1
|
Noshin S, Bairagi RD, Airin S, Debnath D, Rahaman MS, Acharzo AK, Aktar MN, Bourhia M, Salamatullah AM, Islam MA. Synergistic Bioactivity of Aegiceras corniculatum (L.) Blanco and Its Endophytic Fungus Aspergillus: Antioxidant, Antimicrobial, and Cytotoxic Effects. Cell Biochem Biophys 2025; 83:1197-1206. [PMID: 39586960 DOI: 10.1007/s12013-024-01553-w] [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] [Accepted: 09/17/2024] [Indexed: 11/27/2024]
Abstract
The mangrove fungi provide a vast and unexplored source of diverse and unique chemicals and biological properties. The plant Aegiceras corniculatum (L.) Blanco and its endophytic fungus aspergillus species were collected from different sites of the Baleswar river region in Sundarban. Hence, we compared the antioxidant properties of the associated fungus ACSF-1 and the methanolic bark extract of Aegiceras corniculatum (MBAC) by measuring the total phenolic content (TPC), total flavonoid content (TFC), and DPPH free radical assay. Subsequently, antimicrobial activity was measured using the disc diffusion method, and cytotoxic activity was measured using the brine shrimp lethality bioassay. The results showed that MBAC has even more DPPH scavenging activity (IC50 = 44.036 μg/mL), TPC (310.275 mg GAE/g), and TFC (66.275 mg QE/g) in comparison with DPPH scavenging activity (IC50 = 92.542 μg/mL), TPC (234.832 mg GAE/g), and TFC (134.887 mg QE/g) in ACSF-1. The median lethal concentration value (LC50) of MBAC and ACSF-1 was found to be 43.93 μg/mL and 336.84 μg/mL, respectively. Moreover, MBAC showed a dose-dependent antimicrobial response to Escherichia coli and Staphylococcus aureus, whereas ACSF-1 was found to have activity against Bacillus subtilis and S. aureus. These results emphasize the unique pharmacological characteristics of both the plant and fungus, indicating their potential usefulness in various therapeutic fields.
Collapse
Affiliation(s)
- Sharika Noshin
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Rahul Dev Bairagi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Sadia Airin
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Dipa Debnath
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Md Sohanur Rahaman
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Amit Kumar Acharzo
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Most Nazmin Aktar
- School of Natural Sciences, Macquarie University Sydney, Sydney, NSW, 2109, Australia
- Computational Biology Research laboratory, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Mohammed Bourhia
- Department of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, Ibn Zohr University, Laayoune, Morocco
| | - Ahmad Mohammad Salamatullah
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Md Amirul Islam
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh.
- Department of Pharmacy, East West University, Dhaka, Bangladesh.
| |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 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
|
3
|
Wu Y, Xu ZZ, Kong C, Zhang SS, Lin XL, Zhang S, Liu LY, Sun F, Lin HW, Wang SP. Discovery of cycloheptapeptides phakefusins A-E from the marine sponge Phakellia fusca based on molecular networking. PHYTOCHEMISTRY 2025; 229:114248. [PMID: 39197714 DOI: 10.1016/j.phytochem.2024.114248] [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: 05/13/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024]
Abstract
Guided by a probe-based molecular networking strategy, five undescribed cycloheptapeptides, phakefusins A-E (1-5), were isolated from the marine sponge Phakellia fusca. Compounds 1 and 2 contain the nonproteinogenic amino acid residues of dioxindolyalanine (Dioia) and β-3-oxindolylalanine (Oia), respectively. Compound 3 possesses a unique methionine sulfoxide, whereas compound 5 includes a glutamic acid ethyl ester unit. Their structures were elucidated through NMR spectroscopy, HR-MS/MS analysis, and the advanced Marfey's method. By synthesizing the (S, S/R)-Oia standard through tryptophan oxidation, we determined the configuration of this amino acid in compound 2 using the advanced Marfey's method. These cycloheptapeptides were evaluated for their antitumor, antibacterial, and antioxidant activities. Compound 1 showed moderate cytotoxicity against MCF-7 and PC9 cells, with IC50 values of 6.8 and 9.6 μM, respectively, while compounds 2-5 demonstrated potential antioxidant effects by upregulating HO-1, NQO1, and SOD2 levels, as well as inducing Nrf2 activation.
Collapse
Affiliation(s)
- Ying Wu
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhao-Ze Xu
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Can Kong
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shuai-Shuai Zhang
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xin-Li Lin
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Si Zhang
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Li-Yun Liu
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fan Sun
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hou-Wen Lin
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Shu-Ping Wang
- Marine Drug Integrated Innovation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zhao X, Xi X, Zhang M, Lv M, Zhang X, Lu Y, Wang L, Chen Y. Structure-Activity Relationship Study of Majusculamide D: Overcoming Metabolic Instability and Severe Toxicity with a Fluoro Analogue. Mar Drugs 2024; 22:537. [PMID: 39728112 DOI: 10.3390/md22120537] [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: 10/15/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024] Open
Abstract
Majusculamide D, isolated from the marine cyanobacterium Moorea producens, is an anticancer lipopentapeptide consisting of fatty acid, tripeptide, and pyrrolyl proline moieties. In this work, by utilizing a convergent synthetic approach, late-stage modification, and bioisostere strategy, 26 majusculamide D analogues were synthesized, and two (1i and 1j) demonstrated IC50 values < 1 nM against PANC-1 cancer cells. The results summarized a preliminary structure-activity relationship mainly at the C23, C4, C34, and C10 sites. A series of in vitro assays, including wound healing, transwell, clone formation, EdU, and western blot, confirmed that majusculamide D inhibited the migration, invasion, and proliferation of pancreatic cancer cells. The optimized fluorinated analogue 1n demonstrated a notable enhancement in stability during the mouse plasma assay (>50% left after 24 h), exhibited tumor-suppressive effects (51.5% at a dosage of 5 mg/kg), and successfully mitigated the severe toxicity (no mouse dead) observed in the group treated with majusculamide D (3 mice dead) in a xenografted mouse model.
Collapse
Affiliation(s)
- Xiuhe Zhao
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiaonan Xi
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Mingxiao Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Mengxue Lv
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiang Zhang
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yaxin Lu
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Liang Wang
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yue Chen
- College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
7
|
Li Y, Xu C, Weng W, Goel A. Combined treatment with Aronia berry extract and oligomeric proanthocyanidins exhibit a synergistic anticancer efficacy through LMNB1-AKT signaling pathways in colorectal cancer. Mol Carcinog 2024; 63:2145-2157. [PMID: 39282961 DOI: 10.1002/mc.23800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 10/04/2024]
Abstract
Colorectal cancer (CRC) is one of the most prevalent and highly recurrent malignancies worldwide and currently ranks as the second leading cause of cancer-related deaths. The high degree of morbidity and mortality associated with CRC is primarily attributed to the limited effectiveness of current therapeutic approaches and the emergence of chemoresistance to standard treatment modalities. Recent research indicates that several natural products, including Aronia berry extracts (ABE) and oligomeric proanthocyanidins (OPCs), might offer a safe, cost-effective, and multitargeted adjunctive role to cancer treatment. Herein, we hypothesized a combined treatment with ABE and OPCs could synergistically modulate multiple oncogenic pathways in CRC, thereby enhancing their anticancer activity. We initially conducted a series of in vitro experiments to assess the synergistic anticancer effects of ABE and OPCs on CRC cell lines. We demonstrate that these two compounds exhibited a superior synergistic anticancer potential versus individual treatments in enhancing the ability to inhibit cell viability, suppress colony formation, and induce apoptosis (p < 0.05). Consistent with our in vitro findings, we validated this combinatorial anticancer effect in tumor-derived 3D organoids (PDOs; p < 0.01). Using genome-wide transcriptomic profiling, we identified that a specific gene, LMNB1, associated with the cell apoptosis pathway, was found to play a crucial role in exhibiting anticancer effects with these two products. Furthermore, the combined treatment of ABE and OPCs significantly impacted the expression of key proteins involved in apoptosis, including suppressed expression levels of LMNB1 in CRC cell lines (p < 0.05), which resulted in inhibiting downstream AKT phosphorylation. In conclusion, our study provides novel evidence of the synergistic anticancer effects of ABE and OPCs in CRC cells, partially mediated through the regulation of apoptosis and the oncogene LMNB1 within the AKT signaling pathway. These findings have the potential to better appreciate the anticancer potential of natural products in CRC and help improve treatment outcomes in this malignancy.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wenhao Weng
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| |
Collapse
|
8
|
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
|
9
|
Zhou D, Chen D, Wu J, Feng T, Liu P, Xu J. Dicerandrol C Suppresses Proliferation and Induces Apoptosis of HepG2 and Hela Cancer Cells by Inhibiting Wnt/β-Catenin Signaling Pathway. Mar Drugs 2024; 22:278. [PMID: 38921589 PMCID: PMC11204528 DOI: 10.3390/md22060278] [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: 05/24/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Overwhelming evidence points to an aberrant Wnt/β-catenin signaling as a critical factor in hepatocellular carcinoma (HCC) and cervical cancer (CC) pathogenesis. Dicerandrol C (DD-9), a dimeric tetrahydroxanthenone isolated from the endophytic fungus Phomopsis asparagi DHS-48 obtained from mangrove plant Rhizophora mangle via chemical epigenetic manipulation of the culture, has demonstrated effective anti-tumor properties, with an obscure action mechanism. The objective of the current study was to explore the efficacy of DD-9 on HepG2 and HeLa cancer cells and its functional mechanism amid the Wnt/β catenin signaling cascade. Isolation of DD-9 was carried out using various column chromatographic methods, and its structure was elucidated with 1D NMR. The cytotoxicity of DD-9 on HepG2 and HeLa cells was observed with respect to the proliferation, clonality, migration, invasion, apoptosis, cell cycle, and Wnt/β-catenin signaling cascade. We found that DD-9 treatment significantly reduced tumor cell proliferation in dose- and time-dependent manners in HepG2 and HeLa cells. The subsequent experiments in vitro implied that DD-63 could significantly suppress the tumor clonality, metastases, and induced apoptosis, and that it arrested the cell cycle at the G0/G1 phase of HepG2 and HeLa cells. Dual luciferase assay, Western blot, and immunofluorescence assay showed that DD-9 could dose-dependently attenuate the Wnt/β-catenin signaling by inhibiting β-catenin transcriptional activity and abrogating β-catenin translocated to the nucleus; down-regulating the transcription level of β-catenin-stimulated Wnt target gene and the expression of related proteins including p-GSK3-β, β-catenin, LEF1, Axin1, c-Myc, and CyclinD1; and up-regulating GSK3-β expression, which indicates that DD-9 stabilized the β-catenin degradation complex, thereby inducing β-catenin degradation and inactivation of the Wnt/β-catenin pathway. The possible interaction between DD-9 and β-catenin and GSK3-β protein was further confirmed by molecular docking studies. Collectively, DD-9 may suppress proliferation and induce apoptosis of liver and cervical cancer cells, possibly at least in part via GSK3-β-mediated crosstalk with the Wnt/β-catenin signaling axis, providing insights into the mechanism for the potency of DD-9 on hepatocellular and cervical cancer.
