1
|
Li Y, Zhou T, Liu Z, Zhu X, Wu Q, Meng C, Deng Q. Protective effect of antidiabetic drugs against male infertility: evidence from Mendelian randomization. Diabetol Metab Syndr 2025; 17:140. [PMID: 40296064 PMCID: PMC12036310 DOI: 10.1186/s13098-025-01700-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The global prevalence of diabetes has been steadily increasing, with a growing number of younger individuals being affected. Over recent decades, various antidiabetic drugs have been repurposed for treating conditions beyond diabetes. However, the effects of antidiabetic drugs on male infertility (MIF) remain inadequately elucidated. This Mendelian randomization (MR) study aims to clarify the potential impact of antidiabetic drugs on the risk of MIF. METHOD We designed a comprehensive analytical workflow involving two-sample MR and summary-based MR (SMR) to assess the causal relationship between antidiabetic drug targets and MIF. First, instrumental variables were obtained based on HbA1c levels and gene expression levels. Then, MR analysis was performed after selecting positive target genes from four blood glucose level and type 2 diabetes (T2DM) datasets. Finally, we applied SMR analysis to validate and expand upon the previous conclusions. Additionally, sensitivity analyses were conducted to evaluate the robustness of the results. RESULTS Seven drug targets associated with five antidiabetic drugs were identified as significantly related to MIF. In the two-sample MR, the following drugs were found to reduce MIF risk through their respective significant targets: metformin (GPD1: IVW OR 0.007, 95% CI 0.000-0.204, P = 0.004), SGLT2 inhibitors (SGLT2i) (SLC5A1: IVW OR 0.048, 95% CI 0.004-0.585, P = 0.017), insulin and its analogs (IGF1R: IVW OR 0.773, 95% CI 0.648-0.922, P = 0.004), and sulfonylureas (TRPM4: IVW OR 0.869, 95% CI 0.766-0.985, P = 0.028; CTPA1: IVW OR 0.838, 95% CI 0.741-0.947, P = 0.005). In SMR analysis, antidiabetic drugs targeting the genes CPE (P = 0.03, HEIDI = 0.970) and TRPM4 (P = 0.028, HEIDI = 0.746) were found to significantly reduce the risk of MIF. CONCLUSION Our study indicates that metformin, SGLT2i, insulin and its analogs, as well as sulfonylureas, may offer potential therapeutic benefits for MIF. Specifically, six antidiabetic drug target genes GPD1, SLC5A1, IGF1R, TRPM4, CPT1 A, and CPE may play a role in the progression of MIF. These findings have significant implications for the development of personalized precision therapies for MIF.
Collapse
Affiliation(s)
- Yuqi Li
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tao Zhou
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhiyu Liu
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xinyao Zhu
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qilong Wu
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunyang Meng
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Qingfu Deng
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
2
|
Massie PL, Garcia M, Decker A, Liu R, MazloumiBakhshayesh M, Kulkarni D, Justus MP, Gallardo J, Abrums A, Markle K, Pace C, Campen M, Clark RM. Essential and Non-Essential Metals and Metalloids and Their Role in Atherosclerosis. Cardiovasc Toxicol 2025:10.1007/s12012-025-09998-y. [PMID: 40251456 DOI: 10.1007/s12012-025-09998-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Abstract
Peripheral arterial disease (PAD) is becoming more prevalent in the aging developed world and can have significant functional impacts on patients. There is a recent recognition that environmental toxicants such as circulating metals and metalloids may contribute to the pathogenesis of atherosclerotic disease, but the mechanisms are complex. While the broad toxic biologic effects of metals in human systems have been extensively reviewed, the role of non-essential exposure and essential metal aberrancy in PAD specifically is less frequently discussed. This review of the literature describes current scientific knowledge regarding the individual roles several major metals and metalloids play in atherogenesis and highlights areas where a dearth of data exist. The roles of lead (Pb), arsenic (As), cadmium (Cd), iron (Fe), copper (Cu), selenium (Se) are included. Contemporary outcomes of therapeutic trials aimed at chelation therapy of circulating metals to impact cardiovascular outcomes are also discussed. This review highlights the supported notion of differential metal presence within peripheral plaques themselves, although distinguishing their roles within these plaques requires further illumination.
