1
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [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: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
2
|
Wang J, Ma B, Jiang X, Li C, Lin Z, Wang Y, Shi J, Wang G, Cui C. H 2 protects H9c2 cells from hypoxia/reoxygenation injury by inhibiting the Wnt/CX3CR1 signaling pathway. Med Gas Res 2025; 15:339-347. [PMID: 39511756 PMCID: PMC11918467 DOI: 10.4103/mgr.medgasres-d-24-00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 11/15/2024] Open
Abstract
Myocardial ischemia‒reperfusion injury is a severe cardiovascular disease, and its treatment and prevention are crucial for improving patient prognosis and reducing the economic burden. This study aimed to explore the impact of hydrogen (H 2 ) on hypoxia/reoxygenation (H/R) injury in H9c2 cells (derived from rat embryonic heart tissue) induced by hydrogen peroxide (H 2 O 2 ) and to elucidate its underlying mechanism. An H/R injury model was established in H9c2 cells via exposure to 15 μM H 2 O 2 for 3 hours, followed by incubation in a 5% CO 2 atmosphere at 37°C for 24 hours. Then, the cells were treated with H 2 (50%) for 6, 12 or 24 hours. The results demonstrated that H9c2 cells exposed to H 2 O 2 and subjected to H/R injury presented a marked decrease in the cell survival rate, accompanied by severe morphological alterations, such as curling and wrinkling, and elevated lactate dehydrogenase levels. Notably, H 2 mitigated H/R injury induced by H 2 O 2 in a time-dependent manner, improving the morphological damage observed in H9c2 cells and decreasing lactate dehydrogenase levels. Compared with the model group, treatment with H 2 increased the activities of antioxidant enzymes, including catalase, superoxide dismutase, and glutathione peroxidase, while concurrently reducing the level of malondialdehyde, an indicator of cellular damage. Furthermore, H 2 treatment downregulated the expression of inflammatory cytokines and inflammatory-related factors, specifically interleukin-6, high-mobility group box 1, tumor necrosis factor-alpha, and Toll-like receptor 4, in H9c2 cells post-H/R injury. Furthermore, H 2 treatment resulted in a marked decrease in the expression levels of proteins associated with the Wnt/C-X3-C-motif receptor 1 signaling pathway, such as β-catenin, glycogen synthase kinase-3 beta, adenomatous polyposis coli, and Wnt and C-X3-C-motif receptor 1. This observation suggests a potential mechanism for its protective effects against H/R injury. Therefore, H 2 exerts a protective effect against H/R injury in H9c2 cells induced by H 2 O 2 , potentially by inhibiting the activated Wnt/C-X3-C-motif receptor 1 signaling pathway. This inhibition, in turn, prevents the generation of oxidative stress, inflammatory cytokines, and inflammation-associated factors.
Collapse
Affiliation(s)
- Jingsheng Wang
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Bin Ma
- Department of Cardiovascular Medicine, Taian City Taishan District People’s Hospital, Taian, Shandong Province, China
| | - Xue Jiang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Chao Li
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Yumei Wang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Jingfei Shi
- Shandong First Medical University, Jinan, Shandong Province, China
| | - Gang Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Chao Cui
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| |
Collapse
|
3
|
Liu D, Wang C, Chen Y, Huang X, Wen Y, Duan S, Cai Y, Li X, He J, Han K, Li T, Li Y, Xia Z. Protein Kinase C Epsilon Overexpression Protects the Heart Against Doxorubicin-Induced Cardiotoxicity Via Activating SIRT1. Cardiovasc Toxicol 2025; 25:915-928. [PMID: 40327286 PMCID: PMC12116906 DOI: 10.1007/s12012-025-09995-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 04/05/2025] [Indexed: 05/07/2025]
Abstract
Doxorubicin (DOX)-induced cardiotoxicity (DIC) is known to be associated with reduction of cardiac protein kinase C epsilon (PKC-ε). PKC-ε promotes cell survival and protects hearts against various stresses. However, it is unclear whether or not the reduction in cardiac PKC-ε expression plays a causal role in DIC and in particular the potential underlying mechanism whereby PKC-ε may protect against DIC. C57BL/6 mice (8-10-week-old) were either treated with DOX administered intraperitoneally for a duration of 4 weeks to produce cardiotoxicity, or untreated in which mice received the same volume of saline. In vitro, neonatal rat ventricle cardiomyocytes were exposed to DOX for 24 h in the absence or presence of adenovirus overexpressing PKC-ε. Cardiomyocytes in a subgroup were treated with sirtuin-1 (SIRT1) selective inhibitor Ex527. Four weeks after DOX, cardiac contractile function was decreased concomitant with increased serum CK-MB and LDH levels as well as increases in Bax-to-Bcl-2 ratio and Cleaved Caspase 3 proteins expression, while PKC-ε and Sirt1 protein expressions were significantly decreased. In vitro, DOX reduced cardiomyocyte PKC-ε and SIRT1 protein expression, decreased cardiomyocyte viability, and increased LDH release with concomitant increases in oxidative stress and apoptosis. These changes were attenuated by overexpression of PKC-ε. IP study showed that PKC-ε could directly or indirectly bind SIRT1 in cardiomyocytes, and the protect effects of PKC-ε were further canceled by SIRT1 inhibition. In conclusion, activating SIRT1 may represent a major mechanism whereby PKC-ε protects the heart against DOX-induced cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Danyong Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Chunyan Wang
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Yao Chen
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Xiaolei Huang
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Yajie Wen
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Shan Duan
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, China
| | - Xia Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jianfeng He
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Kaijia Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Ting Li
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Yuantao Li
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China.
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Pok Fu Lam Road, Hong Kong, China.
