1
|
Liu H, Xu Y, Liu Y, Han X, Zhao L, Liu Y, Zhang F, Fu Y. Identification of novel ferroptosis-related biomarkers associated with the oxidative stress pathways in ischemic cardiomyopathy. IJC HEART & VASCULATURE 2025; 56:101584. [PMID: 39807364 PMCID: PMC11726793 DOI: 10.1016/j.ijcha.2024.101584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Background Ferroptosis is a cell death process that depends on iron and reactive oxygen species. It significantly contributes to cardiovascular diseases. However, its exact role in ischemic cardiomyopathy (ICM) is still unclear. Methods Using bioinformatics methods, we identified new molecular targets associated with ferroptosis in ICM and conducted various analyses-including correlation analysis, pathway enrichment analysis, protein interaction network construction, and analysis of transcription factor and drug interactions, to reveal the potential mechanisms behind these genes. Results We evaluated two independent training sets of ICM, GSE57338 and GSE5406, comprising 203 ICM samples, and validation sets GSE76701 to examine differentially expressed genes (DEGs) related to ferroptosis. After extracting the intersection of the gene sets and ferroptosis-related genes, 53 DEGs were identified. Enrichment analyses showed that the alterations in ferroptosis-related DEGs were mainly enriched in oxidative stress response, and immune-related pathways. Furthermore, 11 hub genes were identified using protein-protein interaction network analysis. The key interactions between 11 hub genes were more pronounced in protein localization during ICM development. In addition, we construct a hub gene and transcription factor interaction network and a small molecule drug-gene interaction network. We found that among these hub genes, the N-acetylneuraminate outer membrane channel(NANC) gene is positively correlated with most of the small-molecule drugs used to treat ICM, and its high expression might increase resistance. Conclusions Ferroptosis exists in ICM and and is associated with oxidative stress. This association suggests that ferroptosis may facilitate the progression of ICM.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - Yuan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yuanmei Liu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - XueJun Han
- Department of Orthopaedics, Jiayuguan Municipal First People’s Hospital, Jiayuguan 735100, PR China
| | - Liping Zhao
- Department of Ophthalmology, Jiayuguan Municipal First People’s Hospital, Jiayuguan 735100, PR China
| | - Yixuan Liu
- College of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Fuchun Zhang
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - Yicheng Fu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| |
Collapse
|
2
|
Wu Y, Yang L, You J, Tian C, Yang S, Li L. Discovery of phenazine derivatives as a new class of non-classical ferroptosis inhibitors and efficacy evaluation on a mouse model of liver injury. Eur J Med Chem 2025; 282:117042. [PMID: 39556893 DOI: 10.1016/j.ejmech.2024.117042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is an iron-dependent regulated cell death, which has been implicated in the onset and progression of numerous diseases. Ferroptosis inhibitors are thought as potential agents for treating these related diseases. However, the majority of currently available ferroptosis inhibitors are antioxidants or iron chelators (called classical ferroptosis inhibitors), which might have potential risks of side effects during clinical use. Herein, we report the discovery of phenazine derivatives as a new class of non-classical ferroptosis inhibitors. Structure-activity relationship of these series compounds led to the discovery of the most active compound 13l with an EC50 value of 0.0007 μM. Mechanistically, 13l could inhibit NCOA4-mediated ferritinophagy, hence protecting cells from ferroptosis. Notably, in the acetaminophen-induced acute liver injury model, 13l showed an excellent therapeutic effect. Overall, this compound reported here could be a promising lead compound for drug discovery targeting ferroptosis.
