER-BBN, Paseo de Belén 19, 47011 Valladolid, Spain
| |
Collapse
|
18
|
Kumar C P, Banumathi, Satyanarayan ND, Prasad SR, Achur RN, Prabhakar BT. A quinoline derivative exerts antineoplastic efficacy against solid tumour by inducing apoptosis and anti-angiogenesis both in vitro and in vivo. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03830-8. [PMID: 39912901 DOI: 10.1007/s00210-025-03830-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025]
Abstract
Cancer is a heterogeneous and multicomplex disease with the highest morbidity and mortality rate. The targeting of tumour progression with drugs is a very well-established treatment strategy. Despite these, due to the failure of commonly used drugs in combating cancer, new drugs need to be screened and established for better therapeutic approach. With this rationale, the current investigation was aimed to develop quinoline compound (QC) derivatives as anti-tumour molecules. In this extended study, a series of QC analogues were subjected to anti proliferative assays through cell-based screening and evaluated its mechanism of action through apoptotic and anti-angiogenic assays. The change in cell behaviour was assessed through gene expression analysis using qRT-PCR and immunoblot analysis. Further, in vivo solid tumour model was developed and the anti-tumour potential of QC-4 was verified with gene expression studies. The results suggested that QC-4 exhibited significant cytotoxic effect, particularly against human lung adenocarcinoma cell lines and murine Ehrlich Ascites Carcinoma cells. The QC-4 induced condensation, nuclear damage and changes in membrane integrity resulted in apoptosis and neovascularisation inhibition. The modulation of apoptotic and angiogenic genes such as BAX, BAD, p53 and MMP-2 and 9 further supported the molecular cause of cytotoxicity induced by QC-4. The regression of in vivo solid tumour with extended survivability warranted the in vitro results and the gene expression patterns were additionally supportive. Overall, the QC-4 analogue exhibits the anti-neoplastic with a multi-target approach, reserving its capacity to be developed into a new class of the anticancer molecules.
Collapse
Affiliation(s)
- Pradeepa Kumar C
- Department of Biochemistry, Jnana Sahyadri, Kuvempu University, Shankaraghatta, 577451, Shimoga, Karnataka, India
| | - Banumathi
- Molecular Biomedicine Laboratory, Post Graduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577 203, Karnataka, India
| | - N D Satyanarayan
- Department of Pharmaceutical Chemistry, Post-Graduate Centre, Kuvempu University, Kadur, 577548, Chikmagalur Dist, Karnataka, India
| | - Sakshith Raghavendra Prasad
- Department of Pharmaceutical Chemistry, Post-Graduate Centre, Kuvempu University, Kadur, 577548, Chikmagalur Dist, Karnataka, India
| | - Rajeshwara N Achur
- Department of Biochemistry, Jnana Sahyadri, Kuvempu University, Shankaraghatta, 577451, Shimoga, Karnataka, India.
- INTI International University, Nilai, Malaysia.
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Post Graduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577 203, Karnataka, India.
| |
Collapse
|
19
|
Deng R, Zong GF, Wang X, Yue BJ, Cheng P, Tao RZ, Li X, Wei ZH, Lu Y. Promises of natural products as clinical applications for cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189241. [PMID: 39674416 DOI: 10.1016/j.bbcan.2024.189241] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Cancer represents a substantial threat to human health and mortality, necessitating the development of novel pharmacological agents with innovative mechanisms of action. Consequently, extensive research has been directed toward discovering new anticancer compounds derived from natural sources, including plants, microbes, and marine organisms. This review offers a comprehensive analysis of natural anticancer agents that are either currently undergoing clinical trials or have been integrated into clinical practice. A comprehensive understanding of the historical origins of natural anticancer agents, alongside traditional targets for tumor treatment and the distinct characteristics of cancer, can significantly facilitate researchers in the discovery and development of innovative anticancer drugs for clinical use. Furthermore, the exploration of microbial and marine sources is currently a prominent area of focus in the clinical application and advancement of new anticancer therapies. Detailed classification and elucidation of the functions and antitumor properties of these natural products are essential. It is imperative to comprehensively summarize and comprehend the natural anticancer drugs that have been and continue to be utilized in clinical settings.
Collapse
Affiliation(s)
- Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Nanjing Integrated Traditional Chinese And Western Medicine Hospital, Nanjing 210018. China
| | - Gang-Fan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Bing-Jie Yue
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Rui-Zhi Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhong-Hong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
20
|
El-Seedi HR, Refaey MS, Elias N, El-Mallah MF, Albaqami FMK, Dergaa I, Du M, Salem MF, Tahir HE, Dagliaa M, Yosri N, Zhang H, El-Seedi AH, Guo Z, Khalifa SAM. Marine natural products as a source of novel anticancer drugs: an updated review (2019-2023). NATURAL PRODUCTS AND BIOPROSPECTING 2025; 15:13. [PMID: 39853457 PMCID: PMC11759743 DOI: 10.1007/s13659-024-00493-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/16/2024] [Indexed: 01/26/2025]
Abstract
Marine natural products have long been recognized as a vast and diverse source of bioactive compounds with potential therapeutic applications, particularly in oncology. This review provides an updated overview of the significant advances made in the discovery and development of marine-derived anticancer drugs between 2019 and 2023. With a focus on recent research findings, the review explores the rich biodiversity of marine organisms, including sponges, corals, algae, and microorganisms, which have yielded numerous compounds exhibiting promising anticancer properties. Emphasizing the multifaceted mechanisms of action, the review discusses the molecular targets and pathways targeted by these compounds, such as cell cycle regulation, apoptosis induction, angiogenesis inhibition, and modulation of signaling pathways. Additionally, the review highlights the innovative strategies employed in the isolation, structural elucidation, and chemical modification of marine natural products to enhance their potency, selectivity, and pharmacological properties. Furthermore, it addresses the challenges and opportunities associated with the development of marine-derived anticancer drugs, including issues related to supply, sustainability, synthesis, and clinical translation. Finally, the review underscores the immense potential of marine natural products as a valuable reservoir of novel anticancer agents and advocates for continued exploration and exploitation of the marine environment to address the unmet medical needs in cancer therapy.
Collapse
Affiliation(s)
- Hesham R El-Seedi
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, 42351, Madinah, Saudi Arabia.
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China.
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu Education Department, Jiangsu University, Nanjing, 210024, China.
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom, 31100107, Egypt.
| | - Mohamed S Refaey
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Nizar Elias
- Department of Laboratory Medicine, Faculty of Medicine, University of Kalamoon, P.O. Box 222, Dayr Atiyah, Syria
| | - Mohamed F El-Mallah
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom, 31100107, Egypt
| | - Faisal M K Albaqami
- Biology Department, Faculty of Science, Islamic University of Madinah, 42351, Madinah, Saudi Arabia.
| | | | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, China
| | - Mohamed F Salem
- Department of Environmental Biotechnology, Genetic Engineering and Biotechnology Research Institute, GEBRI, University of Sadat City, P.O.Box:79, Sadat City, Egypt
| | - Haroon Elrasheid Tahir
- Agricultural Product Processing and Storage Lab, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Maria Dagliaa
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131, Naples, NA, Italy
| | - Nermeen Yosri
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
- Chemistry Department of Medicinal and Aromatic Plants, Research Institute of Medicinal and Aromatic Plants (RIMAP), Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Hongcheng Zhang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Awg H El-Seedi
- International IT College of Sweden Stockholm, Arena Academy, Hälsobrunnsgatan 6, 11361, Stockholm, Sweden
| | - Zhiming Guo
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - 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.
| |
Collapse
|
21
|
Vujović T, Paradžik T, Babić Brčić S, Piva R. Unlocking the Therapeutic Potential of Algae-Derived Compounds in Hematological Malignancies. Cancers (Basel) 2025; 17:318. [PMID: 39858100 PMCID: PMC11763723 DOI: 10.3390/cancers17020318] [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: 12/21/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Algae are a rich source of bioactive compounds that have a wide range of beneficial effects on human health and can show significant potential in the treatment of hematological malignancies such as leukemia, lymphoma, and multiple myeloma. These diseases often pose a therapeutic challenge despite recent advances in treatment (e.g., the use of immunomodulatory drugs, proteasome inhibitors, CD38 monoclonal antibodies, stem cell transplant, and targeted therapy). A considerable number of patients experience relapses or resistance to the applied therapies. Algal compounds, alone or in combination with chemotherapy or other more advanced therapies, have exhibited antitumor and immunomodulatory effects in preclinical studies that may improve disease outcomes. These include the ability to induce apoptosis, inhibit tumor growth, and improve immune responses. However, most of these studies are conducted in vitro, often without in vivo validation or clinical trials. This paper summarizes the current evidence on the in vitro effects of algae extracts and isolated compounds on leukemia, lymphoma, and myeloma cell lines. In addition, we address the current advances in the application of algae-derived compounds as targeted drug carriers and their synergistic potential against hematologic malignancies.