Collapse
Affiliation(s)
- Pierce L Massie
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Marcus Garcia
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Aerlin Decker
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Milad MazloumiBakhshayesh
- Department of Biomedical Engineering, School of Engineering, University of New Mexico, Albuquerque, USA
| | - Deepali Kulkarni
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Matthew P Justus
- Department of Biomedical Engineering, School of Engineering, University of New Mexico, Albuquerque, USA
| | - Jorge Gallardo
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Avalon Abrums
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Kristin Markle
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Carolyn Pace
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Matthew Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Ross M Clark
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA.
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, USA.
| |
Collapse
|
3
|
Ban T, Dong X, Ma Z, Jin J, Li J, Cui Y, Fu Y, Wang Y, Xue Y, Tong T, Zhang K, Han Y, Shen M, Zhao Y, Zhao L, Xiong L, Lv H, Liu Y, Huo R. Brg1 and RUNX1 synergy in regulating TRPM4 channel in mouse cardiomyocytes. Front Pharmacol 2024; 15:1494205. [PMID: 39726787 PMCID: PMC11669506 DOI: 10.3389/fphar.2024.1494205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/08/2024] [Indexed: 12/28/2024] Open
Abstract
Background Transient Receptor Potential Melastatin 4 (TRPM4), a non-selective cation channel, plays a critical role in cardiac conduction abnormalities. Brg1, an ATP-dependent chromatin remodeler, is essential for regulating gene expression in both heart development and disease. Our previous studies demonstrated Brg1 impacted on cardiac sodium/potassium channels and electrophysiological stability, its influence on TRPM4 expression and function remained unexplored. Methods We investigated the role of Brg1 in regulating TRPM4 expression and function through overexpression and knockdown experiments in mouse cardiomyocytes and TRPM4-overexpressing HEK293 cells by western blot, qPCR, immunofluorescence staining and patch clamp techniques. Cardiomyocytes were exposed to hypoxia for 12 h to mimic cardiac stress, and Brg1 inhibition was performed to assess its impact on TRPM4 under hypoxia. Bioinformatic analyses (STRING and JASPAR databases), Co-immunoprecipitation (Co-IP), dual luciferase reporter assays, and Chromatin Immunoprecipitation (ChIP) were employed to study the interaction between Brg1, RUNX1, and TRPM4 transcription regulation. Results Brg1 positively regulated TRPM4 expression in mouse cardiomyocytes and modulated TRPM4 current in TRPM4-overexpressing HEK293 cells. Brg1 inhibition markedly diminishes TRPM4's hyperexpression in cardiomyocytes exposed to hypoxia. Integrative analyses utilizing STRNG databases and Protein Data Bank unveiled a putative interaction between Brg1 and the transcription factor RUNX1, and we substantiated the interaction between Brg1 and RUNX1. Several binding sites of RUNX1 with the TRPM4 promoter region were predicted by the JASPAR database, and empirical validation substantiated Brg1 modulated TRPM4 promoter activity via RUNX1 engagement. ChIP confirmed that Brg1 interacted with RUNX1 forming a transcriptional complex that located in TRPM4 promoter. Conclusion Our study demonstrated that Brg1 and RUNX1 formed a transcriptional complex that modulated TRPM4 expression and function, especially under hypoxic conditions. These findings provided new insights into TRPM4 regulation and highlighted its potential as a therapeutic target for cardiac hypoxia-related disorders.
