1
|
Wang X, Li FJ, Cheng Y, Chen S, Zhu S, Zhang Y, Reiter RJ, Ashrafizadeh M, Lin J, Wang G, Lin L, Ren J. Activation of protein kinase B rescues against thapsigargin-elicited cardiac dysfunction through regulation of NADPH oxidase and ferroptosis. Chem Biol Interact 2025; 405:111292. [PMID: 39477182 DOI: 10.1016/j.cbi.2024.111292] [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: 08/28/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Endoplasmic reticulum (ER) stress is a known contributor to cardiac remodeling and contractile dysfunction. Although NADPH oxidase has been implicated in ER stress-induced organ damage, its specific role in myocardial complications resulting from ER stress remains unclear. This study aimed to investigate the possible involvement of NADPH oxidase in ER stress-induced myocardial abnormalities and to evaluate the impact of Akt constitutive activation on these myocardial defects. Mice with cardiac-specific overexpression of active mutant of Akt (Myr-Akt) and their wild-type (WT) littermates were treated with ER stress instigator thapsigargin (1 mg/kg, i. p. 72 hrs) before evaluating myocardial morphology and function. Our results noted that thapsigargin significantly impaired echocardiographic parameters and cell shortening indices, including elevated LVESD, decreased ejection fraction, fractional shortening, peak shortening, electrically-stimulated intracellular Ca2+ release, and cardiomyocyte survival. These functional deteriorations were accompanied by upregulation of NADPH oxidase, O2- production, mitochondrial damage, carbonyl formation, lipid peroxidation, apoptosis, and interstitial fibrosis, with unchanged myocardial size. Constitutive Akt hyperactivation did not generate any response on myocardial morphology and function, although it greatly suppressed or nullified thapsigargin-induced myocardial remodeling and dysfunction. Thapsigargin also triggered dephosphorylation of Akt and its downstream signal GSK3β, along with development of ferroptosis, all of which were nullified by Akt hyperactivation. In vitro studies further revealed that thapsigargin provoked cardiomyocyte mechanical anomalies and lipid peroxidation, similar to in vivo results. These effects were reverted by inhibitors of NADPH oxidase and ferroptosis (apocynin and LIP1). Collectively, our data denote an important protective role for Akt hyperactivation in thapsigargin-evoked myocardial anomalies, likely through NADPH oxidase-mediated regulation of ferroptosis.
Collapse
Affiliation(s)
- Xiaohu Wang
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Henan Provincial People's Hospital Heart Center, Zhengzhou, 451464, China
| | - Feng-Juan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China
| | - Yong Cheng
- Department of Cardiovascular Medicine, The Sixth People's Hospital of Zhengzhou, Zhengzhou, Henan, 450000, China
| | - Shuying Chen
- Department of Cardiology, Xinfeng People's Hospital, Shaoguan, 511199, China
| | - Shuyi Zhu
- The Cardiovascular Medicine Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 451162, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX, USA
| | - Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, 200072, China.
| | - Ling Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
2
|
Wang Y, Su H, Lin X, Dai C, Cheng Q, Deng Z, Yang Y, Pu X. 1,25-(OH) 2D 3 improves SD rats high-altitude pulmonary edema by inhibiting ferroptosis and ferritinophagy in alveolar epithelial cells. J Steroid Biochem Mol Biol 2024; 247:106663. [PMID: 39681240 DOI: 10.1016/j.jsbmb.2024.106663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/21/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND This study investigates the protective effects and potential mechanisms of 1,25-(OH)2D3 against high-altitude pulmonary edema (HAPE). METHODS Hypoxia-induced rats were administered 1,25-(OH)2D3 for 24, 48, and 72 hours, and we observed lung tissue injury and pulmonary edema. Immunohistochemistry (IHC) and Western blot analyses were employed to analyze the expression of markers associated with ferroptosis and ferritinophagy in rat lungs. Metabolomics analysis was conducted to investigate changes in serum lipid metabolites. We validated the mechanism of action of 1,25-(OH)2D3 in type II alveolar epithelial cells induced by hypoxia. RESULTS Our results demonstrated that hypoxic exposure significantly altered sodium-water transport in the lungs, leading to edema formation. The degree of pulmonary edema was most pronounced at 48 hours of hypoxi. Treatment with 1,25-(OH)2D3 improved lung function and reduced the degree of pulmonary edema in hypoxic rats. Hypoxia-induced increases in 4-HNE and MDA levels in the lungs, along with iron accumulation, were observed. Hypoxia also resulted in elevated levels of NCOA4, LC3Ⅱ, and FTH1 proteins in the lungs. Furthermore, treatment with 1,25-(OH)2D3 significantly inhibited ferroptosis and ferritinophagy in the lungs after hypoxia. The levels of lipid metabolites, such as L-Aspartic acid and L-Fucose, were significantly elevated in the serum of hypoxic rats. After 1,25-(OH)2D3 treatment, these levels exhibited a significant reduction. CONCLUSION In hypoxic type II alveolar epithelial cells, 1,25-(OH)2D3 improved hypoxia-induced sodium-water transport, ferroptosis, and ferritinophagy, which were reversed by the autophagy agonist Rapamycin.By modulating ferroptosis and ferritinophagy, 1,25-(OH)2D3 mitigated the deleterious effects of hypoxia on pulmonary function.
Collapse
Affiliation(s)
- Yaxuan Wang
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Hong Su
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Xue Lin
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China.
| | - Chongyang Dai
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Qian Cheng
- Qinghai University, Xining, Qinghai Province 810016, China.
| | | | - Yangyang Yang
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Xiaoyan Pu
- Qinghai University, Xining, Qinghai Province 810016, China.
| |
Collapse
|
3
|
Yang D, Wang X, Sun Y, Shao Y, Shi X. Identification and experimental validation of genes associated with programmed cell death in dendritic cells of the thyroid tissue in Hashimoto's thyroiditis. Int Immunopharmacol 2024; 142:113083. [PMID: 39260305 DOI: 10.1016/j.intimp.2024.113083] [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: 11/19/2023] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Hashimoto's thyroiditis (HT) is a chronic autoimmune disorder. As antigen-presenting cells, dendritic cells(DCs) play a pivotal role in inducing programmed cell death (PCD) types, contributing to immune disorders. This study aimed to identify genes associated with multiple PCD pathways in dendritic cells within the thyroid tissue of patients with HT. METHODS The single-cell RNA-sequencing dataset HRA001684 was obtained from the National Genomics Data Center (NGDC) to calculate the area under the curve (AUC) scores for PCD-related genes. Additionally, mRNA sequencing datasets GSE138198 and HRA001684 were sourced from the Gene Expression Omnibus(GEO) and NGDC, respectively. Differentially expressed genes (DEGs) were identified by comparing normal and HT groups in GSE138198 and HRA001684. The intersection of these DEGs with PCD-related genes led to the identification of 17 PCD-related DEGs(PCDDEGs). RESULTS AUC scores revealed that DCs in HT exhibited significantly elevated levels of necroptosis, ferroptosis, pyroptosis, autophagy, and PANoptosis, expressing six key PCDDEGs: TNFAIP3, CYBB, PTPN6, STAT1, TGFB1, and NLRP3. These genes displayed an AUC>0.8 for HT in the GSE29315, GSE138198, and HRA001684 datasets, confirming their diagnostic accuracy. Moreover, their expression was positively correlated with the serum levels of thyroid peroxidase and thyroglobulin antibodies, while the expression of all PCDDEGs was negatively correlated with the abundance of thyroid follicular epithelial cells. qRT-PCR, WB, IHC, and IF experiments further confirmed the differences in PCDDEGs gene and protein levels in HT patients. DISCUSSION These findings highlight the crucial role of DCs in mediating PCD within the thyroid tissues of HT patients. The identified PCDDEGs-TNFAIP3, CYBB, PTPN6, STAT1, TGFB1, and NLRP3-may significantly contribute to HT pathogenesis through PCD pathways.
Collapse
Affiliation(s)
- Dongyu Yang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, PR China
| | - Xichang Wang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, PR China
| | - Ying Sun
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, PR China
| | - Ying Shao
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, PR China
| | - Xiaoguang Shi
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, PR China.
| |
Collapse
|
4
|
Sun Q, Lopaschuk GD. What Is the Link Between Ferroptosis and Cardiac Hypertrophy? Can J Cardiol 2024:S0828-282X(24)01234-0. [PMID: 39631500 DOI: 10.1016/j.cjca.2024.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
5
|
Pan C, Zhao H, Cai X, Wu M, Qin B, Li J. The connection between autophagy and ferroptosis in AKI: recent advances regarding selective autophagy. Ren Fail 2024; 46:2379601. [PMID: 39099238 DOI: 10.1080/0886022x.2024.2379601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Acute kidney injury (AKI) is a significant issue in public health, displaying a high occurrence rate and mortality rate. Ferroptosis, a form of programmed cell death (PCD), is characterized by iron accumulation and intensified lipid peroxidation. Recent studies have demonstrated the pivotal significance of ferroptosis in AKI caused by diverse stimuli, including ischemia-reperfusion injury (IRI), sepsis and toxins. Autophagy, a multistep process that targets damaged organelles and macromolecules for degradation and recycling, also plays an essential role in AKI. Previous research has demonstrated that autophagy deletion in proximal tubules could aggravate tubular injury and renal function loss, indicating the protective function of autophagy in AKI. Consequently, finding ways to stimulate autophagy has become a crucial therapeutic strategy. The recent discovery of the role of selective autophagy in influencing ferroptosis has identified new therapeutic targets for AKI and has highlighted the importance of understanding the cross-talk between autophagy and ferroptosis. This study aims to provide an overview of the signaling pathways involved in ferroptosis and autophagy, focusing on the mechanisms and functions of selective autophagy and autophagy-dependent ferroptosis. We hope to establish a foundation for future investigations into the interaction between autophagy and ferroptosis in AKI as well as other diseases.
Collapse
Affiliation(s)
- Chunyu Pan
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hairui Zhao
- Department of Nephrology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaojing Cai
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manyi Wu
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bowen Qin
- Department of Nephrology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Nephrology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [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: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
7
|
Zheng K, Qian Y, Wang H, Song D, You H, Hou B, Han F, Zhu Y, Feng F, Lam SM, Shui G, Li X. Combinatorial lipidomics and proteomics underscore erythrocyte lipid membrane aberrations in the development of adverse cardio-cerebrovascular complications in maintenance hemodialysis patients. Redox Biol 2024; 78:103389. [PMID: 39486359 PMCID: PMC11563940 DOI: 10.1016/j.redox.2024.103389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024] Open
Abstract
Patients on maintenance hemodialysis exhibit a notably higher risk of cardio-cerebrovascular complications that constitute the major cause of death. Preceding studies have reported conflicting associations between traditional lipid measures and clinical outcome in dialysis patients. In this prospective longitudinal study, we utilized quantitative lipidomics to elucidate, at molecular resolution, changes in lipidome profiles of erythrocyte and plasma samples collected from maintenance hemodialysis patients followed up for 86 months (≈7 years). Primary outcome was defined as cardiovascular-related deaths or new-onset cardio-cerebrovascular events. Cox regression model uncovered plasma/erythrocyte lipids associated with incident cardio-cerebrovascular events in the erythrocyte cohort (n = 117 patients, 37 events) and plasma cohort (n = 45 patients, 11 events), respectively. Both the erythrocyte lipid panel [PA 40:5, PI 34:2, PC 42:6, AUC = 0.83] and plasma lipid panel [PC O-34:1, GM3 18:1; O2/25:0, TG 44:1(16:1_28:0), AUC = 0.94] significantly improved the prediction of cardio-cerebrovascular-related outcome compared to the base model comprising age, sex and dialysis vintage alone. Our findings underscore the pathophysiological significance of anionic phospholipid accretion in erythrocytes in the development of cardio-cerebrovascular complications in dialysis patients. In particular, distorted membrane lipid asymmetry leads to compromised membrane deformability, aberrant cell-cell interactions and altered glutathione metabolism in the erythrocytes of high-risk individuals even at relatively early stage of hemodialysis. Our findings thus underscore the importance of maintaining the RBC pool to lower the risk of cardio-cerebrovascular complications in dialysis patients.