Collapse
Affiliation(s)
- Tamara Vujović
- Laboratory for Aquaculture Biotechnology, Division of Materials Chemistry, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (T.V.); (S.B.B.)
| | - Tina Paradžik
- Department of Physical Chemistry, Rudjer Boskovic Insitute, 10000 Zagreb, Croatia;
| | - Sanja Babić Brčić
- Laboratory for Aquaculture Biotechnology, Division of Materials Chemistry, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (T.V.); (S.B.B.)
| | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| |
Collapse
|
22
|
Lee EG, Yim SK, Kang SM, Ahn BJ, Kim CK, Lee M, Tark D, Lee GH. Phlorofucofuroeckol-A: A Natural Compound with Potential to Attenuate Inflammatory Diseases Caused by Airborne Fine Dust. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:165. [PMID: 39859147 PMCID: PMC11767036 DOI: 10.3390/medicina61010165] [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: 11/22/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Background and Objectives: Persistent exposure to airborne fine dust (FD) particles contributing to air pollution has been linked to various human health issues, including respiratory inflammation, allergies, and skin diseases. We aimed to identify potential seaweed anti-inflammatory bioactive reagents and determine their effects on systemic inflammatory responses induced by FD particles. Materials and Methods: While exploring anti-inflammatory bioactive reagents, we purified compounds with potential anti-inflammatory effects from the seaweed extracts of Ecklonia cava, Ecklonia stolonifera, and Codium fragile. Structural analyses of the purified compounds siphonaxanthin (Sx), fucoxanthin (Fx), dieckol (Dk), and phlorofucofuroeckol-A (PFF-A) were performed using NMR and LC-MS/MS. Results: Notably, these compounds, especially PFF-A, showed significant protective effects against FD-induced inflammatory responses in RAW 264.7 cells without cytotoxicity. Further investigation of inflammatory-associated signaling demonstrated that PFF-A inhibited IL-1β expression by modulating the NF-κB/MAPK signal pathway in FD-induced RAW 264.7 cells. Additionally, gene profiling revealed the early activation of various signature genes involved in the production of inflammatory cytokines and chemokines using gene profiling. Treatment with PFF-A markedly reduced the expression levels of pro-inflammatory and apoptosis-related genes and even elevated the Bmp gene families. Conclusions: These results suggested that PFF-A is a potential natural therapeutic candidate for managing FD-induced inflammatory response.
Collapse
Affiliation(s)
- Eun-Gyeong Lee
- Laboratory for Infection Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea; (E.-G.L.); (S.-M.K.)
| | - Sung-Kun Yim
- Marine Biotechnology Research Center, Jeonnam Bioindustry Foundation, Wando-gun 59108, Republic of Korea; (S.-K.Y.); (B.J.A.)
| | - Sang-Min Kang
- Laboratory for Infection Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea; (E.-G.L.); (S.-M.K.)
| | - Byung Jae Ahn
- Marine Biotechnology Research Center, Jeonnam Bioindustry Foundation, Wando-gun 59108, Republic of Korea; (S.-K.Y.); (B.J.A.)
| | - Chang-Kwon Kim
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea; (C.-K.K.); (M.L.)
| | - Mina Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea; (C.-K.K.); (M.L.)
| | - Dongseob Tark
- Laboratory for Infection Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea; (E.-G.L.); (S.-M.K.)
| | - Gun-Hee Lee
- Laboratory for Infection Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea; (E.-G.L.); (S.-M.K.)
| |
Collapse
|
23
|
Debnath A, Mazumder R, Singh AK, Singh RK. Identification of novel cyclin-dependent kinase 4/6 inhibitors from marine natural products. PLoS One 2025; 20:e0313830. [PMID: 39813224 PMCID: PMC11734976 DOI: 10.1371/journal.pone.0313830] [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] [Received: 09/01/2024] [Accepted: 10/31/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) are crucial regulators of cell cycle progression and represent important therapeutic targets in breast cancer. This study employs a comprehensive computational approach to identify novel CDK4/6 inhibitors from marine natural products. We utilized structure-based virtual screening of the CMNPD database and MNP library, followed by rigorous filtering based on drug-likeness criteria, PAINS filter, ADME properties, and toxicity profiles. From an initial hit of 9,497 compounds, 2,344 passed drug-likeness and PAINS filters. Further ADME filtering yielded 50 compounds, of which 25 exhibited non-toxic profiles. These 25 candidates underwent consensus molecular docking using seven distinct algorithms: AutoDockTools 4.2, idock, LeDock, Qvina 2, Smina, AutoDock Vina 1.2.0, PLANTS, and rDock. Based on these results, six top-scoring compounds were selected for comprehensive 500 nanosecond all-atom molecular dynamics simulations to evaluate their structural stability and interactions with CDK4/6. Our analysis revealed that compounds CMNPD11585 and CMNPD2744 demonstrated superior stability in their interactions with CDK4/6, exhibiting lower RMSD and RMSF values, more favorable binding free energies, and persistent hydrogen bonding patterns. These compounds also showed lower Solvent Accessible Surface Area values, indicating better compatibility with the CDK4/6 active site. Subsequent in-vitro studies using MTT assays on MCF-7 breast cancer cells confirmed the cytotoxic effects of these compounds, with CMNPD11585 showing the highest potency, followed by CMNPD2744.
Collapse
Affiliation(s)
- Abhijit Debnath
- Noida Institute of Engineering and Technology [Pharmacy Institute], Institutional Area, Greater Noida, Uttar Pradesh, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology [Pharmacy Institute], Institutional Area, Greater Noida, Uttar Pradesh, India
| | - Anil Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajesh Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
24
|
Murakami R, Mori T, Murata K, Fuwa H. Total Synthesis of Exiguolide Stereoisomers: Impact of Stereochemical Permutation on Reactivity, Conformation, and Biological Activity. J Org Chem 2025; 90:753-767. [PMID: 39718544 DOI: 10.1021/acs.joc.4c02707] [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: 12/25/2024]
Abstract
(-)-Exiguolide is a marine macrolide natural product with potent anticancer activity. In this study, the total synthesis of exiguolide stereoisomers, (9R)-exiguolide, (9R,13S)-exiguolide, and (9R,13S,19R)-exiguolide, was achieved by capitalizing on our macrocyclization/transannular pyran cyclization strategy. The impact of the stereochemical permutation on the reactivity of advanced intermediates, the conformation of the macrocyclic skeleton, and the antiproliferative activity against human cancer cells were investigated in detail. The total synthesis of (9R,13S)-exiguolide and (9R,13S,19R)-exiguolide was completed in much the same way as that of the parent natural product using stereoisomeric building blocks. Nevertheless, the reactivity of the (9R,13S)- and (9R,13S,19R)-series of intermediates in macrocyclic ring-closing metathesis and transannular pyran-forming reactions was significantly different from that of naturally configured counterparts. The conformation of exiguolide stereoisomers, deduced by means of NMR spectroscopic analysis and DFT calculations, was clearly different from that of the parent natural product. Evaluation of the antiproliferative activity of exiguolide and its stereoisomers suggested the importance of the stereochemistry of the macrocyclic skeleton.
