1
|
Wang Z, Liu S, Shi J, Chen D, Li S, Yu S, Liu S, Yang K, Zhang W, Gao X, Zhang S. Empagliflozin ameliorates RSL3-induced ferroptosis in vascular endothelial cells via the NRF2/HO-1 pathway. BMC Cardiovasc Disord 2025; 25:437. [PMID: 40483457 PMCID: PMC12145636 DOI: 10.1186/s12872-025-04890-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Endothelial cell dysfunction is a fundamental injury of atherosclerosis cardiovascular disease (ASCVD), closely linked to ferroptosis, which is a novel type of cell death induced by iron-dependent lipid peroxidation. Several clinical trials have suggested that empagliflozin, a selective inhibitor of sodium glucose co-transporter 2, reduces the risk of hospitalization for heart failure and cardiovascular death in patients with type 2 diabetes. However, little is known about the mechanism of EMPA in endothelial cell ferroptosis in ASCVD. The aim of the present study was to evaluate the potential mechanism of EMPA against ferroptosis induced by RAS-selective lethal 3 (RSL3) in endothelial cells.EA.hy926 human umbilical vein endothelial cells were cultured in vitro and were divided into four groups: The Control, RSL3, RSL3 + low-concentration EMPA intervention and RSL3 + high-concentration EMPA intervention groups. Iron-dependent lipid peroxidation was assessed by detecting the fluorescence intensity of ferrous ion (Fe2+), lipid reactive oxygen species (ROS) and content of malondialdehyde (MDA). The expression of ferroptosis-related genes was assessed by RT-qPCR and western blotting. siRNA nuclear factor erythroid 2-related factor 2 (NRF2) was transfected into EA.hy926 cells to measure the expression of target genes.It was demonstrated that the level of MDA, Fe2+ and lipid ROS was higher in the RSL3-treated group compared with the EMPA intervention group, while EMPA markedly mitigated that effect. In addition, EMPA can reverse the RSL3-induced low expression of glutathione peroxidase 4 (GPX4) and high expression of ACSL4 in endothelial cells, as evidenced by the upregulation of nuclear factor transcription factor nuclear factor, erythroid 2 (NFE2)-related factor 2 and heme oxygenase-1 expression, while siNRF2 transfection impaired the antiferroptosis effect of EMPA. The present study indicated that EMPA may inhibit RSL3-induced ferroptosis in endothelial cells by activating the NRF2/heme oxygenase 1 gene pathway.
Collapse
Affiliation(s)
- ZiLin Wang
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - SiMiao Liu
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - JiaHao Shi
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - Di Chen
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - ShaoLin Li
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - ShengQin Yu
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - SiXu Liu
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - KaiJing Yang
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - Wan Zhang
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - Xue Gao
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China
| | - ShuYing Zhang
- Affiliated Zhongshan Hospital of Dalian university, Dalian, 116001, China.
| |
Collapse
|
2
|
Liu Y, Tian X, Jia C, Cheng X, Cui C, Li C, Yang S. The role of CXCL16 in atherosclerosis: from mechanisms to therapy. Front Immunol 2025; 16:1555438. [PMID: 40491927 PMCID: PMC12146295 DOI: 10.3389/fimmu.2025.1555438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Atherosclerosis (AS), as the primary pathological basis of cardiovascular and cerebrovascular diseases, is closely associated with chemokines in its occurrence and progression. CXCL16 establishes a new link between chemokines and AS. We briefly introduced the structural characteristics of CXCL16 and its specific receptor CXCR6, as well as related signaling pathways. Furthermore, the significant role of CXCL16 in the progression of AS was elaborated from the perspective of pathological mechanisms and signal pathways. Meanwhile, we objectively evaluated the potential arterial protective effects of CXCL16. Finally, we discussed various novel therapeutic strategies to alleviate AS by targeting the inhibition of CXCL16 and its regulatory pathways. This review systematically summarizes the multifaceted roles of CXCL16 in AS, providing theoretical foundations and research insights for the precise prevention and treatment of AS.
Collapse
Affiliation(s)
- Yue Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xintao Tian
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chunyan Jia
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xinrui Cheng
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Changxing Cui
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cuiping Li
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shaonan Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
3
|
Zaidi SA, Fan Z, Chauhdari T, Ding Y. MicroRNA regulatory dynamic, emerging diagnostic and therapeutic frontier in atherosclerosis. Microvasc Res 2025; 160:104818. [PMID: 40368159 DOI: 10.1016/j.mvr.2025.104818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
MicroRNAs (miRNAs), a class of non-coding RNAs, are pivotal post-transcriptional regulators of gene expression with profound implications in the pathogenesis of atherosclerosis (AS). As a progressive arterial disease driven by vascular cells dysfunction, lipid dysregulation and subsequent chronic inflammation, AS remains a leading cause of global morbidity. Recent studies have demonstrated how important miRNAs are in regulating central biological processes in the vascular wall, such as endothelial function, vascular smooth muscle cell (VSMC) phenotypic switching, and macrophage polarization. This review provides comprehensive insight into the role of miRNAs in the development and complexity of atherosclerotic plaques according to their effects on endothelial cells, macrophages, and VSMCs. We also go over the growing prospects of miRNAs as therapeutic targets and diagnostic biomarkers, providing information to be used in the study of vascular diseases. Lastly, we address recent complications and potential applications of miRNA-based approaches in clinical practice.
Collapse
Affiliation(s)
- Syeda Armana Zaidi
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Zhiyu Fan
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Talha Chauhdari
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| |
Collapse
|
4
|
Ouyang M, Wu J, Hu X, Liu C, Zhou D. Decoding the power of saponins in ferroptosis regulation and disease intervention: a review. J Pharm Pharmacol 2025; 77:593-608. [PMID: 39673380 DOI: 10.1093/jpp/rgae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/05/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVES This review endeavors to elucidate the complex interplay underlying diseases associated with ferroptosis and to delineate the multifaceted mechanisms by which triterpenoid and steroidal saponins modulate this form of cell death. METHODS A meticulous examination of the literature was undertaken, drawing from an array of databases including Web of Science, PubMed, and Wiley Library, with a focus on the keywords "ferroptosis," "saponin," "cancer," "inflammation," "natural products," and "signaling pathways." KEY FINDINGS Ferroptosis represents a distinctive mode of cell death that holds considerable promise for the development of innovative therapeutic strategies targeting a wide range of diseases, especially cancer and inflammatory disorders. This review reveals the nuanced interactions between saponins and critical signaling pathways, including system Xc--GSH-GPX4, Nrf2, p53, and mTOR. These interactions highlight the dual capacity of saponins to modulate ferroptosis, thereby offering fresh perspectives for therapeutic intervention. CONCLUSIONS The insights garnered from this review significantly advance our comprehension of the dynamic relationship between saponins and ferroptosis. By shedding light on these mechanisms, this work sets the stage for leveraging these insights in the creation of pioneering approaches to disease treatment, marking a significant stride in the evolution of therapeutic modalities.
Collapse
Affiliation(s)
- Min Ouyang
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| | - Jianhua Wu
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| | - Xizhuo Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Changfu Liu
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| | - Dan Zhou
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| |
Collapse
|
5
|
Wang T, Wang X, Ren W, Sun Z, Zhang Y, Wu N, Diao H. Cardiomyocyte proliferation: Advances and insights in macrophage-targeted therapy for myocardial injury. Genes Dis 2025; 12:101332. [PMID: 39935606 PMCID: PMC11810708 DOI: 10.1016/j.gendis.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/20/2024] [Indexed: 02/13/2025] Open
Abstract
In the mammalian heart, cardiomyocytes undergo a transient window of proliferation that leads to regenerative impairment, limiting cardiomyocyte proliferation and myocardial repair capacity. Cardiac developmental patterns exacerbate the progression of heart disease characterized by myocardial cell loss, ultimately leading to cardiac dysfunction and heart failure. Myocardial infarction causes the death of partial cardiomyocytes, which triggers an immune response to remove debris and restore tissue integrity. Interestingly, when transient myocardial injury triggers irreversible loss of cardiomyocytes, the subsequent macrophages responsible for proliferation and regeneration have a unique immune phenotype that promotes the formation of pre-existing new cardiomyocytes. During mammalian regeneration, mononuclear-derived macrophages and self-renewing resident cardiac macrophages provide multiple cytokines and molecular signals that create a regenerative environment and cellular plasticity capacity in postnatal cardiomyocytes, a pivotal strategy for achieving myocardial repair. Consistent with other human tissues, cardiac macrophages originating from the embryonic endothelium produce a hierarchy of contributions to monocyte recruitment and fate specification. In this review, we discuss the novel functions of macrophages in triggering cardiac regeneration and repair after myocardial infarction and provide recent advances and prospective insights into the phenotypic transformation and heterogeneous features involving cardiac macrophages. In conclusion, macrophages contribute critically to regeneration, repair, and remodeling, and are challenging targets for cardiovascular therapeutic interventions.
Collapse
Affiliation(s)
- Tao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Weibin Ren
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Hongyan Diao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
6
|
Zhang Y, Chen H, Chen Q, Zaitoun M, Cheng Y, Ge J, Feng Q. Acrolein-Triggered Ferroptosis and Protection by Intermittent Fasting via the AMPK/NRF2-CLOCK/BMAL1 Pathway. TOXICS 2025; 13:369. [PMID: 40423448 DOI: 10.3390/toxics13050369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025]
Abstract
Environmental pollution significantly exacerbates various diseases, particularly those affecting the cardiovascular and respiratory systems. Our previous studies have shown that acrolein, an environmental pollutant, promotes atherosclerosis by downregulating the circadian clock genes (CLOCK/BMAL1) and disrupting circadian rhythm. We have also found that intermittent fasting (IF), closely linked to the circadian clock, may mitigate atherosclerosis induced by acrolein. Ferroptosis, a newly identified form of regulated cell death, is associated with the acceleration of atherosclerotic development, but its relationship with the circadian clock is not well understood. In this study, we explored the potential of IF to alleviate ferroptosis by modulating the circadian clock. Our in vivo experiments revealed that IF reversed ferroptosis and upregulated CLOCK/BMAL1 in APOE-/- mice. In human umbilical vein endothelial cells (HUVECs), we discovered that acrolein-induced ferroptosis leads to cell death, while short-term starvation (STS, IF cell model) reversed this effect. Acrolein also suppressed the expression of AMP-activated protein kinase (AMPK), nuclear factor erythroid 2-related factor 2 (NRF2), and CLOCK/BMAL1, which were restored by subsequent STS treatments. Additionally, the overexpression of CLOCK/BMAL1 mitigated ferroptosis, consistent with findings from CLOCK gene knockout experiments. Notably, CLOCK/BMAL1 and AMPK/NRF2 were found to be mutually regulated. Concurrently, the AMPK and NRF2 signaling pathways may be interdependent and act in concert. In conclusion, our findings suggest that IF modulates the CLOCK/BMAL1-AMPK/NRF2 pathway to alleviate acrolein-induced ferroptosis, offering a potential strategy to address health issues related to environmental pollution.
Collapse
Affiliation(s)
- Yuandie Zhang
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Hong Chen
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Qianfeng Chen
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Margaret Zaitoun
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Ying Cheng
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Jierong Ge
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Qing Feng
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
7
|
He L, Chen S, Zhu X, He F. Gamma-glutamyl transferase to high-density lipoprotein cholesterol ratio: A valuable predictor of coronary heart disease incidence. Nutr Metab Cardiovasc Dis 2025; 35:103775. [PMID: 39638678 DOI: 10.1016/j.numecd.2024.10.013] [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: 06/02/2024] [Revised: 09/25/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND AIM Existing studies have found that serological markers for predicting coronary heart disease (CHD) have relatively low predictive value for the severity of coronary arteries and the types of CHD. GGT to HDL-C ratio (GHR) has been shown to be associated with T2DM and non-alcoholic fatty liver disease. Therefore, we explore the relationship among GHR, CHD and its subgroups. METHODS AND RESULTS The study retrospectively analyzed 2703 participants from August 2022 to August 2023. The patients were divided into CHD group (N = 1911) and control group (N = 792) according to the diagnostic criteria of CHD. Adjustments for all covariates found that GHR was an independent risk factor for CHD (OR: 1.025, 95 % CI 1.016-1.033) and had the highest AUC of 0.767 (95 % CI 0.744-0.790) in identifying CHD. Additionally, GHR was significantly associated with multi-vessel CHD (OR: 1.018, 95 % CI 1.012-1.023) and showed excellent diagnostic capability for patients with multi-vessel CHD (AUC: 0.638). Moreover, compared with chronic coronary syndromes (CCS) and unstable angina (UA) groups, the level of GHR was significantly increased in acute myocardial infarction (AMI) (ST elevation myocardial infarction and Non-ST elevation myocardial infarction) group (P < 0.05). GHR had the higher AUC in STMETI [0.819 (95 % CI 0.796-0.854)] and NASTEMI [0.792 (95 % CI 0.766-0.816)] than the CCS and UA groups. CONCLUSIONS Our study analyses found that GHR is an independent risk factor for CHD and can predict the severity of coronary artery stenosis. Moreover, GHR has a high predictive value for AMI than CCS and UA in CHD patients.
