1
|
Zhang Y, Wei Y, Han X, Shi L, Yu H, Ji X, Gao Y, Gao Q, Zhang L, Duan Y, Li W, Yuan Y, Shi J, Cheng L, Li Y. Faecalibacterium prausnitzii prevents age-related heart failure by suppressing ferroptosis in cardiomyocytes through butyrate-mediated LCN2 regulation. Gut Microbes 2025; 17:2505119. [PMID: 40364435 PMCID: PMC12080280 DOI: 10.1080/19490976.2025.2505119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/20/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025] Open
Abstract
Aging is a primary driver of the escalating prevalence of heart failure (HF). Age-associated gut microbiota dysbiosis has been implicated in various age-related diseases, yet its role in age-related HF remains largely unexplored. In this study, we sought to explore the potential link between age-related gut microbiota alterations and HF in the elderly. We analyzed a publicly available single-cell sequencing dataset, which revealed markedly increased ferroptosis activity in cardiac myocytes of elderly individuals compared to their younger counterparts. Notably, treatment with the ferroptosis inhibitor, ferrostatin-1, mitigated cardiac ferroptosis and prevented cardiac dysfunction in aging rats. Furthermore, fecal microbiota transplantation from elderly HF patients significantly increased cardiac ferroptosis activity and induced cardiac dysfunction in healthy recipient rats. Integrated 16S rRNA sequencing and PCR quantification revealed a marked depletion of Faecalibacterium prausnitzii (F. prausnitzii) in elderly individuals, with a more pronounced decline in elderly patients with HF. Oral administration of F. prausnitzii or its metabolite butyrate effectively attenuated age-related HF through inhibiting ferroptosis. Additionally, gene-editing techniques were employed to generate F. prausnitzii BCoAT mutant deficient in butyrate production. Intriguingly, the protective effect was lost in the butyrate-deficient F. prausnitzii strain. Mechanistically, butyrate reduced intracellular iron accumulation and suppressed ferroptosis by downregulating LCN2 expression in senescent cardiomyocytes. Our findings highlight the critical role of aged microbiota-induced ferroptosis in HF and propose F. prausnitzii or butyrate may serve as potential targets for the prevention and treatment of age-related HF.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Wei
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xuejie Han
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ling Shi
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hui Yu
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuanrui Ji
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yunlong Gao
- Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin, China
| | - Qianhui Gao
- Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin, China
| | - Linwei Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yu Duan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenpeng Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yue Yuan
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jing Shi
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Wang J, Ma B, Jiang X, Li C, Lin Z, Wang Y, Shi J, Wang G, Cui C. H 2 protects H9c2 cells from hypoxia/reoxygenation injury by inhibiting the Wnt/CX3CR1 signaling pathway. Med Gas Res 2025; 15:339-347. [PMID: 39511756 PMCID: PMC11918467 DOI: 10.4103/mgr.medgasres-d-24-00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 11/15/2024] Open
Abstract
Myocardial ischemia‒reperfusion injury is a severe cardiovascular disease, and its treatment and prevention are crucial for improving patient prognosis and reducing the economic burden. This study aimed to explore the impact of hydrogen (H 2 ) on hypoxia/reoxygenation (H/R) injury in H9c2 cells (derived from rat embryonic heart tissue) induced by hydrogen peroxide (H 2 O 2 ) and to elucidate its underlying mechanism. An H/R injury model was established in H9c2 cells via exposure to 15 μM H 2 O 2 for 3 hours, followed by incubation in a 5% CO 2 atmosphere at 37°C for 24 hours. Then, the cells were treated with H 2 (50%) for 6, 12 or 24 hours. The results demonstrated that H9c2 cells exposed to H 2 O 2 and subjected to H/R injury presented a marked decrease in the cell survival rate, accompanied by severe morphological alterations, such as curling and wrinkling, and elevated lactate dehydrogenase levels. Notably, H 2 mitigated H/R injury induced by H 2 O 2 in a time-dependent manner, improving the morphological damage observed in H9c2 cells and decreasing lactate dehydrogenase levels. Compared with the model group, treatment with H 2 increased the activities of antioxidant enzymes, including catalase, superoxide dismutase, and glutathione peroxidase, while concurrently reducing the level of malondialdehyde, an indicator of cellular damage. Furthermore, H 2 treatment downregulated the expression of inflammatory cytokines and inflammatory-related factors, specifically interleukin-6, high-mobility group box 1, tumor necrosis factor-alpha, and Toll-like receptor 4, in H9c2 cells post-H/R injury. Furthermore, H 2 treatment resulted in a marked decrease in the expression levels of proteins associated with the Wnt/C-X3-C-motif receptor 1 signaling pathway, such as β-catenin, glycogen synthase kinase-3 beta, adenomatous polyposis coli, and Wnt and C-X3-C-motif receptor 1. This observation suggests a potential mechanism for its protective effects against H/R injury. Therefore, H 2 exerts a protective effect against H/R injury in H9c2 cells induced by H 2 O 2 , potentially by inhibiting the activated Wnt/C-X3-C-motif receptor 1 signaling pathway. This inhibition, in turn, prevents the generation of oxidative stress, inflammatory cytokines, and inflammation-associated factors.