Collapse
Affiliation(s)
- Reika Murakami
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Tomo Mori
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Keisuke Murata
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Haruhiko Fuwa
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| |
Collapse
|
25
|
Bajhal S, Seenivasan B, Arockiaraj J, Sundaramurthy A. Antimicrobial Potentials of Haliclona fibulata Derived Compounds for Combatting Drug-Resistant Bacterial Infections in Zebrafish Model. Chem Biodivers 2025:e202402717. [PMID: 39780661 DOI: 10.1002/cbdv.202402717] [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: 10/23/2024] [Revised: 01/01/2025] [Accepted: 01/08/2025] [Indexed: 01/11/2025]
Abstract
The rising threat of antimicrobial resistance among pathogens highlights the critical need for novel antimicrobial agents. This study explores the potential of natural products by investigating n-hexane extracts from the marine sponge Haliclona fibulata (HF) for their antibacterial efficacy. The well diffusion method of HF extract showed significant antibacterial activity against Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus with inhibition zones of 20 ± 0.66, 27 ± 0.58, and 22 ± 0.33 mm, respectively. Scanning electron microscopy investigations confirmed the disruptive impact of the extracts on bacterial cell morphology. Further investigations using P. aeruginosa-infected zebrafish model revealed that HF treatment improved survival rates in a dose-dependent manner. Additionally, oxidative stress and apoptosis were significantly reduced in the 100 µg/mL (HF100) treatment group, alongside downregulation of inflammatory markers such as il-1β, nf-kβ, tnf-α, and inos. Histological assessments showed reduced intestinal damage and fewer circulating macrophages in HF100-treated zebrafish larvae. Identifying potent bioactive compounds within the extracts suggests that HF could be a valuable source of natural antimicrobials, warranting further exploration for developing treatments against pathogenic infections.
Collapse
Affiliation(s)
- Sakshi Bajhal
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
- Biomaterials Research Laboratory (BMRL), Department of Chemical Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| | - Boopathi Seenivasan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| | - Anandhakumar Sundaramurthy
- Biomaterials Research Laboratory (BMRL), Department of Chemical Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, India
| |
Collapse
|
26
|
Ji R, Zha X, Zhou S. Marine Fungi: A Prosperous Source of Novel Bioactive Natural Products. Curr Med Chem 2025; 32:992-1006. [PMID: 37885109 DOI: 10.2174/0109298673266304231015070956] [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: 06/08/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023]
Abstract
As the number of viruses, bacteria, and tumors that are resistant to drugs continues to rise, there is a growing need for novel lead compounds to treat them. Marine fungi, due to their unique secondary metabolic pathways and vast biodiversity, have become a crucial source for lead compounds in drug development. This review utilizes bibliometric methods to analyze the research status of natural products from marine fungi in the past decade, revealing the hotspots and trends in this field from Web of Science database. Furthermore, this review summarizes the biological activities and effects on molecular mechanisms of novel natural compounds isolated from marine fungi in the past five years. These novel compounds belong to six different structural classes, such as alkaloids, terpenoids, anthraquinones, polyketones, etc. They also exhibited highly potent biological properties, including antiviral, antitumor, antibacterial, antiinflammatory, and other properties. This review demonstrates the hotspots and trends of marine fungi research in recent years, as well as the variety of chemical structure and biological activities of their natural products, and it may provide guidance for those interested in discovering new drugs from marine fungi and specific targeting mechanisms.
Collapse
Affiliation(s)
- Rong Ji
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Key Laboratory of Tropical Molecular Pharmacology and Advanced Diagnostic Technology, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199, China
| | - Xiangru Zha
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Key Laboratory of Tropical Molecular Pharmacology and Advanced Diagnostic Technology, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199, China
| | - Songlin Zhou
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Key Laboratory of Tropical Molecular Pharmacology and Advanced Diagnostic Technology, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199, China
| |
Collapse
|
27
|
Manivannan HP, Veeraraghavan VP, Francis AP. Identification of Novel Marine Bioactive Compound as Potential Multiple Inhibitors in Triple-negative Breast Cancer - An in silico Approach. Curr Comput Aided Drug Des 2025; 21:375-402. [PMID: 38231067 DOI: 10.2174/0115734099287118240102112337] [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: 10/14/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer lacking specific receptors, with dysregulated and overactivated Hedgehog (Hh) and mTOR/PI3K/AKT signaling pathways as potential therapeutic targets. OBJECTIVE This study aimed to identify potential inhibitors among 53 alkaloids derived from 9 marine bryozoans using in silico approaches. It sought to analyze their impact on key signaling targets and their potential for future experimental validation. METHODS In this research, selected targets were evaluated for protein-protein interactions, coexpression survival, and expression profiles. The protein expression was validated through the Human Protein Atlas (HPA) database and druggability through DGIdb. Online web servers were employed to assess drug-likeness, physiochemical properties, pharmacokinetics, and toxicological characteristics of the compounds. Molecular docking and dynamic simulations were carried out for ligand-protein interactions. Common Pharmacophore features, bioavailability, bioactivity, and biological activity spectrum (BAS) were also analyzed. RESULTS Out of the 13 compounds studied, 10 displayed strong binding affinity with binding energies ranging from >-6.5 to <-8 Kcal/mol across all targets. Molecular dynamics simulations provided insights into Amathamide E's stability and conformational changes. Pharmacophore modeling revealed common features in 14 compounds potentially responsible for their biological activity. CONCLUSION Our findings indicate the potential of marine-derived compounds as TNBC inhibitors. Further in vitro and in vivo validation is necessary to establish their effectiveness and explore their role as novel anti-TNBC agents.
Collapse
Affiliation(s)
- Hema Priya Manivannan
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | - Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| |
Collapse
|
28
|
Afkhami H, Yarahmadi A, Bostani S, Yarian N, Haddad MS, Lesani SS, Aghaei SS, Zolfaghari MR. Converging frontiers in cancer treatment: the role of nanomaterials, mesenchymal stem cells, and microbial agents-challenges and limitations. Discov Oncol 2024; 15:818. [PMID: 39707033 DOI: 10.1007/s12672-024-01590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/14/2024] [Indexed: 12/23/2024] Open
Abstract
Globally, people widely recognize cancer as one of the most lethal diseases due to its high mortality rates and lack of effective treatment options. Ongoing research into cancer therapies remains a critical area of inquiry, holding significant social relevance. Currently used treatment, such as chemotherapy, radiation, or surgery, often suffers from other problems like damaging side effects, inaccuracy, and the lack of ability to clear tumors. Conventional cancer therapies are usually imprecise and ineffective and usually develop resistance to treatments and cancer recurs. Cancer patients need fresh and innovative treatment that can reduce side effects while maximizing effectiveness. In recent decades several breakthroughs in these, and other areas of medical research, have paved the way for new avenues of fighting cancer including more focused and more effective alternatives. This study reviews exciting possibilities for mesenchymal stem cells (MSCs), nanomaterials, and microbial agents in the modern realm of cancer treatment. Nanoparticles (NPs) have demonstrated surprisingly high potential. They improve drug delivery systems (DDS) significantly, enhance imaging techniques remarkably, and target cancer cells selectively while protecting healthy tissues. MSCs play a double role in tissue repair and are a vehicle for novel cancer treatments such as gene treatments or NPs loaded with therapeutic agents. Additionally, therapies utilizing microbial agents, particularly those involving bacteria, offer an inventive approach to cancer treatment. This review investigates the potential of nanomaterials, MSCs, and microbial agents in addressing the shortcomings of conventional cancer therapies. We will also discuss the challenges and limitations of using these therapeutic approaches.