Collapse
Affiliation(s)
- Li He
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China.
| | - Sisi Chen
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China
| | - Xuan Zhu
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China
| | - Fang He
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China
| |
Collapse
|
8
|
Zhang Y, Long T, Wei B, Zhou H, Yin X, Chen Z, Di Fazio P, Li W, Zhou H. A bibliometric analysis of the literature published on autophagy, ferroptosis, necroptosis, and pyroptosis in cardiovascular disease from 2009 to 2023. J Thorac Dis 2025; 17:2537-2562. [PMID: 40400924 PMCID: PMC12090173 DOI: 10.21037/jtd-2025-682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 05/23/2025]
Abstract
Background Programmed cell death (PCD) plays a pivotal role in the development and progression of cardiovascular disease (CVD), which remains the leading cause of mortality worldwide. Among the various types of PCD, autophagy, ferroptosis, necroptosis, and pyroptosis have garnered increasing attention due to their involvement in inflammation, oxidative stress, and cardiomyocyte survival. Although numerous studies have explored the underlying mechanisms of these pathways, their therapeutic potential in clinical practice remains limited. With the rapid growth of publications in this field, a comprehensive understanding of research trends and influential studies is essential to guide future investigations. This study aimed to characterize the progress and research hotspots of autophagy in CVD, ferroptosis in CVD, necroptosis in CVD, and pyroptosis in CVD through a bibliometric analysis to provide a comprehensive overview of PCD in CVD. Methods Publications from January 1, 2009, to December 31, 2023, were analyzed using the "bibliometrix" R package to assess research output, key contributors, and influential journals in each field. Results For the topic of autophagy in CVD, 6,426 articles published by 4,891 institutions from 90 countries/regions were retrieved. For the topic of necroptosis in CVD, 393 articles from 616 organizations in 53 countries/regions were retrieved. For the topic of pyroptosis in CVD, 640 publications from 754 institutions in 48 countries/regions were retrieved. Finally, for the topic of ferroptosis in CVD, 687 articles from 827 institutions in 49 countries/regions were retrieved. Key contributors included Adriana A (22 publications on necroptosis), Ge J, and Ye B (8 publications each on pyroptosis), and Ren J (lead contributor in autophagy and ferroptosis, with 120 and 10 publications, respectively). The most frequently co-cited journals were Cell, Nature, Free Radical Biology and Medicine, and the Journal of Biological Chemistry. Conclusions This bibliometric analysis highlights the growing interest in PCD in CVD research, with autophagy and pyroptosis being the central themes. Future studies should examine therapeutic strategies targeting ferroptosis and necroptosis to improve CVD treatment. The findings provide a roadmap for researchers to navigate emerging research hotspots and foster interdisciplinary collaboration.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Tianyi Long
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bo Wei
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Huan Zhou
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xinhai Yin
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhangrong Chen
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Pietro Di Fazio
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| | - Wei Li
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Haiyan Zhou
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| |
Collapse
|
9
|
Jiang Y, Zhou HZ, Zhang JX, Li KQ, Wang JH. LINC00926, Regulated by TCF12, Modulates the Ubiquitination of GPX4 to Regulate Ferroptosis by Interacting with STUB1 in HUVECs. Mol Biotechnol 2025:10.1007/s12033-025-01441-5. [PMID: 40295445 DOI: 10.1007/s12033-025-01441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
LINC00926 has been identified as an upregulated lncRNA in patients with coronary heart disease (CHD) through high-throughput sequencing. This study aimed to explore the biological role of LINC00926 in vascular endothelial cell ferroptosis and its underlying mechanisms. For in vitro experiments, HUVECs were exposed to hypoxic conditions. Our results showed an upregulation of LINC00926 expression, a decrease in GPX4 and GSH levels, and an increase in MDA and ROS levels in hypoxia-treated HUVECs. Furthermore, the ferroptosis inhibitor (ferrostatin-1) reversed the decrease in cell viability induced by hypoxia, suggesting that hypoxia treatment triggered GPX4-mediated ferroptosis in HUVECs. These variations were further exacerbated when LINC00926 was overexpressed, but were partially mitigated when LINC00926 was silenced. Notably, LINC00926 had no effect on GPX4 mRNA levels. Our data proved that LINC00926 modulated the ubiquitination and degradation of GPX4 via STUB1, thereby promoting hypoxia-induced HUVEC ferroptosis. Additionally, ChIP and luciferase reporter gene assays confirmed that TCF12 protein enhanced the transcriptional activity of LINC00926 promoter, hinting TCF12 is an upstream regulator of LINC00926. Besides, LINC00926 also enhanced the stability of TCF12 mRNA to promote TCF12 expression. Moreover, TCF12 acted as a regulator of ferroptosis in hypoxia-induced HUVECs. Finally, rescue experiments determined the role of the TCF12/LINC00926/GPX4 axis in ferroptosis of HUVECs upon hypoxic stimulation. In conclusion, this study demonstrated that the TCF12/LINC00926/GPX4 axis plays a regulatory role in hypoxia-induced ferroptosis of HUVECs, offering a promising target for the treatment of CHD.
Collapse
Affiliation(s)
- Yong Jiang
- School of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin, 132013, Jilin, China.
| | - Han-Zhu Zhou
- School of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin, 132013, Jilin, China
| | - Jun-Xuan Zhang
- School of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin, 132013, Jilin, China
| | - Kai-Qi Li
- School of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin, 132013, Jilin, China
| | - Jia-He Wang
- School of Clinical Medicine, Jilin Medical University, Jilin, 132013, China
| |
Collapse
|
10
|
Liu Y, Yu Z, Lu Y, Liu Y, Chen L, Li J. Progress in the study of the mechanism of ferroptosis in coronary heart disease and clinical intervention strategies. Front Cardiovasc Med 2025; 12:1545231. [PMID: 40308274 PMCID: PMC12040834 DOI: 10.3389/fcvm.2025.1545231] [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: 12/14/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Coronary heart disease (CHD), a serious cardiovascular condition with complex and diverse pathogenesis, has recently seen increased attention to the role of ferroptosis-a novel iron-dependent form of programmed cell death. This review synthesizes current research on ferroptosis mechanisms in CHD and emerging clinical intervention strategies. Ferroptosis is characterized by dysregulated iron metabolism, lipid peroxidation, and reactive oxygen species (ROS) accumulation, processes intimately linked to CHD pathophysiology. Under ischemic and hypoxic conditions commonly seen in coronary artery disease (CAD), cardiomyocytes become particularly susceptible to ferroptosis, resulting in cellular dysfunction and diminished cardiac performance. Mechanistic studies have revealed that altered expression of iron metabolism-related proteins (including GPX4, FTH1, TfR1, and HO-1), accumulation of lipid peroxidation products, and disruption of antioxidant defense systems (particularly the Nrf2/GPX4 pathway) are central to ferroptosis progression in cardiac tissue. Clinically, both specific ferroptosis inhibitors (such as Ferrostatin-1) and traditional medicine components (such as Puerarin) have emerged as promising therapeutic candidates, showing cardioprotective effects in experimental models. However, research into ferroptosis mechanisms in CHD remains in its early stages, with significant questions regarding its relationship with other cell death pathways and the clinical efficacy of ferroptosis-targeting interventions requiring further investigation. Future research directions should include in-depth mechanistic exploration and the development of more effective, safer clinical interventions targeting the ferroptosis pathway in cardiovascular disease.
Collapse
Affiliation(s)
- Yingzhi Liu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Zixuan Yu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Yuwen Lu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Yue Liu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Lingli Chen
- Hunan Key Laboratory of Pathogeny Biology of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jie Li
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
11
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
12
|
Shen Y, Zhang C, Jiang X, Li X, Chen B, Jiang W. Capsiate attenuates atherosclerosis by activating Nrf2/GPX4 pathway and reshaping the intestinal microbiota in ApoE -/- mice. Microbiol Spectr 2025; 13:e0315524. [PMID: 40029381 PMCID: PMC11960139 DOI: 10.1128/spectrum.03155-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
Atherosclerosis (AS) is the basis of cardiovascular diseases (CVDs) and remains the major contributor to death worldwide. Capsiate is derived from sweet pepper fruit and exhibits numerous pharmacological activities. The objective of this study was to elucidate the protective role of capsiate in atherosclerosis by examining its effect and the underlying regulatory pathways. Here, we showed that capsiate treatment alleviates atherosclerosis in atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice. We found that capsiate effectively reduced the plaque area and body weight compared to the Model group. Capsiate inhibited inflammatory response by downregulating phosphoinositide 3-kinase/protein kinase B/nuclear factor-κB pathway. Additionally, further investigation indicated that capsiate could regulate lipid levels in mice via reducing the expressions of 3-hydroxy-3-methylglutaryl coenzyme A reductase and low-density lipoprotein receptor, and increasing the expression of recombinant cytochrome P450 7A1. Furthermore, capsiate effectively activated transient receptor potential vanilloid subfamily member 1 in ApoE-/- mice fed a high-fat diet. The microbial sequencing demonstrated capsiate administration significantly regulated the gut microbiota disturbance and increased some beneficial bacterial (Lachnospiraceae NK4A136 group) levels in ApoE-/- mice. Human umbilical vein endothelial cells (HUVECs) were exposed to oxidized low-density lipoprotein (ox-LDL) to stimulate atherosclerotic endothelial damage in vitro. Our study revealed that capsiate inhibited ox-LDL-induced HUVECs injury and inflammation. We further investigated the effects of capsiate on ferroptosis in vivo and in vitro; it was found that capsiate exhibited anti-ferroptosis through regulating nuclear factor erythroid 2-related factor 2/glutathione peroxidase 4 pathway. Interestingly, ML385 reversed the anti-ferroptosis effect of capsiate in HUVECs. Taken together, our findings suggest a promising use of small-molecule drug capsiate for the treatment of AS and related CVDs. IMPORTANCE Capsiate has been found to inhibit fat accumulation, promote energy metabolism, and exhibit anti-inflammatory and antioxidative properties. However, there has still been no study on the ferroptosis and gut microbiota of capsiate in atherosclerosis (AS) mouse models. Our study is the first to report on the reshaping of the structure of the gut microbiota by capsiate in AS, and to explore the potential mechanism underlying the improvement of AS. In this study, we demonstrated that capsiate could effectively alleviate high-fat diet-induced AS in apolipoprotein E-deficient mice by inhibiting inflammatory response, improving serum lipid profiles, activating transient receptor potential vanilloid subfamily member 1 pathway, and suppressing ferroptosis. Moreover, the study reported the potential of gut microbiota as mediators of capsiate therapy for AS in animal models. Therefore, these findings may provide robust experimental support for the clinical use of capsiate for AS treatment.
Collapse
Affiliation(s)
- Yongbin Shen
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanqi Zhang
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Jiang
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianwei Li
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Chen
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weiliang Jiang
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Dabravolski SA, Churov AV, Beloyartsev DF, Kovyanova TI, Lyapina IN, Sukhorukov VN, Orekhov AN. The role of NRF2 function and regulation in atherosclerosis: an update. Mol Cell Biochem 2025:10.1007/s11010-025-05233-y. [PMID: 40025257 DOI: 10.1007/s11010-025-05233-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
Atherosclerosis, a chronic inflammatory disease of the arteries, remains a leading cause of cardiovascular morbidity and mortality worldwide. This review examines the molecular mechanisms underlying NRF2 role in atherosclerosis, focusing on the recently defined intricate interplay between autophagy, the nuclear factor erythroid 2-related factor 2 (NRF2) pathway, microRNAs (miRNAs), and genes regulating NRF2 with atheroprotective effects. The NRF2/autophagy axis emerges as a critical regulator of cellular responses to oxidative stress and inflammation in atherosclerosis, with key players including Heat Shock Protein 90 (HSP90), Neuropeptide Y (NPY), and Glutaredoxin 2 (GLRX2). MiRNAs are identified as potent regulators of gene expression in atherosclerosis, impacting NRF2 signalling and disease susceptibility. Additionally, genes such as Prenyl diphosphate synthase subunit 2 (PDSS2), Sulfiredoxin1 (Srxn1), and Isocitrate dehydrogenase 1 (IDH1) are implicated in NRF2-dependent atheroprotective pathways. Future research directions include elucidating the complex interactions between these molecular pathways, evaluating novel therapeutic targets in preclinical and clinical settings, and addressing challenges related to drug delivery and patient heterogeneity. Despite limitations, this review underscores the potential for targeted interventions aimed at modulating NRF2/autophagy signalling and miRNA regulatory networks to mitigate atherosclerosis progression and improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, 2161002, Karmiel, Israel.