Collapse
Affiliation(s)
- Ke Zheng
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujun Qian
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China; Department of Nephrology, Jiangsu Province Hospital/The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyun Wang
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dan Song
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Xuemei Li
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
8
|
Zeng Q, Jiang T. Molecular mechanisms of ferroptosis in cardiovascular disease. Mol Cell Biochem 2024; 479:3181-3193. [PMID: 38374233 DOI: 10.1007/s11010-024-04940-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/12/2024] [Indexed: 02/21/2024]
Abstract
Ferroptosis is a newly recognized type of regulated cell death that is characterized by the accumulation of iron and lipid peroxides in cells. Studies have shown that ferroptosis plays a significant role in the pathogenesis of various diseases, including cardiovascular diseases. In cardiovascular disease, ferroptosis is associated with ischemia-reperfusion injury, myocardial infarction, heart failure, and atherosclerosis. The molecular mechanisms underlying ferroptosis include the iron-dependent accumulation of lipid peroxidation products, glutathione depletion, and dysregulation of lipid metabolism, among others. This review aims to summarize the current knowledge of the molecular mechanisms of ferroptosis in cardiovascular disease and discuss the potential therapeutic strategies targeting ferroptosis as a treatment for cardiovascular disease.
Collapse
Affiliation(s)
- Qun Zeng
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Tingting Jiang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| |
Collapse
|
9
|
Xu C, Wu M, Yu W, Xie D, Wang Q, Chen B, Xi Y, Yu L, Yan Y, Yamamoto T, Koyama H, Zhao H, Cheng J. High Uric Acid Orchestrates Ferroptosis to Promote Cardiomyopathy Via ROS-GPX4 Signaling. Antioxid Redox Signal 2024; 41:1134-1149. [PMID: 39113539 DOI: 10.1089/ars.2023.0473] [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] [Indexed: 08/29/2024]
Abstract
Aims: High uric acid (HUA), as a pro-oxidant, plays a significant role in the pathophysiology of cardiovascular disease. Studies have indicated that elevated uric acid levels can adversely affect cardiovascular health. Nevertheless, the impact of hyperuricemia on cardiomyopathy remains uncertain. Further research is needed to elucidate the relationship between HUA and cardiomyopathy, shedding light on its potential implications for heart health. Results: We demonstrated that uricase knockout (Uox-KO) mice accelerated the development of cardiomyopathy, causing significantly impaired cardiac function and myocardial fibrosis. Meanwhile, the mitochondrial morphology was destroyed, the lipid peroxidation products increased in number and the antioxidant function was weakened. In addition, we evaluated the effects of ferrostatin-1 (Fer-1), the ferroptosis inhibitor. Myocardial damage can be reversed by the Fer-1 treatment caused by HUA combined with doxorubicin (DOX) treatment. Benzbromarone, a uric acid-lowering drug, decreases myocardial fibrosis, and ferroptosis by alleviating hyperuricemia in Uox-KO mice by DOX administration. In vitro, we observed that the activity of cardiomyocytes treated with HUA combined with DOX decreased significantly, and lipid reactive oxygen species (ROS) increased significantly. Afterward, we demonstrated that HUA can promote oxidative stress in DOX, characterized by increased mitochondrial ROS, and downregulate protein levels of glutathione peroxidase 4 (GPX4). N-acetyl-l-cysteine, an antioxidant, inhibits the process by which HUA promotes DOX-induced ferroptosis by increasing the GPX4 expression. Innovation: We verified that HUA can exacerbate myocardial damage. This has clinical implications for the treatment of cardiac damage in patients with hyperuricemia. Conclusions: Our data suggested that HUA promotes the cardiomyopathy. HUA promotes DOX-induced ferroptosis by increasing oxidative stress and downregulating GPX4. Antioxid. Redox Signal. 41, 1134-1149.
Collapse
Affiliation(s)
- Chenxi Xu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Institute of Metabolism and Cell Death, Helmholtz Munich, 85764 Neuherberg, Bavaria, Germany
| | - Mengni Wu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Wei Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Qiang Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Binyang Chen
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yuemei Xi
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Linqian Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yunbo Yan
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hong Zhao
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, China
| |
Collapse
|
10
|
Li FJ, Hu H, Wu L, Luo B, Zhou Y, Ren J, Lin J, Reiter RJ, Wang S, Dong M, Guo J, Peng H. Ablation of mitophagy receptor FUNDC1 accentuates septic cardiomyopathy through ACSL4-dependent regulation of ferroptosis and mitochondrial integrity. Free Radic Biol Med 2024; 225:75-86. [PMID: 39326685 DOI: 10.1016/j.freeradbiomed.2024.09.039] [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: 09/04/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Sepsis evokes compromised myocardial function prompting heart failure albeit target therapy remains dismal. Our study examined the possible role of mitophagy receptor FUNDC1 in septic cardiomyopathy. A sepsis model was established using cecal ligation and puncture (CLP) in FUNDC1 knockout (FUNDC1-/-) and WT mice prior to the evaluation of cardiac morphology, echocardiographic and cardiomyocyte contractile, oxidative stress, apoptosis, necroptosis, and ferroptosis. RNAseq analysis depicted discrepant patterns in mitophagy, oxidative stress and ferroptosis between CLP-challenged and control murine hearts. Septic patients displayed cardiac injury alongside low plasma FUNDC1 and iron levels. CLP evoked interstitial fibrosis, cardiac dysfunction (lowered ejection fraction, fractional shortening, shortening/relengthening velocity, peak shortening and electrically-stimulated intracellular Ca2+ rise, alongside increased LV end systolic diameter and relengthening duration), O2- buildup, apoptosis, necroptosis, and ferroptosis (downregulated GPX4 and SLC7A11), the responses of which were accentuated by FUNDC1 ablation. In particular, levels of lipid peroxidation enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) were upregulated following CLP procedure, with a more pronounced response in FUNDC1-/- mice. Co-immunoprecipitation and interaction interface revealed an evident interaction between FUNDC1 and ACSL4. In vitro studies revealed that the endotoxin lipopolysaccharide provoked cardiomyocyte contractile and lipid peroxidation anomalies, the responses were reversed by the mitophagy inducer oleanolic acid, inhibition of ACSL4 and ferroptosis. These findings favor a role for FUNDC1-ACSL4-ferroptosis cascade in septic cardiomyopathy.
Collapse
Affiliation(s)
- Feng-Juan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China
| | - Huantao Hu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Liangyan Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou,510630,China
| | - Bijun Luo
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX, USA
| | - Shuyi Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Maolong Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jun Guo
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China.
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China; Department of Geriatrics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
11
|
Tian M, Huang X, Li M, Lou P, Ma H, Jiang X, Zhou Y, Liu Y. Ferroptosis in diabetic cardiomyopathy: from its mechanisms to therapeutic strategies. Front Endocrinol (Lausanne) 2024; 15:1421838. [PMID: 39588340 PMCID: PMC11586197 DOI: 10.3389/fendo.2024.1421838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is defined as structural and functional cardiac abnormalities in diabetes, and cardiomyocyte death is the terminal event of DCM. Ferroptosis is iron-dependent oxidative cell death. Evidence has indicated that iron overload and ferroptosis play important roles in the pathogenesis of DCM. Mitochondria, an important organelle in iron homeostasis and ROS production, play a crucial role in cardiomyocyte ferroptosis in diabetes. Studies have shown some anti-diabetic medicines, plant extracts, and ferroptosis inhibitors might improve DCM by alleviating ferroptosis. In this review, we systematically reviewed the evidence of ferroptosis in DCM. Anti-ferroptosis might be a promising therapeutic strategy for the treatment of DCM.
Collapse
Affiliation(s)
- Meimei Tian
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinli Huang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pingping Lou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Xinli Jiang
- Department of Ophthalmology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaru Zhou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Liu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
12
|
Liu Y, Zhang Z, Fang Y, Liu C, Zhang H. Ferroptosis in Osteoarthritis: Current Understanding. J Inflamm Res 2024; 17:8471-8486. [PMID: 39529997 PMCID: PMC11552513 DOI: 10.2147/jir.s493001] [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] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease in elderly people that is characterized by cartilage loss and abrasion, leading to joint pain and dysfunction. The aetiology of OA is complicated and includes abnormal mechanical stress, a mild inflammatory environment, chondrocyte senescence and apoptosis, and changes in chondrocyte metabolism. Ferroptosis is a regulated cell death modality characterized by the excessive accumulation of lipid peroxidation and mitochondrial dysfunction. The role of ferroptosis in OA pathogenesis has aroused researchers' attention in the past two years, and there is mounting evidence indicating that ferroptosis is destructive. However, the impact of ferroptosis on OA and how the regulators of ferroptosis affect OA development are unclear. Here, we reviewed the current understanding of ferroptosis in OA pathogenesis and summarized several drugs and compounds targeting ferroptosis in OA treatment. The accumulation of intracellular iron, the trigger of Fenton reaction, the excessive production of ROS, the peroxidation of PUFA-PLs, and mitochondrial and membrane damage are involved in chondrocyte ferroptosis. System Xc - and GPX4 are the most important regulators that control ferroptosis. Several compounds, such as DFO and Fer-1, have been proven effective in preventing ferroptosis and slowing OA progression on animal models. Collectively, targeting ferroptosis shows great potential in treating OA.
Collapse
Affiliation(s)
- Yikai Liu
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People’s Republic of China
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Zian Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Yuan Fang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Chang Liu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Haining Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| |
Collapse
|
13
|
Zhu J, Liang J. The role of COX2 deficiency attenuates cardiac damage in acute myocardial infarction. BMC Cardiovasc Disord 2024; 24:623. [PMID: 39511505 PMCID: PMC11542414 DOI: 10.1186/s12872-024-04233-y] [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: 03/19/2024] [Accepted: 10/03/2024] [Indexed: 11/15/2024] Open
Abstract
Cardiac cell damage frequently occurs as a consequence of acute myocardial infarction (AMI), a critical complication of coronary atherosclerotic heart disease. There is an escalating recognition of the association between COX2 and myocardial damage induced by ischemia. The objective of this study is to investigate the inhibitory effect of the COX2 on cardiomyocyte damage in the context of AMI. To create an AMI model, mice with the genetic background of wild-type C57BL6/J (WT) and COX2-/- mice were utilized. The left anterior descending (LAD) coronary artery in their hearts was obstructed, and subsequent assessment of hemodynamic parameters and heart function was conducted. Notably, increased levels of COX2 were observed in AMI mice. Through correlational analysis between COX2 expression and cardiac function following AMI, it was revealed that COX2 knockout mice had smaller infarct sizes, better cardiac performance, and suppressed levels of reactive oxygen species (ROS) compared to WT mice. Additionally, we discovered that COX2 knockout mice exhibited significantly higher mRNA levels of smooth muscle actin, collagen I, and collagen III than normal mice with AMI. Conversely, the levels of superoxide dismutase (SOD), malondialdehyde (MDA) were higher but iron content was decreased in COX2 knockout mice compared to normal mice with AMI.In summary, our research demonstrates that the downregulation of COX2 enhances cardiac tissue recovery in the context of AMI.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Cardiology, The Second People's Hospital of Foshan, Foshan, Guangdong, China
| | - Jianqiu Liang
- Department of Cardiology, The Second People's Hospital of Foshan, Foshan, Guangdong, China.
| |
Collapse
|
14
|
Yang Y, Zhang X, Yang Y, Gao P, Fan W, Zheng T, Yang W, Tang Y, Cai K. A two-pronged approach to inhibit ferroptosis of MSCs caused by the iron overload in postmenopausal osteoporosis and promote osseointegration of titanium implant. Bioact Mater 2024; 41:336-354. [PMID: 39161794 PMCID: PMC11331706 DOI: 10.1016/j.bioactmat.2024.07.024] [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: 03/31/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a prevalent condition among elderly women. After menopause, women exhibit decreased iron excretion, which is prone to osteoporosis. To design a specific titanium implant for PMOP, we first analyze miRNAs and DNA characteristics of postmenopausal patients with and without osteoporosis. The results indicate that iron overload disrupts iron homeostasis in the pathogenesis of PMOP. Further experiments confirm that iron overload can cause lipid peroxidation and ferroptosis of MSCs, thus breaking bone homeostasis. Based on the findings above, we have designed a novel Ti implant coated with nanospheres of caffeic acid (CA) and deferoxamine (DFO). CA can bind on the Ti surface through the two adjacent phenolic hydroxyls and polymerize into polycaffeic acid (PCA) dimer, as well as the PCA nanospheres with the repetitive 1,4-benzodioxan units. DFO was grafted with PCA through borate ester bonds. The experimental results showed that modified Ti can inhibit the ferroptosis of MSCs in the pathological environment of PMOP and promote osseointegration in two main ways. Firstly, DFO was released under high oxidative stress, chelating with excess iron and decreasing the labile iron pool in MSCs. Meanwhile, CA and DFO activated the KEAP1/NRF2/HMOX1 pathway in MSCs and reduced the level of intracellular lipid peroxidation. So, the ferroptosis of MSCs is inhibited by promoting the SLC7A11/GSH/GPX4 pathway. Furthermore, the remained CA coating on the Ti surface could reduce the extracellular oxidative stress and glutathione level. This study offers a novel inspiration for the specific design of Ti implants in the treatment of PMOP.