Collapse
Affiliation(s)
- Tao Ban
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianhui Dong
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Ziyue Ma
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Jing Jin
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Jing Li
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yunfeng Cui
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yuyang Fu
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yongzhen Wang
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yadong Xue
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Tingting Tong
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Kai Zhang
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yuxuan Han
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Meimei Shen
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yu Zhao
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Ling Zhao
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Lingzhao Xiong
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Hongzhao Lv
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Yang Liu
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| | - Rong Huo
- Harbin Medical University and Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin, China
| |
Collapse
|
4
|
Gao Z, Lv J, Tong TT, Zhang K, Han YX, Zhao Y, Shen MM, Liu Y, Ban T, Sun Y. Role of the transient receptor potential melastatin 4 in inhibition effect of arsenic trioxide on the tumor biological features of colorectal cancer cell. PeerJ 2024; 12:e17559. [PMID: 38854798 PMCID: PMC11160432 DOI: 10.7717/peerj.17559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024] Open
Abstract
Background To investigate the effects of arsenic trioxide (ATO) on human colorectal cancer cells (HCT116) growth and the role of transient receptor potential melastatin 4 (TRPM4) channel in this process. Methods The viability of HCT116 cells was assessed using the CCK-8 assay. Western blot analysis was employed to examine the protein expression of TRPM4. The apoptosis of HCT116 cells was determined using TUNEL and Flow cytometry. Cell migration was assessed through the cell scratch recovery assay and Transwell cell migration assay. Additionally, Transwell cell invasion assay was performed to determine the invasion ability of HCT116 cells. Results ATO suppressed the viability of HCT116 cells in a dose-dependent manner, accompanied by a decline in cell migration and invasion, and an increase in apoptosis. 9-phenanthroline (9-Ph), a specific inhibitor of TRPM4, abrogated the ATO-induced upregulation of TRPM4 expression. Additionally, blocking TRPM4 reversed the effects of ATO on HCT116 cells proliferation, including restoration of cell viability, migration and invasion, as well as the inhibition of apoptosis. Conclusion ATO inhibits CRC cell growth by inducing TRPM4 expression, our findings indicate that ATO is a promising therapeutic strategy and TRPM4 may be a novel target for the treatment of CRC.
Collapse
Affiliation(s)
- Zhan Gao
- General Medical Department, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Jing Lv
- Department of Pediatric Dentistry, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ting-Ting Tong
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Kai Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yu-Xuan Han
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yu Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Mei-Mei Shen
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
| | - Tao Ban
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, and Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang, China
| | - Yu Sun
- Harbin Medical University Science Park, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
Ye F, Liu D, Zhang J. Transient receptor potential channel TRPM4 favors oxidized low-density lipoprotein-induced coronary endothelial cell dysfunction via a mechanism involving ferroptosis. Tissue Cell 2024; 86:102290. [PMID: 38103473 DOI: 10.1016/j.tice.2023.102290] [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: 08/31/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Accelerating the repair of damaged endothelium can effectively inhibit the progression of atherosclerosis (AS). Transient receptor potential channel TRPM4 is a non-selective cation channel activated by internal Ca2+, which is expressed in endothelial cells. This study aimed to reveal the potential role of TRPM4 in AS along with the mechanism. Human coronary artery endothelial cells (HCAECs) induced by ox-LDL was regarded as an in vitro model. The impacts of TRPM4 knockdown on cellular inflammation response, oxidative stress, normal endothelial function and lipid peroxidation were evaluated. Given that ferroptosis promotes AS progression, the effects of TRPM4 on intracellular iron ions and ferroptosis-related proteins was determined. Afterwards, HCAECs were treated with ferroptosis inducer erastin, and the influence of ferroptosis in the cellular model was revealed. TRPM4 was elevated in response to ox-LDL treatment in HCAECs. TRPM4 knockdown reduced the inflammation response, oxidative stress and lipid peroxidation caused by ox-LDL, and maintained the normal function of HCAECs. Erastin treatment destroyed the impacts of TRPM4 knockdown that are beneficial for cells to resist ox-LDL, showing the enhancement of the above adverse factors. Together, this study found that TRPM4 knockdown reduced ox-LDL-induced inflammation, oxidative stress, and dysfunction in HCAECs, possibly via a mechanism involving Fe2+ and ferroptosis-related proteins.