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A. V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Tatiana I Kovyanova
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Irina N Lyapina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, Kemerovo, Russia, 650002
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| |
Collapse
|
14
|
Wang Z, Sun W, Zhang K, Ke X, Wang Z. New insights into the relationship of mitochondrial metabolism and atherosclerosis. Cell Signal 2025; 127:111580. [PMID: 39732307 DOI: 10.1016/j.cellsig.2024.111580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Atherosclerotic cardiovascular and cerebrovascular diseases are the number one killer of human health. In view of the important role of mitochondria in the formation and evolution of atherosclerosis, our manuscript aims to comprehensively elaborate the relationship between mitochondria and the formation and evolution of atherosclerosis from the aspects of mitochondrial dynamics, mitochondria-organelle interaction (communication), mitochondria and cell death, mitochondria and vascular smooth muscle cell phenotypic switch, etc., which is combined with genome, transcriptome and proteome, in order to provide new ideas for the pathogenesis of atherosclerosis and the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Zexun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China
| | - Wangqing Sun
- Department of Radiology, Yixing Tumor Hospital, Yixing 214200, China
| | - Kai Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
15
|
Sil R, Chakraborti AS. Major heme proteins hemoglobin and myoglobin with respect to their roles in oxidative stress - a brief review. Front Chem 2025; 13:1543455. [PMID: 40070406 PMCID: PMC11893434 DOI: 10.3389/fchem.2025.1543455] [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: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress is considered as the root-cause of different pathological conditions. Transition metals, because of their redox-active states, are capable of free radical generation contributing oxidative stress. Hemoglobin and myoglobin are two major heme proteins, involved in oxygen transport and oxygen storage, respectively. Heme prosthetic group of heme proteins is a good reservoir of iron, the most abundant transition metal in human body. Although iron is tightly bound in the heme pocket of these proteins, it is liberated under specific circumstances yielding free ferrous iron. This active iron can react with H2O2, a secondary metabolite, forming hydroxyl radical via Fenton reaction. Hydroxyl radical is the most harmful free radical among all the reactive oxygen species. It causes oxidative stress by damaging lipid membranes, proteins and nucleic acids, activating inflammatory pathways and altering membrane channels, resulting disease conditions. In this review, we have discussed how heme-irons of hemoglobin and myoglobin can promote oxidative stress under different pathophysiological conditions including metabolic syndrome, diabetes, cardiovascular, neurodegenerative and renal diseases. Understanding the association of heme proteins to oxidative stress may be important for knowing the complications as well as therapeutic management of different pathological conditions.
Collapse
Affiliation(s)
| | - Abhay Sankar Chakraborti
- Department of Biophysics, Molecular Biology and Bioinformatics, University College of Science, University of Calcutta, Kolkata, India
| |
Collapse
|
16
|
Zhu Z, Wu X, Zhang J, Zhu M, Tian M, Zhao P. Advances in understanding ferroptosis mechanisms and their impact on immune cell regulation and tumour immunotherapy. Discov Oncol 2025; 16:153. [PMID: 39930297 PMCID: PMC11811334 DOI: 10.1007/s12672-025-01911-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Ferroptosis is a novel mode of iron-dependent non-apoptotic cell death that occurs mainly due to excessive accumulation of lipid peroxides. Numerous studies in recent years have shown that ferroptosis plays a vital role in the organism and has important interactions with immune cells. Ferroptosis has been shown to have great potential in tumour therapy through studying its mechanism of action. In addition, ferroptosis plays a major role in many types of tumour cells that can potently suppress the tumourigenesis and metastasis, provide a basis for the treatment of many malignant tumour diseases and become a novel therapeutic modality of antitumour immunity in the clinic. Current tumour immunotherapy for ferroptosis in combination with other conventional oncological modalities is not well elaborated. In this paper, we mainly discuss the connection of ferroptosis with immune cells and their mediated tumour immunotherapy in order to provide a better theoretical basis and new thinking about ferroptosis mediated antitumour immunity.
Collapse
Affiliation(s)
- Zengjun Zhu
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, China
| | - Xuanxuan Wu
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, China
| | - Jian Zhang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255036, China
| | - Minghui Zhu
- Department of Clinical Laboratory, Huantai County People's Hospital, Zibo, 256400, China
| | - Maojin Tian
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255036, China.
| | - Peiqing Zhao
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255036, China.
| |
Collapse
|
17
|
Xi C, Zhou J, Zheng X, Fu X, Xie M. Sodium aescinate-induced hepatotoxicity via ATF4/GSH/GPX4 axis-mediated ferroptosis. Sci Rep 2025; 15:1141. [PMID: 39774712 PMCID: PMC11706965 DOI: 10.1038/s41598-024-79723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sodium aescinate (SA), a natural plant extract with various bioactivities, is widely used to treat oedema and inflammation in clinics. However, adverse events, including liver injury, kidney injury, and phlebitis, have been reported in patients with SA in recent years. In this study, we used BALB/c mice and L02 cells to evaluate the role of ferroptosis in SA-induced liver injury. SA significantly increased AST, ALT, MDA and Fe2+, decreased GSH levels, and induced pathological changes in the liver in vivo. SA also reduced the viability of L02 cells and induced LDH release, intracellular cysteine reduction, GSH depletion, iron accumulation, ROS production, and lipid peroxidation, indicating that SA causes ferroptosis. In addition, SA inhibited transcriptional activity of activating transcription factor 4 (ATF4) and subsequently reduced the expression of the downstream genes xCT (solute carrier family 7a member 11, SLC7A11) and Cystathionine gamma-lyase (CTH) which play vital roles in GSH biosynthesis. Interestingly, the cytotoxic effects of SA were effectively attenuated by ATF4 overexpression, while they were significantly aggravated by ATF4 silencing. These results revealed that SA triggers hepatocyte ferroptosis by inhibiting the activity of ATF4, which causes an oxidative imbalance.
Collapse
Affiliation(s)
- Chen Xi
- Pharmaceutical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China.
| | - Xin Zheng
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Xiaoyi Fu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| |
Collapse
|
18
|
Ni Y, Liu L, Jiang F, Wu M, Qin Y. JAG1/Notch Pathway Inhibition Induces Ferroptosis and Promotes Cataractogenesis. Int J Mol Sci 2025; 26:307. [PMID: 39796164 PMCID: PMC11719987 DOI: 10.3390/ijms26010307] [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: 11/21/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Cataracts remain the leading cause of visual impairment worldwide, yet the underlying molecular mechanisms, particularly in age-related cataracts (ARCs), are not fully understood. The Notch signaling pathway, known for its critical role in various degenerative diseases, may also contribute to ARC pathogenesis, although its specific involvement is unclear. This study investigates the role of Notch signaling in regulating ferroptosis in lens epithelial cells (LECs) and its impact on ARC progression. RNA sequencing of anterior lens capsule samples from ARC patients revealed a significant downregulation of Notch signaling, coupled with an upregulation of ferroptosis-related genes. Notch1 expression decreased, while ferroptosis markers increased in an age-dependent manner. In vitro, upregulation of Notch signaling alleviated ferroptosis by decreasing ferritin heavy chain 1 (FTH1) and p53 levels while enhancing the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11). Conversely, inhibition of Notch signaling exacerbated ferroptosis, as evidenced by reduced Nrf2, GPX4, and SLC7A11 expression. These findings suggest that downregulation of Notch signaling promotes ferroptosis in LECs by impairing the Nrf2/GPX4 antioxidant pathway, thereby contributing to ARC development. This study offers new insights into ARC pathogenesis and highlights the Notch signaling pathway as a potential therapeutic target for preventing or mitigating ARC progression.
Collapse
Affiliation(s)
- Yan Ni
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; (Y.N.) (L.L.); (F.J.)
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510033, China
| | - Liangping Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; (Y.N.) (L.L.); (F.J.)
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510033, China
| | - Fanying Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; (Y.N.) (L.L.); (F.J.)
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510033, China
| | - Mingxing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; (Y.N.) (L.L.); (F.J.)
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510033, China
| | - Yingyan Qin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; (Y.N.) (L.L.); (F.J.)
- Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510033, China
| |
Collapse
|
19
|
Zeng Y, Fu S, Xia Y, Meng G, Xu X. Itchy E3 Ubiquitin Ligase-Mediated Ubiquitination of Ferritin Light Chain Contributes to Endothelial Ferroptosis in Atherosclerosis. Int J Mol Sci 2024; 25:13524. [PMID: 39769287 PMCID: PMC11677933 DOI: 10.3390/ijms252413524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
This research seeks to investigate the function and fundamental mechanisms of Itchy E3 ubiquitin ligase (ITCH), a HECT (homologous to E6AP carboxyl terminus)-type E3 ubiquitin ligase, in endothelial ferroptosis, particularly in the context of atherosclerosis, which has been underexplored. The levels of ITCH protein in the aortas of mice with atherosclerosis were analyzed. Constructs for ITCH RNA interference were generated and introduced into human aortic endothelial cells (HAECs). The findings indicated that ITCH protein expression was elevated in atherosclerotic mice and HAECs exposed to oxidized low-density lipoprotein (ox-LDL). ITCH downregulation significantly mitigated ox-LDL-induced endothelial injury and dysfunction. Reducing ITCH expression inhibited ox-LDL-induced endothelial ferroptosis. This study also revealed that ITCH mediates ox-LDL-induced ubiquitin-dependent degradation of ferritin light chain (FTL) in HAECs. The protective impact of ITCH knockdown against ox-LDL-induced ferroptosis and endothelial injury was reversed by FTL siRNA. Additionally, in vivo experiments showed that inhibiting ITCH reduced atherosclerosis progression and reversed ferroptosis in the aorta, with an associated increase in FTL protein expression in the aortas of mice. This study demonstrates that ITCH interacts with and regulates the stability of the FTL protein via the ubiquitin-proteasome system, contributing to ox-LDL-induced ferroptosis and endothelial cell dysfunction. Targeting components of the ITCH-FTL pathway holds potential as a therapeutic strategy against atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Xiaole Xu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, China; (Y.Z.); (S.F.); (Y.X.); (G.M.)
| |
Collapse
|
20
|
Zeng T, Lei GL, Yu ML, Zhang TY, Wang ZB, Wang SZ. The role and mechanism of various trace elements in atherosclerosis. Int Immunopharmacol 2024; 142:113188. [PMID: 39326296 DOI: 10.1016/j.intimp.2024.113188] [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: 06/28/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Atherosclerosis is a slow and complex disease that involves various factors, including lipid metabolism disorders, oxygen-free radical production, inflammatory cell infiltration, platelet adhesion and aggregation, and local thrombosis. Trace elements play a crucial role in human health. Many trace elements, especially metallic ones, not only maintain the normal functions of organs but also participate in basic metabolic processes. The latest studies have revealed a close correlation between trace elements and the occurrence and progression of atherosclerosis. The imbalance of these trace elements can induce atherosclerosis or accelerate its progression through various mechanisms, which poses a significant threat to human health. Therefore, exploring the specific mechanism of trace elements on atherosclerosis is highly significant. In this review, we summarized the roles and mechanisms of iron, copper, zinc, magnesium, and selenium homeostasis and imbalance in atherosclerosis development, in order to identify novel targets and therapeutic strategies for treating atherosclerosis.