Collapse
Affiliation(s)
- Yunjie Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, 610041, China
| | - Lu Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, 610041, China
| | - Jing You
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chenyu Tian
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shengyong Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, 610041, China.
| |
Collapse
|
3
|
Nandha SR, Checker R, Patwardhan RS, Sharma D, Sandur SK. Anti-oxidants as therapeutic agents for oxidative stress associated pathologies: future challenges and opportunities. Free Radic Res 2025:1-25. [PMID: 39764687 DOI: 10.1080/10715762.2025.2450504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Free radicals have been implicated in the pathogenesis of cancer along with cardiovascular, neurodegenerative, pulmonary and inflammatory disorders. Further, the relationship between oxidative stress and disease is distinctively established. Clinical trials using anti-oxidants for the prevention of disease progression have indicated some beneficial effects. However, these trials failed to establish anti-oxidants as therapeutic agents due to lack of efficacy. This is attributed to the fact that living systems are under dynamic redox control wherein their redox behavior is compartmentalized and simple aggregation of redox couples, distributed throughout the system, is of miniscule importance while determining their overall redox state. Further, free radical metabolism is intriguingly complex as they play plural roles segregated in a spatio-temporal manner. Depending on quality, quantity and site of generation, free radicals exhibit beneficial or harmful effects. Use of nonspecific, non-targeted, general ROS scavengers lead to systemic elimination of all types of ROS and interferes in cellular signaling. Failure of anti-oxidants to act as therapeutic agents lies in this oversimplification of extremely dynamic cellular redox environment as a static and non-compartmentalized redox state. Rather than generalizing the term "oxidative stress" if we can identify the "type of oxidative stress" in different types of diseases, a targeted and more specific anti-oxidant therapy may be developed. In this review, we discuss the concept of redox dynamics, role and type of oxidative stress in disease conditions, and current status of anti-oxidants as therapeutic agents. Further, we probe the possibility of developing novel, targeted and efficacious anti-oxidants with drug-like properties.
Collapse
Affiliation(s)
- Shivani R Nandha
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
4
|
Pan M, Zhang L, Chang S, Jiang X, Shen J, Feng X, Xu F, Zha X, Chen X, Fan X. Poly-l-arginine promotes ferroptosis in asthmatic airway epithelial cells by modulating PBX1/GABARAPL1 axis. Int J Biol Macromol 2025; 286:138478. [PMID: 39645127 DOI: 10.1016/j.ijbiomac.2024.138478] [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: 09/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Eosinophils play a featured role among inflammatory cells participating in the onset and development of asthma. Activated eosinophils release several cytotoxic granular proteins, such as major basic protein (MBP), posing a significant threat to airway epithelium. Ferroptosis, a novel form of cell death, is gaining recognition for its involvement in asthma pathogenesis, though the specific mechanisms remain largely unknown. Herein, we revealed that poly-l-arginine (PLA), an MBP mimic, induced ferroptosis in airway epithelium by downregulating γ-aminobutyric acid receptor-associated protein-like 1 (GABARAPL1). Reduced GABARAPL1 expression was further confirmed in ovalbumin (OVA)-induced asthma mice and PLA-treated human airway organoids (hAOs). Mechanistically, PLA activated mechanistic target of rapamycin complex 1 (mTORC1) signaling, inhibiting pre-B-cell leukemia transcription factor 1 (PBX1), which in turn leads to transcriptional downregulation of GABARAPL1. Furthermore, MBP extracted from eosinophils, similar to PLA, induced ferroptosis in airway epithelial cells, as well as modulating mTORC1/PBX1/GABARAPL1 pathway. Finally, Ferrostatin-1 treatment or GABARAPL1 overexpression alleviated ferroptosis and airway inflammation in asthmatic mice. Overall, our findings highlight the cell communication between eosinophils and airway epithelial cells. MBP modulates the mTORC1/PBX1/GABARAPL1 axis, thereby serving as a significant contributor to ferroptosis in airway epithelium and airway inflammation. This suggests that suppressing ferroptosis in airway epithelium or targeting eosinophils and MBP could lead to novel therapeutic strategies for asthma management.
Collapse
Affiliation(s)
- Min Pan
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Ling Zhang
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Shuang Chang
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Xueqin Jiang
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, China
| | - Jiapan Shen
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Xiaoxia Feng
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Fangzhou Xu
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Xu Chen
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China.
