101
|
Genel ME, Adacan K, Selvi S, Kutucu DE, Uvez A, Armutak EI, Sengul A, Ulukaya E, Gurevin EG. Apoptosis-inducing, anti-angiogenic and anti-migratory effects of a dinuclear Pd(II) complex on breast cancer: A promising novel compound. Microvasc Res 2024; 151:104619. [PMID: 37898331 DOI: 10.1016/j.mvr.2023.104619] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/14/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Because of the high mortality and morbidity rate of breast cancer, successful management of the disease requires synthesis of novel compounds. To this end, ongoing attempts to create new candidates include synthesis of multinuclear metal complexes. The high DNA binding affinity and cytotoxic activity of these complexes makes them promising as breast cancer treatments. This study investigated anti-growth/cytotoxic effect of the dinuclear Pd(II) complex on breast cancer cell lines (MCF-7, MDA-MB-231) using various methods of staining, flow cytometry, and immunoblotting. The study conducted colony formation, invasion, and migration assays were to assess the effect of the complex on metastasis. Increased caspase-3/7 levels and positive annexin V staining were observed in both cell lines, proving apoptosis. Altered TNFR1 and TRADD expression with caspase-8 cleavage followed by BCL-2 inactivation with loss of mitochondrial membrane potential confirmed the presence of apoptosis in MCF-7 and MDA-MB-231, regardless of p53 expression status. The results implied anti-migration properties. Finally, the study used the CAM assay to assess antiangiogenic properties and showed that the complex inhibited angiogenesis. The study concluded the dinuclear Pd(II) complex warrants further in vivo experiments to show its potential in the treatment of breast cancer.
Collapse
Affiliation(s)
- Merve Erkisa Genel
- Istinye University, Molecular Cancer Research Center (ISUMKAM), Istanbul, Turkey; Istanbul Health and Technology University, Faculty of Medicine, Department of Medical Biology, Istanbul, Turkey
| | - Kaan Adacan
- Istinye University, Molecular Cancer Research Center (ISUMKAM), Istanbul, Turkey; Istinye University, Faculty of Science and Art, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Selin Selvi
- Istinye University, Molecular Cancer Research Center (ISUMKAM), Istanbul, Turkey
| | - Deniz Erol Kutucu
- Istanbul University, Faculty of Science, Department of Biology, Istanbul, Turkey
| | - Ayca Uvez
- Istanbul University-Cerrahpasa, Faculty of Veterinary Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Elif Ilkay Armutak
- Istanbul University-Cerrahpasa, Faculty of Veterinary Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Abdurrahman Sengul
- Bulent Ecevit University, Faculty of Science and Art, Department of Chemistry, Zonguldak, Turkey
| | - Engin Ulukaya
- Istinye University, Molecular Cancer Research Center (ISUMKAM), Istanbul, Turkey; Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey.
| | - Ebru Gurel Gurevin
- Istanbul University, Faculty of Science, Department of Biology, Istanbul, Turkey.
| |
Collapse
|
102
|
Siddique R, Mehmood MH, Shehzad MA. Current antioxidant medicinal regime and treatments used to alleviate oxidative stress in infertility issues. FUNDAMENTAL PRINCIPLES OF OXIDATIVE STRESS IN METABOLISM AND REPRODUCTION 2024:287-315. [DOI: 10.1016/b978-0-443-18807-7.00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
103
|
Giridhara Prema S, Chandrasekaran J, Kanekar S, George M, Prasad TSK, Raju R, Dagamajalu S, Balaya RDA. Cisplatin and Procaterol Combination in Gastric Cancer? Targeting Checkpoint Kinase 1 for Cancer Drug Discovery and Repurposing by an Integrated Computational and Experimental Approach. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:8-23. [PMID: 38190280 DOI: 10.1089/omi.2023.0163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Checkpoint kinase 1 (CHK1), a serine/threonine kinase, plays a crucial role in cell cycle arrest and is a promising therapeutic target for drug development against cancers. CHK1 coordinates cell cycle checkpoints in response to DNA damage, facilitating repair of single-strand breaks, and maintains the genome integrity in response to replication stress. In this study, we employed an integrated computational and experimental approach to drug discovery and repurposing, aiming to identify a potent CHK1 inhibitor among existing drugs. An e-pharmacophore model was developed based on the three-dimensional crystal structure of the CHK1 protein in complex with CCT245737. This model, characterized by seven key molecular features, guided the screening of a library of drugs through molecular docking. The top 10% of scored ligands were further examined, with procaterol emerging as the leading candidate. Procaterol demonstrated interaction patterns with the CHK1 active site similar to CHK1 inhibitor (CCT245737), as shown by molecular dynamics analysis. Subsequent in vitro assays, including cell proliferation, colony formation, and cell cycle analysis, were conducted on gastric adenocarcinoma cells treated with procaterol, both as a monotherapy and in combination with cisplatin. Procaterol, in synergy with cisplatin, significantly inhibited cell growth, suggesting a potentiated therapeutic effect. Thus, we propose the combined application of cisplatin and procaterol as a novel potential therapeutic strategy against human gastric cancer. The findings also highlight the relevance of CHK1 kinase as a drug target for enhancing the sensitivity of cytotoxic agents in cancer.
Collapse
Affiliation(s)
- Suchitha Giridhara Prema
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Jaikanth Chandrasekaran
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Saptami Kanekar
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Mejo George
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | | | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | | |
Collapse
|
104
|
Ghorbanpour M, Shayanfar A, Soltani B. Copper pyrazole complexes as potential anticancer agents: Evaluation of cytotoxic response against cancer cells and their mechanistic action at the molecular level. Coord Chem Rev 2024; 498:215459. [DOI: 10.1016/j.ccr.2023.215459] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
105
|
Lateef Al-Awsi GR, Arshed U, Arif A, Ramírez-Coronel AA, Alhassan MS, Mustafa YF, Rahman FF, Zabibah RS, Gupta J, Iqbal MS, Iswanto AH, Farhood B. The Chemoprotective Potentials of Alpha-lipoic Acid against Cisplatin-induced Ototoxicity: A Systematic Review. Curr Med Chem 2024; 31:3588-3603. [PMID: 37165582 DOI: 10.2174/0929867330666230509162513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/08/2023] [Accepted: 04/05/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE Ototoxicity is one of the major adverse effects of cisplatin therapy which restrict its clinical application. Alpha-lipoic acid administration may mitigate cisplatin-induced ototoxicity. In the present study, we reviewed the protective potentials of alpha-lipoic acid against the cisplatin-mediated ototoxic adverse effects. METHODS Based on the PRISMA guideline, we performed a systematic search for the identification of all relevant studies in various electronic databases up to June 2022. According to the inclusion and exclusion criteria, the obtained articles (n=59) were screened and 13 eligible articles were finally included in the present study. RESULTS The findings of in-vitro experiments showed that cisplatin treatment significantly reduced the auditory cell viability in comparison with the control group; nevertheless, the alpha-lipoic acid co-administration protected the cells against the reduction of cell viability induced by cisplatin treatment. Moreover, the in-vivo results of the auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) tests revealed a decrease in DPOAE and an increase in ABR threshold of cisplatin-injected animals; however, it was shown that alpha-lipoic acid co-treatment had an opposite pattern on the evaluated parameters. Other findings demonstrated that cisplatin treatment could significantly induce the biochemical and histopathological alterations in inner ear cells/tissue; in contrast, alpha-lipoic acid co-treatment ameliorated the cisplatin-mediated biochemical and histological changes. CONCLUSION The findings of audiometry, biochemical parameters, and histological evaluation showed that alpha-lipoic acid co-administration alleviates the cisplatin-induced ototoxicity. The protective role of alpha-lipoic acid against the cisplatin-induced ototoxicity can be due to different mechanisms of anti-oxidant, anti-apoptotic, anti-inflammatory activities, and regulation of cell cycle progression.
Collapse
Affiliation(s)
| | - Uzma Arshed
- Gujranwala Medical College, Gujranwala, Pakistan
| | - Anam Arif
- Gujranwala Medical College, Gujranwala, Pakistan
| | | | - Muataz S Alhassan
- Division of Advanced Nanomaterial Technologies, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul-41001, Iraq
| | - Ferry Fadzlul Rahman
- Public Health Department, Universitas Muhammadiyah Kalimantan Timur, Samarinda, Indonesia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Acim Heri Iswanto
- Public Health Department, Faculty of Health Science, University of Pembangunan Nasional Veteran Jakarta, Jakarta, Indonesia
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
106
|
Li F, Wang H, Xiang Y, Li R, Li C, Zhong R, Li J, Xiong S, Liang L, He J, Liang W. Comparison of different maintenance regimens following first-line immunochemotherapy for advanced non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:2381-2391. [PMID: 38205212 PMCID: PMC10775013 DOI: 10.21037/tlcr-23-489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024]
Abstract
Background Immunochemotherapy is the standard first-line treatment for non-small cell lung cancer (NSCLC). However, the ideal combination strategy and maintenance regimen remain uncertain. This study aims to compare the clinical efficacy of different first-line maintenance regimens for advanced EGFR/ALK (epidermal growth factor receptor/anaplastic lymphoma Kinase) negative NSCLC and explore the eligibility of chemo-free maintenance. Methods We conducted a retrospective evaluation of 1,510 EGFR/ALK negative NSCLC patients who received immune checkpoint inhibitors (ICIs) treatment in our center from 2019 to 2021. Patients who had controlled disease after 2-6 cycles of first-line ICIs in combination with platinum-based doublet chemotherapy with or without anti-angiogenesis were included. Four maintenance regimens were analyzed: ICIs plus platinum-free chemotherapy with (group 1, I+C+A) or without anti-angiogenesis maintenance (group 2, I+C), single-agent ICIs maintenance (group 3, I) or ICIs plus anti-angiogenesis maintenance (group 4, I+A). For group 3-4, rechallenge with initial chemo-agents was given upon the first progression, those who achieved controlled disease were repeatedly followed by another chemo-free period. The primary outcome was progression-free survival (PFS). Notably, for group 3-4, PFS was characterized as the duration between treatment initiation and failure of rechallenge (last disease progression). Results In total, 140 eligible patients in the maintenance phase were analyzed, with 20, 40, 42, and 38 patients in groups 1 to 4, respectively, displaying comparable baselines. Median PFS was similar in the I+C+A maintenance group (22.6 months), I+C maintenance group (21.0 months), and I+A maintenance group (21.5 months), whereas PFS was inferior in group 4 with I maintenance alone (13.4 months). Median chemo-free duration were 6.3 months in I maintenance group, while 13.5 months in I+A maintenance group. During the maintenance period of group 1 to 4, 25%, 25%, 19%, and 42% of patients experienced partial response (PR) again, respectively. Fifty-five percent, 65%, 48% and 61% of patients sustained durable disease control at the end of follow-up. In group 4, 39% of patients received progressive disease (PD) and rechallenge initial chemo-agents. Fifty percent of patients achieved PR and resumed to chemo-free maintenance. Conclusions We showed that following first-line immunochemotherapy, chemo-free maintenance by ICIs plus anti-angiogenesis and on-demand chemo-rechallenge provided comparable efficacy to chemo-on maintenance in terms of PFS, thus allowing the minimization of cytotoxic drugs without compromising therapeutic effectiveness. In addition, anti-angiogenesis is essential during chemo-free maintenance.
Collapse
Affiliation(s)
- Feng Li
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Huiting Wang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Yang Xiang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Run Li
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ran Zhong
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Shan Xiong
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Lixian Liang
- Cancer Centre, Faculty of Health Sciences, Macau, China
- MoE Frontiers Science Centre for Precision Oncology, University of Macau, Macau, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| |
Collapse
|
107
|
Guo L, Li P, Li J, Gong Y, Li X, Wen T, Wu X, Yang X, Liu Z. Potent Half-Sandwich 16-/18-Electron Iridium(III) and Ruthenium(II) Anticancer Complexes with Readily Available Amine-Imine Ligands. Inorg Chem 2023; 62:21379-21395. [PMID: 38096360 DOI: 10.1021/acs.inorgchem.3c03471] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
The synthesis and biological evaluation of stable 16-electron half-sandwich complexes have remained scarce. We herein present the different coordination modes (16-electron or 18-electron) between half-sandwich iridium(III) complexes and ruthenium(II) complexes derived from the same amine-imine ligands chelating hybrid sp3-N/sp2-N donors. The 16-electron iridium(III) and 18-electron ruthenium(II) complexes with different counteranions were obtained and identified by various techniques. The promising cytotoxicity of these complexes against A549 lung cancer cells, cisplatin-resistant A549/DPP cells, cervical carcinoma HeLa cells, and human hepatocellular liver carcinoma HepG2 cells was observed with IC50 values ranging from 5.4 to 16.3 μM. Moreover, these complexes showed a certain selectivity (selectivity index: 2.1-3.7) toward A549 cells and BEAS-2B normal cells. The variation of metal center, counteranion, 16/18-electron coordination mode, and ligand substituents showed little influence on the cytotoxicity and selectivity of these complexes. The mechanism of action study showed that these complexes could target mitochondria, induce the depolarization of the mitochondrial membrane, and promote the generation of intracellular reactive oxygen species (ROS). Further, the induction of cell apoptosis and the perturbation of the cell cycle in the G0/G1 phase were also observed for these complexes. Overall, it seems that the redox mechanism dominated the anticancer efficacy of these complexes.
Collapse
Affiliation(s)
- Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Jiaxing Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Xiaoyuan Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Tingjun Wen
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Xinxin Wu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Xinyi Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| |
Collapse
|
108
|
Siva M, Das K, Guha S, Sivagnanam S, Das G, Saha A, Stewart A, Maity B, Das P. Liposomes Containing Zinc-Based Chemotherapeutic Drug Block Proliferation and Trigger Apoptosis in Breast Cancer Cells. ACS APPLIED BIO MATERIALS 2023; 6:5310-5323. [PMID: 37988654 DOI: 10.1021/acsabm.3c00587] [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] [Indexed: 11/23/2023]
Abstract
Platinum-based chemotherapeutic drugs are effective in killing malignant cells but often trigger drug resistance or off-target side effects. Unlike platinum, zinc is used as an endogenous cofactor for several cellular enzymes and may, thus, display increased biocompatibility. In this present study, we have rationally designed and synthesized two substituted phenanthro[9,10-d]imidazole-based ligands L1 and L2 with pyridine and quinoline substitution at the 2 position and their corresponding Zn(II) complexes; (L1)2Zn and (L2)2Zn, which are characterized by standard analytical and spectroscopic methods. (L2)2Zn, but not (L1)2Zn has intrinsic fluorescence, indicating its potential utility in imaging applications. To facilitate cellular uptake, we generated liposomal formations with a phospholipid DMPC (1,2-Dimyristoyl-sn-glycero-3-phosphocholine) through molecular self-assembly. These liposomal formulations Lip-(L1)2Zn and Lip-(L2)2Zn were able to enter breast cancer cells, induce DNA fragmentation, arrest the cell cycle at the G0/G1 phase, decrease proliferation, and promote apoptosis by activating the DNA damage response. Importantly, both Lip-(L1)2Zn and Lip-(L2)2Zn decreased the size of breast cancer cell-based spheroids, indicating they may be capable of suppressing tumor growth. Our work represents an important proof-of-concept exercise demonstrating that successful liposomal formation of phenanthro[9,10-d]imidazole-based Zn(II) complexes with inherent optical properties have great promise for the development of imaging probes and efficient anticancer drugs.