- Doctoral Training Platform for Research and Translation, BoShiWan, GuanChong Village, Shuanghe Town, ZhongXiang, 431913, Hubei, China.
| |
Collapse
|
4
|
Li Z, Chang Y, He D, Dong K, Zhang H, Wang S. Human antigen R -mediated autophagy-related gene 3 methylation enhances autophagy-driven ferroptosis in Crohn's disease colitis. Int Immunopharmacol 2025; 154:114565. [PMID: 40174340 DOI: 10.1016/j.intimp.2025.114565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND AND AIMS Crohn's disease (CD) is a chronic inflammatory disorder that can affect any part of the gastrointestinal tract, with the exact etiology remaining unclear. Recent studies have implicated the role of human antigen R (HuR) in the pathogenesis of various inflammatory diseases, including CD. However, the role of HuR in the modulation of CD remains underexplored. Therefore, this study aimed to investigate the mechanistic involvement of HuR in CD. METHODS We established colitis models using human intestinal epithelial cells and lipopolysaccharide and dextran sulfate sodium-induced mice. Additionally, by knocking out HuR in both cell and animal models, we validated the role of HuR in autophagy and ferroptosis. The role of HuR in regulating ferroptosis accompanied by autophagy activation in CD was detected using ELISA, flow cytometry, immunofluorescence, transmission electron microscopy, Western blot, and RT-qPCR. The demethylation level of ATG3 and the stability of ATG3 mRNA regulated by HuR were detected using immunofluorescence, RIP, and MeRIP-qPCR. The effect of HuR on DSS-induced colitis was evaluated using DAI score, H&E staining, TUNEL staining, and immunohistochemistry. RESULTS The results show that HuR expression is significantly increased in CD colonic inflammation. Compared with the control group, the model group mice exhibited decreased levels of lipid peroxidation markers glutathione and superoxide dismutase, elevated malondialdehyde and reactive oxygen species levels, and reduced expression of iron-related proteins glutathione peroxidase 4, ferritin heavy chain protein 1, and solute carrier family 7 member 11. Additionally, the expression of autophagy-related proteins microtubule-associated protein 1 A/1B-light chain 3, beclin-1, and autophagy related 3 (ATG3) was upregulated, while p62 expression was downregulated. In both in vitro and in vivo models, HuR knockout reversed these changes induced by lipopolysaccharide and dextran sulfate sodium, concomitant with improved tissue pathology. Mechanistically, HuR enhances autophagy-mediated ferroptosis in CD colonic inflammation by regulating ATG3 methylation and mRNA stability. CONCLUSION HuR accelerates colonic inflammation in CD by regulating ATG3 methylation, which enhances autophagy-mediated ferroptosis. Knockout of HuR alleviates Crohn's colitis. This finding provides a potential therapeutic target for the treatment of CD.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Hepatobiliary Pancreas Surgery, Nanshan District People's Hospital, Shenzhen, Guangdong 518052, China
| | - Yunxiang Chang
- Department of Hepatobiliary Pancreas Surgery, Nanshan District People's Hospital, Shenzhen, Guangdong 518052, China
| | - Di He
- Department of Hepatobiliary Pancreas Surgery, Nanshan District People's Hospital, Shenzhen, Guangdong 518052, China
| | - Kai Dong
- Department of Hepatobiliary Pancreas Surgery, Nanshan District People's Hospital, Shenzhen, Guangdong 518052, China
| | - Hongzhen Zhang
- Department of Hepatobiliary Pancreas Surgery, Nanshan District People's Hospital, Shenzhen, Guangdong 518052, China
| | - Shikai Wang
- Department of Hepatobiliary Pancreas Surgery, Nanshan District People's Hospital, Shenzhen, Guangdong 518052, China..
| |
Collapse
|
5
|
Al-Maamari A, Sultan M, Wu S, Zhang T, Wang C, Han B, Duan Y, Ding SS, Chen N, Zhang H, Sun F, Chen X, Yu D, Su S. Activation of sigma 1 receptor attenuates doxorubicin-induced cardiotoxicity by alleviating oxidative stress, mitochondria dysfunction, ER stress-related apoptosis, and autophagy impairment. Int J Biol Macromol 2025; 310:143549. [PMID: 40294674 DOI: 10.1016/j.ijbiomac.2025.143549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The cardiotoxic effects of doxorubicin (DOX) are a significant clinical challenge. This study explored the mechanisms underlying the cardioprotective effects of fluvoxamine (FLU) in DOX-induced cardiotoxicity. In vitro and in vivo models of DOX-induced cardiotoxicity were established using H9c2 cardiomyocytes and BALB/c mice, respectively. The cardioprotective effects of FLU were evaluated using echocardiography, electrocardiography, HE staining, myocardial injury indicators, immunohistochemical staining, immunofluorescence staining, and western blotting. Small interfering RNA (siRNA) targeting the sigma-1 receptor (Sig1R), the Sig1R inhibitor haloperidol (HALO), and the nuclear factor erythroid 2-related factor 2 inhibitor ML385 were used to verify the mechanism of FLU in H9c2 cells, and molecular docking was performed to compare the binding affinities of DOX and FLU to Sig1R. DOX treatment significantly decreased Sig1R expression, leading to cardiac dysfunction, endoplasmic reticulum stress, apoptosis, impaired autophagy, oxidative stress, and mitochondrial dysfunction. Treatment with FLU mitigated these effects. Molecular docking simulations revealed that FLU showed better binding affinity for Sig1R than DOX. Moreover, the beneficial effects of FLU in DOX-induced cardiotoxicity were reduced in the presence of HALO, ML385, or Sig1R siRNA. Our study indicates that FLU effectively reduces DOX-induced cardiotoxicity by activating Sig1R, highlighting its potential as a therapeutic agent against chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ahmed Al-Maamari
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Marwa Sultan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shang Wu
- Breast Cancer Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Tao Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Chuchu Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Boye Han
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yuxin Duan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shan-Shan Ding
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Nannan Chen
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Fangyi Sun
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xueyan Chen
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Ding Yu
- Cardiovascular Intensive Care Unit, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050023, PR China
| | - Suwen Su
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.