Collapse
Affiliation(s)
- Fengxiang Ye
- Cardiology Department, Xuzhou Renci Hospital, Xuzhou, Jiangsu 221000, China
| | - Dongtao Liu
- Cardiology Department, Xuzhou Renci Hospital, Xuzhou, Jiangsu 221000, China
| | - Junjie Zhang
- Cath Lab, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
6
|
Xue Y, Tong T, Zhang Y, Huang H, Zhao L, Lv H, Xiong L, Zhang K, Han Y, Fu Y, Wang Y, Huo R, Wang N, Ban T. miR-133a-3p/TRPM4 axis improves palmitic acid induced vascular endothelial injury. Front Pharmacol 2024; 14:1340247. [PMID: 38269270 PMCID: PMC10806017 DOI: 10.3389/fphar.2023.1340247] [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: 11/17/2023] [Accepted: 12/14/2023] [Indexed: 01/26/2024] Open
Abstract
Background: Vascular endothelial injury is a contributing factor to the development of atherosclerosis and the resulting cardiovascular diseases. One particular factor involved in endothelial cell apoptosis and atherosclerosis is palmitic acid (PA), which is a long-chain saturated fatty acid. In addition, transient receptor potential melastatin 4 (TRPM4), a non-selective cation channel, plays a significant role in endothelial dysfunction caused by various factors related to cardiovascular diseases. Despite this, the specific role and mechanisms of TRPM4 in atherosclerosis have not been fully understood. Methods: The protein and mRNA expressions of TRPM4, apoptosis - and inflammation-related factors were measured after PA treatment. The effect of TRPM4 knockout on the protein and mRNA expression of apoptosis and inflammation-related factors was detected. The changes of intracellular Ca2+, mitochondrial membrane potential, and reactive oxygen species were detected by Fluo-4 AM, JC-1, and DCFH-DA probes, respectively. To confirm the binding of miR-133a-3p to TRPM4, a dual luciferase reporter gene assay was conducted. Finally, the effects of miR-133a-3p and TRPM4 on intracellular Ca2+, mitochondrial membrane potential, and reactive oxygen species were examined. Results: Following PA treatment, the expression of TRPM4 increases, leading to calcium overload in endothelial cells. This calcium influx causes the assemblage of Bcl-2, resulting in the opening of mitochondrial calcium channels and mitochondrial damage, ultimately triggering apoptosis. Throughout this process, the mRNA and protein levels of IL-1β, ICAM-1, and VCAM1 significantly increase. Database screenings and luciferase assays have shown that miR-133a-3p preferentially binds to the 3'UTR region of TRPM4 mRNA, suppressing TRPM4 expression. During PA-induced endothelial injury, miR-133a-3p is significantly decreased, but overexpression of miR-133a-3p can attenuate the progression of endothelial injury. On the other hand, overexpression of TRPM4 counteracts the aforementioned changes. Conclusion: TRPM4 participates in vascular endothelial injury caused by PA. Therefore, targeting TRPM4 or miR-133a-3p may offer a novel pharmacological approach to preventing endothelial injury.