Collapse
Affiliation(s)
- Mallayasamy Siva
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Kiran Das
- Department of Systems Biology, Centre of Biomedical Research (CBMR), SGPGI campus, Raebareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Subhabrata Guha
- Department of Signal Transduction and Biogenic Amines, Chittaranajan National Cancer Institute, 37, S.P.Mukherjee Road, Kolkata, West Bengal 700 026, India
| | - Subramaniyam Sivagnanam
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Gaurav Das
- Department of Signal Transduction and Biogenic Amines, Chittaranajan National Cancer Institute, 37, S.P.Mukherjee Road, Kolkata, West Bengal 700 026, India
| | - Abhijit Saha
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Biswanath Maity
- Department of Systems Biology, Centre of Biomedical Research (CBMR), SGPGI campus, Raebareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
109
|
Rickard BP, Overchuk M, Tulino J, Tan X, Ligler FS, Bae-Jump VL, Fenton SE, Rizvi I. Exposure to select PFAS and PFAS mixtures alters response to platinum-based chemotherapy in endometrial cancer cell lines. Environ Health 2023; 22:87. [PMID: 38098045 PMCID: PMC10720226 DOI: 10.1186/s12940-023-01034-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Exposure to per- and poly-fluoroalkyl substances (PFAS) has been associated with significant alterations in female reproductive health. These include changes in menstrual cyclicity, timing of menarche and menopause, and fertility outcomes, as well as increased risk of endometriosis, all of which may contribute to an increased risk of endometrial cancer. The effect of PFAS on endometrial cancer cells, specifically altered treatment response and biology, however, remains poorly studied. Like other gynecologic malignancies, a key contributor to lethality in endometrial cancer is resistance to chemotherapeutics, specifically to platinum-based agents that are used as the standard of care for patients with advanced-stage and/or recurrent disease. OBJECTIVES To explore the effect of environmental exposures, specifically PFAS, on platinum-based chemotherapy response and mitochondrial function in endometrial cancer. METHODS HEC-1 and Ishikawa endometrial cancer cells were exposed to sub-cytotoxic nanomolar and micromolar concentrations of PFAS/PFAS mixtures and were treated with platinum-based chemotherapy. Survival fraction was measured 48-h post-chemotherapy treatment. Mitochondrial membrane potential was evaluated in both cell lines following exposure to PFAS ± chemotherapy treatment. RESULTS HEC-1 and Ishikawa cells displayed differing outcomes after PFAS exposure and chemotherapy treatment. Cells exposed to PFAS appeared to be less sensitive to carboplatin, with instances of increased survival fraction, indicative of platinum resistance, observed in HEC-1 cells. In Ishikawa cells treated with cisplatin, PFAS mixture exposure significantly decreased survival fraction. In both cell lines, increases in mitochondrial membrane potential were observed post-PFAS exposure ± chemotherapy treatment. DISCUSSION Exposure of endometrial cancer cell lines to PFAS/PFAS mixtures had varying effects on response to platinum-based chemotherapies. Increased survival fraction post-PFAS + carboplatin treatment suggests platinum resistance, while decreased survival fraction post-PFAS mixture + cisplatin exposure suggests enhanced therapeutic efficacy. Regardless of chemotherapy sensitivity status, mitochondrial membrane potential findings suggest that PFAS exposure may affect endometrial cancer cell mitochondrial functioning and should be explored further.
Collapse
Affiliation(s)
- Brittany P Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC, 27599, USA
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, Durham, NC, 27709, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA; Engineering Building III, North Carolina State University, Raleigh, NC, 27606, USA
| | - Justin Tulino
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA; Engineering Building III, North Carolina State University, Raleigh, NC, 27606, USA
| | - Xianming Tan
- Department of Biostatistics, University of North Carolina School of Public Health, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Frances S Ligler
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Victoria L Bae-Jump
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, 450 West Drive, Chapel Hill, NC, 27599, USA
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC, 27599, USA
| | - Suzanne E Fenton
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC, 27599, USA
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, Durham, NC, 27709, USA
| | - Imran Rizvi
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC, 27599, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA; Engineering Building III, North Carolina State University, Raleigh, NC, 27606, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, 450 West Drive, Chapel Hill, NC, 27599, USA.
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
110
|
Guidotti L, Tomassi E, Marracci S, Lai M, Lapi D, Pesi R, Pucci L, Novellino E, Albi E, Garcia-Gil M. Effects of Nutraceuticals on Cisplatin-Induced Cytotoxicity in HEI-OC1 Cells. Int J Mol Sci 2023; 24:17416. [PMID: 38139245 PMCID: PMC10743635 DOI: 10.3390/ijms242417416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Cisplatin is a chemotherapeutic drug for the treatment of several solid tumors, whose use is limited by its nephrotoxicity, neurotoxicity, ototoxicity, and development of resistance. The toxicity is caused by DNA cross-linking, increase in reactive oxygen species and/or depletion of cell antioxidant defenses. The aim of the work was to study the effect of antioxidant compounds (Lisosan G, Taurisolo®) or hydrogen sulfide (H2S)-releasing compounds (erucin) in the auditory HEI-OC1 cell line treated with cisplatin. Cell viability was determined using the MTT assay. Caspase and sphingomyelinase activities were measured by fluorometric and colorimetric methods, respectively. Expression of transcription factors, apoptosis hallmarks and genes codifying for antioxidant response proteins were measured by Western blot and/or RT-qPCR. Lisosan G, Taurisolo® and erucin did not show protective effects. Sodium hydrosulfide (NaHS), a donor of H2S, increased the viability of cisplatin-treated cells and the transcription of heme oxygenase 1, superoxide dismutase 2, NAD(P)H quinone dehydrogenase type 1 and the catalytic subunit of glutamate-cysteine ligase and decreased reactive oxygen species (ROS), the Bax/Bcl2 ratio, caspase-3, caspase-8 and acid sphingomyelinase activity. Therefore, NaHS might counteract the cytotoxic effect of cisplatin by increasing the antioxidant response and by reducing ROS levels and caspase and acid sphingomyelinase activity.
Collapse
Affiliation(s)
- Lorenzo Guidotti
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
| | - Elena Tomassi
- Institute of Agricultural Biology and Biotechnology, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (E.T.); (L.P.)
| | - Silvia Marracci
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
| | - Michele Lai
- Retrovirus Centre, Department of Translational Medicine and New Technologies in Medicine and Surgery, University of Pisa, Strada Statale del Brennero 2, 56127 Pisa, Italy;
| | - Dominga Lapi
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
| | - Rossana Pesi
- Biochemistry Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy;
| | - Laura Pucci
- Institute of Agricultural Biology and Biotechnology, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (E.T.); (L.P.)
| | - Ettore Novellino
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, Interno Orto Botanico, University of Perugia, Via Romana, 06126 Perugia, Italy;
| | - Mercedes Garcia-Gil
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
111
|
Sahu A, Pradhan D, Veer B, Kumar S, Singh R, Raza K, Rizvi MA, Jain AK, Verma S. In silico screening, synthesis, characterization and biological evaluation of novel anticancer agents as potential COX-2 inhibitors. Daru 2023; 31:119-133. [PMID: 37454036 PMCID: PMC10624798 DOI: 10.1007/s40199-023-00467-x] [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: 01/06/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Cyclooxygenase enzyme is frequently overexpressed in various types of cancer and found to play a crucial role in poor prognosis in cancer patients. In current research, we have reported the new COX-2 inhibitors for cancer treatment using computer-aided drug design and experimental validation. METHODS A total of 12,795 compounds from the different databases were used to screen against the COX-2 enzyme. It perceived three new compounds with better binding affinity to the enzyme. Afterwards, physicochemical properties and in silico bioactivity were assessed for efficacy, safety, and structural features required for binding. The molecules were synthesized and confirmed by spectroscopic techniques. Later on, molecules were evaluated for their anti-cancer activity using MCF-7, MDA-MB-231 and SiHa cancer cell lines. RESULTS Compound ZINC5921547 and ZINC48442590 (4a, and 4b) reduced the MCF-7, MDA-MB-231, and SiHa cells proliferation potently than parent compounds. The PG-E2 estimation shown, both compounds act through the COX-2 PGE2 axis. Compound 4a and 4b block the cell cycle at G1-S phase and induce cancer cell death. CONCLUSIONS We concluded that compounds 4a and 4b effectively promotes cancer cell death via COX-2 PGE2 axis, and further in vivo studies can be evaluated for development in both compounds as anticancer agents. The compilation of this information will help us to generate better outcome through robust computational methods. The high-quality experimental results may pave the way for identifying effective drug candidates for cancer treatment.
Collapse
Affiliation(s)
- Ankita Sahu
- Tumor Biology, ICMR-National Institute of Pathology, New Delhi, 110029, India
| | - Dibyabhaba Pradhan
- Indian Biological Data Center, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Babita Veer
- Department of Applied Chemistry, Delhi Technological University, New Delhi, 110042, India
| | - Sumit Kumar
- Tumor Biology, ICMR-National Institute of Pathology, New Delhi, 110029, India
| | - Ram Singh
- Department of Applied Chemistry, Delhi Technological University, New Delhi, 110042, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India
| | - Moshahid A Rizvi
- Department of Bioscience, Jamia Millia Islamia, New Delhi, 110025, India
| | - Arun Kumar Jain
- Biomedical Informatics Centre, ICMR-National Institute of Pathology, New Delhi, 110029, India
| | - Saurabh Verma
- Tumor Biology, ICMR-National Institute of Pathology, New Delhi, 110029, India.
| |
Collapse
|
112
|
Mudi A, Ray S, Bera M, Dolai M, Das M, Kundu P, Laha S, Choudhuri I, Chandra Samanta B, Bhattacharyya N, Maity T. A multi-spectroscopic and molecular docking approach for DNA/protein binding study and cell viability assay of first-time reported pendent azide bearing Cu(II)-quercetin and dicyanamide bearing Zn(II)-quercetin complexes. Heliyon 2023; 9:e22712. [PMID: 38125469 PMCID: PMC10731082 DOI: 10.1016/j.heliyon.2023.e22712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023] Open
Abstract
In the current study, one new quercetin-based Zn(II) complex [Zn(Qr)(CNNCN)(H2O)2] (Complex 1) which is developed by condensation of quercetin with ZnCl2 in the presence of NaN(CN)2 and Cu(II) complex [Cu(Qr)N3(CH3OH)(H2O)] (complex 2) which is developed by the condensation reaction of quercetin and CuCl2 in presence of NaN3, are thoroughly examined in relation to their use in biomedicine. The results of several spectroscopic studied confirm the structure of both the complexes and the Density Functional Theory (DFT) study helps to optimize the structure of complex 1 and 2. After completion of the identification process, DNA and Human Serum Albumin (HSA) binding efficacy of both the investigated complexes are performed by implementing a long range of biophysical studies and a thorough analysis of the results unveils that complex 1 has better interaction efficacy with the macromolecules than complex 2. The binding efficacy of complex 1 is comparatively higher towards both macromolecules because of its pure groove binding mode during interaction with DNA and the presence of an extra H-bond during connection with HSA. The experimental host-guest binding results is fully validated by molecular docking study. Interestingly complex 1 shows better antioxidant properties than complex 2, as well as quercetin, and it has strong anticancer property with minimal damage to normal cells, which is proved by the MTT assay study. Better DNA and HSA binding efficacy of 1 may be the reason for the better anticancer property of complex 1.
Collapse
Affiliation(s)
- Anupam Mudi
- Department of Botany, Behala College, Behala, India
| | - Shubham Ray
- Department of Chemistry, Prabhat Kumar College, Contai, Contai, Purba Medinipur, 721404, India
| | - Manjushree Bera
- Department of Nutrition, Prabhat Kumar College, Contai, Contai, Purba Medinipur, 721404, India
| | - Malay Dolai
- Department of Chemistry, Prabhat Kumar College, Contai, Contai, Purba Medinipur, 721404, India
| | - Manik Das
- Department of Chemistry, Prabhat Kumar College, Contai, Contai, Purba Medinipur, 721404, India
| | - Pronab Kundu
- Department of Chemistry, Presidency University, Yelahanka, Bengaluru, 560064, India
| | | | | | | | | | - Tithi Maity
- Department of Chemistry, Prabhat Kumar College, Contai, Contai, Purba Medinipur, 721404, India
| |
Collapse
|
113
|
Karthick K, Abinaya M, Shankar T, Swarnalatha K. In Vitro and In Silico Screening of Benzimidazole-Based Ruthenium(II) Complexes as Potent ALK Inhibitor for Cancer Prevention. Appl Biochem Biotechnol 2023; 195:7397-7413. [PMID: 37000352 DOI: 10.1007/s12010-023-04435-8] [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: 03/15/2023] [Indexed: 04/01/2023]
Abstract
Two new heteroleptic Ru(II) polypyridyl complexes, [Ru(bpy)2(B)]Cl2 (RBB) (bpy = 2,2'-bipyridine and B = 4,4'-bis(benzimidazolyl)-2,2'-bipyridine) and [Ru(phen)2(B)]Cl2 (RPB), were synthesized, and the structural features were confirmed by the analytical and spectral tools such as FT-IR, 1H-NMR, and UV-Visible spectroscopy. We have explored the possibility of improving the selectivity of cytotoxic Ru(II) complex and their preliminary biological evaluation against MCF-7 and MG-63 cell lines and clinical pathogens. The results of the antimicrobial screening show that the ligand and complexes have a range of abilities against the species of bacteria and fungi that were tested. The anti-inflammatory activity of the compounds was found to be in the range of 30-75%. Molecular docking study was performed for these ligand and complexes to evaluate and analyze the anti-lymphoma cancer activity. Molecular docking score and the rank revealed the bonding affinity towards the site of interaction of the oncoprotein anaplastic lymphoma kinase (ALK).