Collapse
Affiliation(s)
- Tao Zeng
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Guan-Lan Lei
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Mei-Ling Yu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Ting-Yu Zhang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| |
Collapse
|
21
|
Chen H, Wu H, Wang Q, Zhang H. IFIT2 mediates iron retention and cholesterol efflux in atherosclerosis. Int Immunopharmacol 2024; 142:113131. [PMID: 39276454 DOI: 10.1016/j.intimp.2024.113131] [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: 06/20/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Abnormalities in iron and lipid metabolism are recognized as key contributors to atherosclerosis (AS). Therefore, this study proposes to characterize the biomarker related to iron and lipid metabolism in AS using bioinformatics, animal, and cell experiments. METHODS The limma package was utilized to identify differentially expressed genes (DEGs) in GSE70126 and GSE70619 datasets, and biomarkers were screened using enrichment analysis and PPI networks. IFIT2 was knocked down using shRNA lentivirus in a high fat diet (HFD)-induced APOE-/- AS model to investigate its effects of IFIT2 on the pathology, iron retention, and lipid accumulation. Iron storage-related and cholesterol efflux-related proteins were evaluated following exogenous modulation of IFIT2 expression in ox-LDL-induced foamy macrophages. RESULTS Compared to non-foamy macrophages from the aorta, 189 and 4152 DEGs were identified in foamy macrophages within the GSE70126 and GSE70619 datasets, respectively. Moreover, intersecting DEGs may modulate immune responses, cell adhesion, vascular permeability, and oxidative stress through NF-kappa B, Wnt, TNF and HIF-1 signaling pathways. Notably, IFIT2 was significantly upregulated in foamy macrophages and AS models. In vivo, IFIT2 co-localized with foamy macrophages, and its knockdown led to reductions in plasma lipid levels, plaque area, immune infiltration, iron retention, and lipid accumulation. In vitro, IFIT2 knockdown alleviated the ox-LDL-induced increase in iron storage-related proteins (Ferritin-L and Ferritin-H) and iron (Fe2+ and Fe3+) in foamy macrophages. Furthermore, IFIT2 knockdown reduced lipid accumulation and upregulated cholesterol efflux-related proteins (PPARγ, LXRα, ABCA1, and ABCG1) in foamy macrophages. CONCLUSION IFIT2 knockdown attenuates iron retention and lipid accumulation in AS plaques, and facilitated cholesterol efflux from foamy macrophages via the PPARγ/LXRα/ABCA1-ABCG1 pathway.
Collapse
Affiliation(s)
- Haoqiang Chen
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China
| | - Haiyan Wu
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China; Faculty of Life Science and Technology, Kunming University of Science and Technology, No.727 Jingming South Road, Kunming 650500, P.R.China
| | - Qian Wang
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China.
| | - Hong Zhang
- Department of Cardiovascular Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, Yunnan 650032, P.R.China.
| |
Collapse
|
22
|
Liu LY, He SJ, Luo J, Huang JK, Yuan JX, Yuan CJ, Zhang JL. Network pharmacology, molecular docking and experimental study on the mechanism of Curcumin's anti-ferroptosis in melanocytes. Biochem Biophys Res Commun 2024; 736:150871. [PMID: 39461013 DOI: 10.1016/j.bbrc.2024.150871] [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: 08/23/2024] [Revised: 10/02/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Ferroptosis is a form of regulated nonapoptotic cell death associated with iron-dependent lipid peroxidation, closely associated with Vitiligo. Although the impact of Curcumin (Cur), a polyphenolic compound derived from the plant Curcuma longa Linn, on vitiligo has been established, the specific role and potential mechanistic pathways through which Cur modulates ferroptosis in vitiligo remain elusive. In this study, the critical targets and potential mechanisms of Cur in treating vitiligo were predicted by network pharmacology and molecule docking. Then, the effects of Cur on Erastin-induced ferroptosis were investigated in melanocytes induced by Erastin in vitro. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of Cur acting on Vitiligo found that these intersection genes are associated with the vitiligo oxidative stress pathway, including nuclear factor erythroid 2-related factor 2(Nrf2)/Heme Oxygenase 1(HO-1) signaling pathway. Further molecular docking shows that Cur has a good binding effect with Nrf2(the binding energy of Cur and Nrf2 protein is -6 kcal/mol). Through the CCK8 assay, showed that 10 μM Cur treatment 24 h after Erastin significantly improved cell viability In vitro. Then we found that Erastin induced cell death, ROS production, the mitochondrial membrane potential(MMP) decreased, Superoxide dismutase (SOD) and Glutathione (GSH) levels reduced, Malonaldehyde (MDA) and iron ion accumulation in melanocytes. In addition, the expression of glutathione peroxidase 4(GPX4) mRNA and protein was inhibited, while the expression of acyl-CoA synthetase long-chain family member 4(ACSL4), Transferrin Receptor Protein 1(TFR1) mRNA and protein was increased. However, the damage induced by Erastin was significantly relieved by Cur and Fer-1 treatment. Mechanistically, Cur treatment significantly promoted nuclear translocation of transcriptional factor Nrf2 and HO-1 expression. Interestingly, pretreatment with ML385, a selective Nrf2 inhibitor, counteracted anti-ferroptosis effects induced by Cur treatment. Taken together, these results demonstrate that Cur inhibits ferroptosis by regulating the Nrf2/HO-1 pathway to protect melanocytes.
Collapse
Affiliation(s)
- Lyu-Ye Liu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 300000, China
| | - Si-Jia He
- Department of Dermatology, Tianjin Public Security Hospital, Tianjin, 300000, China
| | - Jing Luo
- Department of Pathogen Biology and Immunology, Basic Medical College, Tianjin Medical University, Tianjin, 300000, China
| | - Jun-Kai Huang
- Department of Pathogen Biology and Immunology, Basic Medical College, Tianjin Medical University, Tianjin, 300000, China
| | - Jin-Xiang Yuan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 300000, China
| | - Chuan-Jian Yuan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 300000, China
| | - Jun-Ling Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300000, China.
| |
Collapse
|
23
|
Min XL, Lin SX, Zhao XH, Zhao Q, Li YF, Li XH, Liu XY, Cao Y, Sun YL, Zeng Y. Mechanisms of METTL14-Mediated m6A Modification in Promoting Iron Overload-Induced Lipid Peroxidative Damage in Vascular Endothelial Cells to Aggravate Atherosclerosis. J Biochem Mol Toxicol 2024; 38:e70066. [PMID: 39588760 DOI: 10.1002/jbt.70066] [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: 04/24/2024] [Revised: 10/22/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024]
Abstract
Atherosclerosis (AS) is a chronic multifactorial disease with damage to vascular endothelial cells (VECs). This study sought to delve into the mechanism of methyltransferase-like 14 (METTL14) in iron overload-induced lipid peroxidative damage in AS. AS mouse model and cell model were established. Levels of METTL14/circRNA coded by the Arhgap12 (circARHGAP12)/Aspartate β-hydroxylase (ASPH) were determined. AS plaque area/lipid deposition/lipid metabolism in AS mice and iron overload in VECs were evaluated. N6-methyladenosine (m6A) level and METTL14 enrichment and human antigen R (HuR) in circARHGAP12 or ASPH were measured. The mRNA stability of circARHGAP12 or ASPH was analyzed. We observed that METTL14 was upregulated in AS mice. METTL14 downregulation reduced plaque area/lipid deposition/iron overload/peroxidative damage in AS mice. In cell models, METTL14 downregulation could VEC injury/iron overload/lipid peroxidative damage. Mechanically, METTL14 increased the stability and expression of circARHGAP12 through m6A modification, further stabilized ASPH mRNA, and promoted ASPH transcription by binding to HuR. Overexpression of circARHGAP12 or inhibition of ASPH averted the protective role of METTL14 downregulation against iron overload-induced peroxidative damage in AS. In conclusion, METTL14-mediated m6A modification upregulated circARHGAP12 and ASPH to aggravate overload-induced lipid peroxidative damage and facilitate AS progression.
Collapse
Affiliation(s)
- Xiao-Li Min
- Department of Cerebrovascular Diseases, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Si-Xian Lin
- Department of Cerebrovascular Diseases, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Hong Zhao
- Department of Cerebrovascular Diseases, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Zhao
- Department of Cerebrovascular Diseases, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yun-Fei Li
- Department of Cerebrovascular Diseases, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xu-Hui Li
- Department of Neurosurgery, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Yong Liu
- Department of Cardiology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi Cao
- Department of Cerebrovascular Diseases, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu-Long Sun
- Department of Neurology, Baoshan people's hospital, Baoshan, China
| | - Yong Zeng
- Department of Psychiatry, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
24
|
Elkammash A, Zaki A, Tawfik O, Gouda S. Ferroptosis: A Key Driver in Atherosclerosis Progression and Arterial Disease. Rev Cardiovasc Med 2024; 25:441. [PMID: 39742226 PMCID: PMC11683713 DOI: 10.31083/j.rcm2512441] [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: 06/19/2024] [Revised: 09/08/2024] [Accepted: 09/20/2024] [Indexed: 01/03/2025] Open
Abstract
Atherosclerosis (AS) is a growing global health epidemic and is the leading cause of cardiovascular health problems, including ischemic stroke, coronary artery disease, and peripheral vascular disease. Despite extensive research on the underlying mechanisms of AS, iron remains an under-investigated mediator in the atherosclerotic process. Iron's involvement in AS is primarily linked to the iron-induced programmed cell death process known as ferroptosis. Ferroptosis is initiated in endothelial cells when iron overload triggers the Fenton reaction, resulting in the production of reactive oxygen species (ROS) and lipid peroxides. This oxidative stress damages cellular components, ultimately leading to cell death. The review examines the role of iron overload and ferroptosis in the progression and instability of atherosclerotic plaques. Additionally, we explore the potential therapeutic roles of iron chelators and ROS scavengers in mitigating the adverse effects of ferroptosis. The findings indicate that ferroptosis contributes significantly to the progression and instability of atherosclerotic plaques by promoting oxidative damage and cellular dysfunction. Iron chelators and ROS scavengers have shown promise in reducing ferroptosis-induced damage in endothelial cells. These therapeutic agents can potentially stabilize atherosclerotic plaques and prevent the progression of AS. Ferroptosis is a critical yet under-explored pathway in the development and progression of atherosclerosis. Targeting iron-induced oxidative stress through iron chelation and ROS scavenging presents a promising therapeutic strategy for mitigating the adverse effects of ferroptosis on atherosclerotic plaque stability. Further research is needed to validate these therapeutic approaches and better understand the molecular mechanisms underlying ferroptosis in atherosclerosis.
Collapse
Affiliation(s)
- Amr Elkammash
- Department of Cardiology, Bristol Heart Institute, BS2 8HW Bristol, UK
| | - Abrar Zaki
- Department of General Medicine, Eastbourne District General Hospital, BN21 2UD East Sussex, UK
| | - Omar Tawfik
- Department of Cardiology, Bristol Heart Institute, BS2 8HW Bristol, UK
| | - Sherif Gouda
- Department of Cardiology, Royal Gwent Hospital, NP20 2UB Newport, UK
| |
Collapse
|
25
|
Lu Y, Xie X, Luo L. Ferroptosis crosstalk in anti-tumor immunotherapy: molecular mechanisms, tumor microenvironment, application prospects. Apoptosis 2024; 29:1914-1943. [PMID: 39008197 DOI: 10.1007/s10495-024-01997-8] [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] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Immunotherapies for cancer, specifically immune checkpoint inhibition (ICI), have shown potential in reactivating the body's immune response against tumors. However, there are challenges to overcome in addressing drug resistance and improving the effectiveness of these treatments. Recent research has highlighted the relationship between ferroptosis and the immune system within immune cells and the tumor microenvironment (TME), suggesting that combining targeted ferroptosis with immunotherapy could enhance anti-tumor effects. This review explores the potential of using immunotherapy to target ferroptosis either alone or in conjunction with other therapies like immune checkpoint blockade (ICB) therapy, radiotherapy, and nanomedicine synergistic treatments. It also delves into the roles of different immune cell types in promoting anti-tumor immune responses through ferroptosis. Together, these findings provide a comprehensive understanding of synergistic immunotherapy focused on ferroptosis and offer innovative strategies for cancer treatment.