| |
Collapse
|
6
|
Shi Y, Cai J, Chen L, Cheng H, Song X, Xue J, Xu R, Ma J, Ge J. AIG1 protects against doxorubicin-induced cardiomyocyte ferroptosis and cardiotoxicity by promoting ubiquitination-mediated p53 degradation. Theranostics 2025; 15:4931-4954. [PMID: 40303337 PMCID: PMC12036874 DOI: 10.7150/thno.108410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/15/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Doxorubicin (DOX) is a widely employed chemotherapeutic drug, while its clinical use is limited by the lethal cardiotoxicity. Previous studies highlighted the critical role of cardiomyocyte ferroptosis in the pathogenesis of DOX-induced cardiotoxicity (DIC). Androgen-induced gene 1 (AIG1) is perceived as a key regulator of oxidative stress-mediated cell death. Nonetheless, it remains elusive whether AIG1 is involved in the progression of DOX-induced cardiomyocyte ferroptosis and cardiotoxicity. Methods: C57BL/6 male mice were repeatedly administrated with DOX at an accumulative dosage of 20 mg/kg to establish a chronic DIC model. Global AIG1 knockout mice and AAV9-mediated cardiac-specific AIG1 knockdown or overexpression mice were utilized to evaluate the precise role of AIG1 in DIC. Additionally, the effects of AIG1 on cardiomyocyte ferroptosis were further investigated following DOX stimulation. Results: Ferroptosis played a pivotal role in DIC in both in vivo and in vitro settings. DOX exposure significantly reduced AIG1 expression levels in cardiomyocytes. Global AIG1 knockout or cardiac-specific AIG1 knockdown mice exhibited deteriorated cardiac function, adverse cardiac remodeling following DOX insult. Moreover, AIG1 deficiency aggravated DOX-evoked ferroptosis and oxidative stress in cardiomyocytes, whereas cardiac-specific overexpression of AIG1 conferred the protective effects manifested by the inhibition of cardiomyocyte ferroptosis and improvements in cardiac performance and remodeling under DOX challenge. Mechanistically, AIG1 directly interacted with the Pirh2 E3 ubiquitin ligase to promote the ubiquitination of p53, a key protein governing ferroptosis during DIC, thereby accelerating its degradation. Cardiac-specific Pirh2 knockdown markedly exacerbated DOX-induced ferroptosis by enhancing p53 activity in cardiomyocytes. Furthermore, the pharmacological administration of a highly selective p53 inhibitor PFT-α effectively ameliorated DIC in mice by inhibiting cardiomyocyte ferroptosis and substantially abrogated the deleterious cardiac effects associated with AIG knockout under DOX challenge. Conclusion: Our findings defined the critical cardioprotective role of AIG1 in DIC by alleviating cardiomyocyte ferroptosis in a Pirh2/p53 axis-dependent manner. Targeting the novelly identified AIG1-Pirh2-p53 signaling axis presents a promising approach to prevent DIC.
Collapse
Affiliation(s)
- Yuekai Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jieru Cai
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lu Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Hao Cheng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaoyue Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junqiang Xue
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Rende Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jianying Ma
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Cheng Q, Zhou L, Fan X, Ma M, Zhang C, Zha X, Zhang Y. FTO-mediated Nrf2 demethylation alleviates high glucose-induced oxidative stress and apoptosis in retinal pigment epithelial cells. Mol Biol Rep 2025; 52:289. [PMID: 40053164 DOI: 10.1007/s11033-025-10400-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: 03/05/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND N6-methyladenosine (m6A) modification contributes to the development of diabetic retinopathy (DR). This study aimed to reveal the role and downstream regulatory signaling of an m6A demethylase fat mass and obesity-associated gene (FTO) in high glucose-induced damage of retinal pigment epithelial cells. METHODS AND RESULTS By stimulating ARPE-19 cells with different concentrations of glucose (0 mM-50 mM), we observed that FTO expression was significantly downregulated, while m6A modification level was upregulated in a glucose concentration-dependent manner in ARPE-19 cells. Then, ARPE-19 cells were transfected with FTO knockdown or overexpression vector, and administrated with high glucose (25mM) to perform functional verification experiments. FTO overexpression recovered cell viability, inhibited cell apoptosis, elevated GSH/GSSG ratio, but reduced MDA and ROS levels in high glucose-induced cells, while FTO knockdown further exacerbated high glucose-triggered oxidative stress and apoptotic cell death. Additionally, FTO overexpression upregulated the expression of NF-E2-related factor 2 (Nrf2) and activated the antioxidant heme oxygenase 1 (HO-1) signal through m6A demethylation on Nrf2 in high glucose-treated ARPE-19 cells. Finally, we proved that knockdown of Nrf2 or HO-1 reversed the protective effects of FTO overexpression on high glucose-treated ARPE-19 cells. CONCLUSION Altogether, the study demonstrated that FTO ameliorates high glucose-triggered oxidative stress and cell apoptosis through activating the Nrf2/HO-1 signaling pathway in an m6A-dependent manner.
Collapse
Affiliation(s)
- Quan Cheng
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China
| | - Liqiong Zhou
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China
| | - Xinyu Fan
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China
| | - Minjun Ma
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China
| | - Chunhui Zhang
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China
| | - Xu Zha
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China.
| | - Yuanping Zhang
- Ophthalmology Department, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, Yunnan, 650000, China.
| |
Collapse
|
8
|
Filcenkova L, Reisbitzer A, Joseph BP, Weber V, Carloni P, Rossetti G, Krauß S. Application of a novel RNA-protein interaction assay to develop inhibitors blocking RNA-binding of the HuR protein. Front Genet 2025; 16:1549304. [PMID: 40110045 PMCID: PMC11921777 DOI: 10.3389/fgene.2025.1549304] [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: 12/20/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
RNA-protein interactions play an important regulatory role in several biological processes. For example, the RNA-binding protein HuR (human antigen R) binds to its target mRNAs and regulates their translation, stability, and subcellular localization. HuR is involved in the pathogenic processes of various diseases. Thus, small molecules blocking RNA-binding of HuR may be useful in a variety of diseases. Previously, we identified STK018404 as a small molecule targeting the HuR-RNA interaction. Based on this study we identified optimized compounds by exploiting combined structure-based and ligand-based computational approaches. To test a series of these compounds, we developed a novel readout system for the HuR-RNA interaction. Traditional methods to detect RNA-protein interaction come with some disadvantages: they require significant reagent optimization and may be difficult to optimize for weakly expressed RNA molecules. The readout often requires amplification. Thus, these methods are not well suited for quantitative analysis of RNA-protein interactions. To achieve an easy-to-perform, rapid, and robust detection of RNA-protein binding, we applied a split luciferase reporter system, to detect the interaction between HuR and its target RNA. We expressed one luciferase fragment as a fusion protein with HuR. The second luciferase fragment was Streptavidin-coated and coupled to a biotinylated RNA-oligo comprising an AU-rich HuR-binding element. The binding between HuR and its target RNA-oligo then allowed reconstitution of the functional luciferase that was detectable by luminescence. Using the split luciferase reporter system, we present here a series of optimized compounds that we developed.