Collapse
Affiliation(s)
- Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Shoroq Bostani
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | - Nahid Yarian
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | | | - Shima Sadat Lesani
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | | | | |
Collapse
|
29
|
Ravichandiran P, Martyna A, Kochanowicz E, Maroli N, Kubiński K, Masłyk M, Boguszewska-Czubara A, Ramesh T. In Vitro and In Vivo Biological Evaluation of Novel 1,4-Naphthoquinone Derivatives as Potential Anticancer Agents. ChemMedChem 2024; 19:e202400495. [PMID: 39136593 DOI: 10.1002/cmdc.202400495] [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: 06/29/2024] [Revised: 08/13/2024] [Indexed: 10/16/2024]
Abstract
A novel library of naphthoquinone derivatives (3-5 aa) was synthesized and evaluated for their anticancer properties. Specifically, compounds 5 i, 5 l, 5 o, 5 q, 5 r, 5 s, 5 t, and 5 v demonstrated superior cytotoxic activity against the cancer cell lines that were studied. All the studied compounds exhibited a higher selectivity index (SI) and a favourable safety profile than the standard drug doxorubicin. Notably, compound 5 v displayed a greater cytotoxic effect on MCF-7 cells (IC50=1.2 μM, and 0.9 μM at 24 h and 48 h, respectively) compared to the standard drug doxorubicin (IC50=2.4 μM, and 2.1 μM at 24 h and 48 h, respectively). To further investigate the mechanism of cytotoxic effect, additional anticancer studies were conducted with 5 v in MCF-7 cells. The studies are including morphological changes, AO/EB (acridine orange/ethidium bromide) double staining, apoptosis analysis, cell colony assay, SDS-PAGE and Western blotting, cell cycle analysis, and detecting reactive oxygen species (ROS) assay. The findings showed that 5 v triggered cytotoxic effects in MCF-7 cells through the initiation of cell cycle arrest at the G1/S phase and necrosis. In vivo ecotoxicity studies indicated that 5 v had lower toxicity towards zebrafish larvae (LC50=50.15 μM) and had an insignificant impact on cardiac functions. In vivo xenotransplantation of MCF-7 cells in zebrafish larvae demonstrated a significant reduction in tumour volume in the xenograft. Approximately 95 % of the zebrafish larvae with 5 v xenografts survived after 10 days of the treatment. Finally, a computational modelling study was conducted on four protein receptors, namely ER, EFGR, BRCA1, and VEFGR2. The findings highlight the importance of the aminonaphthoquinone moiety, amide linkage, and propyl thio moiety in enhancing the anticancer properties. 5 v exhibited superior drug-likeness features and docking scores (-9.1, -7.1, -8.9, and -10.9 kcal/mol) compared to doxorubicin (-7.2, -6.1, -6.9, and -7.3 kcal/mol) against ER, EFGR, BRCA1, and VEGFR2 receptors, respectively. Therefore, the notable antitumor effects of naphthoquinone derivatives (3-5 aa) suggest that these molecular frameworks may play a role in the development of promising anticancer agents for cancer treatment.
Collapse
Affiliation(s)
- Palanisamy Ravichandiran
- R&D Education Center for Whole Life Cycle R&D of Fuel Cell Systems, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Department of Life Science, Department of Energy Storage/Conversion Engineering of Graduate School, Hydrogen and Fuel Cell Research Center, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Present Address: Analytical, HP Green R & D Centre, Hindustan Petroleum Corporation Limited, KIADB Industrial Area, Devangundi, Hoskote, Bengaluru, Karnataka, 562114, India
| | - Aleksandra Martyna
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Elżbieta Kochanowicz
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Nikhil Maroli
- Department of Physics and Astronomy, University of Delaware, Newark, DE, 19716, USA
| | - Konrad Kubiński
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Ul. Chodźki 4 A, 20-093, Lublin, Poland
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
30
|
Ren Q, Chen G, Wan Q, Xiao L, Zhang Z, Feng Y. Unravelling the role of natural and synthetic products as DNA topoisomerase inhibitors in hepatocellular carcinoma. Bioorg Chem 2024; 153:107860. [PMID: 39442463 DOI: 10.1016/j.bioorg.2024.107860] [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: 05/17/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Topoisomerase is a ubiquitous enzyme in the control of DNA chain topology. There have been extensive research on topoisomerase inhibitors derived from natural sources, which act as partial inducers of tumor cell apoptosis. However, their specific efficacy in treating hepatocellular carcinoma is relatively unexplored. Hence, this comprehensive review focuses on the structural characteristics and anti-cancer properties of topoisomerase inhibitors in hepatocellular carcinoma. Furthermore, this review is also elucidating the mechanism of action, structure-activity relationships, therapeutic limitations, stage of clinical trials of described classes of natural bioactive compounds as well as their potential application in cancer chemotherapies. This broad understanding of topoisomerase medical biology will provide indispensable framework for enhancing the efficiency of rational anti-hepatocellular carcinoma drug discovery.
Collapse
Affiliation(s)
- Qing Ren
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Qi Wan
- Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Liangman Xiao
- Acupuncture Rehabilitation Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhitong Zhang
- Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.
| |
Collapse
|
31
|
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] [MESH Headings] [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
|
32
|
Ding M, He M, Li D, Ding S, Dong C, Zhao H, Song H, Hong K, Zhu H. A Marine-Derived Small Molecule Inhibits Prostate Cancer Growth by Promoting Endoplasmic Reticulum Stress Induced Apoptosis and Autophagy. Phytother Res 2024; 38:6004-6022. [PMID: 39474779 DOI: 10.1002/ptr.8354] [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: 05/01/2024] [Revised: 09/09/2024] [Accepted: 09/22/2024] [Indexed: 12/13/2024]
Abstract
MHO7 (6-epi-ophiobolin G), a novel component extracted from a mangrove fungus, exhibits significant anticancer effects against breast cancer. However, the precise mechanism underlying the anticancer effects of MHO7 in prostate cancer (PCa) is yet to be fully elucidated. Therefore, this study was undertaken to assess the effect of MHO7 on PCa cells and elucidate its underlying mechanism. A series of in vitro experiments were conducted, including Cell Counting Kit-8, and plate clone formation assays, flow cytometry analysis, electron microscopy, immunofluorescence staining, western blotting, and molecular dynamics simulation. Additionally, in vivo tumor xenograft models were employed. Our findings revealed that MHO7 could induce cellular autophagy at low concentration (2 μM) and apoptosis at relatively high concentration (4 and 8 μM), leading to significant PCa cell growth inhibition. Furthermore, MHO7 triggered endoplasmic reticulum (ER) stress, which subsequently stimulated autophagy and apoptosis via IRE1α/XBP-1s signaling pathway activation. Notably, IRE1α knockdown markedly reduced MHO7-induced autophagy and apoptosis. Moreover, MHO7 targeted the IRE1α protein, thereby enhancing its stability. MHO7 also exhibited substantial anticancer activity in tumor xenograft models. Our study revealed that MHO7 holds considerable potential as an anticancer agent against PCa, attributable to its activation of ER stress-induced autophagy and apoptosis at different concentrations, facilitated by the upregulation of IRE1α expression.
Collapse
Affiliation(s)
- Mao Ding
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mu He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dan Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Shuaishuai Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chenjia Dong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Hongchao Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huajie Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Kui Hong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Hengcheng Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
33
|
Lee JH, Choi BK, Kim M, Shin HJ, Park SJ. A Lucknolide Derivative Induces Mitochondrial ROS-Mediated G2/M Arrest and Apoptotic Cell Death in B16F10 Mouse Melanoma Cells. Mar Drugs 2024; 22:533. [PMID: 39728108 DOI: 10.3390/md22120533] [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: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Melanoma is an aggressive skin cancer with a high risk of cancer-related deaths, and inducing apoptosis in melanoma cells is a promising therapeutic strategy. This study investigates the anti-tumor potential of a novel lucknolide derivative LA-UC as a therapeutic candidate for melanoma. Lucknolide A (LA), a tricyclic ketal-lactone metabolite isolated from marine-derived Streptomyces sp., was chemically modified by introducing a 10-undecenoyl group to synthesize LA-UC. LA-UC preferentially inhibited the proliferation of melanoma cells, including B16F10, while exerting minimal effects on normal melanocytes or other tumor cell types, indicating the selective action of LA-UC against melanoma cells. LA-UC decreased G2/M checkpoint proteins, including cyclin B1 and Cdc2, while activating caspase-3 and caspase-9, resulting in G2/M cell cycle arrest and inducing apoptotic cell death in B16F10 cells. The addition of a pan-caspase inhibitor confirmed the caspase-dependent mechanism of LA-UC-induced cell death. Additionally, LA-UC elevated mitochondrial ROS levels, leading to mitochondrial membrane disruption, upregulation of pro-apoptotic proteins, and DNA damage in melanoma cells. The ROS scavenger N-acetylcysteine reduced LA-UC-induced mitochondrial ROS accumulation, mitochondrial membrane disruption, DNA damage, and apoptosis. Collectively, these findings suggest that LA-UC induces G2/M cell cycle arrest and caspase-dependent apoptosis in B16F10 cells through excessive mitochondrial ROS generation, membrane impairment, and DNA damage, highlighting its potential as a promising therapeutic candidate for melanoma treatment.