Collapse
Affiliation(s)
- Yulu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xianhui Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yao Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Pengfei Gao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wuzhe Fan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tao Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Weihu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yu Tang
- Orthopedics Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
15
|
Li Z, Xing J. Nuclear factor erythroid 2-related factor-mediated signaling alleviates ferroptosis during cerebral ischemia-reperfusion injury. Biomed Pharmacother 2024; 180:117513. [PMID: 39341075 DOI: 10.1016/j.biopha.2024.117513] [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: 07/25/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiac arrest (CA) is a significant challenge for emergency physicians worldwide and leads to increased morbidity and mortality rates. The poor prognosis of CA primarily stems from the complexity and irreversibility of cerebral ischemia-reperfusion injury (CIRI). Ferroptosis, a form of programmed cell death characterized by iron overload and lipid peroxidation, plays a crucial role in the progression and treatment of CIRI. In this review, we highlight the mechanisms of ferroptosis within the context of CIRI, focusing on its role as a key contributor to neuronal damage and dysfunction post-CA. We explore the crucial involvement of the nuclear factor erythroid 2-related factor (Nrf2)-mediated signaling pathway in modulating ferroptosis-associated processes during CIRI. Through comprehensive analysis of the regulatory role of Nrf2 in the cellular responses to oxidative stress, we highlight its potential as a therapeutic target for mitigating ferroptotic cell death and improving the neurological prognosis of patients experiencing CA. Furthermore, we discuss interventions targeting the Kelch-like ECH-associated protein 1/Nrf2/antioxidant response element pathway, including the use of traditional Chinese medicine and Western medicine, which demonstrate potential for attenuating ferroptosis and preserving neuronal function in CIRI. Owing to the limitations in the safety, specificity, and effectiveness of Nrf2-targeted drugs, as well as the technical difficulties and ethical constraints in obtaining the results related to the brain pathological examination of patients, most of the studies focusing on Nrf2-related regulation of ferroptosis in CIRI are still in the basic research stage. Overall, this review aims to provide a comprehensive understanding of the mechanisms underlying ferroptosis in CIRI, offering insights into novel therapeutics aimed at enhancing the clinical outcomes of patients with CA.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
16
|
Wu A, Yang H, Xiao T, Gu W, Li H, Chen P. COPZ1 regulates ferroptosis through NCOA4-mediated ferritinophagy in lung adenocarcinoma. Biochim Biophys Acta Gen Subj 2024; 1868:130706. [PMID: 39181476 DOI: 10.1016/j.bbagen.2024.130706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Ferroptosis, a type of autophagy-dependent cell death, has been implicated in the pathogenesis of lung adenocarcinoma (LUAD). This study aimed to investigate the involvement of coatomer protein complex I subunit zeta 1 (COPZ1) in ferroptosis and ferritinophagy in LUAD. METHODS Publicly available human LUAD sample data were obtained from the TCGA database to analyze the association of COPZ1 expression with LUAD grade and patient survival. Clinical samples of LUAD and para-carcinoma tissues were collected. COPZ1-deficient LUAD cell model and xenograft model were established. These models were analyzed to evaluate tumor growth, lipid peroxidation levels, mitochondrial structure, autophagy activation, and iron metabolism. RESULTS High expression of COPZ1 was indicative of malignancy and poor overall survival. Clinical LUAD tissues showed increased COPZ1 expression and decreased nuclear receptor coactivator 4 (NCOA4) expression. COPZ1 knockdown inhibited xenograft tumor growth and induced apoptosis. COPZ1 knockdown elevated the levels of ROS, Fe2+ and lipid peroxidation. COPZ1 knockdown also caused mitochondrial shrinkage. Liproxstatin-1, deferoxamine, and z-VAD-FMK reversed the effects of COPZ1 knockdown on LUAD cell proliferation and ferroptosis. Furthermore, COPZ1 was directly bound to NCOA4. COPZ1 knockdown restricted FTH1 expression and promoted NCOA4 and LC3 expression. NCOA4 knockdown reversed the regulation of iron metabolism, lipid peroxidation, and mitochondrial structure induced by COPZ1 knockdown. COPZ1 knockdown induced the translocation of ferritin to lysosomes for degradation, whereas NCOA4 knockdown disrupted this process. CONCLUSION This study provides novel evidence that COPZ1 regulates NCOA4-mediated ferritinophagy and ferroptosis. These findings provide new insights into the pathogenesis and potential treatment of LUAD.
Collapse
Affiliation(s)
- Anbang Wu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongmin Yang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tengfei Xiao
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wangnin Gu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - He Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; College of pharmacy, Changsha Medical University, Changsha 410219, China.
| | - Pan Chen
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
17
|
Liu Z, Liu C, Fan C, Li R, Zhang S, Liu J, Li B, Zhang S, Guo L, Wang X, Qi Z, Shen Y. E3 ubiquitin ligase DTX2 fosters ferroptosis resistance via suppressing NCOA4-mediated ferritinophagy in non-small cell lung cancer. Drug Resist Updat 2024; 77:101154. [PMID: 39366066 DOI: 10.1016/j.drup.2024.101154] [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: 07/11/2024] [Revised: 09/10/2024] [Accepted: 09/21/2024] [Indexed: 10/06/2024]
Abstract
Non-small cell lung cancer (NSCLC) remains the foremost contributor to cancer-related fatalities globally, with limited effective therapeutic modalities. Recent research has shed light on the role of ferroptosis in various types of cancers, offering a potential avenue for improving cancer therapy. Herein, we identified E3 ubiquitin ligase deltex 2 (DTX2) as a potential therapeutic target candidate implicated in promoting NSCLC cell growth by inhibiting ferroptosis. Our investigation revealed a significant upregulation of DTX2 in NSCLC cells and tissues, which was correlated with poor prognosis. Downregulation of DTX2 suppressed NSCLC cell growth both in vitro and in vivo, while its overexpression accelerated cell proliferation. Moreover, knockdown of DTX2 promoted ferroptosis in NSCLC cells, which was mitigated by DTX2 overexpression. Mechanistically, we uncovered that DTX2 binds to nuclear receptor coactivator 4 (NCOA4), facilitating its ubiquitination and degradation via the K48 chain, which subsequently dampens NCOA4-driven ferritinophagy and ferroptosis in NSCLC cells. Notably, DTX2 knockdown promotes cisplatin-induced ferroptosis and overcomes drug resistance of NSCLC cells. These findings underscore the critical role of DTX2 in regulating ferroptosis and NCOA4-mediated ferritinophagy, suggesting its potential as a novel therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Zhuang Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin Medical University, Tianjin 300203, China
| | - Chang Liu
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Caihong Fan
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Runze Li
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Shiqi Zhang
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Jia Liu
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Bo Li
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lihong Guo
- Department of Gastroenterology, Shengli Oilfield Central Hospital 257000, China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin Medical University, Tianjin 300203, China.
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300000, China; Department of Gastroenterology, Shengli Oilfield Central Hospital 257000, China; The First Department of Critical Care Medicine, The First Affiliated Hospital, Shihezi University 832000, China.
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
18
|
Deng Y, Zeng L, Liu H, Zuo A, Zhou J, Yang Y, You Y, Zhou X, Peng B, Lu H, Ji S, Wang M, Lai Y, Kwan HY, Sun X, Wang Q, Zhao X. Silibinin attenuates ferroptosis in acute kidney injury by targeting FTH1. Redox Biol 2024; 77:103360. [PMID: 39326069 PMCID: PMC11462067 DOI: 10.1016/j.redox.2024.103360] [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: 08/18/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Acute kidney injury (AKI) is primarily caused by renal ischemia-reperfusion injury (IRI), which is one of the most prevalent triggers. Currently, preventive and therapeutic measures remain limited. Ferroptosis plays a significant role in the pathophysiological process of IRI-induced AKI and is considered a key target for improving its outcomes. Silibinin, a polyphenolic flavonoid, possesses diverse pharmacological properties and is widely used as an effective therapeutic agent for liver diseases. Recent studies have reported that silibinin may improves kidney diseases, though the underlying mechanism remain unclear. In this study, we investigated whether silibinin protects against IRI-induced AKI and explored its mechanism of action. Our findings indicated that pretreatment with silibinin alleviated renal dysfunction, pathological damage, and inflammation in IRI-AKI mice. Furthermore, the results demonstrated that silibinin inhibited ferroptosis both in vivo and in vitro. Proteome microarrays were used to identify silibinin's target, and our results revealed that silibinin binds to FTH1. This binding affinity was confirmed through molecular docking, SPRi, CETSA, and DARTS. Additionally, co-IP assays demonstrated that silibinin disrupted the NCOA4-FTH1 interaction, inhibiting ferritinophagy. Finally, the inhibitory effects of silibinin on ferroptosis were reversed by knocking down FTH1 in vitro. In conclusion, our study shows that silibinin effectively alleviates AKI by targeting FTH1 to reduce ferroptosis, suggesting that silibinin could be developed as a potential therapeutic agent for managing and treating AKI.
Collapse
Affiliation(s)
- Yijian Deng
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Liying Zeng
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Huaxi Liu
- Boai Hospital of Zhongshan, Zhongshan, Guangdong, 528403, China
| | - Anna Zuo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jie Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ying Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yanting You
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xinghong Zhou
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, 523000, China
| | - Baizhao Peng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hanqi Lu
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, 523000, China
| | - Shuai Ji
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ming Wang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Yigui Lai
- People's Hospital of Yangjiang, Yangjiang, 529500, China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaomin Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Qi Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Xiaoshan Zhao
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
19
|
Zeng L, Jin X, Xiao QA, Jiang W, Han S, Chao J, Zhang D, Xia X, Wang D. Ferroptosis: action and mechanism of chemical/drug-induced liver injury. Drug Chem Toxicol 2024; 47:1300-1311. [PMID: 38148561 DOI: 10.1080/01480545.2023.2295230] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/12/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023]
Abstract
Drug-induced liver injury (DILI) is characterized by hepatocyte injury, cholestasis injury, and mixed injury. The liver transplantation is required for serious clinical outcomes such as acute liver failure. Current studies have found that many mechanisms were involved in DILI, such as mitochondrial oxidative stress, apoptosis, necroptosis, autophagy, ferroptosis, etc. Ferroptosis occurs when hepatocytes die from iron-dependent lipid peroxidation and plays a key role in DILI. After entry into the liver, where some drugs or chemicals are metabolized, they convert into hepatotoxic substances, consume reduced glutathione (GSH), and decrease the reductive capacity of GSH-dependent GPX4, leading to redox imbalance in hepatocytes and increase of reactive oxygen species (ROS) and lipid peroxidation level, leading to the undermining of hepatocytes; some drugs facilitated the autophagy of ferritin, orchestrating the increased ion level and ferroptosis. The purpose of this review is to summarize the role of ferroptosis in chemical- or drug-induced liver injury (chemical/DILI) and how natural products inhibit ferroptosis to prevent chemical/DILI.
Collapse
Affiliation(s)
- Li Zeng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Xueli Jin
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Qing-Ao Xiao
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Wei Jiang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Shanshan Han
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Jin Chao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Ding Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Xuan Xia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Department of Physiology and Pathophysiology, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Decheng Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| |
Collapse
|
20
|
Yuan C, Ma T, Liu M, Zeng X, Tang G, Xing Y, Zhang T. Ferroptosis, oxidative stress and hearing loss: Mechanistic insights and therapeutic opportunities. Heliyon 2024; 10:e38553. [PMID: 39512327 PMCID: PMC11541459 DOI: 10.1016/j.heliyon.2024.e38553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Hearing loss, a prevalent sensory impairment, poses significant challenges worldwide. Recent research has shed light on the intricate interplay between ferroptosis, a newly recognized form of regulated cell death characterized by iron-dependent lipid peroxidation, and oxidative stress in the pathogenesis of hearing loss. In this review, we delve into the mechanisms underlying ferroptosis and oxidative stress in various forms of hearing loss, including age-related hearing loss (ARHL), noise-induced hearing loss (NIHL) ototoxic drug-induced hearing loss and genetic hearing loss. We discuss the pivotal role of molecules such as FSP1, ACSL4, LKB1-AMPK, and Nrf2 in modulating these pathways in hearing loss. Furthermore, we explore emerging therapeutic strategies targeting the antioxidant system and ferroptosis, including iron chelators, lipid peroxide inhibitors, and antioxidants, highlighting their potential in mitigating hearing loss progression. By elucidating the molecular mechanisms underlying ferroptosis and oxidative stress, this review offers insights into novel therapeutic avenues for the treatment of hearing loss and underscores the importance of targeting these pathways to preserve auditory function.