Collapse
Affiliation(s)
- Dongdong Zhou
- Collaborative Innovation Center of Ecological Civilization, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China; (D.Z.); (D.C.); (J.W.); (T.F.); (P.L.)
| | - Dandan Chen
- Collaborative Innovation Center of Ecological Civilization, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China; (D.Z.); (D.C.); (J.W.); (T.F.); (P.L.)
| | - Jingwan Wu
- Collaborative Innovation Center of Ecological Civilization, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China; (D.Z.); (D.C.); (J.W.); (T.F.); (P.L.)
| | - Ting Feng
- Collaborative Innovation Center of Ecological Civilization, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China; (D.Z.); (D.C.); (J.W.); (T.F.); (P.L.)
| | - Pinghuai Liu
- Collaborative Innovation Center of Ecological Civilization, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China; (D.Z.); (D.C.); (J.W.); (T.F.); (P.L.)
- Research and Utilization on Seaweed Biological Resources Key Laboratory of Haikou, Haikou 570228, China
| | - Jing Xu
- Collaborative Innovation Center of Ecological Civilization, School of Chemistry and Chemical Engineering, Hainan University, Haikou 570228, China; (D.Z.); (D.C.); (J.W.); (T.F.); (P.L.)
| |
Collapse
|
10
|
Yang QB, Liang LF. Spongia Sponges: Unabated Sources of Novel Secondary Metabolites. Mar Drugs 2024; 22:213. [PMID: 38786604 PMCID: PMC11123444 DOI: 10.3390/md22050213] [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: 03/31/2024] [Revised: 04/27/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
Marine sponges of the genus Spongia have proven to be unabated sources of novel secondary metabolites with remarkable scaffold diversities and significant bioactivities. The discovery of chemical substances from Spongia sponges has continued to increase over the last few years. The current work provides an up-to-date literature survey and comprehensive insight into the reported metabolites from the members of the genus Spongia, as well as their structural features, biological activities, and structure-activity relationships when available. In this review, 222 metabolites are discussed based on published data from the period from mid-2015 to the beginning of 2024. The compounds are categorized into sesquiterpenes, diterpenes, sesterterpenes, meroterpenes, linear furanoterpenes, steroids, alkaloids, and other miscellaneous substances. The biological effects of these chemical compositions on a vast array of pharmacological assays including cytotoxic, anti-inflammatory, antibacterial, neuroprotective, protein tyrosine phosphatase 1B (PTP1B)-inhibitory, and phytoregulating activities are also presented.
Collapse
Affiliation(s)
| | - Lin-Fu Liang
- College of Chemistry and Chemical Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| |
Collapse
|
11
|
Chen Q, Xu N, Zhao C, He Y, Kam SHT, Wu X, Huang P, Yang M, Wong CTT, Radis-Baptista G, Tang B, Fan G, Gong G, Lee SMY. A new invertebrate NPY-like polypeptide, ZoaNPY, from the Zoanthus sociatus, as a novel ligand of human NPY Y2 receptor rescues vascular insufficiency via PLC/PKC and Src- FAK-dependent signaling pathways. Pharmacol Res 2024; 203:107173. [PMID: 38580186 DOI: 10.1016/j.phrs.2024.107173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Our recent multi-omics studies have revealed rich sources of novel bioactive proteins and polypeptides from marine organisms including cnidarians. In the present study, we initially conducted a transcriptomic analysis to review the composition profile of polypeptides from Zoanthus sociatus. Then, a newly discovered NPY-like polypeptide-ZoaNPY was selected for further in silico structural, binding and virtually pharmacological studies. To evaluate the pro-angiogenic effects of ZoaNPY, we employed an in vitro HUVECs model and an in vivo zebrafish model. Our results indicate that ZoaNPY, at 1-100 pmol, enhances cell survival, migration and tube formation in the endothelial cells. Besides, treatment with ZoaNPY could restore a chemically-induced vascular insufficiency in zebrafish embryos. Western blot results demonstrated the application of ZoaNPY could increase the phosphorylation of proteins related to angiogenesis signaling including PKC, PLC, FAK, Src, Akt, mTOR, MEK, and ERK1/2. Furthermore, through molecular docking and surface plasmon resonance (SPR) verification, ZoaNPY was shown to directly and physically interact with NPY Y2 receptor. In view of this, all evidence showed that the pro-angiogenic effects of ZoaNPY involve the activation of NPY Y2 receptor, thereby activating the Akt/mTOR, PLC/PKC, ERK/MEK and Src- FAK-dependent signaling pathways. Furthermore, in an excision wound model, the treatment with ZoaNPY was shown to accelerate the wound healing process in mice. Our findings provide new insights into the discovery and development of novel pro-angiogenic drugs derived from NPY-like polypeptides in the future.
Collapse
Affiliation(s)
- Qian Chen
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao
| | - Nan Xu
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao
| | - Chen Zhao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao
| | - Yulin He
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China
| | - Sandy Hio Tong Kam
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao
| | - Xue Wu
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao
| | - Pan Huang
- Kunming Institute of Zoology, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Min Yang
- Kunming Institute of Zoology, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Clarence Tsun Ting Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 999077, Hong Kong, SAR China
| | | | - Benqin Tang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China
| | - Guangyi Fan
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
| | - Guiyi Gong
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China.
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, Macao; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, SAR China.
| |
Collapse
|
12
|
Wang Y, Guo X, Huang C, Shi C, Xiang X. Biomedical potency and mechanisms of marine polysaccharides and oligosaccharides: A review. Int J Biol Macromol 2024; 265:131007. [PMID: 38508566 DOI: 10.1016/j.ijbiomac.2024.131007] [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: 12/12/2023] [Revised: 03/11/2024] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
Derived from bountiful marine organisms (predominantly algae, fauna, and microorganisms), marine polysaccharides and marine oligosaccharides are intricate macromolecules that play a significant role in the growth and development of marine life. Recently, considerable attention has been paid to marine polysaccharides and marine oligosaccharides as auspicious natural products due to their promising biological attributes. Herein, we provide an overview of recent advances in the miscellaneous biological activities of marine polysaccharides and marine oligosaccharides that encompasses their anti-cancer, anti-inflammatory, antibacterial, antiviral, antioxidant, anti-diabetes mellitus, and anticoagulant properties. Furthermore, we furnish a concise summary of the underlying mechanisms governing the behavior of these biological macromolecules. We hope that this review inspires research on marine polysaccharides and marine oligosaccharides in medicinal applications while offering fresh perspectives on their broader facets.
Collapse
Affiliation(s)
- Yi Wang
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Xueying Guo
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Chunxiao Huang
- School of Clinical Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Chuanqin Shi
- Center of Translational Medicine, Zibo Central Hospital, Zibo 255020, China.
| | - Xinxin Xiang
- Center of Translational Medicine, Zibo Central Hospital, Zibo 255020, China.
| |
Collapse
|
13
|
Wu T, Shi Y, Yang T, Zhao P, Yang Z, Yang B. Polymer-DNA assembled nanoflower for targeted delivery of dolastatin-derived microtubule inhibitors. RSC Adv 2024; 14:9602-9608. [PMID: 38516154 PMCID: PMC10956646 DOI: 10.1039/d3ra08146j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
Dolastatin derivatives possess excellent anticancer activity and have been translated into clinical trials for cancer therapy. Drug delivery systems enable dolastatin derivatives to break the limitation of instability during blood circulation and ineffective cell internalization in the application. Nevertheless, their potential has not been thoroughly established because of the limited loading efficacy and complicated chemical modification. Herein, we rationally propose a rolling circle amplification-based polymer-DNA assembled nanoflower for targeted and efficient delivery of dolastatin-derived drugs to achieve efficient anticancer therapy. The polymer-DNA assembled nanoflower with targeted aptamer conjugate is widely applicable for loading dolastatin-derived drugs with high encapsulation efficiency. The developed monomethyl auristatin E (MMAE) loaded PN@M exhibited increased cellular uptake and enhanced inhibitory effect, especially in multidrug-resistant tumor cells. The results of in vivo anticancer effects indicate that nanoflower as a dolastatin derivatives delivery system holds considerable potential for the treatment of malignant cancer.