Collapse
Affiliation(s)
- Kamaraj Karthick
- Photochemistry Research Laboratory, Department of Chemistry, Manonmaniam Sundaranar University, Abishekapatti, Tirunelveli, 627012, Tamil Nadu, India
| | - Muthukumar Abinaya
- Photochemistry Research Laboratory, Department of Chemistry, Manonmaniam Sundaranar University, Abishekapatti, Tirunelveli, 627012, Tamil Nadu, India
| | - Thangaraj Shankar
- Photochemistry Research Laboratory, Department of Chemistry, Manonmaniam Sundaranar University, Abishekapatti, Tirunelveli, 627012, Tamil Nadu, India
| | - Kalaiyar Swarnalatha
- Photochemistry Research Laboratory, Department of Chemistry, Manonmaniam Sundaranar University, Abishekapatti, Tirunelveli, 627012, Tamil Nadu, India.
| |
Collapse
|
114
|
Martínez-Martel I, Pol O. A Novel Therapy for Cisplatin-Induced Allodynia and Dysfunctional and Emotional Impairments in Male and Female Mice. Antioxidants (Basel) 2023; 12:2063. [PMID: 38136183 PMCID: PMC10741113 DOI: 10.3390/antiox12122063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Patients undergoing chemotherapy with cisplatin (CIS) develop neuropathy in addition to other symptoms such as, anxiety, depression, muscle wasting and body weight loss. This symptomatology greatly weakens patients and may even lead to adjournment of chemotherapy. The protecting actions of molecular hydrogen in many neurological illnesses have been described, but its effect on the functional and emotional deficiencies caused by CIS has not been assessed. In C57BL/6J male and female mice injected with CIS, we examined the impact of the prophylactic treatment with hydrogen-rich water (HRW) on: (i) the tactile and cold allodynia, (ii) the deficits of grip strength and weight loss, (iii) the anxiodepressive-like behaviors and (iv) the inflammatory and oxidative reactions incited by CIS in the dorsal root ganglia (DRG) and prefrontal cortex (PFC). The results demonstrate that the mechanical allodynia and the anxiodepressive-like comportment provoked by CIS were similarly manifested in both sexes, whereas the cold allodynia, grip strength deficits and body weight loss produced by this chemotherapeutic agent were greater in female mice. Nonetheless, the prophylactic treatment with HRW prevented the allodynia and the functional and emotional impairments resulting from CIS in both sexes. This treatment also inhibited the inflammatory and oxidative responses activated by CIS in the DRG and PFC in both sexes, which might explain the therapeutic actions of HRW in male and female mice. In conclusion, this study revealed the plausible use of HRW as a new therapy for the allodynia and physical and mental impairments linked with CIS and its possible mechanism of action.
Collapse
Affiliation(s)
- Ignacio Martínez-Martel
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Olga Pol
- Grup de Neurofarmacologia Molecular, Institut de Recerca Sant Pau, Sant Quintí 77-79, 08041 Barcelona, Spain
- Grup de Neurofarmacologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
115
|
Abdoul-Latif FM, Ainane A, Houmed Aboubaker I, Mohamed J, Ainane T. An Overview of Cancer in Djibouti: Current Status, Therapeutic Approaches, and Promising Endeavors in Local Essential Oil Treatment. Pharmaceuticals (Basel) 2023; 16:1617. [PMID: 38004482 PMCID: PMC10674319 DOI: 10.3390/ph16111617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Djibouti, a developing economy, grapples with significant socioeconomic obstacles and the prevalence of infectious pathologies, including certain forms of neoplasms. These challenges are exacerbated by limited access to affordable medical technologies for diagnosis, coupled with a lack of preventive interventions, particularly in disadvantaged areas. The attention devoted to local phytotherapeutic treatments underscores the uniqueness of Djibouti's flora, resulting from its distinctive geographical position. International focus specifically centers on harnessing this potential as a valuable resource, emphasizing the phytoconstituents used to counter pathologies, notably carcinomas. This comprehensive overview covers a broad spectrum, commencing with an examination of the current state of knowledge, namely an in-depth investigation of oncological risk factors. Essential elements of control are subsequently studied, highlighting the fundamental prerequisites for effective management. The significance of dietary habits in cancer prevention and support is explored in depth, while traditional methods are examined, highlighting the cultural significance of indigenous essential oil therapies and encouraging further research based on the promising results.
Collapse
Affiliation(s)
- Fatouma Mohamed Abdoul-Latif
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti P.O. Box 486, Djibouti;
| | - Ayoub Ainane
- Superior School of Technology of Khenifra (EST-Khenifra), University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| | | | - Jalludin Mohamed
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti P.O. Box 486, Djibouti;
| | - Tarik Ainane
- Superior School of Technology of Khenifra (EST-Khenifra), University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| |
Collapse
|
116
|
Abirami A, Devan U, Ramesh R, Antony Joseph Velanganni A, Grzegorz Małecki J. Naphthoyl benzhydrazine-decorated binuclear arene Ru(II) complexes as anticancer agents targeting human breast cancer cells. Dalton Trans 2023; 52:16376-16387. [PMID: 37870147 DOI: 10.1039/d3dt02552g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Breast cancer is the most dangerous type in women and its fatality rate has increased over the past decade. To develop more potent and target-specific breast cancer drugs, six arene ruthenium(II) complexes (1-6) containing naphthoyl benzhydrazine ligands (NL1-NL3) were synthesized and characterized by analytical and spectroscopic (infrared, UV-visible, NMR and HR-MS) methods. The SC-XRD analysis of 1 and 6 demonstrates the bis N^O bidentate binding nature of ligands to ruthenium ions and a pseudo-octahedral geometry around the Ru(II) ion. Solution stability studies using UV-Vis spectroscopy evidenced the instantaneous hydrolysis of the complexes to form monoaquated species in a solution of 1 : 9 (v/v) DMSO/phosphate buffer. All the complexes were screened for their in vitro antiproliferative activities against different human breast cancer cells, including MCF-7, SkBr3, MDA-MB-468, MDA-MB-231, and non-cancerous HEK-293 cells, by an MTT assay, and they displayed good cancer cell growth inhibitory capacity with low IC50 values. Notably, complexes 2 and 5 comprising methoxy and p-cymene groups exhibited excellent cytotoxicity towards SkBr3 cells compared to clinical drug cisplatin. AO-EB and HOECHST-33342 staining assays revealed apoptotic morphological changes in complex-treated cancer cells. Further, reactive oxygen species and mitochondrial membrane potential assays validated that the complexes induce apoptotic cell death via an intrinsic mitochondrial pathway with ROS production. In addition, the apoptotic induction and the quantification of late apoptosis were established with the aid of western blot and flow cytometry analysis, respectively.
Collapse
Affiliation(s)
- Arunachalam Abirami
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli - 620 024, India.
| | - Umapathy Devan
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli - 620 024, India
| | - Rengan Ramesh
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli - 620 024, India.
| | - Arockiam Antony Joseph Velanganni
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli - 620 024, India
| | - Jan Grzegorz Małecki
- Department of Crystallography, Institute of Chemistry, University of Silesia, Katowice, Poland
| |
Collapse
|
117
|
Xin Y, Jiang Q, Liu C, Qiu J. Plumbagin has an inhibitory effect on the growth of TSCC PDX model and it enhances the anticancer efficacy of cisplatin. Aging (Albany NY) 2023; 15:12225-12250. [PMID: 37925175 PMCID: PMC10683608 DOI: 10.18632/aging.205175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/02/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Head and neck squamous cell carcinomas are the sixth most common malignant tumors worldwide. Tongue squamous cell carcinoma is a common malignant tumor of this type, and it is associated with poor prognosis, a high rate of recurrence and a low survival rate. Plumbagin is derived from Plumbago zeylanica L, several studies report that plumbagin could inhibit cell, tumor metastasis, induce apoptosis in various cancer cells. Patient-derived xenograft (PDX) model can maintain the heterogeneity and microenvironment of human tumors, is a powerful research tool for developing potentially effective therapies for TSCC. METHODS Tumor tissues obtained from TSCC patients were implanted into immunodeficient mice to establish TSCC PDX models. Subsequently, the PDX models were used to evaluate the anti-tumor effects of plumbagin on TSCC. Furthermore, we conducted next-generation sequencing (NGS) and explored the mRNA expression profiles between the treatment and control groups. We selected eight mRNAs related to the characteristics and prognosis of TSCC patients for further analysis. RESULTS Plumbagin could inhibit the growth of TSCC PDX models and inhibit expression of Akt/mTOR pathway. In addition, plumbagin was shown to increase drug sensitivity to cisplatin. The eight mRNAs selected for further analysis, AXL, SCG5, VOPP1, DCBLD2 and DRAM1 are cancer-promoting genes, DUSP1, AQP5 and BLNK are cancer suppressor genes. And they were related to the diagnosis, growth, prognosis, and immune cell infiltration in TSCC patients. CONCLUSION Plumbagin exhibits an inhibitory effect on the growth of the PDX model of TSCC. Moreover, plumbagin enhances the inhibitory effects of cisplatin.
Collapse
Affiliation(s)
- Yuqi Xin
- Department of Stomatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qingkun Jiang
- Department of Stomatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chenshu Liu
- Department of Stomatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jiaxuan Qiu
- Department of Stomatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
118
|
Gandioso A, Izquierdo-García E, Mesdom P, Arnoux P, Demeubayeva N, Burckel P, Saubaméa B, Bosch M, Frochot C, Marchán V, Gasser G. Ru(II)-Cyanine Complexes as Promising Photodynamic Photosensitizers for the Treatment of Hypoxic Tumours with Highly Penetrating 770 nm Near-Infrared Light. Chemistry 2023; 29:e202301742. [PMID: 37548580 DOI: 10.1002/chem.202301742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/08/2023]
Abstract
Light-activated treatments, such as photodynamic therapy (PDT), provide temporal and spatial control over a specific cytotoxic response by exploiting toxicity differences between irradiated and dark conditions. In this work, a novel strategy for developing near infrared (NIR)-activatable Ru(II) polypyridyl-based photosensitizers (PSs) was successfully developed through the incorporation of symmetric heptamethine cyanine dyes in the metal complex via a phenanthrimidazole ligand. Owing to their strong absorption in the NIR region, the PSs could be efficiently photoactivated with highly penetrating NIR light (770 nm), leading to high photocytotoxicities towards several cancer cell lines under both normoxic and hypoxic conditions. Notably, our lead PS (Ru-Cyn-1), which accumulated in the mitochondria, exhibited a good photocytotoxic activity under challenging low-oxygen concentration (2 % O2 ) upon NIR light irradiation conditions (770 nm), owing to a combination of type I and II PDT mechanisms. The fact that the PS Protoporphyrin IX (PpIX), the metabolite of the clinically approved 5-ALA PS, was found inactive under the same challenging conditions positions Ru-Cyn-1 complex as a promising PDT agent for the treatment of deep-seated hypoxic tumours.
Collapse
Affiliation(s)
- Albert Gandioso
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Eduardo Izquierdo-García
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Martí i Franquès 1-11, 08028, Barcelona, Spain
| | - Pierre Mesdom
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | | | | | - Pierre Burckel
- Université de Paris, Institut de physique du globe de Paris, CNRS, 75005, Paris, France
| | - Bruno Saubaméa
- Cellular and Molecular Imaging platform, US25 Inserm, UAR3612 CNRS, Faculté de Pharmacie de Paris, Université Paris Cité, 75006, Paris, France
| | - Manel Bosch
- Unitat de Microscòpia Òptica Avançada, Centres Científics i Tecnològics, Universitat de Barcelona (CCiTUB), Av. Diagonal, 643, Barcelona, 08028, Spain
| | - Céline Frochot
- Université de Lorraine, CNRS, LRGP, 54000, Nancy, France
| | - Vicente Marchán
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Martí i Franquès 1-11, 08028, Barcelona, Spain
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| |
Collapse
|
119
|
Bai J, Wang H, Li C, Liu L, Wang J, Sun C, Zhang Q. A novel mitochondria-targeting compound exerts therapeutic effects against melanoma by inducing mitochondria-mediated apoptosis and autophagy in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2023; 38:2608-2620. [PMID: 37466182 DOI: 10.1002/tox.23896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/17/2023] [Accepted: 07/01/2023] [Indexed: 07/20/2023]
Abstract
Melanoma is the most invasive skin cancer, with a high mortality rate. However, existing therapeutic drugs have side effects, low reactivity, and lead to drug resistance. As the power source in cells, mitochondria play an important role in the survival of cancer cells and are an important target for tumor therapy. This study aimed to develop a new anti-melanoma compound that targets mitochondria, evaluate its effect on the proliferation and metastasis of melanoma cells, and explore its mechanism of action. The novel mitochondria-targeting compound, SCZ0148, was synthesized by modifying the structure of cyanine. Then, A375 and B16 cells were incubated with different concentrations of SCZ0148, and different doses of SCZ0148 were administered to A375 and B16 xenograft zebrafish. The results showed that SCZ0148 targeted mitochondria, had dose- and time-dependent effects on the proliferation of melanoma cell lines, and had no obvious side effects on normal cells. In addition, SCZ0148 induced melanoma cell apoptosis through the reactive oxygen species-mediated mitochondrial pathway of apoptosis and promoted autophagy. SCZ0148 significantly inhibited the migration of melanoma cells via a matrix metalloprotein 9-mediated pathway. Similarly, SCZ0148 inhibited melanoma cell proliferation in a concentration-dependent manner in vivo. In summary, SCZ0148 may be a novel anti-melanoma compound that targets mitochondria.