Collapse
Affiliation(s)
- Larissa Filcenkova
- Institute of Biology, Human Biology/Neurobiology, University of Siegen, Siegen, Germany
| | - Annika Reisbitzer
- Institute of Biology, Human Biology/Neurobiology, University of Siegen, Siegen, Germany
| | - Benjamin Philipp Joseph
- Institute for Neuroscience and Medicine and Institute for Advanced Simulations (INM-9/IAS-5), Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, Germany
| | - Verena Weber
- Institute for Neuroscience and Medicine and Institute for Advanced Simulations (INM-9/IAS-5), Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, Germany
| | - Paolo Carloni
- Institute for Neuroscience and Medicine and Institute for Advanced Simulations (INM-9/IAS-5), Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, Germany
| | - Giulia Rossetti
- Institute for Neuroscience and Medicine and Institute for Advanced Simulations (INM-9/IAS-5), Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Department of Neurology, RWTH Aachen University, Aachen, Germany
- Juelich Supercomputing Center (JSC), Forschungszentrum Jülich, Jülich, Germany
| | - Sybille Krauß
- Institute of Biology, Human Biology/Neurobiology, University of Siegen, Siegen, Germany
| |
Collapse
|
9
|
Zhou SM, Shi Y, Li JY, Wang N, Zeng Y, Chen HQ, Tan YP, Deng SW, Liu QQ, Huang XQ, Wang YQ, Zhou ZY, Liu WB. Bisphenol F induces spermatogenic cell ferroptosis via FTO-mediated m6A regulation of FTH1. Free Radic Biol Med 2025; 229:364-373. [PMID: 39848345 DOI: 10.1016/j.freeradbiomed.2025.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Bisphenol F (BPF) has become a new risk factor for male semen quality, but its specific mechanism is still unclear. Therefore, this study explored the potential mechanism of BPF affecting male semen quality from the perspective of ferroptosis and m6A RNA methylation. In vivo experiments showed that BPF destroyed the structure of seminiferous tubules, reduced the layers of spermatogenic cells, and reduced semen quality in mice. Moreover, BPF reduced cell viability and induced ferroptosis in GC-2 cells in vitro. Meanwhile, BPF inhibited the expression of fat mass and obesity-associated gene (FTO). Therefore, we constructed differential expression model of FTO and detected key indicators of ferroptosis such as Fe2+, malondialdehyde, and lipid peroxide. The results found that FTO was important in inhibiting BPF-induced ferroptosis in GC-2 cells. Mechanistically, we found that the m6A modification level on ferritin heavy chain 1 (FTH1) mRNA increased after interfering with FTO by MeRIP assay. Moreover, the RIP assay showed that both YTH N6-methyladenosine RNA binding protein F1 (YTHDF1) and YTH N6-methyladenosine RNA binding protein F2 (YTHDF2) could bind FTH1 mRNA to regulate its expression. This study suggests that FTO regulates the expression of FTH1 in YTHDF1 and YTHDF2 dependent manner and mediates ferroptosis in spermatogenic cells, thus alleviating the reproductive damage induced by BPF.
Collapse
Affiliation(s)
- Shi-Meng Zhou
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yu Shi
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jiang-Ying Li
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Na Wang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Yong Zeng
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hong-Qiang Chen
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yu-Pei Tan
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Shuang-Wu Deng
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Qing-Qing Liu
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Breast and Thyroid Surgery, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Xin-Qiao Huang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yi-Qi Wang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zi-Yuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wen-Bin Liu
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
10
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
11
|
Wang H, Han J, Kong H, Ma C, Zhang XA. The Emerging Role of m6A and Programmed Cell Death in Cardiovascular Diseases. Biomolecules 2025; 15:247. [PMID: 40001550 PMCID: PMC11853213 DOI: 10.3390/biom15020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
N6-methyladenosine (m6A) is the most prevalent internal chemical modification in eukaryotic messenger RNA (mRNA), significantly impacting its lifecycle through dynamic and reversible processes involving methyltransferase, demethylase, and binding proteins. These processes regulate mRNA stability, splicing, nuclear export, translation, and degradation. Programmed cell death (PCD), a tightly controlled process encompassing apoptosis, pyroptosis, ferroptosis, autophagy, and necroptosis, plays a crucial role in maintaining cellular homeostasis, tissue development, and function. Recently, m6A modification has emerged as a significant research area due to its role in regulating PCD and its implications in cardiovascular diseases (CVDs). In this review, we delve into the intricate relationship between various PCD types and m6A modification, emphasizing their pivotal roles in the initiation and progression of CVDs such as myocardial ischemia-reperfusion (I/R), atherosclerosis (AS), pulmonary hypertension (PH), cardiomyopathy, doxorubicin (Dox)-induced cardiotoxicity (DIC), heart failure (HF), and myocardial infarction (MI). Our findings underscore the potential of elucidating the roles of m6A and PCD in CVD to pave new pathways for prevention and treatment strategies.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.); (H.K.)
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.); (H.K.)
| | - Hui Kong
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.); (H.K.)
- College of Exercise and Health, Shanghai Sport University, Shanghai 200438, China
| | - Ce Ma
- Sports Training Teaching and Research Office, Shenyang Sport University, Shenyang 110102, China;
| | - Xin-an Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.); (H.K.)
| |
Collapse
|
12
|
Wu Z, Zhang Y, Zhong W, Wu K, Zhong T, Jiang T. Targeting ferroptosis: a promising approach for treating lung carcinoma. Cell Death Discov 2025; 11:33. [PMID: 39875356 PMCID: PMC11775225 DOI: 10.1038/s41420-025-02308-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
Lung carcinoma incidence and fatality rates remain among the highest on a global scale. The efficacy of targeted therapies and immunotherapies is commonly compromised by the emergence of drug resistance and other factors, resulting in a lack of durable therapeutic benefits. Ferroptosis, a distinct pattern of cell death marked by the buildup of iron-dependent lipid peroxides, has been shown to be a novel and potentially more effective treatment for lung carcinoma. However, the mechanism and regulatory network of ferroptosis are exceptionally complex, and many unanswered questions remain. In addition, research on ferroptosis in the diagnosis and treatment of lung cancer has been growing exponentially. Therefore, it is necessary to provide a thorough summary of the latest advancements in the field of ferroptosis. Here, we comprehensively analyze the mechanisms underlying the preconditions of ferroptosis, the defense system, and the associated molecular networks. The potential strategies of ferroptosis in the treatment of lung carcinoma are also highlighted. Targeting ferroptosis improves tumor cell drug resistance and enhances the effectiveness of targeted drugs and immunotherapies. These findings may shed fresh light on the diagnosis and management of lung carcinoma, as well as the development of drugs related to ferroptosis.