Collapse
Affiliation(s)
- Tiantian Wu
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University Guangzhou 510091 China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University Haikou 571199 China
| | - Yanqiang Shi
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University Guangzhou 510091 China
| | - Tao Yang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University Haikou 571199 China
| | - Pengxuan Zhao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University Haikou 571199 China
| | - Zhu Yang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University Fuzhou 350005 China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University Fuzhou 350212 China
| | - Bin Yang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University Guangzhou 510091 China
| |
Collapse
|
14
|
Zhang C, Liu F, Zhang Y, Song C. Macrocycles and macrocyclization in anticancer drug discovery: Important pieces of the puzzle. Eur J Med Chem 2024; 268:116234. [PMID: 38401189 DOI: 10.1016/j.ejmech.2024.116234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/10/2024] [Accepted: 02/11/2024] [Indexed: 02/26/2024]
Abstract
Increasing disease-related proteins have been identified as novel therapeutic targets. Macrocycles are emerging as potential solutions, bridging the gap between conventional small molecules and biomacromolecules in drug discovery. Inspired by successful macrocyclic drugs of natural origins, macrocycles are attracting more attention for enhanced binding affinity and target selectivity. Due to the conformation constraint and structure preorganization, macrocycles can reach bioactive conformations more easily than parent acyclic compounds. Also, rational macrocyclization combined with sequent structural modification will help improve oral bioavailability and combat drug resistance. This review introduces various strategies to enhance membrane permeability in macrocyclization and subsequent modification, such as N-methylation, intramolecular hydrogen bonding modulation, isomerization, and reversible bicyclization. Several case studies highlight macrocyclic inhibitors targeting kinases, HDAC, and protein-protein interactions. Finally, some macrocyclic agents targeting tumor microenvironments are illustrated.
Collapse
Affiliation(s)
- Chao Zhang
- Laboratory for Food and Medicine Homologous Natural Resources Development and Utilization, Belgorod College of Food Sciences, Dezhou University, Dezhou, 253023, China
| | - Fenfen Liu
- Laboratory for Food and Medicine Homologous Natural Resources Development and Utilization, Belgorod College of Food Sciences, Dezhou University, Dezhou, 253023, China
| | - Youming Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
| | - Chun Song
- Laboratory for Food and Medicine Homologous Natural Resources Development and Utilization, Belgorod College of Food Sciences, Dezhou University, Dezhou, 253023, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
| |
Collapse
|
15
|
Angulo-Elizari E, Henriquez-Figuereo A, Morán-Serradilla C, Plano D, Sanmartín C. Unlocking the potential of 1,4-naphthoquinones: A comprehensive review of their anticancer properties. Eur J Med Chem 2024; 268:116249. [PMID: 38458106 DOI: 10.1016/j.ejmech.2024.116249] [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: 01/15/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Cancer encompasses a group of pathologies with common characteristics, high incidence, and prevalence in all countries. Although there are treatments available for this disease, they are not always effective or safe, often failing to achieve the desired results. This is why it is necessary to continue the search for new therapies. One of the strategies for obtaining new antitumor drugs is the use of 1,4-naphthoquinone as a scaffold in synthetic or natural products with antitumor activity. This review focuses on compiling studies related to the antitumor activity of 1,4-naphthoquinone and its natural and synthetic derivatives over the last 10 years. The work describes the main natural naphthoquinones with antitumor activity and classifies the synthetic naphthoquinones based on the structural modifications made to the scaffold. Additionally, the formation of metal complexes using naphthoquinones as a ligand is considered. After a thorough review, 197 synthetic compounds with potent biological activity against cancer have been classified according to their chemical structures and their mechanisms of action have been described.
Collapse
Affiliation(s)
- Eduardo Angulo-Elizari
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Andreina Henriquez-Figuereo
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Cristina Morán-Serradilla
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Daniel Plano
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| | - Carmen Sanmartín
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| |
Collapse
|
16
|
Yi Y, Zhou B, Man T, Xu Z, Tang H, Li J, Sun Z. Resveratrol Inhibits Nasopharyngeal Carcinoma (NPC) by Targeting the MAPK Signaling Pathway. Anticancer Agents Med Chem 2024; 24:1207-1219. [PMID: 38988166 DOI: 10.2174/0118715206319761240705115109] [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: 04/19/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND With conventional cancer treatments facing limitations, interest in plant-derived natural products as potential alternatives is increasing. Although resveratrol has demonstrated antitumor effects in various cancers, its impact and mechanism on nasopharyngeal carcinoma remain unclear. OBJECTIVE This study aimed to systematically investigate the anti-cancer effects of resveratrol on nasopharyngeal carcinoma using a combination of experimental pharmacology, network pharmacology, and molecular docking approaches. METHODS CCK-8, scratch wound, and transwell assays were employed to confirm the inhibitory effect of resveratrol on the proliferation, migration, and invasion of nasopharyngeal carcinoma cells. H&E and TUNEL stainings were used to observe the morphological changes and apoptosis status of resveratrol-treated cells. The underlying mechanisms were elucidated using a network pharmacology approach. Immunohistochemistry and Western blotting were utilized to validate key signaling pathways. RESULTS Resveratrol inhibited the proliferation, invasion, and migration of nasopharyngeal carcinoma cells, ultimately inducing apoptosis in a time- and dose-dependent manner. Network pharmacology analysis revealed that resveratrol may exert its anti-nasopharyngeal carcinoma effect mainly through the MAPK pathway. Immunohistochemistry results from clinical cases showed MAPK signaling activation in nasopharyngeal carcinoma tissues compared to adjacent tissues. Western blotting validated the targeting effect of resveratrol, demonstrating significant inhibition of the MAPK signaling pathway. Furthermore, molecular docking supported its multi-target role with MAPK, TP53, PIK3CA, SRC, etc. Conclusion: Resveratrol has shown promising potential in inhibiting human nasopharyngeal carcinoma cells by primarily targeting the MAPK pathway. These findings position resveratrol as a potential therapeutic agent for nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yujuan Yi
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Bo Zhou
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Tengjun Man
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Zihan Xu
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Hong Tang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jia Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zheng Sun
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| |
Collapse
|
17
|
Wang Z, Xu Z, Yang X, Li M, Yip RCS, Li Y, Chen H. Current application and modification strategy of marine polysaccharides in tissue regeneration: A review. BIOMATERIALS ADVANCES 2023; 154:213580. [PMID: 37634336 DOI: 10.1016/j.bioadv.2023.213580] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023]
Abstract
Marine polysaccharides (MPs) are exceptional bioactive materials that possess unique biochemical mechanisms and pharmacological stability, making them ideal for various tissue engineering applications. Certain MPs, including agarose, alginate, carrageenan, chitosan, and glucan have been successfully employed as biological scaffolds in animal studies. As carriers of signaling molecules, scaffolds can enhance the adhesion, growth, and differentiation of somatic cells, thereby significantly improving the tissue regeneration process. However, the biological benefits of pure MPs composite scaffold are limited. Therefore, physical, chemical, enzyme modification and other methods are employed to expand its efficacy. Chemically, the structural properties of MPs scaffolds can be altered through modifications to functional groups or molecular weight reduction, thereby enhancing their biological activities. Physically, MPs hydrogels and sponges emulate the natural extracellular matrix, creating a more conducive environment for tissue repair. The porosity and high permeability of MPs membranes and nanomaterials expedite wound healing. This review explores the distinctive properties and applications of select MPs in tissue regeneration, highlighting their structural versatility and biological applicability. Additionally, we provide a brief overview of common modification strategies employed for MP scaffolds. In conclusion, MPs have significant potential and are expected to be a novel regenerative material for tissue engineering.
Collapse
Affiliation(s)
- Zhaokun Wang
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Zhiwen Xu
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Xuan Yang
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Man Li
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China.
| | - Ryan Chak Sang Yip
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Yuanyuan Li
- Department of Food Science, Cornell University, Stocking Hall, Ithaca, NY 14853, USA.
| | - Hao Chen
- Marine College, Shandong University, NO. 180 Wenhua West Road, Gao Strict, Weihai 264209, China; The Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, NO. 1800 Lihu Road, Wuxi 214122, China.
| |
Collapse
|
18
|
Fan R, Wang S, Wu Y, Feng Y, Gao M, Cao Y, Ma X, Xie S, Wang C, Gao L, Wang Y, Dai F. Activation of endoplasmic reticulum stress by harmine suppresses the growth of esophageal squamous cell carcinoma. Phytother Res 2023; 37:4655-4673. [PMID: 37525965 DOI: 10.1002/ptr.7933] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
The worldwide overall 5-year survival rate of esophageal squamous cell carcinoma (ESCC) patients is less than 20%, and novel therapeutic strategies for these patients are urgently needed. Harmine is a natural β-carboline alkaloid, which received great interest in cancer research because of its biological and anti-tumor activities. The aim of this study is to examine the effects of harmine on ESCC and its mechanism. We investigated the effects of harmine on proliferation, cell cycle, apoptosis, and tumor growth in vivo. RNA sequencing (RNA-seq), real-time PCR, and western blotting were used to detect the mechanism. Harmine inhibited ESCC cell growth in vitro and tumor growth in vivo. Differentially expressed genes in harmine-treated ESCC cells were mainly involved in protein processing in the endoplasmic reticulum (ER). Real-time PCR and western blotting confirmed harmine-induced cellular ER stress. CRISPR-Cas9 knockout of C/EBP homologous protein (CHOP) abolished harmine-induced expression of death receptor 5 and apoptosis. Harmine also induced the expression of CHOP-mediated sestrin-2, which in turn contributes to autophagosome formation via suppressing the AMP-activated protein kinase-protein kinase B-mammalian target of rapamycin signaling pathway. In conclusion, our results demonstrate that harmine inhibits the growth of ESCC through its regulation of ER stress, suggesting that it is a promising candidate for ESCC treatment.