Collapse
Affiliation(s)
- Jun Bai
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
| | - Hailan Wang
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, China
| | - Chenwen Li
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
| | - Li Liu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianv Wang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qingbi Zhang
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
120
|
Dutta S, Mahalanobish S, Saha S, Mandal M, Begam S, Sadhukhan P, Ghosh S, Brahmachari G, Sil PC. Biological evaluation of the novel 3,3'-((4-nitrophenyl)methylene)bis(4-hydroxy-2H-chromen-2-one) derivative as potential anticancer agents via the selective induction of reactive oxygen species-mediated apoptosis. Cell Signal 2023; 111:110876. [PMID: 37640193 DOI: 10.1016/j.cellsig.2023.110876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/02/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Selective initiation of programmed cell death in cancer cells than normal cells is reflected as an attractive chemotherapeutic strategy. In the current study, a series of synthetic bis-coumarin derivatives were synthesized possessing reactive oxygen species (ROS) modulating functional groups and examined in four cancerous and two normal cell lines for their cytotoxic ability using MTT assay. Among these compounds, 3 l emerged as the most promising derivative in persuading apoptosis in human renal carcinoma cells (SKRC-45) among diverse cancer cell lines. 3 l causes significantly less cytotoxicity to normal kidney cells compared to cisplatin. This compound was able to induce apoptosis and cell-cycle arrest by modulating the p53 mediated apoptotic pathways via the generation of ROS, decreasing mitochondrial membrane potential, and causing DNA fragmentation. Unlike cisplatin, the 3 l derivative was found to inhibit the nuclear localisation of NF-κB in SKRC-45 cells. It was also found to reduce the proliferation, survival and migration ability of SKRC-45 cells by downregulating COX-2/ PTGES2 cascade and MMP-2. In an in vivo tumor model, 3 l showed an anticancer effect by reducing the mean tumor mass, volume and inducing caspase-3 activation, without affecting kidney function. Further studies are needed to establish 3 l as a promising anti-cancer drug candidate.
Collapse
Affiliation(s)
- Sayanta Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sushweta Mahalanobish
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sukanya Saha
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Mullicka Mandal
- Laboratory of Natural Products and Organic Synthesis, Department of Chemistry, Visva-Bharati (a Central University), Santiniketan 731 235, West Bengal, India
| | - Sanchari Begam
- Laboratory of Natural Products and Organic Synthesis, Department of Chemistry, Visva-Bharati (a Central University), Santiniketan 731 235, West Bengal, India
| | - Pritam Sadhukhan
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sumit Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Goutam Brahmachari
- Laboratory of Natural Products and Organic Synthesis, Department of Chemistry, Visva-Bharati (a Central University), Santiniketan 731 235, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India.
| |
Collapse
|
121
|
Ruiz de Porras V, Figols M, Font A, Pardina E. Curcumin as a hepatoprotective agent against chemotherapy-induced liver injury. Life Sci 2023; 332:122119. [PMID: 37741319 DOI: 10.1016/j.lfs.2023.122119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Despite significant advances in cancer therapeutics, chemotherapy remains the cornerstone of treatment for many tumors. Importantly, however, chemotherapy-induced toxicity, including hepatotoxicity, can lead to the interruption or discontinuation of potentially effective therapy. In recent years, special attention has been paid to the search for complementary therapies to mitigate chemotherapy-induced toxicity. Although there is currently a lack of specific interventions to mitigate or prevent hepatotoxicity in chemotherapy-treated patients, the polyphenol compound curcumin has emerged as a potential strategy to overcome this adverse effect. Here we review, firstly, the molecular and physiological mechanisms and major risk factors of chemotherapy-induced hepatotoxicity. We then present an overview of how curcumin has the potential to mitigate hepatotoxicity by targeting specific molecular mechanisms. Hepatotoxicity is a well-described side effect of cytotoxic drugs that can limit their clinical application. Inflammation and oxidative stress are the most common mechanisms involved in hepatotoxicity. Several studies have shown that curcumin could prevent and/or palliate chemotherapy-induced liver injury, mainly due to its anti-inflammatory, antioxidant, antifibrotic and hypolipidemic properties. Further clinical investigation using bioavailable curcumin formulations is warranted to demonstrate its efficacy as an hepatoprotective agent in cancer patients.
Collapse
Affiliation(s)
- Vicenç Ruiz de Porras
- Grup de Recerca en Toxicologia (GRET), Unitat de Toxicologia, Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Avda Joan XXIII s/n, 08028 Barcelona, Spain; CARE program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain; Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain.
| | - Mariona Figols
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243, Manresa, Barcelona, Spain
| | - Albert Font
- CARE program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain; Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain; Medical Oncology Department, Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
| | - Eva Pardina
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
122
|
Kumari P, Ghosh S, Acharya S, Mitra P, Roy S, Ghosh S, Maji M, Singh S, Mukherjee A. Cytotoxic Imidazolyl-Mesalazine Ester-Based Ru(II) Complexes Reduce Expression of Stemness Genes and Induce Differentiation of Oral Squamous Cell Carcinoma. J Med Chem 2023; 66:14061-14079. [PMID: 37831489 DOI: 10.1021/acs.jmedchem.3c01092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The aggressiveness and recurrence of cancer is linked to cancer stem cells (CSCs), but drugs targeting CSCs may not succeed in the clinic due to the lack of a distinct CSC subpopulation. Clinical Pt(II) drugs can increase stemness. We screened 15 RuII or IrIII complexes with mesalazine or 3-aminobenzoate Schiff bases of the general formulas [Ru(p-cym)L]+, [Ru(p-cym)L], and [Ir(Cp*)L]+ (L = L1-L9) and found three complexes (2, 12, and 13) that are active against oral squamous cell carcinoma (OSCC) CSCs. There is a putative oncogenic role of transcription factors (viz. NOTCH1, SOX2, c-MYC) to enhance the stemness. Our work shows that imidazolyl-mesalazine ester-based RuII complexes inhibit growth of CSC-enriched OSCC 3D spheroids at low micromolar doses (2 μM). Complexes 2, 12, and 13 reduce stemness gene expression and induce differentiation markers (Involucrin, CK10) in OSCC 3D cultures. The imidazolyl-mesalazine ester-based RuII complex 13 shows the strongest effect. Downregulating c-MYC suggests that RuII complexes may target c-MYC-driven cancers.
Collapse
Affiliation(s)
- Pragya Kumari
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Subhashis Ghosh
- National Institute of Biomedical Genomics, Kalyani-741251, West Bengal, India
| | - Sourav Acharya
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Paromita Mitra
- National Institute of Biomedical Genomics, Kalyani-741251, West Bengal, India
| | - Souryadip Roy
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Shilpendu Ghosh
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Moumita Maji
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| | - Sandeep Singh
- National Institute of Biomedical Genomics, Kalyani-741251, West Bengal, India
| | - Arindam Mukherjee
- Department of Chemical Sciences and Centre for Advance Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, Nadia, West Bengal, India
| |
Collapse
|
123
|
AbdelSamad AL, El-Saadi MT, Gouda AM, AboulMagd AM. Pyrrolizine/indolizine-bearing (un)substituted isoindole moiety: design, synthesis, antiproliferative and MDR reversal activities, and in silico studies. RSC Adv 2023; 13:30753-30770. [PMID: 37869384 PMCID: PMC10587743 DOI: 10.1039/d3ra05310e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
Two new series of pyrrolizine/indolizine derivative-bearing (un)substituted isoindole moiety were designed and synthesized. The anticancer potential of the new compounds was evaluated against hepatocellular carcinoma (HepG-2), colorectal carcinoma, colon cancer (HCT-116), and breast cancer (MCF-7) cell lines. Compounds 6d and 6o were the most potent derivatives with IC50 values ranging from 6.02 to 13.87 μM against HePG-2, HCT-116, and MCF-7 cell lines. Moreover, methyl analog of the fluoro-substituted indolizine derivative 6m revealed significant antiproliferative activity against HePG-2, HCT-116, and MCF-7 cancer cell lines with IC50 values of 11.97, 28.37, and 19.87 μM, respectively. The most active anticancer analogs, 6d, 6m, and 6o, were inspected for their putative mechanism of action by estimating their epidermal growth factor receptor (EGFR) and cyclin-dependent kinase (CDK 2) inhibitory activities. Thus, compound 6o displayed the most inhibitory activity against EGFR and CDK 2 with IC50 values of 62 and 118 nM, respectively. Additionally, the quantitative real-time PCR analysis for the P-glycoprotein effect of compounds 6d, 6m, and 6o was performed, in which compound 6o illustrated significant down-regulation of P-gp against the HepG-2 cell line by 0.2732 fold. Mechanistic studies for the most active compounds involving the reversal doxorubicin (DOX) effect of compounds 6d, 6m, and 6o were performed, which illustrated cytotoxic activity with IC50 22.27, 3.88, and 8.79 μM, respectively. Moreover, the apoptotic activity of the most active derivative 6o on HCT-116 cancer cells showed accumulation in the G1 and S phases of the cell cycle.
Collapse
Affiliation(s)
- Amr L AbdelSamad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University in Beni-Suef (NUB) Beni-Suef 62513 Egypt
| | - Mohammed T El-Saadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Beni-Suef University Beni-Suef 62514 Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Kantra Branch Ismailia Egypt
| | - Ahmed M Gouda
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Beni-Suef University Beni-Suef 62514 Egypt
| | - Asmaa M AboulMagd
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University in Beni-Suef (NUB) Beni-Suef 62513 Egypt
| |
Collapse
|
124
|
Zhou M, Boulos JC, Omer EA, Rudbari HA, Schirmeister T, Micale N, Efferth T. Two palladium (II) complexes derived from halogen-substituted Schiff bases and 2-picolylamine induce parthanatos-type cell death in sensitive and multi-drug resistant CCRF-CEM leukemia cells. Eur J Pharmacol 2023; 956:175980. [PMID: 37567459 DOI: 10.1016/j.ejphar.2023.175980] [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: 05/26/2023] [Revised: 07/29/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
The use of cisplatin and its derivatives in cancer treatment triggered the interest in metal-containing complexes as potential novel anticancer agents. Palladium (II)-based complexes have been synthesized in recent years with promising antitumor activity. Previously, we described the synthesis and cytotoxicity of palladium (II) complexes containing halogen-substituted Schiff bases and 2-picolylamine. Here, we selected two palladium (II) complexes with double chlorine-substitution or double iodine-substitution that displayed the best cytotoxicity in drug-sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 leukemia cells for further biological investigation. Surprisingly, these compounds did not significantly induce apoptotic cell death. This study aims to reveal the major mode of cell death of these two palladium (II) complexes. We performed annexin V-FITC/PI staining and flow cytometric mitochondrial membrane potential measurement followed by western blotting, immunofluorescence microscopy, and alkaline single cell electrophoresis (comet assay). J4 and J6 still induced neither apoptosis nor necrosis in both leukemia cell lines. They also insufficiently induced autophagy as evidenced by Beclin and p62 detection in western blotting. Interestingly, J4 and J6 induced a novel mode of cell death (parthanatos) as mainly demonstrated in CCRF-CEM cells by hyper-activation of poly(ADP-ribose) polymerase 1 (PARP) and poly(ADP-ribose) (PAR) using western blotting, flow cytometric measurement of mitochondrial membrane potential collapse, nuclear translocation of apoptosis-inducing factor (AIF) by immunofluorescence microscopy, and DNA damage by alkaline single cell electrophoresis (comet assay). AIF translocation was also observed in CEM/ADR5000 cells. Thus, parthanatos was the predominant mode of cell death induced by J4 and J6, which explains the high cytotoxicity in CCRF-CEM and CEM/ADR5000 cells. J4 and J6 may be interesting drug candidates and deserve further investigations to overcome resistance of tumors against apoptosis. This study will promote the design of further novel palladium (II)-based complexes as chemotherapeutic agents.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, Staudinger Weg 5, 55128, Mainz, Germany
| | - Joelle C Boulos
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, Staudinger Weg 5, 55128, Mainz, Germany
| | - Ejlal A Omer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, Staudinger Weg 5, 55128, Mainz, Germany
| | - Hadi Amiri Rudbari
- Department of Chemistry, University of Isfahan, Isfahan, 81746-73441, Iran
| | - Tanja Schirmeister
- Department of Medicinal Chemistry, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, Staudinger Weg 5, 55128, Mainz, Germany
| | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 1-98166, Messina, Italy
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, Staudinger Weg 5, 55128, Mainz, Germany.
| |
Collapse
|
125
|
Macuer-Guzmán J, Giovagnoli-Vicuña C, Bernal G, Lobos-González L, de la Fuente-Ortega E, Araya-Castillo M, Ibáñez C. Annona cherimola Seed Extracts Trigger an Early Apoptosis Response and Selective Anticlonogenic Activity against the Human Gastric Carcinoma Cell Line SNU-1. Molecules 2023; 28:6906. [PMID: 37836749 PMCID: PMC10574650 DOI: 10.3390/molecules28196906] [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: 06/15/2023] [Revised: 08/19/2023] [Accepted: 09/05/2023] [Indexed: 10/15/2023] Open
Abstract
The aim of this study was to evaluate, for the first time, the antiproliferative, apoptotic and diminishing effects of the anchored growth-independent capacity of an ethanol macerate extract from the Annona cherimola seed (EMCHS) in the human gastric cancer cell line SNU-1. The cells treated with EMCHS (20 μg/mL) significantly reduced the capacity to form clones of the tumor cell. Moreover, 50 μg/mL of EMCHS extract induced apoptosis, as was shown by the Annexin-V assay. UHPLC-MS/MS analysis detected two acetogenins (Annonacinone and Annonacin) in the EMCHS, which could be largely responsible for its selective antiproliferative effect. The identification of fatty acids by GC-FID showed the presence of eight fatty acids, among which was, oleic acid, which has recognized activity as an adjuvant in antitumor treatments. Taken together, our results indicate that the EMCHS seems promising for use as a natural therapy against gastric cancer disease.