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, China.
| |
Collapse
|
5
|
Ma C, Xu C, Zheng M, Zhang S, Liu Q, Lyu J, Pang X, Wang Y. Utilizing Lactic Acid Bacteria to Improve Hyperlipidemia: A Comprehensive Analysis from Gut Microbiota to Metabolic Pathways. Foods 2024; 13:4058. [PMID: 39767000 PMCID: PMC11675396 DOI: 10.3390/foods13244058] [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: 10/19/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Hyperlipidemia poses significant risks for cardiovascular diseases, with emerging evidence underscoring the critical role of gut microbiota in metabolic regulation. This study explores Lactobacillus casei CAAS36, a probiotic strain with promising cholesterol-lowering capabilities, assessing its impact on hyperlipidemic hamsters. Utilizing 1H NMR-based metabolomics and 16S rRNA gene sequencing, we observed that L. casei CAAS36 treatment not only altered metabolic pathways but also reshaped gut microbiota composition. Notably, the treatment restored the balance between Firmicutes and Bacteroidetes and significantly increased the abundance of propionate-producing Muribaculaceae. Metabolically, L. casei CAAS36 administration led to the normalization of key lipid markers, including reductions in total cholesterol, LDL-C, and triglycerides (29.9%, 29.4% and 32.6%), while enhancing the protective HDL-C levels. These effects were accompanied by significant increases in beneficial metabolites such as propionate and succinate, which are known for their roles in preventing metabolic disorders. These findings highlight the dual regulatory effects of L. casei CAAS36 on the metabolic profile and gut microbiota, suggesting a substantial potential for this probiotic in the management of hyperlipidemia and possibly other metabolic diseases. Future applications may include its use as a natural therapeutic agent in clinical settings, aiming to reduce reliance on conventional pharmaceuticals and their associated side effects.
Collapse
Affiliation(s)
- Changlu Ma
- Department of Food and Bio-Engineering, Beijing Vocational College of Agriculture, Beijing 102442, China;
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Chen Xu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Mumin Zheng
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Shuwen Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Qifeng Liu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China;
| | - Jiaping Lyu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Xiaoyang Pang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Yinghong Wang
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China;
| |
Collapse
|
6
|
Yan Z, Zhao W, Zhao N, Liu Y, Yang B, Wang L, Liu J, Wang D, Wang J, Jiao X, Cao J, Li J. PRMT1 alleviates isoprenaline-induced myocardial hypertrophy by methylating SRSF1. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39659162 DOI: 10.3724/abbs.2024175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Myocardial hypertrophy (MH) is an important factor contributing to severe cardiovascular disease. Previous studies have demonstrated that specific deletion of the protein arginine methyltransferase 1 (PRMT1) leads to MH, but the exact mechanism remains unclear. Serine/arginine-rich splicing factor 1 (SRSF1) affects the development and progression of cardiovascular disease by selectively splicing downstream signaling proteins. The present study is designed to determine whether PRMT1 is involved in MH by regulating SRSF1 and, if so, to explore the underlying mechanisms. Adult male mice and H9C2 cardiomyocytes are treated with isoprenaline (ISO) to establish MH models. The expression levels of PRMT1 are significantly decreased in the ISO-induced MH models, and inhibiting PRMT1 worsens MH, whereas overexpression of PRMT1 ameliorates MH. SRSF1 serves as the downstream target of PRMT1, and its expression is markedly elevated in MH. Moreover, SRSF1 increases the mRNA expressions of CaMKIIδ A and CaMKIIδ B, decreases the mRNA expression of CaMKIIδ C by altering the selective splicing of CaMKIIδ, and further participates in MH. In addition, there is an interaction between PRMT1 and SRSF1, whereby PRMT1 reduces the phosphorylation level of SRSF1 via methylation, thus further altering its functional activity and eventually improving MH. Our present study demonstrates that PRMT1 relieves MH by methylating SRSF1, which is expected to provide a new theoretical basis for the pathogenic mechanism of MH and potential drug targets for reducing MH and associated cardiovascular disease.