Collapse
Affiliation(s)
- Ziyang Wu
- School of Life Sciences, Zhuhai College of Science and Technology, Zhuhai, Guangdong, China
| | - Yan Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wendi Zhong
- School of Life Sciences, Zhuhai College of Science and Technology, Zhuhai, Guangdong, China
| | - Kunjian Wu
- School of Life Sciences, Zhuhai College of Science and Technology, Zhuhai, Guangdong, China
| | - Tian Zhong
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao
| | - Tao Jiang
- School of Life Sciences, Zhuhai College of Science and Technology, Zhuhai, Guangdong, China.
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao.
| |
Collapse
|
13
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
14
|
Jing C, Wu Y, Zhang Y, Zhu Z, Zhang Y, Liu Z, Sun D. Epigenetic regulation and post-translational modifications of ferroptosis-related factors in cardiovascular diseases. Clin Epigenetics 2025; 17:4. [PMID: 39799367 PMCID: PMC11724467 DOI: 10.1186/s13148-024-01809-5] [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: 11/15/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
As an important element of the human body, iron participates in numerous physiological and biochemical reactions. In the past decade, ferroptosis (a form of iron-dependent regulated cell death) has been reported to contribute to the pathogenesis and progression of various diseases. The stability of iron in cardiomyocytes is crucial for the maintenance of normal physiological cardiac activity. Ferroptosis has been detected in many cardiovascular diseases (CVDs), including coronary heart disease, myocardial ischemia-reperfusion injury, heart failure, and chemotherapy-induced myocardial damage. In cardiomyocytes, epigenetic regulation and post-translational modifications regulate the expression of ferroptosis-related factors, maintain iron homeostasis, and participate in the progression of CVDs. Currently, there is no detailed mechanism to explain the relationship between epigenetic regulation and ferroptosis in CVDs. In this review, we provide an initial summary of the core mechanisms of ferroptosis in cardiomyocytes, with first focus on the epigenetic regulation and expression of ferroptosis-related factors in the context of common cardiovascular diseases. We anticipate that the new insights into the pathogenesis of CVDs provided here will inspire the development of clinical interventions to specifically target the active sites of these factors, reducing the harmfulness of ferroptosis to human health.
Collapse
Affiliation(s)
- Chunlu Jing
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
- Dalian Medical University, Dalian, 116044, Liaoning, People's Republic of China
| | - Yupeng Wu
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
| | - Yuzhu Zhang
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
| | - Zaihan Zhu
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
| | - Yong Zhang
- Department of Urology, The People's Hospital of Liaoning Province, The People's Hospital of China Medical University, 33 Wenyi Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Zhen Liu
- Department of Urology, The People's Hospital of Liaoning Province, The People's Hospital of China Medical University, 33 Wenyi Road, Shenhe District, Shenyang, 110016, People's Republic of China.
| | - Dandan Sun
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China.
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China.
| |
Collapse
|
15
|
Meng W, Li L. N6-methyladenosine modification of SPOP relieves ferroptosis and diabetic cardiomyopathy by enhancing ubiquitination of VDAC3. Free Radic Biol Med 2025; 226:216-229. [PMID: 39549880 DOI: 10.1016/j.freeradbiomed.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/17/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Understanding the pathogenesis of diabetic cardiomyopathy (DCM), a common microvascular complication affecting the heart, is crucial for identifying new therapeutic targets and intervention strategies for DCM. Our study revealed a significant downregulation in Speckle-type POZ protein (SPOP) expression in DCM, while the overexpression of SPOP improved DCM-induced myocardial dysfunction, injury, fibrosis, hypertrophy, and ferroptosis. Mechanistically, SPOP facilitated the degradation of voltage-dependent anion channel 3 (VDAC3) by enhancing its ubiquitination. M6A demethylase AlkB homolog 5 (ALKBH5) reduced the mRNA stability of SPOP by decreasing m6A modification in its 3'UTR. The m6A reader insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) enhanced the stability of SPOP mRNA through recognition of m6A-modified SPOP 3'UTR. Furthermore, ALKBH5 promoted ferroptosis by inhibiting SPOP-induced VDAC3 degradation, while IGF2BP2 inhibited ferroptosis via activation of SPOP-induced VDAC3 degradation in high glucose-treated neonatal mouse ventricular cardiomyocytes (NMVCs). Overall, our study has unveiled a novel role of SPOP in the pathogenesis of ferroptosis and DCM, thereby significantly advancing our understanding of the involvement of ferroptosis during the progression of DCM. Moreover, this discovery offers promising potential therapeutic interventions targeting DCM.
Collapse
Affiliation(s)
- Wei Meng
- Department of Geriatric, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, 646000, China
| | - Linghua Li
- Department of Electrocardiography and Electroencephalography, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, 646000, China.
| |
Collapse
|
16
|
Shi H, Zou Y, Li Y, Li Y, Liu B. Neuregulin-1 reduces Doxorubicin-induced cardiotoxicity by upregulating YAP to inhibit senescence. Int Immunopharmacol 2024; 143:113278. [PMID: 39405937 DOI: 10.1016/j.intimp.2024.113278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024]
Abstract
The cardiotoxicity of Doxorubicin (Dox) limits its clinical application, creating an urgent need to investigate its underlying mechanism and develop effective therapies. Senescence plays an important role in Dox-induced cardiotoxicity (DIC). Recently, Neuregulin-1 (NRG1) was found to regulate Yes-associated protein (YAP), which was reported to inhibit senescence, suggesting that NRG1 might be used to treat DIC by inhibiting senescence through YAP regulation. We examined the changes and regulatory roles of YAP and senescence in Dox cardiotoxicity and whether NRG1 could reduce DIC in chronic DIC mice and Dox-treated H9c2 cells. Our study revealed that sustained small doses of Dox impaired cardiac function and H9c2 cell viability, induced myocardial senescence, and inhibited YAP expression. Conversely, high levels of YAP inhibited Dox-induced senescence in H9c2 cells, indicating that Dox promotes myocardial senescence by inhibiting YAP. In addition, we found that exogenous NRG1 inhibited the phosphorylation of LATS1 and MST1, thereby inhibiting YAP phosphorylation and promote the nuclear translocation of YAP, inhibiting senescence and attenuating Dox-induced cardiotoxicity. YAP knockdown or inhibition of YAP binding to TEA domain transcription factor protein (TEAD)blocks the protective effects of NRG1. In conclusion, our study suggests that Dox-induced myocardial senescence through YAP inhibition is one of the pathological mechanisms of its cardiotoxicity. Additionally, NRG1 reduces DIC by upregulating YAP to inhibit senescence.