Collapse
Affiliation(s)
- Johan Macuer-Guzmán
- Laboratorio de Silvigenómica y Biotecnología, Departamento de Biología, Facultad de Ciencias, Universidad de La Serena, Avenida Raúl Bitrán 1305, Casilla 599, La Serena 1700000, Chile;
- Facultad de Ciencias Agropecuarias, Universidad del Alba, Cuatro Esquinas 060, La Serena 1700000, Chile
| | - Claudia Giovagnoli-Vicuña
- Departamento de Química Inorgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna 4860, Macul, Santiago 7810000, Chile;
| | - Giuliano Bernal
- Laboratorio de Biología Molecular y Celular del Cáncer (CáncerLab), Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo 1781421, Chile;
| | - Lorena Lobos-González
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Avenida las Condes 12438, Lo Barnechea, Santiago 7710162, Chile;
| | - Erwin de la Fuente-Ortega
- Laboratorio de Estrés Celular y Enfermedades Crónicas no Transmisibles, Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo 1781421, Chile;
| | - Michael Araya-Castillo
- Centro de Investigación y Desarrollo Tecnológico de Algas (CIDTA), Facultad de Ciencias del Mar, Universidad Católica del Norte, Larrondo 1281, Coquimbo 1781421, Chile;
| | - Cristian Ibáñez
- Laboratorio de Silvigenómica y Biotecnología, Departamento de Biología, Facultad de Ciencias, Universidad de La Serena, Avenida Raúl Bitrán 1305, Casilla 599, La Serena 1700000, Chile;
- Instituto Multidisciplinario de Investigación y Postgrado, Universidad de La Serena, Avenida Raúl Bitrán 1305, Casilla 599, La Serena 1700000, Chile
| |
Collapse
|
126
|
Ramadan SA, Kamel EM, Alruhaimi RS, Bin-Ammar A, Ewais MA, Khowailed AA, Hassanein EH, Mahmoud AM. An integrated phytochemical, in silico and in vivo approach to identify the protective effect of Caroxylon salicornicum against cisplatin hepatotoxicity. Saudi Pharm J 2023; 31:101766. [PMID: 37731943 PMCID: PMC10507235 DOI: 10.1016/j.jsps.2023.101766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/27/2023] [Indexed: 09/22/2023] Open
Abstract
Cisplatin (CIS) is a chemotherapeutic medication for the treatment of cancer. However, hepatotoxicity is among the adverse effects limiting its use. Caroxylon salicornicum is traditionally used for treating inflammatory diseases. In this investigation, three flavonoids, four coumarins, and three sterols were detected in the petroleum ether fraction of C. salicornicum (PEFCS). The isolated phytochemicals exhibited binding affinity toward Keap1, NF-κB, and SIRT1 in silico. The hepatoprotective role of PEFCS (100, 200 and 400 mg/kg) was investigated in vivo. Rats received PEFCS for 14 days and CIS on day 15. CIS increased ALT, AST and ALP and caused tissue injury along with increased ROS, MDA, and NO. Hepatic NF-κB p65, pro-inflammatory mediators, Bax and caspase-3 were increased in CIS-treated animals while antioxidants and Bcl-2 were decreased. PEFCS mitigated hepatocyte injury, and ameliorated transaminases, ALP, oxidative stress (OS) and inflammatory markers. PEFCS downregulated pro-apoptosis markers and boosted Bcl-2 and antioxidants. In addition, PEFCS upregulated Nrf2, HO-1, and SIRT1 in CIS-administered rats. In conclusion, PEFCS is rich in beneficial phytoconstituents and conferred protection against liver injury by attenuating OS and inflammation and upregulating Nrf2 and SIRT1.
Collapse
Affiliation(s)
| | | | - Reem S. Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Albandari Bin-Ammar
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Saudi Arabia
| | - Madeha A. Ewais
- Physiology Department, Faculty of Medicine, Beni-Suef University, Egypt
| | | | - Emad H.M. Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Egypt
| | - Ayman M. Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Egypt
| |
Collapse
|
127
|
Shi T, Yuan Z, He Y, Zhang D, Chen S, Wang X, Yao L, Shao J, Wang X. Competition between p53 and YY1 determines PHGDH expression and malignancy in bladder cancer. Cell Oncol (Dordr) 2023; 46:1457-1472. [PMID: 37326803 DOI: 10.1007/s13402-023-00823-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/17/2023] Open
Abstract
PURPOSE Serine metabolism is frequently dysregulated in many types of cancers and the tumor suppressor p53 is recently emerging as a key regulator of serine metabolism. However, the detailed mechanism remains unknown. Here, we investigate the role and underlying mechanisms of how p53 regulates the serine synthesis pathway (SSP) in bladder cancer (BLCA). METHODS Two BLCA cell lines RT-4 (WT p53) and RT-112 (p53 R248Q) were manipulated by applying CRISPR/Cas9 to examine metabolic differences under WT and mutant p53 status. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) and non-targeted metabolomics analysis were adopted to identify metabolomes changes between WT and p53 mutant BLCA cells. Bioinformatics analysis using the cancer genome atlas and Gene Expression Omnibus datasets and immunohistochemistry (IHC) staining was used to investigate PHGDH expression. Loss-of-function of PHGDH and subcutaneous xenograft model was adopted to investigate the function of PHGDH in mice BLCA. Chromatin immunoprecipitation (Ch-IP) assay was performed to analyze the relationships between YY1, p53, SIRT1 and PHGDH expression. RESULTS SSP is one of the most prominent dysregulated metabolic pathways by comparing the metabolomes changes between wild-type (WT) p53 and mutant p53 of BLCA cells. TP53 gene mutation shows a positive correlation with PHGDH expression in TCGA-BLCA database. PHGDH depletion disturbs the reactive oxygen species homeostasis and attenuates the xenograft growth in the mouse model. Further, we demonstrate WT p53 inhibits PHGDH expression by recruiting SIRT1 to the PHGDH promoter. Interestingly, the DNA binding motifs of YY1 and p53 in the PHGDH promoter are partially overlapped which causes competition between the two transcription factors. This competitive regulation of PHGDH is functionally linked to the xenograft growth in mice. CONCLUSION YY1 drives PHGDH expression in the context of mutant p53 and promotes bladder tumorigenesis, which preliminarily explains the relationship between high-frequency mutations of p53 and dysfunctional serine metabolism in bladder cancer.
Collapse
Affiliation(s)
- Tiezhu Shi
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Zhihao Yuan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Yanying He
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
| | - Dongliang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Siteng Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Xiongjun Wang
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
| | - Linli Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, 200080, Shanghai, China.
| | - Jialiang Shao
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China.
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China.
| |
Collapse
|
128
|
Terry S, Gommet C, Kerangueven AC, Leguet M, Thévenin V, Berthelot M, Begoud L, Windenberger F, Lainee P. Activity in Group-Housed Home Cages of Mice as a Novel Preclinical Biomarker in Oncology Studies. Cancers (Basel) 2023; 15:4798. [PMID: 37835492 PMCID: PMC10571829 DOI: 10.3390/cancers15194798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Improving experimental conditions in preclinical animal research is a major challenge, both scientifically and ethically. Automated digital ventilated cages (DVC®) offer the advantage of continuous monitoring of animal activity in their home-cage. The potential utility of this technology remains understudied and deserves investigation in the field of oncology. METHODS Using the DVC® platform, we sought to determine if the continuous assessment of locomotor activity of mice in their home cages can serve as useful digital readout in the monitoring of animals treated with the reference oncology compounds cisplatin and cyclophosphamide. SCID mice of 14 weeks of age were housed in DVC® cages in groups of four and followed with standard and digital examination before and after treatment over a 17-day total period. RESULTS DVC® detected statistically significant effects of cisplatin on the activity of mice in the short and long term, as well as trends for cyclophosphamide. The activity differences between the vehicle- and chemotherapy-treated groups were especially marked during the nighttime, a period when animals are most active and staff are generally not available for regular checks. Standard clinical parameters, such as body weight change and clinical assessment during the day, provided additional and complementary information. CONCLUSION The DVC® technology enabled the home cage monitoring of mice and non-invasive detection of animal activity disturbances. It can easily be integrated into a multimodal monitoring approach to better capture the different effects of oncology drugs on anti-tumor efficacy, toxicity, and safety and improve translation to clinical studies.
Collapse
Affiliation(s)
| | - Céline Gommet
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Anne-Cécile Kerangueven
- Biostatistics & Programming, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (A.-C.K.); (F.W.)
| | - Mickaël Leguet
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Vincent Thévenin
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Mickaël Berthelot
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Laurent Begoud
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Fanny Windenberger
- Biostatistics & Programming, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (A.-C.K.); (F.W.)
| | - Pierre Lainee
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| |
Collapse
|
129
|
Guo L, Li P, Li J, Gong Y, Li X, Liu Y, Yu K, Liu Z. Half-Sandwich Iridium(III), Rhodium(III), and Ruthenium(II) Complexes Chelating Hybrid sp 2-N/sp 3-N Donor Ligands to Achieve Improved Anticancer Selectivity. Inorg Chem 2023; 62:15118-15137. [PMID: 37671819 DOI: 10.1021/acs.inorgchem.3c02118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
The biological efficacy of half-sandwich platinum group organometallic complexes of the formula [(η5-Cpx)/(η6-arene)M(XY)Cl]0/+ (XY = bidentate ligands; Cpx = functionalized cyclopentadienyl; M = Ir, Rh, Ru, Os) has received considerable attention due to the significance of the metal center, chelating ligand, and Cpx/arene moieties in defining their anticancer potency and selectivity. With a facile access to the BIAN-derived imine-amine ligands using alkylaluminum as the reductant, we herein described the preparation and characterization of 16 half-sandwich Ir(III), Rh(III), and Ru(II) complexes chelating the hybrid sp2-N/sp3-N donor ligand. A nonplanar five-member metallacycle was confirmed by X-ray single-crystal structures of Ir1-Ir3, Ir7, Rh1, Ru1, and Ru4. The attempt to prepare imine-amido complexes using a base as the deprotonating agent led to the mixture of imine-amine complexes, within which the leaving group Cl- was displaced, and 16-electron imine-amido complexes without Cl-. The half-sandwich imine-amine complexes in this system underwent rapid hydrolysis in aqueous solution, exhibited weak photoluminescence, and showed the ability of binding to CT-DNA and BSA. The cytotoxicity of all imine-amine complexes against A549 lung cancer cell lines, HeLa cervical cancer cell lines, and 4T1 mouse breast cancer cells was determined by an MTT assay. The IC50 values of these complexes were in a range of 5.71-67.28 μM. Notably, most of these complexes displayed improved selectivity toward A549 cancer cells versus noncancerous BEAS-2B cells in comparison with the corresponding α-diimine complexes chelating the sp2-N/sp2-N donor ligand, which have been shown no selectivity in our previous report. The anticancer selectivity of these complexes appeared to be related to the redox-based mechanism including the catalytic oxidation of NADH to NAD+, reactive oxygen species (ROS) generation, and depolarization of the mitochondrial membrane. Further, inducing apoptosis of these complexes in A549 cancer cells and BEAS-2B normal cells also correlated with their anticancer selectivity, indicating the apoptosis mode of cell death in this system. In addition, these complexes could enter A549 cells via energy-dependent pathway and were able to impede the in vitro migration of A549 cells.
Collapse
Affiliation(s)
- Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Jiaxing Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Xiaoyuan Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yiming Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Kaijian Yu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| |
Collapse
|
130
|
Li H, Cheng S, Zhai J, Lei K, Zhou P, Cai K, Li J. Platinum based theranostics nanoplatforms for antitumor applications. J Mater Chem B 2023; 11:8387-8403. [PMID: 37581251 DOI: 10.1039/d3tb01035j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Platinum (Pt) based nanoplatforms are biocompatible nanoagents with photothermal antitumor performance, while exhibiting excellent radiotherapy sensitization properties. Pt-nanoplatforms have extensive research prospects in the realm of cancer treatment due to their highly selective and minimally invasive treatment mode with low damage, and integrated diagnosis and treatment with image monitoring and collaborative drug delivery. Platinum based anticancer chemotherapeutic drugs can kill tumor cells by damaging DNA through chemotherapy. Meanwhile, Pt-nanoplatforms also have good electrocatalytic activity, which can mediate novel electrodynamic therapy. Simultaneously, Pt(II) based compounds also have potential as photosensitizers in photodynamic therapy for malignant tumors. Pt-nanoplatforms can also modulate the immunosuppressive environment and synergistically ablate tumor cells in combination with immune checkpoint inhibitors. This article reviews the research progress of platinum based nanoplatforms in new technologies for cancer therapy, starting from widely representative examples of platinum based nanoplatforms in chemotherapy, electrodynamic therapy, photodynamic therapy, photothermal therapy, and immunotherapy. Finally, multimodal imaging techniques of platinum based nanoplatforms for biomedical diagnosis are briefly discussed.
Collapse
Affiliation(s)
- Heying Li
- College of Medical Technology and Engineering, The 1st Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China.
| | - Shaowen Cheng
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Jingming Zhai
- College of Medical Technology and Engineering, The 1st Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China.
| | - Kun Lei
- College of Medical Technology and Engineering, The 1st Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China.
| | - Ping Zhou
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Jinghua Li
- College of Medical Technology and Engineering, The 1st Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China.
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
131
|
Thilak
Babu L, Paira P. CuAAC "Click"-Derived Luminescent 2-(2-(4-(4-(Pyridin-2-yl)-1 H-1,2,3-triazol-1-yl)butoxy)phenyl)benzo[ d]thiazole-Based Ru(II)/Ir(III)/Re(I) Complexes as Anticancer Agents. ACS OMEGA 2023; 8:32382-32395. [PMID: 37720792 PMCID: PMC10500652 DOI: 10.1021/acsomega.3c01639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/13/2023] [Indexed: 09/19/2023]
Abstract
To enhance the cytoselective behavior of the complexes, we intended to develop a CuAAC "click"-derived synthetic protocol for the preparation of 2-(2-(4-(4-(pyridin-2-yl)-1H-1,2,3-triazol-1-yl)butoxy)phenyl)benzo[d]thiazole-based Ru(II)/Ir(III)/Re(I) complexes, and their cytotoxicity against three different cancer cell lines (MCF-7, HeLa, and U87MG) in consort with one normal cell line (HEK-293) was evaluated. In our detailed investigations, the significant cytotoxic nature of the Ru(II) complex 7a compared to Ir(III) and Re(I) complexes (7b and 7c, respectively) was observed. Complex 7a was capable of MCF-7 cell apoptosis via the inhibition of both S- and G2/M-phase cell cycle arrest in association with a substantial quantity of ROS production and DNA intercalation.