Collapse
Affiliation(s)
- Yining Lu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xiaoting Xie
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
26
|
Wang R, Geng J. The melatonin-FTO-ATF4 signaling pathway protects granulosa cells from cisplatin-induced chemotherapeutic toxicity by suppressing ferroptosis. J Assist Reprod Genet 2024; 41:3503-3516. [PMID: 39388020 PMCID: PMC11707222 DOI: 10.1007/s10815-024-03276-6] [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: 07/09/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
PURPOSE In cisplatin-induced premature ovarian failure (POF) mice, granulosa cells showed a high level of ferroptosis. Previous research has indicated that the fat mass and obesity-associated protein/activating transcription factor 4 (FTO/ATF4) axis was involved in the regulation of ferroptosis. The purpose of this study was to explore the role of the FTO/ATF4 axis in cisplatin-induced ferroptosis in granulosa cell. METHODS The extent of ferroptosis was assessed by transmission electron microscopy (TEM) and ROS, GPX, GSH, and MDA assays. Western blotting was used to evaluate the protein expression levels of ferroptosis-related molecules. Ferroptosis activator and inhibitor were also used. RESULTS We found that ferroptosis increased in a concentration-dependent manner in cisplatin-induced injured granulosa cells, accompanied by the downregulation of FTO. In addition, gain- and loss-of-function studies showed that FTO affects ferroptosis in injured cells by regulating ATF4 expression. Ferrostatin-1 inhibited the effect of FTO downregulation on injured granulosa cells ferroptosis, and erastin reversed the protective effect of FTO on ferroptosis in injured granulosa cells. Finally, melatonin was used, and we found that melatonin reduced ferroptosis in cisplatin-induced injured granulosa cells by upregulating FTO expression. CONCLUSION Our study demonstrated that cisplatin induced granulosa cell ferroptosis by downregulating the expression of FTO. ATF4 was identified as a downstream target of FTO, and overexpression of ATF4 reversed the effects of decreased FTO on ferroptosis. Additionally, melatonin mitigates the cytotoxic effects of cisplatin by upregulating FTO expression. The melatonin-FTO-ATF4 signaling pathway plays a vital role in the treatment of cisplatin-induced POF.
Collapse
Affiliation(s)
- Rongli Wang
- 1Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China.
| | - Jing Geng
- 1Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
27
|
Zhu L, Zhang J, Fan W, Su C, Jin Z. Identification of iron metabolism-related genes in coronary heart disease and construction of a diagnostic model. Front Cardiovasc Med 2024; 11:1409605. [PMID: 39610972 PMCID: PMC11602506 DOI: 10.3389/fcvm.2024.1409605] [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: 03/30/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024] Open
Abstract
Background Coronary heart disease is a common cardiovascular disease, yferroptosiset its relationship with iron metabolism remains unclear. Methods Gene expression data from peripheral blood samples of patients with coronary heart disease and a healthy control group were utilized for a comprehensive analysis that included differential expression analysis, weighted gene co-expression network analysis, gene enrichment analysis, and the development of a logistic regression model to investigate the associations and differences between the groups. Additionally, the CIBERSORT algorithm was employed to examine the composition of immune cell types within the samples. Results Eight central genes were identified as being both differentially expressed and related to iron metabolism. These central genes are mainly involved in the cellular stress response. A logistic regression model based on the central genes achieved an AUC of 0.64-0.65 in the diagnosis of coronary heart disease. A higher proportion of M0 macrophages was found in patients with coronary heart disease, while a higher proportion of CD8T cells was observed in the normal control group. Conclusion The study identified important genes related to iron metabolism in the pathogenesis of coronary heart disease and constructed a robust diagnostic model. The results suggest that iron metabolism and immune cells may play a significant role in the development of coronary heart disease, providing a basis for further research.
Collapse
Affiliation(s)
| | | | | | | | - Zhi Jin
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
28
|
Jiang M, Zhao W, Wu L, Zhu G. Screening of m6A-associated ferroptosis-related genes in atherosclerosis based on WGCNA. Front Cardiovasc Med 2024; 11:1469805. [PMID: 39529974 PMCID: PMC11550986 DOI: 10.3389/fcvm.2024.1469805] [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: 07/24/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Background N6-methyladenosine (m6A) has been shown to mediate ferroptosis but its role in atherosclerosis (AS) is unclear. Methods Differentially expressed m6A-associated ferroptosis-related genes (DE-m6A-Ferr-RGs) were obtained using differential expression analysis and Pearson correlation analysis. Weighted gene co-expression network analysis (WGCNA) was also performed. The intersection of the module genes and the DE-m6A-Ferr-RGs were recorded as candidate m6A-Ferr-related signature genes. Finally, the m6A-Ferr-related signature genes were screened using least absolute shrinkage and selection operator (LASSO) analysis. Expression validation, receiver operating characteristic ( mapping, and immune correlation analysis were also performed based on the m6A-Ferr-related signature genes. The expression of m6A-Ferr-related signature genes was further validated using a real-time polymerase chain reaction (RT-qPCR). Results In total, 6,167 differentially expressed genes were intersected with 24 m6A- and 259 ferroptosis-related genes, respectively, resulting in 113 DE-m6A-Ferr-RGs obtained using Pearson's correlation analysis. The module genes obtained from the WGCNA and the 113 DE-m6A-Ferr-RGs were intersected to obtain 48 candidate m6A-Ferr-related signature genes. LASSO analysis was performed and six m6A-Ferr-related signature genes were screened. In addition, the area under the curve values of all six m6A-Ferr-related signature genes were greater than 0.7, indicating that they had potential diagnostic value. Furthermore, the RT-qPCR results revealed that the expression of SLC3A2, NOX4, and CDO1 was consistent with the transcriptome level. Moreover, there was a significant difference in two types of immune cells between the AS and control groups. Naive B cells, CD8+ T cells, regulatory T cells, and activated natural killer cells were positively correlated with CDO1 and NOX4 but negatively correlated with ATG7, CYBB, and SLC3A2. Conclusion In total, three m6A-Ferr-related signature genes (NOX4, CDO1, and SLC3A2) were obtained through a series of bioinformatics analyses and an RT-qPCR.
Collapse
Affiliation(s)
| | | | | | - Guofu Zhu
- Cardiology Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
29
|
Wu W, Wen F, Hu J, Li L. Overexpression of ATF4 Inhibits Ferroptosis to Alleviate Anxiety Disorders by Activating the TGF-β Signaling Pathway. Neuropsychiatr Dis Treat 2024; 20:1969-1983. [PMID: 39430656 PMCID: PMC11491069 DOI: 10.2147/ndt.s480782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
Background Anxiety disorders seriously impair patients' mental health and quality of life, with limited effectiveness of current treatments. Dysregulation of activating transcription factor 4 (ATF4) is involved in various mental diseases, but the research on its potential roles in alleviating anxiety disorders remains limited. Methods ATF4 was screened out by bioinformatic analysis and its expression was verified in vivo. Mice were treated with 21 d of chronic restraint stress to establish the anxiety mice model. The anxiolytic effect of ATF4 was assessed by a battery of behavior tests and evaluation of hippocampal tissue damage after overexpressing ATF4. Ferroptosis-related indicators were detected by enzyme-linked immunosorbent assay and Western blotting. Then the transforming growth factor beta (TGF-β) signaling pathway was predicted as the downstream regulatory pathway of ATF4 by bioinformatic methods. Western blotting was conducted to detect the protein expression level of TGF-β1, small mothers against decapentaplegic 3 (Smad3), and phospho-Smad3 (p-Smad3). Results ATF4 was screened out as a ferroptosis-related anxiolytic gene after bioinformatics analysis and was down-regulated in the anxiety mice model. Mice with ATF4 overexpression spent more time in the open arms in the elevated plus-maze test, appeared more frequently in the central area in the open-field test, and decreased the immobility time in the forced swimming and tail suspension tests. Hippocampal tissue damage was alleviated, ferroptosis was suppressed, and the levels of TGF-β1 and p-Smad3/Smad3 were increased by AFT4 overexpression. Conclusion ATF4 overexpression can repress ferroptosis to improve anxiety disorders by activating the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Wentao Wu
- Department of Psychiatry, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Fei Wen
- Department of Psychiatry, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Jiaxin Hu
- Department of Psychiatry, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Leijun Li
- Department of Psychiatry, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou City, Guangdong Province, People’s Republic of China
| |
Collapse
|
30
|
Zhang J, Liu J, Li Y, Zhang X, Yang C. Regulatory Role and Molecular Mechanism of Mammalian Sterile 20-Like Kinase 1 in 1-Methyl-4-Phenylpyridinium Ion-Induced Parkinson's Disease Cell Model. Rejuvenation Res 2024; 27:154-162. [PMID: 39003528 DOI: 10.1089/rej.2024.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024] Open
Abstract
Parkinson's disease (PD) is a multifactorial degenerative disease in the elder. Given the involvement of mammalian sterile 20-like kinase 1 (MST1) in PD, this article was to illustrate the mechanism of MST1 in 1-methyl-4-phenylpyridinium ion (MPP+)-induced PD cell model. Cells were treated with different concentrations of MPP+ to establish a PD cell model. Reverse transcription-quantitative polymerase chain reaction and Western blot revealed that MST1 expression and iron ion concentration increased, but cellular viability decreased with MPP+ concentration. Inhibition of MST1 decreased ferroptosis; increased cellular viability, iron ion content, and levels of glutathione peroxidase 4; and decreased reactive oxygen species and lactate dehydrogenase release. Upregulation of ferroptosis levels using ferroptosis agonist Erastin reduced the protective effect of MST1 inhibition on PD cells. Mechanistically, dual-luciferase analysis identified that miR-23b-3p targeted MST1 and inhibited its expression. Overexpression of miR-23b-3p inhibited MST1 levels, thereby reducing cellular ferroptosis and attenuating MPP+-induced cell injury. Collectively, MST1 expression increased with increasing MPP+ concentration, and miR-23b-3p targeted MST1 to reduce ferroptosis and MPP+-induced cell injury.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Neurology, Jingmen People's Hospital, Jingmen, China
| | - Jie Liu
- Department of Neurology, Jingmen People's Hospital, Jingmen, China
| | - Yongle Li
- Department of Neurology, Jingmen People's Hospital, Jingmen, China
| | - Xuexian Zhang
- Department of Neurology, Jingmen People's Hospital, Jingmen, China
| | - Chunxiang Yang
- Department of Neurology, Jingmen People's Hospital, Jingmen, China
| |
Collapse
|
31
|
Guo Z, Zhang W, Gao H, Li Y, Li X, Yang X, Fan L. High expression levels of haem oxygenase-1 promote ferroptosis in macrophage-derived foam cells and exacerbate plaque instability. Redox Biol 2024; 76:103345. [PMID: 39255694 PMCID: PMC11414708 DOI: 10.1016/j.redox.2024.103345] [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: 08/08/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Plaque rupture with consequent thrombosis is the leading cause of acute cardiovascular events, during which macrophage death is a hallmark. Ferroptosis is a pivotal intermediate link between early and advanced atherosclerosis. Existing evidence indicates the involvement of macrophage ferroptosis in plaque vulnerability; however, the exact mechanism remains elusive. The aim of this study was to explore key ferroptosis-related genes (FRGs) involved in plaque progression and the underlying molecular mechanisms involved. The expression landscape of FRGs was obtained from atherosclerosis-related GEO datasets. Molecular mechanism studies of ferroptosis were performed using bone marrow-derived macrophages (BMDMs) and macrophage-derived foam cells (MDFCs). Bioinformatics analysis and immunohistochemistry revealed that macrophage haem oxygenase-1 (HMOX1) is the key FRG involved in plaque destabilization. Hypoxic conditions induced a significant increase in Hmox1 expression in MDFCs but not in macrophages. In addition, the beneficial or deleterious effects of Hmox1 were dependent on the degree of Hmox1 induction. Hmox1 overexpression drove inflammatory responses and ferroptotic oxidative stress in MDFCs and aggravated the plaque burden in atherosclerotic model mice. Further mechanistic investigations demonstrated that hypoxia-mediated degradation of egl-9 family hypoxia-inducible factor 3 (Egln3) stabilized Hif1a, which subsequently promoted Hmox1 transcription. Our findings suggest that high Hmox1 expression under hypoxia is deleterious to MDFC viability and plaque stability, providing a reference for the management of acute cardiovascular events.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wan Zhang
- Department of Vascular Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Hongxia Gao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Yang Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Xiaohu Yang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Longhua Fan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China.
| |
Collapse
|
32
|
Jin B, Zhang Z, Zhang Y, Yang M, Wang C, Xu J, Zhu Y, Mi Y, Jiang J, Sun Z. Ferroptosis and myocardial ischemia-reperfusion: mechanistic insights and new therapeutic perspectives. Front Pharmacol 2024; 15:1482986. [PMID: 39411064 PMCID: PMC11473306 DOI: 10.3389/fphar.2024.1482986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a significant factor in the development of cardiac dysfunction following a myocardial infarction. Ferroptosis, a type of regulated cell death driven by iron and marked by lipid peroxidation, has garnered growing interest for its crucial involvement in the pathogenesis of MIRI.This review comprehensively examines the mechanisms of ferroptosis, focusing on its regulation through iron metabolism, lipid peroxidation, VDAC signaling, and antioxidant system dysregulation. We also compare ferroptosis with other forms of cell death to highlight its distinct characteristics. Furthermore, the involvement of ferroptosis in MIRI is examined with a focus on recent discoveries concerning ROS generation, mitochondrial impairment, autophagic processes, ER stress, and non-coding RNA regulation. Lastly, emerging therapeutic strategies that inhibit ferroptosis to mitigate MIRI are reviewed, providing new insights into potential clinical applications.