Collapse
Affiliation(s)
- Chenyang Yuan
- Department of Otorhinolaryngology Head and Neck surgery, The First Hospital affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianyu Ma
- Department of Otorhinolaryngology Head and Neck surgery, The First Hospital affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Mengting Liu
- Department of Otorhinolaryngology Head and Neck surgery, The First Hospital affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoyun Zeng
- Department of Otorhinolaryngology Head and Neck surgery, The First Hospital affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Gongrui Tang
- Department of Otorhinolaryngology Head and Neck surgery, The First Hospital affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Yazhi Xing
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Rd, Shanghai, 200233, China
| | - Tianhong Zhang
- Department of Otorhinolaryngology Head and Neck surgery, The First Hospital affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
21
|
Kim JM, Kim DH, Kim WT, Shin SC, Cheon YI, Park GC, Lee HW, Lee BJ. Amifostine and Melatonin Prevent Acute Salivary Gland Dysfunction 10 Days After Radiation Through Anti-Ferroptosis and Anti-Ferritinophagy Effects. Int J Mol Sci 2024; 25:11613. [PMID: 39519165 PMCID: PMC11546762 DOI: 10.3390/ijms252111613] [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: 09/09/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Irradiation of the head and neck inevitably leads to decreased salivary gland function. It is postulated that radiation generates excessive reactive oxygen species (ROS) and reduces salivary gland function by ferroptosis, a new cell death mechanism; however, research in this area is currently lacking. In this study, we investigated the effects of amifostine and melatonin on acute salivary gland dysfunction and ferroptosis. Thirty-two Sprague Dawley rats were divided into four groups: control, radiation, radiation + amifostine, and radiation + melatonin. ROS; iron levels; glutathione peroxidase 4; 4-hydroxynonenal; various cytokines; and fibrosis and salivary gland functional markers were measured. Western blotting was used to detect ferritinophagy. After irradiation, we observed an increase in iron levels, ROS generation, oxidized glutathione, lipid peroxidation, fibrosis, and salivary gland dysfunction and a decrease in glutathione peroxidase 4 in salivary gland tissue. Treatment with amifostine or melatonin decreased the ferroptotic response and improved acute salivary gland function 10 days after radiation. The increase in iron levels associated with ferritinophagy was reduced after treatment with amifostine or melatonin. Our results demonstrate that radiation-induced acute salivary gland dysfunction is associated with ferroptosis and ferritinophagy. Amifostine and melatonin inhibit radiation-induced ferroptosis and ferritinophagy in the salivary gland and prevent acute salivary gland dysfunction 10 days after radiation.
Collapse
Affiliation(s)
- Ji-Min Kim
- Pusan National University Medical Research Institute, Pusan National University School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiation Oncology, Pusan National University School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Won-Taek Kim
- Department of Radiation Oncology, Pusan National University School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Sung-Chan Shin
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yong-il Cheon
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Gi-Cheol Park
- Department of Otolaryngology-Head and Neck Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Hyoun-Wook Lee
- Department of Pathology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Byung-Joo Lee
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
22
|
Zhang X, Liu Z, Li Z, Qi L, Huang T, Li F, Li M, Wang Y, Ma Z, Gao Y. Ferroptosis pathways: Unveiling the neuroprotective power of cistache deserticola phenylethanoid glycosides. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118465. [PMID: 38944360 DOI: 10.1016/j.jep.2024.118465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cistanche deserticola is a kind of parasitic plant living in the roots of desert trees. It is a rare Chinese medicine, which has the effect of tonifying kidney Yang, benefiting essence and blood and moistening the intestinal tract. Cistache deserticola phenylethanoid glycoside (PGS), an active component found in Cistanche deserticola Ma, have potential kidney tonifying, intellectual enhancing, and neuroprotective effects. Cistanche total glycoside capsule has been marketed to treat vascular dementia disease. AIM OF THE STUDY To identify the potential renal, intellectual enhancing and neuroprotective effects of PGS and explore the exact targets and mechanisms of PGS. MATERIALS AND METHODS This study systematically investigated the four types of pathways leading to ferroptosis through transcriptome, metabolome, ultrastructure and molecular biology techniques and explored the molecular mechanism by which multiple PGS targets and pathways synergistically exert neuroprotective effects on hypoxia. RESULTS PGS alleviated learning and memory dysfunction and pathological injury in mice exposed to hypobaric hypoxia by attenuating hypobaric hypoxia-induced hippocampal histopathological damage, impairing blood‒brain barrier integrity, increasing oxidative stress levels, and increasing the expression of cognitive proteins. PGS reduced the formation of lipid peroxides and improved ferroptosis by upregulating the GPX-4/SCL7A311 axis and downregulating the ACSL4/LPCAT3/LOX axis. PGS also reduced ferroptosis by facilitating cellular Fe2+ efflux and regulating mitochondrial Fe2+ transport and effectively antagonized cell ferroptosis induced by erastin (a ferroptosis inducer). CONCLUSIONS This study demonstrated the mechanism by which PGS prevents hypobaric hypoxic nerve injury through four types of ferroptosis pathways, achieved neuroprotective effects and alleviated learning and memory dysfunction in hypobaric hypoxia mice. This study provides a theoretical basis for the development and application of PGS.
Collapse
Affiliation(s)
- Xianxie Zhang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zuoxu Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zhihui Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Ling Qi
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Tianke Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Fang Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Maoxing Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Yuguang Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zengchun Ma
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Yue Gao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China.
| |
Collapse
|
23
|
Bao L, Zhao Y, Duan S, Wu K, Shan R, Liu Y, Yang Y, Chen Q, Song C, Li W. Ferroptosis is involved in Staphylococcus aureus-induced mastitis through autophagy activation by endoplasmic reticulum stress. Int Immunopharmacol 2024; 140:112818. [PMID: 39083924 DOI: 10.1016/j.intimp.2024.112818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Cell death caused by severe Staphylococcus aureus (S. aureus) infection is a fatal threat to humans and animals. However, whether ferroptosis, an iron-dependent form of cell death, is involved in S. aureus-induced cell death and its role in S. aureus-induced diseases are unclear. Using a mouse mastitis model and mammary epithelial cells (MMECs), we investigated the role of ferroptosis in the pathogenesis of S. aureus infection. The results revealed that S. aureus-induced ferroptosis in vivo and in vitro as demonstrated by dose-dependent increases in cell death; the level of malondialdehyde (MDA), the final product of lipid peroxidation; and dose-dependent decrease the production of the antioxidant glutathione (GSH). Treatment with typical inhibitors of ferroptosis, including ferrostatin-1 (Fer-1) and deferiprone (DFO), significantly inhibited S. aureus-induced death in MMECs. Mechanistically, treatment with S. aureus activated the protein kinase RNA-like ER kinase (PERK)-eukaryotic initiation factor 2, α subunit (eIF2α)-activating transcription factor 4 (ATF4)-C/EBP homologous protein (CHOP) pathway, which subsequently upregulated autophagy and promoted S. aureus-induced ferroptosis. The activation of autophagy degraded ferritin, resulting in iron dysregulation and ferroptosis. In addition, we found that excessive reactive oxygen species (ROS) production induced ferroptosis and activated endoplasmic reticulum (ER) stress, manifesting as elevated p-PERK-p-eIF2α-ATF4-CHOP pathway protein levels. Collectively, our findings indicate that ferroptosis is involved in S. aureus-induced mastitis via ER stress-mediated autophagy activation, implying a potential strategy for the prevention of S. aureus-associated diseases by targeting ferroptosis. In conclusion, the ROS-ER stress-autophagy axis is involved in regulating S. aureus-induced ferroptosis in MMECs. These findings not only provide a new potential mechanism for mastitis induced by S. aureus but also provide a basis for the treatment of other ferroptotic-related diseases.
Collapse
Affiliation(s)
- Lijuan Bao
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yihong Zhao
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Shiyu Duan
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Keyi Wu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Ruping Shan
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yi Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yang Yang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Qiujie Chen
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Changlong Song
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China.
| | - Wenjia Li
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China.
| |
Collapse
|
24
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-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] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Chen X, Chen H, Zhang P, Ju Q, Wu Z, Xu N, Bi Q, Yang S, Ji J, Yu D, Zhao Y. Coke oven emissions exacerbate allergic asthma by promoting ferroptosis in airway epithelial cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135542. [PMID: 39154481 DOI: 10.1016/j.jhazmat.2024.135542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Epidemiological studies have shown that coke oven emissions (COEs) affect the deterioration of asthma, but has not been proven by experimental results. In this study, we found for the first time that COEs exacerbate allergen house dust mite (HDM)-induced allergic asthma in the mouse model. The findings reveal that airway inflammation, airway remodeling and allergic reaction were aggravated in the COE + HDM combined exposure group compared with the individual exposure group. Mechanism studies indicated higher levels of iron and MDA in the COE + HDM combined exposure group, along with increased expression of Ptgs2 and reduced GPX4 expression. Iron chelator deferoxamine (DFO) effectively inhibited ferroptosis induced by COE synergistically with HDM in vitro. Further studies highlighted the role of ferritinophagy in the COE + HDM-induced ferroptosis. 3-methyladenine (3-MA) could inhibit ferroptosis in the COE + HDM exposure group. Interestingly, we injected DFO intraperitoneally into mice in the combined exposure group and found DFO could significantly inhibit the COE-exacerbated ferroptosis and allergic asthma. Our findings link ferroptosis with COE-exacerbated allergic asthma, implying that ferroptosis may have important therapeutic potential for asthma in patients with occupational exposure of COE.
Collapse
Affiliation(s)
- Xian Chen
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Hongguang Chen
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Pimei Zhang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Qiang Ju
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Zhaoxu Wu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Nuo Xu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Qing Bi
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Shuaishuai Yang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Jing Ji
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Dianke Yu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China
| | - Yanjie Zhao
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, Shandong 266021, China.
| |
Collapse
|
26
|
He X, Wang Z, Ge Q, Sun S, Li R, Wang B. Lactylation of nuclear receptor coactivator 4 promotes ferritinophagy and glycolysis of neuronal cells after cerebral ischemic injury. Neuroreport 2024; 35:895-903. [PMID: 39166386 DOI: 10.1097/wnr.0000000000002080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Ischemic stroke remains a major cause of disability and mortality. Nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy is involved in cerebral ischemic injury. Additionally, lactylation regulates the progression of ischemia injury. This study aimed to investigate the impact of NCOA4 on ferritinophagy and glycolysis of hippocampal neuron cells and its lactylation modification. Middle cerebral artery occlusion (MCAO) mouse and oxygen-glucose deprivation (OGD)-treated HT22 cell models were generated. Ferritinophagy was evaluated via detecting ferrous iron (Fe 2+ ), glutathione, malondialdehyde, and protein levels. Glycolysis was assessed by examining the glucose consumption, lactate production, and extracellular acidification rate. The lactylation was evaluated using immunoprecipitation and immunoblotting. Brain injury in vivo was analyzed by measuring brain infarct and neurological function. The results showed that NCOA4 expression was increased in the blood of patients with acute ischemia stroke, the peri-infarct region of the brain in MCAO mice (increased percentage: 142.11%) and OGD-treated cells (increased percentage: 114.70%). Knockdown of NCOA4 inhibited ferritinophagy and glycolysis of HT22 cells induced by OGD. Moreover, OGD promoted the lactylation of NCOA4 at lysine (K)450 sites, which enhanced NCOA4 protein stability. Additionally, interfering with NCOA4 attenuated brain infarction and neurological dysfunction in MCAO mice. Lactylation of NCOA4 at K450 sites promotes ferritinophagy and glycolysis of hippocampal neuron cells, thereby accelerating cerebral ischemic injury. These findings suggest a novel pathogenesis of ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyin He
- Department of Neurology, The Air Force Hospital of Southern Theater Command, PLA, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
27
|
Wu X, Du F, Zhang A, Zhang G, Xu R, Du X. KDELR2 is necessary for chronic obstructive pulmonary disease airway Mucin5AC hypersecretion via an IRE1α/XBP-1s-dependent mechanism. J Cell Mol Med 2024; 28:e70125. [PMID: 39365189 PMCID: PMC11451269 DOI: 10.1111/jcmm.70125] [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: 11/02/2023] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
Airway mucus hypersecretion, a crucial pathological feature of chronic obstructive pulmonary disease (COPD), contributes to the initiation, progression, and exacerbation of this disease. As a macromolecular mucin, the secretory behaviour of Mucin5AC (MUC5AC) is highly dependent on a series of modifying and folding processes that occur in the endoplasmic reticulum (ER). In this study, we focused on the ER quality control protein KDEL receptor (KDELR) and demonstrated that KDELR2 and MUC5AC were colocalized in the airway epithelium of COPD patients and COPD model rats. In addition, knockdown of KDELR2 markedly reduced the expression of MUC5AC both in vivo and in vitro and knockdown of ATF6 further decreased the levels of KDELR2. Furthermore, pretreatment with 4μ8C, an IRE1α inhibitor, led to a partial reduction in the expression of KDELR2 and MUC5AC both in vivo and in vitro, which indicated the involvement of IRE1α/XBP-1s in the upstream signalling cascade. Our study revealed that KDELR2 plays a crucial role in airway MUC5AC hypersecretion in COPD, which might be dependent on ATF6 and IRE1α/XBP-1s upstream signalling.