Collapse
Affiliation(s)
- Jae Hyeop Lee
- BB21 Plus Program, Department of Chemistry, Pukyong National University, Busan 48513, Republic of Korea
| | - Byeoung-Kyu Choi
- Department of Bio-Convergence Engineering, Dongyang Mirae University, Seoul 08221, Republic of Korea
| | - Minsoo Kim
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Japan
| | - Hee Jae Shin
- Marine Natural Products Laboratory, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Yeongdo-gu, Busan 49111, Republic of Korea
| | - Sun Joo Park
- BB21 Plus Program, Department of Chemistry, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
34
|
Xu Y, Wang Q, Xu G, Xu Y, Mou Y. Screening, optimization, and ADMET evaluation of HCJ007 for pancreatic cancer treatment through active learning and dynamics simulation. Front Chem 2024; 12:1482758. [PMID: 39654652 PMCID: PMC11626003 DOI: 10.3389/fchem.2024.1482758] [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: 08/18/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
In this study, we leveraged a sophisticated active learning model to enhance virtual screening for SQLE inhibitors. The model's improved predictive accuracy identified compounds with significant advantages in binding affinity and thermodynamic stability. Detailed analyses, including molecular dynamics simulations and ADMET profiling, were conducted, particularly focusing on compounds CMNPD11566 and its derivative HCJ007. CMNPD11566 showed stable interactions with SQLE, while HCJ007 exhibited improved binding stability and more frequent interactions with key residues, indicating enhanced dynamic adaptability and overall binding effectiveness. ADMET data comparison highlighted HCJ007s superior profile in terms of lower toxicity and better drug-likeness. Our findings suggest HCJ007 as a promising candidate for SQLE inhibition, with significant improvements over CMNPD11566 in various pharmacokinetic and safety parameters. The study underscores the efficacy of computational models in drug discovery and the importance of comprehensive preclinical evaluations.
Collapse
Affiliation(s)
- YunYun Xu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiang Wang
- General Surgery, Tiantai People’s Hospital, Taizhou, Zhejiang, China
| | - GaoQiang Xu
- General Surgery, Tiantai People’s Hospital, Taizhou, Zhejiang, China
| | - YouJian Xu
- General Surgery, Tiantai People’s Hospital, Taizhou, Zhejiang, China
| | - YiPing Mou
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Abdelhafez OH, Elmaidomy AH, Hisham M, Glaeser SP, Kämpfer P, Wu J, Abdelmohsen UR. Hyrtios sp.-associated Cladosporium sp. UR3 as a potential source of antiproliferative metabolites. BMC Microbiol 2024; 24:445. [PMID: 39487417 PMCID: PMC11529160 DOI: 10.1186/s12866-024-03560-6] [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: 03/03/2024] [Accepted: 09/30/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Sponge-associated microorganisms are promising resources for the production of bioactive compounds with cytotoxic potential. The main goal of our study is to isolate the fungal endophytes from the Red Sea sponge Hyrtios sp. followed by investigating their cytotoxicity against number of cell lines. RESULTS The fungal strain UR3 was isolated from the Red Sea sponge using Sabouraud dextrose agar media. It was identified based on partial 18 S rRNA gene and ITS sequence analyses as Cladosporium sp. UR3. The in vitro cytotoxic potential of the ethyl acetate extract of the fungal isolate was evaluated using MTT assay against three cancer cell lines: CACO2, MCF7, and HEPG2. Metabolomics profiling of the obtained ethyl acetate extract using LC-HR-ESI-MS, along with molecular docking and pharmacological network studies for the dereplicated compounds were performed to explore its chemical profile and the possible cytotoxic mechanism of the sponge-associated fungi. CONCLUSION These results highlighted the role of sponge-associated fungi as a fruitful resource for the discovery of cytotoxic metabolites.
Collapse
Affiliation(s)
- Omnia Hesham Abdelhafez
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia City, Minia, Egypt
| | - Abeer H Elmaidomy
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Mohamed Hisham
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New-Minia, 61512, Egypt
| | - Stefanie P Glaeser
- Institute of Applied Microbiology, Justus-Liebig University Gießen, Gießen, Germany
| | - Peter Kämpfer
- Institute of Applied Microbiology, Justus-Liebig University Gießen, Gießen, Germany
| | - Jun Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, College of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia City, Minia, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt.
| |
Collapse
|
36
|
Marasinghe CK, Yoon SD, Je JY. Two peptides LLRLTDL and GYALPCDCL inhibit foam cell formation through activating PPAR-γ/LXR-α signaling pathway in oxLDL-treated RAW264.7 macrophages. Biofactors 2024; 50:1161-1175. [PMID: 38760074 DOI: 10.1002/biof.2075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 03/12/2024] [Indexed: 05/19/2024]
Abstract
Foam cell formation plays a pivotal role in atherosclerosis-associated cardiovascular diseases. Bioactive peptides generated from marine sources have been found to provide multifunctional health advantages. In the present study, we investigated the anti-atherosclerotic effects of LLRLTDL (Bu1) and GYALPCDCL (Bu2) peptides, isolated from ark shell protein hydrolysates by assessing their inhibitory effect on oxidized LDL (oxLDL)-induced foam cell formation. The two peptides showed a promising anti-atherosclerotic effect by inhibiting foam cell formation, which was evidenced by inhibiting lipid accumulation in oxLDL-treated RAW264.7 macrophages and oxLDL-treated primary human aortic smooth muscle cells (HASMC). Two peptides effectively reduced total cholesterol, free cholesterol, cholesterol ester, and triglyceride levels by upregulating cholesterol efflux and downregulating cholesterol influx. Expression of cholesterol influx-related proteins such as SR-A1 and CD36 were reduced, whereas cholesterol efflux-related proteins such as ATP-binding cassette transporter ABCA-1 and ABCG-1 were highly expressed. In addition, Bu1 and Bu2 peptides increased PPAR-γ and LXR-α expression. However, PPAR-γ siRNA transfection reversed the foam cell formation inhibitory activity of Bu1 and Bu2 peptides. Furthermore, the synergistic effect of Bu1 and Bu2 peptides on foam cell formation inhibition was observed with PPAR-γ agonist thiazolidinediones, indicating that PPAR-γ signaling pathway plays a key role in foam cell formation of macrophages. Beyond their impact on foam cell formation, Bu1 and Bu2 peptides demonstrated anti-inflammatory potential by inhibiting the generation of pro-inflammatory cytokines and nitric oxide and NF-κB nuclear activation. Taken together, these results suggest that Bu1 and Bu2 peptides may be useful for atherosclerosis and associated anti-inflammatory therapies.
Collapse
Affiliation(s)
| | - Soon-Do Yoon
- Department of Biomolecular and Chemical Engineering, Chonnam National University, Yeosu, Jeonnam, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
37
|
Liu Y, Lu T, Li R, Xu R, Baranenko D, Yang L, Xiao D. Discovery of Jaspamycin from marine-derived natural product based on MTA3 to inhibit hepatocellular carcinoma progression. Sci Rep 2024; 14:25294. [PMID: 39455636 PMCID: PMC11511890 DOI: 10.1038/s41598-024-75205-7] [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: 03/19/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Studies have underscored the pivotal role of metastasis-associated protein 3 (MTA3) as a cancer regulator, yet its potential as a drug target across cancers necessitates comprehensive evaluation. In this study, we analyzed MTA3 expression profiles to ascertain its diagnostic and prognostic value in pan-cancers, probing associations with genetic variations and immunological characteristics. Notably, liver hepatocellular carcinoma (LIHC) exhibited the most significant correlation with MTA3. By transfection of siRNA, interference of MTA3 affected HepG2 and Hepa1-6 cell viability and migration. Through drug screening and drug-likeness evaluation among marine-derived natural products, Jaspamycin was identified as a potential hepatocellular carcinoma treatment by targeting MTA3. By applying in vitro and in vivo experiment, the inhibitory effects of Jaspamycin on hepatocellular carcinoma viability, migration, and tumor progression were observed. To assess the potential of MTA3 as an anticancer drug target, MTA3 overexpression plasmid was transfected together with Jaspamycin treatment, and observed that MTA3 upregulation counteracted the inhibitory effects of Jaspamycin on hepatocarcinoma cell proliferation and migration, underscoring the efficacy of MTA3 as a drug target in hepatocellular carcinoma drug screening. This study highlights the clinical significance of MTA3 in pan-cancer, particularly in hepatocellular carcinoma. Additionally, it identifies Jaspamycin, a marine-derived compound with promising pharmacological properties, as an effective inhibitor of MTA3 activity, suggesting its potential for hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Yihan Liu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150001, China
| | - Tong Lu
- Medical Technology Department, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, China
| | - Runze Li
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Rui Xu
- Cancer Hospital, Shenzhen Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China
| | - Denis Baranenko
- School of Life Sciences, Faculty of Ecotechnologies, ITMO University, St. Petersburg, 197101, Russia
| | - Lida Yang
- Heilongjiang Nursing Collage, Harbin, Heilongjiang, 150086, China
| | - Dan Xiao
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China.