Collapse
Affiliation(s)
- Henghe Shi
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yifei Zou
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yinghao Li
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yangxue Li
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China.
| |
Collapse
|
17
|
Benak D, Sevcikova A, Holzerova K, Hlavackova M. FTO in health and disease. Front Cell Dev Biol 2024; 12:1500394. [PMID: 39744011 PMCID: PMC11688314 DOI: 10.3389/fcell.2024.1500394] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Fat mass and obesity-associated (FTO) protein, a key enzyme integral to the dynamic regulation of epitranscriptomic modifications in RNAs, significantly influences crucial RNA lifecycle processes, including splicing, export, decay, and translation. The role of FTO in altering the epitranscriptome manifests across a spectrum of physiological and pathological conditions. This review aims to consolidate current understanding regarding the implications of FTO in health and disease, with a special emphasis on its involvement in obesity and non-communicable diseases associated with obesity, such as diabetes, cardiovascular disease, and cancer. It also summarizes the established molecules with FTO-inhibiting activity. Given the extensive impact of FTO on both physiology and pathophysiology, this overview provides illustrative insights into its roles, rather than an exhaustive account. A proper understanding of FTO function in human diseases could lead to new treatment approaches, potentially unlocking novel avenues for addressing both metabolic disorders and malignancies. The evolving insights into FTO's regulatory mechanisms hold great promise for future advancements in disease treatment and prevention.
Collapse
Affiliation(s)
| | | | | | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
18
|
Xiong L, Xiao Q, Chen R, Huang L, Gao J, Wang L, Li G, Li Y. Histone deacetylase 9 promotes osteogenic trans-differentiation of vascular smooth muscle cells via ferroptosis in chronic kidney disease vascular calcification. Ren Fail 2024; 46:2422435. [PMID: 39500708 PMCID: PMC11539403 DOI: 10.1080/0886022x.2024.2422435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
Vascular calcification, a common complication of chronic kidney disease (CKD), remains an unmet therapeutic challenge. The trans-differentiation of vascular smooth muscle cells (VSMCs) into osteoblast-like cells is crucial in the pathogenesis of vascular calcification in CKD. Despite ferroptosis promotes vascular calcification in CKD, the upstream or downstream regulatory mechanisms involved remains unclear. In this study, we aimed to investigate the regulatory mechanism involved in ferroptosis in CKD vascular calcification. Transcriptome sequencing revealed a potential relationship between HDAC9 and ferroptosis in CKD vascular calcification. Subsequently, we observed increased expression of HDAC9 in calcified arteries of patients undergoing hemodialysis and in a rat model of CKD. We further demonstrated that knockout of HDAC9 attenuates osteogenic trans-differentiation and ferroptosis in VSMCs stimulated by high calcium and phosphorus. In addition, RSL3 exacerbated ferroptosis and osteogenic trans-differentiation of VSMCs exposed to high levels of calcium and phosphorus, and these effects were suppressed to some extent by HDAC9 knockout. In summary, our findings suggest that HDAC9 promotes VSMCs osteogenic trans-differentiation involving ferroptosis, providing new insights for the therapy of CKD vascular calcification.
Collapse
MESH Headings
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/etiology
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Rats
- Cell Transdifferentiation
- Humans
- Histone Deacetylases/metabolism
- Histone Deacetylases/genetics
- Ferroptosis
- Male
- Osteogenesis
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Disease Models, Animal
- Rats, Sprague-Dawley
- Cells, Cultured
Collapse
Affiliation(s)
- Lin Xiong
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Qiong Xiao
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Rong Chen
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Liming Huang
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Jun Gao
- Department of Toxicological Inspection Sichuan Provincial Centre for Disease Prevention and Control, Chengdu, China
| | - Li Wang
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Guisen Li
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Yi Li
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
19
|
Tan YT, Li T, Wang RB, Liu ZK, Ma MY, Huang RZ, Mo HY, Luo SY, Lin JF, Xu RH, Ju HQ. WTAP weakens oxaliplatin chemosensitivity of colorectal cancer by preventing PANoptosis. Cancer Lett 2024; 604:217254. [PMID: 39270768 DOI: 10.1016/j.canlet.2024.217254] [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: 06/11/2024] [Revised: 08/17/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
As the most abundant post-transcriptional modification in eukaryotes, N6-methyladenosine (m6A) plays a crucial role in cancer cell proliferation, invasion and chemoresistance. However, its specific effects on chemosensitivity to oxaliplatin-based regimens and the impact of these drugs on m6A methylation levels in colorectal cancer (CRC) remain largely unexplored. In this study, we demonstrated that the m6A methyltransferase Wilms tumor 1-associating protein (WTAP) weakens oxaliplatin chemosensitivity in HCT116 and DLD1 cells. Mechanistically, oxaliplatin treatment upregulated WTAP expression, preventing multiple forms of cell death simultaneously, a process known as PANoptosis, by decreasing intracellular oxidative stress through maintaining the expression of nuclear factor erythroid-2-related factor 2 (NRF2), a major antioxidant response element, in an m6A-dependent manner. In addition, high WTAP expression in CRC patients is associated with a poor prognosis and reduced benefit from standard chemotherapy by clinical data analysis of The Cancer Genome Atlas (TCGA) database and patient cohort study. These findings suggest that targeting WTAP-NRF2-PANoptosis axis could enhance the antitumor efficacy of oxaliplatin-based chemotherapy in CRC treatment.