Collapse
Affiliation(s)
- Xiaojuan Wu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of Respiratory and Critical Care MedicineSuining Central HospitalSuiningSichuanChina
| | - Fawang Du
- Department of Respiratory and Critical Care MedicineSuining Central HospitalSuiningSichuanChina
| | - Aijie Zhang
- Basic Laboratory, Key Laboratory of Metabolic DiseasesSuining Central HospitalSuiningChina
| | - Guoyue Zhang
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Rui Xu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xianzhi Du
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
28
|
Zheng D, Jin S, Liu PS, Ye J, Xie X. Targeting ferroptosis by natural products in pathophysiological conditions. Arch Toxicol 2024; 98:3191-3208. [PMID: 38987487 DOI: 10.1007/s00204-024-03812-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Ferroptosis is a form of cell death that is induced by iron-mediated accumulation of lipid peroxidation. The involvement of ferroptosis in different pathophysiological conditions has offered new perspectives on potential therapeutic interventions. Natural products, which are widely recognized for their significance in drug discovery and repurposing, have shown great promise in regulating ferroptosis by targeting various ferroptosis players. In this review, we discuss the regulatory mechanisms of ferroptosis and its implications in different pathological conditions. We dissect the interactions between natural products and ferroptosis in cancer, ischemia/reperfusion, neurodegenerative diseases, acute kidney injury, liver injury, and cardiomyopathy, with an emphasis on the relevance of ferroptosis players to disease targetability.
Collapse
Affiliation(s)
- Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Pu-Ste Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Jianping Ye
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
29
|
Feng F, Luo R, Mu D, Cai Q. Ferroptosis and Pyroptosis in Epilepsy. Mol Neurobiol 2024; 61:7354-7368. [PMID: 38383919 DOI: 10.1007/s12035-024-04018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/02/2024] [Indexed: 02/23/2024]
Abstract
Epilepsy is sudden, recurrent, and transient central nervous system dysfunction caused by abnormal discharge of neurons in the brain. Ferroptosis and pyroptosis are newly discovered ways of programmed cell death. One of the characteristics of ferroptosis is the oxidative stress generated by lipid peroxides. Similarly, pyroptosis has unique pro-inflammatory properties. As both oxidative stress and neuroinflammation are significant contributors to the pathogenesis of epilepsy, increasing evidence shows that ferroptosis and pyroptosis are closely related to epilepsy. This article reviews the current comprehension of ferroptosis and pyroptosis and elucidates potential mechanisms by which ferroptosis and pyroptosis may contribute to epilepsy. In addition, we also highlight the possible interactions between ferroptosis and pyroptosis because they reportedly coexist in many diseases, and increasing studies have demonstrated the convergence of pathways between the two. This is of great significance for explaining the occurrence and development of epilepsy and provides a new therapeutic perspective for the treatment of epilepsy.
Collapse
Affiliation(s)
- Fan Feng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pediatrics, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatrics, Key Laboratory of Development and Maternal and Child Diseases of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Rong Luo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pediatrics, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatrics, Key Laboratory of Development and Maternal and Child Diseases of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pediatrics, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatrics, Key Laboratory of Development and Maternal and Child Diseases of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Qianyun Cai
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Pediatrics, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu, Sichuan, China.
- Department of Pediatrics, Key Laboratory of Development and Maternal and Child Diseases of Sichuan Province, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Zhang Y, Hu K, Shang Z, Yang X, Cao L. Ferroptosis: Regulatory mechanisms and potential targets for bone metabolism: A review. Medicine (Baltimore) 2024; 103:e39158. [PMID: 39331895 PMCID: PMC11441915 DOI: 10.1097/md.0000000000039158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2024] Open
Abstract
Bone homeostasis is a homeostasis process constructed by osteoblast bone formation and osteoclast bone resorption. Bone homeostasis imbalance and dysfunction are the basis for the development of various orthopedic diseases such as osteoporosis, osteoarthritis, and steroid-induced avascular necrosis of femoral head. Previous studies have demonstrated that ferroptosis can induce lipid peroxidation through the generation of reactive oxygen species, activate a number of signaling pathways, and participate in the regulation of osteoblast bone formation and osteoclast bone resorption, resulting in bone homeostasis imbalance, which is an important factor in the pathogenesis of many orthopedic diseases, but the mechanism of ferroptosis is still unknown. In recent years, it has been found that, in addition to iron metabolism and intracellular antioxidant system imbalance, organelle dysfunction is also a key factor affecting ferroptosis. This paper takes this as the starting point, reviews the latest literature reports at home and abroad, elaborates the pathogenesis and regulatory pathways of ferroptosis and the relationship between ferroptosis and various organelles, and summarizes the mechanism by which ferroptosis mediates bone homeostasis imbalance, with the aim of providing new directions for the research related to ferroptosis and new ideas for the prevention and treatment of bone and joint diseases.
Collapse
Affiliation(s)
- Yongjie Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kangyi Hu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengya Shang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaorui Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linzhong Cao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- The Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
31
|
Guo D, Liu Z, Zhou J, Ke C, Li D. Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9947. [PMID: 39337436 PMCID: PMC11432010 DOI: 10.3390/ijms25189947] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to promote neuronal development, establish neural architecture, and shape the central nervous system (CNS), although the role of PCD extends beyond the CNS. Abnormalities in PCD signaling cascades contribute to the irreversible loss of neuronal cells and function, leading to the onset and progression of neurodegenerative diseases. In this review, we summarize the molecular processes and features of different modalities of PCD, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other novel forms of PCD, and their effects on the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Additionally, we examine the key factors involved in these PCD signaling pathways and discuss the potential for their development as therapeutic targets and strategies. Therefore, therapeutic strategies targeting the inhibition or facilitation of PCD signaling pathways offer a promising approach for clinical applications in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Dong Guo
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Zhihao Liu
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Jinglin Zhou
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Chongrong Ke
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Daliang Li
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| |
Collapse
|
32
|
Zhang X, Dong X, Jie H, Li S, Li H, Su Y, Li L, Kang L, Dong B, Zhang Y. Downregulation of the (pro)renin receptor alleviates ferroptosis-associated cardiac pathological changes via the NCOA 4-mediated ferritinophagy pathway in diabetic cardiomyopathy. Int Immunopharmacol 2024; 138:112605. [PMID: 38963979 DOI: 10.1016/j.intimp.2024.112605] [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: 03/14/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Ferroptosis, characterized by the accumulation of reactive oxygen species and lipid peroxidation, is involved in various cardiovascular diseases. (Pro)renin receptor (PRR) in performs as ligands in the autophagic process, and its function in diabetic cardiomyopathy (DCM) is not fully understood. We investigated whether PRR promotes ferroptosis through the nuclear receptor coactivator 4 (NCOA 4)-mediated ferritinophagy pathway and thus contributes to DCM. We first established a mouse model of DCM with downregulated and upregulated PRR expression and used a ferroptosis inhibitor. Myocardial inflammation and fibrosis levels were then measured, cardiac function and ferroptosis-related indices were assessed. In vitro, neonatal rat ventricular primary cardiomyocytes were cultured with high glucose and transfected with recombinant adenoviruses knocking down or overexpressing the PRR, along with a ferroptosis inhibitor and small interfering RNA for the ferritinophagy receptor, NCOA4. Ferroptosis levels were measured in vitro. The results showed that the knockdown of PRR not only alleviated cardiomyocyte ferroptosis in vivo but also mitigated the HG-induced ferroptosis in vitro. Moreover, administration of Fer-1 can inhibit HG-induced ferroptosis. NCOA4 knockdown blocked the effect of PRR on ferroptosis and improved cell survival. Our result indicated that inhibition of PRR and NCOA4 expression provides a new therapeutic strategy for the treatment of DCM. The effect of PRR on the pathological process of DCM in mice may be in promoting cardiomyocyte ferroptosis through the NCOA 4-mediated ferritinophagy pathway.
Collapse
Affiliation(s)
- XinYu Zhang
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China
| | - XueFei Dong
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China
| | - HaiPeng Jie
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China
| | - ShengNan Li
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China
| | - HuiXin Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China; Department of Cardiology, Shandong University of Traditional Chinese Medicine, Jinan 250021, China
| | - YuDong Su
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China; Department of Cardiology, Shandong University of Traditional Chinese Medicine, Jinan 250021, China
| | - Lei Li
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China
| | - Li Kang
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China; Department of Cardiology, Shandong University of Traditional Chinese Medicine, Jinan 250021, China.
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250021, China.
| |
Collapse
|
33
|
Zhu X, Wang L, Wang K, Yao Y, Zhou F. Erdafitinib promotes ferroptosis in human uveal melanoma by inducing ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling axis. Free Radic Biol Med 2024; 222:552-568. [PMID: 38971541 DOI: 10.1016/j.freeradbiomed.2024.07.002] [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: 05/14/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Uveal melanoma (UM) is a rare yet lethal primary intraocular malignancy affecting adults. Analysis of data from The Cancer Genome Atlas (TCGA) database revealed that FGFR1 expression was increased in UM tumor tissues and was linked to aggressive behavior and a poor prognosis. This study assessed the anti-tumor effects of Erdafitinib, a selective pan-FGFR inhibitor, in both in vitro and in vivo UM models. Erdafitinib exhibited a robust anti-cancer activity in UM through inducing ferroptosis in the FGFR1-dependent manner. Transcriptomic data revealed that Erdafitinib mediated its anti-cancer effects via modulating the ferritinophagy/lysosome biogenesis. Subsequent research revealed that Erdafitinib exerted its effects by reducing the expression of FGFR1 and inhibiting the activity of mTORC1 in UM cells. Concurrently, it enhanced the dephosphorylation, nuclear translocation, and transcriptional activity of TFEB. The aggregation of TFEB in nucleus triggered FTH1-dependent ferritinophagy, leading to lysosomal activation and iron overload. Conversely, the overexpression of FGFR1 served to mitigate the effects of Erdafitinib on ferritinophagy, lysosome biogenesis, and the activation of the mTORC1/TFEB signaling pathway. In vivo experiments have convincingly shown that Erdafitinib markedly curtails tumor growth in an UM xenograft mouse model, an effect that is closely correlated with a decrease in FGFR1 expression levels. The present study is the first to demonstrate that Erdafitinib powerfully induces ferroptosis in UM by orchestrating the ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling. Consequently, Erdafitinib emerges as a strong candidate for clinical trial investigation, and FGFR1 emerges as a novel and promising therapeutic target in the treatment of UM.