Collapse
Affiliation(s)
- Jingsheng Wang
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Bin Ma
- Department of Cardiovascular Medicine, Taian City Taishan District People’s Hospital, Taian, Shandong Province, China
| | - Xue Jiang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Chao Li
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Yumei Wang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Jingfei Shi
- Shandong First Medical University, Jinan, Shandong Province, China
| | - Gang Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Chao Cui
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| |
Collapse
|
3
|
Liu H, Zhang C, Zhang L, Shen J, He K, Huang B. Selenomethionine suppresses lung tumor growth without hepatorenal toxicity in mice via the induction of apoptosis-ferroptosis and angiogenesis inhibition. Tissue Cell 2025; 96:102956. [PMID: 40382949 DOI: 10.1016/j.tice.2025.102956] [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: 09/01/2024] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
The high global mortality rate of lung cancer underscores the urgent need for novel therapeutic strategies. Selenium (Se), an essential trace element, exhibits tumor-suppressive properties across malignancies. This study systematically evaluated the antitumor efficacy of selenomethionine (SeMet) compared with that of selenocysteine (SeCys) in Lewis lung carcinoma (LLC) cells in vitro and in tumor-bearing mice in vivo. Compared with SeCys (IC50 = 45.89 μM), SeMet demonstrated superior cytotoxicity against LLC cells (IC50 = 30.19 μM) and significantly inhibited proliferation and migration by inducing apoptosis and ferroptosis. In vivo, SeMet treatment inhibited tumour growth by 50.87 % through the suppression of angiogenesis, outperforming SeCys (27.3 %). Transcriptomic analysis revealed the downregulation of proangiogenic chemokines (Cxcl1, Cxcl2, Cxcl3, Cxcl5) and the upregulation of antitumor chemokines (Cxcl9, Cxcl16) in SeMet-treated tumors. Additionally, SeMet enhanced the activity of antioxidant enzymes (T-SOD and GSH-PX) and reduced the levels of proinflammatory cytokines (IL-6 and TNF-α) without hepatorenal toxicity. These findings establish SeMet as a multifaceted therapeutic candidate for lung cancer through dual induction of apoptosis-ferroptosis and angiogenesis inhibition.
Collapse
Affiliation(s)
- Houru Liu
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China; Traditional Chinese Medicine Research Center, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Caiyun Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China; Traditional Chinese Medicine Research Center, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Lei Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China; Traditional Chinese Medicine Research Center, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Jie Shen
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China; Traditional Chinese Medicine Research Center, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Kan He
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China; Traditional Chinese Medicine Research Center, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China.
| | - Bei Huang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China; Traditional Chinese Medicine Research Center, School of Life Sciences, Anhui University, Hefei, Anhui 230601, China.
| |
Collapse
|
4
|
Shahid MM, Hohman G, Eldeeb M. Fine-Tuning Ferroptosis by Modulating GPX4 and Its Potential in Mitigating Neuronal Degeneration in Parkinson's Disease. Chembiochem 2025; 26:e202401052. [PMID: 40353519 DOI: 10.1002/cbic.202401052] [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: 12/20/2024] [Revised: 02/02/2025] [Indexed: 05/14/2025]
Abstract
The increasing prevalence of neurodegenerative diseases necessitates the development of novel approaches to study, diagnose, and treat these devastating disorders. Accordingly, there is a critical need to precisely address the gap in the biochemical and physiological mechanisms that underlie neurodegenerative diseases to promote advancements in therapeutic interventions. Parkinson's Disease (PD), the second most common neurodegenerative disorder after Alzheimer's, demands further research focused on unravelling the rather intricate molecular mechanisms that drive its progression upon different cell signaling cues. While alpha-synuclein aggregation and mitochondrial dysfunction are two cellular hallmarks of the molecular pathophysiology of PD, few drugs are currently in clinical trials for treatment of PD, which warrants further studies to identify new therapeutic molecular targets. Herein, we briefly highlight some of the reported roles of ferroptosis, a modality of cell death that is driven by iron-dependent phospholipid peroxidation, and its regulation by glutathione peroxidase 4 (GPX4). We discuss the interconnectedness between lipid peroxidation and GPX4 regulation in the context of molecular pathogenesis of PD. Future studies are imperative in investigating the physiological role of ferroptosis and the translational impact of ferroptosis-specific modulators in studying PD biology.