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan, 450007, China.
- School of Medicine and Health, Harbin Institute of Technology, No. 92, Xidazhi Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
38
|
Chagaleti BK, Baby K, Peña-Corona SI, Leyva-Gómez G, S M S, Naveen NR, Jose J, Aldahish AA, Sharifi-Rad J, Calina D. Anti-cancer properties of Sansalvamide A, its derivatives, and analogs: an updated review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7337-7351. [PMID: 38739152 DOI: 10.1007/s00210-024-03129-0] [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: 02/20/2024] [Accepted: 04/29/2024] [Indexed: 05/14/2024]
Abstract
As peptide-based therapies gain recognition for their potential anti-cancer activity, cyclic peptides like Sansalvamide A, a marine-derived cyclic depsipeptide, have emerged as a potential anti-cancer agent due to their potent activity against various cancer types in preclinical studies. This review offers a comprehensive overview of Sansalvamide A, including its sources, structure-activity relationship, and semi-synthetic derivatives. The review also aims to outline the mechanisms through which Sansalvamide A and its analogs exert their anti-proliferative effects and to discuss the need for enhancements in pharmacokinetic profiles for better clinical utility. An extensive literature search was conducted, focusing on studies that detailed the anti-cancer activity of Sansalvamide A, its pharmacokinetics, and mechanistic pathways. Data from both in vitro and in vivo studies were collated and analyzed. Sansalvamide A and its analogs demonstrated significant anti-cancer activity across various cancer models, mediated through Hsp 90 inhibition, Topoisomerase inhibition, and G0/G1 cell cycle arrest. However, their pharmacokinetic properties were identified as a significant limitation, requiring improvement for effective clinical translation. Despite its notable anti-cancer effects, the utility of Sansalvamide A is currently limited by its pharmacokinetic characteristics. Therefore, while Sansalvamide A exhibits promise as an anti-cancer agent, there is a compelling need for further clinical and toxicological studies and optimization of its pharmacokinetic profile to fully exploit its therapeutic potential alongside modern cancer therapies.
Collapse
Affiliation(s)
- Bharat Kumar Chagaleti
- Department of Pharmaceutical Chemistry, Akshaya Institute of Pharmacy, Tumkur, Karnataka, India
| | - Krishnaprasad Baby
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sindhoor S M
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - N Raghavendra Naveen
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B.G. Nagar, Bellur, Karnataka, India
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India.
| | - Afaf Ahmed Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 61441, Kingdom of Saudi Arabia
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| |
Collapse
|
39
|
Ameen HM, Jayadev A, Prasad G, Nair DI. Seagrass Meadows: Prospective Candidates for Bioactive Molecules. Molecules 2024; 29:4596. [PMID: 39407526 PMCID: PMC11478234 DOI: 10.3390/molecules29194596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Seagrass meadows consist of angiosperms that thrive fully submerged in marine environments and form distinct ecosystems. They provide essential support for many organisms, acting as nursery grounds for species of economic importance. Beyond their ecological roles, seagrasses and their associated microbiomes are rich sources of bioactive compounds with the potential to address numerous human healthcare challenges. Seagrasses produce bioactive molecules responding to physical, chemical, and biological environmental changes. These activities can treat microbe-borne diseases, skin diseases, diabetes, muscle pain, helminthic diseases, and wounds. Seagrasses also offer potential secondary metabolites that can be used for societal benefits. Despite numerous results on their presence and bioactive derivatives, only a few studies have explored the functional and therapeutic properties of secondary metabolites from seagrass. With the increasing spread of epidemics and pandemics worldwide, the demand for alternative drug sources and drug discovery has become an indispensable area of research. Seagrasses present a reliable natural source, making this an opportune moment for further exploration of their pharmacological activities with minimal side effects. This review provides a comprehensive overview of the biochemical, phytochemical, and biomedical applications of seagrasses globally over the last two decades, highlighting the prospective areas of future research for identifying biomedical applications.
Collapse
Affiliation(s)
- Hazeena M. Ameen
- Postgraduate Department of Environmental Sciences, All Saints’ College (Affiliated to the University of Kerala), Thiruvananthapuram 695007, India;
| | - Ayona Jayadev
- Postgraduate Department of Environmental Sciences, All Saints’ College (Affiliated to the University of Kerala), Thiruvananthapuram 695007, India;
| | - Geena Prasad
- Department of Mechanical Engineering, Amrita Vishwa Vidyapeetham, Amritapuri 641112, India
| | - Deepa Indira Nair
- Department of Engineering Technologies, Swinburne University of Technology, Melbourne 3122, Australia;
| |
Collapse
|
40
|
Chen Z, Wu J, Wang N, Li T, Wu H, Wu H, Xiang W. Isolation, Characterization, Moisturization and Anti-HepG2 Cell Activities of a Novel Polysaccharide from Cyanobacterium aponinum. Molecules 2024; 29:4556. [PMID: 39407483 PMCID: PMC11478272 DOI: 10.3390/molecules29194556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 10/20/2024] Open
Abstract
Polysaccharides from cyanobacteria are extensively reported for their complex structures, good biocompatibility, and diverse bioactivities, but only a few cyanobacterial species have been exploited for the biotechnological production of polysaccharides. According to our previous study, the newly isolated marine cyanobacterium Cyanobacterium aponinum SCSIO-45682 was a good candidate for polysaccharide production. This work provided a systematic study of the extraction optimization, isolation, structural characterization, and bioactivity evaluation of polysaccharides from C. aponinum SCSIO-45682. Results showed that the crude polysaccharide yield of C. aponinum reached 17.02% by hot water extraction. The crude polysaccharides showed a porous and fibrous structure, as well as good moisture absorption and retention capacities comparable to that of sodium alginate. A homogeneous polysaccharide (Cyanobacterium aponinum polysaccharide, CAP) was obtained after cellulose DEAE-52 column and Sephadex G-100 column purification. CAP possessed a high molecular weight of 4596.64 kDa. It was mainly composed of fucose, galactose, and galacturonic acid, with a molar ratio of 15.27:11.39:8.64. The uronic acid content and sulfate content of CAP was 12.96% and 18.06%, respectively. Furthermore, CAP showed an in vitro growth inhibition effect on human hepatocellular carcinoma (HepG2) cells. The above results indicated the potential of polysaccharides from the marine cyanobacterium C. aponinum SCSIO-45682 as a moisturizer and anticancer addictive applied in cosmetical and pharmaceutical industries.
Collapse
Affiliation(s)
- Zishuo Chen
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayi Wu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Guangzhou 511466, China
| | - Na Wang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Basic Medical Sciences, Heyang Medical School, University of South China, Hengyang 421001, China
| | - Tao Li
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
| | - Houbo Wu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
| | - Hualian Wu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
| | - Wenzhou Xiang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Institution of South China Sea Ecology and Environmental Engineering, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (Z.C.); (J.W.); (N.W.); (T.L.); (H.W.)
| |
Collapse
|
41
|
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
|
42
|
Jeevithan L, Diao X, Hu J, Elango J, Wu W, Mate Sanchez de Val JE, Rajendran S, Sundaram T, Rajamani Sekar SK. Recent advancement of novel marine fungi derived secondary metabolite fibrinolytic compound FGFC in biomedical applications: a review. Front Cell Infect Microbiol 2024; 14:1422648. [PMID: 39359937 PMCID: PMC11445226 DOI: 10.3389/fcimb.2024.1422648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
For several decades, products derived from marine natural sources (PMN) have been widely identified for several therapeutic applications due to their rich sources of bioactive sub-stances, unique chemical diversity, biocompatibility and excellent biological activity. For the past 15 years, our research team explored several PMNs, especially fungi fibrinolytic compounds (FGFCs). FGFC is an isoindolone alkaloid derived from marine fungi, also known as staplabin analogs or Stachybotrys microspora triprenyl phenol (SMTP). For instance, our previous studies explored different types of FGFCs such as FGFC 1, 2, 3 and 4 from the marine fungi Stachybotrys longispora FG216 derived metabolites. The derivatives of FGFC are potentially employed in several disease treatments, mainly for stroke, cancer, ischemia, acute kidney injury, inflammation, cerebral infarction, thrombolysis and hemorrhagic activities, etc. Due to the increasing use of FGFCs in pharmaceutical and biomedical applications, it is important to understand the fundamental signaling concept of FGFCs. Hence, for the first time, this review collectively summarizes the background, types, mode of action and biological applications of FGFCs and their current endeavors for future therapies.