Collapse
Affiliation(s)
- Ronghui Fan
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Senzhen Wang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yalan Wu
- School of Biomedical Sciences, Heart and Vascular Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yongli Feng
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Mengke Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Yue Cao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Xiaoxuan Ma
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Songqiang Xie
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Chaojie Wang
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| | - Lei Gao
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan University, Kaifeng, Henan, China
| | - Yanming Wang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Fujun Dai
- Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, Henan, China
| |
Collapse
|
19
|
Yan XY, Zhang L, Yang QB, Ge ZY, Liang LF, Guo YW. Genus Litophyton: A Hidden Treasure Trove of Structurally Unique and Diversely Bioactive Secondary Metabolites. Mar Drugs 2023; 21:523. [PMID: 37888458 PMCID: PMC10608288 DOI: 10.3390/md21100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Marine soft corals are prolific sources of various natural products that have served as a wealthy reservoir of diverse chemical scaffolds with potential as new drug leads. The genus Litophyton contains almost 100 species but only a small proportion of them has been chemically investigated, which calls for more attentions from global researchers. In the current work, 175 secondary metabolites have been discussed, drawing from published data spanning almost five decades, up to July 2023. The studied species of the genus Litophyton resided in various tropical and temperate regions and encompassed a broad range of biologically active natural products including terpenes, steroids, nitrogen-containing metabolites, lipids, and other metabolites. A wide spectrum of pharmacological effects of these compounds had been evaluated, such as cytotoxic, antiviral, antibacterial, antifungal, anti-malarial, antifeedant, anti-inflammatory, molluscicidal, PTP1B inhibitory, insect growth inhibitory, and neuroprotective activities. This review aims to offer an up-to-date survey of the literature and provide a comprehensive understanding of the chemical structures, taxonomical distributions, and biological activities of the reported metabolites from the title genus whenever available.
Collapse
Affiliation(s)
- Xian-Yun Yan
- College of Materials Science and Engineering, Central South University of Forestry and Technology, 498 South Shaoshan Road, Changsha 410004, China; (X.-Y.Y.); (L.Z.); (Q.-B.Y.); (Z.-Y.G.)
| | - Ling Zhang
- College of Materials Science and Engineering, Central South University of Forestry and Technology, 498 South Shaoshan Road, Changsha 410004, China; (X.-Y.Y.); (L.Z.); (Q.-B.Y.); (Z.-Y.G.)
| | - Qi-Bin Yang
- College of Materials Science and Engineering, Central South University of Forestry and Technology, 498 South Shaoshan Road, Changsha 410004, China; (X.-Y.Y.); (L.Z.); (Q.-B.Y.); (Z.-Y.G.)
| | - Zeng-Yue Ge
- College of Materials Science and Engineering, Central South University of Forestry and Technology, 498 South Shaoshan Road, Changsha 410004, China; (X.-Y.Y.); (L.Z.); (Q.-B.Y.); (Z.-Y.G.)
| | - Lin-Fu Liang
- College of Materials Science and Engineering, Central South University of Forestry and Technology, 498 South Shaoshan Road, Changsha 410004, China; (X.-Y.Y.); (L.Z.); (Q.-B.Y.); (Z.-Y.G.)
| | - Yue-Wei Guo
- School of Medicine, Shanghai University, 99 Shangda Road, Bao Shan District, Shanghai 200444, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, 198 Binhai East Road, High-tech Zone, Yantai 264117, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
20
|
Cao XZ, Zhang BQ, Wang CF, Yin JN, Haider W, Said G, Wei MY, Lu L. A Terphenyllin Derivative CHNQD-00824 from the Marine Compound Library Induced DNA Damage as a Potential Anticancer Agent. Mar Drugs 2023; 21:512. [PMID: 37888447 PMCID: PMC10608154 DOI: 10.3390/md21100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
With the emergence of drug resistance and the consequential high morbidity and mortality rates, there is an urgent need to screen and identify new agents for the effective treatment of cancer. Terphenyls-a group of aromatic hydrocarbons consisting of a linear 1,4-diaryl-substituted benzene core-has exhibited a wide range of biological activities. In this study, we discovered a terphenyllin derivative-CHNQD-00824-derived from the marine compound library as a potential anticancer agent. The cytotoxic activities of the CHNQD-00824 compound were evaluated against 13 different cell lines with IC50 values from 0.16 to 7.64 μM. Further study showed that CHNQD-00824 inhibited the proliferation and migration of cancer cells, possibly by inducing DNA damage. Acridine orange staining demonstrated that CHNQD-00824 promoted apoptosis in zebrafish embryos. Notably, the anti-cancer effectiveness was verified in a doxycin hydrochloride (DOX)-induced liver-specific enlargement model in zebrafish. With Solafinib as a positive control, CHNQD-00824 markedly suppressed tumor growth at concentrations of 2.5 and 5 μM, further highlighting its potential as an effective anticancer agent.
Collapse
Affiliation(s)
- Xi-Zhen Cao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Bo-Qi Zhang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Cui-Fang Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Jun-Na Yin
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Waqas Haider
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Gulab Said
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Department of Chemistry, Women University Swabi, Swabi 23430, Pakistan
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Ling Lu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| |
Collapse
|
21
|
Wang H, Liu Y, Cui J, Tong M, Guan W, Cao Z, Gao X, Han X, Xian X, Li J, Zhao L. Effects of Scutellaria strigillosa Hemsl. extract on HepG2 cell proliferation and apoptosis through binding to aspartate β-hydroxylase. Biochem Biophys Res Commun 2023; 668:62-69. [PMID: 37244036 DOI: 10.1016/j.bbrc.2023.05.077] [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: 04/20/2023] [Revised: 05/10/2023] [Accepted: 05/20/2023] [Indexed: 05/29/2023]
Abstract
This study aims to examine the impacts of Scutellaria strigillosa Hemsl. (SSH) on the proliferation, apoptosis of human hepatoma cell HepG2 and screen the bioactive components. We found that SSH extract inhibited HepG2 proliferation, arrested cell division prior to S phase. Additionally, SSH extract exposure induced apoptosis, and increased the proportions of late apoptotic cells. Specifically, we focus on the inhibitory effect of SSH extract on aspartate β-hydroxylase, a key therapeutic target of hepatocellular carcinoma closely related with the proliferation and apoptosis of HepG2. We found SSH extract with notable inhibitory activity against aspartate β-hydroxylase, elucidated the main bioactive constituents by HPLC-Q-TOF/MS and Molecular docking analysis. In conclusion, these results provided the antiproliferative and proapoptotic effects of SSH on HepG2 cell, elucidated the main bioactive constituents based on aspartate β-hydroxylase inhibition. These data revealed the potential value of SSH and its bioactive components for the prevention and treatment of liver cancer for the first time.
Collapse
Affiliation(s)
- Hairong Wang
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Yuan Liu
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Jiawen Cui
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Miaomiao Tong
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Wenlong Guan
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Zhi Cao
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Xiaoli Gao
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Xiaopeng Han
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Xiaomeng Xian
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China
| | - Jiankun Li
- The Forth Affiliated Hospital of Hebei Medical University, Health Road No. 12, Shijiazhuang, 050011, China.
| | - Lili Zhao
- Hebei Medical University, Zhongshan Road No. 361, Shijiazhuang, 050017, China.
| |
Collapse
|
22
|
Lee TY, Yang W, Cha DS, Han YT. Synthesis of a natural quinoline alkaloid isolated from the deep-sea-derived fungus and its potential as a therapeutic for Parkinson's disease. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2023; 25:446-455. [PMID: 35980025 DOI: 10.1080/10286020.2022.2104259] [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: 05/30/2022] [Revised: 07/17/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
2-(Quinoline-8-carboxamido)benzoic acid (2-QBA; 1) is a natural quinoline alkaloid isolated from the deep-sea-derived fungus Aspergillus sp. SCSIO06786. Alkaloid 1 was synthesized by an amidation reaction of 8-quinolinecaroxylic acid with methyl anthranilate, followed by hydrolysis. The neuroprotective properties of 1 were evaluated using a Caenorhabditis elegans Parkinson's disease model, which revealed that 1 significantly ameliorated 1-methyl-4-phenylpyridinium (MPP+)-induced dopaminergic neurodegeneration in a dose-dependent manner. MPP+-induced behavioral defects in worms, including impaired locomotion and basal slowing ability, were restored by treatment with 1. We further demonstrated that treatment with 1 modulates the formation of neurotoxic α-synuclein oligomers by suppressing α-synuclein expressions and enhancing proteasome activity. These results suggest that 1 is a promising therapeutic candidate for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Tae Yeon Lee
- College of Pharmacy, Dankook University, Cheonan 31116, South Korea
| | - Wooin Yang
- College of Pharmacy, Woosuk University, Wanju-gun 55338, South Korea
| | - Dong Seok Cha
- College of Pharmacy, Woosuk University, Wanju-gun 55338, South Korea
| | - Young Taek Han
- College of Pharmacy, Dankook University, Cheonan 31116, South Korea
| |
Collapse
|
23
|
Lin K, Huang L, Zhang Y, Chen M, Li Z, Yung KKL, Lv S, Pan Q, Zhang W, Fu J, Li W, Deng Q. The Antiangiogenic and Antitumor Effects of Scoparasin B in Non-Small-Cell Lung Cancer. JOURNAL OF NATURAL PRODUCTS 2023; 86:368-379. [PMID: 36692021 DOI: 10.1021/acs.jnatprod.2c00979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Angiogenesis and vasculogenic mimicry (VM) are crucial for the growth and metastasis of non-small-cell lung cancer (NSCLC). Most tumor angiogenesis inhibitors mainly target endothelial cell-mediated angiogenesis, ignoring tumor-cell-mediated VM and frequently leading to tumor recurrence and metastasis. Thus, development of bioactive molecules interfering with both tumor angiogenesis and VM is necessary. Identifying novel angiogenesis inhibitors from natural products is a promising strategy. Scoparasin B, a pimarane diterpene extracted from a marine-derived fungus, Eutypella sp. F0219, has an antibacterial effect. However, its effect on angiogenesis and VM remains unexplored. In this study, we first certified that scoparasin B showed a strong inhibition effect on angiogenesis and the VM process in vitro and ex vivo. Moreover, scoparasin B prominently impeded tumor growth, angiogenesis, and VM in an NCI-H1299 xenograft model. Further study revealed that scoparasin B restrained tumor angiogenesis and VM by reducing the VEGF-A level and suppressing the VEGF-A/VEGFR2 signaling pathway. This study first demonstrated scoparasin B inhibited tumor angiogenesis, VM, and tumor growth of NSCLC and revealed its underlying mechanism. These new findings further support the potential of scoparasin B as a novel angiogenesis inhibitor and give a hint for further exploring potential angiogenesis inhibitors from natural products.