Collapse
Affiliation(s)
- Zi Yan
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- MOE Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Wenhui Zhao
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Naixin Zhao
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Yufeng Liu
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Bowen Yang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan 030001, China
| | - Jingyi Liu
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan 030001, China
| | - Deping Wang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- MOE Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Jin Wang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- MOE Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Xiangying Jiao
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- MOE Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Jimin Cao
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- MOE Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Jianguo Li
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- MOE Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, China
- Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou 510515, China
| |
Collapse
|
7
|
Zhu XM, Xu Y, Zhang J. Cardiometabolic Index is associated with heart failure: a cross-sectional study based on NHANES. Front Med (Lausanne) 2024; 11:1507100. [PMID: 39717172 PMCID: PMC11663657 DOI: 10.3389/fmed.2024.1507100] [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: 10/07/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction Heart failure is a complex syndrome characterized by impaired cardiac function. Despite improvements in treatment, the prevalence of heart failure continues to rise. The Cardiometabolic Index (CMI), a novel measure combining abdominal obesity and lipid levels, has emerged as a potential predictor of cardiac metabolic risk. Methods We analyzed data from the National Health and Nutrition Examination Survey (NHANES) involving 22,586 participants to investigate the association between CMI and heart failure. Multivariable logistic regression models and RCS analysis were used to explore the association between heart failure and CMI after adjusting for potential confounders. Subgroup analyses were performed among populations with different demographic and clinical characteristics. Results Our results revealed a significant positive correlation between CMI and heart failure, with odds ratios of 2.77 and 1.87 for the highest quartile after adjusting for confounders. Subgroup analyses indicated heightened risks among older adults and those with hypertension or diabetes. ROC curve analysis demonstrated that CMI offers good diagnostic value for heart failure, surpassing traditional measures like BMI. Discussion Our findings suggest that CMI is a valuable tool for assessing the risk of heart failure, particularly in individuals with increased abdominal obesity or abnormal lipid profiles. This highlights the importance of addressing cardiac metabolic health in both prevention and treatment strategies for heart failure. Future research should focus on exploring causal relationships and refining predictive models that incorporate CMI to enhance early detection and intervention.
Collapse
Affiliation(s)
| | | | - Jie Zhang
- Department of Cardiology, Xishan People's Hospital of Wuxi City, Wuxi, China
| |
Collapse
|
8
|
Ge Q, Zhang T, Yu J, Lu X, Xiao S, Zhang T, Qing T, Xiao Z, Zeng L, Luo L. A new perspective on targeting pulmonary arterial hypertension: Programmed cell death pathways (Autophagy, Pyroptosis, Ferroptosis). Biomed Pharmacother 2024; 181:117706. [PMID: 39581144 DOI: 10.1016/j.biopha.2024.117706] [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: 05/05/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiovascular disease characterized by elevated pulmonary vascular resistance, progressive increases in pulmonary artery pressures, ultimately leading to right-sided heart failure, and potentially mortality. Pulmonary vascular remodeling is pivotal in PAH onset and progression. While targeted drug therapies have notably ameliorated PAH prognosis, current medications primarily focus on vascular vasodilation, with limited ability to reverse pulmonary vascular remodeling fundamentally, resulting in suboptimal patient prognoses. Cellular death in pulmonary vasculature, once thought to be confined to apoptosis and necrosis, has evolved with the identification of pyroptosis, autophagy, and ferroptosis, revealing their association with vascular injury in PAH. These novel forms of regulated cellular death impact reactive oxygen species (ROS) generation, calcium stress, and inflammatory cascades, leading to pulmonary vascular cell loss, exacerbating vascular injury, and mediating adverse remodeling, inflammation, immune anomalies, and current emerging mechanisms (such as endothelial-mesenchymal transition, abnormal energy metabolism, and epigenetic regulation) in the pathogenesis of PAH. This review comprehensively delineates the roles of autophagy, pyroptosis, and ferroptosis in PAH, elucidating recent advances in their involvement and regulation of vascular injury. It juxtaposes their distinct functions in PAH and discusses the interplay of these programmed cell deaths in pulmonary vascular injury, highlighting the benefits of combined targeted therapies in mitigating pulmonary arterial hypertension-induced vascular injury, providing novel insights into targeted treatments for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Jiangbiao Yu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Xuelin Lu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Sijie Xiao
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Ting Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tao Qing
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China.