Collapse
Affiliation(s)
- Michael M Shahid
- Department of Chemistry, Illinois State University, Normal, IL, USA
| | - Grace Hohman
- Department of Chemistry, Illinois State University, Normal, IL, USA
| | - Mohamed Eldeeb
- Department of Chemistry, Illinois State University, Normal, IL, USA
| |
Collapse
|
5
|
Xin W, Yang H, Heng X, Xu T, Zhang K, Zhao Y, Liu Y, Han D, Wu Y, Zhang W, He M, Pu L, Shen Y, Qu X, Sun N, Ye C. Sauchinone preserves cardiac function in doxorubicin-induced cardiomyopathy by inhibiting the NLRP3 inflammasome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156624. [PMID: 40068294 DOI: 10.1016/j.phymed.2025.156624] [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: 10/19/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Doxorubicin (Dox)-induced cardiomyopathy (DIC) is characterized by severe myocardial damage that can progress to dilated cardiomyopathy and potentially lead to heart failure. No effective prevention or treatment strategies are available for DIC. Sauchinone, a diastereomeric lignan isolated from Saururus chinensis, is known for its notable anti-inflammatory effects. However, a paucity of research on sauchinone in relation to heart disease exists, particularly regarding its role in DIC, which remains unclear. PURPOSE This study aimed to assess the therapeutic potential of sauchinone in alleviating cardiac injury and elucidate its potential molecular mechanism in DIC. METHODS Male C57BL/6J mice were used to construct chronic and acute DIC models in vivo. The mice were administered sauchinone intragastrically concurrently with the first injection of Dox to evaluate the therapeutic effect of sauchinone on DIC. H9c2, a rat cardiomyocyte cell line, was treated with various concentrations of sauchinone in conjunction with Dox to assess the protective effects of sauchinone on cardiomyocyte injury in vitro. RESULTS Supplementation with exogenous sauchinone mitigated Dox-induced cardiac atrophy, cardiac fibrosis, and ventricular remodeling, while preserving cardiac function. Sauchinone reduced Dox-induced abnormal apoptosis both in vitro and in vivo. Additionally, sauchinone restored mitochondrial function and decreased reactive oxygen species levels, which may be attributed to its activation of nuclear factor erythroid 2-related factor 2 (NRF2) signaling, thereby attenuating Dox-induced oxidative damage. Furthermore, sauchinone significantly inhibited the activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome and reduced the cardiac infiltration of inflammatory factors, thereby alleviating oxidative stress and inhibiting the progression of DIC. The NLRP3 agonist nigericin abolished DIC progression, while the NLRP3 antagonist MCC950 further enhanced the beneficial effects of sauchinone on DIC progression both in vivo and in vitro. CONCLUSIONS The key novel finding of the present study is that the use of sauchinone, a diastereomeric lignan isolated from Saururus chinensis, effectively limits the progression of DIC. Specifically, sauchinone not only alleviates Dox-induced chronic cardiac injury but also significantly delays the progression of acute DIC. Mechanistically, inactivation of the NLRP3 inflammasome and NRF2-mediated antioxidant pathways have been identified as two critical signaling pathways regulated by sauchinone, which plays a vital role in blocking the progression of DIC. Sauchinone holds promise as a potential therapeutic approach for DIC or dilated cardiomyopathy.