Collapse
Affiliation(s)
- Kaili Lin
- School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China
- Golden Meditech Center for Neuro Regeneration Sciences, HKBU, Kowloon Tong, Hong Kong 999077, China
| | - Lijuan Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Minshan Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ken Kin Lam Yung
- Department of Biology & Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong, China
| | - Sha Lv
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qianrong Pan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Weisong Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jijun Fu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wanshan Li
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Qiudi Deng
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
24
|
Chen Q, Ruan D, Shi J, Du D, Bian C. The multifaceted roles of natural products in mitochondrial dysfunction. Front Pharmacol 2023; 14:1093038. [PMID: 36860298 PMCID: PMC9968749 DOI: 10.3389/fphar.2023.1093038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
Mitochondria are the primary source of energy production in cells, supporting the metabolic demand of tissue. The dysfunctional mitochondria are implicated in various diseases ranging from neurodegeneration to cancer. Therefore, regulating dysfunctional mitochondria offers a new therapeutic opportunity for diseases with mitochondrial dysfunction. Natural products are pleiotropic and readily obtainable sources of therapeutic agents, which have broad prospects in new drug discovery. Recently, many mitochondria-targeting natural products have been extensively studied and have shown promising pharmacological activity in regulating mitochondrial dysfunction. Hence, we summarize recent advances in natural products in targeting mitochondria and regulating mitochondrial dysfunction in this review. We discuss natural products in terms of their mechanisms on mitochondrial dysfunction, including modulating mitochondrial quality control system and regulating mitochondrial functions. In addition, we describe the future perspective and challenges in the development of mitochondria-targeting natural products, emphasizing the potential value of natural products in mitochondrial dysfunction.
Collapse
Affiliation(s)
| | | | - Jiayan Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Gynecology and Obstetrics, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Dongru Du
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Gynecology and Obstetrics, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
25
|
Fisher JF, Mobashery S. β-Lactams from the Ocean. Mar Drugs 2023; 21:86. [PMID: 36827127 PMCID: PMC9963991 DOI: 10.3390/md21020086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
The title of this essay is as much a question as it is a statement. The discovery of the β-lactam antibiotics-including penicillins, cephalosporins, and carbapenems-as largely (if not exclusively) secondary metabolites of terrestrial fungi and bacteria, transformed modern medicine. The antibiotic β-lactams inactivate essential enzymes of bacterial cell-wall biosynthesis. Moreover, the ability of the β-lactams to function as enzyme inhibitors is of such great medical value, that inhibitors of the enzymes which degrade hydrolytically the β-lactams, the β-lactamases, have equal value. Given this privileged status for the β-lactam ring, it is therefore a disappointment that the exemplification of this ring in marine secondary metabolites is sparse. It may be that biologically active marine β-lactams are there, and simply have yet to be encountered. In this report, we posit a second explanation: that the value of the β-lactam to secure an ecological advantage in the marine environment might be compromised by its close structural similarity to the β-lactones of quorum sensing. The steric and reactivity similarities between the β-lactams and the β-lactones represent an outside-of-the-box opportunity for correlating new structures and new enzyme targets for the discovery of compelling biological activities.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry & Biochemistry, 354 McCourtney Hall, University of Note Dame, Notre Dame, IN 46656-5670, USA
| | - Shahriar Mobashery
- Department of Chemistry & Biochemistry, 354 McCourtney Hall, University of Note Dame, Notre Dame, IN 46656-5670, USA
| |
Collapse
|
26
|
Xing H, Zhu X, Liao J, Kong Y, Lu Y, Zhao D, Li N, Chen X, Qin Z. Pharmacokinetic study of Strongylocentrotus nudus egg polysaccharide in rats and beagles using a 3H-labeling method. Front Pharmacol 2023; 14:1109084. [PMID: 36937847 PMCID: PMC10017969 DOI: 10.3389/fphar.2023.1109084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Strongylocentrotus nudus egg polysaccharide (SEP) extracted from sea urchins has potential anticancer activity. However, little is known about its pharmacokinetic properties. To investigate the pharmacokinetics of SEP, it was radiolabeled with tritium. Furthermore, a sensitive, selective, and rapid liquid scintillation counter (LSC) method for quantifying 3H-SEP in biological matrix was validated. The lower quantification limit of the method was 4 Bq. The relative standard deviations (RSDs) of the intra- and inter-day precision were <3.0% and <3.9%, respectively. 3H-SEP was successfully applied to investigate the pharmacokinetics of SEP after intravenous administration of 20, 40, and 80 mg/kg (40 μCi/kg) in rats and 5, 10, and 20 mg/kg (6 μCi/kg) in beagles. The AUC(0-t) of SEP at three different doses was 487.81 ± 39.99 mg/L*h, 1,003.10 ± 95.94 mg/L*h, and 2,188.84 ± 137.73 mg/L*h in rats and 144.12 ± 3.78 mg/L*h, 322.62 ± 28.03 mg/L*h, and 754.17 ± 37.79 mg/L*h in beagles. The terminal elimination half-life (t1/2) of SEP was longer in beagles (204.29 ± 139.34 h) than in rats (35.48 ± 6.04 h). The concentration of SEP in plasma declined rapidly in both rats and beagles. All the study results provide detailed pharmacokinetic profiles of SEP in two kinds of animals, which will be helpful for further development.
Collapse
Affiliation(s)
- Han Xing
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Application and Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Xiaojie Zhu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Jianmin Liao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Ying Kong
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yayuan Lu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Ning Li
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- *Correspondence: Zhiying Qin, ; Xijing Chen,
| | - Zhiying Qin
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Application and Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhiying Qin, ; Xijing Chen,
| |
Collapse
|
27
|
Two New Alkaloids and a New Butenolide Derivative from the Beibu Gulf Sponge-Derived Fungus Penicillium sp. SCSIO 41413. Mar Drugs 2022; 21:md21010027. [PMID: 36662200 PMCID: PMC9864330 DOI: 10.3390/md21010027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022] Open
Abstract
Marine sponge-derived fungi have been proven to be a prolific source of bioactive natural products. Two new alkaloids, polonimides E (1) and D (2), and a new butenolide derivative, eutypoid F (11), were isolated from the Beibu Gulf sponge-derived fungus, Penicillium sp. SCSIO 41413, together with thirteen known compounds (3-10, 12-16). Their structures were determined by detailed NMR, MS spectroscopic analyses, and electronic circular dichroism (ECD) analyses. Butenolide derivatives 11 and 12 exhibited inhibitory effect against the enzyme PI3K with IC50 values of 1.7 μM and 9.8 μM, respectively. The molecular docking was also performed to understand the inhibitory activity, while 11 and 12 showed obvious protein/ligand-binding effects to the PI3K protein. Moreover, 4 and 15 displayed obvious inhibitory activity against LPS-induced NF-κB activation in RAW264.7 cells at 10 µM.
Collapse
|
28
|
Potential of Compounds Originating from the Nature to Act in Hepatocellular Carcinoma Therapy by Targeting the Tumor Immunosuppressive Microenvironment: A Review. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010195. [PMID: 36615387 PMCID: PMC9822070 DOI: 10.3390/molecules28010195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022]
Abstract
Hepatocellular carcinoma (HCC), the most prevalent subtype of liver cancer, is the second main reason for cancer-related deaths worldwide. In recent decades, sufficient evidence supported that immunotherapy was a safe and effective treatment option for HCC. However, tolerance and frequent recurrence and metastasis occurred in patients after immunotherapy due to the complicated crosstalk in the tumor immunosuppressive microenvironment (TIME) in HCC. Therefore, elucidating the TIME in HCC and finding novel modulators to target TIME for attenuating immune suppression is critical to optimize immunotherapy. Recently, studies have shown the potentially immunoregulatory activities of natural compounds, characterized by multiple targets and pathways and low toxicity. In this review, we concluded the unique role of TIME in HCC. Moreover, we summarized evidence that supports the hypothesis of natural compounds to target TIME to improve immunotherapy. Furthermore, we discussed the comprehensive mechanisms of these natural compounds in the immunotherapy of HCC. Accordingly, we present a well-grounded review of the naturally occurring compounds in cancer immunotherapy, expecting to shed new light on discovering novel anti-HCC immunomodulatory drugs from natural sources.
Collapse
|
29
|
Liu L, Qian X, Yang T, Fang D, Qin Z, Ren B, Li G. Cyclopiazonic Acid and Okaramine Analogues, Including Chlorinated Compounds, from Chrysosporium undulatum YT-1. JOURNAL OF NATURAL PRODUCTS 2022; 85:2547-2556. [PMID: 36268672 DOI: 10.1021/acs.jnatprod.2c00445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Eight new cyclopiazonic acid (1-8) and five new okaramine (9-13) alkaloids together with 13 known compounds were isolated from the fungus Chrysosporium undulatum YT-1. Compounds 2, 4, 5, 7, 10, 11, and 13 were chlorinated indole alkaloids. The structures of compounds 1-13 were elucidated by HRESIMS and NMR spectroscopic data. Their relative and absolute configurations were established by J-based configuration analysis, NOESY, NOEDIFF experiments, ECD spectroscopic data, and biogenetic considerations. Compound 4 inhibited the growth of Bacillus subtilis with an MIC value of 6.3 μg/mL. Compounds 9-11 exhibited strong insecticidal capacity against the third instar larvae of silkworm and cotton bollworm (LD50: ≤7.56 μg/g). At 40 μM, compound 1 showed obvious neuroprotection to the PC12 cells with 6-OHDA treatment.