| |
Collapse
|
9
|
Huang M, Liu Y, Zhang L, Wang S, Wang X, He Z. Advancements in Research on Mesenchymal Stem-Cell-Derived Exosomal miRNAs: A Pivotal Insight into Aging and Age-Related Diseases. Biomolecules 2024; 14:1354. [PMID: 39595531 PMCID: PMC11592330 DOI: 10.3390/biom14111354] [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: 09/12/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into various cell types and play a crucial role in repairing aging tissues and diseased organs. Aging manifests as a gradual loss of cellular, tissue, and organ function, leading to the progression of pathologies. Exosomes (Exos) are extracellular vesicles secreted by cells, which maintain cellular homeostasis, clear cellular debris, and facilitate communication between cells and organs. This review provides a comprehensive summary of the mechanisms for the synthesis and sorting of MSC-Exo miRNAs and summarizes the current research status of MSCs-Exos in mitigating aging and age-related diseases. It delves into the underlying molecular mechanisms, which encompass antioxidative stress, anti-inflammatory response, and the promotion of angiogenesis. Additionally, this review also discusses potential challenges in and future strategies for advancing MSC-Exo miRNA-based therapies in the treatment of aging and age-related diseases.
Collapse
Affiliation(s)
- Minglei Huang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Ye Liu
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Longze Zhang
- Scientific Research Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi 563000, China;
| | - Shuangmin Wang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
10
|
Zhou D, Yang Y, Han R, He J, Liu D, Xia W, Cai Y, Perek B, Xia Z. Ferroptosis and its Potential Determinant Role in Myocardial Susceptibility to Ischemia/Reperfusion Injury in Diabetes. Rev Cardiovasc Med 2024; 25:360. [PMID: 39484139 PMCID: PMC11522832 DOI: 10.31083/j.rcm2510360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 11/03/2024] Open
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is a major cause of cardiac death particularly in patients with diabetes. When the coronary artery is partially or completely blocked, restoration of blood perfusion can normally be achieved within a certain time due to the development of advanced techniques such as percutaneous coronary intervention (PCI) and coronary artery bypass grafting (CABG) surgery. However, cardiac tissue injury may aggravate progressively even after the ischemic myocardium is restored to normal perfusion. MIRI is often associated with various forms of cell death, including apoptosis, autophagy, programmed necrosis, pyroptosis, and ferroptosis, among others. Ferroptosis is known as iron-dependent cell death that is distinct from other programmed modes of cell death. Ferroptosis is under constitutive control by glutathione peroxidase 4 (GPX4), and the reduction of GPX4 may result in ferroptosis even if iron homeostasis is physiologically maintained. The essences of ferroptosis are substantial iron accumulation and lipid peroxidation that trigger cell death. Under impaired antioxidant system, cellular reactive oxygen species (ROS) accumulation leads to lipid peroxidation which consequently results in ferroptosis. Ferroptosis shares a few common features with several types of cell death and interplays with various forms of cell death such as autophagy and apoptosis in the development of cardiovascular diseases. More and more recent studies have demonstrated that ferroptosis plays an important role in MIRI. However, few studies have addressed the relative importance of ferroptosis in MIRI relative to other forms of cell deaths. In this review, we summarized the basic aspects and advances regarding the molecular pathogenesis of ferroptosis, evaluated its role in MIRI, and propose that the levels of ferroptosis may function as a major determinant of myocardial susceptibility to ischemia/reperfusion injury (IRI) in general and of the enhanced vulnerability to MIRI specifically in diabetes.