Collapse
Affiliation(s)
- Yadong Xue
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tingting Tong
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuyao Zhang
- Department of Anatomy, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Haijun Huang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ling Zhao
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongzhao Lv
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lingzhao Xiong
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Kai Zhang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuxuan Han
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuyang Fu
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yongzhen Wang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Rong Huo
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ning Wang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tao Ban
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology) at College of Pharmacy, Harbin Medical University, Harbin, China
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
- National-Local Joint Engineering Laboratory of Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, The National Development and Reform Commission, Harbin, China
| |
Collapse
|
7
|
Abstract
Arsenic toxicity is a major concern due to its deleterious consequences for human health. Rapid industrialization also has weakened the quality of the environment by introducing pollutants that may disrupt balanced ecosystems, adversely and irreversibly impacting humans, plants, and animals. Arsenic, an important toxicant among all environmental hazards, can lead to several detrimental effects on cells and organs, impacting the overall quality of life. Nevertheless, arsenic also has a rich history as a chemotherapeutic agent used in ancient days for the treatment of diseases such as malaria, cancer, plague, and syphilis when other chemotherapeutic agents were yet to be discovered. Arsenicosis-mediated disorders remain a serious problem due to the lack of effective therapeutic options. Initially, chelation therapy was used to metabolically eliminate arsenic by forming a complex, but adverse effects limited their pharmacological use. More recently, plant-based products have been found to provide significant relief from the toxic effects of arsenic poisoning. They act by different mechanisms affecting various cellular processes. Phytoconstituents such as curcumin, quercetin, diallyl trisulfide, thymoquinone, and others act via various molecular pathways, primarily by attenuating oxidative damage, membrane damage, DNA damage, and proteinopathies. Nonetheless, most of the phytochemicals reviewed here protect against the adverse effects of metal or metalloid exposure, supporting their consideration as alternatives to chelation therapy. These agents, if used prophylactically and in conjunction with other chemotherapeutic agents, may provide an effective approach for management of arsenic toxicity. In a few instances, such strategies like coadministration of phytochemicals with a known chelating agent have led to more pronounced elimination of arsenic from the body with lesser off-site adverse effects. This is possible because combination treatment ensures the use of a reduced dose of chelating agent with a phytochemical without compromising treatment. Thus, these therapies are more practical than conventional therapeutic agents in ameliorating arsenic-mediated toxicity. This review summarizes the potential of phytochemicals in alleviating arsenic toxicity on the basis of available experimental and clinical evidence.
Collapse
Affiliation(s)
- Sabiya Samim Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | - Ankita Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226020, India
| | - Swaran J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226020, India
| |
Collapse
|
8
|
Farzan SF, Eunus HM, Haque SE, Sarwar G, Hasan AR, Wu F, Islam T, Ahmed A, Shahriar M, Jasmine F, Kibriya MG, Parvez F, Karagas MR, Chen Y, Ahsan H. Arsenic exposure from drinking water and endothelial dysfunction in Bangladeshi adolescents. ENVIRONMENTAL RESEARCH 2022; 208:112697. [PMID: 35007543 PMCID: PMC8917065 DOI: 10.1016/j.envres.2022.112697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 05/12/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide, with ∼80% of CVD-related deaths occurring in low- and middle-income countries. Growing evidence suggests that chronic arsenic exposure may contribute to CVD through its effect on endothelial function in adults. However, few studies have examined the influence of arsenic exposure on cardiovascular health in children and adolescents. To examine arsenic's relation to preclinical markers of endothelial dysfunction, we enrolled 200 adolescent children (ages 15-19 years; median 17) of adult participants in the Health Effects of Arsenic Longitudinal Study (HEALS), in Araihazar, Bangladesh. Participants' arsenic exposure was determined by recall of lifetime well usage for drinking water. As part of HEALS, wells were color-coded to indicate arsenic level (<10 μg/L, 10-50 μg/L, >50 μg/L). Endothelial function was measured by recording fingertip arterial pulsatile volume change and reactive hyperemia index (RHI) score, an independent CVD risk factor, was calculated from these measurements. In linear regression models adjusted for participant's sex, age, education, maternal education, land ownership and body weight, individuals who reported always drinking water from wells with >50 μg/L arsenic had a 11.75% lower level of RHI (95% CI: -21.26, -1.09, p = 0.03), as compared to participants who drank exclusively from wells with ≤50 μg/L arsenic. Sex-stratified analyses suggest that these associations were stronger in female participants. As compared to individuals who drank exclusively from wells with ≤50 μg/L arsenic, the use of wells with >50 μg/L arsenic was associated with 14.36% lower RHI (95% CI: -25.69, -1.29, p = 0.03) in females, as compared to 5.35% lower RHI (95% CI: -22.28, 15.37, p = 0.58) in males for the same comparison. Our results suggest that chronic arsenic exposure may be related to endothelial dysfunction in adolescents, especially among females. Further work is needed to confirm these findings and examine whether these changes may increase risk of later adverse cardiovascular health events.