Collapse
Affiliation(s)
- Wenxu Xin
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Hai Yang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Xinyu Heng
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Tao Xu
- Department of Geriatrics, Sir Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ke Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Yining Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Yankui Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Deshen Han
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Yueyue Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Wei Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Meiqi He
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Lin Pu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Yicong Shen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Xiuxia Qu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China.
| | - Ning Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China.
| | - Chao Ye
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; Department of Cardiology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
6
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
7
|
Tang P, Huang R, Zhong X, Chen X, Lei Y. A comprehensive review on selenium and blood pressure: Recent advances and research perspectives. J Trace Elem Med Biol 2025; 88:127607. [PMID: 39908739 DOI: 10.1016/j.jtemb.2025.127607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Globally, approximately 31.1 % of adults are affected by hypertension(HTN), and there is currently no effective treatment for this condition. Selenium (Se), an essential trace element in the human body, has been shown to play a role in various biological processes, including anti-inflammation, antioxidative stress, anti-ferroptosis, and regulation of immune response. Research suggests that Se may have potential hypotensive effects. OBJECTIVE This review aims to comprehensively investigate the relationship between Se and blood pressure(BP), elucidate the mechanisms through which Se influences BP, and explore its prospective applications in clinical practice. METHODS We conducted a systematic search on PubMed for a thorough review of articles concerning the relationship between Se and BP, as well as the mechanisms by which Se may lower BP. RESULTS AND CONCLUSIONS Although some findings indicate that Se might increase BP, its anti-inflammatory, anti-oxidant, anti-vascular remodeling, anti-atherosclerotic, anti-ferroptosis, and regulation of immune response effects suggest that maintaining an appropriate level of Se may contribute to BP reduction and possibly lower the risk of pregnancy-induced hypertension(PIH). While Se shows promise in the management of HTN, further exploration is necessary for its development. Future studies should clarify the mechanisms involved and identify relevant targets through clinical research, which may provide adjunctive therapies for HTN.
Collapse
Affiliation(s)
- Pusong Tang
- Cardiovascular Disease Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei Minzu University, Enshi, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, China
| | - Rui Huang
- Cardiovascular Disease Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, China
| | - Xing Zhong
- Cardiovascular Disease Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei Minzu University, Enshi, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, China
| | - Xin Chen
- Cardiovascular Disease Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, China
| | - Yuhua Lei
- Cardiovascular Disease Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, China.
| |
Collapse
|
8
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
9
|
Fan S, Wang K, Zhang T, Deng D, Shen J, Zhao B, Fu D, Chen X. Mechanisms and Therapeutic Potential of GPX4 in Pain Modulation. Pain Ther 2025; 14:21-45. [PMID: 39503961 PMCID: PMC11751247 DOI: 10.1007/s40122-024-00673-8] [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: 08/14/2024] [Accepted: 10/04/2024] [Indexed: 01/23/2025] Open
Abstract
Pain, a complex symptom encompassing both sensory and emotional dimensions, constitutes a significant global public health issue. Oxidative stress is a pivotal factor in the complex pathophysiology of pain, with glutathione peroxidase 4 (GPX4) recognized as a crucial antioxidant enzyme involved in both antioxidant defense mechanisms and ferroptosis pathways. This review systematically explores GPX4's functions across various pain models, including neuropathic, inflammatory, low back, and cancer-related pain. Specifically, the focus includes GPX4's physiological roles, antioxidant defense mechanisms, regulation of ferroptosis, involvement in signal transduction pathways, and metabolic regulation. By summarizing current research, we highlight the potential of GPX4-targeted therapies in pain management.
Collapse
Affiliation(s)
- Shiwen Fan
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China
| | - Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Jiwei Shen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Bowen Zhao
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
10
|
Guo X, Liang XJ, Liu JL, Li ZH, You Z, Zhao D, Song Y, Li L, Song XQ. Precise Carrier-Free Pt(IV)-Nanobombs for Apoptosis/Ferroptosis Synergistic Tumor Therapy: A New Effective Method to Obtain Good Chemotherapy and Low Toxicity. J Med Chem 2025; 68:387-404. [PMID: 39680635 DOI: 10.1021/acs.jmedchem.4c02034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The emerged apoptosis/ferroptosis synergistic platinum-based therapy has attracted a lot of attention but is far from clinic use due to high systemic toxicity. Herein, a series of novel precise carrier-free self-assembled platinum(IV) nanoparticles with lipid regulation effect named FSPNPs (5NPs-8NPs) were constructed via connecting fenofibrate acid (FA) to cisplatin or oxaliplatin-derived platinum(IV)-intermediates with disulfide bonds. FSPNPs can be stimulated by high-glutathione/ascorbic acid and acidity environment to produce an "explosion-like" cascade release process. Cell-activity showed precision of FSPNPs, which accumulated more in tumor cells and inhibited cell proliferation. Especially, 5NPs have higher cell selectivity than cisplatin. FSPNPs downregulated glutathione/glutathione peroxidase 4, increased reactive oxygen species/lipid peroxidation/malondialdehyde, induced DNA damage/S-phase arrest, and regulated p53/Bcl-2/Bax to trigger the apoptosis/ferroptosis hybrid pathway. The released FA and derivates were docked into the peroxisome proliferator-activated receptor α with activating cholesterol metabolism to destroy membrane integrity. FSPNPs also showed good biocompatibility and superior antitumor activity with no observable tissue damage.