Collapse
Affiliation(s)
- Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China.
| | - Ying Yao
- Department of Pharmacy, Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, Jiangsu Province, China.
| | - Fanfan Zhou
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
34
|
Zhou Y, Pang N, Li W, Li Q, Luo J, Gu Y, Hu Q, Ding YJ, Sun Y, Pan J, Gao M, Xiao Y, Ma S, Hao Y, Xing H, Fang EF, Ling W, Zhang Z, Yang L. Inhibition of ethanol-induced eNAMPT secretion attenuates liver ferroptosis through BAT-Liver communication. Redox Biol 2024; 75:103274. [PMID: 39059204 PMCID: PMC11327441 DOI: 10.1016/j.redox.2024.103274] [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: 06/25/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND & AIMS Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) has long been recognized as an adipokine. However, the exact role of eNAMPT in alcoholic liver disease (ALD) and its relevance to brown adipose tissue (BAT) remain largely unknown. This study aimed to evaluate the impact of eNAMPT on liver function and the underlying mechanisms involved in BAT-Liver communication. METHODS Serum eNAMPT levels were detected in the serum of both ALD patients and mice. Chronic and binge ethanol feeding was used to induce alcoholic liver injury in mice. An eNAMPT antibody, a coculture model of brown adipocytes and hepatocytes, and BAT-specific Nampt knockdown mice were used to investigate the role of eNAMPT in ALD. RESULTS Serum eNAMPT levels are elevated in ALD patients and are significantly positively correlated with the liver injury index. In ALD mice, neutralizing eNAMPT reduced the elevated levels of circulating eNAMPT induced by ethanol and attenuated liver injury. In vitro experiments revealed that eNAMPT induced hepatocyte ferroptosis through the TLR4-dependent mitochondrial ROS-induced ferritinophagy pathway. Furthermore, ethanol stimulated eNAMPT secretion from brown adipocytes but not from other adipocytes. In the coculture model, ethanol-induced release of eNAMPT from brown adipocytes promoted hepatocyte ferroptosis. In BAT-specific Nampt-knockdown mice, ethanol-induced eNAMPT secretion was significantly reduced, and alcoholic liver injury were attenuated. These effects can be reversed by intraperitoneal injection of eNAMPT. CONCLUSION Inhibition of ethanol-induced eNAMPT secretion from BAT attenuates liver injury and ferroptosis. Our study reveals a previously uncharacterized critical role of eNAMPT-mediated BAT-Liver communication in ALD and highlights its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yujia Zhou
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Nengzhi Pang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenli Li
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Immunization Programs, Guangzhou Huadu District Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Qiuyan Li
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Luo
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingying Gu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qianrong Hu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Women Health Care, Guangzhou Baiyun District Maternal and Child Health Hospital, Guangzhou, Guangdong, China
| | - Yi Jie Ding
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Sun
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jie Pan
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengqi Gao
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Xiao
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sixi Ma
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanxu Hao
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Huichun Xing
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, China; Peking University Ditan Teaching Hospital, Beijing, China
| | - Evendro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
| | - Wenhua Ling
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenfeng Zhang
- Department of Radiology, Translational Medicine Center and Guangdong Provincial Education Department, Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Lili Yang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
35
|
Wang S, Wu Y, Yang F, Hsu F, Zhang K, Hung J. NCI677397 targeting USP24-mediated induction of lipid peroxidation induces ferroptosis in drug-resistant cancer cells. Mol Oncol 2024; 18:2255-2276. [PMID: 38140768 PMCID: PMC11467797 DOI: 10.1002/1878-0261.13574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/20/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer represents a profound challenge to healthcare systems and individuals worldwide. The development of multiple drug resistance is a major problem in cancer therapy and can result in progression of the disease. In our previous studies, we developed small-molecule inhibitors targeting ubiquitin-specific peptidase 24 (USP24) to combat drug-resistant lung cancer. Recently, we found that the USP24 inhibitor NCI677397 induced ferroptosis, a type of programmed cell death, in drug-resistant cancer cells by increasing lipid reactive oxygen species (ROS) levels. In the present study, we investigated the molecular mechanisms and found that the targeting of USP24 by NCI677397 increased gene expression of most lipogenesis-related genes, such as acyl-CoA synthetase long-chain family member 4 (ACSL4), and activated autophagy. In addition, the activity of several antioxidant enzymes, such as glutathione peroxidase 4 (GPX4) and dihydrofolate reductase (DHFR), was inhibited by NCI677397 treatment via an increase in protein degradation, thereby inducing lipid ROS production and lipid peroxidation. In summary, we demonstrated that NCI677397 induced a marked increase in lipid ROS levels, subsequently causing lipid peroxidation and leading to the ferroptotic death of drug-resistant cancer cells. Our study provides new insights into the clinical use of USP24 inhibitors as ferroptosis inducers (FINs) to block drug resistance during chemotherapy.
Collapse
Affiliation(s)
- Shao‐An Wang
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Yu‐Chih Wu
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Feng‐Ming Yang
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Feng‐Lin Hsu
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Kuan Zhang
- Cardiovascular Research InstituteUniversity of California, San FranciscoCAUSA
| | - Jan‐Jong Hung
- Department of Biotechnology and Bioindustry SciencesNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
36
|
Ye H, Wu L, Liu Y. Iron metabolism in doxorubicin-induced cardiotoxicity: From mechanisms to therapies. Int J Biochem Cell Biol 2024; 174:106632. [PMID: 39053765 DOI: 10.1016/j.biocel.2024.106632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Doxorubicin (DOX) is an anti-tumor agent for chemotherapy, but its use is often hindered by the severe and life-threatening side effect of cardiovascular toxicity. In recent years, studies have focused on dysregulated iron metabolism and ferroptosis, a unique type of cell death induced by iron overload, as key players driving the development of DOX-induced cardiotoxicity (DIC). Recent advances have demonstrated that DOX disturbs normal cellular iron metabolism, resulting in excessive iron accumulation and ferroptosis in cardiomyocytes. This review will explore how dysregulated iron homeostasis and ferroptosis drive the progression of DIC. We will also discuss the current approaches to target iron metabolism and ferroptosis to mitigate DIC. Besides, we will discuss the limitations and challenges for clinical translation for these therapeutic regimens.
Collapse
Affiliation(s)
- Hua Ye
- Department of Burns & Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China.
| | - Lin Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yanmei Liu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
37
|
Moscovicz F, Taborda C, Fernández F, Borda N, Auzmendi J, Lazarowski A. Ironing out the Links: Ferroptosis in epilepsy and SUDEP. Epilepsy Behav 2024; 157:109890. [PMID: 38905915 DOI: 10.1016/j.yebeh.2024.109890] [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: 02/26/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/23/2024]
Abstract
Iron is a crucial element for almost all organisms because it plays a vital role in oxygen transport, enzymatic processes, and energy generation due to its electron transfer capabilities. However, its dysregulation can lead to a form of programmed cell death known as ferroptosis, which is characterized by cellular iron accumulation, reactive oxygen species (ROS) production, and unrestricted lipid peroxidation. Both iron and ferroptosis have been identified as key players in the pathogenesis of various neurodegenerative diseases. While in epilepsy this phenomenon remains relatively understudied, seizures can be considered hypoxic-ischemic episodes resulting in increased ROS production, lipid peroxidation, membrane disorganization, and cell death. All of this is accompanied by elevated intracellular free Fe2+ concentration and hemosiderin precipitation, as existing reports suggest a significant accumulation of iron in the brain and heart associated with epilepsy. Generalized tonic-clonic seizures (GTCS), a primary risk factor for Sudden Unexpected Death in Epilepsy (SUDEP), not only have an impact on the brain but also lead to cardiogenic dysfunctions associated with "Iron Overload and Cardiomyopathy" (IOC) and "Epileptic heart" characterized by electrical and mechanical dysfunction and a high risk of malignant bradycardia. In line with this phenomenon, studies conducted by our research group have demonstrated that recurrent seizures induce hypoxia in cardiomyocytes, resulting in P-glycoprotein (P-gp) overexpression, prolonged Q-T interval, severe bradycardia, and hemosiderin precipitation, correlating with an elevated spontaneous death ratio. In this article, we explore the intricate connections among ferroptosis, epilepsy, and SUDEP. By synthesizing current knowledge and drawing insights from recent publications, this study provides a comprehensive understanding of the molecular underpinnings. Furthermore, this review offers insights into potential therapeutic avenues and outlines future research directions.
Collapse
Affiliation(s)
- F Moscovicz
- University of Buenos Aires, Faculty of Pharmacy and Biochemistry, Institute of Phisiopatology and Clinical Biochemistry (INFIBIOC), Applied Neurobiology Lab, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Argentina.
| | - C Taborda
- University of Buenos Aires, Faculty of Pharmacy and Biochemistry, Institute of Phisiopatology and Clinical Biochemistry (INFIBIOC), Applied Neurobiology Lab, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Argentina
| | - F Fernández
- University of Buenos Aires, Faculty of Pharmacy and Biochemistry, Institute of Phisiopatology and Clinical Biochemistry (INFIBIOC), Applied Neurobiology Lab, Buenos Aires, Argentina
| | - N Borda
- University of Buenos Aires, Faculty of Pharmacy and Biochemistry, Institute of Phisiopatology and Clinical Biochemistry (INFIBIOC), Applied Neurobiology Lab, Buenos Aires, Argentina
| | - J Auzmendi
- University of Buenos Aires, Faculty of Pharmacy and Biochemistry, Institute of Phisiopatology and Clinical Biochemistry (INFIBIOC), Applied Neurobiology Lab, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Argentina.
| | - A Lazarowski
- University of Buenos Aires, Faculty of Pharmacy and Biochemistry, Institute of Phisiopatology and Clinical Biochemistry (INFIBIOC), Applied Neurobiology Lab, Buenos Aires, Argentina.
| |
Collapse
|
38
|
Yin Z, Lv Z, Yang L, Li C, Teng F, Liang W. The nuclear receptor coactivator 4 regulates ferritinophagy induced by vibrio splendidus in coelomocytes of Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109745. [PMID: 38960105 DOI: 10.1016/j.fsi.2024.109745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
Iron homeostasis is vital for the host's defense against pathogenic invasion and the ferritinophagy is a crucial mechanism in maintaining intracellular iron homeostasis by facilitating the degradation and recycling of stored iron. The nuclear receptor coactivator 4 (NCOA4) serves as a ferritinophagy receptor, facilitating the binding and delivery of ferritin to the autophagosome and lysosome. However, NCOA4 of the sea cucumber Apostichopus japonicus (AjNCOA4) has not been reported until now. In this study, we identified and characterized AjNCOA4 in A. japonicus. This gene encodes a polypeptide containing 597 amino acids with an open reading frame of 1794 bp. The inferred amino acid sequence of AjNCOA4 comprises an ARA70 domain. Furthermore, a multiple sequence alignment demonstrated varying degrees of sequence homology between AjNCOA4 from A. japonicus and other NCOA4 orthologs. The phylogenetic tree of NCOA4 correlates with the established timeline of metazoan evolution. Expression analysis revealed that AjNCOA4 is expressed in all tested tissues, including the body wall, muscle, intestine, respiratory tree, and coelomocytes. Following challenge with Vibrio splendidus, the coelomocytes exhibited a significant increase in AjNCOA4 mRNA levels, peaking at 24 h. We successfully obtained recombinant AjNCOA4 protein through prokaryotic expression and prepared a specific polyclonal antibody. Immunofluorescence and co-immunoprecipitation experiments demonstrated an interaction between AjNCOA4 and AjFerritin in coelomocytes. RNA interference-mediated knockdown of AjNCOA4 expression resulted in elevated iron ion levels in coelomocytes. Bacterial stimulation enhanced ferritinophagy in coelomocytes, while knockdown of AjNCOA4 reduced the occurrence of ferritinophagy. These findings suggest that AjNCOA4 modulates ferritinophagy induced by V. splendidus in coelomocytes of A. japonicus.