Collapse
Affiliation(s)
- Lavanya Thilak
Babu
- Department of Chemistry,
School of Advanced Sciences, Vellore Institute
of Technology, Vellore 632014, Tamil Nadu, India
| | - Priyankar Paira
- Department of Chemistry,
School of Advanced Sciences, Vellore Institute
of Technology, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
132
|
Sun L, Ji WX, Li Y, Li ZL, Duan CC, Xia BR, Xiao L. The PAPSS1 gene is a modulator of response to cisplatin by regulating estrogen receptor alpha signaling activity in ovarian cancer cells. J Ovarian Res 2023; 16:187. [PMID: 37684671 PMCID: PMC10486135 DOI: 10.1186/s13048-023-01262-7] [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: 03/01/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Cancer cells may develop resistance to cisplatin by various mechanisms. Yet, the exact mechanism of cisplatin in ovarian cancer remains unclear. Recent studies have shown that 3'-phospoadenosine 5'-phosphosulfate synthase 1 (PAPSS1) inhibition combined with low-dose cisplatin increases DNA damage. The aim of this study was to determine the value of targeting PAPSS1 as a cisplatin modulator in epithelial ovarian cancer (EOC). RESULTS Increased expression of PAPSS1 was observed in both EOC cells and tissues. Also, its higher nuclear expression was distinctly associated with FIGO (The International Federation of Gynecology and Obstetrics) stage, histological subtype, metastasis, and recurrence. Down-regulation of the PAPSS1 gene increased the cisplatin sensitivity of EOC in vitro and in vivo. Expression of PAPSS1 was negatively correlated with estrogen receptor α (ERα) in EOC. Also, low nuclear PAPSS1 and high nuclear ERα expression in EOC were associated with longer overall survival and progression-free survival in all ovarian cancer and ovarian cancer patients who received platinum-based chemotherapy. PAPSS1 silencing increased the activity of ERα-signaling in EOC cells, thus sensitizing tumors to cisplatin. CONCLUSIONS These findings characterize a novel interplay between PAPSS1-mediated sulfation and ERα-signaling in EOC cisplatin resistance. PAPSS1 may be exploited as a cisplatin-sensitizing therapeutic target.
Collapse
Affiliation(s)
- Lei Sun
- Department of Obstetrics & Gynecology, the First Affiliated Hospital, Anhui Medical University, Hefei, 230020, Anhui, P. R. China
| | - Wei-Xue Ji
- Department of Obstetrics & Gynecology, the First Affiliated Hospital, Anhui Medical University, Hefei, 230020, Anhui, P. R. China
| | - Yan Li
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, Hubei, P. R. China
| | - Ze-Lian Li
- Department of Obstetrics & Gynecology, the First Affiliated Hospital, Anhui Medical University, Hefei, 230020, Anhui, P. R. China
| | - Can-Can Duan
- Department of Obstetrics & Gynecology, the First Affiliated Hospital, Anhui Medical University, Hefei, 230020, Anhui, P. R. China
| | - Bai-Rong Xia
- Department of Gynecology Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, P. R. China.
| | - Lan Xiao
- Department of Obstetrics & Gynecology, the First Affiliated Hospital, Anhui Medical University, Hefei, 230020, Anhui, P. R. China.
| |
Collapse
|
133
|
Wu M, Xue L, Chen Y, Tang W, Guo Y, Xiong J, Chen D, Zhu Q, Fu F, Wang S. Inhibition of checkpoint kinase prevents human oocyte apoptosis induced by chemotherapy and allows enhanced tumour chemotherapeutic efficacy. Hum Reprod 2023; 38:1769-1783. [PMID: 37451671 DOI: 10.1093/humrep/dead145] [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: 12/30/2022] [Revised: 05/26/2023] [Indexed: 07/18/2023] Open
Abstract
STUDY QUESTION Could inhibition of the checkpoint kinase (CHEK) pathway protect human oocytes and even enhance the anti-tumour effects, during chemotherapy? SUMMARY ANSWER CHEK inhibitors prevented apoptosis of human oocytes induced by chemotherapy and even enhanced the anti-tumour effects. WHAT IS KNOWN ALREADY CHEK inhibitors showed ovarian protective effects in mice during chemotherapy, while their role in human oocytes is unclear. STUDY DESIGN, SIZE, DURATION This experimental study evaluated the ovarian reserve of young patients (120 patients) with cancer, exposed or not exposed to taxane and platinum (TP)-combined chemotherapy. Single RNA-sequencing analysis of human primordial oocytes from 10 patients was performed to explore the mechanism of oocyte apoptosis induced by TP chemotherapy. The damaging effects of paclitaxel (PTX) and cisplatin on human oocytes were also evaluated by culturing human ovaries in vitro. A new mouse model that combines human ovarian xenotransplantation and patient-derived tumour xenografts was developed to explore adjuvant therapies for ovarian protection. The mice were randomly allocated to four groups (10 mice for each group): control, cisplatin, cisplatin + CK1 (CHEK1 inhibitor, SCH 900776), and cisplatin + CK2 (CHEK2 inhibitor, BML277). PARTICIPANTS/MATERIALS, SETTING, METHODS In the prospective cohort study, human ovarian follicles were counted and serum AMH levels were evaluated. RNA-sequencing analysis was conducted, and staining for follicular damage (phosphorylated H2AX histone; γH2AX), terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labelling (TUNEL) assays and assessments of apoptotic biomarkers (western blot and immunofluorescence) were conducted in human ovaries. After the treatments, histological analysis was performed on human ovarian samples to investigate follicular populations, and oocyte damage was measured by γH2AX staining, BAX staining, and TUNEL assays. At the same time, the tumours were evaluated for volume, weight, and apoptosis levels. MAIN RESULTS AND THE ROLE OF CHANCE Patients who received TP chemotherapy showed decreased ovarian reserves. Single RNA-sequencing analysis of human primordial oocytes indicated that TP chemotherapy induced apoptosis of human primordial oocytes by causing CHEK-mediated TAp63α phosphorylation. In vitro culture of human ovaries showed greater damaging effects on oocytes after cisplatin treatment compared with that after PTX treatment. Using the new animal model, CHEK1/2 inhibitors prevented the apoptosis of human oocytes induced by cisplatin and even enhanced its anti-tumour effects. This protective effect appeared to be mediated by inhibiting DNA damage via the CHEK-TAp63α pathway and by generation of anti-apoptotic signals in the oocytes. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This was a preclinical study performed with human ovarian samples, and clinical research is required for validation. WIDER IMPLICATIONS OF THE FINDINGS These findings highlight the therapeutic potential of CHEK1/2 inhibitors as a complementary strategy for preserving fertility in female cancer patients. STUDY FUNDING/COMPETING INTEREST(S) This work was financially supported by the National Natural Science Foundation of China (nos. 82001514 and 81902669) and the Fundamental Research Funds for the Central Universities (2021yjsCXCY087). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Qingqing Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| |
Collapse
|
134
|
Yang Y, Wang P, Ji Z, Xu X, Zhang H, Wang Y. Polysaccharide‑platinum complexes for cancer theranostics. Carbohydr Polym 2023; 315:120997. [PMID: 37230639 DOI: 10.1016/j.carbpol.2023.120997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 05/27/2023]
Abstract
Platinum anticancer drugs have been explored and developed in recent years to reduce systematic toxicities and resist drug resistance. Polysaccharides derived from nature have abundant structures as well as pharmacological activities. The review provides insights on the design, synthesis, characterization and associating therapeutic application of platinum complexes with polysaccharides that are classified by electronic charge. The complexes give birth to multifunctional properties with enhanced drug accumulation, improved tumor selectivity and achieved synergistic antitumor effect in cancer therapy. Several techniques developing polysaccharides-based carriers newly are also discussed. Moreover, the lasted immunoregulatory activities of innate immune reactions triggered by polysaccharides are summarized. Finally, we discuss the current shortcomings and outline potential strategies for improving platinum-based personalized cancer treatment. Using platinum-polysaccharides complexes for improving the immunotherapy efficiency represents a promising framework in future.
Collapse
Affiliation(s)
- Yunxia Yang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Province Engineering Research Center of Agricultural Breeding Pollution Control and Resource, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng 224007, China.
| | - Pengge Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Zengrui Ji
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China.
| | - Hongmei Zhang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Yanqing Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China.
| |
Collapse
|
135
|
Hannon Barroeta P, O'Sullivan MJ, Zisterer DM. The role of the Nrf2/GSH antioxidant system in cisplatin resistance in malignant rhabdoid tumours. J Cancer Res Clin Oncol 2023; 149:8379-8391. [PMID: 37079050 PMCID: PMC10374708 DOI: 10.1007/s00432-023-04734-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
PURPOSE Malignant rhabdoid tumour (MRT) is a rare and aggressive childhood malignancy that occurs in the kidneys or central nervous system and is associated with very poor prognosis. Chemoresistance is a major issue in the treatment of this malignancy leading to an urgent need for a greater understanding of its underlying mechanisms in MRT and novel treatment strategies for MRT patients. The balance between oxidative stress mediated by reactive oxygen species (ROS) and the antioxidant system has become a subject of interest in cancer therapy research. Studies have implicated key players of the antioxidant system in chemotherapeutic including the well-known antioxidant glutathione (GSH) and the transcription factor nuclear erythroid-related factor-2 (Nrf2). METHODS: This study evaluated the role of these components in the response of MRT cells to treatment with the commonly used chemotherapeutic agent, cisplatin. RESULTS This study characterised the basal levels of GSH, ROS and Nrf2 in a panel of MRT cell lines and found a correlation between the expression profile of the antioxidant defence system and cisplatin sensitivity. Results showed that treatment with ROS scavenger N-acetylcysteine (NAC) protected cells from cisplatin-induced ROS and apoptosis. Interestingly, depleting GSH levels with the inhibitor buthionine sulphoximine (BSO) enhanced cisplatin-induced ROS and sensitised cells to cisplatin. Lastly, targeting Nrf2 with the small molecule inhibitor ML385 or by siRNA diminished GSH levels, enhanced ROS and sensitised resistant MRT cells to cisplatin. CONCLUSIONS These results suggest that targeting the Nrf2/GSH antioxidant system may present a novel therapeutic strategy to combat chemoresistance in rhabdoid tumours.
Collapse
Affiliation(s)
- Patricia Hannon Barroeta
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse St, Dublin, D02 R590, Ireland.
| | - Maureen J O'Sullivan
- The National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse St, Dublin, D02 R590, Ireland
| |
Collapse
|
136
|
Hata M, Ueno J, Hitomi Y, Kodera M. Roles of DNA Target in Cancer Cell-Selective Cytotoxicity by Dicopper Complexes with DNA Target/Ligand Conjugates. ACS OMEGA 2023; 8:28690-28701. [PMID: 37576680 PMCID: PMC10413468 DOI: 10.1021/acsomega.3c03387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
The DNA target/ligand conjugates (HLX, X = Pn and Mn, n = 1-3) were synthesized where various lengths of -CONH(CH2CH2O)nCH2CH2NHCO- linkers with a 9-phenanthrenyl (P) or methyl (M) terminal as DNA targets replace the methyl group of 2,6-di(amide-tether cyclen)-p-cresol ligand (HL). DNA binding, DNA cleavage, cellular uptake, and cytotoxicity of [Cu2(μ-OH)(LX)](ClO4)2 (1X) are examined and compared with those of [Cu2(μ-OH)(L)](ClO4)2 (1) to clarify roles of DNA targets. Upon reaction of 1X with H2O2, μ-1,1-O2H complexes are formed for DNA cleavage. 1P1, 1P2, and 1P3 are 22-, 11-, 3-fold more active for conversion of Form II to III in the cleavage of supercoiled plasmid DNA with H2O2 than 1, where the short P-linker may fix a dicopper moiety within a small number of base pairs to facilitate DNA double-strand breaks (dsb). This enhances the proapoptotic activity of 1P1, 1P2, and 1P3, which are 30-, 12-, and 9.9-fold cytotoxic against HeLa cells than 1. DNA dsb and cytotoxicity are 44% correlated in 1P1-3 but 5% in 1M1-3, suggesting specific DNA binding of P-linkers and nonspecific binding of M-linkers in biological cells. 1P1-3 exert cancer cell-selective cytotoxicity against lung and pancreas cancer and normal cells where the short P-linker enhances the selectivity, but 1M1-3 do not. Intracellular visualization, apoptosis assay, and caspase activity assay clarify mitochondrial apoptosis caused by 1P1-3. The highest cancer cell selectivity of 1P1 may be enabled by the short P-linker promoting dsb of mitochondrial DNA with H2O2 increased by mitochondrial dysfunction in cancer cells.
Collapse
Affiliation(s)
- Machi Hata
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Jin Ueno
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Yutaka Hitomi
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Masahito Kodera
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| |
Collapse
|
137
|
Wang J, Liu Q, Zhao Y, Fu J, Su J. Tumor Cells Transmit Drug Resistance via Cisplatin-Induced Extracellular Vesicles. Int J Mol Sci 2023; 24:12347. [PMID: 37569723 PMCID: PMC10418773 DOI: 10.3390/ijms241512347] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Cisplatin is a first-line clinical agent used for treating solid tumors. Cisplatin damages the DNA of tumor cells and induces the production of high levels of reactive oxygen species to achieve tumor killing. Tumor cells have evolved several ways to tolerate this damage. Extracellular vesicles (EVs) are an important mode of information transfer in tumor cells. EVs can be substantially activated under cisplatin treatment and mediate different responses of tumor cells under cisplatin treatment depending on their different cargoes. However, the mechanism of action of tumor-cell-derived EVs under cisplatin treatment and their potential cargoes are still unclear. This review considers recent advances in cisplatin-induced release of EVs from tumor cells, with the expectation of providing a new understanding of the mechanisms of cisplatin treatment and drug resistance, as well as strategies for the combined use of cisplatin and other drugs.
Collapse
Affiliation(s)
| | | | | | | | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China; (J.W.); (Q.L.); (Y.Z.); (J.F.)
| |
Collapse
|
138
|
Balikci Cicek I, Colak C, Yologlu S, Kucukakcali Z, Ozhan O, Taslidere E, Danis N, Koc A, Parlakpinar H, Akbulut S. Nephrotoxicity Development of a Clinical Decision Support System Based on Tree-Based Machine Learning Methods to Detect Diagnostic Biomarkers from Genomic Data in Methotrexate-Induced Rats. APPLIED SCIENCES 2023; 13:8870. [DOI: 10.3390/app13158870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
Background: The purpose of this study was to carry out the bioinformatic analysis of lncRNA data obtained from the genomic analysis of kidney tissue samples taken from rats with nephrotoxicity induced by methotrexate (MTX) and from rats without pathology and modeling with the tree-based machine learning method. Another aim of the study was to identify potential biomarkers for the diagnosis of nephrotoxicity and to provide a better understanding of the nephrotoxicity formation process by providing the interpretability of the model with explainable artificial intelligence methods as a result of the modeling. Methods: To identify potential indicators of drug-induced nephrotoxicity, 20 female Wistar albino rats were separated into two groups: MTX-treated and the control. Kidney tissue samples were collected from the rats, and genomic, histological, and immunohistochemical analyses were performed. The dataset obtained as a result of genomic analysis was modeled with random forest (RF), a tree-based method. Modeling results were evaluated with sensitivity (Se), specificity (Sp), balanced accuracy (B-Acc), negative predictive value (Npv), accuracy (Acc), positive predictive value (Ppv), and F1-score performance metrics. The local interpretable model-agnostic annotations (LIME) method was used to determine the lncRNAs that could be biomarkers for nephrotoxicity by providing the interpretability of the RF model. Results: The outcomes of the histological and immunohistochemical analyses conducted in the study support the conclusion that MTX use caused kidney injury. According to the results of the bioinformatics analysis, 52 lncRNAs showed different expressions in the groups. As a result of modeling with RF for lncRNAs selected with Boruta variable selection, the B-Acc, Acc, Sp, Se, Npv, Ppv, and F1-score were 88.9%, 90%, 90.9%, 88.9%, 90.9%, 88.9%, and 88.9%, respectively. lncRNAs with id rnaXR_591534.3 rnaXR_005503408.1, rnaXR_005495645.1, rnaXR_001839007.2, rnaXR_005492056.1, and rna_XR_005492522.1. The lncRNAs with the highest variable importance values produced from RF modeling can be used as nephrotoxicity biomarker candidates. Furthermore, according to the LIME results, the high level of lncRNAs with id rnaXR_591534.3 and rnaXR_005503408.1 particularly increased the possibility of nephrotoxicity. Conclusions: With the possible biomarkers resulting from the analyses in this study, it can be ensured that the procedures for the diagnosis of drug-induced nephrotoxicity can be carried out easily, quickly, and effectively.