Collapse
Affiliation(s)
- Yue-Tao Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Ting Li
- Department of Gastroenterology and Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, PR China
| | - Ruo-Bing Wang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Ze-Kun Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China; Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Meng-Yao Ma
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Ren-Ze Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Hai-Yu Mo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Shu-Yu Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Jin-Fei Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China; Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Rui-Hua Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, PR China.
| | - Huai-Qiang Ju
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China.
| |
Collapse
|
20
|
Gao Q, Chen JM, Li CSZ, Zhan JY, Yin XD, Li BS, Dong HL, Luo LX, Li ZL. CDKN1A promotes Cis-induced AKI by inducing cytoplasmic ROS production and ferroptosis. Food Chem Toxicol 2024; 193:115003. [PMID: 39353481 DOI: 10.1016/j.fct.2024.115003] [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: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND OBJECTIVE This study focuses on investigating the role of CDKN1A in cisplatin-induced AKI (acute kidney injury, AKI) and its potential as a biomarker for early diagnosis and therapeutic intervention by integrating bioinformatics analysis, machine learning, and experimental validation. METHODS We analyzed the GSE85957 dataset to find genes that changed between control and cisplatin-treated rats. Using bioinformatics and machine learning, we found 13 important genes related to ferroptosis and the P53 pathway. The key gene, CDKN1A, was identified using various algorithms. We then tested how reducing CDKN1A in human kidney cells affected cell health, ROS, and iron levels. We also checked how CDKN1A changes the levels of proteins linked to ferroptosis using Q-PCR and Western Blot. RESULTS CDKN1A was found to negatively regulate the G1/S phase transition and was associated with ferroptosis in p53 signaling. Experiments in human renal tubular epithelial cells (HK-2) and rat NRK-52E cells showed that CDKN1A knockdown mitigated cisplatin-induced cell injury by reducing oxidative stress and ferroptosis. CONCLUSION Our integrated approach identified CDKN1A as a biomarker for cisplatin-induced AKI. Its regulation could be key in AKI pathogenesis, offering new therapeutic insights and aiding in early diagnosis and intervention.
Collapse
Affiliation(s)
- Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, China
| | - Jun-Ming Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Chen-Sui-Zi Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, China
| | - Jia-Yi Zhan
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xue-Dong Yin
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ben-Shang Li
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, China
| | - Hong-Liang Dong
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - Lian-Xiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Zhi-Ling Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
21
|
Jiang W, Yu L, Mu N, Zhang Z, Ma H. MG53 inhibits ferroptosis by targeting the p53/SLC7A11/GPX4 pathway to alleviate doxorubicin-induced cardiotoxicity. Free Radic Biol Med 2024; 223:224-236. [PMID: 39111582 DOI: 10.1016/j.freeradbiomed.2024.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Doxorubicin (DOX) is an anthracycline medication that is commonly used to treat solid tumors. However, DOX has limited clinical efficacy due to known cardiotoxicity. Ferroptosis is involved in DOX-induced cardiotoxicity (DIC). Although mitsugumin-53 (MG53) has cardioprotective effects and is expected to attenuate myocardial ischemic injury, its ability to inhibit DOX-induced ferroptosis has not been extensively studied. This research aims to investigate the pathophysiological impact of MG53 on DOX induced ferroptosis. Here, MG53 levels were evaluated in relation to the extent of ferroptosis by establishing in vivo and in vitro DIC mouse models. Additionally, myocardial specific MG53 overexpressing mice were used to study the effect of MG53 on cardiac function in DIC mice. The study found that the MG53 expression decreased in DOX treated mouse hearts or cardiomyocytes. However, MG53-overexpressing improved cardiac function in the DIC model and effectively reduced myocardial ferroptosis by increasing solute carrier family 7 member 11 (SLC7A11) and Glutathione peroxidase 4 (GPX4) levels, which were decreased by DOX. Mechanistically, MG53 binds to tumor suppressor 53 (p53) to regulate its ubiquitination and degradation. Ferroptosis induced by DOX was prevented by either MG53 overexpression or p53 knockdown in cardiomyocytes. The modulation of the p53/SLC7A11/GPX4 pathway by overexpression of MG53 can alleviate DOX induced ferroptosis. The study indicates that MG53 can provide protection against DIC by increasing p53 ubiquitination. These results highlight the previously unidentified role of MG53 in inhibiting ferroptosis to prevent DIC.
Collapse
Affiliation(s)
- Wenhua Jiang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zihui Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China.
| | - Heng Ma
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China; Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
22
|
Chen C, Wang J, Zhang S, Zhu X, Hu J, Liu C, Liu L. Epigenetic regulation of diverse regulated cell death modalities in cardiovascular disease: Insights into necroptosis, pyroptosis, ferroptosis, and cuproptosis. Redox Biol 2024; 76:103321. [PMID: 39186883 PMCID: PMC11388786 DOI: 10.1016/j.redox.2024.103321] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024] Open
Abstract
Cell death constitutes a critical component of the pathophysiology of cardiovascular diseases. A growing array of non-apoptotic forms of regulated cell death (RCD)-such as necroptosis, ferroptosis, pyroptosis, and cuproptosis-has been identified and is intimately linked to various cardiovascular conditions. These forms of RCD are governed by genetically programmed mechanisms within the cell, with epigenetic modifications being a common and crucial regulatory method. Such modifications include DNA methylation, RNA methylation, histone methylation, histone acetylation, and non-coding RNAs. This review recaps the roles of DNA methylation, RNA methylation, histone modifications, and non-coding RNAs in cardiovascular diseases, as well as the mechanisms by which epigenetic modifications regulate key proteins involved in cell death. Furthermore, we systematically catalog the existing epigenetic pharmacological agents targeting novel forms of RCD and their mechanisms of action in cardiovascular diseases. This article aims to underscore the pivotal role of epigenetic modifications in precisely regulating specific pathways of novel RCD in cardiovascular diseases, thus offering potential new therapeutic avenues that may prove more effective and safer than traditional treatments.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China.
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| |
Collapse
|
23
|
Zeng H, Xu J, Wu R, Wang X, Jiang Y, Wang Q, Guo J, Xiao F. FTO alleviated ferroptosis in septic cardiomyopathy via mediating the m6A modification of BACH1. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167307. [PMID: 38897256 DOI: 10.1016/j.bbadis.2024.167307] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Sepsis is a global health challenge that results in systemic inflammation, oxidative stress, and multi-organ dysfunction, with the heart being particularly susceptible. This study aimed to elucidate the effect of FTO, a key regulator in m6A methylation in septic cardiomyopathy, and its potential therapeutic implications. Cellular and animal models of septic myocardial injury were established. Moreover, it was revealed that ferroptosis, which is a form of programmed necrosis occurring with iron dependence, was activated within cardiomyocytes during septic conditions. The overexpression of FTO-suppressed ferroptosis alleviated heart inflammation and dysfunction and improved survival rates in vivo. However, the protective effects of FTO were attenuated by the overexpression of BACH1, which is a molecule negatively correlated with FTO. Mechanistically, FTO modulated the m6A modification of BACH1, suggesting a complex interplay in the regulation of cardiomyocyte damage and sepsis. Our findings reveal the potential of targeting the FTO/BACH1 axis and ferroptosis inhibitors as therapeutic strategies for sepsis-induced cardiac injuries.