Collapse
Affiliation(s)
- Lakshmi Jeevithan
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Catolica San-Antonio de Murcia, Murcia, Spain
| | - Xiaozhen Diao
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jiudong Hu
- Shanghai Sixth People’s Hospital, affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jeevithan Elango
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Catolica San-Antonio de Murcia, Murcia, Spain
- Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Wenhui Wu
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jose Eduardo Mate Sanchez de Val
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Catolica San-Antonio de Murcia, Murcia, Spain
| | | | - Thanigaivel Sundaram
- Department of Biotechnology, Faculty of Science & Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | | |
Collapse
|
43
|
Lianou AD, Lianos GD, Schizas D, Machairas N, Mitsis M, Alexiou GA. Natural Compounds and Cancer: Current Evidences. MAEDICA 2024; 19:621-628. [PMID: 39553354 PMCID: PMC11565147 DOI: 10.26574/maedica.2024.19.3.621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Natural compounds are constantly gaining ground in the treatment of various chronic diseases and in cancer research. Recent efforts have been focusing on them due to their special features consisting of low toxicity and high bioavailability. These compounds have already demonstrated important antitumor activity against several cancers in vitro through several mechanisms, including cell viability reduction, suppression of cell proliferation, cell death induction and cell cycle arrest. Herewith, we reviewed natural compounds that can be potentially used for head and neck cancer, glioblastoma and gastrointestinal cancers.
Collapse
Affiliation(s)
| | - Georgios D Lianos
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - Dimitrios Schizas
- 1nd Department of Propaedeutic Surgery, General Hospital Laiko, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Machairas
- 2nd Department of Propaedeutic Surgery, General Hospital Laiko, National and Kapodistrian University of Athens, Athens, Greece
| | - Michail Mitsis
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - George A Alexiou
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
44
|
Zare A, Ablakimova N, Kaliyev AA, Mussin NM, Tanideh N, Rahmanifar F, Tamadon A. An update for various applications of Artificial Intelligence (AI) for detection and identification of marine environmental pollutions: A bibliometric analysis and systematic review. MARINE POLLUTION BULLETIN 2024; 206:116751. [PMID: 39053264 DOI: 10.1016/j.marpolbul.2024.116751] [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: 06/10/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Marine environmental pollution is one of the growing concerns of humans all over the world. Therefore, managing these marine pollutants has been a crucial matter for scientists in recent decades. Thus, researchers have tried to implement artificial intelligence (AI) to handle marine environmental pollutants. Therefore, in this manuscript, we performed a bibliometric analysis to understand the main applications of AI for managing marine environments. Therefore, we examined both PubMed online database and Google Scholar to find any research articles that discuss the applications of AI in managing marine environmental pollution. Ultimately, we found that AI can detect, locate, and even predict aquatic contaminants like oil fingerprinting, oil spills, oil spill damage, oil slicks, forecasting marine water quality, water quality development, harmful algal blooms, benthic sediment toxicity, as well as detection of marine debris with high accuracy.
Collapse
Affiliation(s)
| | - Nurgul Ablakimova
- Department of Pharmacology, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan
| | - Asset Askerovich Kaliyev
- Department of Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan
| | - Nadiar Maratovich Mussin
- Department of Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan.
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran; Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amin Tamadon
- Department for Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| |
Collapse
|
45
|
Makhloufi H, Pinon A, Champavier Y, Saliba J, Millot M, Fruitier-Arnaudin I, Liagre B, Chemin G, Mambu L. In Vitro Antiproliferative Activity of Echinulin Derivatives from Endolichenic Fungus Aspergillus sp. against Colorectal Cancer. Molecules 2024; 29:4117. [PMID: 39274965 PMCID: PMC11397142 DOI: 10.3390/molecules29174117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/16/2024] Open
Abstract
The endolichenic fungus Aspergillus sp. was isolated from the lichen Xanthoparmelia conspersa harvested in France. Aspergillus sp. was grown on a solid culture medium to ensure the large-scale production of the fungus with a sufficient mass of secondary metabolites. The molecular network analysis of extracts and subfractions enabled the annotation of 22 molecules, guiding the purification process. The EtOAc extract displayed an antiproliferative activity of 3.2 ± 0.4 µg/mL at 48 h against human colorectal cancer cells (HT-29) and no toxicity at 30 µg/mL against human triple-negative breast cancer (TNBC) cells (MDA-MB-231) and human embryonic kidney (HEK293) non-cancerous cells. Among the five prenylated compounds isolated, of which four are echinulin derivatives, compounds 1 and 2 showed the most important activity, with IC50 values of 1.73 µM and 8.8 µM, respectively, against HT-29 cells.
Collapse
Affiliation(s)
- Hind Makhloufi
- LABCiS, UR 22722, Faculté de Pharmacie, Univ. Limoges, F-87000 Limoges, France
| | - Aline Pinon
- LABCiS, UR 22722, Faculté de Pharmacie, Univ. Limoges, F-87000 Limoges, France
| | - Yves Champavier
- Univ. Limoges, CNRS, Inserm, CHU Limoges, BISCEm, UAR 2015, US 42, F-87025 Limoges, France
| | - Jennifer Saliba
- Laboratoire LIENSs, Université de La Rochelle, UMR CNRS 7266, F-17000 La Rochelle, France
| | - Marion Millot
- LABCiS, UR 22722, Faculté de Pharmacie, Univ. Limoges, F-87000 Limoges, France
| | | | - Bertrand Liagre
- LABCiS, UR 22722, Faculté de Pharmacie, Univ. Limoges, F-87000 Limoges, France
| | - Guillaume Chemin
- LABCiS, UR 22722, Faculté de Pharmacie, Univ. Limoges, F-87000 Limoges, France
| | - Lengo Mambu
- LABCiS, UR 22722, Faculté de Pharmacie, Univ. Limoges, F-87000 Limoges, France
| |
Collapse
|
46
|
Chuang YT, Yen CY, Tang JY, Wu KC, Chang FR, Tsai YH, Chien TM, Chang HW. Marine anticancer drugs in modulating miRNAs and antioxidant signaling. Chem Biol Interact 2024; 399:111142. [PMID: 39019423 DOI: 10.1016/j.cbi.2024.111142] [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: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Several marine drugs exert anticancer effects by inducing oxidative stress, which becomes overloaded and kills cancer cells when redox homeostasis is imbalanced. The downregulation of antioxidant signaling induces oxidative stress, while its upregulation attenuates oxidative stress. Marine drugs have miRNA-modulating effects against cancer cells. However, the potential antioxidant targets of such drugs have been rarely explored. This review aims to categorize the marine-drug-modulated miRNAs that downregulate their antioxidant targets, causing oxidative stress in anticancer treatments. We also categorize the downregulation of oxidative-stress-inducing miRNAs in antioxidant protection among non-cancer cells. We summarize the putative antioxidant targets of miRNA-modulating marine drugs by introducing a bioinformatics tool (miRDB). Finally, the marine drugs affecting antioxidant targets are surveyed. In this way, the connections between marine drugs and their modulating miRNA and antioxidant targets are innovatively categorized to provide a precise network for exploring their potential anticancer functions and protective effects on non-cancer cells.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei, 11031, Taiwan; Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, 71004, Taiwan.
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung, 900392, Taiwan.
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung, 907101, Taiwan.
| | - Tsu-Ming Chien
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan; Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
47
|
Shi Y, Sun XQ, Zhang JX, Zhang RH, Hong K, Xue YX, Qiu H, Liu L. New Cytotoxic γ-Lactam Alkaloids from the Mangrove-Derived Fungus Talaromyces hainanensis sp. nov. Guided by Molecular Networking Strategy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:17431-17443. [PMID: 39021257 DOI: 10.1021/acs.jafc.4c03959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The fungus Talaromyces hainanensis, isolated from the mangrove soil, was characterized as a novel species by morphology observation and phylogenetic analyses. Four new γ-lactam alkaloids talaroilactams A-D (1-4) and two reported compounds harzianic acid (5) and isoharzianic acid (6) were identified from the fungus T. hainanensis WHUF0341, assisted by OSMAC along with molecular networking approaches. Their structures were determined through ECD calculations and spectroscopic analyses. Moreover, the biosynthetic route of 1-4 was also proposed. Compound 1 displayed potent cytotoxicity against HepG2 cell lines, with an IC50 value of 10.75 ± 1.11 μM. In addition, network pharmacology was employed to dissect the probable mechanisms contributing to the antihepatocellular carcinoma effects of compound 1, revealing that cytotoxicity was mainly associated with proteolysis, negative regulation of autophagy, inflammatory response, and the renin-angiotensin system. These results not only expanded the chemical space of natural products from the mangrove associated fungi but also afforded promising lead compounds for developing the antihepatocellular carcinoma agents.