Collapse
Affiliation(s)
- Shohreh F Farzan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | | | | | | | | | - Fen Wu
- Department of Population Health, New York University, New York, NY, USA
| | | | | | - Mohammad Shahriar
- UChicago Research Bangladesh, Dhaka, Bangladesh; Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Farzana Jasmine
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Faruque Parvez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Yu Chen
- Department of Population Health, New York University, New York, NY, USA
| | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
9
|
Kovács ZM, Dienes C, Hézső T, Almássy J, Magyar J, Bányász T, Nánási PP, Horváth B, Szentandrássy N. Pharmacological Modulation and (Patho)Physiological Roles of TRPM4 Channel—Part 1: Modulation of TRPM4. Pharmaceuticals (Basel) 2022; 15:ph15010081. [PMID: 35056138 PMCID: PMC8781449 DOI: 10.3390/ph15010081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential melastatin 4 is a unique member of the TRPM protein family and, similarly to TRPM5, is Ca2+-sensitive and permeable to monovalent but not divalent cations. It is widely expressed in many organs and is involved in several functions by regulating the membrane potential and Ca2+ homeostasis in both excitable and non-excitable cells. This part of the review discusses the pharmacological modulation of TRPM4 by listing, comparing, and describing both endogenous and exogenous activators and inhibitors of the ion channel. Moreover, other strategies used to study TRPM4 functions are listed and described. These strategies include siRNA-mediated silencing of TRPM4, dominant-negative TRPM4 variants, and anti-TRPM4 antibodies. TRPM4 is receiving more and more attention and is likely to be the topic of research in the future.
Collapse
Affiliation(s)
- Zsigmond Máté Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence:
| |
Collapse
|
10
|
Negri S, Faris P, Moccia F. Reactive Oxygen Species and Endothelial Ca 2+ Signaling: Brothers in Arms or Partners in Crime? Int J Mol Sci 2021; 22:ijms22189821. [PMID: 34575985 PMCID: PMC8465413 DOI: 10.3390/ijms22189821] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
An increase in intracellular Ca2+ concentration ([Ca2+]i) controls virtually all endothelial cell functions and is, therefore, crucial to maintain cardiovascular homeostasis. An aberrant elevation in endothelial can indeed lead to severe cardiovascular disorders. Likewise, moderate amounts of reactive oxygen species (ROS) induce intracellular Ca2+ signals to regulate vascular functions, while excessive ROS production may exploit dysregulated Ca2+ dynamics to induce endothelial injury. Herein, we survey how ROS induce endothelial Ca2+ signals to regulate vascular functions and, vice versa, how aberrant ROS generation may exploit the Ca2+ handling machinery to promote endothelial dysfunction. ROS elicit endothelial Ca2+ signals by regulating inositol-1,4,5-trisphosphate receptors, sarco-endoplasmic reticulum Ca2+-ATPase 2B, two-pore channels, store-operated Ca2+ entry (SOCE), and multiple isoforms of transient receptor potential (TRP) channels. ROS-induced endothelial Ca2+ signals regulate endothelial permeability, angiogenesis, and generation of vasorelaxing mediators and can be exploited to induce therapeutic angiogenesis, rescue neurovascular coupling, and induce cancer regression. However, an increase in endothelial [Ca2+]i induced by aberrant ROS formation may result in endothelial dysfunction, inflammatory diseases, metabolic disorders, and pulmonary artery hypertension. This information could pave the way to design alternative treatments to interfere with the life-threatening interconnection between endothelial ROS and Ca2+ signaling under multiple pathological conditions.