Collapse
Affiliation(s)
- Zhiqun Yin
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Zhimeng Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Lei Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, 266071, China.
| | - Fei Teng
- College of Mathematics and Computer, Jilin Normal University, Siping, Jilin, China
| | - Weikang Liang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
39
|
Li FJ, Abudureyimu M, Zhang ZH, Tao J, Ceylan AF, Lin J, Yu W, Reiter RJ, Ashrafizadeh M, Guo J, Ren J. Inhibition of ER stress using tauroursodeoxycholic acid rescues obesity-evoked cardiac remodeling and contractile anomalies through regulation of ferroptosis. Chem Biol Interact 2024; 398:111104. [PMID: 38906502 DOI: 10.1016/j.cbi.2024.111104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
Interrupted ER homeostasis contributes to the etiology of obesity cardiomyopathy although it remains elusive how ER stress evokes cardiac anomalies in obesity. Our study evaluated the impact of ER stress inhibition on cardiac anomalies in obesity. Lean and ob/ob obese mice received chemical ER chaperone tauroursodeoxycholic acid (TUDCA, 50 mg/kg/d, p.o.) for 35 days prior to evaluation of glucose sensitivity, echocardiographic, myocardial geometric, cardiomyocyte mechanical and subcellular Ca2+ property, mitochondrial integrity, oxidative stress, apoptosis, and ferroptosis. Intracellular Ca2+ governing domains including sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) were monitored by45Ca2+uptake and immunoblotting. Our results noted that TUDCA alleviated myocardial remodeling (fibrosis, hypertrophy, enlarged LVESD), echocardiographic anomalies (compromised fractional shortening and ejection fraction), cardiomyocyte contractile dysfunction (amplitude and velocity of cell shortening, relengthening time) and intracellular Ca2+ anomalies (compromised subcellular Ca2+ release, clearance and SERCA function), mitochondrial damage (collapsed membrane potential, downregulated mitochondrial elements and ultrastructural alteration), ER stress (GRP78, eIF2α and ATF4), oxidative stress, apoptosis and ferroptosis [downregulated SLC7A11, GPx4 and upregulated transferrin receptor (TFRC)] without affecting global glucose sensitivity and serum Fe2+ in obese mice. Obesity-evoked change in HSP90, phospholamban and Na+-Ca2+ exchanger was spared by the chemical ER chaperone. Moreover, in vitro results noted that TUDCA, PERK inhibitor GSK2606414, TFRC neutralizing antibody and ferroptosis inhibitor LIP1 mitigated palmitic acid-elicited changes in lipid peroxidation and mechanical function. Our findings favored a role for ferroptosis in obesity cardiomyopathy downstream of ER stress.
Collapse
Affiliation(s)
- Feng-Juan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China
| | - Miyesaier Abudureyimu
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China
| | - Zeng-Hui Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Asli F Ceylan
- Ankara Yildirim Beyazit University, Faculty of Medicine, Department of Medical Pharmacology, Bilkent, Ankara, Turkey
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Wei Yu
- Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, 437100, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX, USA
| | - Milad Ashrafizadeh
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jun Guo
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510660, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
40
|
Shang Y, Zhao M, Chen S, Chen Y, Liu X, Zhou F, Li Y, Long M, Xu K, Ding Z, Wang L. Tetrastigma hemsleyanum polysaccharide combined with doxorubicin promote ferroptosis and immune function in triple-negative breast cancer. Int J Biol Macromol 2024; 275:133424. [PMID: 38945330 DOI: 10.1016/j.ijbiomac.2024.133424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
The absence of effective therapeutic targets poses considerable obstacles to the treatment of triple-negative breast cancer (TNBC). This study aimed to explore the function and mechanism of polysaccharides derived from the aerial parts of Tetrastigma hemsleyanum (THP) for the treatment of TNBC. THP exerts notable anti-TNBC effects when used alone, and its combination with Doxorubicin (DOX) effectively augments the sensitivity of TNBC cells to DOX. Through RNA sequencing, Fe2+ assays, western blotting, and transmission electron microscopy, THP was identified as a natural inducer of ferroptosis and ferritinophagy through the xCT/GSH/GPX4 and Nrf2/NCOA4/FTH1 pathways. Further research revealed that the THP branched-chain hexose directly binds to the xCT protein to inhibit its expression and promotes ferroptosis. In vivo experiments confirmed the role of THP in inducing ferroptosis and showed that THP improves the tumor microenvironment and immune function by increasing the ratio of CD4+ and CD8+ T cells to regulatory T cells and modulating cytokine levels. As demonstrated by electrocardiography, blood chemistry, and histological analyses, THP alleviates organ toxicity caused by DOX. Overall, these results suggest that THP has significant clinical potential as a natural macromolecular drug and may provide a safe and effective treatment strategy for TNBC when combined with DOX.
Collapse
Affiliation(s)
- Yini Shang
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Pathophysiology, Medical College of Southeast University, Nanjing 210009, China
| | - Mengjia Zhao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Senmiao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yuchi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xia Liu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yiping Li
- Department of Pathophysiology, Medical College of Southeast University, Nanjing 210009, China
| | - Min Long
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Keying Xu
- Department of Pathophysiology, Medical College of Southeast University, Nanjing 210009, China
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Lihong Wang
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Pathophysiology, Medical College of Southeast University, Nanjing 210009, China.
| |
Collapse
|
41
|
Hong Y, Yuan Q, Wang L, Yang Z, Xu P, Guan X, Chen C. Integrative bioinformatics analysis to identify ferroptosis-related genes in non-obstructive azoospermia. J Assist Reprod Genet 2024; 41:2145-2161. [PMID: 38902567 PMCID: PMC11339017 DOI: 10.1007/s10815-024-03155-0] [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: 03/05/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
PURPOSE The objective of this study was to discern ferroptosis-related genes (FRGs) linked to non-obstructive azoospermia and investigate the associated molecular mechanisms. METHOD A dataset related to azoospermia was retrieved from the Gene Expression Omnibus database, and FRGs were sourced from GeneCards. Ferroptosis-related differentially expressed genes (FRDEGs) were discerned. Subsequently, these genes underwent analyses encompassing Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, as well as protein-protein interaction (PPI) networks and assessments of functional similarity. Following the identification of hub genes, an exploration of immune infiltration, single-cell expression, diagnostic utility, and interactions involving hub genes, RNA-binding proteins (RBPs), transcription factors (TFs), microRNAs (miRNAs), and drugs was conducted. RESULTS A total of 35 differentially expressed FRGs were discerned. These genes demonstrated enrichment in functions and pathways associated with ferroptosis. From the PPI network, eight hub genes were selected. Functional similarity analysis highlighted the potential pivotal roles of HMOX1 and GPX4 in azoospermia. Analysis of immune cell infiltration indicated a significant decrease in activated dendritic cells in the azoospermia group, with notable correlations between hub genes, particularly SAT1 and HMGCR, and immune cell infiltration. Unique expression patterns of hub genes across various cell types in the human testis were observed, with GPX4 prominently enriched in spermatid/sperm. Eight hub genes exhibited robust diagnostic value (AUC > 0.75). Lastly, a comprehensive hub gene-miRNA-TF-RBP-drug network was constructed. CONCLUSION In summary, our investigation unveiled eight FRDEGs associated with azoospermia, which hold potential as biomarkers for the diagnosis and treatment of azoospermia.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China
| | - Qichao Yuan
- Department of Pediatric Urology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China
| | - Lingfei Wang
- Department of Pediatric Urology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China
| | - Zihan Yang
- Department of Pediatric Urology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China
| | - Peiyu Xu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China
| | - Xiaoju Guan
- Department of Pediatric Urology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China.
| | - Congde Chen
- Department of Pediatric Urology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
42
|
Zuo Z, Luo M, Liu Z, Liu T, Wang X, Huang X, Li S, Wu H, Pan Q, Chen T, Yang L, Liu HF. Selenium nanoparticles alleviate renal ischemia/reperfusion injury by inhibiting ferritinophagy via the XBP1/NCOA4 pathway. Cell Commun Signal 2024; 22:376. [PMID: 39061070 PMCID: PMC11282718 DOI: 10.1186/s12964-024-01751-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Acute kidney injury (AKI) is closely related to lysosomal dysfunction and ferroptosis in renal tubular epithelial cells (TECs), for which effective treatments are urgently needed. Although selenium nanoparticles (SeNPs) have emerged as promising candidates for AKI therapy, their underlying mechanisms have not been fully elucidated. Here, we investigated the effect of SeNPs on hypoxia/reoxygenation (H/R)-induced ferroptosis and lysosomal dysfunction in TECs in vitro and evaluated their efficacy in a murine model of ischemia/reperfusion (I/R)-AKI. We observed that H/R-induced ferroptosis was accompanied by lysosomal Fe2+ accumulation and dysfunction in TECs, which was ameliorated by SeNPs administration. Furthermore, SeNPs protected C57BL/6 mice against I/R-induced inflammation and ferroptosis. Mechanistically, we found that lysosomal Fe2+ accumulation and ferroptosis were associated with the excessive activation of NCOA4-mediated ferritinophagy, a process mitigated by SeNPs through the upregulation of X-box binding protein 1 (XBP1). Downregulation of XBP1 promoted ferritinophagy and partially counteracted the protective effects of SeNPs on ferroptosis inhibition in TECs. Overall, our findings revealed a novel role for SeNPs in modulating ferritinophagy, thereby improving lysosomal function and attenuating ferroptosis of TECs in I/R-AKI. These results provide evidence for the potential application of SeNPs as therapeutic agents for the prevention and treatment of AKI.
Collapse
Affiliation(s)
- Zhenying Zuo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mianna Luo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhongyu Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ting Liu
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Xi Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Huang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shangmei Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hongluan Wu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
43
|
Yamada K, St Croix C, Stolz DB, Tyurina YY, Tyurin VA, Bradley LR, Kapralov AA, Deng Y, Zhou X, Wei Q, Liao B, Fukuda N, Sullivan M, Trudeau J, Ray A, Kagan VE, Zhao J, Wenzel SE. Compartmentalized mitochondrial ferroptosis converges with optineurin-mediated mitophagy to impact airway epithelial cell phenotypes and asthma outcomes. Nat Commun 2024; 15:5818. [PMID: 38987265 PMCID: PMC11237105 DOI: 10.1038/s41467-024-50222-2] [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: 07/11/2023] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
A stable mitochondrial pool is crucial for healthy cell function and survival. Altered redox biology can adversely affect mitochondria through induction of a variety of cell death and survival pathways, yet the understanding of mitochondria and their dysfunction in primary human cells and in specific disease states, including asthma, is modest. Ferroptosis is traditionally considered an iron dependent, hydroperoxy-phospholipid executed process, which induces cytosolic and mitochondrial damage to drive programmed cell death. However, in this report we identify a lipoxygenase orchestrated, compartmentally-targeted ferroptosis-associated peroxidation process which occurs in a subpopulation of dysfunctional mitochondria, without promoting cell death. Rather, this mitochondrial peroxidation process tightly couples with PTEN-induced kinase (PINK)-1(PINK1)-Parkin-Optineurin mediated mitophagy in an effort to preserve the pool of functional mitochondria and prevent cell death. These combined peroxidation processes lead to altered epithelial cell phenotypes and loss of ciliated cells which associate with worsened asthma severity. Ferroptosis-targeted interventions of this process could preserve healthy mitochondria, reverse cell phenotypic changes and improve disease outcomes.
Collapse
Affiliation(s)
- Kazuhiro Yamada
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, 545-8585, Japan
| | - Claudette St Croix
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Laura R Bradley
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alexander A Kapralov
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yanhan Deng
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiuxia Zhou
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Qi Wei
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Bo Liao
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Otolaryngology-Head & Neck Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nobuhiko Fukuda
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Mara Sullivan
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - John Trudeau
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Anuradha Ray
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jinming Zhao
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
44
|
Feng Z, Luan M, Zhu W, Xing Y, Ma X, Wang Y, Jia Y. Targeted ferritinophagy in gastrointestinal cancer: from molecular mechanisms to implications. Arch Toxicol 2024; 98:2007-2018. [PMID: 38602537 DOI: 10.1007/s00204-024-03745-y] [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: 02/27/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Gastrointestinal cancer is a significant global health burden, necessitating the development of novel therapeutic strategies. Emerging evidence has highlighted the potential of targeting ferritinophagy as a promising approach for the treatment of gastrointestinal cancer. Ferritinophagy is a form of selective autophagy that is mediated by the nuclear receptor coactivator 4 (NCOA4). This process plays a crucial role in regulating cellular iron homeostasis and has been implicated in various pathological conditions, including cancer. This review discusses the molecular mechanisms underlying ferritinophagy and its relevance to gastrointestinal cancer. Furthermore, we highlight the potential therapeutic implications of targeting ferritinophagy in gastrointestinal cancer. Several approaches have been proposed to modulate ferritinophagy, including small molecule inhibitors and immunotherapeutic strategies. We discuss the advantages and challenges associated with these therapeutic interventions and provide insights into their potential clinical applications.
Collapse
Affiliation(s)
- Zhaotian Feng
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yanfei Jia
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China.