Collapse
Affiliation(s)
- Dongcheng Zhou
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
| | - Yuhui Yang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
| | - Ronghui Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
| | - Jianfeng He
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
| | - Danyong Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
| | - Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Bartłomiej Perek
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China
- Doctoral Training Platform for Research and Translation, 431913 Zhongxiang, Hubei, China
| |
Collapse
|
11
|
Tang Y, Liang H, Su L, Xue X, Zhan J. Ferroptosis: a new perspective on the pathogenesis of radiation-induced cataracts. Front Public Health 2024; 12:1449216. [PMID: 39220446 PMCID: PMC11363423 DOI: 10.3389/fpubh.2024.1449216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Ionizing radiation is a significant risk factor for cataracts, but the pathogenesis of radiation-induced cataracts remains incompletely understood. Ferroptosis, an iron-dependent form of programmed cell death discovered in recent years, has gained increasing attention for its role in various diseases. This article systematically reviews research progress on ionizing radiation, ferroptosis, age-related cataracts, and radiation-induced cataracts. It proposes the "ferroptosis hypothesis" for the pathogenesis of radiation-induced cataracts. Through ionization and oxidative stress effects, ionizing radiation leads to elevated free iron levels and exacerbated lipid peroxidation in lens cells, activating the ferroptosis pathway and resulting in lens opacity. The involvement of ferroptosis in the development of age-related cataracts suggests that it may also be an important pathogenic mechanism of radiation-induced cataracts. Targeting the ferroptosis pathway may be a novel strategy for preventing and treating radiation-induced cataracts. Furthermore, developing new ferroptosis-specific inhibitors with improved targeting and pharmacokinetic properties is also an essential direction for research on preventing and treating radiation-induced cataracts. The study of ferroptosis provides new insights into the mechanism and management of radiation-induced cataracts, potentially transforming radiation-induced cataracts from "inevitable" to "preventable and treatable."
Collapse
Affiliation(s)
| | | | | | - Xiangming Xue
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan, China
| | - Jingming Zhan
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan, China
| |
Collapse
|
12
|
Lv Q, Lin J, Huang H, Ma B, Li W, Chen J, Wang M, Wang X, Fu G, Xiao Y. Nanosponge for Iron Chelation and Efflux: A Ferroptosis-Inhibiting Approach for Myocardial Infarction Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305895. [PMID: 38671590 PMCID: PMC11220697 DOI: 10.1002/advs.202305895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 04/11/2024] [Indexed: 04/28/2024]
Abstract
Myocardial infarction (MI), a consequence of coronary artery occlusion, triggers the degradation of ferritin, resulting in elevated levels of free iron in the heart and thereby inducing ferroptosis. Targeting myocardial ferroptosis through the chelation of excess iron has therapeutic potential for MI treatment. However, iron chelation in post ischemic injury areas using conventional iron-specific chelators is hindered by ineffective myocardial intracellular chelation, rapid clearance, and high systemic toxicity. A chitosan-desferrioxamine nanosponge (CDNS) is designed by co-crosslinking chitosan and deferoxamine through noncovalent gelation to address these challenges. This architecture facilitates direct iron chelation regardless of deferoxamine (DFO) release due to its sponge-like porous hydrogel structure. Upon cellular internalization, CDNS can effectively chelate cellular iron and facilitate the efflux of captured iron, thereby inhibiting ferroptosis and associated oxidative stress and lipid peroxidation. In MI mouse models, myocardial injection of CDNS promotes sustainable retention and the suppression of ferroptosis in the infarcted heart. This intervention improves cardiac function and alleviates adverse cardiac remodeling post-MI, leading to decreased oxidative stress and the promotion of angiogenesis due to ferroptosis inhibition by CDNS in the infarcted heart. This study reveals a nanosponge-based nanomedicine targeting myocardial ferroptosis with efficient iron chelation and efflux, offering a promising MI treatment.