Collapse
Affiliation(s)
- Binwei Jin
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhiming Zhang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yang Zhang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Minjun Yang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Cheng Wang
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Cardiology, Taizhou hospital of Zhejiang Province, Shaoxing University, Linhai, China
| | - Jiayi Xu
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Cardiology, Taizhou hospital of Zhejiang Province, Shaoxing University, Linhai, China
| | - Yu Zhu
- Medical Research Center, Taizhou Hospital of Zhejiang Province, Linhai, China
| | - Yafei Mi
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Cardiology, Taizhou hospital of Zhejiang Province, Shaoxing University, Linhai, China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
33
|
Xi C, Zhang J, Liu H, Tao S, Xie Y, Liu J, Tong C, Tian D, Ye H, Zhang X. Can Omega-3 prevent the accidence of stroke: a mendelian randomization study. Hereditas 2024; 161:30. [PMID: 39232799 PMCID: PMC11375838 DOI: 10.1186/s41065-024-00329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND The lipid-lowering effects of Omega-3 fatty acids have been widely reported, yet their impact on ischemic stroke remains controversial. Reports on the protective effects of unsaturated fatty acids, such as Omega-6 and Omega-7, as well as saturated fatty acids in cardiovascular diseases, including hypertension and ischemic stroke, are less frequent. OBJECTIVES This study aims to identify fatty acids associated with blood pressure and ischemic stroke through Mendelian randomization. Besides, it seeks to determine whether specific fatty acids can prevent ischemic stroke by managing blood pressure and revealing the specific mechanisms of this action. METHODS This research involved downloading relevant data from websites and extracting SNPs that met the standard criteria as instrumental variables. Simultaneously, the 'MR-PRESSO' package and 'Mendelian Randomization' package were used to eliminate confounding SNPs that could bias the study results. Then, inverse variance weighting and the weighted median were employed as primary analysis methods, accompanied by sensitivity analysis to assess the validity of the causal relationships. Initially, multivariable Mendelian randomization was used to identify fatty acids linked to blood pressure and the incidence of ischemic stroke. The causal link between certain fatty acids and the initiation of ischemic stroke was then investigated using bidirectional and mediator Mendelian randomization techniques. Stepwise Regression and the Product of Coefficients Method in mediator Mendelian randomization were utilized to ascertain whether specific fatty acids reduce ischemic stroke risk by lowering blood pressure. RESULTS Multivariable Mendelian randomization analysis indicated a potential inverse correlation between Omega-3 intake and both blood pressure and ischemic stroke. Consequently, Omega-3 was selected as the exposure, with blood pressure and ischemic stroke-related data as outcomes, for further bidirectional and mediation Mendelian Randomization analyses. Bidirectional Mendelian Randomization revealed that Omega-3 significantly influences DBP (P = 1.01e-04) and IS (P = 0.016). It also showed that DBP and SBP significantly affect LAS, SVS, CES, IS, and LS. Mediator Mendelian Randomization identified five established mediating pathways: Omega-3-Diastolic blood pressure-Small vessel stroke, Omega-3-Diastolic blood pressure-Cardioembolic stroke, Omega-3-Diastolic blood pressure-Lacunar stroke, Omega-3-Diastolic blood pressure-Large artery atherosclerosis stroke, and Omega-3-Diastolic blood pressure-Ischemic stroke. Of these, four pathways are complete mediation, and one pathway is partial mediation. CONCLUSIONS The findings suggest that Omega-3 may indirectly reduce the incidence of ischemic stroke by lowering blood pressure. Thus, blood pressure modulation might be one of the mechanisms through which Omega-3 prevents ischemic stroke. In summary, incorporating an increased intake of Omega-3 in the diet can serve as one of the dietary intervention strategies for patients with hypertension. Additionally, it can act as an adjunctive therapy for the prevention of ischemic strokes and their complications.
Collapse
Affiliation(s)
- Chongcheng Xi
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhang
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haihui Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sian Tao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xie
- School of Acupuncture- Moxibustion and Tuina, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Jibin Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changqing Tong
- School of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Dong Tian
- School of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Hua Ye
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiaobo Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
34
|
Zhang M, Li J, Hu W. The complex interplay between ferroptosis and atherosclerosis. Biomed Pharmacother 2024; 178:117183. [PMID: 39079265 DOI: 10.1016/j.biopha.2024.117183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024] Open
Abstract
Atherosclerosis, characterized by the accumulation of plaque within the arterial walls, is an intricate cardiovascular disease that often results in severe health issues. Recent studies have emphasized the importance of ferroptosis, a controlled type of cell death dependent on iron, as a critical factor in this disease state. Ferroptosis, distinguished by its reliance on iron and the accumulation of lipid hydroperoxides, offers a unique insight into the pathology of atherosclerotic lesions. This summary encapsulates the current knowledge of the intricate role ferroptosis plays in the onset and progression of atherosclerosis. It explores the molecular processes through which lipid peroxidation and iron metabolism contribute to the development of atheromatous plaques and evaluates the possibility of utilizing ferroptosis as a novel treatment approach for atherosclerosis. By illuminating the intricate relationship between ferroptosis-related processes and atherosclerosis, this review paves the way for future clinical applications and personalized medicine approaches aimed at alleviating the effects of atherosclerosis.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangping Li
- Department of Oncological Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Hu
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
35
|
Gao HX, Jiang J, Yang CY, Xu JF, He Q, Hu YW. Zinc finger translocation‑associated protein promotes ferroptosis through the upregulation of ACSL4 expression in vascular endothelial cells. Exp Ther Med 2024; 28:334. [PMID: 39011065 PMCID: PMC11247542 DOI: 10.3892/etm.2024.12623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/17/2024] [Indexed: 07/17/2024] Open
Abstract
Numerous studies have reported the potential involvement of ferroptosis in the development of atherosclerosis (AS). Acyl-CoA synthetase long chain family member 4 (ACSL4) is an essential component in the promotion of ferroptosis. The present study aimed to investigate the role of ACSL4 and zinc finger translocation-associated protein (ZFTA) in the regulation of endothelial cell ferroptosis in AS. Human umbilical vein endothelial cells (HUVECs) with ACSL4 knockout were generated using CRISPR/Cas9 technology. To assess ferroptosis, malondialdehyde concentration, iron content and reactive oxygen species levels were quantified in the present study. In addition, western blot analysis was conducted to explore the potential mechanisms underlying ACSL4 and ZFTA in the modulation of ferroptosis in HUVECs. The results of the present study demonstrated that the expression levels of ACSL4 and ZFTA were significantly increased in human atherosclerotic plaques. In addition, ACSL4 knockout led to a reduced susceptibility to ferroptosis, while ZFTA contributed to ferroptosis in HUVECs. Results of the present study also demonstrated that ZFTA overexpression upregulated ACSL4 expression in HUVECs, whereas ZFTA knockdown led to decreased ACSL4 expression. Co-transfection experiments demonstrated that the ZTFA overexpression-mediated increase in ferroptosis was reversed following ACSL4 knockdown. Collectively, results of the present study highlighted that ACSL4 mediated the effects of ZFTA on the ferroptosis of HUVECs. Thus, the present study demonstrated the potential role of ACSL4 and ZFTA in the regulation of ferroptosis, and highlighted that ferroptosis-related pathways may act as potential targets in the treatment of AS.
Collapse
Affiliation(s)
- Hui-Xin Gao
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Jun Jiang
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Chun-Yan Yang
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Jin-Fu Xu
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Qing He
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, P.R. China
| |
Collapse
|
36
|
Wang X, Chen Y, Meng H, Ruan J, Meng F. SREBP-1-mediated lipogenesis confers resistance to ferroptosis and improves endothelial injury. FASEB J 2024; 38:e23806. [PMID: 38970404 DOI: 10.1096/fj.202400721r] [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: 04/01/2024] [Revised: 06/15/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024]
Abstract
Atherosclerosis refers to a disease characterized by the formation of lipid plaque deposits within arterial walls, leading to reduced blood flow or blockage of blood outflow. The process of endothelial injury induced by oxidized low-density lipoprotein (ox-LDL) is considered the initial stage of atherosclerosis. Ferroptosis is a form of iron-dependent, non-apoptotic cell death, and current research suggests its association with coronary artery disease (CAD). In this study, we observed a correlation between reduced expression of SREBP-1 and the occurrence of stable CAD. Additionally, during the process of endothelial injury induced by ox-LDL, we also noted decreased expression of the SREBP-1/SCD1/FADS2 and involvement in the ferroptosis process. Mechanistically, ox-LDL induced endothelial injury by inhibiting the lipid biosynthesis process mediated by the SREBP-1/SCD1/FADS2, thereby inducing lipid peroxidation and ferroptosis. On the contrary, overexpression of SREBP-1 or supplementation with monounsaturated fatty acids counteracted iron accumulation, mitochondrial damage, and lipid peroxidation-induced ferroptosis, thereby improving endothelial injury. Our study indicated that the decreased expression of peripheral blood SREBP-1 mRNA is an independent risk factor for stable CAD. Furthermore, in endothelial cells, the lipid biosynthesis process mediated by SREBP-1 could ameliorate endothelial injury by resisting ferroptosis. The study has been registered with the Chinese Clinical Trial Registry, which serves as a primary registry in the World Health Organization International Clinical Trials Registry Platform (ChiCTR2300074315, August 3rd, 2023).
Collapse
Affiliation(s)
- Xue Wang
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yanqiu Chen
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Heyu Meng
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianjun Ruan
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fanbo Meng
- China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Liu X, Meng Q, Shi S, Geng X, Wang E, Li Y, Lin F, Liang X, Xi X, Han W, Fan H, Zhou X. Cardiac-derived extracellular vesicles improve mitochondrial function to protect the heart against ischemia/reperfusion injury by delivering ATP5a1. J Nanobiotechnology 2024; 22:385. [PMID: 38951822 PMCID: PMC11218245 DOI: 10.1186/s12951-024-02618-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: 07/27/2023] [Accepted: 02/28/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Numerous studies have confirmed the involvement of extracellular vesicles (EVs) in various physiological processes, including cellular death and tissue damage. Recently, we reported that EVs derived from ischemia-reperfusion heart exacerbate cardiac injury. However, the role of EVs from healthy heart tissue (heart-derived EVs, or cEVs) on myocardial ischemia-reperfusion (MI/R) injury remains unclear. RESULTS Here, we demonstrated that intramyocardial administration of cEVs significantly enhanced cardiac function and reduced cardiac damage in murine MI/R injury models. cEVs treatment effectively inhibited ferroptosis and maintained mitochondrial homeostasis in cardiomyocytes subjected to ischemia-reperfusion injury. Further results revealed that cEVs can transfer ATP5a1 into cardiomyocytes, thereby suppressing mitochondrial ROS production, alleviating mitochondrial damage, and inhibiting cardiomyocyte ferroptosis. Knockdown of ATP5a1 abolished the protective effects of cEVs. Furthermore, we found that the majority of cEVs are derived from cardiomyocytes, and ATP5a1 in cEVs primarily originates from cardiomyocytes of the healthy murine heart. Moreover, we demonstrated that adipose-derived stem cells (ADSC)-derived EVs with ATP5a1 overexpression showed much better efficacy on the therapy of MI/R injury compared to control ADSC-derived EVs. CONCLUSIONS These findings emphasized the protective role of cEVs in cardiac injury and highlighted the therapeutic potential of targeting ATP5a1 as an important approach for managing myocardial damage induced by MI/R injury.
Collapse
Affiliation(s)
- Xuan Liu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shanshan Shi
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xuedi Geng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Enhao Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yinzhen Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fang Lin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoting Liang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoling Xi
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wei Han
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Huimin Fan
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China.
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China.