Collapse
Affiliation(s)
- Hua Zeng
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junmei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Rui Wu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xin Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yaqing Jiang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qing Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jiali Guo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feng Xiao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
24
|
Liu Y, Wu H, Zhou G, Zhang D, Yang Q, Li Y, Yang X, Sun J. Role of M6a Methylation in Myocardial Ischemia-Reperfusion Injury and Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024; 24:918-928. [PMID: 39026038 DOI: 10.1007/s12012-024-09898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide, with acute myocardial infarction and anticancer drug-induced cardiotoxicity being the significant factors. The most effective treatment for acute myocardial infarction is rapid restoration of coronary blood flow by thrombolytic therapy or percutaneous coronary intervention. However, myocardial ischemia-reperfusion injury (MI/RI) after reperfusion therapy is common in acute myocardial infarction, thus affecting the prognosis of patients with acute myocardial infarction. There is no effective treatment for MI/RI. Anthracyclines such as Doxorubicin (DOX) have limited clinical use due to their cardiotoxicity, and the mechanism of DOX-induced cardiac injury is complex and not yet fully understood. N6-methyladenosine (m6A) plays a crucial role in many biological processes. Emerging evidence suggests that m6A methylation plays a critical regulatory role in MI/RI and DOX-induced cardiotoxicity (DIC), suggesting that m6A may serve as a novel biomarker and therapeutic target for MI/RI and DIC. M6A methylation may mediate the pathophysiological processes of MI/RI and DIC by regulating cellular autophagy, apoptosis, oxidative stress, and inflammatory response. In this paper, we first focus on the relationship between m6A methylation and MI/RI, then further elucidate that m6A methylation may mediate the pathophysiological process of MI/RI through the regulation of cellular autophagy, apoptosis, oxidative stress, and inflammatory response. Finally, briefly outline the roles played by m6A in DIC, which will provide a new methodology and direction for the research and treatment of MI/RI and DIC.
Collapse
Affiliation(s)
- Yanfang Liu
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Hui Wu
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China.
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China.
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China.
| | - Gang Zhou
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Dong Zhang
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Qingzhuo Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yi Li
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Xiaoting Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Jianfeng Sun
- Department of Vascular Surgery, The First College of Medical Science, Yichang Central People's Hospital, China Three Gorges University, Hubei, 443000, China
| |
Collapse
|
25
|
Ren C, Cao Z, Liu Y, Wang R, Lin C, Wang Z. Medicinal chemistry aspects of fat mass and obesity associated protein: structure, function and inhibitors. Future Med Chem 2024; 16:1705-1726. [PMID: 39101588 PMCID: PMC11370915 DOI: 10.1080/17568919.2024.2380245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Adiposity and obesity-related proteins (FTO), the earliest identified mRNA N6-methyladenosine (m6A) demethylases, are known to play crucial roles in several biological processes. Therefore, FTO is a promising target for anticancer treatment. Understanding the biological functions and regulatory mechanisms of FTO targets can serve as guidelines for drug development. Despite significant efforts to develop FTO inhibitors, no specific small-molecule inhibitors have entered clinical trials so far. In this manuscript, we review the relationship between FTO and various cancers, the small-molecule inhibitors developed against FTO targets from the perspective of medicinal chemistry and other fields, and describe their structural optimization process and structure-activity relationship, providing clues for their future development direction.
Collapse
Affiliation(s)
- Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Zhi Cao
- Medical Quality Control & Evaluation Department, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Yang Liu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Rui Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Congcong Lin
- Department of Pharmaceutics, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zishu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| |
Collapse
|
26
|
Liu J, Liu H, Deng L, Wang T, Li L, Chen Y, Qu L, Zou W. Protective Role of Dioscin against Doxorubicin-Induced Chronic Cardiotoxicity: Insights from Nrf2-GPX4 Axis-Mediated Cardiac Ferroptosis. Biomolecules 2024; 14:422. [PMID: 38672439 PMCID: PMC11047995 DOI: 10.3390/biom14040422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Recent evidence suggests that ferroptosis, an iron-facilitated cell death with excessive lipid peroxidation, is a critical mechanism underlying doxorubicin (DOX)-induced cardiotoxicity (DIC). Although dioscin has been reported to improve acute DIC, direct evidence is lacking to clarify the role of dioscin in chronic DIC and its potential mechanism in cardiac ferroptosis. In this study, we used chronic DIC rat models and H9c2 cells to investigate the potential of dioscin to mitigate DIC by inhibiting ferroptosis. Our results suggest that dioscin significantly improves chronic DIC-induced cardiac dysfunction. Meanwhile, it significantly inhibited DOX-induced ferroptosis by reducing Fe2+ and lipid peroxidation accumulation, maintaining mitochondrial integrity, increasing glutathione peroxidase 4 (GPX4) expression, and decreasing acyl-CoA synthetase long-chain family 4 (ACSL4) expression. Through transcriptomic analysis and subsequent validation, we found that the anti-ferroptotic effects of dioscin are achieved by regulating the nuclear factor-erythroid 2-related factor 2 (Nrf2)/GPX4 axis and Nrf2 downstream iron metabolism genes. Dioscin further downregulates nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) and upregulates expression of frataxin (FXN) and ATP-binding cassette B8 (ABCB8) to limit mitochondrial Fe2+ and lipid peroxide accumulation. However, Nrf2 inhibition diminishes the anti-ferroptotic effects of dioscin, leading to decreased GPX4 expression and increased lipid peroxidation. This study is a compelling demonstration that dioscin can effectively reduce DIC by inhibiting ferroptosis, which is dependent on the Nrf2/GPX4 pathway modulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Liping Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (J.L.); (H.L.); (L.D.); (T.W.); (L.L.); (Y.C.)
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (J.L.); (H.L.); (L.D.); (T.W.); (L.L.); (Y.C.)
| |
Collapse
|