Collapse
Affiliation(s)
- Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jun Lin
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
- Department of Cardiovascular SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Boxuan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Wujiao Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jiawen Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced StudiesZhejiang UniversityHangzhou310058China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yun Xiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| |
Collapse
|
13
|
Wang X, Lin Z, Tang X, Xie M, Li T, Zhou J. Matrine induces cardiotoxicity by promoting ferroptosis through the Nrf2 antioxidant system in H9c2 cells. Toxicol Lett 2024; 397:11-22. [PMID: 38723914 DOI: 10.1016/j.toxlet.2024.05.001] [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: 02/03/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Matrine (MT) has shown promising efficacy in various cancers and chronic hepatitis; however, its clinical application is limited because of its side effects. Our previous studies have indicated that MT can induce severe hepatotoxicity and nephrotoxicity. The current study aimed to investigate its cardiotoxicity and potential underlying mechanisms in H9c2 cells. Our results showed that MT induced H9c2 cell death and disrupted the cellular membrane integrity. Moreover, MT decreased glutathione (GSH) and cysteine (Cys) levels, and increased Fe2+, lipid peroxidation, reactive oxygen species (ROS), and MDA levels, ultimately leading to ferroptosis. Interestingly, these phenomena were alleviated by the ferroptosis inhibitor Fer-1, whereas MT-induced ferroptosis was exacerbated by the ferroptosis agonist RSL3. In addition, MT significantly reduced FTH, Nrf2, xCT, GPX4, and FSP1 protein levels and inhibited the transcriptional activity of Nrf2 while increasing TFR1 protein levels. Supplementation with Nrf2 agonist (Dimethyl fumarate, DMF) or selenium (Sodium selenite, SS) and CoQ10 alleviated MT-induced cytotoxic effects in H9c2 cells. These results suggest that ferroptosis, which is mediated by an imbalance in the Nrf2 antioxidant system, is involved in MT-induced cardiac toxicity.
Collapse
Affiliation(s)
- Xi Wang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Zixiong Lin
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Xinyi Tang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Ting Li
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China.
| |
Collapse
|
14
|
Linjacki S, Wang Y, Baath N, Mantle D, Yang G. H 2S Protects from Rotenone-Induced Ferroptosis by Stabilizing Fe-S Clusters in Rat Cardiac Cells. Cells 2024; 13:371. [PMID: 38474335 PMCID: PMC10931451 DOI: 10.3390/cells13050371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Hydrogen sulfide (H2S) has been recently recognized as an important gasotransmitter with cardioprotections, and iron is vital for various cellular activities. This study explored the regulatory role of H2S on iron metabolism and mitochondrial functions in cultured rat cardiac cells. Rotenone, a mitochondrial complex I inhibitor, was used for establishing an in vitro model of ischemic cell damage. It was first found that rotenone induced oxidative stress and lipid peroxidation and decreased mitochondrial membrane potential and ATP generation, eventually causing cell death. The supplement of H2S at a physiologically relevant concentration protected from rotenone-induced ferroptotic cell death by reducing oxidative stress and mitochondrial damage, maintaining GPx4 expression and intracellular iron level. Deferiprone, an iron chelator, would also protect from rotenone-induced ferroptosis. Further studies demonstrated that H2S inhibited ABCB8-mediated iron efflux from mitochondria to cytosol and promoted NFS1-mediated Fe-S cluster biogenesis. It is also found that rotenone stimulated iron-dependent H2S generation. These results indicate that H2S would protect cardiac cells from ischemic damage through preserving mitochondrial functions and intracellular Fe-S cluster homeostasis.
Collapse
Affiliation(s)
- Sara Linjacki
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Yuehong Wang
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Navjeet Baath
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Devin Mantle
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Guangdong Yang
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|