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
38
|
Aierken Y, He H, Li R, Lin Z, Xu T, Zhang L, Wu Y, Liu Y. Inhibition of Slc39a14/Slc39a8 reduce vascular calcification via alleviating iron overload induced ferroptosis in vascular smooth muscle cells. Cardiovasc Diabetol 2024; 23:186. [PMID: 38812011 PMCID: PMC11138056 DOI: 10.1186/s12933-024-02224-z] [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: 02/21/2024] [Accepted: 04/03/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Vascular calcification (VC) is an independent risk factor for cardiovascular diseases. Recently, ferroptosis has been recognised as a novel therapeutic target for cardiovascular diseases. Although an association between ferroptosis and vascular calcification has been reported, the role and mechanism of iron overload in vascular calcification are still poorly understood. Specifically, further in-depth research is required on whether metalloproteins SLC39a14 and SLC39a8 are involved in ferroptosis induced by iron overload. METHODS R language was employed for the differential analysis of the dataset, revealing the correlation between ferroptosis and calcification. The experimental approaches encompassed both in vitro and in vivo studies, incorporating the use of iron chelators and models of iron overload. Additionally, gain- and loss-of-function experiments were conducted to investigate iron's effects on vascular calcification comprehensively. Electron microscopy, immunofluorescence, western blotting, and real-time polymerase chain reaction were used to elucidate how Slc39a14 and Slc39a8 mediate iron overload and promote calcification. RESULTS Ferroptosis was observed in conjunction with vascular calcification (VC); the association was consistently confirmed by in vitro and in vivo studies. Our results showed a positive correlation between iron overload in VSMCs and calcification. Iron chelators are effective in reversing VC and iron overload exacerbates this process. The expression levels of the metal transport proteins Slc39a14 and Slc39a8 were significantly upregulated during calcification; the inhibition of their expression alleviated VC. Conversely, Slc39a14 overexpression exacerbates calcification and promotes intracellular iron accumulation in VSMCs. CONCLUSIONS Our research demonstrates that iron overload occurs during VC, and that inhibition of Slc39a14 and Slc39a8 significantly relieves VC by intercepting iron overload-induced ferroptosis in VSMCs, providing new insights into the VC treatment.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Animals
- Cation Transport Proteins/metabolism
- Cation Transport Proteins/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
- Iron Chelating Agents/pharmacology
- Iron Chelating Agents/therapeutic use
- Signal Transduction
- Male
- Humans
- Iron/metabolism
- Iron Overload/metabolism
- Iron Overload/pathology
Collapse
Affiliation(s)
- Yierpani Aierken
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Huqiang He
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Runwen Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Zipeng Lin
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Tongjie Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Li Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
39
|
Zhang W, Liu Y, Liao Y, Zhu C, Zou Z. GPX4, ferroptosis, and diseases. Biomed Pharmacother 2024; 174:116512. [PMID: 38574617 DOI: 10.1016/j.biopha.2024.116512] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/03/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
GPX4 (Glutathione peroxidase 4) serves as a crucial intracellular regulatory factor, participating in various physiological processes and playing a significant role in maintaining the redox homeostasis within the body. Ferroptosis, a form of iron-dependent non-apoptotic cell death, has gained considerable attention in recent years due to its involvement in multiple pathological processes. GPX4 is closely associated with ferroptosis and functions as the primary inhibitor of this process. Together, GPX4 and ferroptosis contribute to the pathophysiology of several diseases, including sepsis, nervous system diseases, ischemia reperfusion injury, cardiovascular diseases, and cancer. This review comprehensively explores the regulatory roles and impacts of GPX4 and ferroptosis in the development and progression of these diseases, with the aim of providing insights for identifying potential therapeutic strategies in the future.
Collapse
Affiliation(s)
- Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yang Liu
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
40
|
Pecchillo Cimmino T, Punziano C, Panico I, Petrone Z, Cassese M, Faraonio R, Barresi V, Esposito G, Ammendola R, Cattaneo F. Formyl-Peptide Receptor 2 Signaling Modulates SLC7A11/xCT Expression and Activity in Tumor Cells. Antioxidants (Basel) 2024; 13:552. [PMID: 38790657 PMCID: PMC11118824 DOI: 10.3390/antiox13050552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer cells exhibit high levels of oxidative stress and consequently require a high amount of cysteine for glutathione synthesis. Solute Carrier Family 7 Member 11 (SLC7A11), or xCT, mediates the cellular uptake of cystine in exchange for intracellular glutamate; imported extracellular cystine is reduced to cysteine in the cytosol through a NADPH-consuming reduction reaction. SLC7A11/xCT expression is under the control of stress-inducing conditions and of several transcription factors, such as NRF2 and ATF4. Formyl-peptide receptor 2 (FPR2) belongs to the FPR family, which transduces chemotactic signals mediating either inflammatory or anti-inflammatory responses according to the nature of its ligands and/or FPR2 binding with other FPR isoforms. The repertoire of FPR2 agonists with anti-inflammatory activities comprises WKYMVm peptide and Annexin A1 (ANXA1), and the downstream effects of the intracellular signaling cascades triggered by FPR2 include NADPH oxidase (NOX)-dependent generation of reactive oxygen species. Herein, we demonstrate that stimulation of CaLu-6 cells with either WKYMVm or ANXA1: (i) induces the redox-regulated activation of SLC7A11/xCT; (ii) promotes the synthesis of glutathione; (iii) prevents lipid peroxidation; and (iv) favors NRF2 nuclear translocation and activation. In conclusion, our overall results demonstrate that FPR2 agonists and NOX modulate SLC7A11/xCT expression and activity, thereby identifying a novel regulative pathway of the cystine/glutamate antiport that represents a new potential therapeutical target for the treatment of human cancers.
Collapse
Affiliation(s)
- Tiziana Pecchillo Cimmino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Carolina Punziano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Iolanda Panico
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Zeudi Petrone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Myrhiam Cassese
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Vincenza Barresi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| |
Collapse
|
41
|
Ishida M, Sakai C, Kobayashi Y, Ishida T. Cigarette Smoking and Atherosclerotic Cardiovascular Disease. J Atheroscler Thromb 2024; 31:189-200. [PMID: 38220184 PMCID: PMC10918046 DOI: 10.5551/jat.rv22015] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024] Open
Abstract
The detrimental effects of cigarette smoking on cardiovascular health, particularly atherosclerosis and thrombosis, are well established, and more detailed mechanisms continue to emerge. As the fundamental pathophysiology of the adverse effects of smoking, endothelial dysfunction, inflammation, and thrombosis are considered to be particularly important. Cigarette smoke induces endothelial dysfunction, leading to impaired vascular dilation and hemostasis regulation. Factors contributing to endothelial dysfunction include reduced bioavailability of nitric oxide, increased levels of superoxide anion, and endothelin release. Chronic inflammation of the vascular wall is a central pathogenesis of smoking-induced atherosclerosis. Smoking systemically elevates inflammatory markers and induces the expression of adhesion molecules and cytokines in various tissues. Pattern recognition receptors and damage-associated molecular patterns play crucial roles in the mechanism underlying smoking-induced inflammation. Smoking-induced DNA damage and activation of innate immunity, such as the NLRP3 inflammasome, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, and Toll-like receptor 9, are shown to amplify inflammatory cytokine expression. Cigarette smoke-induced oxidative stress and inflammation influence platelet adhesion, aggregation, and coagulation via adhesion molecule upregulation. Furthermore, it affects the coagulation cascade and fibrinolysis balance, causing thrombus formation. Matrix metalloproteinases contribute to plaque vulnerability and atherothrombotic events. The impact of smoking on inflammatory cells and adhesion molecules further intensifies the risk of atherothrombosis. Collectively, exposure to cigarette smoke exerts profound effects on endothelial function, inflammation, and thrombosis, contributing to the development and progression of atherosclerosis and atherothrombotic cardiovascular diseases. Understanding these intricate mechanisms highlights the urgent need for smoking cessation to protect cardiovascular health. This comprehensive review investigates the multifaceted mechanisms through which smoking contributes to these life-threatening conditions.
Collapse
Affiliation(s)
- Mari Ishida
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chiemi Sakai
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yusuke Kobayashi
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
42
|
Pan Q, Chen C, Yang YJ. Top Five Stories of the Cellular Landscape and Therapies of Atherosclerosis: Current Knowledge and Future Perspectives. Curr Med Sci 2024; 44:1-27. [PMID: 38057537 DOI: 10.1007/s11596-023-2818-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/22/2023] [Indexed: 12/08/2023]
Abstract
Atherosclerosis (AS) is characterized by impairment and apoptosis of endothelial cells, continuous systemic and focal inflammation and dysfunction of vascular smooth muscle cells, which is documented as the traditional cellular paradigm. However, the mechanisms appear much more complicated than we thought since a bulk of studies on efferocytosis, transdifferentiation and novel cell death forms such as ferroptosis, pyroptosis, and extracellular trap were reported. Discovery of novel pathological cellular landscapes provides a large number of therapeutic targets. On the other side, the unsatisfactory therapeutic effects of current treatment with lipid-lowering drugs as the cornerstone also restricts the efforts to reduce global AS burden. Stem cell- or nanoparticle-based strategies spurred a lot of attention due to the attractive therapeutic effects and minimized adverse effects. Given the complexity of pathological changes of AS, attempts to develop an almighty medicine based on single mechanisms could be theoretically challenging. In this review, the top stories in the cellular landscapes during the initiation and progression of AS and the therapies were summarized in an integrated perspective to facilitate efforts to develop a multi-targets strategy and fill the gap between mechanism research and clinical translation. The future challenges and improvements were also discussed.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Cheng Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
43
|
Luo X, Wang Y, Zhu X, Chen Y, Xu B, Bai X, Weng X, Xu J, Tao Y, Yang D, Du J, Lv Y, Zhang S, Hu S, Li J, Jia H. MCL attenuates atherosclerosis by suppressing macrophage ferroptosis via targeting KEAP1/NRF2 interaction. Redox Biol 2024; 69:102987. [PMID: 38100883 PMCID: PMC10761782 DOI: 10.1016/j.redox.2023.102987] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/03/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Micheliolide (MCL), which is the active metabolite of parthenolide, has demonstrated promising clinical application potential. However, the effects and underlying mechanisms of MCL on atherosclerosis are still unclear. METHOD ApoE-/- mice were fed with high fat diet, with or without MCL oral administration, then the plaque area, lipid deposition and collagen content were determined. In vitro, MCL was used to pretreat macrophages combined by ox-LDL, the levels of ferroptosis related proteins, NRF2 activation, mitochondrial function and oxidative stress were detected. RESULTS MCL administration significantly attenuated atherosclerotic plaque progress, which characteristics with decreased plaque area, less lipid deposition and increased collagen. Compared with HD group, the level of GPX4 and xCT in atherosclerotic root macrophages were increased in MCL group obviously. In vitro experiment demonstrated that MCL increased GPX4 and xCT level, improved mitochondrial function, attenuated oxidative stress and inhibited lipid peroxidation to suppress macrophage ferroptosis induced with ox-LDL. Moreover, MCL inhibited KEAP1/NRF2 complex formation and enhanced NRF2 nucleus translocation, while the protective effect of MCL on macrophage ferroptosis was abolished by NRF2 inhibition. Additionally, molecular docking suggests that MCL may bind to the Arg483 site of KEAP1, which also contributes to KEAP1/NRF2 binding. Furthermore, Transfection Arg483 (KEAP1-R483S) mutant plasmid can abrogate the anti-ferroptosis and anti-oxidative effects of MC in macrophages. KEAP1-R483S mutation also limited the protective effect of MCL on atherosclerosis progress and macrophage ferroptosis in ApoE-/- mice. CONCLUSION MCL suppressed atherosclerosis by inhibiting macrophage ferroptosis via activating NRF2 pathway, the related mechanism is through binding to the Arg483 site of KEAP1 competitively.
Collapse
Affiliation(s)
- Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Yuehong Wang
- State Key Laboratory of Systems Medicine for Cancer, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Cancer Institute, Shanghai, 200127, PR China
| | - Xinxin Zhu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Yuwu Chen
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Biyi Xu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Xiaoxuan Bai
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Xiuzhu Weng
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Jinmei Xu
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China
| | - Yangyang Tao
- Department of Ultrasound, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China
| | - Dan Yang
- Department of Forensic Medicine, Harbin Medical University, Harbin, 150001, PR China
| | - Jie Du
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Ying Lv
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Shan Zhang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Sining Hu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Ji Li
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; National Key Laboratory of Frigid Zone Cardiovascular Diseases; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, PR China.
| |
Collapse
|
44
|
Luan Y, Yang Y, Luan Y, Liu H, Xing H, Pei J, Liu H, Qin B, Ren K. Targeting ferroptosis and ferritinophagy: new targets for cardiovascular diseases. J Zhejiang Univ Sci B 2024; 25:1-22. [PMID: 38163663 PMCID: PMC10758208 DOI: 10.1631/jzus.b2300097] [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: 02/13/2023] [Accepted: 05/21/2023] [Indexed: 01/03/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading factor driving mortality worldwide. Iron, an essential trace mineral, is important in numerous biological processes, and its role in CVDs has raised broad discussion for decades. Iron-mediated cell death, namely ferroptosis, has attracted much attention due to its critical role in cardiomyocyte damage and CVDs. Furthermore, ferritinophagy is the upstream mechanism that induces ferroptosis, and is closely related to CVDs. This review aims to delineate the processes and mechanisms of ferroptosis and ferritinophagy, and the regulatory pathways and molecular targets involved in ferritinophagy, and to determine their roles in CVDs. Furthermore, we discuss the possibility of targeting ferritinophagy-induced ferroptosis modulators for treating CVDs. Collectively, this review offers some new insights into the pathology of CVDs and identifies possible therapeutic targets.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, China
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Han Xing
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No. 3 Provincial People's Hospital, Zhengzhou 450052, China
| | - Hengdao Liu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China. ,
| | - Bo Qin
- Center for Translational Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China. ,
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China.