Collapse
Affiliation(s)
- Xu Guo
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Xue-Jiao Liang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Jia-Le Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Zhi-Hui Li
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Zhihao You
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Dandan Zhao
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Yali Song
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Longfei Li
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| | - Xue-Qing Song
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmacy, Hebei University, Baoding 071002, Hebei, P. R. China
| |
Collapse
|
11
|
Wang B, Wang J, Liu C, Li C, Meng T, Chen J, Liu Q, He W, Liu Z, Zhou Y. Ferroptosis: Latest evidence and perspectives on plant-derived natural active compounds mitigating doxorubicin-induced cardiotoxicity. J Appl Toxicol 2025; 45:135-158. [PMID: 39030835 DOI: 10.1002/jat.4670] [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: 06/08/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy drug widely used in clinical settings, acting as a first-line treatment for various malignant tumors. However, its use is greatly limited by the cardiotoxicity it induces, including doxorubicin-induced cardiomyopathy (DIC). The mechanisms behind DIC are not fully understood, but its potential biological mechanisms are thought to include oxidative stress, inflammation, energy metabolism disorders, mitochondrial damage, autophagy, apoptosis, and ferroptosis. Recent studies have shown that cardiac injury induced by DOX is closely related to ferroptosis. Due to their high efficacy, availability, and low side effects, natural medicine treatments hold strong clinical potential. Currently, natural medicines have been shown to mitigate DOX-induced ferroptosis and ease DIC through various functions such as antioxidation, iron ion homeostasis correction, lipid metabolism regulation, and mitochondrial function improvement. Therefore, this review summarizes the mechanisms of ferroptosis in DIC and the regulation by natural plant products, with the expectation of providing a reference for future research and development of inhibitors targeting ferroptosis in DIC. This review explores the mechanisms of ferroptosis in doxorubicin-induced cardiomyopathy (DIC) and summarizes how natural plant products can alleviate DIC by inhibiting ferroptosis through reducing oxidative stress, correcting iron ion homeostasis, regulating lipid metabolism, and improving mitochondrial function.
Collapse
Affiliation(s)
- Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wang He
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
12
|
Li Y, Liu C, Fang B, Chen X, Wang K, Xin H, Wang K, Yang SM. Ferroptosis, a therapeutic target for cardiovascular diseases, neurodegenerative diseases and cancer. J Transl Med 2024; 22:1137. [PMID: 39710702 DOI: 10.1186/s12967-024-05881-6] [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: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The identification of ferroptosis represents a pivotal advancement in the field of cell death research, revealing an entirely novel mechanism of cellular demise and offering new insights into the initiation, progression, and therapeutic management of various diseases. Ferroptosis is predominantly induced by intracellular iron accumulation, lipid peroxidation, or impairments in the antioxidant defense system, culminating in membrane rupture and consequent cell death. Studies have associated ferroptosis with a wide range of diseases, and by enhancing our comprehension of its underlying mechanisms, we can formulate innovative therapeutic strategies, thereby providing renewed hope for patients.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cuiyun Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bo Fang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Su-Min Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
13
|
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
|
14
|
Jumabayi W, Reyimu A, Zheng R, Paerhati P, Rahman M, Zou X, Xu A. Ferroptosis: A new way to intervene in the game between Mycobacterium tuberculosis and macrophages. Microb Pathog 2024; 197:107014. [PMID: 39396689 DOI: 10.1016/j.micpath.2024.107014] [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: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the main pathogen responsible for the high mortality and morbidity of tuberculosis (TB) worldwide, primarily targets and invades macrophages. Infected macrophages activate a series of immune mechanisms to clear Mtb, however, Mtb evades host immune surveillance through subtle immune escape strategies to create a microenvironment conducive to its own proliferation, growth, and dissemination, while inducing immune cell death. The course of TB is strongly correlated with the form of cell death, including apoptosis, pyroptosis, and necrosis. Recent studies have revealed that ferroptosis, a novel type of programmed cell death characterized by iron-dependent lipid peroxidation, is closely linked to the regulatory mechanisms of TB. The central role of ferroptosis in the pathologic process of TB is increasingly becoming a focal point for exploring new therapeutic targets in this field. This paper will delve into the dynamic game between Mtb and host immune cells, especially the role of ferroptosis in the pathogenesis of TB. At the same time, this paper will analyze the regulatory pathways of ferroptosis and provide unique insights and innovative perspectives for TB therapeutic strategies based on the ferroptosis mechanism. This study not only expands the theoretical basis of TB treatment, but also points out the direction of future drug development, providing new possibilities for overcoming this global health problem.