Collapse
Affiliation(s)
- Ying Shi
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Qi Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin-Xin Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruo-Han Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kui Hong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Department of Radiation and Medical Oncology, Zhongnan Hospital, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Ya-Xin Xue
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Department of Radiation and Medical Oncology, Zhongnan Hospital, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hui Qiu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Department of Radiation and Medical Oncology, Zhongnan Hospital, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Ling Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
48
|
Wang S, Li X, Yang W, Huang R. Exploring the secrets of marine microorganisms: Unveiling secondary metabolites through metagenomics. Microb Biotechnol 2024; 17:e14533. [PMID: 39075735 PMCID: PMC11286668 DOI: 10.1111/1751-7915.14533] [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] [Received: 01/29/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024] Open
Abstract
Marine microorganisms are increasingly recognized as primary producers of marine secondary metabolites, drawing growing research interest. Many of these organisms are unculturable, posing challenges for study. Metagenomic techniques enable research on these unculturable microorganisms, identifying various biosynthetic gene clusters (BGCs) related to marine microbial secondary metabolites, thereby unveiling their secrets. This review comprehensively analyses metagenomic methods used in discovering marine microbial secondary metabolites, highlighting tools commonly employed in BGC identification, and discussing the potential and challenges in this field. It emphasizes the key role of metagenomics in unveiling secondary metabolites, particularly in marine sponges and tunicates. The review also explores current limitations in studying these metabolites through metagenomics, noting how long-read sequencing technologies and the evolution of computational biology tools offer more possibilities for BGC discovery. Furthermore, the development of synthetic biology allows experimental validation of computationally identified BGCs, showcasing the vast potential of metagenomics in mining marine microbial secondary metabolites.
Collapse
Affiliation(s)
- Shaoyu Wang
- Institute of Marine Science and TechnologyShandong UniversityQingdaoShandongChina
- Qingdao Key Laboratory of Ocean Carbon Sequestration and Negative Emission TechnologyShandong UniversityQingdaoChina
| | - Xinyan Li
- Institute of Marine Science and TechnologyShandong UniversityQingdaoShandongChina
- Qingdao Key Laboratory of Ocean Carbon Sequestration and Negative Emission TechnologyShandong UniversityQingdaoChina
| | - Weiqin Yang
- School of Computer Science and TechnologyShandong UniversityQingdaoShandongChina
| | - Ranran Huang
- Institute of Marine Science and TechnologyShandong UniversityQingdaoShandongChina
- Qingdao Key Laboratory of Ocean Carbon Sequestration and Negative Emission TechnologyShandong UniversityQingdaoChina
- Global Ocean Negative Carbon Emissions (ONCE) Program AllianceQingdaoChina
| |
Collapse
|
49
|
Visuddho V, Halim P, Helen H, Muhar AM, Iqhrammullah M, Mayulu N, Surya R, Tjandrawinata RR, Ribeiro RIMA, Tallei TE, Taslim NA, Kim B, Syahputra RA, Nurkolis F. Modulation of Apoptotic, Cell Cycle, DNA Repair, and Senescence Pathways by Marine Algae Peptides in Cancer Therapy. Mar Drugs 2024; 22:338. [PMID: 39195454 DOI: 10.3390/md22080338] [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: 05/25/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Marine algae, encompassing both macroalgae and microalgae, have emerged as a promising and prolific source of bioactive compounds with potent anticancer properties. Despite their significant therapeutic potential, the clinical application of these peptides is hindered by challenges such as poor bioavailability and susceptibility to enzymatic degradation. To overcome these limitations, innovative delivery systems, particularly nanocarriers, have been explored. Nanocarriers, including liposomes, nanoparticles, and micelles, have demonstrated remarkable efficacy in enhancing the stability, solubility, and bioavailability of marine algal peptides, ensuring controlled release and prolonged therapeutic effects. Marine algal peptides encapsulated in nanocarriers significantly enhance bioavailability, ensuring more efficient absorption and utilization in the body. Preclinical studies have shown promising results, indicating that nanocarrier-based delivery systems can significantly improve the pharmacokinetic profiles and therapeutic outcomes of marine algal peptides. This review delves into the diverse anticancer mechanisms of marine algal peptides, which include inducing apoptosis, disrupting cell cycle progression, and inhibiting angiogenesis. Further research focused on optimizing nanocarrier formulations, conducting comprehensive clinical trials, and continued exploration of marine algal peptides holds great promise for developing innovative, effective, and sustainable cancer therapies.
Collapse
Affiliation(s)
- Visuddho Visuddho
- Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Muhammad Iqhrammullah
- Postgraduate Program of Public Health, Universitas Muhammadiyah Aceh, Banda Aceh 23123, Indonesia
| | - Nelly Mayulu
- Department of Nutrition, Faculty of Health Science, Muhammadiyah Manado University, Manado 95249, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta 11480, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Department of Biotechnology, Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta 12930, Indonesia
| | | | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia
| |
Collapse
|
50
|
Nova P, Cunha SA, Costa-Pinto AR, Gomes AM. Chemical and Antioxidant Properties of Solvent and Enzyme-Assisted Extracts of Fucus vesiculosus and Porphyra dioica. Mar Drugs 2024; 22:319. [PMID: 39057428 PMCID: PMC11278442 DOI: 10.3390/md22070319] [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: 06/14/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Extraction strategies impact the efficiency and nature of extracted compounds. This work assessed the chemical composition and antioxidant capacity of ethanolic, hydroethanolic, and aqueous versus enzyme-assisted extracts (isolated or with the sequential use of alcalase®, cellulase®, and viscozyme®) of the macroalgae Fucus vesiculosus (brown, Phaeophyceae) and Porphyra dioica (red, Rhodophyta. For both macroalgae, enzyme-assisted extraction (EAE) was the most efficient process compared to solvent-assisted extraction (SAE), independent of solvent. Fucus vesiculosus extraction yields were higher for EAE than for SAE (27.4% to 32.2% and 8.2% to 30.0%, respectively). Total phenolics content (TPC) was at least 10-fold higher in EAE extracts (229.2 to 311.3 GAE/gextract) than in SAE (4.34 to 19.6 GAE/gextract) counterparts and correlated well with antioxidant capacity (ABTS and ORAC methods), with EAE achieving values up to 8- and 2.6-fold higher than those achieved by SAE, respectively. Porphyra dioica followed F. vesiculosus's trend for extraction yields (37.5% to 51.6% for EAE and 5.7% to 35.1% for SAE), TPC, although of a lower magnitude, (0.77 to 8.95 GAE/gextract for SE and 9.37 to 14.73 GAE/gextract for EAE), and antioxidant capacity. Aqueous extracts registered the highest DPPH values for both macroalgae, with 2.3 µmol TE/gextract and 13.3 µmol TE/gextract for F. vesiculosus and P. dioica, respectively. EAE was a more efficient process in the extraction of soluble protein and reducing sugars in comparison to SAE. Furthermore, an improved effect of enzyme-assisted combinations was observed for almost all analyzed parameters. This study shows the promising application of enzyme-assisted extraction for the extraction of valuable compounds from F. vesiculosus and P.dioica, making them excellent functional ingredients for a wide range of health and food industrial applications.
Collapse
Affiliation(s)
- Paulo Nova
- CBQF—Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (P.N.); (S.A.C.); (A.M.G.)
- i3S—Instituto de Investigacão e Inovacão em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Sara A. Cunha
- CBQF—Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (P.N.); (S.A.C.); (A.M.G.)
| | - Ana R. Costa-Pinto
- i3S—Instituto de Investigacão e Inovacão em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Instituto de Patologia Molecular e Imunologia da Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Maria Gomes
- CBQF—Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (P.N.); (S.A.C.); (A.M.G.)
| |
Collapse
|