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
45
|
Gu Y, Cui M, Wang W, Zhang J, Wang H, Zheng C, Lei L, Ji M, Chen W, Xu Y, Wang P. Visualization of the Ferroptosis in Atherosclerotic Plaques with Nanoprobe Engineered by Macrophage Cell Membranes. Anal Chem 2024; 96:281-291. [PMID: 38153251 DOI: 10.1021/acs.analchem.3c03999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Atherosclerosis (AS) is the root cause of cardiovascular diseases. Ferroptosis is characterized by highly iron-dependent lipid peroxidation and has been reported to play an important role in the pathogenesis of AS. Visualization of the ferroptosis process in atherosclerotic plaques is of great importance for diagnosing and treating AS. In this work, the rationally designed fluorescent probe FAS1 exhibited excellent advantages including large Stokes shift, sensitivity to environmental viscosity, good photostability, and improved water solubility. It also could co-locate with commercial lipid droplets (LDs) probes (BODIPY 493/503) well in RAW264.7 cells treated by the ferroptosis inducer. After self-assembly into nanoparticles and then encapsulation with macrophage membranes, the engineered FAS1@MM NPs could successfully target the atherosclerotic plaques in Western diet-induced apolipoprotein E knockout (ApoE-/-) mice and reveal the association of ferroptosis with AS through fluorescence imaging in vivo. This study may provide additional insights into the roles of ferroptosis in the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Yinhui Gu
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyuan Cui
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Weizhi Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Jiaqi Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Huizhe Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Zheng
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Lijuan Lei
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610044, China
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing 100050, China
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
46
|
Zhou D, Lu P, Mo X, Yang B, Chen T, Yao Y, Xiong T, Yue L, Yang X. Ferroptosis and metabolic syndrome and complications: association, mechanism, and translational applications. Front Endocrinol (Lausanne) 2024; 14:1248934. [PMID: 38260171 PMCID: PMC10800994 DOI: 10.3389/fendo.2023.1248934] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic syndrome is a medical condition characterized by several metabolic disorders in the body. Long-term metabolic disorders raise the risk of cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM). Therefore, it is essential to actively explore the aetiology of metabolic syndrome (MetS) and its comorbidities to provide effective treatment options. Ferroptosis is a new form of cell death that is characterized by iron overload, lipid peroxide accumulation, and decreased glutathione peroxidase 4(GPX4) activity, and it involves the pathological processes of a variety of diseases. Lipid deposition caused by lipid diseases and iron overload is significant in metabolic syndrome, providing the theoretical conditions for developing ferroptosis. Recent studies have found that the major molecules of ferroptosis are linked to common metabolic syndrome consequences, such as T2DM and atherosclerosis. In this review, we first discussed the mechanics of ferroptosis, the regulatory function of inducers and inhibitors of ferroptosis, and the significance of iron loading in MetS. Next, we summarized the role of ferroptosis in the pathogenesis of MetS, such as obesity, type 2 diabetes, and atherosclerosis. Finally, we discussed relevant ferroptosis-targeted therapies and raised some crucial issues of concern to provide directions for future Mets-related treatments and research.
Collapse
Affiliation(s)
- Dongmei Zhou
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Peipei Lu
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xianglai Mo
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Bing Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Ting Chen
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - You Yao
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Tian Xiong
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Lin Yue
- School of Nursing, Hunan University of Medicine, Huaihua, China
| | - Xi Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
47
|
Hu Y, Gu X, Zhang Y, Ma W, Sun L, Wang C, Ren B. Adrenomedullin, transcriptionally regulated by vitamin D receptors, alleviates atherosclerosis in mice through suppressing AMPK-mediated endothelial ferroptosis. ENVIRONMENTAL TOXICOLOGY 2024; 39:199-211. [PMID: 37688783 DOI: 10.1002/tox.23958] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 09/11/2023]
Abstract
PURPOSE Vitamin D receptors (VDR) play important roles in cardiovascular, immune, metabolic and other functions. Activation of VDR may help improve endothelial dysfunction, atherosclerosis, vascular calcification, and cardiac hypertrophy. However, the specific target genes and mechanisms of VDR in improving Human Umbilical Vein Endothelial Cell (HUVEC) functions remain unclear. This study aims to investigate the function and mechanism of VDR in HUVECs. METHODS Endothelial dysfunction cell model was constructed by oxidized low-density lipoprotein (ox-LDL). An animal model of atherosclerosis was established in male homozygous Apoe-/- mice (6 weeks) on a high fat diet for 6 weeks. The relationship between VDR and adrenomedullin (ADM) was studied by bioinformatics analysis, ChIP, and luciferase reporter gene analysis. Endothelial cell function was evaluated by Transwell migration and Tube Formation tests. Ferroptosis was detected by measuring intracellular iron content, levels of oxidative stress markers, and ferroptosis related proteins. RESULTS Overexpression of VDR in HUVECs inhibits ox-LDL-induced endothelial dysfunction and ferroptosis. VDR binds to the ADM promoter sequence and regulates the transcription of ADM. Inhibition of ADM promotes ox-LDL-induced endothelial dysfunction and ferroptosis. ADM regulates ox-LDL-induced endothelial dysfunction and ferroptosis through the AMPK signaling pathway. Overexpression of VDR in Apoe-/- mice inhibited lipid deposition and plaque area in atherosclerotic mice. CONCLUSION VDR inhibits ox-LDL-induced endothelial dysfunction and ferroptosis by regulating ADM transcription and acting on AMPK signaling pathway. Overexpression of VDR in Apoe-/- mice reduced lipid deposition and plaque area in the thoracic aorta of atherosclerotic mice.
Collapse
Affiliation(s)
- Yanchao Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xu Gu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Weidong Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Lijun Sun
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Congxia Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Bincheng Ren
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
48
|
Li S, Wen P, Zhang D, Li D, Gao Q, Liu H, Di Y. PGAM5 expression levels in heart failure and protection ROS-induced oxidative stress and ferroptosis by Keap1/Nrf2. Clin Exp Hypertens 2023; 45:2162537. [PMID: 36780919 DOI: 10.1080/10641963.2022.2162537] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
OBJECTIVES As a common and frequently occurring disease, heart failure has been paid more and more attention, but the mechanism of its occurrence and development is still unclear. This study investigated that PGAM5 expression levels in heart failure and its underlying mechanisms in vivo and in vitro. METHODS The inhibition of PGAM5 mRNA expression levels in patients with heart failure was compared with the normal group. RESULTS The serum of PGAM5 mRNA expression was negative correlation with collagen I and collagen III in patients with heart failure. PGAM5 mRNA and protein expression in the heart tissue of mice with heart failure were down-regulated at a time-dependent rate. The inhibition of PGAM5 presented heart failure in the model. PGAM5 reduced inflammation and inhibited ROS-induced oxidative stress in models of heart failure. PGAM5 reduced Ferroptosis in models of heart failure. PGAM5 regulated Keap1/Nrf2 signaling pathway. IP also showed that PGAM5 protein combined with the Keap1 protein. PGAM5 could increase Keap1 protein ubiquitination. Keap1 inhibition affected the effects of PGAM5 in model of heart failure. CONCLUSIONS We conclude that the protection of PGAM5 reduced ROS-induced oxidative stress and ferroptosis by the Keap1/Nrf2 signaling pathway in heart failure, suggesting that targeting this mechanism of PGAM5 may be a feasible strategy to treat heart failure.
Collapse
Affiliation(s)
- Shuangfei Li
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| | - Ping Wen
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| | - Dayong Zhang
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| | - Decai Li
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| | - Qidong Gao
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| | - Hong Liu
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| | - Yunfeng Di
- Department of Cardiovascular Medicine, Sichuan Mianyang 404 Hospital, Mianyang, China
| |
Collapse
|
49
|
Yan S, Shi Q, Ma H, Xu Q. Study on mechanism of Zhenwu Decoction in treatment of heart failure based on network pharmacology: A review. Medicine (Baltimore) 2023; 102:e36073. [PMID: 37986291 PMCID: PMC10659699 DOI: 10.1097/md.0000000000036073] [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: 06/29/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023] Open
Abstract
To explore the mechanism of Zhenwu Decoction (ZWD) in the treatment of heart failure (HF) by network pharmacology analysis, so as to provide a basis for the innovation and application of drugs. The effective components and targets of 5 Chinese herbal medicines in ZWD were retrieved by TCM Pharmacology Database and Analysis Platform (TCMSP).Gene card, OMIM and TTD databases were used to obtain the disease targets of HF, and the intersection with the targets of ZWD was obtained. We used Cytoscape3.9.1 software to construct a drug-active component-disease-target interaction network for ZWD treatment of HF, and performed protein-protein interaction (PPI) network and topology analysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses were performed. Fifty-nine effective components and 229 targets of ZWD were screened. Among them, ZWD for HF has 27 active components and 38 common targets, and the core targets of PPI are IL-6, ATK1 and TNF. Pathway enrichment analysis included lipid and atherosclerotic and TNF signaling pathways. This study preliminarily clarified the main active components, targets and related pathways of ZWD in the treatment of HF, and laid a foundation for further study of the pharmacological effects of ZWD.
Collapse
Affiliation(s)
- Sai Yan
- The second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Qingchun Shi
- Baotou Mongolian Medicine Hospital of Traditional Chinese Medicine, Baotou, China
| | - Hongtao Ma
- The second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Qian Xu
- The second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
50
|
Xing M, Ma X, Wang X, Wang H, Xie M, Zhang Z, Zhou J. Emodin disrupts the Notch1/Nrf2/GPX4 antioxidant system and promotes renal cell ferroptosis. J Appl Toxicol 2023; 43:1702-1718. [PMID: 37393915 DOI: 10.1002/jat.4509] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/04/2023]
Abstract
Emodin has been demonstrated to possess multiple pharmacological activities. However, emodin has also been reported to induce nephrotoxicity at high doses and with long-term use, and the underlying mechanism has not been fully disclosed. The current study aimed to investigate the roles of oxidative stress and ferroptosis in emodin-induced kidney toxicity. Mice were intraperitoneally treated with emodin, and NRK-52E cells were exposed to emodin in the presence or absence of treatment with Jagged1, SC79, or t-BHQ. Emodin significantly upregulated the levels of blood urea nitrogen, serum creatinine, malondialdehyde, and Fe2+ , reduced the levels of superoxide dismutase and glutathione, and induced pathological changes in the kidneys in vivo. Moreover, the viability of NRK-52E cells treated with emodin was reduced, and emodin induced iron accumulation, excessive reactive oxygen species production, and lipid peroxidation and depolarized the mitochondrial membrane potential (ΔΨm). In addition, emodin treatment downregulated the activity of neurogenic locus notch homolog protein 1 (Notch1), reduced the nuclear translocation of nuclear factor erythroid-2 related factor 2 (Nrf2), and decreased glutathione peroxidase 4 protein levels. However, Notch1 activation by Jagged1 pretreatment, Akt activation by SC79 pretreatment, or Nrf2 activation by t-BHQ pretreatment attenuated the toxic effects of emodin in NRK-52E cells. Taken together, these results revealed that emodin-induced ferroptosis triggered kidney toxicity through inhibition of the Notch1/Nrf2/glutathione peroxidase 4 axis.
Collapse
Affiliation(s)
- Miao Xing
- School of Medicine, Yichun University, Yichun, China
| | - Xiaoyu Ma
- School of Medicine, Yichun University, Yichun, China
| | - Xi Wang
- School of Medicine, Yichun University, Yichun, China
| | - Haoze Wang
- School of Medicine, Yichun University, Yichun, China
| | - Minjuan Xie
- School of Medicine, Yichun University, Yichun, China
| | - Ziwen Zhang
- School of Medicine, Yichun University, Yichun, China
| | - Jie Zhou
- School of Medicine, Yichun University, Yichun, China
| |
Collapse
|