Collapse
Affiliation(s)
- Ipek Balikci Cicek
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Cemil Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Saim Yologlu
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Zeynep Kucukakcali
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Onural Ozhan
- Department of Pharmacology, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Elif Taslidere
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Nefsun Danis
- Department of Medical Biology and Genetics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Ahmet Koc
- Department of Medical Biology and Genetics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Pharmacology, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| | - Sami Akbulut
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
- Department of Surgery, Faculty of Medicine, Inonu University, 44280 Malatya, Turkey
| |
Collapse
|
139
|
Campos MC, Barbosa IR, Guedes GP, Echevarria A, Echevarria-Lima J, Chaves OA. Novel Zn(II)-complex with hybrid chalcone-thiosemicarbazone ligand: Synthesis, characterization, and inhibitory effect on HTLV-1-infected MT-2 leukemia cells. J Inorg Biochem 2023; 245:112239. [PMID: 37148641 DOI: 10.1016/j.jinorgbio.2023.112239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023]
Abstract
Chalcone and thiosemicarbazone have attracted attention due to their easy synthetic procedure and high success in the development of antiviral and antitumor, however, there are few biological data on the evaluation of chalcone-thiosemicarbazone hybrids and their complexation with metal ions. In this sense, the present work reports the synthesis and characterization of the hybrid (Z)-2-((E)-3-(4-chlorophenyl)-1-phenylallylidene)hydrazine-1-carbothioamide (CTCl) and its Zn(II)-complex (CTCl-Zn). The compounds were cell-based evaluated in terms of cytotoxicity against human T-cell lymphotropic virus type 1 (HTLV-1) infected leukemia cells (MT-2) and the experimental data were correlated with molecular docking calculations. The ligand and Zn(II)-complex were easily synthesized with a good yield - 57% and 79%, respectively. The dynamic of E/Z isomers with respect to the imine bond configuration of CTCl was evidenced by 1H NMR experiments in DMSO‑d6, while the X-ray diffraction of CTCl-Zn showed that Zn(II) ion is tetracoordinated to two ligands in a bidentate mode and the metal ion lies on an intermediate geometry between the see-saw and trigonal pyramid. The ligand and complex exhibited low toxicity and the Zn(II)-complex is more cytotoxic than the ligand, with the corresponding IC50 value of 30.01 and 47.06 μM. Both compounds had a pro-apoptotic effect without the release of reactive oxygen species (ROS) and they can interact with DNA via minor grooves driven by van der Waals forces.
Collapse
Affiliation(s)
- Maria Clara Campos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Igor Resendes Barbosa
- Instituto de Química, Departamento de Química Orgânica, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | | | - Aurea Echevarria
- Instituto de Química, Departamento de Química Orgânica, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil.
| | - Juliana Echevarria-Lima
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Otávio Augusto Chaves
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil; CQC-IMS, Departamento de Química, Universidade de Coimbra, Rua Larga s/n, Coimbra, Portugal.
| |
Collapse
|
140
|
Dorairaj DP, Haribabu J, Dharmasivam M, Malekshah RE, Mohamed Subarkhan MK, Echeverria C, Karvembu R. Ru(II)- p-Cymene Complexes of Furoylthiourea Ligands for Anticancer Applications against Breast Cancer Cells. Inorg Chem 2023; 62:11761-11774. [PMID: 37459067 DOI: 10.1021/acs.inorgchem.3c00757] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Half-sandwich Ru(II) complexes containing nitro-substituted furoylthiourea ligands, bearing the general formula [(η6-p-cymene)RuCl2(L)] (1-6) and [(η6-p-cymene)RuCl(L)(PPh3)]+ (7--12), have been synthesized and characterized. In contrast to the spectroscopic data which revealed monodentate coordination of the ligands to the Ru(II) ion via a "S" atom, single crystal X-ray structures revealed an unusual bidentate N, S coordination with the metal center forming a four-membered ring. Interaction studies by absorption, emission, and viscosity measurements revealed intercalation of the Ru(II) complexes with calf thymus (CT) DNA. The complexes showed good interactions with bovine serum albumin (BSA) as well. Further, their cytotoxicity was explored exclusively against breast cancer cells, namely, MCF-7, T47-D, and MDA-MB-231, wherein all of the complexes were found to display more pronounced activity than their ligand counterparts. Complexes 7-12 bearing triphenylphosphine displayed significant cytotoxicity, among which complex 12 showed IC50 values of 0.6 ± 0.9, 0.1 ± 0.8, and 0.1 ± 0.2 μM against MCF-7, T47-D, and MDA-MB-231 cell lines, respectively. The most active complexes were tested for their mode of cell death through staining assays, which confirmed apoptosis. The upregulation of apoptotic inducing and downregulation of apoptotic suppressing proteins as inferred from the western blot analysis also corroborated the apoptotic mode of cell death. The active complexes effectively generated reactive oxygen species (ROS) in MDA-MB-231 cells as analyzed from the 2',7'-dichlorofluorescein diacetate (DCFH-DA) staining. Finally, in vivo studies of the highly active complexes (6 and 12) were performed on the mice model. Histological analyses revealed that treatment with these complexes at high doses of up to 8 mg/kg did not induce any visible damage to the tested organs.
Collapse
Affiliation(s)
| | - Jebiti Haribabu
- Faculty of Medicine, University of Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
| | - Mahendiran Dharmasivam
- Department of Chemistry, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Rahime Eshaghi Malekshah
- Medical Biomaterial Research Centre (MBRC), Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Mohamed Kasim Mohamed Subarkhan
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310018, P. R. China
| | - Cesar Echeverria
- Faculty of Medicine, University of Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India
| |
Collapse
|
141
|
Kuo YY, Chen WT, Lin GB, Lu CH, Chao CY. Study on the effect of a triple cancer treatment of propolis, thermal cycling-hyperthermia, and low-intensity ultrasound on PANC-1 cells. Aging (Albany NY) 2023; 15:7496-7512. [PMID: 37506229 PMCID: PMC10457055 DOI: 10.18632/aging.204916] [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: 02/10/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023]
Abstract
To reduce side effects and enhance treatment efficacy, study on combination therapy for pancreatic cancer, a deadly cancer, has gained much attraction in recent years. In this study, we propose a novel triple treatment combining propolis and two physical stimuli-thermal cycling-hyperthermia (TC-HT) and low-intensity ultrasound (US). The study found that, after the triple treatment, the cell viability of a human cancer cell line PANC-1 decreased to a level 80% less than the control, without affecting the normal pancreatic cells. Another result was excessive accumulation of reactive oxygen species (ROS) after the triple treatment, leading to the amplification of apoptotic pathway through the MAPK family and mitochondrial dysfunction. This study, to the best of our knowledge, is the first attempt to combine TC-HT, US, and a natural compound in cancer treatment. The combination of TC-HT and US also promotes the anticancer effect of the heat-sensitive chemotherapy drug cisplatin on PANC-1 cells. It is expected that optimized parameters for different agents and different types of cancer will expand the methodology on oncological therapy in a safe manner.
Collapse
Affiliation(s)
- Yu-Yi Kuo
- Department of Physics, Lab for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Wei-Ting Chen
- Department of Physics, Lab for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Guan-Bo Lin
- Department of Physics, Lab for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Chueh-Hsuan Lu
- Department of Physics, Lab for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Chih-Yu Chao
- Department of Physics, Lab for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
142
|
Li J, Wang Z, Chen Z, Xue X, Lin K, Chen H, Pan L, Yuan Y, Ma Z. Silver complexes with substituted terpyridines as promising anticancer metallodrugs and their crystal structure, photoluminescence, and DNA interactions. Dalton Trans 2023; 52:9607-9621. [PMID: 37377144 DOI: 10.1039/d2dt03463h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Six silver hexafluoroantimonate complexes (1-6) with 4'-(4'-substituted-phenyl)-2,2':6',2''-terpyridine compounds bearing hydrogen (L1), methyl (L2), methylsulfonyl (L3), chloro (L4), bromo (L5) and iodo (L6) were prepared and characterized by 1H NMR, 13C NMR, IR, elemental analysis and single crystal X-ray diffraction. All the compounds exhibit interesting photoluminescence properties in the solid state and solution. In vitro data demonstrate that all of them show higher antiproliferative activities than cisplatin against three human carcinoma cell lines, A549, Eca-109 and MCF-7. Compound 3 exhibits the lowest IC50 value (2.298 μM) against A549 cell lines, which is 2.963 μM for 4 against Eca-109 and 1.830 μM for 1 against MCF-7. For silver halogen-substituted terpyridine compounds, their anticancer activities decrease following the sequence of -Cl, -Br, and -I substituents. The comparison results show that their anticancer activity is significantly higher than that of their free ligands. The DNA interaction was studied by fluorescence titration, circular dichroism spectroscopy and molecular modeling methods. Spectrophotometric results reveal that the compounds have strong affinity binding with DNA as intercalators and molecular docking studies indicate that the binding is contributed by the π-π stacking and hydrogen bonds. The DNA binding ability of the complexes has been correlated with their anticancer activities, which could potentially provide a new rationale for the future design of terpyridine-based metal complexes with antitumor potential.
Collapse
Affiliation(s)
- Jiahe Li
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, People's Republic of China.
- National Engineering Research Center for Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning 530007, People's Republic of China
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisbon 1049-001, Portugal
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, People's Republic of China
| | - Zhiyuan Wang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, People's Republic of China.
| | - Zhongting Chen
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, People's Republic of China
| | - Xingyong Xue
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, People's Republic of China.
| | - Kejuan Lin
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, People's Republic of China.
| | - Hailan Chen
- School of Animal Science and Technology, Guangxi University, Nanning 530004, People's Republic of China
| | - Lixia Pan
- National Engineering Research Center for Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning 530007, People's Republic of China
| | - Yulin Yuan
- Department of Laboratory Medicine, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, People's Republic of China.
| | - Zhen Ma
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, People's Republic of China.
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisbon 1049-001, Portugal
| |
Collapse
|
143
|
Chen H, Xie G, Luo Q, Yang Y, Hu S. Regulatory miRNAs, circRNAs and lncRNAs in cell cycle progression of breast cancer. Funct Integr Genomics 2023; 23:233. [PMID: 37432486 DOI: 10.1007/s10142-023-01130-z] [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: 05/14/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 07/12/2023]
Abstract
Breast cancer is a complex and heterogeneous disease that poses a significant public health concern worldwide, and it remains a major challenge despite advances in treatment options. One of the main properties of cancer cells is the increased proliferative activity that has lost regulation. Dysregulation of various positive and negative modulators in the cell cycle has been identified as one of the driving factors of breast cancer. In recent years, non-coding RNAs have garnered much attention in the regulation of cell cycle progression, with microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs) being of particular interest. MiRNAs are a class of highly conserved and regulatory small non-coding RNAs that play a crucial role in the modulation of various cellular and biological processes, including cell cycle regulation. CircRNAs are a novel form of non-coding RNAs that are highly stable and capable of modulating gene expression at posttranscriptional and transcriptional levels. LncRNAs have also attracted considerable attention because of their prominent roles in tumor development, including cell cycle progression. Emerging evidence suggests that miRNAs, circRNAs and lncRNAs play important roles in the regulation of cell cycle progression in breast cancer. Herein, we summarized the latest related literatures in breast cancer that emphasize the regulatory roles of miRNAs, circRNAs and lncRNAs in cell cycle progress of breast cancer. Further understanding of the precise roles and mechanisms of non-coding RNAs in breast cancer cell cycle regulation could lead to the development of new diagnostic and therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Huan Chen
- Department of Clinical Laboratory, Wuhan Institute of Technology Hospital, Wuhan Institute of Technology, Wuhan, China
| | - Guoping Xie
- Department of Clinical Laboratory, The Second Staff Hospital of Wuhan Iron and Steel (Group) Corporation, Wuhan, China
| | - Qunying Luo
- Department of Internal Medicine-Neurology, Huarun Wuhan Iron and Steel General Hospital, Wuhan, China
| | - Yisha Yang
- Luoyang Campus, Henan Vocational College of Agriculture, Luoyang, China
| | - Siheng Hu
- Department of Clinical Laboratory, Honggangcheng Street Community Health Service Center, Wuhan, China.