Collapse
Affiliation(s)
- Lingyan Liu
- Key Laboratory of Environmental and Applied Microbiology, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xueqing Qian
- Key Laboratory of Environmental and Applied Microbiology, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Tao Yang
- Key Laboratory of Environmental and Applied Microbiology, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Dongmei Fang
- Key Laboratory of Environmental and Applied Microbiology, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zhangyi Qin
- Key Laboratory of Environmental and Applied Microbiology, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Bo Ren
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China
| | - Guoyou Li
- Key Laboratory of Environmental and Applied Microbiology, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan 610041, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
30
|
Gao S, Jiang X, Wang L, Jiang S, Luo H, Chen Y, Peng C. The pathogenesis of liver cancer and the therapeutic potential of bioactive substances. Front Pharmacol 2022; 13:1029601. [PMID: 36278230 PMCID: PMC9581229 DOI: 10.3389/fphar.2022.1029601] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Liver cancer is the third most common cause of cancer-related deaths in the world and has become an urgent problem for global public health. Bioactive substances are widely used for the treatment of liver cancer due to their widespread availability and reduced side effects. This review summarizes the main pathogenic factors involved in the development of liver cancer, including metabolic fatty liver disease, viral infection, and alcoholic cirrhosis, and focuses on the mechanism of action of bioactive components such as polysaccharides, alkaloids, phenols, peptides, and active bacteria/fungi. In addition, we also summarize transformation methods, combined therapy and modification of bioactive substances to improve the treatment efficiency against liver cancer, highlighting new ideas in this field.
Collapse
Affiliation(s)
- Song Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xingyue Jiang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liang Wang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shanshan Jiang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyuan Luo
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Yan Chen, ; Cheng Peng,
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Yan Chen, ; Cheng Peng,
| |
Collapse
|
31
|
Li W, Zhang C, Zhang HE, Dong R, Liu JY, Wang CM, Wang M, Wang YW, Wang C, Zhang Y, Shi L, Xu Y, Sun LP. Design, synthesis, and anticancer evaluation of ammosamide B with pyrroloquinoline derivatives as novel BRD4 inhibitors. Bioorg Chem 2022; 127:105917. [PMID: 35738217 DOI: 10.1016/j.bioorg.2022.105917] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/21/2022] [Accepted: 05/26/2022] [Indexed: 11/02/2022]
Abstract
Bromodomain-containing protein 4 (BRD4), which is a member of the bromodomain and extra-terminal domain (BET) family, plays an important role in the regulation of gene expression as the "reader" of epigenetic regulation. BRD4 has become a promising target to treat cancer, because the up-regulation of BRD4 expression is closely associated with the occurrence and development of various cancers. At present, several BRD4 inhibitors are in clinical trials for cancer therapy, but no BRD4 inhibitors are on the market. Here, we designed and synthesized a series of compounds bearing pyrrolo[4,3,2-de]quinolin-2(1H)-one scaffold through structural modification of natural products ammosamide B, which is a natural pyrroloquinoline derivative reported for its potential antitumor activity. All target compounds were evaluated for their BRD4 BD1 inhibition activities via the protein thermal shift assays or AlphaSceen assay. The representative compound 49 showed potent activity (IC50 = 120 nM). The co-crystal of compound 49 with BRD4 BD1 was solved to study the structure activity relationship, which showed that 49 could combine with the acetyl lysine binding site and formed a hydrogen bond with the conserved residue Asn140. The results demonstrate that compound 49 is worthy of further investigation as a promising BRD4 inhibitor.
Collapse
Affiliation(s)
- Wen Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Cheng Zhang
- Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Hong-En Zhang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ru Dong
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jing-Ying Liu
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Chun-Meng Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Min Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yu-Wei Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Chao Wang
- Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Yan Zhang
- Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Lei Shi
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yong Xu
- Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou 510530, China.
| | - Li-Ping Sun
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
32
|
Püsküllüoğlu M, Michalak I. An ocean of possibilities: a review of marine organisms as sources of nanoparticles for cancer care. Nanomedicine (Lond) 2022; 17:1695-1719. [PMID: 36562416 DOI: 10.2217/nnm-2022-0206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Seas and oceans have been explored for the last 70 years in search of new compounds that can support the battle against cancer. Marine polysaccharides can act as nanomaterials for medical applications and marine-derived bioactive compounds can be applied for the biosynthesis of metallic and nonmetallic nanoparticles. Nanooncology can be used in numerous fields including diagnostics, serving as drug carriers or acting as drugs. This review focuses on marine-derived nanoparticles with potential oncological applications. It classifies organisms used for nanoparticle production, explains the production process, presents different types of nanoparticles with prospective applications in oncology, describes the molecular pathways responsible for numerous nanomedicine applications, tags areas of nanoparticle implementation in oncology and speculates about future directions.
Collapse
Affiliation(s)
- Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, Kraków, 31-115, Poland
| | - Izabela Michalak
- Wrocław University of Science & Technology, Department of Advanced Material Technologies, Smoluchowskiego 25, Wrocław, 50-370, Poland
| |
Collapse
|
33
|
Investigating the structure-activity relationship of marine polycyclic batzelladine alkaloids as promising inhibitors for SARS-CoV-2 main protease (Mpro). Comput Biol Med 2022; 147:105738. [PMID: 35777088 PMCID: PMC9212445 DOI: 10.1016/j.compbiomed.2022.105738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/04/2022] [Accepted: 06/11/2022] [Indexed: 11/24/2022]
Abstract
Over a span of two years ago, since the emergence of the first case of the novel coronavirus (SARS-CoV-2) in China, the pandemic has crossed borders causing serious health emergencies, immense economic crisis and impacting the daily life worldwide. Despite the discovery of numerous forms of precautionary vaccines along with other recently approved orally available drugs, yet effective antiviral therapeutics are necessarily needed to hunt this virus and its variants. Historically, naturally occurring chemicals have always been considered the primary source of beneficial medications. Considering the SARS-CoV-2 main protease (Mpro) as the duplicate key element of the viral cycle and its main target, in this paper, an extensive virtual screening for a focused chemical library of 15 batzelladine marine alkaloids, was virtually examined against SARS-CoV-2 main protease (Mpro) using an integrated set of modern computational tools including molecular docking (MDock), molecule dynamic (MD) simulations and structure-activity relationships (SARs) as well. The molecular docking predictions had disclosed four promising compounds including batzelladines H–I (8–9) and batzelladines F-G (6–7), respectively according to their prominent ligand-protein energy scores and relevant binding affinities with the (Mpro) pocket residues. The best two chemical hits, batzelladines H–I (8–9) were further investigated thermodynamically though studying their MD simulations at 100 ns, where they showed excellent stability within the accommodated (Mpro) pocket. Moreover, SARs studies imply the crucial roles of the fused tricyclic guanidinic moieties, its degree of unsaturation, position of the N–OH functionality and the length of the side chain as a spacer linking between two active sites, which disclosed fundamental structural and pharmacophoric features for efficient protein-ligand interaction. Such interesting findings are greatly highlighting further in vitro/vivo examinations regarding those marine natural products (MNPs) and their synthetic equivalents as promising antivirals.
Collapse
|
34
|
Two New Phenylhydrazone Derivatives from the Pearl River Estuary Sediment-Derived Streptomyces sp. SCSIO 40020. Mar Drugs 2022; 20:md20070449. [PMID: 35877742 PMCID: PMC9323291 DOI: 10.3390/md20070449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Two new phenylhydrazone derivatives and one new alkaloid, penzonemycins A–B (1–2) and demethylmycemycin A (3), together with three known compounds including an alkaloid (4) and two sesquiterpenoids (5–6), were isolated from the Streptomyces sp. SCSIO 40020 obtained from the Pearl River Estuary sediment. Their structures and absolute configurations were assigned by 1D/2D NMR, mass spectroscopy and X-ray crystallography. Compound 1 was evaluated in four human cancer cell lines by the SRB method and displayed weak cytotoxicity in three cancer cell lines, with IC50 values that ranged from 30.44 to 61.92 µM, which were comparable to those of the positive control cisplatin. Bioinformatic analysis of the putative biosynthetic gene cluster indicated a Japp–Klingemann coupling reaction involved in the hydrazone formation of 1 and 2.
Collapse
|
35
|
Ahn MH, Shin JA, Yang SO, Choi WS, Jang S, Kang SC, Cho SD. Metabolite profiling of a Sargassum micracanthum methanol extract with in vitro efficacy against human head and neck squamous cell carcinoma aggressiveness. Arch Oral Biol 2022; 137:105386. [PMID: 35272061 DOI: 10.1016/j.archoralbio.2022.105386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Extracts from the brown algae Sargassum micracanthum have documented anti-viral, anti-oxidant, and anti-inflammatory activities as well as potential anti-tumor efficacy against several cancer types. Here, we evaluated the inhibitory effect and molecular mechanisms of methanol extract of S. micracanthum (MESM) on the aggressiveness of human head and neck squamous cell carcinoma (HNSCC) using in vitro cell culture-based models. DESIGN To test the potential efficacy of MESM on the migratory and invasive properties of HNSCC cells, we used wound healing, transwell cell migration and invasion assays. Proteome profiling and functional in silico analysis were applied to investigate the possible modes of action by MESM. We also examined the metabolite profiling of MESM using gas chromatography/mass spectrometry. RESULTS MESM inhibited the motility of human HNSCC cell lines as well as invasiveness without influencing cell survival. Proteome profiling identified 19 oncogenic proteins significantly downregulated by MESM treatment. Protein-protein interaction network and gene ontology analyses revealed that Tie2 and associated angiogenic signaling pathway components were significantly enriched among these downregulated oncogenic proteins, which was confirmed by validating the reduced Tie2 expression in MESM treatment groups. Metabolite profiling of MESM identified six-carbon sugar alcohols such as D-sorbitol and/or D-mannitol as the main bioactive compounds. D-sorbitol and D-mannitol effectively reduced Tie2 expression and the aggressiveness of human HNSCC cell lines. CONCLUSIONS These findings suggest that six-carbon sugar alcohols in MESM have promising anti-cancer efficacy for the treatment of human HNSCC and further identify Tie2 signaling components as potential treatment targets.