Collapse
Affiliation(s)
- Wuerken Jumabayi
- The Third Clinical Medical College (Affiliated Cancer Hospital) of Xinjiang Medical University, Urumqi, China
| | | | | | | | | | | | - Aimin Xu
- The First People's Hospital of Kashi, Kashi, China.
| |
Collapse
|
15
|
Li H, Wang M, Huang Y. Anthracycline-induced cardiotoxicity: An overview from cellular structural perspective. Biomed Pharmacother 2024; 179:117312. [PMID: 39167843 DOI: 10.1016/j.biopha.2024.117312] [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: 05/13/2024] [Revised: 07/28/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Anthracyclines are broad-spectrum anticancer drugs, but their clinical use is limited due to their severe cardiotoxicity. Anthracycline-induced cardiotoxicity (AIC) remains a significant cause of heart disease-related mortality in many cancer survivors. The underlying mechanisms of AIC have been explored over the past few decades. Reactive oxygen species and drug-induced inhibition of topoisomerase II beta are well-studied mechanisms, with mitochondria being a prominently investigated organelle. Emerging mechanisms such as ferroptosis, Ca2+ overload, autophagy and inflammation mediators have been implicated in recent years. In this review, our goal is to summarize and update the roles of various mechanisms in AIC, focusing on different cellular levels and further explore promising therapeutic approaches targeting these organelles or pathways.
Collapse
Affiliation(s)
- Hansheng Li
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province 430060, China.
| | - Meilun Wang
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province 430060, China.
| | - Yan Huang
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province 430060, China.
| |
Collapse
|
16
|
Zhong S, Wang Z, Yang J, Jiang D, Wang K. Ferroptosis-related oxaliplatin resistance in multiple cancers: Potential roles and therapeutic Implications. Heliyon 2024; 10:e37613. [PMID: 39309838 PMCID: PMC11414570 DOI: 10.1016/j.heliyon.2024.e37613] [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: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Oxaliplatin (OXA)-based therapy is effective in the treatment of multiple cancers. However, primary or acquired OXA resistance remains an emerging challenge for its clinical application. Ferroptosis is an iron-dependent mode of cell death that has been demonstrated to play an essential role in the chemoresistance of many drugs, including OXA. In particular, dysregulation of SLC7A11-GPX4, one of the major antioxidant systems of ferroptosis, was found in the OXA resistance of colorectal cancer (CRC) and hepatocellular carcinoma (HCC). In addition, Nrf2, the upstream regulator of GPX4 and many other antioxidant factors, is also involved in the OXA resistance of CRC and HCC. Inhibition of SLC7A11-GPX4 or Nrf2 by genetic deletion of pharmaceutical inhibition could significantly reverse OXA resistance. Long noncoding RNA (lncRNA) also participates in chemoresistance and ferroptosis of cancer cells. Specifically, LINC01134 promotes the recruitment of Nrf2 to the promoter of GPX4, thereby exerting transcriptional regulation of GPX4, which eventually increases the OXA sensitivity of HCC through upregulation of ferroptosis. On the other hand, a novel lncRNA DACT3-AS1 sensitizes gastric cancer cells to OXA through miR-181a-5p/sirtuin 1(SIRT1)-mediated ferroptosis. Therapies based on ferroptosis or a combination of OXA and ferroptosis enhancers could provide new therapeutic insights to overcome OXA resistance. In the present review, we present the current understanding of ferroptosis-related OXA resistance, highlight ferroptosis pathogenesis in OXA chemoresistance, and summarize available therapies that target OXA resistance by enhancing ferroptosis.