| |
Collapse
|
144
|
Rahman MM, Islam MR, Akash S, Hossain ME, Tumpa AA, Abrar Ishtiaque GM, Ahmed L, Rauf A, Khalil AA, Al Abdulmonem W, Simal-Gandara J. Pomegranate-specific natural compounds as onco-preventive and onco-therapeutic compounds: Comparison with conventional drugs acting on the same molecular mechanisms. Heliyon 2023; 9:e18090. [PMID: 37519687 PMCID: PMC10372646 DOI: 10.1016/j.heliyon.2023.e18090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/18/2023] [Accepted: 07/06/2023] [Indexed: 08/01/2023] Open
Abstract
Pomegranate, scientifically known as Punica granatum, has been a traditional medicinal remedy since ancient times. Research findings have shown that using pomegranate extracts can positively affect a variety of signaling pathways, including those involved in angiogenesis, inflammation, hyperproliferation, cellular transformation, the beginning stages of tumorigenesis, and lastly, a reduction in the final stages of metastasis and tumorigenesis. This is due to the fact that pomegranate extracts are rich in polyphenols, which are known to inhibit the activity of certain signaling pathways. In the United States, cancer is the second biggest cause of death after heart disease. The number of fatalities caused by cancer in the United States escalates yearly. Altering one's diet, getting involved in regular physical activity, and sustaining a healthy body weight are three easy steps an individual may follow to lower their cancer risk. Simply garnishing one's diet with vegetables and fruits has the potential to avert at least 20% of all cancer diagnoses and around 200,000 deaths caused by cancer each year. Vegetables, fruits, and other dietary constituents, such as minerals and phytochemicals, are currently being researched for their potential to prevent cancer. It is being done because they are safe, have minimal toxicity, possess antioxidant properties, and are universally accepted as dietary supplements. Ancient civilizations used the fruit of pomegranate (Punica granatum L.) to prevent and cure a number of diseases. The anti-tumorigenic, anti-inflammatory and anti-proliferative qualities of pomegranate have been shown in studies with the fruit, juice, extract, and oil of the pomegranate. Pomegranate has the capacity to affect several signaling pathways, which implies that it may have the potential to be employed not only as a chemopreventive agent but also as a chemotherapeutic drug. This article elaborates on some recent preclinical and clinical research which shows that pomegranate seems to have a role in the prevention and treatment of a number of cancers, including but not limited to breast, bladder, skin, prostate, colon, and lung cancer, among others.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md Emon Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | | | - Limon Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, 54000, Pakistan
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine Qassim University, Buraydah, Saudi Arabia
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, Faculty of Science, E32004 Ourense, Spain
| |
Collapse
|
145
|
Sami DH, Soliman AS, Khowailed AA, Alruhaimi RS, Hassanein EHM, Kamel EM, Mahmoud AM. The protective effect of 7-hydroxycoumarin against cisplatin-induced liver injury is mediated via attenuation of oxidative stress and inflammation and upregulation of Nrf2/HO-1 pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:80181-80191. [PMID: 37291353 DOI: 10.1007/s11356-023-27879-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/19/2023] [Indexed: 06/10/2023]
Abstract
Cisplatin (CIS) is an effective chemotherapy against different solid cancers. However, the adverse effects, including hepatotoxicity, limit its clinical use. 7-hydroxycoumarin (7-HC) possesses antioxidant and hepatoprotective activities, but its protective effect against CIS hepatotoxicity has not been investigated. This study evaluated the effect of 7-HC on liver injury, oxidative stress (OS), and inflammation provoked by CIS. Rats received 7-HC (25, 50, and 100 mg/kg) orally for 2 weeks followed by intraperitoneal injection of CIS (7 mg/kg) at day 15. CIS increased serum transaminases, alkaline phosphatase (ALP), and bilirubin and provoked tissue injury accompanied by elevated reactive oxygen species (ROS), malondialdehyde (MDA), and nitric oxide (NO). Liver nuclear factor (NF)-κB p65, inducible NO synthase (iNOS), pro-inflammatory cytokines, Bax, and caspase-3 were upregulated, and antioxidant defenses and Bcl-2 were decreased in CIS-treated rats, while 7-HC prevented liver injury and ameliorated OS, inflammatory and apoptosis markers. In addition, 7-HC enhanced nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase (HO)-1 in CIS-administered rats and in silico studies revealed its binding affinity toward HO-1. In conclusion, 7-HC protected against CIS hepatotoxicity by mitigating OS and inflammatory response and modulating Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Demiana H Sami
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Ayman S Soliman
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Akef A Khowailed
- Physiology Department, Faculty of Medicine, Cairo University, Giza, 12613, Egypt
| | - Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Assiut, 71524, Egypt
| | - Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Ayman M Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt.
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK.
| |
Collapse
|
146
|
Sato K, Miyauchi Y, Xu X, Kon R, Ikarashi N, Chiba Y, Hosoe T, Sakai H. Platinum-based anticancer drugs-induced downregulation of myosin heavy chain isoforms in skeletal muscle of mouse. J Pharmacol Sci 2023; 152:167-177. [PMID: 37257944 DOI: 10.1016/j.jphs.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023] Open
Abstract
Cisplatin, a platinum-based anticancer drug used frequently in cancer treatment, causes skeletal muscle atrophy. It was predicted that the proteolytic pathway is enhanced as the mechanism of this atrophy. Therefore, we investigated whether a platinum-based anticancer drug affects the expression of the major proteins of skeletal muscle, myosin heavy chain (MyHC). Mice were injected with cisplatin or oxaliplatin for four consecutive days. C2C12 myotubes were treated using cisplatin and oxaliplatin. Administration of platinum-based anticancer drug reduced quadriceps mass and muscle strength compared to the control group. Protein levels of all MyHC isoforms were reduced in the platinum-based anticancer drug groups. However, only Myh2 (MyHC-IIa) gene expression in skeletal muscle of mice treated with platinum-based anticancer drugs was found to be reduced. Treatment of C2C12 myotubes with platinum-based anticancer drugs reduced the protein levels of all MyHCs, and treatment with the proteasome inhibitor MG-132 restored this reduction. The expression of Mef2c, which was predicted to act upstream of Myh2, was reduced in the skeletal muscle of mice treated systemically with platinum-based anticancer drug. Degradation of skeletal muscle MyHCs by proteasomes may be a factor that plays an important role in muscle mass loss in platinum-based anticancer drug-induced muscle atrophy.
Collapse
Affiliation(s)
- Ken Sato
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Yu Miyauchi
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Xinran Xu
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Risako Kon
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Nobutomo Ikarashi
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Tomoo Hosoe
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan; Department of Bioregulatory Science, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Hiroyasu Sakai
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan.
| |
Collapse
|
147
|
Hamaya S, Oura K, Morishita A, Masaki T. Cisplatin in Liver Cancer Therapy. Int J Mol Sci 2023; 24:10858. [PMID: 37446035 DOI: 10.3390/ijms241310858] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor and is often diagnosed at an unresectable advanced stage. Systemic chemotherapy as well as transarterial chemoembolization (TACE) and hepatic arterial infusion chemotherapy (HAIC) are used to treat advanced HCC. TACE and HAIC have long been the standard of care for patients with unresectable HCC but are limited to the treatment of intrahepatic lesions. Systemic chemotherapy with doxorubicin or chemohormonal therapy with tamoxifen have also been considered, but neither has demonstrated survival benefits. In the treatment of unresectable advanced HCC, cisplatin is administered transhepatic arterially for local treatment. Subsequently, for cisplatin-refractory cases due to drug resistance, a shift to systemic therapy with a different mechanism of action is expected to produce new antitumor effects. Cisplatin is also used for the treatment of liver tumors other than HCC. This review summarizes the action and resistance mechanism of cisplatin and describes the treatment of the major hepatobiliary cancers for which cisplatin is used as an anticancer agent, with a focus on HCC.
Collapse
Affiliation(s)
- Sae Hamaya
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| | - Kyoko Oura
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| |
Collapse
|
148
|
Nana AB, Marimuthu T, Wamwangi D, Kondiah PPD, Choonara YE. Design and Evaluation of Composite Magnetic Iron-Platinum Nanowires for Targeted Cancer Nanomedicine. Biomedicines 2023; 11:1857. [PMID: 37509497 PMCID: PMC10377173 DOI: 10.3390/biomedicines11071857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
The purpose of the study was to synthesize and investigate the influence of geometrical structure, magnetism, and cytotoxic activity on core-shell platinum and iron-platinum (Fe/Pt) composite nanowires (NWs) for potential application in targeted chemotherapeutic approaches. The Pt-NWs and Fe/Pt composite NWs were synthesized via template electrodeposition, using anodic aluminum oxide (AAO) membranes. The Fe/Pt composite NWs (Method 1) was synthesized using two electrodeposition steps, allowing for greater control of the diameter of the NW core. The Fe/Pt composite NWs (Method 2) was synthesized by pulsed electrodeposition, using a single electrolytic bath. The properties of the synthesized NWs were assessed by high-resolution transmission electron microscopy (HRTEM), Raman spectroscopy, powder X-ray diffraction (XRD), inductively coupled plasma-optical emission spectrometry (ICP-OES), vibrating-sample magnetometry (VSM), and surface charge (zeta potential). A microscopy image analysis of the NWs revealed the presence of high-aspect-ratio NWs with nominal diameters of 40-50 nm and lengths of approximately <4 µm. The obtained powder XRD patterns confirmed the presence of a polycrystalline structure for both Pt NWs and Fe/Pt composite NWs. The potential utility of the synthesized NW nanoplatforms for anticancer activity was investigated using Tera 1 cells and Mouse 3T3 cells. Pt-NWs displayed modest cytotoxic activity against Tera 1 cells, while the Fe/Pt composite NWs (both Methods 1 and 2) demonstrated enhanced cytotoxic activity compared to the Pt-NWs on Tera 1 cells. The Fe/Pt composite NWs (Method 1) displayed ferromagnetic behavior and enhanced cytotoxic activity compared to Pt-NWs on Tera 1 cells, thus providing a sound basis for future magnetically targeted chemotherapeutic applications.
Collapse
Affiliation(s)
- Abu Bakr Nana
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Thashree Marimuthu
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Daniel Wamwangi
- School of Physics, Materials Physics Research Institute, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa
| | - Pierre P D Kondiah
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
149
|
Gonçalves YG, Becceneri AB, Graminha AE, Miranda VM, Rios RR, Rinaldi-Neto F, Costa MS, Gonçalves ACR, Deflon VM, Yoneyama KAG, Maia PIS, Franca EF, Cominetti MR, Silva RS, Von Poelhsitz G. New ruthenium(II) complexes with cyclic thio- and semicarbazone: evaluation of cytotoxicity and effects on cell migration and apoptosis of lung cancer cells. Dalton Trans 2023. [PMID: 37377063 DOI: 10.1039/d3dt00750b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
We describe the synthesis, physicochemical characterization, and in vitro antitumor assays of four novel analogous ruthenium(II) complexes with general formula cis-[RuII(N-L)(P-P)2]PF6, where P-P = bis(diphenylphosphine)methane (dppm, in complexes 1 and 2) or bis(diphenylphosphine)ethane (dppe, in complexes 3 and 4) and N-L = 5,6-diphenyl-4,5-dihydro-2H-[1,2,4]triazine-3-thione (Btsc, in complexes 1 and 3) or 5,6-diphenyltriazine-3-one (Bsc, in complexes 2 and 4). The data were consistent with cis arrangement of the biphosphine ligands. For the Btsc and Bsc ligands, the data pointed to monoanionic bidentate coordination to ruthenium(II) through N,S and N,O, respectively. Single-crystal X-ray diffraction showed that complex 1 crystallized in the monoclinic system, space group P21/c. Determination of the cytotoxicity profiles of complexes 1-4 gave SI values ranging from 1.19 to 3.50 against the human lung adenocarcinoma cell line A549 and the non-tumor lung cell line MRC-5. Although the molecular docking studies suggested that the interaction between DNA and complex 4 was energetically favorable, the experimental results showed that they interacted weakly. Overall, our results demonstrated that these novel ruthenium(II) complexes have interesting in vitro antitumor potential and this study may contribute to further studies in medicinal inorganic chemistry.
Collapse
Affiliation(s)
- Yasmim G Gonçalves
- Chemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil.
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Amanda B Becceneri
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Angelica E Graminha
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
- Gerontology Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Victor M Miranda
- Grupo de Química Inorgânica Estrutural e Biológica, Chemistry Institute of São Carlos, Universidade de São Paulo, USP - São Carlos, São Carlos, SP, Brazil
| | - Rafaella R Rios
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francisco Rinaldi-Neto
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mônica S Costa
- Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Ana C R Gonçalves
- Exacts, Natural Sciences, and Education Institute, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Victor M Deflon
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kelly A G Yoneyama
- Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Pedro I S Maia
- Exacts, Natural Sciences, and Education Institute, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Eduardo F Franca
- Laboratório de Cristalografia e Química Computacional, Chemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Márcia R Cominetti
- Gerontology Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Roberto S Silva
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
150
|
Zakaria NH, Saad N, Che Abdullah CA, Mohd Esa N. The Antiproliferative Effect of Chloroform Fraction of Eleutherine bulbosa (Mill.) Urb. on 2D- and 3D-Human Lung Cancer Cells (A549) Model. Pharmaceuticals (Basel) 2023; 16:936. [PMID: 37513848 PMCID: PMC10384492 DOI: 10.3390/ph16070936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Since lung cancer is the leading cause of cancer-related death worldwide, research is being conducted to discover anticancer agents as its treatment. Eleutherine bulbosa, a Dayak folklore medicine, exhibited anticancer effects against several cancer cells; however, its anticancer potency against lung cancer cells has not been explored yet. This study aims to determine the anticancer potency of E. bulbosa bulbs against lung cancer cells (A549) using 2D and 3D culture models, as well as determine its active compounds using gas chromatography-mass spectrometry (GC-MS) analysis. Three fractions of E. bulbosa bulbs, namely chloroform, n-hexane, and ethyl acetate, were tested for cytotoxicity using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium-bromide (MTT) and CellTiter-Glo. The antiproliferative effects of the most cytotoxic fraction against the 2D culture model were determined by a clonogenic survival assay and propidium iodide/Hoechst 33342 double staining, whereas the effects against the 3D culture model were determined by microscopy, flow cytometry, and gene expression analysis. The chloroform fraction is the most cytotoxic against A549 cells than other fractions, and it inhibited colony formation and induced apoptosis of A549 cells. The chloroform fraction also inhibited the growth of the A549 spheroid by suppressing the spheroid size, inducing apoptosis, reducing the proportion of CD44 lung cancer stem cells, causing arrest at the S phase of the cell cycle, and suppressing the expression of the SOX2 and MYC genes. Furthermore, the GC-MS analysis detected 20 active compounds in the chloroform fraction, including the major compounds of eleutherine and isoeleutherine. In conclusion, the chloroform fraction of E. bulbosa bulbs exhibit its antiproliferative effect on 2D and 3D culture models of A549 cells, suggesting it could be a lung cancer chemopreventive agent.
Collapse
Affiliation(s)
- Nur Hannan Zakaria
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Norazalina Saad
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Che Azurahanim Che Abdullah
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Department of Physics, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Materials Synthesis and Characterization Laboratory (MSCL), Institute of Nanoscience and Nanotechnology (ION2), Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Norhaizan Mohd Esa
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|