Collapse
|
11
|
Xue Y, Li J, Zhang Y, Li D, Yuan L, Cheng Y, Liu S, Hacker CE. Assessing the vulnerability and adaptation strategies of wild camel to climate change in the Kumtag Desert of China. Glob Ecol Conserv 2021. [DOI: 10.1016/j.gecco.2021.e01725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
12
|
Shi J, Wang X, Chen L, Deng H, Zhang M. HBCD, TBECH, and BTBPE exhibit cytotoxic effects in human vascular endothelial cells by regulating mitochondria function and ROS production. ENVIRONMENTAL TOXICOLOGY 2021; 36:1674-1682. [PMID: 33974337 DOI: 10.1002/tox.23163] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 06/12/2023]
Abstract
Brominated flame retardants (BFRs), such as, 1,2,5,6-tetrabromocyclooctane (HBCD), 1,2-dibromo-4-(1,2-dibromopropyl)cyclohexane (TBECH), and 1 1,2-bis-(2,4,6-tribromophenoxy)ethane (BTBPE), have garnered increasing attention due to their potent biological effects. In the present study, the toxicity of HBCD, TBECH, and BTBPE in human vascular endothelial cells (ECs) was explored. The data showed that HBCD, TBECH, and BTBPE induced cytotoxicity, namely dose-dependent cell viability reduction, cell membrane permeability and apoptosis increase, migration, and lumen formation inhibition. Moreover, HBCD was found to be more toxic than BTBPE or TBECH. Exposure to HBCD, TBECH, and BTBPE led to the production of reactive oxygen species, mitochondrial superoxide generation, and mitochondrial membrane potential collapse, implying that reactive stress caused the cytotoxicity. The ATP content, glutathione content, superoxide dismutase, and MDA activities were reduced, indicating that mitochondrial dysfunction may be the key mechanisms responsible for apoptosis. The present study suggested that mitochondria are a new target of BFRs in ECs and further deepened our understanding of the developmental toxicity of BFRs.
Collapse
Affiliation(s)
- Jun Shi
- Shanghai East Hospital, Key Laboratory of Yangtze River Water Environment Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Xueting Wang
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Chen
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiping Deng
- Shanghai East Hospital, Key Laboratory of Yangtze River Water Environment Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Min Zhang
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Tsai CL, Tsai CW, Chang WS, Lin JC, Hsia TC, Bau DAT. Protective Effects of Baicalin on Arsenic Trioxide-induced Oxidative Damage and Apoptosis in Human Umbilical Vein Endothelial Cells. In Vivo 2021; 35:155-162. [PMID: 33402461 DOI: 10.21873/invivo.12243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIM Arsenic trioxide (As2O3) is an environmental pollutant. However, the detailed mechanisms about As2O3-induced loss of endothelial integrity are unknown. This study aimed at investigating how As2O3 causes endothelial dysfunction and whether baicalin can reverse such dysfunction. MATERIALS AND METHODS Human umbilical vein endothelial cells (HUVECs) were used to examine As2O3-induced oxidative stress, and apoptosis. The influence of baicalin on As2O3-induced endothelial dysfunction were investigated. RESULTS The viability of HUVECs was inhibited by As2O3 and cells underwent apoptosis. As2O3 treatment increased NADPH oxidase activity, and elevated the level of reactive oxygen species (ROS). Formamidopyrimidine DNA-glycosylase- and endonuclease III-digestible adducts were accumulated. Baicalin reversed As2O3-induced apoptosis and As2O3-suppressed cell viability. Baicalin caused a decrease in NADPH oxidase activity, and re-balanced the ROS level. As2O3-induced formamidopyrimidine DNA-glycosylase- and endonuclease III-digestible adducts were down-regulated. CONCLUSION Baicalin was found to have the potential capacity to protect endothelial cells from As2O3-induced cytotoxicity.