Collapse
Affiliation(s)
- Sijia Zhong
- Department of Gastrointestinal Surgery, the First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Zihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, 110122, China
| | - Jiaxi Yang
- Department of Gastrointestinal Surgery, the First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Di Jiang
- China University of Petroleum (East China), 66 Changjiang West Road, Qingdao, 266580, China
| | - Kewei Wang
- Department of Gastrointestinal Surgery, the First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| |
Collapse
|
17
|
Li Y, Li H, Zhang K, Xu C, Wang J, Li Z, Zhou Y, Liu S, Zhao X, Li Z, Yang F, Hu W, Jing Y, Wu P, Zhang J, Shi C, Zhang R, Jiang W, Xing N, Wen W, Han D, Qin W. Genetically Engineered Membrane-Coated Nanoparticles for Enhanced Prostate-Specific Membrane Antigen Targeting and Ferroptosis Treatment of Castration-Resistant Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401095. [PMID: 38946578 PMCID: PMC11434221 DOI: 10.1002/advs.202401095] [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: 01/30/2024] [Revised: 05/27/2024] [Indexed: 07/02/2024]
Abstract
Conventional androgen deprivation therapy (ADT) targets the androgen receptor (AR) inhibiting prostate cancer (PCa) progression; however, it can eventually lead to recurrence as castration-resistant PCa (CRPC), which has high mortality rates and lacks effective treatment modalities. The study confirms the presence of high glutathione peroxidase 4 (GPX4) expression, a key regulator of ferroptosis (i.e., iron-dependent program cell death) in CRPC cells. Therefore, inducing ferroptosis in CRPC cells might be an effective therapeutic modality for CRPC. However, nonspecific uptake of ferroptosis inducers can result in undesirable cytotoxicity in major organs. Thus, to precisely induce ferroptosis in CRPC cells, a genetic engineering strategy is proposed to embed a prostate-specific membrane antigen (PSMA)-targeting antibody fragment (gy1) in the macrophage membrane, which is then coated onto mesoporous polydopamine (MPDA) nanoparticles to produce a biomimetic nanoplatform. The results indicate that the membrane-coated nanoparticles (MNPs) exhibit high specificity and affinity toward CRPC cells. On further encapsulation with the ferroptosis inducers RSL3 and iron ions, MPDA/Fe/RSL3@M-gy1 demonstrates superior synergistic effects in highly targeted ferroptosis therapy eliciting significant therapeutic efficacy against CRPC tumor growth and bone metastasis without increased cytotoxicity. In conclusion, a new therapeutic strategy is reported for the PSMA-specific, CRPC-targeting platform for ferroptosis induction with increased efficacy and safety.
Collapse
Affiliation(s)
- Yu Li
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral, Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Hongji Li
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jingwei Wang
- Department of Medicine Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, No.169 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Zeyu Li
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yike Zhou
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Xiaolong Zhao
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Zhengxuan Li
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Wei Hu
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yuming Jing
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Peng Wu
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jingliang Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, No.169 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Rui Zhang
- The State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, No.169 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Wenkai Jiang
- State Key Laboratory of Oral, Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Nianzeng Xing
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Department of Urology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Air Force Medical University, No.169 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Air Force Medical University, No.127 Western Changle Road, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
18
|
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
|
19
|
Lin H, Wang L, Jiang X, Wang J. Glutathione dynamics in subcellular compartments and implications for drug development. Curr Opin Chem Biol 2024; 81:102505. [PMID: 39053236 PMCID: PMC11722958 DOI: 10.1016/j.cbpa.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
Glutathione (GSH) is a pivotal tripeptide antioxidant essential for maintaining cellular redox homeostasis and regulating diverse cellular processes. Subcellular compartmentalization of GSH underscores its multifaceted roles across various organelles including the cytosol, mitochondria, endoplasmic reticulum, and nucleus, each exhibiting distinct regulatory mechanisms. Perturbations in GSH dynamics contribute to pathophysiological conditions, emphasizing the clinical significance of understanding its intricate regulation. This review consolidates current knowledge on subcellular GSH dynamics, highlighting its implications in drug development, particularly in covalent drug design and antitumor strategies targeting intracellular GSH levels. Challenges and future directions in deciphering subcellular GSH dynamics are discussed, advocating for innovative methodologies to advance our comprehension and facilitate the development of precise therapeutic interventions based on GSH modulation.