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China.
| |
Collapse
|
45
|
Su Y, Jiao Y, Cai S, Xu Y, Wang Q, Chen X. The molecular mechanism of ferroptosis and its relationship with Parkinson's disease. Brain Res Bull 2024; 213:110991. [PMID: 38823725 DOI: 10.1016/j.brainresbull.2024.110991] [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] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024]
Abstract
Neurodegenerative diseases such as Parkinson's disease (PD) have complex pathogenetic mechanisms. Genetic, age, and environmental factors are all related to PD. Due to the unclear pathogenesis of PD and the lack of effective cure methods, it is urgent to find new targets for treating PD patients. Ferroptosis is a form of cell death that is reliant on iron and exhibits distinct morphological and mechanistic characteristics compared to other types of cell death. It encompasses a range of biological processes, including iron/lipid metabolism and oxidative stress. In recent years, research has found that ferroptosis plays a crucial role in the pathophysiological processes of neurodegenerative diseases and stroke. Therefore, ferroptosis is also closely related to PD, This article reviews the core mechanisms of ferroptosis and elucidates the correlation between PD and ferroptosis. In addition, new compounds that have emerged in recent years to exert anti PD effects by inhibiting the ferroptosis signaling pathway were summarized. I hope to further elaborate the relationship between ferroptosis and PD through the review of this article, and provide new strategies for developing PD treatments targeting ferroptosis.
Collapse
Affiliation(s)
- Yan Su
- Department of neurology, The First Affiliated hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Yue Jiao
- Department of neurology, The First Affiliated hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Sheng Cai
- Department of neurology, The First Affiliated hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Yang Xu
- Department of neurology, The First Affiliated hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Qi Wang
- Department of neurology, The First Affiliated hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Xianwen Chen
- Department of neurology, The First Affiliated hospital of Anhui Medical University, Hefei, Anhui, 230001, China.
| |
Collapse
|
46
|
Yan H, Shen X, Yao Y, Khan SA, Ma S, Johnson CH. A machine learning and drug repurposing approach to target ferroptosis in colorectal cancer stratified by sex and KRAS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600340. [PMID: 38979294 PMCID: PMC11230177 DOI: 10.1101/2024.06.24.600340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The landscape of sex differences in Colorectal Cancer (CRC) has not been well characterized with respect to the mechanisms of action for oncogenes such as KRAS. However, our recent study showed that tumors from male patients with KRAS mutations have decreased iron-dependent cell death called ferroptosis. Building on these findings, we further examined ferroptosis in CRC, considering both sex of the patient and KRAS mutations, using public databases and our in-house CRC tumor cohort. Through subsampling inference and variable importance analysis (VIMP), we identified significant differences in gene expression between KRAS mutant and wild type tumors from male patients. These genes suppress (e.g., SLC7A11 ) or drive (e.g., SLC1A5 ) ferroptosis, and these findings were further validated with Gaussian mixed models. Furthermore, we explored the prognostic value of ferroptosis regulating genes and discovered sex- and KRAS-specific differences at both the transcriptional and metabolic levels by random survival forest with backward elimination algorithm (RSF-BE). Of note, genes and metabolites involved in arginine synthesis and glutathione metabolism were uniquely associated with prognosis in tumors from males with KRAS mutations. Additionally, drug repurposing is becoming popular due to the high costs, attrition rates, and slow pace of new drug development, offering a way to treat common and rare diseases more efficiently. Furthermore, increasing evidence has shown that ferroptosis inhibition or induction can improve drug sensitivity or overcome chemotherapy drug resistance. Therefore, we investigated the correlation between gene expression, metabolite levels, and drug sensitivity across all CRC primary tumor cell lines using data from the Genomics of Drug Sensitivity in Cancer (GDSC) resource. We observed that ferroptosis suppressor genes such as DHODH , GCH1 , and AIFM2 in KRAS mutant CRC cell lines were resistant to cisplatin and paclitaxel, underscoring why these drugs are not effective for these patients. The comprehensive map generated here provides valuable biological insights for future investigations, and the findings are supported by rigorous analysis of large-scale publicly available data and our in-house cohort. The study also emphasizes the potential application of VIMP, Gaussian mixed models, and RSF-BE models in the multi-omics research community. In conclusion, this comprehensive approach opens doors for leveraging precision molecular feature analysis and drug repurposing possibilities in KRAS mutant CRC.
Collapse
|
47
|
Gao X, Mao H, Zhao L, Li X, Liao Y, Li W, Li H, Chen Y. Nuciferine Protects Cochlear Hair Cells from Ferroptosis through Inhibiting NCOA4-Mediated Ferritinophagy. Antioxidants (Basel) 2024; 13:714. [PMID: 38929153 PMCID: PMC11201048 DOI: 10.3390/antiox13060714] [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: 04/17/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Cisplatin is a widely used antineoplastic drug for treating various types of cancers. However, it can cause severe side effects, such as bilateral and irreversible hearing loss, which significantly impacts quality of life. Ferroptosis, an iron-dependent form of programmed cell death, has been implicated in the pathogenesis of cisplatin-induced ototoxicity. Here, we investigated the effects of nuciferine, a natural active ingredient isolated from lotus species, on the ferroptosis of cochlear hair cells. Firstly, our results demonstrated that nuciferine can protect hair cells against RSL3-induced and cisplatin-induced damage. Secondly, nuciferine treatment reduced ferrous iron (Fe2+) overload in cochlear hair cells via inhibiting NCOA4-mediated ferritinophagy. Inhibition of ferritinophagy by knocking down Ncoa4 alleviated cisplatin-induced ototoxicity. Importantly, nuciferine treatment mitigated cochlear hair cell loss and damage to ribbon synapse, and improved mouse hearing function in an acute cisplatin-induced hearing loss model. Our findings highlight the role of NCOA4-mediated ferritinophagy in the pathogenesis of cisplatin-induced ototoxicity and provide evidence for nuciferine as a promising protective agent for treating cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Xian Gao
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
| | - Huanyu Mao
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
| | - Liping Zhao
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
| | - Xiang Li
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
| | - Yaqi Liao
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- Department of Otorhinolaryngology Head and Neck Surgery, The Third People’s Hospital of Hubei Province, Wuhan 430030, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
- The Institutes of Brain Science, The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
- The Institutes of Brain Science, The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology, Department of Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Shanghai 200031, China
| |
Collapse
|
48
|
Jiang J, Zhou X, Chen H, Wang X, Ruan Y, Liu X, Ma J. 18β-Glycyrrhetinic acid protects against deoxynivalenol-induced liver injury via modulating ferritinophagy and mitochondrial quality control. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134319. [PMID: 38657511 DOI: 10.1016/j.jhazmat.2024.134319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024]
Abstract
Deoxynivalenol (DON), a widespread mycotoxin, represents a substantial public health hazard due to its propensity to contaminate agricultural produce, leading to both acute and chronic health issues in humans and animals upon consumption. The role of ferroptosis in DON-induced hepatic damage remains largely unexplored. This study investigates the impact of 18β-glycyrrhetinic acid (GA), a prominent constituent of glycyrrhiza, on DON hepatotoxicity and elucidates the underlying mechanisms. Our results indicate that GA effectively attenuates liver injury inflicted by DON. This was achieved by inhibiting nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and ferroptosis, as well as by adjusting mitochondrial quality control (MQC). Specifically, GA curtails ferritinophagy by diminishing NCOA4 expression without affecting the autophagic flux. At a molecular level, GA binds to and stabilizes programmed cell death protein 4 (PDCD4), thereby inhibiting its ubiquitination and subsequent degradation. This stabilization of PDCD4 leads to the downregulation of NCOA4 via the JNK-Jun-NCOA4 axis. Knockdown of PDCD4 weakened GA's protective action against DON exposure. Furthermore, GA improved mitochondrial function and limited excessive mitophagy and mitochondrial division induced by DON. Disrupting GA's modulation of MQC nullified its anti-ferroptosis effects. Overall, GA offers protection against DON-induced ferroptosis by blocking ferritinophagy and managing MQC. ENVIRONMENTAL IMPLICATION: Food contamination from mycotoxins, is a problem for agricultural and food industries worldwide. Deoxynivalenol (DON), the most common mycotoxins in cereal commodities. A survey in 2023 showed that the positivity rate for DON contamination in food reached more than 70% globally. DON can damage the health of humans whether exposed to high doses for short periods of time or low doses for long periods of time. We have discovered 18β-Glycyrrhetinic acid (GA), a prominent constituent of glycyrrhiza. Liver damage caused by low-dose DON can be successfully treated with GA. This study will support the means of DON control, including antidotes.
Collapse
Affiliation(s)
- Junze Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xintong Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hao Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yongbao Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaohui Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, PR China.
| |
Collapse
|
49
|
Liu Z, Bian Q, Wang D. Exposure to 6-PPD quinone causes ferroptosis activation associated with induction of reproductive toxicity in Caenorhabditis elegans. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134356. [PMID: 38643579 DOI: 10.1016/j.jhazmat.2024.134356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/29/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
Exposure to N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ) caused toxicity on Caenorhabditis elegans, including reproductive toxicity. However, the underlying mechanisms for this induced reproductive toxicity by 6-PPDQ remain largely unclear. We examined possible association of ferroptosis activation with reproductive toxicity of 6-PPDQ. In 1-100 μg/L 6-PPDQ exposed nematodes, Fe2+ content was increased, which was accompanied with enhanced lipid peroxidation, increased malonydialdehyde (MDA) content, and decreased L-glutathione (GSH) content. Exposure to 1-100 μg/L 6-PPDQ decreased expressions of ftn-1 encoding ferritin, ads-1 encoding AGPS, and gpx-6 encoding GPX4 and increased expression of bli-3 encoding dual oxidase. After 6-PPDQ exposure, RNAi of ftn-1 decreased ads-1 and gpx-6 expressions and increased bli-3 expression. RNAi of ftn-1, ads-1, and gpx-6 strengthened alterations in ferroptosis related indicators, and RNAi of bli-3 suppressed changes of ferroptosis related indicators in 6-PPDQ exposed nematodes. Meanwhile, RNAi of ftn-1, ads-1, and gpx-6 induced susceptibility, and RNAi of bli-3 caused resistance to 6-PPDQ reproductive toxicity. Moreover, expressions of DNA damage checkpoint genes (clk-2, mrt-2, and hus-1) could be increased by RNAi of ftn-1, ads-1, and gpx-6 in 6-PPDQ exposed nematodes. Therefore, our results demonstrated activation of ferroptosis in nematodes exposed to 6-PPDQ at environmentally relevant concentrations, and this ferroptosis activation was related to reproductive toxicity of 6-PPDQ.
Collapse
Affiliation(s)
- Zhengying Liu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China
| | - Qian Bian
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Dayong Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China; Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, Shenzhen, China.
| |
Collapse
|
50
|
Wang Y, Hao Y, Yuan L, Tian H, Sun X, Zhang Y. Ferroptosis: a new mechanism of traditional Chinese medicine for treating ulcerative colitis. Front Pharmacol 2024; 15:1379058. [PMID: 38895617 PMCID: PMC11184165 DOI: 10.3389/fphar.2024.1379058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Ulcerative colitis (UC), a subtype of inflammatory bowel disease, manifests with symptoms such as abdominal pain, diarrhea, and mucopurulent, bloody stools. The pathogenesis of UC is not fully understood. At present, the incidence of UC has increased significantly around the world. Conventional therapeutic arsenals are relatively limited, with often poor efficacy and many adverse effects. In contrast, traditional Chinese medicine (TCM) holds promise due to their notable effectiveness, reduced recurrence rates, and minimal side effects. In recent years, significant progress has been made in the basic research on TCM for UC treatment. It has been found that the inhibition of ferroptosis through the intervention of TCM can significantly promote intestinal mucosal healing and reverse UC. The mechanism of action involves multiple targets and pathways. Aim of the review This review summarizes the experimental studies on the targeted regulation of ferroptosis by TCM and its impact on UC in recent years, aiming to provide theoretical basis for the prevention, treatment, and further drug development for UC. Results Ferroptosis disrupts antioxidant mechanisms in intestinal epithelial cells, damages the intestinal mucosa, and participates in the pathological process of UC. TCM acts on various pathways such as Nrf2/HO-1 and GSH/GPX4, blocking the pathological progression of ferroptosis in intestinal epithelial cells, inhibiting pathological damage to the intestinal mucosa, and thereby alleviating UC. Conclusion The diverse array of TCM single herbs, extracts and herbal formulas facilitates selective and innovative research and development of new TCM methods for targeting UC treatment. Although progress has been made in studying TCM compound formulas, single herbs, and extracts, there are still many issues in clinical and basic experimental designs, necessitating further in-depth scientific exploration and research.
Collapse
Affiliation(s)
- Yingyi Wang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanwei Hao
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lingling Yuan
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huaie Tian
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuhui Sun
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|