Collapse
Affiliation(s)
- Min-Hye Ahn
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Ok Yang
- National Instrumentation Center for Environmental Management, Seoul National University, Seoul 08826, Republic of Korea
| | - Won-Sil Choi
- National Instrumentation Center for Environmental Management, Seoul National University, Seoul 08826, Republic of Korea
| | - Soojin Jang
- Department of Oral Pathology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Se-Chan Kang
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea.
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
36
|
Carbone D, Vestuto V, Ferraro MR, Ciaglia T, Pecoraro C, Sommella E, Cascioferro S, Salviati E, Novi S, Tecce MF, Amodio G, Iraci N, Cirrincione G, Campiglia P, Diana P, Bertamino A, Parrino B, Ostacolo C. Metabolomics-assisted discovery of a new anticancer GLS-1 inhibitor chemotype from a nortopsentin-inspired library: From phenotype screening to target identification. Eur J Med Chem 2022; 234:114233. [DOI: 10.1016/j.ejmech.2022.114233] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/21/2022]
|
37
|
Huang W, Shi L, Liu M, Zhang Z, Liu F, Long T, Wen S, Huang D, Wang K, Zhou R, Fang W, Hu H, Ke S. Design, Synthesis, and Cytotoxic Activity of Novel Natural Arylsulfonamide-Inspired Molecules. Molecules 2022; 27:1479. [PMID: 35268580 PMCID: PMC8911723 DOI: 10.3390/molecules27051479] [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: 01/14/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Primary arylsulfonamide functional groups feature prominently in diverse pharmaceuticals. However, natural arylsulfonamides are relatively infrequent. In this work, two novel arylsulfonamide natural products were first synthesized, and then a series of novel molecules derived from natural arylsulfonamides were designed and synthesized, and their in vitro cytotoxic activities against A875, HepG2, and MARC145 cell lines were systematically evaluated. The results indicate that some of these arylsulfonamide derivatives exhibit significantly good cytotoxic activity against the tested cell lines compared with the control 5-fluorouracil (5-FU), such as compounds 10l, 10p, 10q, and 10r. In particular, the potential molecule 10q, containing a carbazole moiety, exhibited the highest inhibitory activity against all tested cell lines, with IC50 values of 4.19 ± 0.78, 3.55 ± 0.63, and 2.95 ± 0.78 μg/mL, respectively. This will offer the potential to discover novel drug-like compounds from the sparsely populated area of natural products that can lead to effective anticancer agents.
Collapse
Affiliation(s)
- Wenbo Huang
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Liqiao Shi
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Manli Liu
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zhigang Zhang
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Fang Liu
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Tong Long
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Shaohua Wen
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Daye Huang
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Kaimei Wang
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Ronghua Zhou
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Wei Fang
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Hongtao Hu
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Shaoyong Ke
- Key Laboratory of Microbial Pesticides, Ministry of Agriculture and Rural Affairs, Wuhan 430064, China; (W.H.); (L.S.); (M.L.); (Z.Z.); (F.L.); (T.L.); (S.W.); (D.H.); (K.W.); (R.Z.)
- National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| |
Collapse
|
38
|
Chen DL, Chen MY, Hou Y, Wang CH, Sun ZC, Yang Y, Liang HQ, Ma GX, Xu XD, Wei JH. Cadinane-Type Sesquiterpenoids with Cytotoxic Activity from the Infected Stems of the Semi-mangrove Hibiscus tiliaceus. JOURNAL OF NATURAL PRODUCTS 2022; 85:127-135. [PMID: 35040320 DOI: 10.1021/acs.jnatprod.1c00849] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Eight new cadinane sesquiterpenoids (1-8), along with two known compounds (9 and 10), were isolated from infected stems of the semi-mangrove plant, Hibiscus tiliaceus. The structures of compounds 1-8 were elucidated through the analysis of their 1D and 2D NMR and MS data, and their absolute configurations were determined by comparing their experimental and calculated ECD spectra and by single-crystal X-ray diffraction. The two confused known compounds (9 and 10) were resolved using single-crystal X-ray crystallography. Compounds 1-3 have novel norsesquiterpene carbon skeletons arising from a ring contraction rearrangement. All obtained isolates were evaluated against the HepG2 and Huh7 cell lines, and compounds 1b, 2b, 4, 6, and 8 showed cytotoxic activity toward both cell lines, with IC50 values ranging from 3.5 to 6.8 μM.
Collapse
Affiliation(s)
- De-Li Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
- Hainan Branch Institute of Medicinal Plant Development (Hainan Provincial Key Laboratory of Resources Conservation and Development of Southern Medicine), Chinese Academy of Medical Sciences & Peking Union Medical College, Haikou 570311, People's Republic of China
| | - Mei-Ying Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
| | - Yong Hou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
| | - Can-Hong Wang
- Hainan Branch Institute of Medicinal Plant Development (Hainan Provincial Key Laboratory of Resources Conservation and Development of Southern Medicine), Chinese Academy of Medical Sciences & Peking Union Medical College, Haikou 570311, People's Republic of China
| | - Zhao-Cui Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
| | - Yun Yang
- Hainan Branch Institute of Medicinal Plant Development (Hainan Provincial Key Laboratory of Resources Conservation and Development of Southern Medicine), Chinese Academy of Medical Sciences & Peking Union Medical College, Haikou 570311, People's Republic of China
| | - Han-Qiao Liang
- Department of Biomedicine, Beijing City University, Beijing 100083, People's Republic of China
| | - Guo-Xu Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
- Hainan Branch Institute of Medicinal Plant Development (Hainan Provincial Key Laboratory of Resources Conservation and Development of Southern Medicine), Chinese Academy of Medical Sciences & Peking Union Medical College, Haikou 570311, People's Republic of China
| | - Xu-Dong Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
| | - Jian-He Wei
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing 100193, People's Republic of China
- Hainan Branch Institute of Medicinal Plant Development (Hainan Provincial Key Laboratory of Resources Conservation and Development of Southern Medicine), Chinese Academy of Medical Sciences & Peking Union Medical College, Haikou 570311, People's Republic of China
| |
Collapse
|
39
|
Panggabean JA, Adiguna SP, Murniasih T, Rahmawati SI, Bayu A, Putra MY. Structure-Activity Relationship of Cytotoxic Natural Products from Indonesian Marine Sponges. REVISTA BRASILEIRA DE FARMACOGNOSIA : ORGAO OFICIAL DA SOCIEDADE BRASILEIRA DE FARMACOGNOSIA 2022; 32:12-38. [PMID: 35034994 PMCID: PMC8740879 DOI: 10.1007/s43450-021-00195-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/15/2021] [Indexed: 11/29/2022]
Abstract
Indonesian marine natural products have been one of the most promising sources in the race to obtain potential drugs for cancer treatment. One of the primary producers of cytotoxic compounds is sponges. However, there are still limited sources of comprehensive reviews related to the relationship between the structure of isolated compounds and their cytotoxic activity. This review remarks the attempt to provide a preliminary guidance from the perspective of structure-activity relationship and its participation on marine natural products research. This guidance is segregated by the compound's classes and their cytotoxic targets to obtain and organized a reliable summary of inter-study of the isolated compounds and their cytotoxicity. Structure-activity relationship is well-known for its ability to tune the bioactivity of a specific compound, especially on synthetic organic chemistry and in silico study but rarely used on natural product chemistry. The present review is intended to narrow down the endless possibilities of cytotoxicity by giving a predictable structure-activity relationship for active compounds. In addition, bioactive framework leads were selected by uncovering a noticeable structure-activity relationship with the intervention of cytotoxic agents from natural sources, especially Indonesian marine sponge. Graphical abstract
Collapse
Affiliation(s)
- Jonathan A. Panggabean
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Bulaksumur, Yogyakarta, 55281 Indonesia
- Research Center for Biotechnology, Research Organization for Life Science, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor KM. 46, Cibinong, Jawa Barat 16911 Bogor, Indonesia
| | - Sya’ban P. Adiguna
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Bulaksumur, Yogyakarta, 55281 Indonesia
- Research Center for Biotechnology, Research Organization for Life Science, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor KM. 46, Cibinong, Jawa Barat 16911 Bogor, Indonesia
| | - Tutik Murniasih
- Research Center for Biotechnology, Research Organization for Life Science, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor KM. 46, Cibinong, Jawa Barat 16911 Bogor, Indonesia
| | - Siti I. Rahmawati
- Research Center for Biotechnology, Research Organization for Life Science, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor KM. 46, Cibinong, Jawa Barat 16911 Bogor, Indonesia
| | - Asep Bayu
- Research Center for Biotechnology, Research Organization for Life Science, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor KM. 46, Cibinong, Jawa Barat 16911 Bogor, Indonesia
| | - Masteria Y. Putra
- Research Center for Biotechnology, Research Organization for Life Science, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor KM. 46, Cibinong, Jawa Barat 16911 Bogor, Indonesia
| |
Collapse
|
40
|
Zhang Y, Xu J, Qiu Z, Guan Y, Zhang X, Zhang X, Chai D, Chen C, Hu Q, Wang W. STK25 enhances hepatocellular carcinoma progression through the STRN/AMPK/ACC1 pathway. Cancer Cell Int 2022; 22:4. [PMID: 34986838 PMCID: PMC8734210 DOI: 10.1186/s12935-021-02421-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Serine/threonine protein kinase 25 (STK25) plays an important role in regulating glucose and insulin homeostasis and in ectopic lipid accumulation. It directly affects the progression and prognosis of nonalcoholic fatty liver disease (NAFLD). However, the effects of STK25 on lipid metabolism in hepatocellular carcinoma (HCC) remain unexplored. The aim of this study was to investigate the role of STK25 in HCC and to elucidate the underlying mechanisms. Methods Immunohistochemistry was used to measure the expression of STK25 in hepatic tissues of HCC patients, and public datasets were used as supplementary material for predicting the expression of STK25 and the prognosis of patients with HCC. The interaction between STK25 and striatin (STRN) was determined by the STRING database, immunohistochemistry and western blot analyses. The involved signaling pathway was detected by the KEGG database and western blot. Moreover, the biological behaviors of the HCC cells were detected by wound healing assays, Transwell invasion assays and oil red O staining. Finally, it was verified again by xenograft model. Results STK25 is highly expressed in HCC patients and is associated with poor prognosis. STK25 knockdown inhibited the HCC cell invasion and proliferation, promotes apoptosis. Consistently, STK25 knockdown inhibited tumor growth in xenograft mouse model. Besides, STK25 deficiency decreased lipid synthesis, energy reserve, epithelial-mesenchymal transition (EMT) by down-regulating lipid metabolism signaling pathway. STRN could reverse the change of lipid metabolism. Conclusions Our results demonstrated that STK25 interacted with STRN to regulates the energy reserve and EMT via lipid metabolism reprogramming. Accordingly, high expression of STK25 may be associated with HCC patients and poor prognosis, which implicates STK25 could be a potential target for lipid metabolism in cancer therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02421-w.