Collapse
Affiliation(s)
- Chung-Lin Tsai
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, R.O.C.,Division of Cardiac and Vascular Surgery, Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Jiunn-Cherng Lin
- Division of Cardiology, Department of Internal Medicine, Taichung Veterans General Hospital Chiayi Branch, Chiayi, Taiwan, R.O.C
| | - Te-Chun Hsia
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, R.O.C.; .,Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
14
|
Bahrami A, Sathyapalan T, Moallem SA, Sahebkar A. Counteracting arsenic toxicity: Curcumin to the rescue? JOURNAL OF HAZARDOUS MATERIALS 2020; 400:123160. [PMID: 32574880 DOI: 10.1016/j.jhazmat.2020.123160] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Arsenicosis leads to various irreversible damages in several organs and is considered to be a carcinogen. The effects of chronic arsenic poisoning are a result of an imbalance between pro- and antioxidant homeostasis, oxidative stress, as well as DNA and protein damage. Curcumin, the polyphenolic pigment extracted from the rhizome of Curcuma longa, is well-known for its pleiotropic medicinal effects. Curcumin has been shown to have ameliorative effects in arsenic-induced genotoxicity, nephrotoxicity, hepatotoxicity, angiogenesis, skin diseases, reproductive toxicity, neurotoxicity, and immunotoxicity. This review aims to summarize the scientific evidence on arsenic toxicity in various organs and the ameliorative effects of curcumin on the arsenic toxicity.
Collapse
Affiliation(s)
- Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, HU3 2JZ, UK
| | - Seyed Adel Moallem
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Simard C, Magaud C, Adjlane R, Dupas Q, Sallé L, Manrique A, Bois P, Faivre JF, Guinamard R. TRPM4 non-selective cation channel in human atrial fibroblast growth. Pflugers Arch 2020; 472:1719-1732. [PMID: 33047172 DOI: 10.1007/s00424-020-02476-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022]
Abstract
Cardiac fibroblasts play an important role in cardiac matrix turnover and are involved in cardiac fibrosis development. Ca2+ is a driving belt in this phenomenon. This study evaluates the functional expression and contribution of the Ca2+-activated channel TRPM4 in atrial fibroblast phenotype. Molecular and electrophysiological investigations were conducted in human atrial fibroblasts in primary culture and in atrial fibroblasts obtained from wild-type and transgenic mice with disrupted Trpm4 gene (Trpm4-/-). A typical TRPM4 current was recorded on human cells (equal selectivity for Na+ and K+, activation by internal Ca2+, voltage sensitivity, conductance of 23.2 pS, inhibition by 9-phenanthrol (IC50 = 6.1 × 10-6 mol L-1)). Its detection rate was 13% on patches at days 2-4 in culture but raised to 100% on patches at day 28. By the same time, a cell growth was observed. This growth was smaller when cells were maintained in the presence of 9-phenanthrol. Similar cell growth was measured on wild-type mice atrial fibroblasts during culture. However, this growth was minimized on Trpm4-/- mice fibroblasts compared to control animals. In addition, the expression of alpha smooth muscle actin increased during culture of atrial fibroblasts from wild-type mice. This was not observed in Trpm4-/- mice fibroblasts. It is concluded that TRPM4 participates in fibroblast growth and could thus be involved in cardiac fibrosis.
Collapse
Affiliation(s)
- Christophe Simard
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Christophe Magaud
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM), Université de Poitiers, CNRS, Poitiers, France
| | - Racim Adjlane
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Quentin Dupas
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Laurent Sallé
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Alain Manrique
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France
| | - Patrick Bois
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM), Université de Poitiers, CNRS, Poitiers, France
| | - Jean-François Faivre
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM), Université de Poitiers, CNRS, Poitiers, France
| | - Romain Guinamard
- Groupe Signalisation, Electrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, EA4650, GIP Cyceron, Université de Caen Normandie, Sciences D, Esplanade de la Paix, 14032, Caen Cedex 5, France.
| |
Collapse
|