Collapse
Affiliation(s)
- Hanfeng Lin
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lingfei Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiqian Jiang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jin Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Decoding ferroptosis: Revealing the hidden assassin behind cardiovascular diseases. Biomed Pharmacother 2024; 176:116761. [PMID: 38788596 DOI: 10.1016/j.biopha.2024.116761] [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: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
The discovery of regulatory cell death processes has driven innovation in cardiovascular disease (CVD) therapeutic strategies. Over the past decade, ferroptosis, an iron-dependent form of regulated cell death driven by excessive lipid peroxidation, has been shown to drive the development of multiple CVDs. This review provides insights into the evolution of the concept of ferroptosis, the similarities and differences with traditional modes of programmed cell death (e.g., apoptosis, autophagy, and necrosis), as well as the core regulatory mechanisms of ferroptosis (including cystine/glutamate transporter blockade, imbalance of iron metabolism, and lipid peroxidation). In addition, it provides not only a detailed review of the role of ferroptosis and its therapeutic potential in widely studied CVDs such as coronary atherosclerotic heart disease, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, cardiomyopathy, and aortic aneurysm but also an overview of the phenomenon and therapeutic perspectives of ferroptosis in lesser-addressed CVDs such as cardiac valvulopathy, pulmonary hypertension, and sickle cell disease. This article aims to integrate this knowledge to provide a comprehensive view of ferroptosis in a wide range of CVDs and to drive innovation and progress in therapeutic strategies in this field.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
21
|
Zhang L, Luo YL, Xiang Y, Bai XY, Qiang RR, Zhang X, Yang YL, Liu XL. Ferroptosis inhibitors: past, present and future. Front Pharmacol 2024; 15:1407335. [PMID: 38846099 PMCID: PMC11153831 DOI: 10.3389/fphar.2024.1407335] [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] [Accepted: 05/06/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic mode of programmed cell death characterized by iron dependence and lipid peroxidation. Since the ferroptosis was proposed, researchers have revealed the mechanisms of its formation and continue to explore effective inhibitors of ferroptosis in disease. Recent studies have shown a correlation between ferroptosis and the pathological mechanisms of neurodegenerative diseases, as well as diseases involving tissue or organ damage. Acting on ferroptosis-related targets may provide new strategies for the treatment of ferroptosis-mediated diseases. This article specifically describes the metabolic pathways of ferroptosis and summarizes the reported mechanisms of action of natural and synthetic small molecule inhibitors of ferroptosis and their efficacy in disease. The paper also describes ferroptosis treatments such as gene therapy, cell therapy, and nanotechnology, and summarises the challenges encountered in the clinical translation of ferroptosis inhibitors. Finally, the relationship between ferroptosis and other modes of cell death is discussed, hopefully paving the way for future drug design and discovery.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yi Lin Luo
- School of Medicine, Yan’an University, Yan’an, China
| | - Yang Xiang
- College of Physical Education, Yan’an University, Yan’an, China
| | - Xin Yue Bai
- School of Medicine, Yan’an University, Yan’an, China
| | | | - Xin Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yan Ling Yang
- School of Medicine, Yan’an University, Yan’an, China
| | - Xiao Long Liu
- School of Medicine, Yan’an University, Yan’an, China
| |
Collapse
|
22
|
Pang Q, Tang Z, Luo L. The crosstalk between oncogenic signaling and ferroptosis in cancer. Crit Rev Oncol Hematol 2024; 197:104349. [PMID: 38626848 DOI: 10.1016/j.critrevonc.2024.104349] [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: 09/30/2023] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Ferroptosis, a novel form of cell death regulation, was identified in 2012. It is characterized by unique features that differentiate it from other types of cell death, including necrosis, apoptosis, autophagy, and pyroptosis. Ferroptosis is defined by an abundance of iron ions and lipid peroxidation, resulting in alterations in subcellular structures, an elevation in reactive oxygen species (ROS), a reduction in glutathione (GSH) levels, and an augmentation in Fe (II) cytokines. Ferroptosis, a regulated process, is controlled by an intricate network of signaling pathways, where multiple stimuli can either enhance or hinder the process. This review primarily examines the defensive mechanisms of ferroptosis and its interaction with the tumor microenvironment. The analysis focuses on the pathways that involve AMPK, p53, NF2, mTOR, System Xc-, Wnt, Hippo, Nrf2, and cGAS-STING. The text discusses the possibilities of employing a combination therapy that targets several pathways for the treatment of cancer. It emphasizes the necessity for additional study in this field.
Collapse
Affiliation(s)
- Qianghu Pang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Zhirou Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang,School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|