Collapse
Affiliation(s)
- Yichao Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junhui Xu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhendong Qiu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yongjun Guan
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - XiaoYi Zhang
- Intensive Care Unit, ZhongNan Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dongqi Chai
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qinyong Hu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
41
|
Abdalla AN, Di Stefano M, Poli G, Tuccinardi T, Bader A, Vassallo A, Abdallah ME, El-Readi MZ, Refaat B, Algarni AS, Ahmad R, Alkahtani HM, Abdel-Aziz AAM, El-Azab AS, Alqathama A. Co-Inhibition of P-gp and Hsp90 by an Isatin-Derived Compound Contributes to the Increase of the Chemosensitivity of MCF7/ADR-Resistant Cells to Doxorubicin. Molecules 2021; 27:molecules27010090. [PMID: 35011321 PMCID: PMC8746493 DOI: 10.3390/molecules27010090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is a complex and multi-drug resistant (MDR) disease, which could result in the failure of many chemotherapeutic clinical agents. Discovering effective molecules from natural products or by derivatization from known compounds is the interest of many research studies. The first objective of the present study is to investigate the cytotoxic combinatorial, chemosensitizing, and apoptotic effects of an isatin derived compound (5,5-diphenylimidazolidine-2,4-dione conjugated with 5-substituted isatin, named HAA2021 in the present study) against breast cancer cells (MCF7) and breast cancer cells resistant to doxorubicin (MCF7/ADR) when combined with doxorubicin. The second objective is to investigate the binding mode of HAA2021 withP-glycoprotein (P-gp) and heat shock protein 90 (Hsp90), and to determine whether their co-inhibition by HAA2021 contribute to the increase of the chemosensitization of MCF7/ADR cells to doxorubicin. The combination of HAA2021, at non-toxic doses, with doxorubicin synergistically inhibited the proliferation while inducing significant apoptosis in MCF7 cells. Moreover, HAA2021 increased the chemosensitization of MCF7/ADR cells to doxorubicin, resulting in increased cytotoxicity/selectivity and apoptosis-inducing efficiency compared with the effect of doxorubicin or HAA2021 alone against MCF7/ADR cells. Molecular modeling showed that two molecules of HAA2021 bind to P-gp at the same time, causing P-gp inhibitory effect of the MDR efflux pump, and accumulation of Rhodamine-123 (Rho123) in MCF7/ADR cells. Furthermore, HAA2021 stably interacted with Hsp90α more efficiently compared with 17-N-allylamino-17-demethoxygeldanamycin (17-AAG), which was confirmed with the surface plasmon resonance (SPR) and molecular modeling studies. Additionally, HAA2021 showed multi-target effects via the inhibition of Hsp90 and nuclear factor kappa B (NF-𝜅B) proteins in MCF7 and MCF7/ADR cells. Results of real time-PCR also confirmed the synergistic co-inhibition of P-gp/Hsp90α genes in MCF7/ADR cells. Further pharmacokinetic and in vivo studies are warranted for HAA2021 to confirm its anticancer capabilities.
Collapse
Affiliation(s)
- Ashraf N. Abdalla
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
- Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
- Correspondence: or
| | - Miriana Di Stefano
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.S.); (G.P.); (T.T.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.S.); (G.P.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.S.); (G.P.); (T.T.)
| | - Ammar Bader
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
| | - Antonio Vassallo
- Dipartimento di Scienze, Università Degli Studi della Basilicata, 85100 Potenza, Italy;
| | - Mohamed E. Abdallah
- Department of Clinical Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.Z.E.-R.)
| | - Mahmoud Zaki El-Readi
- Department of Clinical Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.Z.E.-R.)
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Bassem Refaat
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Alanood S. Algarni
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
| | - Rizwan Ahmad
- Natural Products and Alternative Medicines, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Hamad M. Alkahtani
- College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (H.M.A.); (A.A.-M.A.-A.); (A.S.E.-A.)
| | - Alaa A.-M. Abdel-Aziz
- College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (H.M.A.); (A.A.-M.A.-A.); (A.S.E.-A.)
| | - Adel S. El-Azab
- College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (H.M.A.); (A.A.-M.A.-A.); (A.S.E.-A.)
| | - Aljawharah Alqathama
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
| |
Collapse
|
42
|
Ray B, Ali I, Jana S, Mukherjee S, Pal S, Ray S, Schütz M, Marschall M. Antiviral Strategies Using Natural Source-Derived Sulfated Polysaccharides in the Light of the COVID-19 Pandemic and Major Human Pathogenic Viruses. Viruses 2021; 14:35. [PMID: 35062238 PMCID: PMC8781365 DOI: 10.3390/v14010035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
Only a mere fraction of the huge variety of human pathogenic viruses can be targeted by the currently available spectrum of antiviral drugs. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak has highlighted the urgent need for molecules that can be deployed quickly to treat novel, developing or re-emerging viral infections. Sulfated polysaccharides are found on the surfaces of both the susceptible host cells and the majority of human viruses, and thus can play an important role during viral infection. Such polysaccharides widely occurring in natural sources, specifically those converted into sulfated varieties, have already proved to possess a high level and sometimes also broad-spectrum antiviral activity. This antiviral potency can be determined through multifold molecular pathways, which in many cases have low profiles of cytotoxicity. Consequently, several new polysaccharide-derived drugs are currently being investigated in clinical settings. We reviewed the present status of research on sulfated polysaccharide-based antiviral agents, their structural characteristics, structure-activity relationships, and the potential of clinical application. Furthermore, the molecular mechanisms of sulfated polysaccharides involved in viral infection or in antiviral activity, respectively, are discussed, together with a focus on the emerging methodology contributing to polysaccharide-based drug development.
Collapse
Affiliation(s)
- Bimalendu Ray
- Department of Chemistry, The University of Burdwan, Burdwan 713104, West Bengal, India; (I.A.); (S.J.); (S.M.); (S.P.)
| | - Imran Ali
- Department of Chemistry, The University of Burdwan, Burdwan 713104, West Bengal, India; (I.A.); (S.J.); (S.M.); (S.P.)
| | - Subrata Jana
- Department of Chemistry, The University of Burdwan, Burdwan 713104, West Bengal, India; (I.A.); (S.J.); (S.M.); (S.P.)
| | - Shuvam Mukherjee
- Department of Chemistry, The University of Burdwan, Burdwan 713104, West Bengal, India; (I.A.); (S.J.); (S.M.); (S.P.)
| | - Saikat Pal
- Department of Chemistry, The University of Burdwan, Burdwan 713104, West Bengal, India; (I.A.); (S.J.); (S.M.); (S.P.)
| | - Sayani Ray
- Department of Chemistry, The University of Burdwan, Burdwan 713104, West Bengal, India; (I.A.); (S.J.); (S.M.); (S.P.)
| | - Martin Schütz
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
43
|
Seipp K, Geske L, Opatz T. Marine Pyrrole Alkaloids. Mar Drugs 2021; 19:514. [PMID: 34564176 PMCID: PMC8471394 DOI: 10.3390/md19090514] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022] Open
Abstract
Nitrogen heterocycles are essential parts of the chemical machinery of life and often reveal intriguing structures. They are not only widespread in terrestrial habitats but can also frequently be found as natural products in the marine environment. This review highlights the important class of marine pyrrole alkaloids, well-known for their diverse biological activities. A broad overview of the marine pyrrole alkaloids with a focus on their isolation, biological activities, chemical synthesis, and derivatization covering the decade from 2010 to 2020 is provided. With relevant structural subclasses categorized, this review shall provide a clear and timely synopsis of this area.
Collapse
Affiliation(s)
| | | | - Till Opatz
- Department of Chemistry, Organic Chemistry Section, Johannes Gutenberg University, Duesbergweg 10–14, 55128 Mainz, Germany; (K.S.); (L.G.)
| |
Collapse
|